Skip to main content
Log in

Fibulin-3 knockout mice demonstrate corneal dysfunction but maintain normal retinal integrity

  • Original Article
  • Published:
Journal of Molecular Medicine Aims and scope Submit manuscript

Abstract

Fibulin-3 (F3) is an extracellular matrix glycoprotein found in basement membranes across the body. An autosomal dominant R345W mutation in F3 causes a macular dystrophy resembling dry age-related macular degeneration (AMD), whereas genetic removal of wild-type (WT) F3 protects mice from sub-retinal pigment epithelium (RPE) deposit formation. These observations suggest that F3 is a protein which can regulate pathogenic sub-RPE deposit formation in the eye. Yet the precise role of WT F3 within the eye is still largely unknown. We found that F3 is expressed throughout the mouse eye (cornea, trabecular meshwork (TM) ring, neural retina, RPE/choroid, and optic nerve). We next performed a thorough structural and functional characterization of each of these tissues in WT and homozygous (F3−/−) knockout mice. The corneal stroma in F3−/− mice progressively thins beginning at 2 months, and the development of corneal opacity and vascularization starts at 9 months, which worsens with age. However, in all other tissues (TM, neural retina, RPE, and optic nerve), gross structural anatomy and functionality were similar across WT and F3−/− mice when evaluated using SD-OCT, histological analyses, electron microscopy, scotopic electroretinogram, optokinetic response, and axonal anterograde transport. The lack of noticeable retinal abnormalities in F3−/− mice was confirmed in a human patient with biallelic loss-of-function mutations in F3. These data suggest that (i) F3 is important for maintaining the structural integrity of the cornea, (ii) absence of F3 does not affect the structure or function of any other ocular tissue in which it is expressed, and (iii) targeted silencing of F3 in the retina and/or RPE will likely be well-tolerated, serving as a safe therapeutic strategy for reducing sub-RPE deposit formation in disease.

Key messages

• Fibulins are expressed throughout the body at varying levels.

• Fibulin-3 has a tissue-specific pattern of expression within the eye.

• Lack of fibulin-3 leads to structural deformities in the cornea.

• The retina and RPE remain structurally and functionally healthy in the absence of fibulin-3 in both mice and humans.

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

Similar content being viewed by others

References

  1. Zhang Y, Marmorstein LY (2010) Focus on molecules: fibulin-3 (EFEMP1). Exp Eye Res 90:374–375

    CAS  Google Scholar 

  2. Hulleman JD (2016) Malattia Leventinese/Doyne honeycomb retinal dystrophy: similarities to age-related macular degeneration and potential therapies. Adv Exp Med Biol 854:153–158

    CAS  Google Scholar 

  3. McLaughlin PJ, Bakall B, Choi J, Liu Z, Sasaki T, Davis EC, Marmorstein AD, Marmorstein LY (2007) Lack of fibulin-3 causes early aging and herniation, but not macular degeneration in mice. Hum Mol Genet 16:3059–3070

    CAS  Google Scholar 

  4. Papke CL, Yanagisawa H (2014) Fibulin-4 and fibulin-5 in elastogenesis and beyond: Insights from mouse and human studies. Matrix Biol 37:142–149

    CAS  Google Scholar 

  5. Kumra H, Nelea V, Hakami H, Pagliuzza A, Djokic J, Xu J, Yanagisawa H, Reinhardt DP (2019) Fibulin-4 exerts a dual role in LTBP-4L-mediated matrix assembly and function. Proc Natl Acad Sci U S A 116:20428–20437

    CAS  PubMed Central  Google Scholar 

  6. Li J, Qi C, Liu X, Li C, Chen J, Shi M (2018) Fibulin-3 knockdown inhibits cervical cancer cell growth and metastasis in vitro and in vivo. Sci Rep 8:10594

    PubMed Central  Google Scholar 

  7. Nandhu MS, Kwiatkowska A, Bhaskaran V, Hayes J, Hu B, Viapiano MS (2017) Tumor-derived fibulin-3 activates pro-invasive NF-kappaB signaling in glioblastoma cells and their microenvironment. Oncogene 36:4875–4886

    CAS  PubMed Central  Google Scholar 

  8. Tian H, Liu J, Chen J, Gatza ML, Blobe GC (2015) Fibulin-3 is a novel TGF-beta pathway inhibitor in the breast cancer microenvironment. Oncogene 34:5635–5647

    CAS  PubMed Central  Google Scholar 

  9. Kim IG, Kim SY, Choi SI, Lee JH, Kim KC, Cho EW (2014) Fibulin-3-mediated inhibition of epithelial-to-mesenchymal transition and self-renewal of ALDH+ lung cancer stem cells through IGF1R signaling. Oncogene 33:3908–3917

    CAS  Google Scholar 

  10. Klenotic PA, Munier FL, Marmorstein LY, Anand-Apte B (2004) Tissue inhibitor of metalloproteinases-3 (TIMP-3) is a binding partner of epithelial growth factor-containing fibulin-like extracellular matrix protein 1 (EFEMP1). Implications for macular degenerations. J Biol Chem 279:30469–30473

    CAS  Google Scholar 

  11. Kobayashi N, Kostka G, Garbe JH, Keene DR, Bachinger HP, Hanisch FG, Markova D, Tsuda T, Timpl R, Chu ML et al (2007) A comparative analysis of the fibulin protein family. Biochemical characterization, binding interactions, and tissue localization. J Biol Chem 282:11805–11816

    CAS  Google Scholar 

  12. Hulleman JD, Genereux JC, Nguyen A (2016) Mapping wild-type and R345W fibulin-3 intracellular interactomes. Exp Eye Res 153:165–169

    CAS  PubMed Central  Google Scholar 

  13. Blackburn J, Tarttelin EE, Gregory-Evans CY, Moosajee M, Gregory-Evans K (2003) Transcriptional regulation and expression of the dominant drusen gene FBLN3 (EFEMP1) in mammalian retina. Invest Ophthalmol Vis Sci 44:4613–4621

    Google Scholar 

  14. Stone EM, Lotery AJ, Munier FL, Heon E, Piguet B, Guymer RH, Vandenburgh K, Cousin P, Nishimura D, Swiderski RE et al (1999) A single EFEMP1 mutation associated with both Malattia Leventinese and Doyne honeycomb retinal dystrophy. Nat Genet 22:199–202

    CAS  Google Scholar 

  15. Sohn EH, Wang K, Thompson S, Riker MJ, Hoffmann JM, Stone EM, Mullins RF (2015) Comparison of drusen and modifying genes in autosomal dominant radial drusen and age-related macular degeneration. Retina 35:48–57

    CAS  PubMed Central  Google Scholar 

  16. Michaelides M, Jenkins SA, Brantley MA Jr, Andrews RM, Waseem N, Luong V, Gregory-Evans K, Bhattacharya SS, Fitzke FW, Webster AR (2006) Maculopathy due to the R345W substitution in fibulin-3: distinct clinical features, disease variability, and extent of retinal dysfunction. Invest Ophthalmol Vis Sci 47:3085–3097

    Google Scholar 

  17. Hulleman JD, Kaushal S, Balch WE, Kelly JW (2011) Compromised mutant EFEMP1 secretion associated with macular dystrophy remedied by proteostasis network alteration. Mol Biol Cell 22:4765–4775

    CAS  PubMed Central  Google Scholar 

  18. Hulleman JD, Balch WE, Kelly JW (2012) Translational attenuation differentially alters the fate of disease-associated fibulin proteins. FASEB J 26:4548–4560

    CAS  PubMed Central  Google Scholar 

  19. Marmorstein LY, Munier FL, Arsenijevic Y, Schorderet DF, McLaughlin PJ, Chung D, Traboulsi E, Marmorstein AD (2002) Aberrant accumulation of EFEMP1 underlies drusen formation in Malattia Leventinese and age-related macular degeneration. Proc Natl Acad Sci U S A 99:13067–13072

    CAS  PubMed Central  Google Scholar 

  20. Hulleman JD, Brown SJ, Rosen H, Kelly JW (2013) A high-throughput cell-based Gaussia luciferase reporter assay for identifying modulators of fibulin-3 secretion. J Biomol Screen 18:647–658

    CAS  Google Scholar 

  21. Hulleman JD, Kelly JW (2015) Genetic ablation of N-linked glycosylation reveals two key folding pathways for R345W fibulin-3, a secreted protein associated with retinal degeneration. FASEB J 29:565–575

    CAS  Google Scholar 

  22. Roybal CN, Marmorstein LY, Vander Jagt DL, Abcouwer SF (2005) Aberrant accumulation of fibulin-3 in the endoplasmic reticulum leads to activation of the unfolded protein response and VEGF expression. Invest Ophthalmol Vis Sci 46:3973–3979

    Google Scholar 

  23. Fernandez-Godino R, Garland DL, Pierce EA (2015) A local complement response by RPE causes early-stage macular degeneration. Hum Mol Genet 24:5555–5569

    CAS  PubMed Central  Google Scholar 

  24. Fu L, Garland D, Yang Z, Shukla D, Rajendran A, Pearson E, Stone EM, Zhang K, Pierce EA (2007) The R345W mutation in EFEMP1 is pathogenic and causes AMD-like deposits in mice. Hum Mol Genet 16:2411–2422

    CAS  Google Scholar 

  25. Marmorstein LY, McLaughlin PJ, Peachey NS, Sasaki T, Marmorstein AD (2007) Formation and progression of sub-retinal pigment epithelium deposits in Efemp1 mutation knock-in mice: a model for the early pathogenic course of macular degeneration. Hum Mol Genet 16:2423–2432

    CAS  Google Scholar 

  26. Garland DL, Fernandez-Godino R, Kaur I, Speicher KD, Harnly JM, Lambris JD, Speicher DW, Pierce EA (2014) Mouse genetics and proteomic analyses demonstrate a critical role for complement in a model of DHRD/ML, an inherited macular degeneration. Hum Mol Genet 23:52–68

    CAS  Google Scholar 

  27. Duvvari MR, van de Ven JP, Geerlings MJ, Saksens NT, Bakker B, Henkes A, Neveling K, del Rosario M, Westra D, van den Heuvel LP et al (2016) Whole exome sequencing in patients with the cuticular drusen subtype of age-related macular degeneration. PLoS One 11:e0152047

    PubMed Central  Google Scholar 

  28. Guymer RH, McNeil R, Cain M, Tomlin B, Allen PJ, Dip CL, Baird PN (2002) Analysis of the Arg345Trp disease-associated allele of the EFEMP1 gene in individuals with early onset drusen or familial age-related macular degeneration. Clin Exp Ophthalmol 30:419–423

    Google Scholar 

  29. Meyer KJ, Davis LK, Schindler EI, Beck JS, Rudd DS, Grundstad AJ, Scheetz TE, Braun TA, Fingert JH, Alward WL et al (2011) Genome-wide analysis of copy number variants in age-related macular degeneration. Hum Genet 129:91–100

    Google Scholar 

  30. Mackay DS, Bennett TM, Shiels A (2015) Exome sequencing identifies a missense variant in EFEMP1 co-segregating in a family with autosomal dominant primary open-angle glaucoma. PLoS One 10:e0132529

    PubMed Central  Google Scholar 

  31. Driver SGW, Jackson MR, Richter K, Tomlinson P, Brockway B, Halliday BJ, Markie DM, Robertson SP, Wade EM (2020) Biallelic variants in EFEMP1 in a man with a pronounced connective tissue phenotype. Eur J Hum Genet 28:445–452

    CAS  Google Scholar 

  32. Bizzari S, El-Bazzal L, Nair P, Younan A, Stora S, Mehawej C, El-Hayek S, Delague V, Megarbane A (2020) Recessive marfanoid syndrome with herniation associated with a homozygous mutation in Fibulin-3. Eur J Med Genet 63:103869

    Google Scholar 

  33. Hasegawa A, Yonezawa T, Taniguchi N, Otabe K, Akasaki Y, Matsukawa T, Saito M, Neo M, Marmorstein LY, Lotz MK (2017) Role of fibulin 3 in aging-related joint changes and osteoarthritis pathogenesis in human and mouse knee cartilage. Arthritis Rheum 69:576–585

    CAS  Google Scholar 

  34. Stanton JB, Marmorstein AD, Zhang Y, Marmorstein LY (2017) Deletion of Efemp1 is protective against the development of sub-RPE deposits in mouse eyes. Invest Ophthalmol Vis Sci 58:1455–1461

    CAS  PubMed Central  Google Scholar 

  35. Timpl R, Sasaki T, Kostka G, Chu ML (2003) Fibulins: a versatile family of extracellular matrix proteins. Nat Rev 4:479–489

    CAS  Google Scholar 

  36. Yanagisawa H, Davis EC, Starcher BC, Ouchi T, Yanagisawa M, Richardson JA, Olson EN (2002) Fibulin-5 is an elastin-binding protein essential for elastic fibre development in vivo. Nature 415:168–171

    Google Scholar 

  37. Tsunezumi J, Sugiura H, Oinam L, Ali A, Thang BQ, Sada A, Yamashiro Y, Kuro OM, Yanagisawa H (2018) Fibulin-7, a heparin binding matricellular protein, promotes renal tubular calcification in mice. Matrix Biol 74:5–20

    CAS  Google Scholar 

  38. Nasser W, Amitai-Lange A, Soteriou D, Hanna R, Tiosano B, Fuchs Y, Shalom-Feuerstein R (2018) Corneal-committed cells restore the stem cell pool and tissue boundary following injury. Cell Rep 22:323–331

    CAS  Google Scholar 

  39. Fabiani C, Barabino S, Rashid S, Dana MR (2009) Corneal epithelial proliferation and thickness in a mouse model of dry eye. Exp Eye Res 89:166–171

    CAS  PubMed Central  Google Scholar 

  40. Quigley HA (2011) Glaucoma. Lancet 377:1367–1377

    Google Scholar 

  41. Thompson S, Blodi FR, Larson DR, Anderson MG, Stasheff SF (2019) The Efemp1R345W Macular dystrophy mutation causes amplified circadian and photophobic responses to light in mice. Invest Ophthalmol Vis Sci 60:2110–2117

    CAS  PubMed Central  Google Scholar 

  42. Wyatt MK, Tsai JY, Mishra S, Campos M, Jaworski C, Fariss RN, Bernstein SL, Wistow G (2013) Interaction of complement factor h and fibulin3 in age-related macular degeneration. PLoS One 8:e68088

    CAS  PubMed Central  Google Scholar 

  43. Brubaker RF (1999) Tonometry and corneal thickness. Arch Ophthalmol 117:104–105

    CAS  Google Scholar 

  44. Douglas RM, Alam NM, Silver BD, McGill TJ, Tschetter WW, Prusky GT (2005) Independent visual threshold measurements in the two eyes of freely moving rats and mice using a virtual-reality optokinetic system. Vis Neurosci 22:677–684

    CAS  Google Scholar 

  45. Prusky GT, Alam NM, Beekman S, Douglas RM (2004) Rapid quantification of adult and developing mouse spatial vision using a virtual optomotor system. Invest Ophthalmol Vis Sci 45:4611–4616

    Google Scholar 

  46. Xuan M, Wang S, Liu X, He Y, Li Y, Zhang Y (2016) Proteins of the corneal stroma: importance in visual function. Cell Tissue Res 364:9–16

    CAS  Google Scholar 

  47. Chakravarti S, Petroll WM, Hassell JR, Jester JV, Lass JH, Paul J, Birk DE (2000) Corneal opacity in lumican-null mice: defects in collagen fibril structure and packing in the posterior stroma. Invest Ophthalmol Vis Sci 41:3365–3373

    CAS  PubMed Central  Google Scholar 

  48. Salchow DJ, Gehle P (2019) Ocular manifestations of Marfan syndrome in children and adolescents. Eur J Ophthalmol 29:38–43

    Google Scholar 

  49. Zhang K, Sun X, Chen Y, Zhong Q, Lin L, Gao Y, Hong F (2018) Doyne honeycomb retinal dystrophy/malattia leventinese induced by EFEMP1 mutation in a Chinese family. BMC Ophthalmol 18:318

    PubMed Central  Google Scholar 

  50. Zayas-Santiago A, Cross SD, Stanton JB, Marmorstein AD, Marmorstein LY (2017) Mutant fibulin-3 causes proteoglycan accumulation and impaired diffusion across Bruch’s membrane. Invest Ophthalmol Vis Sci 58:3046–3054

    CAS  PubMed Central  Google Scholar 

  51. Bessant DA, Ali RR, Bhattacharya SS (2001) Molecular genetics and prospects for therapy of the inherited retinal dystrophies. Curr Opin Genet Dev 11:307–316

    CAS  Google Scholar 

  52. Simonelli F, Maguire AM, Testa F, Pierce EA, Mingozzi F, Bennicelli JL, Rossi S, Marshall K, Banfi S, Surace EM et al (2010) Gene therapy for Leber’s congenital amaurosis is safe and effective through 1.5 years after vector administration. Mol Ther 18:643–650

    CAS  Google Scholar 

  53. Jain A, Zode G, Kasetti RB, Ran FA, Yan W, Sharma TP, Bugge K, Searby CC, Fingert JH, Zhang F et al (2017) CRISPR-Cas9-based treatment of myocilin-associated glaucoma. Proc Natl Acad Sci U S A 114:11199–11204

    CAS  PubMed Central  Google Scholar 

  54. Wu J, Bell OH, Copland DA, Young A, Pooley JR, Maswood R, Evans RS, Khaw PT, Ali RR, Dick AD et al (2020) Gene therapy for glaucoma by ciliary body aquaporin 1 disruption using CRISPR-Cas9. Mol Ther 28:820–829

    CAS  PubMed Central  Google Scholar 

  55. Jo DH, Song DW, Cho CS, Kim UG, Lee KJ, Lee K, Park SW, Kim D, Kim JH, Kim JS et al (2019) CRISPR-Cas9-mediated therapeutic editing of Rpe65 ameliorates the disease phenotypes in a mouse model of Leber congenital amaurosis. Sci Adv 5:eaax1210

    CAS  PubMed Central  Google Scholar 

  56. Petroll WM, Kivanany PB, Hagenasr D, Graham EK (2015) Corneal fibroblast migration patterns during intrastromal wound healing correlate with ECM structure and alignment. Invest Ophthalmol Vis Sci 56:7352–7361

    PubMed Central  Google Scholar 

  57. Petroll WM, Weaver M, Vaidya S, McCulley JP, Cavanagh HD (2013) Quantitative 3-dimensional corneal imaging in vivo using a modified HRT-RCM confocal microscope. Cornea 32:e36–e43

    PubMed Central  Google Scholar 

  58. Cai D, Zhu M, Petroll WM, Koppaka V, Robertson DM (2014) The impact of type 1 diabetes mellitus on corneal epithelial nerve morphology and the corneal epithelium. Am J Pathol 184:2662–2670

    CAS  PubMed Central  Google Scholar 

  59. Zhivov A, Stachs O, Stave J, Guthoff RF (2009) In vivo three-dimensional confocal laser scanning microscopy of corneal surface and epithelium. Br J Ophthalmol 93:667–672

    CAS  Google Scholar 

  60. Kivanany PB, Grose KC, Petroll WM (2016) Temporal and spatial analysis of stromal cell and extracellular matrix patterning following lamellar keratectomy. Exp Eye Res 153:56–64

    CAS  PubMed Central  Google Scholar 

  61. Sun N, Shibata B, Hess JF, FitzGerald PG (2015) An alternative means of retaining ocular structure and improving immunoreactivity for light microscopy studies. Mol Vis 21:428–442

    PubMed Central  Google Scholar 

  62. Daniel S, Clark AF, McDowell CM (2018) Subtype-specific response of retinal ganglion cells to optic nerve crush. Cell Death Dis 4:7

    CAS  Google Scholar 

  63. Boatright JH, Dalal N, Chrenek MA, Gardner C, Ziesel A, Jiang Y, Grossniklaus HE, Nickerson JM (2015) Methodologies for analysis of patterning in the mouse RPE sheet. Mol Vis 21:40–60

    PubMed Central  Google Scholar 

  64. Ramadurgum P, Woodard DR, Daniel S, Peng H, Mallipeddi PL, Niederstrasser H, Mihelakis M, Chau VQ, Douglas PM, Posner BA et al (2020) imultaneous control of endogenous and user-defined genetic pathways using unique ecDHFR pharmacological chaperones. Cell Chem Biol 27:622–634 e626

    CAS  Google Scholar 

Download references

Acknowledgments

The authors would like to personally thank Amber Wilkerson, the UTSW Histo-Pathology Core, and the UTSW Electron Microscopy for their assistance with data acquisition and sample processing. The authors would like to also thank Dr. Zainah Alsagoff, MD (Southland Hospital, Invercargill, New Zealand) for performing the ocular exam.

Funding

VQC was supported by a funding from the UT Southwestern Summer Medical Student Research Program. JDH is supported by an endowment from the Roger and Dorothy Hirl Research Fund, a vision research grant from the Karl Kirchgessner Foundation, a BrightFocus Foundation Macular Degeneration Research Grant (M2016200), an NEI R01 grant (EY027785), and a Career Development Award from Research to Prevent Blindness (RPB). Additional support was provided by an NEI Visual Science Core grant (P30 EY030413) and an unrestricted grant from RPB (both to the UT Southwestern Department of Ophthalmology). GZ is supported by an NEI R01 grant (EY026177). WMP is supported by an NEI R01 grant (EY013322).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to John D. Hulleman.

Ethics declarations

Conflict of interest

The authors declare that they have no conflicts of interest.

Additional information

Publisher’s note

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

Electronic supplementary material

Supplemental Figure 1:

Confirmation of genotypes and F3 knockout. (a) Genotyping products for WT, F3+/-, and F3-/- animals. (b) Primer coverage for Sybr green and TaqMan probes. (c) Confirmation of F3 knockdown using Sybr green and Taqman probes. (PDF 690 kb)

Supplemental Figure 2:

Limbal stem cells are present and proliferating in the absence of F3. (a) Representative images of WT and F3-/- corneas stained for K15, Ki67, and DAPI. Both WT and F3-/- show evidence of limbal stem cell (K15 positive) as well as proliferating cells (Ki67 positive) in 8 mo mice. Dashed lines represent limbal limit (n=4) (scale bar = 100 μm). (PDF 2615 kb)

Supplemental Figure 3:

Additional histologic analyses and IOP measurements. (a) Histological comparison of ciliary body (CB) and TM between WT and F3-/- mice is unremarkable. (b) F3-/- mice exhibit lower IOP than WT mice, but these readings are likely an underestimation of the actual IOP measurements due to thinner corneas in F3-/- mice at 6 mo. (c) Representative images of immunostaining shows localization of F3 in the retina in WT as well as absence of F3 staining in F3-/- (scale bar = 100 μm). (PDF 2184 kb)

Supplemental Figure 4:

Absence of F3 does not lead to astrogliosis. (a) Representative images of immunostaining shows GFAP labeled astrocytes in WT and F3-/- retinas (scale bar = 100 μm). (b) Graph representing intensities (AU) shows no differences between WT and F3-/- in 8 mo mice (n=4). (c) Representative images of immunostaining shows Iba-1 labeled microglia in WT and F3-/- (inner and outer) retinas (scale bar = 100 μm). (d) Graph representing cell density (cells/mm2) shows no differences between WT and F3-/- in 8 mo mice (n=4). (PDF 1864 kb)

Supplemental Figure 5:

Normal nerve fiber, ganglion cell, and inner plexiform thickness in a patient with biallelic loss-of-function mutations in F3. (a, b) Normal combined thickness of the nerve fiber layer (NFL), ganglion cell layer (GCL), and inner plexiform layer (IPL) as determined by OCT suggests no indications of inner retinal thinning or glaucoma. (PDF 1306 kb)

Supplemental Figure 6:

Normal structure and tissue thickness surrounding the optic nerve head in a patient with biallelic loss-of-function mutations in F3. (a, b) Normal retinal nerve fiber layer (RNFL) structure and thickness surrounding the optic nerve head suggests a healthy optic disk with no evidence of cupping or dysfunction indicative of glaucoma. (PDF 2137 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Daniel, S., Renwick, M., Chau, V.Q. et al. Fibulin-3 knockout mice demonstrate corneal dysfunction but maintain normal retinal integrity. J Mol Med 98, 1639–1656 (2020). https://doi.org/10.1007/s00109-020-01974-z

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00109-020-01974-z

Keywords

Navigation