#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Optogenetically transduced human ES cell-derived neural progenitors and their neuronal progenies: Phenotypic characterization and responses to optical stimulation


Autoři: Jiwon Ryu aff001;  Philippe F. Y. Vincent aff002;  Nikolaos K. Ziogas aff001;  Leyan Xu aff001;  Shirin Sadeghpour aff001;  John Curtin aff001;  Athanasios S. Alexandris aff001;  Nicholas Stewart aff001;  Richard Sima aff002;  Sascha du Lac aff002;  Elisabeth Glowatzki aff002;  Vassilis E. Koliatsos aff001
Působiště autorů: Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America aff001;  Department of Otolaryngology Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America aff002;  Division of Neuropathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America aff003;  Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America aff004
Vyšlo v časopise: PLoS ONE 14(11)
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pone.0224846

Souhrn

Optogenetically engineered human neural progenitors (hNPs) are viewed as promising tools in regenerative neuroscience because they allow the testing of the ability of hNPs to integrate within nervous system of an appropriate host not only structurally, but also functionally based on the responses of their differentiated progenies to light. Here, we transduced H9 embryonic stem cell-derived hNPs with a lentivirus harboring human channelrhodopsin (hChR2) and differentiated them into a forebrain lineage. We extensively characterized the fate and optogenetic functionality of hChR2-hNPs in vitro with electrophysiology and immunocytochemistry. We also explored whether the in vivo phenotype of ChR2-hNPs conforms to in vitro observations by grafting them into the frontal neocortex of rodents and analyzing their survival and neuronal differentiation. Human ChR2-hNPs acquired neuronal phenotypes (TUJ1, MAP2, SMI-312, and synapsin 1 immunoreactivity) in vitro after an average of 70 days of coculturing with CD1 astrocytes and progressively displayed both inhibitory and excitatory neurotransmitter signatures by immunocytochemistry and whole-cell patch clamp recording. Three months after transplantation into motor cortex of naïve or injured mice, 60–70% of hChR2-hNPs at the transplantation site expressed TUJ1 and had neuronal cytologies, whereas 60% of cells also expressed ChR2. Transplant-derived neurons extended axons through major commissural and descending tracts and issued synaptophysin+ terminals in the claustrum, endopiriform area, and corresponding insular and piriform cortices. There was no apparent difference in engraftment, differentiation, or connectivity patterns between injured and sham subjects. Same trends were observed in a second rodent host, i.e. rat, where we employed longer survival times and found that the majority of grafted hChR2-hNPs differentiated into GABAergic neurons that established dense terminal fields and innervated mostly dendritic profiles in host cortical neurons. In physiological experiments, human ChR2+ neurons in culture generated spontaneous action potentials (APs) 100–170 days into differentiation and their firing activity was consistently driven by optical stimulation. Stimulation generated glutamatergic and GABAergic postsynaptic activity in neighboring ChR2- cells, evidence that hChR2-hNP-derived neurons had established functional synaptic connections with other neurons in culture. Light stimulation of hChR2-hNP transplants in vivo generated complicated results, in part because of the variable response of the transplants themselves. Our findings show that we can successfully derive hNPs with optogenetic properties that are fully transferrable to their differentiated neuronal progenies. We also show that these progenies have substantial neurotransmitter plasticity in vitro, whereas in vivo they mostly differentiate into inhibitory GABAergic neurons. Furthermore, neurons derived from hNPs have the capacity of establishing functional synapses with postsynaptic neurons in vitro, but this outcome is technically challenging to explore in vivo. We propose that optogenetically endowed hNPs hold great promise as tools to explore de novo circuit formation in the brain and, in the future, perhaps launch a new generation of neuromodulatory therapies.

Klíčová slova:

Cell differentiation – Neurons – Transplantation immunology – Yellow fluorescent protein – Light pulses – Axons – Astrocytes – Neuronal differentiation


Zdroje

1. Koliatsos VE, Xu L, Cummings BJ. Stem cell therapies for traumatic brain injury. Regen Med. 2015;10(8):917–20. Epub 2015/11/07. doi: 10.2217/rme.15.62 26542417.

2. Cummings BJ, Uchida N, Tamaki SJ, Salazar DL, Hooshmand M, Summers R, et al. Human neural stem cells differentiate and promote locomotor recovery in spinal cord-injured mice. ProcNatlAcadSciUSA. 2005;102(39):14069–74.

