Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Epigenetic suppression of neuroligin 1 underlies amyloid-induced memory deficiency

Abstract

Amyloid-induced microglial activation and neuroinflammation impair central synapses and memory function, although the mechanism remains unclear. Neuroligin 1 (NLGN1), a postsynaptic protein found in central excitatory synapses, governs excitatory synaptic efficacy and plasticity in the brain. Here we found, in rodents, that amyloid fibril–induced neuroinflammation enhanced the interaction between histone deacetylase 2 and methyl-CpG-binding protein 2, leading to suppressed histone H3 acetylation and enhanced cytosine methylation in the Nlgn1 promoter region and decreased NLGN1 expression, underlying amyloid-induced memory deficiency. Manipulation of microglia-associated neuroinflammation modulated the epigenetic modification of the Nlgn1 promoter, hippocampal glutamatergic transmission and memory function. These findings link neuroinflammation, synaptic efficacy and memory, thus providing insight into the pathogenesis of amyloid-associated diseases.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Epigenetic suppression of NLGN1 underlies amyloid fibril–induced memory deficiency.
Figure 2: Enhanced HDAC2 activity was involved in amyloid fibril–suppressed NLGN1 expression.
Figure 3: Amyloid fibrils induced functional modification of MeCP2 activity in hippocampal CA1 tissue.
Figure 4: Suppression of MeCP2 activity ameliorated amyloid fibril–suppressed NLGN1 expression and memory.
Figure 5: LPS-induced suppression of NLGN1 expression.
Figure 6: LPS impaired hippocampal glutamatergic transmission and memory in naive rats.
Figure 7: Microinjection of minocycline (5 μg per side) ameliorated amyloid fibril–suppressed NLGN1 expression.
Figure 8: Microinjection of minocycline restored hippocampal glutamatergic transmission and memory impaired by amyloid fibrils.

Similar content being viewed by others

References

  1. Hardy, J. & Selkoe, D.J. The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics. Science 297, 353–356 (2002).

    Article  CAS  PubMed  Google Scholar 

  2. Middei, S., Geracitano, R., Caprioli, A., Mercuri, N. & Ammassari-Teule, M. Preserved fronto-striatal plasticity and enhanced procedural learning in a transgenic mouse model of Alzheimer's disease overexpressing mutant hAPPswe. Learn. Mem. 11, 447–452 (2004).

    Article  PubMed Central  PubMed  Google Scholar 

  3. Südhof, T.C. Neuroligins and neurexins link synaptic function to cognitive disease. Nature 455, 903–911 (2008).

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  4. Choi, Y.B. et al. Neurexin-neuroligin transsynaptic interaction mediates learning-related synaptic remodeling and long-term facilitation in Aplysia. Neuron 70, 468–481 (2011).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  5. Ye, X. & Carew, T.J. Transsynaptic coordination of presynaptic and postsynaptic modifications underlying enduring synaptic plasticity. Neuron 70, 379–381 (2011).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  6. Blundell, J. et al. Neuroligin-1 deletion results in impaired spatial memory and increased repetitive behavior. J. Neurosci. 30, 2115–2129 (2010).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  7. Dinamarca, M.C., Weinstein, D., Monasterio, O. & Inestrosa, N.C. The synaptic protein neuroligin-1 interacts with the amyloid beta-peptide. Is there a role in Alzheimer's disease? Biochemistry 50, 8127–8137 (2011).

    Article  CAS  PubMed  Google Scholar 

  8. Saura, C.A., Servian-Morilla, E. & Scholl, F.G. Presenilin/gamma-secretase regulates neurexin processing at synapses. PLoS ONE 6, e19430 (2011).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  9. Guan, J.S. et al. HDAC2 negatively regulates memory formation and synaptic plasticity. Nature 459, 55–60 (2009).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  10. Gräff, J. et al. An epigenetic blockade of cognitive functions in the neurodegenerating brain. Nature 483, 222–226 (2012).

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  11. Lombardi, P.M., Cole, K.E., Dowling, D.P. & Christianson, D.W. Structure, mechanism, and inhibition of histone deacetylases and related metalloenzymes. Curr. Opin. Struct. Biol. 21, 735–743 (2011).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  12. Guy, J., Cheval, H., Selfridge, J. & Bird, A. The role of MeCP2 in the brain. Annu. Rev. Cell Dev. Biol. 27, 631–652 (2011).

