Skip to main content

Advertisement

Log in

Transcriptional repression of the ectodomain sheddase ADAM10 by TBX2 and potential implication for Alzheimer’s disease

  • Original Article
  • Published:
Cellular and Molecular Life Sciences Aims and scope Submit manuscript

Abstract

Background

The ADAM10-mediated cleavage of transmembrane proteins regulates cellular processes such as proliferation or migration. Substrate cleavage by ADAM10 has also been implicated in pathological situations such as cancer or Morbus Alzheimer. Therefore, identifying endogenous molecules, which modulate the amount and consequently the activity of ADAM10, might contribute to a deeper understanding of the enzyme’s role in both, physiology and pathology.

Method

To elucidate the underlying cellular mechanism of the TBX2-mediated repression of ADAM10 gene expression, we performed overexpression, RNAi-mediated knockdown and pharmacological inhibition studies in the human neuroblastoma cell line SH-SY5Y. Expression analysis was conducted by e.g. real-time RT-PCR or western blot techniques. To identify the binding region of TBX2 within the ADAM10 promoter, we used luciferase reporter assay on deletion constructs and EMSA/WEMSA experiments. In addition, we analyzed a TBX2 loss-of-function Drosophila model regarding the expression of ADAM10 orthologs by qPCR. Furthermore, we quantified the mRNA level of TBX2 in post-mortem brain tissue of AD patients.

Results

Here, we report TBX2 as a transcriptional repressor of ADAM10 gene expression: both, the DNA-binding domain and the repression domain of TBX2 were necessary to effect transcriptional repression of ADAM10 in neuronal SH-SY5Y cells. This regulatory mechanism required HDAC1 as a co-factor of TBX2. Transcriptional repression was mediated by two functional TBX2 binding sites within the core promoter sequence (− 315 to − 286 bp). Analysis of a TBX2 loss-of-function Drosophila model revealed that kuzbanian and kuzbanian-like, orthologs of ADAM10, were derepressed compared to wild type. Vice versa, analysis of cortical brain samples of AD-patients, which showed reduced ADAM10 mRNA levels, revealed a 2.5-fold elevation of TBX2, while TBX3 and TBX21 levels were not affected.

Conclusion

Our results characterize TBX2 as a repressor of ADAM10 gene expression and suggest that this regulatory interaction is conserved across tissues and species.

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
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8

Similar content being viewed by others

Availability of data and materials

The datasets supporting the conclusions of this article are included within the article and its additional files.

Abbreviations

AD:

Alzheimer’s disease

ADAM10:

A disintegrin and metalloproteinase 10

A-beta:

Amyloid-beta

APP:

Amyloid precursor protein

BACE-1:

Beta site APP cleaving enzyme-1

CERAD:

Consortium to establish a registry for Alzheimer’s disease

EST:

Expressed sequence tag

EMSA:

Electrophoretic mobility shift assay

FL:

Full-length

HDAC1:

Histone deacetylase 1

kuz :

Kuzbanian

kul :

Kuzbanian-like

NG2:

Nerve glia antigen 2

NL-1:

Neuroligin-1

Omb :

Optomotor-blind

PPAR-alpha:

Peroxisome proliferator activated receptor-alpha

RB1:

Retinoblastoma 1

Rp49 :

Ribosomal protein 49

SA beta-GAL:

Senescence associated beta-galactosidase

TF:

Transcription factor

TPM:

Transcripts per million

XBP-1:

X-box binding protein-1

References

  1. Saftig P, Reiss K (2011) The “A Disintegrin And Metalloproteases” ADAM10 and ADAM17: novel drug targets with therapeutic potential? Eur J Cell Biol 90:527–535

    Article  PubMed  CAS  Google Scholar 

  2. Edwards DR, Handsley MM, Pennington CJ (2008) The ADAM metalloproteinases. Mol Aspects Med 29:258–289

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  3. Anders A, Gilbert S, Garten W, Postina R, Fahrenholz F (2001) Regulation of the alpha-secretase ADAM10 by its prodomain and proprotein convertases. FASEB J 15:1837–1839

    Article  PubMed  CAS  Google Scholar 

  4. Pruessmeyer J, Ludwig A (2009) The good, the bad and the ugly substrates for ADAM10 and ADAM17 in brain pathology, inflammation and cancer. Semin Cell Dev Biol 20:164–174

    Article  PubMed  CAS  Google Scholar 

  5. Endres K, Deller T (2017) Regulation of alpha-secretase ADAM10 in vitro and in vivo: genetic, epigenetic, and protein-based mechanisms. Front Mol Neurosci 10:56

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Sakry D, Neitz A, Singh J, Frischknecht R, Marongiu D, Biname F, Perera SS, Endres K, Lutz B, Radyushkin K et al (2014) Oligodendrocyte precursor cells modulate the neuronal network by activity-dependent ectodomain cleavage of glial NG2. PLoS Biol 12:e1001993

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Suzuki K, Hayashi Y, Nakahara S, Kumazaki H, Prox J, Horiuchi K, Zeng M, Tanimura S, Nishiyama Y, Osawa S et al (2012) Activity-dependent proteolytic cleavage of neuroligin-1. Neuron 76:410–422

    Article  PubMed  CAS  Google Scholar 

  8. Jarriault S, Greenwald I (2005) Evidence for functional redundancy between C. elegans ADAM proteins SUP-17/Kuzbanian and ADM-4/TACE. Dev Biol 287:1–10

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Riddle DL, Blumenthal T, Meyer BJ, Priess JR (1997) Introduction to C. elegans. In: Riddle DL, Blumenthal T, Meyer BJ, Priess JR (eds) C. elegans I. Cold Spring Harbor, New York

    Google Scholar 

  10. Rooke J, Pan D, Xu T, Rubin GM (1996) KUZ, a conserved metalloprotease-disintegrin protein with two roles in Drosophila neurogenesis. Science 273:1227–1231

    Article  PubMed  CAS  Google Scholar 

  11. Wei S, Whittaker CA, Xu G, Bridges LC, Shah A, White JM, Desimone DW (2010) Conservation and divergence of ADAM family proteins in the Xenopus genome. BMC Evol Biol 10:211

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Hartmann D, de Strooper B, Serneels L, Craessaerts K, Herreman A, Annaert W, Umans L, Lübke T, Lena Illert A, von Figura K, Saftig P (2002) The disintegrin/metalloprotease ADAM 10 is essential for Notch signalling but not for alpha-secretase activity in fibroblasts. Hum Mol Genet 11:2615–2624

    Article  PubMed  CAS  Google Scholar 

  13. Jorissen E, Prox J, Bernreuther C, Weber S, Schwanbeck R, Serneels L, Snellinx A, Craessaerts K, Thathiah A, Tesseur I et al (2010) The disintegrin/metalloproteinase ADAM10 is essential for the establishment of the brain cortex. J Neurosci 30:4833–4844

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Esselens CW, Malapeira J, Colome N, Moss M, Canals F, Arribas J (2008) Metastasis-associated C4.4A, a GPI-anchored protein cleaved by ADAM10 and ADAM17. Biol Chem 389:1075–1084

    Article  PubMed  CAS  Google Scholar 

  15. Gavert N, Conacci-Sorrell M, Gast D, Schneider A, Altevogt P, Brabletz T, Ben-Ze’ev A (2005) L1, a novel target of beta-catenin signaling, transforms cells and is expressed at the invasive front of colon cancers. J Cell Biol 168:633–642

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Kohutek ZA, diPierro CG, Redpath GT, Hussaini IM (2009) ADAM-10-mediated N-cadherin cleavage is protein kinase C-alpha dependent and promotes glioblastoma cell migration. J Neurosci 29:4605–4615

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. McCulloch DR, Akl P, Samaratunga H, Herington AC, Odorico DM (2004) Expression of the disintegrin metalloprotease, ADAM-10, in prostate cancer and its regulation by dihydrotestosterone, insulin-like growth factor I, and epidermal growth factor in the prostate cancer cell model LNCaP. Clin Cancer Res 10:314–323

    Article  PubMed  CAS  Google Scholar 

  18. Moss ML, Stoeck A, Yan W, Dempsey PJ (2008) ADAM10 as a target for anti-cancer therapy. Curr Pharm Biotechnol 9:2–8

    Article  PubMed  CAS  Google Scholar 

  19. Wu E, Croucher PI, McKie N (1997) Expression of members of the novel membrane linked metalloproteinase family ADAM in cells derived from a range of haematological malignancies. Biochem Biophys Res Commun 235:437–442

    Article  PubMed  CAS  Google Scholar 

  20. Endres K, Fahrenholz F (2010) Upregulation of the alpha-secretase ADAM10–risk or reason for hope? FEBS J 277:1585–1596

    Article  PubMed  CAS  Google Scholar 

  21. Endres K, Fahrenholz F (2012) Regulation of alpha-secretase ADAM10 expression and activity. Exp Brain Res 217:343–352

    Article  PubMed  CAS  Google Scholar 

  22. Endres K, Fahrenholz F, Lotz J, Hiemke C, Teipel S, Lieb K, Tuscher O, Fellgiebel A (2014) Increased CSF APPs-alpha levels in patients with Alzheimer disease treated with acitretin. Neurology 83:1930–1935

    Article  PubMed  CAS  Google Scholar 

  23. Tippmann F, Hundt J, Schneider A, Endres K, Fahrenholz F (2009) Up-regulation of the alpha-secretase ADAM10 by retinoic acid receptors and acitretin. FASEB J 23:1643–1654

    Article  PubMed  CAS  Google Scholar 

  24. Corrigan F, Vink R, Blumbergs PC, Masters CL, Cappai R, van den Heuvel C (2012) sAPPalpha rescues deficits in amyloid precursor protein knockout mice following focal traumatic brain injury. J Neurochem 122:208–220

    Article  PubMed  CAS  Google Scholar 

  25. Kuhn PH, Wang H, Dislich B, Colombo A, Zeitschel U, Ellwart JW, Kremmer E, Rossner S, Lichtenthaler SF (2010) ADAM10 is the physiologically relevant, constitutive alpha-secretase of the amyloid precursor protein in primary neurons. EMBO J 29:3020–3032

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Milosch N, Tanriover G, Kundu A, Rami A, Francois JC, Baumkotter F, Weyer SW, Samanta A, Jaschke A, Brod F et al (2014) Holo-APP and G-protein-mediated signaling are required for sAPPalpha-induced activation of the Akt survival pathway. Cell Death Dis 5:e1391

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Postina R, Schroeder A, Dewachter I, Bohl J, Schmitt U, Kojro E, Prinzen C, Endres K, Hiemke C, Blessing M et al (2004) A disintegrin-metalloproteinase prevents amyloid plaque formation and hippocampal defects in an Alzheimer disease mouse model. J Clin Invest 113:1456–1464

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Thornton E, Vink R, Blumbergs PC, Van Den Heuvel C (2006) Soluble amyloid precursor protein alpha reduces neuronal injury and improves functional outcome following diffuse traumatic brain injury in rats. Brain Res 1094:38–46

    Article  PubMed  CAS  Google Scholar 

  29. Prinzen C, Muller U, Endres K, Fahrenholz F, Postina R (2005) Genomic structure and functional characterization of the human ADAM10 promoter. FASEB J 19:1522–1524

    Article  PubMed  Google Scholar 

  30. Endres K, Postina R, Schroeder A, Mueller U, Fahrenholz F (2005) Shedding of the amyloid precursor protein-like protein APLP2 by disintegrin-metalloproteinases. FEBS J 272:5808–5820

    Article  PubMed  CAS  Google Scholar 

  31. Corbett GT, Gonzalez FJ, Pahan K (2015) Activation of peroxisome proliferator-activated receptor alpha stimulates ADAM10-mediated proteolysis of APP. Proc Natl Acad Sci USA 112:8445–8450

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  32. Reinhardt S, Schuck F, Grosgen S, Riemenschneider M, Hartmann T, Postina R, Grimm M, Endres K (2014) Unfolded protein response signaling by transcription factor XBP-1 regulates ADAM10 and is affected in Alzheimer’s disease. FASEB J 28:978–997

    Article  PubMed  CAS  Google Scholar 

  33. Harrelson Z, Kelly RG, Goldin SN, Gibson-Brown JJ, Bollag RJ, Silver LM, Papaioannou VE (2004) Tbx2 is essential for patterning the atrioventricular canal and for morphogenesis of the outflow tract during heart development. Development 131:5041–5052

    Article  PubMed  CAS  Google Scholar 

  34. Jacobs JJ, Keblusek P, Robanus-Maandag E, Kristel P, Lingbeek M, Nederlof PM, van Welsem T, van de Vijver MJ, Koh EY, Daley GQ, van Lohuizen M (2000) Senescence bypass screen identifies TBX2, which represses Cdkn2a (p19(ARF)) and is amplified in a subset of human breast cancers. Nat Genet 26:291–299

    Article  PubMed  CAS  Google Scholar 

  35. Kokubo H, Tomita-Miyagawa S, Hamada Y, Saga Y (2007) Hesr1 and Hesr2 regulate atrioventricular boundary formation in the developing heart through the repression of Tbx2. Development 134:747–755

    Article  PubMed  CAS  Google Scholar 

  36. Prince S, Carreira S, Vance KW, Abrahams A, Goding CR (2004) Tbx2 directly represses the expression of the p21(WAF1) cyclin-dependent kinase inhibitor. Cancer Res 64:1669–1674

    Article  PubMed  CAS  Google Scholar 

  37. Vassar R, Bennett BD, Babu-Khan S, Kahn S, Mendiaz EA, Denis P, Teplow DB, Ross S, Amarante P, Loeloff R et al (1999) Beta-secretase cleavage of Alzheimer’s amyloid precursor protein by the transmembrane aspartic protease BACE. Science 286:735–741

    Article  PubMed  CAS  Google Scholar 

  38. Naiche LA, Harrelson Z, Kelly RG, Papaioannou VE (2005) T-box genes in vertebrate development. Annu Rev Genet 39:219–239

    Article  PubMed  CAS  Google Scholar 

  39. Papaioannou VE, Silver LM (1998) The T-box gene family. BioEssays 20:9–19

    Article  PubMed  CAS  Google Scholar 

  40. Chapman DL, Garvey N, Hancock S, Alexiou M, Agulnik SI, Gibson-Brown JJ, Cebra-Thomas J, Bollag RJ, Silver LM, Papaioannou VE (1996) Expression of the T-box family genes, Tbx1-Tbx5, during early mouse development. Dev Dyn 206:379–390

    Article  PubMed  CAS  Google Scholar 

  41. Gibson-Brown JJ, Agulnik SI, Silver LM, Niswander L, Papaioannou VE (1998) Involvement of T-box genes Tbx2-Tbx5 in vertebrate limb specification and development. Development 125:2499–2509

    PubMed  CAS  Google Scholar 

  42. Takabatake Y, Takabatake T, Takeshima K (2000) Conserved and divergent expression of T-box genes Tbx2-Tbx5 in Xenopus. Mech Dev 91:433–437

    Article  PubMed  CAS  Google Scholar 

  43. Douglas NC, Papaioannou VE (2013) The T-box transcription factors TBX2 and TBX3 in mammary gland development and breast cancer. J Mammary Gland Biol Neoplasia 18:143–147

    Article  PubMed  PubMed Central  Google Scholar 

  44. Peres J, Davis E, Mowla S, Bennett DC, Li JA, Wansleben S, Prince S (2010) The highly homologous T-box transcription factors, TBX2 and TBX3, have distinct roles in the oncogenic process. Genes Cancer 1:272–282

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Rowley M, Grothey E, Couch FJ (2004) The role of Tbx2 and Tbx3 in mammary development and tumorigenesis. J Mammary Gland Biol Neoplasia 9:109–118

    Article  PubMed  Google Scholar 

  46. Lingbeek ME, Jacobs JJ, van Lohuizen M (2002) The T-box repressors TBX2 and TBX3 specifically regulate the tumor suppressor gene p14ARF via a variant T-site in the initiator. J Biol Chem 277:26120–26127

    Article  PubMed  CAS  Google Scholar 

  47. He M, Wen L, Campbell CE, Wu JY, Rao Y (1999) Transcription repression by Xenopus ET and its human ortholog TBX3, a gene involved in ulnar-mammary syndrome. Proc Natl Acad Sci USA 96:10212–10217

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  48. Marchler-Bauer A, Derbyshire MK, Gonzales NR, Lu S, Chitsaz F, Geer LY, Geer RC, He J, Gwadz M, Hurwitz DI et al (2015) CDD: NCBI’s conserved domain database. Nucleic Acids Res 43:D222–D226

    Article  PubMed  CAS  Google Scholar 

  49. Demay F, Bilican B, Rodriguez M, Carreira S, Pontecorvi M, Ling Y, Goding CR (2007) T-box factors: targeting to chromatin and interaction with the histone H3 N-terminal tail. Pigment Cell Res 20:279–287

    Article  PubMed  CAS  Google Scholar 

  50. Fischer K, Pflugfelder GO (2015) Putative breast cancer driver mutations in TBX3 cause impaired transcriptional repression. Front Oncol 5:244

    Article  PubMed  PubMed Central  Google Scholar 

  51. Rodriguez M, Aladowicz E, Lanfrancone L, Goding CR (2008) Tbx3 represses E-cadherin expression and enhances melanoma invasiveness. Cancer Res 68:7872–7881

    Article  PubMed  CAS  Google Scholar 

  52. Macindoe I, Glockner L, Vukasin P, Stennard FA, Costa MW, Harvey RP, Mackay JP, Sunde M (2009) Conformational stability and DNA binding specificity of the cardiac T-box transcription factor Tbx20. J Mol Biol 389:606–618

    Article  PubMed  CAS  Google Scholar 

  53. Sinha S, Abraham S, Gronostajski RM, Campbell CE (2000) Differential DNA binding and transcription modulation by three T-box proteins, T, TBX1 and TBX2. Gene 258:15–29

    Article  PubMed  CAS  Google Scholar 

  54. Vance KW, Shaw HM, Rodriguez M, Ott S, Goding CR (2010) The retinoblastoma protein modulates Tbx2 functional specificity. Mol Biol Cell 21:2770–2779

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Vance KW, Carreira S, Brosch G, Goding CR (2005) Tbx2 is overexpressed and plays an important role in maintaining proliferation and suppression of senescence in melanomas. Cancer Res 65:2260–2268

    Article  PubMed  CAS  Google Scholar 

  56. Saito A, Yamashita T, Mariko Y, Nosaka Y, Tsuchiya K, Ando T, Suzuki T, Tsuruo T, Nakanishi O (1999) A synthetic inhibitor of histone deacetylase, MS-27-275, with marked in vivo antitumor activity against human tumors. Proc Natl Acad Sci USA 96:4592–4597

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  57. Hu E, Dul E, Sung CM, Chen Z, Kirkpatrick R, Zhang GF, Johanson K, Liu R, Lago A, Hofmann G et al (2003) Identification of novel isoform-selective inhibitors within class I histone deacetylases. J Pharmacol Exp Ther 307:720–728

    Article  PubMed  CAS  Google Scholar 

  58. Narath R, Ambros IM, Kowalska A, Bozsaky E, Boukamp P, Ambros PF (2007) Induction of senescence in MYCN amplified neuroblastoma cell lines by hydroxyurea. Genes Chromosomes Cancer 46:130–142

    Article  PubMed  CAS  Google Scholar 

  59. Yu X, Li X, Jiang G, Wang X, Chang HC, Hsu WH, Li Q (2013) Isradipine prevents rotenone-induced intracellular calcium rise that accelerates senescence in human neuroblastoma SH-SY5Y cells. Neuroscience 246:243–253

    Article  PubMed  CAS  Google Scholar 

  60. Dimri GP, Lee X, Basile G, Acosta M, Scott G, Roskelley C, Medrano EE, Linskens M, Rubelj I, Pereira-Smith O et al (1995) A biomarker that identifies senescent human cells in culture and in aging skin in vivo. Proc Natl Acad Sci USA 92:9363–9367

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  61. Yang NC, Hu ML (2005) The limitations and validities of senescence associated-beta-galactosidase activity as an aging marker for human foreskin fibroblast Hs68 cells. Exp Gerontol 40:813–819

    Article  PubMed  CAS  Google Scholar 

  62. Li J, Ballim D, Rodriguez M, Cui R, Goding CR, Teng H, Prince S (2014) The anti-proliferative function of the TGF-beta1 signaling pathway involves the repression of the oncogenic TBX2 by its homologue TBX3. J Biol Chem 289:35633–35643

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Grimm S, Pflugfelder GO (1996) Control of the gene optomotor-blind in Drosophila wing development by decapentaplegic and wingless. Science 271:1601–1604

    Article  PubMed  CAS  Google Scholar 

  64. Pflugfelder GO, Eichinger F, Shen J (2017) T-box genes in drosophila limb development. Curr Top Dev Biol 122:313–354

    Article  PubMed  CAS  Google Scholar 

  65. Poeck B, Hofbauer A, Pflugfelder GO (1993) Expression of the Drosophila optomotor-blind gene transcript in neuronal and glial cells of the developing nervous system. Development 117:1017–1029

    PubMed  CAS  Google Scholar 

  66. Lieber T, Kidd S, Young MW (2002) kuzbanian-mediated cleavage of Drosophila Notch. Genes Dev 16:209–221

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Kidd S, Lieber T (2002) Furin cleavage is not a requirement for Drosophila Notch function. Mech Dev 115:41–51

    Article  PubMed  CAS  Google Scholar 

  68. Kovalevich J, Langford D (2013) Considerations for the use of SH-SY5Y neuroblastoma cells in neurobiology. Methods Mol Biol 1078:9–21

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  69. Boogerd KJ, Wong LY, Christoffels VM, Klarenbeek M, Ruijter JM, Moorman AF, Barnett P (2008) Msx1 and Msx2 are functional interacting partners of T-box factors in the regulation of Connexin43. Cardiovasc Res 78:485–493

    Article  PubMed  CAS  Google Scholar 

  70. Christoffels VM, Hoogaars WM, Tessari A, Clout DE, Moorman AF, Campione M (2004) T-box transcription factor Tbx2 represses differentiation and formation of the cardiac chambers. Dev Dyn 229:763–770

    Article  PubMed  CAS  Google Scholar 

  71. Weber S, Niessen MT, Prox J, Lullmann-Rauch R, Schmitz A, Schwanbeck R, Blobel CP, Jorissen E, de Strooper B, Niessen CM, Saftig P (2011) The disintegrin/metalloproteinase Adam10 is essential for epidermal integrity and Notch-mediated signaling. Development 138:495–505

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. Zhang C, Tian L, Chi C, Wu X, Yang X, Han M, Xu T, Zhuang Y, Deng K (2010) Adam10 is essential for early embryonic cardiovascular development. Dev Dyn 239:2594–2602

    Article  PubMed  CAS  Google Scholar 

  73. Kispert A, Hermann BG (1993) The Brachyury gene encodes a novel DNA binding protein. EMBO J 12:4898–4899

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  74. Sen A, Grimm S, Hofmeyer K, Pflugfelder GO (2014) Optomotor-blind in the development of the Drosophila HS and VS lobula plate tangential cells. J Neurogenet 28:250–263

    Article  PubMed  CAS  Google Scholar 

  75. Chen JR, Chatterjee B, Meyer R, Yu JC, Borke JL, Isales CM, Kirby ML, Lo CW, Bollag RJ (2004) Tbx2 represses expression of Connexin43 in osteoblastic-like cells. Calcif Tissue Int 74:561–573

    Article  PubMed  CAS  Google Scholar 

  76. Abrahams A, Parker MI, Prince S (2010) The T-box transcription factor Tbx2: its role in development and possible implication in cancer. IUBMB Life 62:92–102

    PubMed  CAS  Google Scholar 

  77. Paxton C, Zhao H, Chin Y, Langner K, Reecy J (2002) Murine Tbx2 contains domains that activate and repress gene transcription. Gene 283:117–124

    Article  PubMed  CAS  Google Scholar 

  78. Zhu B, Zhang M, Byrum SD, Tackett AJ, Davie JK (2014) TBX2 blocks myogenesis and promotes proliferation in rhabdomyosarcoma cells. Int J Cancer 135:785–797

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  79. Zhu B, Zhang M, Williams EM, Keller C, Mansoor A, Davie JK (2016) TBX2 represses PTEN in rhabdomyosarcoma and skeletal muscle. Oncogene 35:4212–4224

    Article  PubMed  CAS  Google Scholar 

  80. Kaltenbrun E, Greco TM, Slagle CE, Kennedy LM, Li T, Cristea IM, Conlon FL (2013) A Gro/TLE-NuRD corepressor complex facilitates Tbx20-dependent transcriptional repression. J Proteome Res 12:5395–5409

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  81. Lewandowski SL, Janardhan HP, Smee KM, Bachman M, Sun Z, Lazar MA, Trivedi CM (2014) Histone deacetylase 3 modulates Tbx5 activity to regulate early cardiogenesis. Hum Mol Genet 23:3801–3809

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  82. Cunliffe VT (2004) Histone deacetylase 1 is required to repress Notch target gene expression during zebrafish neurogenesis and to maintain the production of motoneurones in response to hedgehog signalling. Development 131:2983–2995

    Article  PubMed  CAS  Google Scholar 

  83. Lightman EG, Harrison MR, Cunliffe VT (2011) Opposing actions of histone deacetylase 1 and Notch signalling restrict expression of erm and fgf20a to hindbrain rhombomere centres during zebrafish neurogenesis. Int J Dev Biol 55:597–602

    Article  PubMed  CAS  Google Scholar 

  84. Rutenberg JB, Fischer A, Jia H, Gessler M, Zhong TP, Mercola M (2006) Developmental patterning of the cardiac atrioventricular canal by Notch and Hairy-related transcription factors. Development 133:4381–4390

    Article  PubMed  CAS  Google Scholar 

  85. Yoon K, Gaiano N (2005) Notch signaling in the mammalian central nervous system: insights from mouse mutants. Nat Neurosci 8:709–715

    Article  PubMed  CAS  Google Scholar 

  86. Pan D, Rubin GM (1997) Kuzbanian controls proteolytic processing of Notch and mediates lateral inhibition during Drosophila and vertebrate neurogenesis. Cell 90:271–280

    Article  PubMed  CAS  Google Scholar 

  87. Sapir A, Assa-Kunik E, Tsruya R, Schejter E, Shilo BZ (2005) Unidirectional Notch signaling depends on continuous cleavage of Delta. Development 132:123–132

    Article  PubMed  CAS  Google Scholar 

  88. Sotillos S, Roch F, Campuzano S (1997) The metalloprotease-disintegrin Kuzbanian participates in Notch activation during growth and patterning of Drosophila imaginal discs. Development 124:4769–4779

    PubMed  CAS  Google Scholar 

  89. Currais A, Hortobagyi T, Soriano S (2009) The neuronal cell cycle as a mechanism of pathogenesis in Alzheimer’s disease. Aging (Albany NY) 1:363–371

    Article  CAS  Google Scholar 

  90. McShea A, Lee HG, Petersen RB, Casadesus G, Vincent I, Linford NJ, Funk JO, Shapiro RA, Smith MA (2007) Neuronal cell cycle re-entry mediates Alzheimer disease-type changes. Biochim Biophys Acta 1772:467–472

    Article  PubMed  CAS  Google Scholar 

  91. Counts SE, Mufson EJ (2017) Regulator of cell cycle (RGCC) expression during the progression of Alzheimer’s disease. Cell Transplant 26:693–702

    Article  PubMed  PubMed Central  Google Scholar 

  92. Schneider MA, Scheffer KD, Bund T, Boukhallouk F, Lambert C, Cotarelo C, Pflugfelder GO, Florin L, Spoden GA (2013) The transcription factors TBX2 and TBX3 interact with human papillomavirus 16 (HPV16) L2 and repress the long control region of HPVs. J Virol 87:4461–4474

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  93. Habets PE, Moorman AF, Clout DE, van Roon MA, Lingbeek M, van Lohuizen M, Campione M, Christoffels VM (2002) Cooperative action of Tbx2 and Nkx2.5 inhibits ANF expression in the atrioventricular canal: implications for cardiac chamber formation. Genes Dev 16:1234–1246

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  94. Thomas P, Smart TG (2005) HEK293 cell line: a vehicle for the expression of recombinant proteins. J Pharmacol Toxicol Methods 51:187–200

    Article  PubMed  CAS  Google Scholar 

  95. Moeenrezakhanlou A, Nandan D, Reiner NE (2008) Identification of a calcitriol-regulated Sp-1 site in the promoter of human CD14 using a combined western blotting electrophoresis mobility shift assay (WEMSA). Biol Proced Online 10:29–35

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  96. Poeck B, Balles J, Pflugfelder GO (1993) Transcript identification in the optomotor-blind locus of Drosophila melanogaster by intragenic recombination mapping and PCR-aided sequence analysis of lethal point mutations. Mol Gen Genet 238:325–332

    Article  PubMed  CAS  Google Scholar 

  97. Pflugfelder GO, Roth H, Poeck B, Kerscher S, Schwarz H, Jonschker B, Heisenberg M (1992) The lethal(1)optomotor-blind gene of Drosophila melanogaster is a major organizer of optic lobe development: isolation and characterization of the gene. Proc Natl Acad Sci USA 89:1199–1203

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  98. Sen A, Gadomski C, Balles J, Abassi Y, Dorner C, Pflugfelder GO (2010) Null mutations in Drosophila Optomotor-blind affect T-domain residues conserved in all Tbx proteins. Mol Genet Genom 283:147–156

    Article  CAS  Google Scholar 

  99. Grimm MO, Grosgen S, Rothhaar TL, Burg VK, Hundsdorfer B, Haupenthal VJ, Friess P, Muller U, Fassbender K, Riemenschneider M et al (2011) Intracellular APP domain regulates serine-palmitoyl-CoA transferase expression and is affected in alzheimer’s disease. Int J Alzheimers Dis 2011:695413

    PubMed  PubMed Central  Google Scholar 

  100. Livak KJ, Schmittgen TD (2001) Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 25:402–408

    Article  PubMed  CAS  Google Scholar 

  101. Steiner H, Kostka M, Romig H, Basset G, Pesold B, Hardy J, Capell A, Meyn L, Grim ML, Baumeister R et al (2000) Glycine 384 is required for presenilin-1 function and is conserved in bacterial polytopic aspartyl proteases. Nat Cell Biol 2:848–851

    Article  PubMed  CAS  Google Scholar 

  102. Fehon RG, Kooh PJ, Rebay I, Regan CL, Xu T, Muskavitch MA, Artavanis-Tsakonas S (1990) Molecular interactions between the protein products of the neurogenic loci Notch and Delta, two EGF-homologous genes in Drosophila. Cell 61:523–534

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by the Federal Ministry of Education and Research (BMBF) in the framework of the National Genome Research Network (NGFN), FKZ01GS08130, 01GS08125 and 01GS08129-5 and by the Alfons Geib Stiftung. We thank K. Hilger, A. Bruns, and S. Schneider (all University Medical Center Mainz, Germany) for technical assistance; we also are grateful to Inka Hoffmann, Fred Eichinger, and Melanie Heyde (University of Mainz, Germany) for preparation of Drosophila wing imaginal discs and to Sven Grösgen (Saarland University, Germany) for experiments regarding human samples; we want to thank Colin Goding (University of Oxford, UK) for murine TBX2 expression constructs and Christian Haass (LMU Munich, Germany) for APP C-terminal antibody 6687. Stock 25706 obtained from the Bloomington Drosophila Stock Center (NIH P40OD018537) was used in this study.

Funding

This work was supported by the German Federal Ministry of Education and Research (BMBF) in the framework of the National Genome Research Network (NGFN) and FKZ01GS08130 and by the Alfons-Geib Stiftung.

Author information

Authors and Affiliations

Authors

Contributions

KE conceived and coordinated the study, drafted the manuscript, performed experiments with dermal fibroblasts and generated the TBX2 binding site deletion mutant. SR carried out the molecular biology and biochemistry studies, performed analysis on EST profiles, conducted the statistical analysis, and helped to draft the manuscript. FS constructed secreted luciferase reporter vectors and helped to revise the manuscript. NS performed overexpression experiments of HDAC1 to assess effects on ADAM10 and luciferase reporter gene assays for analyzing the TBX2 binding site deletion mutant. MG and TH designed and coordinated studies regarding human brain tissue and revised the manuscript. GP provided expression constructs for human DDK-tagged TBX2 and Drosophila larval samples. All authors have read and approved the final version of the manuscript.

Corresponding author

Correspondence to Kristina Endres.

Ethics declarations

Ethical approval and consent to participate

All experiments were conducted in accordance to the official regulations for the care and use of laboratory animals and approved by local authorities (University of Mainz, Germany).

Conflict of interest

The authors declare that they have no competing interests.

Additional information

Publisher's Note

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

Electronic supplementary material

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Reinhardt, S., Schuck, F., Stoye, N. et al. Transcriptional repression of the ectodomain sheddase ADAM10 by TBX2 and potential implication for Alzheimer’s disease. Cell. Mol. Life Sci. 76, 1005–1025 (2019). https://doi.org/10.1007/s00018-018-2998-2

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00018-018-2998-2

Keywords

Navigation