3. Han SSW, Kang DY, Mujtaba T, Rao MS, Fischer I. Grafted lineage-restricted precursors differentiate exclusively into neurons in the adult spinal cord. Experimental neurology. 2002;177(2):360–75. doi: 10.1006/exnr.2002.7995 12429183

4. McDonald JW, Liu XZ, Qu Y, Liu S, Mickey SK, Turetsky D, et al. Transplanted embryonic stem cells survive, differentiate and promote recovery in injured rat spinal cord. Nature Med. 1999;5:1410–2. doi: 10.1038/70986 10581084

5. Nasonkin I, Mahairaki V, Xu L, Hatfield G, Cummings BJ, Eberhart C, et al. Long-term, stable differentiation of human embryonic stem cell-derived neural precursors grafted into the adult mammalian neostriatum. Stem cells. 2009;27(10):2414–26. doi: 10.1002/stem.177 19609935

6. Snyder EY. Grafting immortalized neurons to the CNS. ENGLAND. p. 742–51. doi: 10.1016/0959-4388(94)90018-3 7849531

7. Xu L, Ryugo DK, Pongstaporn T, Johe K, Koliatsos VE. Human neural stem cell grafts in the spinal cord of SOD1 transgenic rats: differentiation and structural integration into the segmental motor circuitry. J Comp Neurol. 2009;514(4):297–309. doi: 10.1002/cne.22022 19326469

8. Xu L, Shen P, Hazel T, Johe K, Koliatsos VE. Dual transplantation of human neural stem cells into cervical and lumbar cord ameliorates motor neuron disease in SOD1 transgenic rats. 2011. p. 222–6. doi: 10.1016/j.neulet.2011.03.017 21402124

9. Yan J, Welsh AM, Bora SH, Snyder EY, Koliatsos VE. Differentiation and tropic/trophic effects of exogenous neural precursors in the adult spinal cord. J Comp Neurol. 2004;480(1):101–14. doi: 10.1002/cne.20344 15514921

10. Yan J, Xu L, Welsh AM, Hatfield G, Hazel T, Johe K, et al. Extensive Neuronal Differentiation of Human Neural Stem Cell Grafts in Adult Rat Spinal Cord. PLoS Med. 2007;4(2):e39. doi: 10.1371/journal.pmed.0040039 17298165

11. Xu L, Yan J, Chen D, Welsh AM, Hazel T, Johe K, et al. Human neural stem cell grafts ameliorate motor neuron disease in SOD-1 transgenic rats. Transplantation. 2006;82(7):865–75. doi: 10.1097/01.tp.0000235532.00920.7a 17038899

12. Yan J, Xu L, Welsh AM, Chen D, Hazel T, Johe K, et al. Combined immunosuppressive agents or CD4 antibodies prolong survival of human neural stem cell grafts and improve disease outcomes in amyotrophic lateral sclerosis transgenic mice. 2006. p. 1976–85. doi: 10.1634/stemcells.2005-0518 16644922

13. Ghosh A, Carnahan J, Greenberg ME. Requirement for BDNF in activity-dependent survival of cortical neurons. Science. 1994;263:1618–23. doi: 10.1126/science.7907431 7907431

14. Prakash N, Cohen-Cory S, Frostig RD. Rapid and opposite effects of BDNF and NGF on the functional organization of the adult cortex in vivo. Nature. 1996;381:702–6. doi: 10.1038/381702a0 8649516

15. Zafra F, Hengerer B, Leibrock J, Thoenen H, Lindholm D. Activity dependent regulation of BDNF and NGF mRNAs in the rat hippocampus is mediated by non-NMDA glutamate receptors. The EMBO journal. 1990;9:3545–50. 2170117

16. Daadi MM, Klausner JQ, Bajar B, Goshen I, Lee-Messer C, Lee SY, et al. Optogenetic Stimulation of Neural Grafts Enhances Neurotransmission and Downregulates the Inflammatory Response in Experimental Stroke Model. 2016. p. 1371–80. doi: 10.3727/096368915X688533 26132738

17. Jensen MB, Yan H, Krishnaney-Davison R, Al SA, Zhang SC. Survival and differentiation of transplanted neural stem cells derived from human induced pluripotent stem cells in a rat stroke model. 2013. p. 304–8. doi: 10.1016/j.jstrokecerebrovasdis.2011.09.008 22078778

18. Tornero D, Tsupykov O, Granmo M, Rodriguez C, Gronning-Hansen M, Thelin J, et al. Synaptic inputs from stroke-injured brain to grafted human stem cell-derived neurons activated by sensory stimuli. 2017. p. 692–706. doi: 10.1093/brain/aww347 28115364

19. Kriks S, Shim JW, Piao J, Ganat YM, Wakeman DR, Xie Z, et al. Dopamine neurons derived from human ES cells efficiently engraft in animal models of Parkinson's disease. 2011. p. 547–51. doi: 10.1038/nature10648 22056989

20. Song J, Lee ST, Kang W, Park JE, Chu K, Lee SE, et al. Human embryonic stem cell-derived neural precursor transplants attenuate apomorphine-induced rotational behavior in rats with unilateral quinolinic acid lesions. 2007. p. 58–61. doi: 10.1016/j.neulet.2007.05.066 17669593

21. Wyatt TJ, Rossi SL, Siegenthaler MM, Frame J, Robles R, Nistor G, et al. Human motor neuron progenitor transplantation leads to endogenous neuronal sparing in 3 models of motor neuron loss. 2011. p. 207230. doi: 10.4061/2011/207230 21716648

22. Fandel TM, Trivedi A, Nicholas CR, Zhang H, Chen J, Martinez AF, et al. Transplanted Human Stem Cell-Derived Interneuron Precursors Mitigate Mouse Bladder Dysfunction and Central Neuropathic Pain after Spinal Cord Injury. 2016. p. 544–57. doi: 10.1016/j.stem.2016.08.020 27666009

23. Fujimoto Y, Abematsu M, Falk A, Tsujimura K, Sanosaka T, Juliandi B, et al. Treatment of a mouse model of spinal cord injury by transplantation of human induced pluripotent stem cell-derived long-term self-renewing neuroepithelial-like stem cells. 2012. p. 1163–73. doi: 10.1002/stem.1083 22419556

24. Diaz-Arrastia R, Kochanek PM, Bergold P, Kenney K, Marx C, Grimes J, et al. Pharmacotherapy of Traumatic Brain Injury: State of the Science and the Road Forward Report of the Department of Defense Neurotrauma Pharmacology Workgroup. 2014. p. 135–58. doi: 10.1089/neu.2013.3019 23968241

25. Chambers SM, Fasano CA, Papapetrou EP, Tomishima M, Sadelain M, Studer L. Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling. Nat Biotechnol. 2009;27(3):275–80. doi: 10.1038/nbt.1529 19252484

26. Wen Z, Nguyen HN, Guo Z, Lalli MA, Wang X, Su Y, et al. Synaptic dysregulation in a human iPS cell model of mental disorders. Nature. 2014;515(7527):414–8. doi: 10.1038/nature13716 25132547

27. Mahairaki V, Ryu J, Peters A, Chang Q, Li T, Park TS, et al. Induced Pluripotent Stem Cells from Familial Alzheimer's Disease Patients Differentiate into Mature Neurons with Amyloidogenic Properties. Stem Cells Dev. 2014;23(24):2996–3010. doi: 10.1089/scd.2013.0511 25027006

28. Xu L, Ryu J, Hiel H, Menon A, Aggarwal A, Rha E, et al. Transplantation of human oligodendrocyte progenitor cells in an animal model of diffuse traumatic axonal injury: survival and differentiation. 2015. p. 93. doi: 10.1186/s13287-015-0087-0 25971252

29. Schildge S, Bohrer C, Beck K, Schachtrup C. Isolation and culture of mouse cortical astrocytes. JVisExp. 2013;(71).

30. Anderson AJ, Haus DL, Hooshmand MJ, Perez H, Sontag CJ, Cummings BJ. Achieving stable human stem cell engraftment and survival in the CNS: is the future of regenerative medicine immunodeficient? RegenMed. 2011;6(3):367–406.

31. Marmarou A, Foda MAA, Vandenbrink W, Campbell J, Kita H, Demetriadou K. A New Model of Diffuse Brain Injury in Rats .1. Pathophysiology and Biomechanics. Journal of neurosurgery. 1994;80(2):291–300. doi: 10.3171/jns.1994.80.2.0291 8283269

32. Xu L, Nguyen JV, Lehar M, Menon A, Rha E, Arena J, et al. Repetitive mild traumatic brain injury with impact acceleration in the mouse: Multifocal axonopathy, neuroinflammation, and neurodegeneration in the visual system. Exp Neurol. 2016. p. 436–49. doi: 10.1016/j.expneurol.2014.11.004 25450468

33. Koliatsos VE, Price DL, Clatterbuck RE. Motor neurons in Onuf's nucleus and its rat homologues express the p75 nerve growth factor receptor: sexual dimorphism and regulation by axotomy. J Comp Neurol. 1994;345(4):510–27. doi: 10.1002/cne.903450404 7962698

34. Colombres M, Henriquez JP, Reig GF, Scheu J, Calderon R, Alvarez A, et al. Heparin activates Wnt signaling for neuronal morphogenesis. J Cell Physiol. 2008;216(3):805–15. doi: 10.1002/jcp.21465 18449906

35. Faravelli I, Bucchia M, Rinchetti P, Nizzardo M, Simone C, Frattini E, et al. Motor neuron derivation from human embryonic and induced pluripotent stem cells: experimental approaches and clinical perspectives. Stem Cell ResTher. 2014;5(4):87.

36. Espuny-Camacho I, Michelsen KA, Gall D, Linaro D, Hasche A, Bonnefont J, et al. Pyramidal neurons derived from human pluripotent stem cells integrate efficiently into mouse brain circuits in vivo. Neuron. 2013;77(3):440–56. doi: 10.1016/j.neuron.2012.12.011 23395372

37. Gaspard N, Bouschet T, Hourez R, Dimidschstein J, Naeije G, van dA, et al. An intrinsic mechanism of corticogenesis from embryonic stem cells. Nature. 2008;455(7211):351–7. doi: 10.1038/nature07287 18716623

38. Boyden ES, Zhang F, Bamberg E, Nagel G, Deisseroth K. Millisecond-timescale, genetically targeted optical control of neural activity. 2005. p. 1263–8. doi: 10.1038/nn1525 16116447

39. Avaliani N, Sorensen AT, Ledri M, Bengzon J, Koch P, Brustle O, et al. Optogenetics reveal delayed afferent synaptogenesis on grafted human-induced pluripotent stem cell-derived neural progenitors. 2014. p. 3088–98. doi: 10.1002/stem.1823 25183299

40. Byers B, Lee HJ, Liu J, Weitz AJ, Lin P, Zhang P, et al. Direct in vivo assessment of human stem cell graft-host neural circuits. NeuroImage. 2015;114:328–37. doi: 10.1016/j.neuroimage.2015.03.079 25936696

41. Cunningham M, Cho JH, Leung A, Savvidis G, Ahn S, Moon M, et al. hPSC-derived maturing GABAergic interneurons ameliorate seizures and abnormal behavior in epileptic mice. 2014. p. 559–73. doi: 10.1016/j.stem.2014.10.006 25517465

42. Nicholas CR, Chen J, Tang Y, Southwell DG, Chalmers N, Vogt D, et al. Functional maturation of hPSC-derived forebrain interneurons requires an extended timeline and mimics human neural development. 2013. p. 573–86. doi: 10.1016/j.stem.2013.04.005 23642366

43. Pina-Crespo JC, Talantova M, Cho EG, Soussou W, Dolatabadi N, Ryan SD, et al. High-frequency hippocampal oscillations activated by optogenetic stimulation of transplanted human ESC-derived neurons. 2012. p. 15837–42. doi: 10.1523/JNEUROSCI.3735-12.2012 23136422

44. Steinbeck JA, Choi SJ, Mrejeru A, Ganat Y, Deisseroth K, Sulzer D, et al. Optogenetics enables functional analysis of human embryonic stem cell-derived grafts in a Parkinson's disease model. Nat Biotechnol. 2015;33(2):204–9. doi: 10.1038/nbt.3124 25580598

45. Weick JP, Johnson MA, Skroch SP, Williams JC, Deisseroth K, Zhang SC. Functional control of transplantable human ESC-derived neurons via optogenetic targeting. Stem cells. 2010;28(11):2008–16. doi: 10.1002/stem.514 20827747

46. Weick JP, Liu Y, Zhang SC. Human embryonic stem cell-derived neurons adopt and regulate the activity of an established neural network. 2011. p. 20189–94. doi: 10.1073/pnas.1108487108 22106298

47. Bryson JB, Machado CB, Crossley M, Stevenson D, Bros-Facer V, Burrone J, et al. Optical control of muscle function by transplantation of stem cell-derived motor neurons in mice. 2014. p. 94–7. doi: 10.1126/science.1248523 24700859

48. Stroh A, Tsai HC, Wang LP, Zhang F, Kressel J, Aravanis A, et al. Tracking stem cell differentiation in the setting of automated optogenetic stimulation. 2011. p. 78–88. doi: 10.1002/stem.558 21280159

49. Colasante G, Lignani G, Rubio A, Medrihan L, Yekhlef L, Sessa A, et al. Rapid Conversion of Fibroblasts into Functional Forebrain GABAergic Interneurons by Direct Genetic Reprogramming. 2015. p. 719–34. doi: 10.1016/j.stem.2015.09.002 26526726

50. Rosen AD. Nonlinear temperature modulation of sodium channel kinetics in GH(3) cells. 2001. p. 391–6. doi: 10.1016/s0005-2736(01)00301-7 11286982

51. Yang F, Zheng J. High temperature sensitivity is intrinsic to voltage-gated potassium channels. 2014. p. e03255. doi: 10.7554/eLife.03255 25030910

52. Koch P, Opitz T, Steinbeck JA, Ladewig J, Br++stle O. A rosette-type, self-renewing human ES cell-derived neural stem cell with potential for in vitro instruction and synaptic integration. Proceedings of the National Academy of Sciences of the United States of America; A rosette-type, self-renewing human ES cell-derived neural stem cell with potential for in vitro instruction and synaptic integration. 2009;106(9):3225–30.

53. Ladewig J, Koch P, Endl E, Meiners B, Opitz T, Couillard-Despres S, et al. Lineage selection of functional and cryopreservable human embryonic stem cell-derived neurons. Stem cells. 2008;26(7):1705–12. doi: 10.1634/stemcells.2008-0007 18420830

54. Niclis JC, Turner C, Durnall J, McDougal S, Kauhausen JA, Leaw B, et al. Long-Distance Axonal Growth and Protracted Functional Maturation of Neurons Derived from Human Induced Pluripotent Stem Cells After Intracerebral Transplantation. 2017. p. 1547–56. doi: 10.1002/sctm.16-0198 28198124

55. Wang DD, Kriegstein AR. Defining the role of GABA in cortical development. J Physiol. 2009;587(Pt 9):1873–9. Epub 2009/01/21. doi: 10.1113/jphysiol.2008.167635 19153158; PubMed Central PMCID: PMC2689328.

56. Frysinger RC, Harper RM. Cardiac and respiratory relationships with neural discharge in the anterior cingulate cortex during sleep-walking states. Exp Neurol. 1986;94(2):247–63. Epub 1986/11/01. doi: 10.1016/0014-4886(86)90100-7 3770117.

57. Herrero JL, Khuvis S, Yeagle E, Cerf M, Mehta AD. Breathing above the brain stem: volitional control and attentional modulation in humans. J Neurophysiol. 2018;119(1):145–59. Epub 2017/09/29. doi: 10.1152/jn.00551.2017 28954895; PubMed Central PMCID: PMC5866472.

58. Pattinson KT, Governo RJ, MacIntosh BJ, Russell EC, Corfield DR, Tracey I, et al. Opioids depress cortical centers responsible for the volitional control of respiration. J Neurosci. 2009;29(25):8177–86. Epub 2009/06/26. doi: 10.1523/JNEUROSCI.1375-09.2009 19553457.

59. Vedyasova OA. Respiratory effects of stimulation of the limbic cortex in rats and their modulation with serotonin. Bull Exp Biol Med. 2005;140(3):265–7. Epub 2005/11/25. doi: 10.1007/s10517-005-0462-4 16307031.


Článok vyšiel v časopise

PLOS One


2019 Číslo 11
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#