    Article  CAS  PubMed  Google Scholar 

  13. Na, E.S., Nelson, E.D., Kavalali, E.T. & Monteggia, L.M. The impact of MeCP2 loss- or gain-of-function on synaptic plasticity. Neuropsychopharmacology 38, 212–219 (2013).

    Article  CAS  PubMed  Google Scholar 

  14. Yoon, H.G., Chan, D.W., Reynolds, A.B., Qin, J. & Wong, J. N-CoR mediates DNA methylation-dependent repression through a methyl CpG binding protein Kaiso. Mol. Cell 12, 723–734 (2003).

    Article  CAS  PubMed  Google Scholar 

  15. Jones, P.L. et al. Methylated DNA and MeCP2 recruit histone deacetylase to repress transcription. Nat. Genet. 19, 187–191 (1998).

    Article  CAS  PubMed  Google Scholar 

  16. Wyss-Coray, T. Inflammation in Alzheimer disease: driving force, bystander or beneficial response? Nat. Med. 12, 1005–1015 (2006).

    CAS  PubMed  Google Scholar 

  17. Grathwohl, S.A. et al. Formation and maintenance of Alzheimer's disease beta-amyloid plaques in the absence of microglia. Nat. Neurosci. 12, 1361–1363 (2009).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  18. Wu, J. et al. Activation of the CB2 receptor system reverses amyloid-induced memory deficiency. Neurobiol. Aging 34, 791–804 (2013).

    Article  CAS  PubMed  Google Scholar 

  19. Solito, E. & Sastre, M. Microglia function in Alzheimer's disease. Front. Pharmacol. 3, 14 (2012).

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  20. Chacón, M.A., Barria, M.I., Soto, C. & Inestrosa, N.C. Beta-sheet breaker peptide prevents Aβ-induced spatial memory impairments with partial reduction of amyloid deposits. Mol. Psychiatry 9, 953–961 (2004).

    Article  PubMed  CAS  Google Scholar 

  21. Sun, B. et al. Imbalance between GABAergic and glutamatergic transmission impairs adult neurogenesis in an animal model of Alzheimer's disease. Cell Stem Cell 5, 624–633 (2009).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  22. Bie, B. et al. Upregulation of nerve growth factor in central amygdala increases sensitivity to opioid reward. Neuropsychopharmacology 37, 2780–2788 (2012).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  23. Suzuki, K. et al. Activity-dependent proteolytic cleavage of neuroligin-1. Neuron 76, 410–422 (2012).

    Article  CAS  PubMed  Google Scholar 

  24. Fischer, A., Sananbenesi, F., Mungenast, A. & Tsai, L.H. Targeting the correct HDAC(s) to treat cognitive disorders. Trends Pharmacol. Sci. 31, 605–617 (2010).

    Article  CAS  PubMed  Google Scholar 

  25. Zhang, Z., Cai, Y.Q., Zou, F., Bie, B. & Pan, Z.Z. Epigenetic suppression of GAD65 expression mediates persistent pain. Nat. Med. 17, 1448–1455 (2011).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  26. Tao, J. et al. Phosphorylation of MeCP2 at serine 80 regulates its chromatin association and neurological function. Proc. Natl. Acad. Sci. USA 106, 4882–4887 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Zhou, Z. et al. Brain-specific phosphorylation of MeCP2 regulates activity-dependent Bdnf transcription, dendritic growth, and spine maturation. Neuron 52, 255–269 (2006).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  28. Bie, B. et al. Nerve growth factor-regulated emergence of functional delta-opioid receptors. J. Neurosci. 30, 5617–5628 (2010).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  29. Nan, X. et al. Transcriptional repression by the methyl-CpG-binding protein MeCP2 involves a histone deacetylase complex. Nature 393, 386–389 (1998).

    Article  CAS  PubMed  Google Scholar 

  30. Jankowsky, J.L. et al. Mutant presenilins specifically elevate the levels of the 42 residue beta-amyloid peptide in vivo: evidence for augmentation of a 42-specific gamma secretase. Hum. Mol. Genet. 13, 159–170 (2004).

