Clin Exp Otorhinolaryngol.  2015 Jun;8(2):83-91. 10.3342/ceo.2015.8.2.83.

Neural-Induced Human Mesenchymal Stem Cells Promote Cochlear Cell Regeneration in Deaf Guinea Pigs

Affiliations
  • 1Department of Physiology, Chonnam National University Medical School, Gwangju, Korea.
  • 2The Brain Korea 21 Project, Center for Biomedical Human Resources at Chonnam National University, Gwangju, Korea.
  • 3Research Institute of Medical Sciences, Chonnam National University, Gwangju, Korea. choyb@chonnam.ac.kr
  • 4Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Medical School, Gwangju, Korea.
  • 5Department of Pathology, Chonnam National University Medical School, Gwangju, Korea.
  • 6Department of Physiology, Chosun University College of Medicine, Gwangju, Korea.

Abstract


OBJECTIVES
In mammals, cochlear hair cell loss is irreversible and may result in a permanent sensorineural hearing loss. Secondary to this hair cell loss, a progressive loss of spiral ganglion neurons (SGNs) is presented. In this study, we have investigated the effects of neural-induced human mesenchymal stem cells (NI-hMSCs) from human bone marrow on sensory neuronal regeneration from neomycin treated deafened guinea pig cochleae.
METHODS
HMSCs were isolated from the bone marrow which was obtained from the mastoid process during mastoidectomy for ear surgery. Following neural induction with basic fibroblast growth factor and forskolin, we studied the several neural marker and performed electrophysiological analysis. NI-hMSCs were transplanted into the neomycin treated deafened guinea pig cochlea. Engraftment of NI-hMSCs was evaluated immunohistologically at 8 weeks after transplantation.
RESULTS
Following neural differentiation, hMSCs expressed high levels of neural markers, ionic channel markers, which are important in neural function, and tetrodotoxin-sensitive voltage-dependent sodium currents. After transplantation into the scala tympani of damaged cochlea, NI-hMSCs-injected animals exhibited a significant increase in the number of SGNs compared to Hanks balanced salt solution-injected animals. Transplanted NI-hMSCs were found within the perilymphatic space, the organ of Corti, along the cochlear nerve fibers, and in the spiral ganglion. Furthermore, the grafted NI-hMSCs migrated into the spiral ganglion where they expressed the neuron-specific marker, NeuN.
CONCLUSION
The results show the potential of NI-hMSCs to give rise to replace the lost cochlear cells in hearing loss mammals.

Keyword

Hair Cells, Auditory; Mesenchymal Stem Cells; Hearing Loss; Cell Differentiation; Transplantation

MeSH Terms

Animals
Bone Marrow
Cell Differentiation
Cochlea
Cochlear Nerve
Colforsin
Ear
Fibroblast Growth Factor 2
Guinea Pigs*
Hair
Hearing Loss
Hearing Loss, Sensorineural
Humans
Ion Channels
Mammals
Mastoid
Mesenchymal Stromal Cells*
Neomycin
Neurons
Organ of Corti
Regeneration*
Scala Tympani
Sensory Receptor Cells
Sodium
Spiral Ganglion
Transplantation
Transplants
Colforsin
Fibroblast Growth Factor 2
Ion Channels
Neomycin
Sodium

Figure

  • Fig. 1 Characterization of bone marrow-derived human mesenchymal stem cells (hMSCs). Primary hMSCs expressed MSC specific markers including CD13 (+), CD44 (+), CD90 (+), and CD166 (+), but did not express hematopoietic stem cell markers including CD14 (-), CD34 (-), and CD45 (-).

  • Fig. 2 In vitro differentiation of human mesenchymal stem cells (hMSCs). hMSCs were induced to differentiate into neural cells in the presence of basic fibroblast growth factor and forskolin for two weeks. (A) Immunocytochemistry revealed that the expressions of neurofilament-L (NF-L), neurofilament-M (NF-M), neurofilament-H (NF-H), microtubule-associated protein (MAP2), neuronal nuclei (NeuN), β-tubulin III (Tuj1), and glial fibrillary acidic protein (GFAP) in neural-induced hMSCs (NI-hMSCs) were increased than those in primary hMSCs. (B) Immunocytochemical data depicted the high ratio of NI-hMSCs expressing above neural markers. The number of positive cells was counted and the ratio to the number of nuclei was analyzed for each antigen (n=8, *P<0.05 compared with primary hMSCs).

  • Fig. 3 Electrophysiological features of the neural-induced human mesenchymal stem cells (NI-hMSCs). (A) hMSCs demonstrated neuronal characteristics after neural differentiation under voltage-clamp recording. The holding potential was -80 mV and depolarizing steps were applied from -80 mV to +40 mV in 10 mV increments. Large voltage-dependent sodium currents were activated evidently from a depolarizing step of -30 mV and blocked reversibly by tetrodotoxin (TTX) 100 nM. In addition, the hMSCs grown with basic fibroblast growth factor (bFGF) and forskolin showed sustained outward potassium currents as well. Peak current-voltage relationship was plotted against the voltages and demonstrated the voltage-dependence of potassium currents (IK) and sodium currents (INa) (n=16). (B) Under current clamp condition, the resting membrane potential of NI-hMSCs was recorded more negatively than that of control hMSCs grown without bFGF and forskolin (n=16, **P<0.01 compare with primary hMSCs). (C) Expression of molecular markers for ion channel subunits was increased after neural differentiation in hMSCs. The mRNA expression of human large-conductance, voltage- and calcium-dependent K+ channel marker, MaxiK; voltage-dependent K+ channel marker, Kv1.4, Kv4.2, and Kv4.3; either-à-go-go K+ channel marker, Eag1 and Eag2; tetrodotoxin-sensitive Na+ channel marker, NE-Na; voltage-dependent L-type Ca2+ channel, alpha 1C subunit marker, CACNA1C; and voltage-dependent T-type Ca2+ channel, alpha 1G subunit marker, CACNA1G were increased in NI-hMSCs, however, that of TTX-insensitive sodium channel marker, SCN5A were not detected. Reverse transcription-polymerase chain reaction assay was repeated five times independently from different cells. The representative data are shown. (D) The intensity of each gene was normalized to GAPDH and these results were repeated at least three times (*P<0.05 compared with primary hMSCs).

  • Fig. 4 Hematoxylin and eosin staining at eight weeks after neural-induced human mesenchymal stem cells (NI-hMSCs) transplantation. (A, B) All turns of cochlea were studied and basal one was magnified as a box, individually. (A) In a Hanks balanced salt solution (HBSS) alone-injected animal, severe loss of sensory hair cells in the organ of Corti and the degeneration of spiral ganglion neurons were observed. (B) However, cochlea of a NI-hMSCs-injected guinea pig demonstrated relatively preserved organ of Corti and spiral ganglion. (C) Quantification of spiral ganglion cell counts demonstrated that cochleae of NI-hMSCs-injected animals exhibited a significant increase in the number of cell body compared to HBSS alone-injected animals (n=8, **P<0.01). Normal animals (n=10) were used as a control. OC, organ of Corti; SG, spiral ganglion; SM, scala media; ST, scala tympani; SV, scala vestibule; IHC, inner hair cell; OHC, outer hair cell. Scale bars indicate 50 µm.

  • Fig. 5 Localization and in vivo differentiation of transplanted human mesenchymal stem cells (hMSCs). (A-D) Immunohistochemical study was performed eight weeks after neural-induced hMSCs (NI-hMSCs) transplantation into 10% neomycin-treated guinea pig inner ear. (A) Transplanted hMSCs stained with human nuclei (hNuclei; red) were found along the cochlear nerve fibers close to the organ of Corti. (B) Human nuclei expressing transplant derived-hMSCs (red) were located in the inner hair cell layer (arrow) and supporting cell layer (arrowhead). (C) NI-hMSCs (green) migrated into the damaged spiral ganglion were stained with microtubule-associated protein (MAP2) (red), and merged as yellow. These results depict that NI-hMSCs have the capacity not only to survive and localize in the inner ear, but also to regenerate or replace the damaged cochlear cell types. (D) Quantification of spiral ganglion neuron (MAP2+) and transplant-derived cell (hNuclei+) counts indicated that transplanted hMSCs were transdifferentiated into mature neurons in the spiral ganglion. CN, cochlear nerve fibers; OC, organ of Corti; SG, spiral ganglion; SL, spiral limbus; SM, scala media; ST, scala tympani; SV, scala vestibule.


Reference

1. Holley MC. Keynote review: the auditory system, hearing loss and potential targets for drug development. Drug Discov Today. 2005; 10. 10(19):1269–1282. PMID: 16214671.
Article
2. Warchol ME, Lambert PR, Goldstein BJ, Forge A, Corwin JT. Regenerative proliferation in inner ear sensory epithelia from adult guinea pigs and humans. Science. 1993; 3. 259(5101):1619–1622. PMID: 8456285.
Article
3. Ernfors P, Kucera J, Lee KF, Loring J, Jaenisch R. Studies on the physiological role of brain-derived neurotrophic factor and neurotrophin-3 in knockout mice. Int J Dev Biol. 1995; 10. 39(5):799–807. PMID: 8645564.
4. Tan J, Shepherd RK. Aminoglycoside-induced degeneration of adult spiral ganglion neurons involves differential modulation of tyrosine kinase B and p75 neurotrophin receptor signaling. Am J Pathol. 2006; 8. 169(2):528–543. PMID: 16877354.
Article
5. Duan M, Agerman K, Ernfors P, Canlon B. Complementary roles of neurotrophin 3 and a N-methyl-D-aspartate antagonist in the protection of noise and aminoglycoside-induced ototoxicity. Proc Natl Acad Sci U S A. 2000; 6. 97(13):7597–7602. PMID: 10861021.
Article
6. Izumikawa M, Minoda R, Kawamoto K, Abrashkin KA, Swiderski DL, Dolan DF, et al. Auditory hair cell replacement and hearing improvement by Atoh1 gene therapy in deaf mammals. Nat Med. 2005; 3. 11(3):271–276. PMID: 15711559.
Article
7. Sage C, Huang M, Karimi K, Gutierrez G, Vollrath MA, Zhang DS, et al. Proliferation of functional hair cells in vivo in the absence of the retinoblastoma protein. Science. 2005; 2. 307(5712):1114–1118. PMID: 15653467.
Article
8. Li H, Roblin G, Liu H, Heller S. Generation of hair cells by stepwise differentiation of embryonic stem cells. Proc Natl Acad Sci U S A. 2003; 11. 100(23):13495–13500. PMID: 14593207.
Article
9. Oshima K, Heller S. Sound from silence. Nat Med. 2005; 3. 11(3):249–250. PMID: 15746932.
Article
10. Kim SS, Yoo SW, Park TS, Ahn SC, Jeong HS, Kim JW, et al. Neural induction with neurogenin1 increases the therapeutic effects of mesenchymal stem cells in the ischemic brain. Stem Cells. 2008; 9. 26(9):2217–2228. PMID: 18617687.
Article
11. Liechty KW, MacKenzie TC, Shaaban AF, Radu A, Moseley AM, Deans R, et al. Human mesenchymal stem cells engraft and demonstrate site-specific differentiation after in utero transplantation in sheep. Nat Med. 2000; 11. 6(11):1282–1286. PMID: 11062543.
Article
12. Dezawa M, Kanno H, Hoshino M, Cho H, Matsumoto N, Itokazu Y, et al. Specific induction of neuronal cells from bone marrow stromal cells and application for autologous transplantation. J Clin Invest. 2004; 6. 113(12):1701–1710. PMID: 15199405.
Article
13. Chen W, Jongkamonwiwat N, Abbas L, Eshtan SJ, Johnson SL, Kuhn S, et al. Restoration of auditory evoked responses by human ES-cell-derived otic progenitors. Nature. 2012; 10. 490(7419):278–282. PMID: 22972191.
Article
14. Parker MA, Corliss DA, Gray B, Anderson JK, Bobbin RP, Snyder EY, et al. Neural stem cells injected into the sound-damaged cochlea migrate throughout the cochlea and express markers of hair cells, supporting cells, and spiral ganglion cells. Hear Res. 2007; 10. 232(1-2):29–43. PMID: 17659854.
Article
15. Revoltella RP, Papini S, Rosellini A, Michelini M, Franceschini V, Ciorba A, et al. Cochlear repair by transplantation of human cord blood CD133+ cells to nod-scid mice made deaf with kanamycin and noise. Cell Transplant. 2008; 17(6):665–678. PMID: 18819255.
Article
16. Nishimura K, Nakagawa T, Ono K, Ogita H, Sakamoto T, Yamamoto N, et al. Transplantation of mouse induced pluripotent stem cells into the cochlea. Neuroreport. 2009; 9. 20(14):1250–1254. PMID: 19625987.
Article
17. Li H, Liu H, Heller S. Pluripotent stem cells from the adult mouse inner ear. Nat Med. 2003; 10. 9(10):1293–1299. PMID: 12949502.
Article
18. Jang S, Cho HH, Cho YB, Park JS, Jeong HS. Functional neural differentiation of human adipose tissue-derived stem cells using bFGF and forskolin. BMC Cell Biol. 2010; 4. 11:25. PMID: 20398362.
Article
19. Cho YB, Cho HH, Jang S, Jeong HS, Park JS. Transplantation of neural differentiated human mesenchymal stem cells into the cochlea of an auditory-neuropathy guinea pig model. J Korean Med Sci. 2011; 4. 26(4):492–498. PMID: 21468255.
Article
20. Selimoglu E. Aminoglycoside-induced ototoxicity. Curr Pharm Des. 2007; 13(1):119–126. PMID: 17266591.
Article
21. Jiang Y, Henderson D, Blackstad M, Chen A, Miller RF, Verfaillie CM. Neuroectodermal differentiation from mouse multipotent adult progenitor cells. Proc Natl Acad Sci U S A. 2003; 9. 100(Suppl 1):11854–11860. PMID: 12925733.
Article
22. Anghileri E, Marconi S, Pignatelli A, Cifelli P, Galie M, Sbarbati A, et al. Neuronal differentiation potential of human adipose-derived mesenchymal stem cells. Stem Cells Dev. 2008; 10. 17(5):909–916. PMID: 18564036.
Article
23. Li GR, Deng XL, Sun H, Chung SS, Tse HF, Lau CP. Ion channels in mesenchymal stem cells from rat bone marrow. Stem Cells. 2006; 6. 24(6):1519–1528. PMID: 16484345.
Article
24. Sharif S, Nakagawa T, Ohno T, Matsumoto M, Kita T, Riazuddin S, et al. The potential use of bone marrow stromal cells for cochlear cell therapy. Neuroreport. 2007; 3. 18(4):351–354. PMID: 17435601.
Article
25. Kamiya K, Fujinami Y, Hoya N, Okamoto Y, Kouike H, Komatsuzaki R, et al. Mesenchymal stem cell transplantation accelerates hearing recovery through the repair of injured cochlear fibrocytes. Am J Pathol. 2007; 7. 171(1):214–226. PMID: 17591967.
Article
26. Matsuoka AJ, Kondo T, Miyamoto RT, Hashino E. Enhanced survival of bone-marrow-derived pluripotent stem cells in an animal model of auditory neuropathy. Laryngoscope. 2007; 9. 117(9):1629–1635. PMID: 17632425.
Article
27. Hu Z, Wei D, Johansson CB, Holmstrom N, Duan M, Frisen J, et al. Survival and neural differentiation of adult neural stem cells transplanted into the mature inner ear. Exp Cell Res. 2005; 1. 302(1):40–47. PMID: 15541724.
Article
28. Chen X, Li Y, Wang L, Katakowski M, Zhang L, Chen J, et al. Ischemic rat brain extracts induce human marrow stromal cell growth factor production. Neuropathology. 2002; 12. 22(4):275–279. PMID: 12564767.
Article
29. Kelly S, Bliss TM, Shah AK, Sun GH, Ma M, Foo WC, et al. Transplanted human fetal neural stem cells survive, migrate, and differentiate in ischemic rat cerebral cortex. Proc Natl Acad Sci U S A. 2004; 8. 101(32):11839–11844. PMID: 15280535.
Article
Full Text Links
  • CEO
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr