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:

C/EBPβ/δ-secretase signaling mediates Parkinson’s disease pathogenesis via regulating transcription and proteolytic cleavage of α-synuclein and MAOB

Abstract

Parkinson’s disease (PD) is characterized by dopaminergic neuronal loss and the presence of intra-neuronal Lewy body (LB) inclusions with aggregated α-synuclein (α-Syn) as the major component. MAOB, a crucial monoamine oxidase for dopamine metabolism, triggers oxidative stress in dopaminergic neurons and α-Syn aggregation. However, the key molecular mechanism that mediates PD pathogenesis remains elusive. Here we show that C/EBPβ acts as an age-dependent transcription factor for both α-Syn and MAOB, and initiates the PD pathologies by upregulating these two pivotal players, in addition to escalating δ-secretase activity to cleave α-Syn and promotes its neurotoxicity. Overexpression of C/EBPβ in human wild-type α-Syn transgenic mice facilitates PD pathologies and elicits motor disorders associated with augmentation of δ-secretase, α-Syn, and MAOB. In contrast, depletion of C/EBPβ from human α-Syn Tg mice abolishes rotenone-elicited PD pathologies and motor impairments via downregulating the expression of these key factors. Hence, our study supports that C/EBPβ/δ-secretase signaling mediates PD pathogenesis via regulating the expression and cleavage of α-Syn and MAOB.

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

Fig. 1: C/EBPβ acts as transcription factors for MAOB and α-Syn.
Fig. 2: C/EBPβ regulates PD neurotoxin-induced MAOB and α-Syn expression in primary neurons.
Fig. 3: C/EBPβ regulates MAOB and α-Syn expression in an age-dependent manner.
Fig. 4: C/EBPβ mediates α-Syn and MAOB expression in human PD neurons and brains.
Fig. 5: Knockout of C/EBPβ attenuates MAOB and α-Syn protein expression.
Fig. 6: C/EBPβ mediates PD pathologies in human α-Syn Tg mice.

Similar content being viewed by others

References

  1. Savitt JM, Dawson VL, Dawson TM. Diagnosis and treatment of Parkinson disease: molecules to medicine. J Clin Invest. 2006;116:1744–54.

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Olanow CW, Brundin P. Parkinson’s disease and alpha synuclein: is Parkinson’s disease a prion-like disorder? Mov Disord. 2013;28:31–40.

    CAS  PubMed  Google Scholar 

  3. Kruger R, Kuhn W, Muller T, Woitalla D, Graeber M, Kosel S, et al. Ala30Pro mutation in the gene encoding alpha-synuclein in Parkinson’s disease. Nat Genet. 1998;18:106–8.

    CAS  PubMed  Google Scholar 

  4. Zarranz JJ, Alegre J, Gomez-Esteban JC, Lezcano E, Ros R, Ampuero I, et al. The new mutation, E46K, of alpha-synuclein causes Parkinson and Lewy body dementia. Ann Neurol. 2004;55:164–73.

    CAS  PubMed  Google Scholar 

  5. Polymeropoulos MH, Lavedan C, Leroy E, Ide SE, Dehejia A, Dutra A, et al. Mutation in the alpha-synuclein gene identified in families with Parkinson’s disease. Science. 1997;276:2045–7.

    CAS  PubMed  Google Scholar 

  6. Simon-Sanchez J, Schulte C, Bras JM, Sharma M, Gibbs JR, Berg D, et al. Genome-wide association study reveals genetic risk underlying Parkinson’s disease. Nat Genet. 2009;41:1308–12.

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Yasuda T, Nakata Y, Mochizuki H. alpha-Synuclein and neuronal cell death. Mol Neurobiol. 2013;47:466–83.

    CAS  PubMed  Google Scholar 

  8. Saura J, Richards JG, Mahy N. Differential age-related changes of MAO-A and MAO-B in mouse brain and peripheral organs. Neurobiol Aging. 1994;15:399–408.