    Article  CAS  PubMed  Google Scholar 

  31. Ransohoff, R.M. & Cardona, A.E. The myeloid cells of the central nervous system parenchyma. Nature 468, 253–262 (2010).

    Article  CAS  PubMed  Google Scholar 

  32. Prinz, M., Priller, J., Sisodia, S.S. & Ransohoff, R.M. Heterogeneity of CNS myeloid cells and their roles in neurodegeneration. Nat. Neurosci. 14, 1227–1235 (2011).

    Article  CAS  PubMed  Google Scholar 

  33. Dean, C. & Dresbach, T. Neuroligins and neurexins: linking cell adhesion, synapse formation and cognitive function. Trends Neurosci. 29, 21–29 (2006).

    Article  CAS  PubMed  Google Scholar 

  34. Ting, J.T., Peca, J. & Feng, G. Functional consequences of mutations in postsynaptic scaffolding proteins and relevance to psychiatric disorders. Annu. Rev. Neurosci. 35, 49–71 (2012).

    Article  CAS  PubMed  Google Scholar 

  35. Chih, B., Engelman, H. & Scheiffele, P. Control of excitatory and inhibitory synapse formation by neuroligins. Science 307, 1324–1328 (2005).

    Article  CAS  PubMed  Google Scholar 

  36. Levinson, J.N. et al. Neuroligins mediate excitatory and inhibitory synapse formation: involvement of PSD-95 and neurexin-1beta in neuroligin-induced synaptic specificity. J. Biol. Chem. 280, 17312–17319 (2005).

    Article  CAS  PubMed  Google Scholar 

  37. Lee, S.T. et al. miR-206 regulates brain-derived neurotrophic factor in Alzheimer disease model. Ann. Neurol. 72, 269–277 (2012).

    Article  CAS  PubMed  Google Scholar 

  38. Sultana, R., Banks, W.A. & Butterfield, D.A. Decreased levels of PSD95 and two associated proteins and increased levels of BCl2 and caspase 3 in hippocampus from subjects with amnestic mild cognitive impairment: insights into their potential roles for loss of synapses and memory, accumulation of Aβ, and neurodegeneration in a prodromal stage of Alzheimer's disease. J. Neurosci. Res. 88, 469–477 (2010).

    CAS  PubMed Central  PubMed  Google Scholar 

  39. Zeng, Y. et al. Epigenetic enhancement of BDNF signaling rescues synaptic plasticity in aging. J. Neurosci. 31, 17800–17810 (2011).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  40. Skene, P.J. et al. Neuronal MeCP2 is expressed at near histone-octamer levels and globally alters the chromatin state. Mol. Cell 37, 457–468 (2010).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  41. Kishi, N. & Macklis, J.D. MECP2 is progressively expressed in post-migratory neurons and is involved in neuronal maturation rather than cell fate decisions. Mol. Cell. Neurosci. 27, 306–321 (2004).

    Article  CAS  PubMed  Google Scholar 

  42. Adkins, N.L. & Georgel, P.T. MeCP2: structure and function. Biochem. Cell Biol. 89, 1–11 (2011).

    Article  CAS  PubMed  Google Scholar 

  43. Samaco, R.C. & Neul, J.L. Complexities of Rett syndrome and MeCP2. J. Neurosci. 31, 7951–7959 (2011).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  44. Cohen, S. et al. Genome-wide activity-dependent MeCP2 phosphorylation regulates nervous system development and function. Neuron 72, 72–85 (2011).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  45. Li, H., Zhong, X., Chau, K.F., Williams, E.C. & Chang, Q. Loss of activity-induced phosphorylation of MeCP2 enhances synaptogenesis, LTP and spatial memory. Nat. Neurosci. 14, 1001–1008 (2011).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  46. Gonzales, M.L., Adams, S., Dunaway, K.W. & LaSalle, J.M. Phosphorylation of distinct sites in MeCP2 modifies cofactor associations and the dynamics of transcriptional regulation. Mol. Cell. Biol. 32, 2894–2903 (2012).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  47. Rutlin, M. & Nelson, S.B. MeCP2: phosphorylated locally, acting globally. Neuron 72, 3–5 (2011).

    Article  CAS  PubMed  Google Scholar 

  48. Kimura, H. & Shiota, K. Methyl-CpG-binding protein, MeCP2, is a target molecule for maintenance DNA methyltransferase, Dnmt1. J. Biol. Chem. 278, 4806–4812 (2003).

    Article  CAS  PubMed  Google Scholar 

  49. Dong, E., Guidotti, A., Grayson, D.R. & Costa, E. Histone hyperacetylation induces demethylation of reelin and 67-kDa glutamic acid decarboxylase promoters. Proc. Natl. Acad. Sci. USA 104, 4676–4681 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Correa, F., Mallard, C., Nilsson, M. & Sandberg, M. Activated microglia decrease histone acetylation and Nrf2-inducible anti-oxidant defence in astrocytes: restoring effects of inhibitors of HDACs, p38 MAPK and GSK3beta. Neurobiol. Dis. 44, 142–151 (2011).

    CAS  PubMed Central  PubMed  Google Scholar 

  51. Paxinos, G. & Watson, C. The Rat Brain in Stereotaxic Coordinates (Academic, New York, 1998).

  52. Shin, R.W. et al. Amyloid beta-protein (Aβ) 1–40 but not Aβ1–42 contributes to the experimental formation of Alzheimer disease amyloid fibrils in rat brain. J. Neurosci. 17, 8187–8193 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Ahmed, T., Enam, S.A. & Gilani, A.H. Curcuminoids enhance memory in an amyloid-infused rat model of Alzheimer's disease. Neuroscience 169, 1296–1306 (2010).

    Article  CAS  PubMed  Google Scholar 

  54. Bie, B., Zhu, W. & Pan, Z.Z. Ethanol-induced delta-opioid receptor modulation of glutamate synaptic transmission and conditioned place preference in central amygdala. Neuroscience 160, 348–358 (2009).

    Article  CAS  PubMed  Google Scholar 

  55. Lubin, F.D., Roth, T.L. & Sweatt, J.D. Epigenetic regulation of BDNF gene transcription in the consolidation of fear memory. J. Neurosci. 28, 10576–10586 (2008).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  56. Bie, B., Peng, Y., Zhang, Y. & Pan, Z.Z. cAMP-mediated mechanisms for pain sensitization during opioid withdrawal. J. Neurosci. 25, 3824–3832 (2005).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  57. Bie, B., Brown, D.L. & Naguib, M. Increased synaptic GluR1 subunits in the anterior cingulate cortex of rats with peripheral inflammation. Eur. J. Pharmacol. 653, 26–31 (2011).

    Article  CAS  PubMed  Google Scholar 

  58. Gallicano, G.I. et al. Desmoplakin is required early in development for assembly of desmosomes and cytoskeletal linkage. J. Cell Biol. 143, 2009–2022 (1998).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  59. DeFelipe, J., Marco, P., Busturia, I. & Merchán-Pérez, A. Estimation of the number of synapses in the cerebral cortex: methodological considerations. Cereb. Cortex 9, 722–732 (1999).

    Article  CAS  PubMed  Google Scholar 

  60. Bie, B., Zhu, W. & Pan, Z.Z. Rewarding morphine-induced synaptic function of delta-opioid receptors on central glutamate synapses. J. Pharmacol. Exp. Ther. 329, 290–296 (2009).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  61. Shipton, O.A. et al. Tau protein is required for amyloid β-induced impairment of hippocampal long-term potentiation. J. Neurosci. 31, 1688–1692 (2011).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

Download references

Acknowledgements

Support of the Lerner Research Institute at the Cleveland Clinic.

Author information

Authors and Affiliations

Authors

Contributions

B.B. and J.W. performed most of the experiments and contributed to a draft of the manuscript. J.W. performed all behavioral testing. H.Y. and J.J.X. conducted some of the molecular experiments. D.L.B. and M.N. designed and directed the project, reviewed the experimental data, performed data analyses and wrote the final manuscript. All authors commented on the final version of the manuscript.

Corresponding author

Correspondence to Mohamed Naguib.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Amyloid fibrils induced memory deficiency and impaired glutamatergic transmission in the hippocampal CA1 neurons.

Significantly extended escape latency (a, n = 10 rats in each group, F(2,27) = 17.0, P<0.0001) and less time spent in the target quadrant (b, n = 10 rats in each group, F(2,27) =8.6, P = 0.001) were observed in the rats microinjected with Aβ1-40 fibrils, but not Aβ40-1 fibrils nor artificial CSF (control). Significantly attenuated input (stimulus intensity) - output (EPSC amplitude) response of evoked EPSC (c, n = 11,11,10 neurons in each group, F(2,29) = 14.5, P<0.0001) and HFS-induced LTP (d, n = 11,9,11 neurons per group, F(2,28) =8.37, P = 0.001) were also observed in the hippocampal CA1 neurons of the rats microinjected with Aβ1-40 fibrils, but not Aβ40-1 fibrils nor artificial CSF. (e) Aβ1-40, but not Aβ40-1, fibrils significantly decreased hippocampal NLGN1 expression (n = 6 rats per group, F(2,15) = 9.3, P = 0.002). Data represent mean ± s.e.m. *, P<0.05, **, P<0.01. Full length blots are presented in supplementary figure 11.

Supplementary Figure 2 Amyloid fibrils preferentially attenuated hippocampal glutamatergic transmission.

(a) Amyloid fibrils (Aβ1-40) significantly increased the paired-pulse ratio of evoked EPSC, indicating decreased presynaptic glutamate release in hippocampal neurons (n = 11 and 9 neurons, F(1,18) = 14.1, P = 0.002), and decreased the exogenous AMPA (1 μM)-induced inward currents, indicating attenuated postsynaptic response in hippocampal CA1 neurons (b, n = 11 and 8 neurons, t = 2.5, P = 0.022). Representative traces of mEPSC and mIPSC of hippocampal CA1 neurons in control and amyloid-injected rats were shown (c). Amyloid fibrils significantly decreased the amplitude (d, n = 13 and 17 neurons, t = 2.8, P = 0.009) and frequency (e, n = 13 and 17 neurons, t = 2.4, P = 0.025) of mEPSC, but not mIPSC, in hippocampal CA1 neurons. (f) Amyloid fibrils decreased the mEPSC/mIPSC ratio of amplitude (n = 17 and 19 neurons, t = 2.3, P = 0.03) and frequency (n = 17 and 19 neurons, t = 4.8, P<0.001) in hippocampal CA1 neurons. (g) Immunofluorescence for inhibitory GABAergic presynaptic marker GAD67 (green) and excitatory glutamatergic presynaptic marker vGLUT1 (violet) in the hippocampal CA1 in control and amyloid-injected rats. Note a marked decrease in vGLUT1, but not GAD67, immunoreactivities was observed (n = 25 sections from 5 rats in each group, t = 2.456, P = 0.39, scale bar = 25 μm). Data represent mean ± s.e.m. *, P<0.05, **, P<0.01.

Supplementary Figure 3 Amyloid fibrils did not change the expression of NLGN2, neurexin-1α, ADAM10, or presenilin 1 in the hippocampal CA1 area, and microinjection of Nlgn1 siRNA significantly decreased the hippocampal expression of NLGN1.

(a) Amyloid fibrils did not significantly change the hippocampal protein levels of NLGN2 (n = 8 and 9 rats per group, t = 0.1, P = 0.907) and neurexin-1α (n = 8 and 9 rats per group, t = 0.3, P = 0.779). (b) Amyloid fibrils did not significantly affect the hippocampal protein levels of ADAM10 (n = 6 and 9 rats per group, t = 0.7, P = 0.497) and presenilin 1 (n = 9 rats per group, t = 0.3, P = 0.763). Significantly decreased mRNA (c, n = 6 rats in each group, F(2,15) = 5.9, P = 0.01) and protein (d, n = 6 rats in each group, F(2,15) = 4.0, P = 0.03) levels of NLGN1 were observed in the hippocampal CA1 area after Nlgn1 siRNA (5 nmol per side), but not scrambled RNA (scRNA) microinjection. Data represent mean ± s.e.m. Full length blots are presented in supplementary figure 11.

Supplementary Figure 4 Amyloid fibrils increased the HDAC2 immunosignal in the hippocampal CA1 neurons, which was attenuated by microinjection of Hdac2 siRNA.

Amyloid fibrils increased the HDAC2 immunosignal in the hippocampal CA1 neurons, which was attenuated by microinjection of Hdac2 siRNA. Microinjection of Hdac2 siRNA (5 nmol per side), but not scrambled RNA (scRNA), significantly attenuated amyloid-upregulated Hdac2 mRNA levels (a, n = 6 rats per group, F(3,20) = 8.8, P = 0.0006) and protein (b, n = 6 rats per group, F(3,20) = 8.1, P<0.001) levels in the hippocampal CA1 area. The expression of HDAC2 was largely colocalized with the immunofluorescence signal of NeuN (c), the marker of neuron, but not GFAP (d), the marker of astrocyte. Microinjection of Hdac2 siRNA, but not scrambled RNA (scRNA), significantly attenuated the amyloid fibrils-upregulated immunofluorescence signal of HDAC2 in the hippocampal CA1 neurons (n = 24 sections from 5 rats in each group, F(3,16) = 11.5, P = 0.0003, Scale bar = 25 μm). (e) Microinjection of Hdac2 siRNA also significantly attenuated amyloid-decreased synapse number in the hippocampal CA1 area. Total synapse density was calculated as the number of synapses per 100 μm2 of neuropil (n = 5 rats per group, F(3,16) = 12.77, P = 0.0002, scale bar = 0.5 μm). Data represent mean ± s.e.m. *, P<0.05, **, P<0.01. Full length blots are presented in supplementary figure 11.

Supplementary Figure 5 Microinjection of suberoylanilide hydroxamic acid (SAHA) significantly restored amyloid-impaired hippocampal NLGN1 expression, glutamatergic transmission and memory.

SAHA (0.3 nmol per side for 3 days) significantly increased histone H3 acetylation across the Nlgn1 promoter region (a, n = 6 rats per group, F(3,20) = 5.3, P = 0.008), mRNA (b, n = 6 rats per group, F(3,20) = 6.7, P = 0.003) and protein (c, n = 8,8,6,6 rats per group, F(3,20) = 9.4, P = 0.0003) levels of the hippocampal NLGN1 in the rats injected with amyloid fibrils. SAHA significantly enhanced evoked EPSC amplitudes (d, n = 11,11,10,9 neurons in each group, F(3,37) = 10.8, P<0.0001) and HFS-induced LTP (e, n = 10,11,9,9 neurons in each group, F(3,35) = 7.96, P = 0.0004) in the hippocampal CA1 neurons of the amyloid-injected rats. It also significantly shortened the escape latencies (f, n = 10 rats per group, F(3,36) = 13.7, P<0.0001) and increased the time spent in the target quadrant (g, n = 10 rats per group, F(3,36) = 8.9, P = 0.0002) for amyloid-injected rats in the Morris water maze test. Data represent mean ± s.e.m. *, P<0.05, **, P<0.01. Full length blots are presented in supplementary figure 11.

Supplementary Figure 6 Amyloid fibrils increased the MeCP2 immunosignal in the hippocampal CA1 neurons, which was attenuated by microinjection of Mecp2 siRNA.

Amyloid-induced increased in the MeCP2 immunosignal was significantly attenuated by microinjection of Mecp2 siRNA (5 nmol per side), but not scrambled RNA (scRNA) (a, n = 24 sections from 5 rats in each group, F(3,16) = 5.8, P = 0.0071, scale bar = 25 μm). The expression of MeCP2 was largely colocalized with the immunofluorescence signal of NeuN (a), the marker of neuron, but not GFAP (b), the marker of astrocyte. Microinjection of Mecp2 siRNA significantly decreased the mRNA (c, n = 6 rats per group, F(3,20) = 10.1, P<0.01) and protein (d, n = 6 rats per group, F(3,20) = 34.4, P<0.0001) levels of hippocampal MeCP2 in the amyloid-injected rats. Microinjection of Mecp2 siRNA also significantly attenuated amyloid-decreased synapse number in the hippocampal CA1 area (e, n = 5 rats per group, F(3,16) = 25.6, P<0.0001, scale bar = 0.5 μm). Total synapse density was calculated as the number of synapses per 100 μm2 of neuropil. *, P<0.05, **, P<0.01. Data represent mean ± s.e.m. Full length blots are presented in supplementary figure 11.

Supplementary Figure 7 Amyloid fibrils induced significant change in mSin3A but not HDAC1 expression in the hippocampal CA1 area.

(a) Significant increase in protein levels of mSin3A (n = 6 and 9 rats per group, t = 5.6, P<0.0001), but not HDAC1 (n = 9 rats in each group, t = 0.9, P = 0.360), in the hippocampal CA1 of amyloid-injected rats. (b) Significant increase in binding of mSin3A (n = 9 rats per group, t = 4.1, P = 0.0008), but not HDAC1 (n = 6 rats per group, t = 0.4, P = 0.718), with MeCP2 in the hippocampal CA1 of amyloid-injected rats. Significant increase in binding of mSin3A (d, n = 6 rats per group, t = 2.7, P = 0.024), but not HDAC1 (c, n = 6 rats per group, t = 0.3, P = 0.772), in Nlgn1 promoter region in the hippocampal CA1 of amyloid-injected rats. Data represent mean ± s.e.m. *, P<0.05, **, P<0.01. Full length blots are presented in supplementary figure 11.

Supplementary Figure 8 Epigenetic modification of NLGN1 expression in APP/PSEN1 mice.

Significantly increased expression of HDAC2 (a, n = 9 mice per group, t = 3.8, P = 0.002) and MeCP2 (n = 5 and 6 mice per group, t = 12.0, P<0.0001) as well as increased formation of complex containing HDAC2 and MeCP2 (b, n = 5 mice per group, t = 4.9, P = 0.001) were observed in the hippocampal CA1 of APP/PSEN1 mice. The occupancy of HDAC2 (c, n = 6 mice per group, t = 2.7, P = 0.023) and MeCP2 (d, n = 6 mice per group, t = 4.1, P = 0.002) were increased in Nlgn1, but not Gpdh, promoter region in these transgenic mice. Significantly decreased histone H3 acetylation (e, n = 6 mice per group, t = 3.1, P = 0.01) and increased cytosine methylation (f, n = 6 mice per group, t = 3.7, P = 0.004) were also observed in Nlgn1, but not Gpdh, promoter region in these transgenic mice. The expression of NLGN1 was decreased in hippocampal CA1 of these transgenic mice (g, n = 8 and 10 mice in each group, t = 7.2, P<0.0001). Data represent mean ± s.e.m. *, P<0.05, **, P<0.01. Full length blots are presented in supplementary figure 11.

Supplementary Figure 9 Microinjection of lipopolysaccharide (LPS, 5 μg per side), like that of amyloid fibrils, induced significant activation of microglia in the hippocampal CA1 area.

Significantly increased CD11b expression was observed in the hippocampal CA1 area of the rats injected with amyloid fibrils (a, n = 3-4 sections from 5 rats, t = 13.5, P<0.0001, scale bar = 40 μm.) or LPS (b, n = 3-4 sections from 5 rats in each group, t = 15.1, P<0.0001). Microinjection of LPS also significantly decreased synapse number in the hippocampal CA1 area (c, n = 5 rats per group, t = 6.862, P = 0.0001, scale bar = 0.5 μm). Total synapse density was calculated as the number of synapses per 100 μm2 of neuropil. Data represent mean ± s.e.m.**, P<0.01.

Supplementary Figure 10 Microinjection of minocycline (5 μg per side) suppressed amyloid fibril—induced microglial activity in the hippocampal CA1 area.

Immunostaining revealed significantly decreased CD11b expression in the hippocampal CA1 area of the modeled rats injected with minocycline (mino) (a, n = 3-4 sections from 5 rats in each group, F(3,72) = 98.9, P<0.0001, **, P<0.01, scale bar = 40 μm). Microinjection of minocycline also prevented amyloid fibril-induced upsurge in IL-1β synthesis (b, n = 6 rats in each group, F(4,25) = 9.4, P=0.001, *, P<0.05). Microinjection of minocycline (5 μg per side) significantly attenuated amyloid-decreased synapse number in the hippocampal CA1 area (c, n = 5 rats per group, F(3,16) = 21.4, P<0.0001, scale bar = 0.5 μm). Total synapse density was calculated as the number of synapses per 100 μm2 of neuropil. Data represent mean ± s.e.m. *, P<0.01, **, P<0.01.

Supplementary Figure 11 Full-length blots.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–11 (PDF 4143 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Bie, B., Wu, J., Yang, H. et al. Epigenetic suppression of neuroligin 1 underlies amyloid-induced memory deficiency. Nat Neurosci 17, 223–231 (2014). https://doi.org/10.1038/nn.3618

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.3618

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing