Summary

ייזום בידול בתאי multipotent otic אב הונצחו

Published: January 02, 2016
doi:

Summary

The current protocols to maintain immortalized multipotent otic progenitor (iMOP) cells and otic differentiation are described. Culture conditions and molecular markers that indicate differentiation into sensory epithelia and spiral ganglion neurons (SGN) are highlighted.

Abstract

Use of human induced pluripotent stem cells (iPSC) or embryonic stem cells (ESC) for cell replacement therapies holds great promise. Several limitations including low yields and heterogeneous populations of differentiated cells hinder the progress of stem cell therapies. A fate restricted immortalized multipotent otic progenitor (iMOP) cell line was generated to facilitate efficient differentiation of large numbers of functional hair cells and spiral ganglion neurons (SGN) for inner ear cell replacement therapies. Starting from dissociated cultures of single iMOP cells, protocols that promote cell cycle exit and differentiation by basic fibroblast growth factor (bFGF) withdrawal were described. A significant decrease in proliferating cells after bFGF withdrawal was confirmed using an EdU cell proliferation assay. Concomitant with a decrease in proliferation, successful differentiation resulted in expression of molecular markers and morphological changes. Immunostaining of Cdkn1b (p27KIP) and Cdh1 (E-cadherin) in iMOP-derived otospheres was used as an indicator for differentiation into inner ear sensory epithelia while immunostaining of Cdkn1b and Tubb3 (neuronal β-tubulin) was used to identify iMOP-derived neurons. Use of iMOP cells provides an important tool for understanding cell fate decisions made by inner ear neurosensory progenitors and will help develop protocols for generating large numbers of iPSC or ESC-derived hair cells and SGNs. These methods will accelerate efforts for generating otic cells for replacement therapies.

Introduction

The organs of the inner ear, the cochlea, utricle, saccule and three semicircular canals, mediate the ability to hear and balance. Within the cochlea, hair cells convert sounds into electrical signals that are relayed to the spiral ganglion neurons (SGN). The SGNs fire action potentials to propagate neural signals through the auditory circuit. Genetic mutations, ototoxic drugs and exposure to loud sounds contribute to hair cell and SGN death that result in hearing loss1-4. Once lost, these cells are not replaced. Use of iPSC and ESC to generate nascent hair cells or SGNs holds great promise for inner ear cell replacement therapies5-8. A flurry of progress has shown that pluripotent stem cells and inner ear derived progenitors can differentiate into hair cells and SGNs at various stages of maturity. Mammalian embryonic stem cells (ESC) and induced pluripotent stem cells (iPSC) can be used to generate functional hair cells and SGNs9-11. Stem cells and progenitor cells derived from the mammalian inner ear have also been shown to form hair cells and neurons with properties of their in vivo cellular counterparts12-16.

Use of iPSC or ESC-derived otic progenitors to replace lost hair cells and SGNs requires efficient differentiation. Improper differentiation or continued proliferation of engrafted stem-derived progenitors in the inner ear can exacerbate inner ear function and pose a tumorigenic risk such as teratomas formation in the inner ear17. There is a clear need for developing culture conditions and understanding differentiation of otic progenitors. One strategy in developing these methods is to recapitulate cell fate decisions made by neurosensory progenitors during inner ear development. Protocols that prevent proliferation and direct otic progenitors into hair cells or SGNs will help improve safety as well as efficacy of replacement therapies.

During development, the inner ear begins with the thickening of surface ectoderm in a restricted region between rhombomeres 5 and 6 to become the otic placode. As the otic placode invaginates to form an otic cup, a collection of cells in the anterior region of the otic cup gives rise to the neural-sensory-competent domain (NSD), which contains precursors of hair cells and neurons of the inner ear18. Fate mapping studies from mouse, chicken and zebrafish developing inner ear suggest multiple populations of neurosensory progenitors that give rise to the sensory hair cells, surrounding supporting cells and otic neurons19-22. The high mobility group transcription factor, Sox2, has been implicated in sensory cell specification and used as a marker for inner ear progenitors23,24. Hypomorphic mutations that decrease Sox2 expression levels in the inner ear result in the loss of the hair cells, supporting cells and SGNs in the cochlea25,26.

To study otic progenitor cells undergoing cell fate decisions, a fate restricted immortalized multipotent otic progenitor (iMOP) cell line from Sox2 expressing cochlear progenitors was previously established. iMOP cells were originally derived from embryonic E12.5-13.5 cochlea and infected with a c-Myc retrovirus27. iMOP cells can continually proliferate as colony forming cells known as otospheres and have the capacity to differentiate into hair cells, supporting cells and SGNs27. Understanding the capacity of iMOP cells to differentiate into distinct otic lineages allows application of these findings to efficiently generate iPSC or ESC-derived hair cells and SGNs. Efficient differentiation protocols will open new avenues for cell replacement therapies of inner ear diseases that are recalcitrant to conventional treatments. A crucial issue in generating otic cells by in vitro cell culture is to have differentiation markers that help determine if cells are undergoing differentiating. Cdkn1b (p27KIP) has been extensively used as an early marker for differentiation in developing inner ear, however, expression of Cdkn1b in iMOP cells and how it correlates to differentiation has not been addressed. In this study, the current culture conditions and how Cdkn1b expression correlates to other markers of iMOP differentiation are described.

Protocol

1. שמירה עצמית התחדשות בתאי IMOP הכן iMOP תקשורת ותרבות: DMEM / F12, תוספת 1X B27, 25 מיקרוגרם / מיליליטר וcarbenecillin 20 ng / ml bFGF. לעשות 50 מיליליטר של תקשורת בתרבות iMOP באמצעות ריאגנטים סטרילי. לחמם 49 מיליליטר של DMEM / F12 בחרוטי 50 מיליליטר באמ?…

Representative Results

bFGF נסיגת ירידות הפצת נשק בתאי IMOP כדי להקטין את יכולת השגשוג של תאי iMOP וליזום התמיינות של תאי iMOP, bFGF הוסר ממדפי התרבויות. כדי לוודא שנסיגת גורם גדילה יורדת התפשטות, התאגדות edu הועסקה כassay…

Discussion

ניטור תרבויות IMOP

פרוטוקול לשמירה על התחדשות עצמית וקידום הבידול של שורת תאי iMOP רומן מתואר ופורמטי ציפוי נוספים כלולים (טבלה 1). כמה שלבים קריטיים המסייעים בהרחבה והתמיינות של תאי iMOP שגרה הם ציינו. בדומה לתרביות ?…

Divulgations

The authors have nothing to disclose.

Acknowledgements

The work was supported in part by the Duncan and Nancy MacMillan Faculty Development Chair Endowment Fund (K.Y.K.), Busch Biomedical Research Grant (K.Y.K.) and the Rutgers Faculty Development Grant (K.Y.K.).

Materials

CoolCell LX Alcohol-Free Cell Freezing Containers BioCision BCS-405
Cryogenic Vials (2 ml)  Corning 430654
1.5 Thickness Glass Coverslip (Round 12 mm) Electron Microscopy Sciences 72230-01
DMEM/F12 Life Technologies 11320-082
Neurobasal Medium Life Technologies 21103
Phosphate Buffered Saline (PBS) pH 7.4 Life Technologies 10010-023
Hank's Balanced Salt Solution (HBSS) Life Technologies 14025-092
B27 Supplement (50X) Serum Free Life Technologies 17504-044 Stored as 1 ml aliquots
L-Glutamine(200 mM)  Life Technologies 25030-081 Stored as 5 ml aliquots
Natural Mouse Laminin Life Technologies 23017-015 Stored as 1 mg/ml aliquots
Click-iT EdU Alexa Fluor 488  Life Technologies C10337
Synth-A-Freeze Cryopreservation Media Life Technologies A12542-01
Prolong Gold Antifade Mountant Life Technologies 47743-736 Stored as 10 mg/ml 100 µl aliquots
Moxi Z Mini Automated Cell Counter Orflo  MXZ001
Moxi Z Cassette Type S Orflo  MXC002
Recombinant Murine Fibroblast Growth Factor, basic (bFGF) Peprotech 450-33 Resuspended in 0.1% BSA in H20 and stored as 20 mg/ml aliquots
Poly-D-Lysine Sigma P7886 Resuspended in 1X PBS and stored as 10 mg/ml 100 µl aliquots
Carbenicillin, Disodium Salt Thermo Fisher Scientific BP2648-1 Resuspended in 10mM Hepes pH 7.4 and stored as 100 mg/ml aliquots
5 ml pipet individually wrapped paperback (200/case) Thermo Fisher Scientific 1367811D
10 ml pipet individually wrapped paperback (200/case) Thermo Fisher Scientific 1367811E
Tissue Culture Treated Biolite 24 -Well Plate Thermo Fisher Scientific 130188
Tissue Culture Treated Biolite 6 -Well Plate Thermo Fisher Scientific 130184 
Tissue Culture Treated 6 cm Dish Thermo Fisher Scientific 130181 
EMD Millipore Millex Sterile Syringe PVDF Filter Pore size: 0.22μm Thermo Fisher Scientific SLGV033RS
TipOne filter pipet tips 0.1-10 ul elongated filter tip USA Scientific 1120-3810
TipOne filter pipet tips 1-20 ul filter tip USA Scientific 1120-1810
TipOne filter pipet tips 1-200 ul  filter tip USA Scientific 1120-8810
TipOne filter pipet tips 101-1000 ul filter tip USA Scientific 1126-7810
15 ml conical tubes sterile 20 bags of 25 tubes (500 tubes) USA Scientific 1475-0511
50 ml conical tubes sterile 20 bags of 25 tubes (500 tubes) USA Scientific 1500-1211

References

  1. Petit, C., & Richardson, G. P. Linking genes underlying deafness to hair-bundle development and function. Nat Neurosci. 12 (6), 703-710 (2009).
  2. Kujawa, S. G., & Liberman, M. C. Adding insult to injury: cochlear nerve degeneration after 'temporary' noise-induced hearing loss. J Neurosci. 29 (45), 14077-14085 (2009).
  3. Kujawa, S. G., & Liberman, M. C. Acceleration of age-related hearing loss by early noise exposure: evidence of a misspent youth. J Neurosci. 26 (7), 2115-2123 (2006).
  4. Huth, M. E., Ricci, A. J., & Cheng, A. G. Mechanisms of aminoglycoside ototoxicity and targets of hair cell protection. Int J Otolaryngol. 2011 937861 (2011).
  5. Brigande, J. V., & Heller, S. Quo vadis, hair cell regeneration? Nat Neurosci. 12 (6), 679-685 (2009).
  6. Groves, A. K. The challenge of hair cell regeneration. Exp Biol Med (Maywood). 235 (4), 434-446 (2010).
  7. Okano, T., & Kelley, M. W. Stem cell therapy for the inner ear: recent advances and future directions. Trends Amplif. 16 (1), 4-18 (2012).
  8. Ronaghi, M., Nasr, M., & Heller, S. Concise review: Inner ear stem cells–an oxymoron, but why? Stem Cells. 30 (1), 69-74 (2012).
  9. Chen, W. et al. Restoration of auditory evoked responses by human ES-cell-derived otic progenitors. Nature. 490 (7419), 278-282 (2012).
  10. Koehler, K. R., Mikosz, A. M., Molosh, A. I., Patel, D., & Hashino, E. Generation of inner ear sensory epithelia from pluripotent stem cells in 3D culture. Nature. (2013).
  11. Oshima, K. et al. Mechanosensitive hair cell-like cells from embryonic and induced pluripotent stem cells. Cell. 141 (4), 704-716 (2010).
  12. Diensthuber, M., Oshima, K., & Heller, S. Stem/progenitor cells derived from the cochlear sensory epithelium give rise to spheres with distinct morphologies and features. J Assoc Res Otolaryngol. 10 (2), 173-190 (2009).
  13. Doetzlhofer, A., White, P., Lee, Y. S., Groves, A., & Segil, N. Prospective identification and purification of hair cell and supporting cell progenitors from the embryonic cochlea. Brain Res. 1091 (1), 282-288 (2006).
  14. Doetzlhofer, A., White, P. M., Johnson, J. E., Segil, N., & Groves, A. K. In vitro growth and differentiation of mammalian sensory hair cell progenitors: a requirement for EGF and periotic mesenchyme. Dev Biol. 272 (2), 432-447 (2004).
  15. Martinez-Monedero, R., Yi, E., Oshima, K., Glowatzki, E., & Edge, A. S. Differentiation of inner ear stem cells to functional sensory neurons. Dev Neurobiol. 68 (5), 669-684 (2008).
  16. Oshima, K., Senn, P., & Heller, S. Isolation of sphere-forming stem cells from the mouse inner ear. Methods Mol Biol. 493 141-162 (2009).
  17. Nishimura, K., Nakagawa, T., Sakamoto, T., & Ito, J. Fates of murine pluripotent stem cell-derived neural progenitors following transplantation into mouse cochleae. Cell Transplant. 21 (4), 763-771 (2012).
  18. Wu, D. K., & Kelley, M. W. Molecular mechanisms of inner ear development. Cold Spring Harb Perspect Biol. 4 (8), a008409 (2012).
  19. Jiang, H. et al. Lineage analysis of the late otocyst stage mouse inner ear by transuterine microinjection of a retroviral vector encoding alkaline phosphatase and an oligonucleotide library. PLoS One. 8 (7), e69314 (2013).
  20. Sapede, D., Dyballa, S., & Pujades, C. Cell lineage analysis reveals three different progenitor pools for neurosensory elements in the otic vesicle. J Neurosci. 32 (46), 16424-16434 (2012).
  21. Satoh, T., & Fekete, D. M. Clonal analysis of the relationships between mechanosensory cells and the neurons that innervate them in the chicken ear. Development. 132 (7), 1687-1697 (2005).
  22. Raft, S. et al. Cross-regulation of Ngn1 and Math1 coordinates the production of neurons and sensory hair cells during inner ear development. Development. 134 (24), 4405-4415 (2007).
  23. Neves, J., Parada, C., Chamizo, M., & Giraldez, F. Jagged 1 regulates the restriction of Sox2 expression in the developing chicken inner ear: a mechanism for sensory organ specification. Development. 138 (4), 735-744 (2011).
  24. Mak, A. C., Szeto, I. Y., Fritzsch, B., & Cheah, K. S. Differential and overlapping expression pattern of SOX2 and SOX9 in inner ear development. Gene Expr Patterns. 9 (6), 444-453 (2009).
  25. Kiernan, A. E. et al. Sox2 is required for sensory organ development in the mammalian inner ear. Nature. 434 (7036), 1031-1035 (2005).
  26. Puligilla, C., Dabdoub, A., Brenowitz, S. D., & Kelley, M. W. Sox2 induces neuronal formation in the developing mammalian cochlea. J Neurosci. 30 (2), 714-722 (2010).
  27. Kwan, K. Y., Shen, J., & Corey, D. P. C-MYC transcriptionally amplifies SOX2 target genes to regulate self-renewal in multipotent otic progenitor cells. Stem Cell Reports. 4 (1), 47-60 (2015).
  28. Dittami, G. M., Sethi, M., Rabbitt, R. D., & Ayliffe, H. E. Determination of mammalian cell counts, cell size and cell health using the Moxi Z mini automated cell counter. J Vis Exp. (64) (2012).
  29. Levenstein, M. E. et al. Basic fibroblast growth factor support of human embryonic stem cell self-renewal. Stem Cells. 24 (3), 568-574 (2006).
  30. Furue, M. K. et al. Heparin promotes the growth of human embryonic stem cells in a defined serum-free medium. Proc Natl Acad Sci U S A. 105 (36), 13409-13414 (2008).
  31. Lotz, S. et al. Sustained levels of FGF2 maintain undifferentiated stem cell cultures with biweekly feeding. PLoS One. 8 (2), e56289 (2013).
  32. Brewer, G. J., Torricelli, J. R., Evege, E. K., & Price, P. J. Optimized survival of hippocampal neurons in B27-supplemented Neurobasal, a new serum-free medium combination. J Neurosci Res. 35 (5), 567-576 (1993).
  33. Watanabe, K. et al. A ROCK inhibitor permits survival of dissociated human embryonic stem cells. Nat Biotechnol. 25 (6), 681-686 (2007).
  34. Ruben, R. J. Development of the inner ear of the mouse: a radioautographic study of terminal mitoses. Acta Otolaryngol. Suppl 220:221-244 (1967).
  35. Matei, V. et al. Smaller inner ear sensory epithelia in Neurog 1 null mice are related to earlier hair cell cycle exit. Dev Dyn. 234 (3), 633-650 (2005).
  36. Lee, Y. S., Liu, F., & Segil, N. A morphogenetic wave of p27Kip1 transcription directs cell cycle exit during organ of Corti development. Development. 133 (15), 2817-2826 (2006).
  37. Chen, P., & Segil, N. p27(Kip1) links cell proliferation to morphogenesis in the developing organ of Corti. Development. 126 (8), 1581-1590 (1999).
  38. Hasson, T. et al. Unconventional myosins in inner-ear sensory epithelia. J Cell Biol. 137 (6), 1287-1307 (1997).
  39. Barclay, M., Ryan, A. F., & Housley, G. D. Type I vs type II spiral ganglion neurons exhibit differential survival and neuritogenesis during cochlear development. Neural Dev. 6 33 (2011).
  40. Bermingham, N. A. et al. Math1: an essential gene for the generation of inner ear hair cells. Science. 284 (5421), 1837-1841 (1999).
  41. Izumikawa, M. et al. Auditory hair cell replacement and hearing improvement by Atoh1 gene therapy in deaf mammals. Nat Med. 11 (3), 271-276 (2005).
  42. Kim, W. Y. et al. NeuroD-null mice are deaf due to a severe loss of the inner ear sensory neurons during development. Development. 128 (3), 417-426 (2001).
  43. Liu, M. et al. Essential role of BETA2/NeuroD1 in development of the vestibular and auditory systems. Genes Dev. 14 (22), 2839-2854 (2000).
  44. Ma, Q., Anderson, D. J., & Fritzsch, B. Neurogenin 1 null mutant ears develop fewer, morphologically normal hair cells in smaller sensory epithelia devoid of innervation. J Assoc Res Otolaryngol. 1 (2), 129-143 (2000).
  45. Pang, Z. P. et al. Induction of human neuronal cells by defined transcription factors. Nature. 476 (7359), 220-223 (2011).
  46. Vierbuchen, T. et al. Direct conversion of fibroblasts to functional neurons by defined factors. Nature. 463 (7284), 1035-1041 (2010).
  47. Zhang, Y. et al. Rapid single-step induction of functional neurons from human pluripotent stem cells. Neuron. 78 (5), 785-798 (2013).
  48. Blanchard, J. W. et al. Selective conversion of fibroblasts into peripheral sensory neurons. Nat Neurosci. 18 (1), 25-35 (2015).
check_url/fr/53692?article_type=t

Play Video

Citer Cet Article
Azadeh, J., Song, Z., Laureano, A. S., Toro-Ramos, A., Kwan, K. Initiating Differentiation in Immortalized Multipotent Otic Progenitor Cells. J. Vis. Exp. (107), e53692, doi:10.3791/53692 (2016).

View Video