    CAS  PubMed  Google Scholar 

  9. Mahy N, Andres N, Andrade C, Saura J. Age-related changes of MAO-A and -B distribution in human and mouse brain. Neurobiology. 2000;8:47–54.

    CAS  PubMed  Google Scholar 

  10. Wei Q, Yeung M, Jurma OP, Andersen JK. Genetic elevation of monoamine oxidase levels in dopaminergic PC12 cells results in increased free radical damage and sensitivity to MPTP. J Neurosci Res. 1996;46:666–73.

    CAS  PubMed  Google Scholar 

  11. Wei Q, Jurma OP, Andersen JK. Increased expression of monoamine oxidase-B results in enhanced neurite degeneration in methamphetamine-treated PC12 cells. J Neurosci Res. 1997;50:618–26.

    CAS  PubMed  Google Scholar 

  12. Chen JF, Steyn S, Staal R, Petzer JP, Xu K, Van Der Schyf CJ, et al. 8-(3-Chlorostyryl)caffeine may attenuate MPTP neurotoxicity through dual actions of monoamine oxidase inhibition and A2A receptor antagonism. J Biol Chem. 2002;277:36040–4.

    CAS  PubMed  Google Scholar 

  13. Grimsby J, Toth M, Chen K, Kumazawa T, Klaidman L, Adams JD, et al. Increased stress response and beta-phenylethylamine in MAOB-deficient mice. Nat Genet. 1997;17:206–10.

    CAS  PubMed  Google Scholar 

  14. Mallajosyula JK, Kaur D, Chinta SJ, Rajagopalan S, Rane A, Nicholls DG, et al. MAO-B elevation in mouse brain astrocytes results in Parkinson’s pathology. PLoS One. 2008;3:e1616.

    PubMed  PubMed Central  Google Scholar 

  15. Siddiqui A, Mallajosyula JK, Rane A, Andersen JK. Ability to delay neuropathological events associated with astrocytic MAO-B increase in a Parkinsonian mouse model: implications for early intervention on disease progression. Neurobiol Dis. 2011;43:527–32.

    CAS  PubMed  Google Scholar 

  16. Lieu CA, Chinta SJ, Rane A, Andersen JK. Age-related behavioral phenotype of an astrocytic monoamine oxidase-B transgenic mouse model of Parkinson’s disease. PLoS One. 2013;8:e54200.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Zhang Z, Kang SS, Liu X, Ahn EH, Zhang Z, He L, et al. Asparagine endopeptidase cleaves alpha-synuclein and mediates pathologic activities in Parkinson’s disease. Nat Struct Mol Biol. 2017;24:632–42.

    PubMed  PubMed Central  Google Scholar 

  18. Kang SS, Ahn EH, Zhang Z, Liu X, Manfredsson FP, Sandoval IM et al. alpha-Synuclein stimulation of monoamine oxidase-B and legumain protease mediates the pathology of Parkinson’s disease. EMBO J. 2018;37:e98878.

  19. Pulido-Salgado M, Vidal-Taboada JM, Saura J. C/EBPbeta and C/EBPdelta transcription factors: basic biology and roles in the CNS. Prog Neurobiol. 2015;132:1–33.

    CAS  PubMed  Google Scholar 

  20. Magalini A, Savoldi G, Ferrari F, Garnier M, Ghezzi P, Albertini A, et al. Role of IL-1 beta and corticosteroids in the regulation of the C/EBP-alpha, beta and delta genes in vivo. Cytokine. 1995;7:753–8.

    CAS  PubMed  Google Scholar 

  21. Poli V. The role of C/EBP isoforms in the control of inflammatory and native immunity functions. J Biol Chem. 1998;273:29279–82.

    CAS  PubMed  Google Scholar 

  22. Wedel A, Ziegler-Heitbrock HW. The C/EBP family of transcription factors. Immunobiology. 1995;193:171–85.

    CAS  PubMed  Google Scholar 

  23. Straccia M, Gresa-Arribas N, Dentesano G, Ejarque-Ortiz A, Tusell JM, Serratosa J, et al. Pro-inflammatory gene expression and neurotoxic effects of activated microglia are attenuated by absence of CCAAT/enhancer binding protein beta. J Neuroinflamm. 2011;8:156.

    CAS  Google Scholar 

  24. Gomez-Santos C, Barrachina M, Gimenez-Xavier P, Dalfo E, Ferrer I, Ambrosio S. Induction of C/EBP beta and GADD153 expression by dopamine in human neuroblastoma cells. Relationship with alpha-synuclein increase and cell damage. Brain Res Bull. 2005;65:87–95.

    CAS  PubMed  Google Scholar 

  25. Wang ZH, Gong K, Liu X, Zhang Z, Sun X, Wei ZZ, et al. C/EBPbeta regulates delta-secretase expression and mediates pathogenesis in mouse models of Alzheimer’s disease. Nat Commun. 2018;9:1784.

    PubMed  PubMed Central  Google Scholar 

  26. Liu Z, Jang SW, Liu X, Cheng D, Peng J, Yepes M, et al. Neuroprotective actions of PIKE-L by inhibition of SET proteolytic degradation by asparagine endopeptidase. Mol Cell. 2008;29:665–78.

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Hatcher JM, Pennell KD, Miller GW. Parkinson’s disease and pesticides: a toxicological perspective. Trends Pharmacol Sci. 2008;29:322–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Betarbet R, Sherer TB, MacKenzie G, Garcia-Osuna M, Panov AV, Greenamyre JT. Chronic systemic pesticide exposure reproduces features of Parkinson’s disease. Nat Neurosci. 2000;3:1301–6.

    CAS  PubMed  Google Scholar 

  29. Cannon JR, Tapias V, Na HM, Honick AS, Drolet RE, Greenamyre JT. A highly reproducible rotenone model of Parkinson’s disease. Neurobiol Dis. 2009;34:279–90.

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Ramsay RR, Salach JI, Dadgar J, Singer TP. Inhibition of mitochondrial NADH dehydrogenase by pyridine derivatives and its possible relation to experimental and idiopathic parkinsonism. Biochem Biophys Res Commun. 1986;135:269–75.

    CAS  PubMed  Google Scholar 

  31. Langston JW, Ballard PA Jr. Parkinson’s disease in a chemist working with 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine. N Engl J Med. 1983;309:310.

    CAS  PubMed  Google Scholar 

  32. Kfoury N, Kapatos G. Identification of neuronal target genes for CCAAT/enhancer binding proteins. Mol Cell Neurosci. 2009;40:313–27.

    CAS  PubMed  Google Scholar 

  33. Burke WJ, Kumar VB, Pandey N, Panneton WM, Gan Q, Franko MW, et al. Aggregation of alpha-synuclein by DOPAL, the monoamine oxidase metabolite of dopamine. Acta Neuropathol. 2008;115:193–203.

    CAS  PubMed  Google Scholar 

  34. Hald A, Lotharius J. Oxidative stress and inflammation in Parkinson’s disease: is there a causal link? Exp Neurol. 2005;193:279–90.

    CAS  PubMed  Google Scholar 

  35. Niranjan R. The role of inflammatory and oxidative stress mechanisms in the pathogenesis of Parkinson’s disease: focus on astrocytes. Mol Neurobiol. 2014;49:28–38.

    CAS  PubMed  Google Scholar 

  36. Caivano M, Gorgoni B, Cohen P, Poli V. The induction of cyclooxygenase-2 mRNA in macrophages is biphasic and requires both CCAAT enhancer-binding protein beta (C/EBP beta) and C/EBP delta transcription factors. J Biol Chem. 2001;276:48693–701.

    CAS  PubMed  Google Scholar 

  37. Reddy ST, Wadleigh DJ, Herschman HR. Transcriptional regulation of the cyclooxygenase-2 gene in activated mast cells. J Biol Chem. 2000;275:3107–13.

    CAS  PubMed  Google Scholar 

  38. Bradley MN, Zhou L, Smale ST. C/EBPbeta regulation in lipopolysaccharide-stimulated macrophages. Mol Cell Biol. 2003;23:4841–58.

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Cardinaux JR, Allaman I, Magistretti PJ. Pro-inflammatory cytokines induce the transcription factors C/EBPbeta and C/EBPdelta in astrocytes. Glia. 2000;29:91–97.

    CAS  PubMed  Google Scholar 

  40. Cardinaux JR, Magistretti PJ. Vasoactive intestinal peptide, pituitary adenylate cyclase-activating peptide, and noradrenaline induce the transcription factors CCAAT/enhancer binding protein (C/EBP)-beta and C/EBP delta in mouse cortical astrocytes: involvement in cAMP-regulated glycogen metabolism. J Neurosci. 1996;16:919–29.

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Ejarque-Ortiz A, Medina MG, Tusell JM, Perez-Gonzalez AP, Serratosa J, Saura J. Upregulation of CCAAT/enhancer binding protein beta in activated astrocytes and microglia. Glia. 2007;55:178–88.

    PubMed  Google Scholar 

  42. Perez-Capote K, Saura J, Serratosa J, Sola C. Expression of C/EBPalpha and C/EBPbeta in glial cells in vitro after inducing glial activation by different stimuli. Neurosci Lett. 2006;410:25–30.

    CAS  PubMed  Google Scholar 

  43. Kapadia R, Tureyen K, Bowen KK, Kalluri H, Johnson PF, Vemuganti R. Decreased brain damage and curtailed inflammation in transcription factor CCAAT/enhancer binding protein beta knockout mice following transient focal cerebral ischemia. J Neurochem. 2006;98:1718–31.

    CAS  PubMed  Google Scholar 

  44. Cortes-Canteli M, Luna-Medina R, Sanz-Sancristobal M, Alvarez-Barrientos A, Santos A, Perez-Castillo A. CCAAT/enhancer binding protein beta deficiency provides cerebral protection following excitotoxic injury. J Cell Sci. 2008;121:1224–34.

    CAS  PubMed  Google Scholar 

  45. Kohutnicka M, Lewandowska E, Kurkowska-Jastrzebska I, Czlonkowski A, Czlonkowska A. Microglial and astrocytic involvement in a murine model of Parkinson’s disease induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Immunopharmacology. 1998;39:167–80.

    CAS  PubMed  Google Scholar 

  46. Kurkowska-Jastrzebska I, Wronska A, Kohutnicka M, Czlonkowski A, Czlonkowska A. The inflammatory reaction following 1-methyl-4-phenyl-1,2,3, 6-tetrahydropyridine intoxication in mouse. Exp Neurol. 1999;156:50–61.

    CAS  PubMed  Google Scholar 

  47. Wu H, Kanatous SB, Thurmond FA, Gallardo T, Isotani E, Bassel-Duby R, et al. Regulation of mitochondrial biogenesis in skeletal muscle by CaMK. Science. 2002;296:349–52.

    CAS  PubMed  Google Scholar 

  48. Mogi M, Togari A, Ogawa M, Ikeguchi K, Shizuma N, Fan D, et al. Effects of repeated systemic administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) to mice on interleukin-1beta and nerve growth factor in the striatum. Neurosci Lett. 1998;250:25–28.

    CAS  PubMed  Google Scholar 

  49. Gao HM, Hong JS, Zhang W, Liu B. Distinct role for microglia in rotenone-induced degeneration of dopaminergic neurons. J Neurosci. 2002;22:782–90.

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Shih JC. Cloning, after cloning, knock-out mice, and physiological functions of MAO A and B. Neurotoxicology. 2004;25:21–30.

    CAS  PubMed  Google Scholar 

  51. Zhang Z, Song M, Liu X, Kang SS, Kwon IS, Duong DM, et al. Cleavage of tau by asparagine endopeptidase mediates the neurofibrillary pathology in Alzheimer’s disease. Nat Med. 2014;20:1254–62.

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Zhang Z, Song M, Liu X, Su Kang S, Duong DM, Seyfried NT, et al. Delta-secretase cleaves amyloid precursor protein and regulates the pathogenesis in Alzheimer’s disease. Nat Commun. 2015;6:8762.

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Negre-Salvayre A, Coatrieux C, Ingueneau C, Salvayre R. Advanced lipid peroxidation end products in oxidative damage to proteins. Potential role in diseases and therapeutic prospects for the inhibitors. Br J Pharmacol. 2008;153:6–20.

    CAS  PubMed  Google Scholar 

  54. Huin V, Buee L, Behal H, Labreuche J, Sablonniere B, Dhaenens CM. Alternative promoter usage generates novel shorter MAPT mRNA transcripts in Alzheimer’s disease and progressive supranuclear palsy brains. Sci Rep. 2017;7:12589.

    PubMed  PubMed Central  Google Scholar 

  55. Zhang Z, Liu X, Schroeder JP, Chan CB, Song M, Yu SP, et al. 7,8-dihydroxyflavone prevents synaptic loss and memory deficits in a mouse model of Alzheimer’s disease. Neuropsychopharmacology. 2014;39:638–50.

    PubMed  Google Scholar 

  56. Rojo AI, Cavada C, de Sagarra MR, Cuadrado A. Chronic inhalation of rotenone or paraquat does not induce Parkinson’s disease symptoms in mice or rats. Exp Neurol. 2007;208:120–6.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work is supported by a grant from the National Institute of Health (RF1, AG051538; RO1, NS105982) to KY, the State Key Program of National Natural Science Foundation of China (No. 81330030) to LC, the National Key Research and Development Program of China (No. 2016YFA0100800) to LC, the Fundamental Research Funds for the Central Universities of China (No. 22120170273) to LC, the National Natural Science Foundation (NSFC) of China (No. 81301042) to ZW, and the Shanghai Pujiang Talent Program, China (No. 19PJ1409200) to ZW. The authors are thankful to Dr Arthur W. English at Department of Cell Biology, Emory University for his critical reading of the manuscript. This study was supported in part by the Rodent Behavioral Core, which is subsidized by the Emory University School of Medicine and is one of the Emory Integrated Core Facilities. Additional support was provided by the Viral Vector Core of the Emory Neuroscience NINDS Core Facilities (P30NS055077). Further support was provided by the Georgia Clinical & Translational Science Alliance of the National Institutes of Health under Award Number UL1TR002378. HPLC study was supported in part by the Emory HPLC Bioanalytical Core, which was supported by the Department of Pharmacology, Emory University School of Medicine and the Georgia Clinical & Translational Science Alliance of the National Institutes of Health under Award Number UL1TR002378. The content is solely the responsibility of the authors and does not necessarily reflect the official views of the National Institutes of Health.

Author information

Authors and Affiliations

Authors

Contributions

KY conceived the project, designed the experiments, analyzed the data, and wrote the manuscript. ZW designed and performed most of the experiments and analyzed the data. YX prepared human PD-iPSCs and the ChIP assay in rat primary neuron. Z-HW preformed the tests in adult and aged Cebpb+/+/Cebpb−/− mice. SSK performed immunochemistry staining. KL and XL prepared primary neurons and assisted with in vivo and in vitro experiments. LJ, XW, and LC assisted with data analysis and interpretation and critically read the manuscript.

Corresponding authors

Correspondence to Liming Cheng or Keqiang Ye.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Additional information

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

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wu, Z., Xia, Y., Wang, Z. et al. C/EBPβ/δ-secretase signaling mediates Parkinson’s disease pathogenesis via regulating transcription and proteolytic cleavage of α-synuclein and MAOB. Mol Psychiatry 26, 568–585 (2021). https://doi.org/10.1038/s41380-020-0687-7

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41380-020-0687-7

This article is cited by

Search

Quick links