Induced Pluripotent Stem Cells in Drug Discovery and Neurodegenerative Disease Modelling
Abstract
:1. Introduction
1.1. Basic Concepts
1.2. Neurodegenerative Disease Modelling by iPSC Technology
1.3. Drug Testing in Neurodegenerative Diseases Using iPSCs
1.4. Advances in Specific Conditions Using iPSCs
1.4.1. Parkinson’s Disease
1.4.2. Alzheimer Disease
1.4.3. Amyotrophic Lateral Sclerosis
1.4.4. Multiple Sclerosis
1.4.5. Huntington’s Disease
2. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Dorsey, E.R.; Constantinescu, R.; Thompson, J.P.; Biglan, K.M.; Holloway, R.G.; Kieburtz, K.; Marshall, F.J.; Ravina, B.M.; Schifitto, G.; Siderowf, A.; et al. Projected number of people with Parkinson disease in the most populous nations, 2005 through 2030. Neurology 2007, 68, 384–386. [Google Scholar] [CrossRef]
- Arthur, K.C.; Calvo, A.; Price, T.R.; Geiger, J.T.; Chiò, A.; Traynor, B.J. Projected increase in amyotrophic lateral sclerosis from 2015 to 2040. Nat. Commun. 2016, 7, 12408. [Google Scholar] [CrossRef]
- Chang, Y.; Kim, J.; Park, H.; Choi, H. Modelling neurodegenerative diseases with 3D brain organoids. Biol. Rev. Camb. Philos. Soc. 2020, 95, 1497–1509. [Google Scholar] [CrossRef]
- Purves, D.; Augustine, G.J.; Fitzpatrick, D.; Hall, W.C.; LaMantia, A.-S.; White, L.E. Neuroscience, 5th ed.; Sinauer Associates: Sunderland, MA, USA, 2012. [Google Scholar]
- Kamm, F.M. Ethical issues in using and not using embryonic stem cells. Stem Cell Rev. 2005, 1, 325–330. [Google Scholar] [CrossRef]
- Gurdon, J.B. The developmental capacity of nuclei taken from intestinal epithelium cells of feeding tadpoles. J. Embryol. Exp. Morphol. 1962, 10, 622–640. [Google Scholar] [CrossRef]
- Wilmut, I.; Schnieke, A.E.; McWhir, J.; Kind, A.J.; Campbell, K.H. Viable offspring derived from fetal and adult mammalian cells. Nature 1997, 385, 810–813. [Google Scholar] [CrossRef]
- Gehring, W.J. Homeo boxes in the study of development. Science 1987, 236, 1245–1252. [Google Scholar] [CrossRef]
- Davis, R.L.; Weintraub, H.; Lassar, A.B. Expression of a single transfected cDNA converts fibroblasts to myoblasts. Cell 1987, 51, 987–1000. [Google Scholar] [CrossRef]
- Evans, M.J.; Kaufman, M.H. Establishment in culture of pluripotential cells from mouse embryos. Nature 1981, 292, 154–156. [Google Scholar] [CrossRef]
- Thomson, J.A.; Itskovitz-Eldor, J.; Shapiro, S.S.; Waknitz, M.A.; Swiergiel, J.J.; Marshall, V.S.; Jones, J.M. Embryonic stem cell lines derived from human blastocysts. Science 1998, 282, 1145–1147. [Google Scholar] [CrossRef]
- Smith, A.G.; Heath, J.K.; Donaldson, D.D.; Wong, G.G.; Moreau, J.; Stahl, M.; Rogers, D. Inhibition of pluripotential embryonic stem cell differentiation by purified polypeptides. Nature 1988, 336, 688–690. [Google Scholar] [CrossRef] [PubMed]
- Takahashi, K.; Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006, 126, 663–676. [Google Scholar] [CrossRef] [PubMed]
- Fusaki, N.; Ban, H.; Nishiyama, A.; Saeki, K.; Hasegawa, M. Efficient induction of transgene-free human pluripotent stem cells using a vector based on Sendai virus, an RNA virus that does not integrate into the host genome. Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 2009, 85, 348–362. [Google Scholar] [CrossRef] [PubMed]
- Hou, P.; Li, Y.; Zhang, X.; Liu, C.; Guan, J.; Li, H.; Zhao, T.; Ye, J.; Yang, W.; Liu, K.; et al. Pluripotent stem cells induced from mouse somatic cells by small-molecule compounds. Science 2013, 341, 651–654. [Google Scholar] [CrossRef] [PubMed]
- Warren, L.; Manos, P.D.; Ahfeldt, T.; Loh, Y.H.; Li, H.; Lau, F.; Ebina, W.; Mandal, P.K.; Smith, Z.D.; Meissner, A.; et al. Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA. Cell Stem Cell 2010, 7, 618–630. [Google Scholar] [CrossRef] [PubMed]
- Aboul-Soud, M.A.M.; Alzahrani, A.J.; Mahmoud, A. Induced Pluripotent Stem Cells (iPSCs)-Roles in Regenerative Therapies, Disease Modelling and Drug Screening. Cells 2021, 10, 2319. [Google Scholar] [CrossRef] [PubMed]
- Verwer, R.W.; Dubelaar, E.J.; Hermens, W.T.; Swaab, D.F. Tissue cultures from adult human postmortem subcortical brain areas. J. Cell Mol. Med. 2002, 6, 429–432. [Google Scholar] [CrossRef]
- Meijboom, K.E.; Abdallah, A.; Fordham, N.P.; Nagase, H.; Rodriguez, T.; Kraus, C.; Gendron, T.F.; Krishnan, G.; Esanov, R.; Andrade, N.S.; et al. CRISPR/Cas9-mediated excision of ALS/FTD-causing hexanucleotide repeat expansion in C9ORF72 rescues major disease mechanisms in vivo and in vitro. Nat. Commun. 2022, 13, 6286. [Google Scholar] [CrossRef]
- Li, C.; Fleck, J.S.; Martins-Costa, C.; Burkard, T.R.; Themann, J.; Stuempflen, M.; Peer, A.M.; Vertesy, Á.; Littleboy, J.B.; Esk, C.; et al. Single-cell brain organoid screening identifies developmental defects in autism. Nature 2023, 621, 373–380. [Google Scholar] [CrossRef]
- Nakazawa, T. Modeling schizophrenia with iPS cell technology and disease mouse models. Neurosci. Res. 2022, 175, 46–52. [Google Scholar] [CrossRef]
- Sharma, A.; Sances, S.; Workman, M.J.; Svendsen, C.N. Multi-lineage Human iPSC-Derived Platforms for Disease Modeling and Drug Discovery. Cell Stem Cell 2020, 26, 309–329. [Google Scholar] [CrossRef] [PubMed]
- Wang, H. Modeling Neurological Diseases With Human Brain Organoids. Front. Synaptic Neurosci. 2018, 10, 15. [Google Scholar] [CrossRef] [PubMed]
- Qian, X.; Nguyen, H.N.; Song, M.M.; Hadiono, C.; Ogden, S.C.; Hammack, C.; Yao, B.; Hamersky, G.R.; Jacob, F.; Zhong, C.; et al. Brain-Region-Specific Organoids Using Mini-bioreactors for Modeling ZIKV Exposure. Cell 2016, 165, 1238–1254. [Google Scholar] [CrossRef]
- Muguruma, K.; Nishiyama, A.; Kawakami, H.; Hashimoto, K.; Sasai, Y. Self-organization of polarized cerebellar tissue in 3D culture of human pluripotent stem cells. Cell Rep. 2015, 10, 537–550. [Google Scholar] [CrossRef] [PubMed]
- Qian, X.; Song, H.; Ming, G.L. Brain organoids: Advances, applications and challenges. Development 2019, 146, dev.166074. [Google Scholar] [CrossRef]
- Pantazis, C.B.; Yang, A.; Lara, E.; McDonough, J.A.; Blauwendraat, C.; Peng, L.; Oguro, H.; Kanaujiya, J.; Zou, J.; Sebesta, D.; et al. A reference human induced pluripotent stem cell line for large-scale collaborative studies. Cell Stem Cell 2022, 29, 1685–1702.e1622. [Google Scholar] [CrossRef]
- Kwart, D.; Gregg, A.; Scheckel, C.; Murphy, E.A.; Paquet, D.; Duffield, M.; Fak, J.; Olsen, O.; Darnell, R.B.; Tessier-Lavigne, M. A Large Panel of Isogenic APP and PSEN1 Mutant Human iPSC Neurons Reveals Shared Endosomal Abnormalities Mediated by APP β-CTFs, Not Aβ. Neuron 2019, 104, 256–270.e255. [Google Scholar] [CrossRef]
- Raja, W.K.; Mungenast, A.E.; Lin, Y.T.; Ko, T.; Abdurrob, F.; Seo, J.; Tsai, L.H. Self-Organizing 3D Human Neural Tissue Derived from Induced Pluripotent Stem Cells Recapitulate Alzheimer’s Disease Phenotypes. PLoS ONE 2016, 11, e0161969. [Google Scholar] [CrossRef]
- Chen, X.; Sun, G.; Tian, E.; Zhang, M.; Davtyan, H.; Beach, T.G.; Reiman, E.M.; Blurton-Jones, M.; Holtzman, D.M.; Shi, Y. Modeling Sporadic Alzheimer’s Disease in Human Brain Organoids under Serum Exposure. Adv. Sci. 2021, 8, e2101462. [Google Scholar] [CrossRef]
- Devine, M.J.; Ryten, M.; Vodicka, P.; Thomson, A.J.; Burdon, T.; Houlden, H.; Cavaleri, F.; Nagano, M.; Drummond, N.J.; Taanman, J.W.; et al. Parkinson’s disease induced pluripotent stem cells with triplication of the α-synuclein locus. Nat. Commun. 2011, 2, 440. [Google Scholar] [CrossRef]
- Woodard, C.M.; Campos, B.A.; Kuo, S.H.; Nirenberg, M.J.; Nestor, M.W.; Zimmer, M.; Mosharov, E.V.; Sulzer, D.; Zhou, H.; Paull, D.; et al. iPSC-derived dopamine neurons reveal differences between monozygotic twins discordant for Parkinson’s disease. Cell Rep. 2014, 9, 1173–1182. [Google Scholar] [CrossRef] [PubMed]
- Schweitzer, J.S.; Song, B.; Herrington, T.M.; Park, T.Y.; Lee, N.; Ko, S.; Jeon, J.; Cha, Y.; Kim, K.; Li, Q.; et al. Personalized iPSC-Derived Dopamine Progenitor Cells for Parkinson’s Disease. N. Engl. J. Med. 2020, 382, 1926–1932. [Google Scholar] [CrossRef] [PubMed]
- Yagi, T.; Ito, D.; Okada, Y.; Akamatsu, W.; Nihei, Y.; Yoshizaki, T.; Yamanaka, S.; Okano, H.; Suzuki, N. Modeling familial Alzheimer’s disease with induced pluripotent stem cells. Hum. Mol. Genet. 2011, 20, 4530–4539. [Google Scholar] [CrossRef]
- Muratore, C.R.; Rice, H.C.; Srikanth, P.; Callahan, D.G.; Shin, T.; Benjamin, L.N.; Walsh, D.M.; Selkoe, D.J.; Young-Pearse, T.L. The familial Alzheimer’s disease APPV717I mutation alters APP processing and Tau expression in iPSC-derived neurons. Hum. Mol. Genet. 2014, 23, 3523–3536. [Google Scholar] [CrossRef] [PubMed]
- Kondo, T.; Imamura, K.; Funayama, M.; Tsukita, K.; Miyake, M.; Ohta, A.; Woltjen, K.; Nakagawa, M.; Asada, T.; Arai, T.; et al. iPSC-Based Compound Screening and In Vitro Trials Identify a Synergistic Anti-amyloid β Combination for Alzheimer’s Disease. Cell Rep. 2017, 21, 2304–2312. [Google Scholar] [CrossRef] [PubMed]
- Xu, X.; Lei, Y.; Luo, J.; Wang, J.; Zhang, S.; Yang, X.J.; Sun, M.; Nuwaysir, E.; Fan, G.; Zhao, J.; et al. Prevention of β-amyloid induced toxicity in human iPS cell-derived neurons by inhibition of Cyclin-dependent kinases and associated cell cycle events. Stem Cell Res. 2013, 10, 213–227. [Google Scholar] [CrossRef]
- Israel, M.A.; Yuan, S.H.; Bardy, C.; Reyna, S.M.; Mu, Y.; Herrera, C.; Hefferan, M.P.; Van Gorp, S.; Nazor, K.L.; Boscolo, F.S.; et al. Probing sporadic and familial Alzheimer’s disease using induced pluripotent stem cells. Nature 2012, 482, 216–220. [Google Scholar] [CrossRef]
- Iovino, M.; Agathou, S.; González-Rueda, A.; Del Castillo Velasco-Herrera, M.; Borroni, B.; Alberici, A.; Lynch, T.; O’Dowd, S.; Geti, I.; Gaffney, D.; et al. Early maturation and distinct tau pathology in induced pluripotent stem cell-derived neurons from patients with MAPT mutations. Brain 2015, 138, 3345–3359. [Google Scholar] [CrossRef]
- Pomeshchik, Y.; Klementieva, O.; Gil, J.; Martinsson, I.; Hansen, M.G.; de Vries, T.; Sancho-Balsells, A.; Russ, K.; Savchenko, E.; Collin, A.; et al. Human iPSC-Derived Hippocampal Spheroids: An Innovative Tool for Stratifying Alzheimer Disease Patient-Specific Cellular Phenotypes and Developing Therapies. Stem Cell Rep. 2020, 15, 256–273. [Google Scholar] [CrossRef]
- Jones, V.C.; Atkinson-Dell, R.; Verkhratsky, A.; Mohamet, L. Aberrant iPSC-derived human astrocytes in Alzheimer’s disease. Cell Death Dis. 2017, 8, e2696. [Google Scholar] [CrossRef]
- Oksanen, M.; Petersen, A.J.; Naumenko, N.; Puttonen, K.; Lehtonen, Š.; Gubert Olivé, M.; Shakirzyanova, A.; Leskelä, S.; Sarajärvi, T.; Viitanen, M.; et al. PSEN1 Mutant iPSC-Derived Model Reveals Severe Astrocyte Pathology in Alzheimer’s Disease. Stem Cell Rep. 2017, 9, 1885–1897. [Google Scholar] [CrossRef] [PubMed]
- Konttinen, H.; Cabral-da-Silva, M.E.C.; Ohtonen, S.; Wojciechowski, S.; Shakirzyanova, A.; Caligola, S.; Giugno, R.; Ishchenko, Y.; Hernández, D.; Fazaludeen, M.F.; et al. PSEN1ΔE9, APPswe, and APOE4 Confer Disparate Phenotypes in Human iPSC-Derived Microglia. Stem Cell Rep. 2019, 13, 669–683. [Google Scholar] [CrossRef] [PubMed]
- Choi, S.H.; Kim, Y.H.; Hebisch, M.; Sliwinski, C.; Lee, S.; D’Avanzo, C.; Chen, H.; Hooli, B.; Asselin, C.; Muffat, J.; et al. A three-dimensional human neural cell culture model of Alzheimer’s disease. Nature 2014, 515, 274–278. [Google Scholar] [CrossRef] [PubMed]
- Egawa, N.; Kitaoka, S.; Tsukita, K.; Naitoh, M.; Takahashi, K.; Yamamoto, T.; Adachi, F.; Kondo, T.; Okita, K.; Asaka, I.; et al. Drug screening for ALS using patient-specific induced pluripotent stem cells. Sci. Transl. Med. 2012, 4, 145ra104. [Google Scholar] [CrossRef] [PubMed]
- Burkhardt, M.F.; Martinez, F.J.; Wright, S.; Ramos, C.; Volfson, D.; Mason, M.; Garnes, J.; Dang, V.; Lievers, J.; Shoukat-Mumtaz, U.; et al. A cellular model for sporadic ALS using patient-derived induced pluripotent stem cells. Mol. Cell Neurosci. 2013, 56, 355–364. [Google Scholar] [CrossRef] [PubMed]
- Sareen, D.; O’Rourke, J.G.; Meera, P.; Muhammad, A.K.; Grant, S.; Simpkinson, M.; Bell, S.; Carmona, S.; Ornelas, L.; Sahabian, A.; et al. Targeting RNA foci in iPSC-derived motor neurons from ALS patients with a C9ORF72 repeat expansion. Sci. Transl. Med. 2013, 5, 208ra149. [Google Scholar] [CrossRef] [PubMed]
- Kiskinis, E.; Sandoe, J.; Williams, L.A.; Boulting, G.L.; Moccia, R.; Wainger, B.J.; Han, S.; Peng, T.; Thams, S.; Mikkilineni, S.; et al. Pathways disrupted in human ALS motor neurons identified through genetic correction of mutant SOD1. Cell Stem Cell 2014, 14, 781–795. [Google Scholar] [CrossRef]
- Kiskinis, E.; Kralj, J.M.; Zou, P.; Weinstein, E.N.; Zhang, H.; Tsioras, K.; Wiskow, O.; Ortega, J.A.; Eggan, K.; Cohen, A.E. All-Optical Electrophysiology for High-Throughput Functional Characterization of a Human iPSC-Derived Motor Neuron Model of ALS. Stem Cell Rep. 2018, 10, 1991–2004. [Google Scholar] [CrossRef]
- Madill, M.; McDonagh, K.; Ma, J.; Vajda, A.; McLoughlin, P.; O’Brien, T.; Hardiman, O.; Shen, S. Amyotrophic lateral sclerosis patient iPSC-derived astrocytes impair autophagy via non-cell autonomous mechanisms. Mol. Brain 2017, 10, 22. [Google Scholar] [CrossRef]
- Ferraiuolo, L.; Meyer, K.; Sherwood, T.W.; Vick, J.; Likhite, S.; Frakes, A.; Miranda, C.J.; Braun, L.; Heath, P.R.; Pineda, R.; et al. Oligodendrocytes contribute to motor neuron death in ALS via SOD1-dependent mechanism. Proc. Natl. Acad. Sci. USA 2016, 113, E6496–E6505. [Google Scholar] [CrossRef]
- García-León, J.A.; Kumar, M.; Boon, R.; Chau, D.; One, J.; Wolfs, E.; Eggermont, K.; Berckmans, P.; Gunhanlar, N.; de Vrij, F.; et al. SOX10 Single Transcription Factor-Based Fast and Efficient Generation of Oligodendrocytes from Human Pluripotent Stem Cells. Stem Cell Rep. 2018, 10, 655–672. [Google Scholar] [CrossRef] [PubMed]
- Najm, F.J.; Madhavan, M.; Zaremba, A.; Shick, E.; Karl, R.T.; Factor, D.C.; Miller, T.E.; Nevin, Z.S.; Kantor, C.; Sargent, A.; et al. Drug-based modulation of endogenous stem cells promotes functional remyelination in vivo. Nature 2015, 522, 216–220. [Google Scholar] [CrossRef] [PubMed]
- Daviaud, N.; Chen, E.; Edwards, T.; Sadiq, S.A. Cerebral organoids in primary progressive multiple sclerosis reveal stem cell and oligodendrocyte differentiation defect. Biol. Open 2023, 12, bio.059845. [Google Scholar] [CrossRef] [PubMed]
- Consortium, H.i. Induced pluripotent stem cells from patients with Huntington’s disease show CAG-repeat-expansion-associated phenotypes. Cell Stem Cell 2012, 11, 264–278. [Google Scholar] [CrossRef]
- Xu, X.; Tay, Y.; Sim, B.; Yoon, S.I.; Huang, Y.; Ooi, J.; Utami, K.H.; Ziaei, A.; Ng, B.; Radulescu, C.; et al. Reversal of Phenotypic Abnormalities by CRISPR/Cas9-Mediated Gene Correction in Huntington Disease Patient-Derived Induced Pluripotent Stem Cells. Stem Cell Rep. 2017, 8, 619–633. [Google Scholar] [CrossRef] [PubMed]
- Nekrasov, E.D.; Vigont, V.A.; Klyushnikov, S.A.; Lebedeva, O.S.; Vassina, E.M.; Bogomazova, A.N.; Chestkov, I.V.; Semashko, T.A.; Kiseleva, E.; Suldina, L.A.; et al. Manifestation of Huntington’s disease pathology in human induced pluripotent stem cell-derived neurons. Mol. Neurodegener. 2016, 11, 27. [Google Scholar] [CrossRef] [PubMed]
- Linville, R.M.; Nerenberg, R.F.; Grifno, G.; Arevalo, D.; Guo, Z.; Searson, P.C. Brain microvascular endothelial cell dysfunction in an isogenic juvenile iPSC model of Huntington’s disease. Fluids Barriers CNS 2022, 19, 54. [Google Scholar] [CrossRef] [PubMed]
- Tropepe, V.; Hitoshi, S.; Sirard, C.; Mak, T.W.; Rossant, J.; van der Kooy, D. Direct neural fate specification from embryonic stem cells: A primitive mammalian neural stem cell stage acquired through a default mechanism. Neuron 2001, 30, 65–78. [Google Scholar] [CrossRef]
- Shi, Y.; Kirwan, P.; Smith, J.; Robinson, H.P.; Livesey, F.J. Human cerebral cortex development from pluripotent stem cells to functional excitatory synapses. Nat. Neurosci. 2012, 15, 477–486. [Google Scholar] [CrossRef]
- Gunhanlar, N.; Shpak, G.; van der Kroeg, M.; Gouty-Colomer, L.A.; Munshi, S.T.; Lendemeijer, B.; Ghazvini, M.; Dupont, C.; Hoogendijk, W.J.G.; Gribnau, J.; et al. A simplified protocol for differentiation of electrophysiologically mature neuronal networks from human induced pluripotent stem cells. Mol. Psychiatry 2018, 23, 1336–1344. [Google Scholar] [CrossRef]
- Bardy, C.; van den Hurk, M.; Eames, T.; Marchand, C.; Hernandez, R.V.; Kellogg, M.; Gorris, M.; Galet, B.; Palomares, V.; Brown, J.; et al. Neuronal medium that supports basic synaptic functions and activity of human neurons in vitro. Proc. Natl. Acad. Sci. USA 2015, 112, E2725–E2734. [Google Scholar] [CrossRef] [PubMed]
- Dimos, J.T.; Rodolfa, K.T.; Niakan, K.K.; Weisenthal, L.M.; Mitsumoto, H.; Chung, W.; Croft, G.F.; Saphier, G.; Leibel, R.; Goland, R.; et al. Induced pluripotent stem cells generated from patients with ALS can be differentiated into motor neurons. Science 2008, 321, 1218–1221. [Google Scholar] [CrossRef] [PubMed]
- Jovanovic, V.M.; Weber, C.; Slamecka, J.; Ryu, S.; Chu, P.H.; Sen, C.; Inman, J.; De Sousa, J.F.; Barnaeva, E.; Hirst, M.; et al. A defined roadmap of radial glia and astrocyte differentiation from human pluripotent stem cells. Stem Cell Rep. 2023, 18, 1701–1720. [Google Scholar] [CrossRef] [PubMed]
- Krencik, R.; Weick, J.P.; Liu, Y.; Zhang, Z.J.; Zhang, S.C. Specification of transplantable astroglial subtypes from human pluripotent stem cells. Nat. Biotechnol. 2011, 29, 528–534. [Google Scholar] [CrossRef]
- Velasco, S.; Kedaigle, A.J.; Simmons, S.K.; Nash, A.; Rocha, M.; Quadrato, G.; Paulsen, B.; Nguyen, L.; Adiconis, X.; Regev, A.; et al. Individual brain organoids reproducibly form cell diversity of the human cerebral cortex. Nature 2019, 570, 523–527. [Google Scholar] [CrossRef]
- Nguyen, H.N.; Byers, B.; Cord, B.; Shcheglovitov, A.; Byrne, J.; Gujar, P.; Kee, K.; Schüle, B.; Dolmetsch, R.E.; Langston, W.; et al. LRRK2 mutant iPSC-derived DA neurons demonstrate increased susceptibility to oxidative stress. Cell Stem Cell 2011, 8, 267–280. [Google Scholar] [CrossRef]
- Kikuchi, T.; Morizane, A.; Doi, D.; Magotani, H.; Onoe, H.; Hayashi, T.; Mizuma, H.; Takara, S.; Takahashi, R.; Inoue, H.; et al. Human iPS cell-derived dopaminergic neurons function in a primate Parkinson’s disease model. Nature 2017, 548, 592–596. [Google Scholar] [CrossRef]
- Tao, Y.; Zhang, S.C. Neural Subtype Specification from Human Pluripotent Stem Cells. Cell Stem Cell 2016, 19, 573–586. [Google Scholar] [CrossRef]
- Tcw, J.; Wang, M.; Pimenova, A.A.; Bowles, K.R.; Hartley, B.J.; Lacin, E.; Machlovi, S.I.; Abdelaal, R.; Karch, C.M.; Phatnani, H.; et al. An Efficient Platform for Astrocyte Differentiation from Human Induced Pluripotent Stem Cells. Stem Cell Rep. 2017, 9, 600–614. [Google Scholar] [CrossRef]
- Garcia-Leon, J.A.; Vitorica, J.; Gutierrez, A. Use of human pluripotent stem cell-derived cells for neurodegenerative disease modeling and drug screening platform. Future Med. Chem. 2019, 11, 1305–1322. [Google Scholar] [CrossRef]
- García-León, J.A.; Verfaillie, C.M. Stem Cell-Derived Oligodendroglial Cells for Therapy in Neurological Diseases. Curr. Stem Cell Res. Ther. 2016, 11, 569–577. [Google Scholar] [CrossRef]
- Ogawa, S.; Tokumoto, Y.; Miyake, J.; Nagamune, T. Induction of oligodendrocyte differentiation from adult human fibroblast-derived induced pluripotent stem cells. In Vitro Cell Dev. Biol. Anim. 2011, 47, 464–469. [Google Scholar] [CrossRef] [PubMed]
- Pouya, A.; Satarian, L.; Kiani, S.; Javan, M.; Baharvand, H. Human induced pluripotent stem cells differentiation into oligodendrocyte progenitors and transplantation in a rat model of optic chiasm demyelination. PLoS ONE 2011, 6, e27925. [Google Scholar] [CrossRef] [PubMed]
- Wang, S.; Bates, J.; Li, X.; Schanz, S.; Chandler-Militello, D.; Levine, C.; Maherali, N.; Studer, L.; Hochedlinger, K.; Windrem, M.; et al. Human iPSC-derived oligodendrocyte progenitor cells can myelinate and rescue a mouse model of congenital hypomyelination. Cell Stem Cell 2013, 12, 252–264. [Google Scholar] [CrossRef] [PubMed]
- Douvaras, P.; Wang, J.; Zimmer, M.; Hanchuk, S.; O’Bara, M.A.; Sadiq, S.; Sim, F.J.; Goldman, J.; Fossati, V. Efficient generation of myelinating oligodendrocytes from primary progressive multiple sclerosis patients by induced pluripotent stem cells. Stem Cell Rep. 2014, 3, 250–259. [Google Scholar] [CrossRef] [PubMed]
- Piao, J.; Major, T.; Auyeung, G.; Policarpio, E.; Menon, J.; Droms, L.; Gutin, P.; Uryu, K.; Tchieu, J.; Soulet, D.; et al. Human embryonic stem cell-derived oligodendrocyte progenitors remyelinate the brain and rescue behavioral deficits following radiation. Cell Stem Cell 2015, 16, 198–210. [Google Scholar] [CrossRef] [PubMed]
- Pandya, H.; Shen, M.J.; Ichikawa, D.M.; Sedlock, A.B.; Choi, Y.; Johnson, K.R.; Kim, G.; Brown, M.A.; Elkahloun, A.G.; Maric, D.; et al. Differentiation of human and murine induced pluripotent stem cells to microglia-like cells. Nat. Neurosci. 2017, 20, 753–759. [Google Scholar] [CrossRef] [PubMed]
- Abud, E.M.; Ramirez, R.N.; Martinez, E.S.; Healy, L.M.; Nguyen, C.H.H.; Newman, S.A.; Yeromin, A.V.; Scarfone, V.M.; Marsh, S.E.; Fimbres, C.; et al. iPSC-Derived Human Microglia-like Cells to Study Neurological Diseases. Neuron 2017, 94, 278–293.e279. [Google Scholar] [CrossRef]
- Hansen, D.V.; Hanson, J.E.; Sheng, M. Microglia in Alzheimer’s disease. J. Cell Biol. 2018, 217, 459–472. [Google Scholar] [CrossRef]
- Lancaster, M.A.; Renner, M.; Martin, C.A.; Wenzel, D.; Bicknell, L.S.; Hurles, M.E.; Homfray, T.; Penninger, J.M.; Jackson, A.P.; Knoblich, J.A. Cerebral organoids model human brain development and microcephaly. Nature 2013, 501, 373–379. [Google Scholar] [CrossRef]
- Lee, C.T.; Bendriem, R.M.; Wu, W.W.; Shen, R.F. 3D brain Organoids derived from pluripotent stem cells: Promising experimental models for brain development and neurodegenerative disorders. J. Biomed. Sci. 2017, 24, 59. [Google Scholar] [CrossRef] [PubMed]
- Tomaskovic-Crook, E.; Crook, J.M. Clinically Amendable, Defined, and Rapid Induction of Human Brain Organoids from Induced Pluripotent Stem Cells. Methods Mol. Biol. 2019, 1576, 13–22. [Google Scholar] [CrossRef] [PubMed]
- Huang, W.K.; Wong, S.Z.H.; Pather, S.R.; Nguyen, P.T.T.; Zhang, F.; Zhang, D.Y.; Zhang, Z.; Lu, L.; Fang, W.; Chen, L.; et al. Generation of hypothalamic arcuate organoids from human induced pluripotent stem cells. Cell Stem Cell 2021, 28, 1657–1670.e1610. [Google Scholar] [CrossRef] [PubMed]
- Marion, R.M.; Strati, K.; Li, H.; Tejera, A.; Schoeftner, S.; Ortega, S.; Serrano, M.; Blasco, M.A. Telomeres acquire embryonic stem cell characteristics in induced pluripotent stem cells. Cell Stem Cell 2009, 4, 141–154. [Google Scholar] [CrossRef] [PubMed]
- Suhr, S.T.; Chang, E.A.; Tjong, J.; Alcasid, N.; Perkins, G.A.; Goissis, M.D.; Ellisman, M.H.; Perez, G.I.; Cibelli, J.B. Mitochondrial rejuvenation after induced pluripotency. PLoS ONE 2010, 5, e14095. [Google Scholar] [CrossRef] [PubMed]
- Grenier, K.; Kao, J.; Diamandis, P. Three-dimensional modeling of human neurodegeneration: Brain organoids coming of age. Mol. Psychiatry 2020, 25, 254–274. [Google Scholar] [CrossRef]
- Miller, J.D.; Ganat, Y.M.; Kishinevsky, S.; Bowman, R.L.; Liu, B.; Tu, E.Y.; Mandal, P.K.; Vera, E.; Shim, J.W.; Kriks, S.; et al. Human iPSC-based modeling of late-onset disease via progerin-induced aging. Cell Stem Cell 2013, 13, 691–705. [Google Scholar] [CrossRef]
- Tang, Y.; Liu, M.L.; Zang, T.; Zhang, C.L. Direct Reprogramming Rather than iPSC-Based Reprogramming Maintains Aging Hallmarks in Human Motor Neurons. Front. Mol. Neurosci. 2017, 10, 359. [Google Scholar] [CrossRef]
- Garcia, T.Y.; Gutierrez, M.; Reynolds, J.; Lamba, D.A. Modeling the Dynamic AMD-Associated Chronic Oxidative Stress Changes in Human ESC and iPSC-Derived RPE Cells. Invest. Ophthalmol. Vis. Sci. 2015, 56, 7480–7488. [Google Scholar] [CrossRef]
- Lee, G.; Ramirez, C.N.; Kim, H.; Zeltner, N.; Liu, B.; Radu, C.; Bhinder, B.; Kim, Y.J.; Choi, I.Y.; Mukherjee-Clavin, B.; et al. Large-scale screening using familial dysautonomia induced pluripotent stem cells identifies compounds that rescue IKBKAP expression. Nat. Biotechnol. 2012, 30, 1244–1248. [Google Scholar] [CrossRef]
- Naryshkin, N.A.; Weetall, M.; Dakka, A.; Narasimhan, J.; Zhao, X.; Feng, Z.; Ling, K.K.; Karp, G.M.; Qi, H.; Woll, M.G.; et al. Motor neuron disease. SMN2 splicing modifiers improve motor function and longevity in mice with spinal muscular atrophy. Science 2014, 345, 688–693. [Google Scholar] [CrossRef] [PubMed]
- Wainger, B.J.; Kiskinis, E.; Mellin, C.; Wiskow, O.; Han, S.S.; Sandoe, J.; Perez, N.P.; Williams, L.A.; Lee, S.; Boulting, G.; et al. Intrinsic membrane hyperexcitability of amyotrophic lateral sclerosis patient-derived motor neurons. Cell Rep. 2014, 7, 1–11. [Google Scholar] [CrossRef] [PubMed]
- McNeish, J.; Gardner, J.P.; Wainger, B.J.; Woolf, C.J.; Eggan, K. From Dish to Bedside: Lessons Learned While Translating Findings from a Stem Cell Model of Disease to a Clinical Trial. Cell Stem Cell 2015, 17, 8–10. [Google Scholar] [CrossRef] [PubMed]
- Poewe, W.; Seppi, K.; Tanner, C.M.; Halliday, G.M.; Brundin, P.; Volkmann, J.; Schrag, A.E.; Lang, A.E. Parkinson disease. Nat. Rev. Dis. Primers 2017, 3, 17013. [Google Scholar] [CrossRef] [PubMed]
- Sethi, K.D. The impact of levodopa on quality of life in patients with Parkinson disease. Neurologist 2010, 16, 76–83. [Google Scholar] [CrossRef] [PubMed]
- Brundin, P.; Strecker, R.; Clarke, D.; Widner, H.; Nilsson, O.; Åstedt, B.; Lindvall, O.; Björklund, A. Chapter 57 Can human fetal dopamine neuron grafts provide a therapy for Parkinson’s disease? In Progress in Brain Research; Elsevier: Amsterdam, The Netherlands, 1988; Volume 78, pp. 441–448. [Google Scholar]
- Barker, R.A.; Parmar, M.; Kirkeby, A.; Björklund, A.; Thompson, L.; Brundin, P. Are Stem Cell-Based Therapies for Parkinson’s Disease Ready for the Clinic in 2016? J. Park. Dis. 2016, 6, 57–63. [Google Scholar] [CrossRef]
- Yasuhara, T.; Kameda, M.; Sasaki, T.; Tajiri, N.; Date, I. Cell Therapy for Parkinson’s Disease. Cell Transplant. 2017, 26, 1551–1559. [Google Scholar] [CrossRef]
- Stoddard-Bennett, T.; Reijo Pera, R. Treatment of Parkinson’s Disease through Personalized Medicine and Induced Pluripotent Stem Cells. Cells 2019, 8, 26. [Google Scholar] [CrossRef]
- Polanco, J.C.; Li, C.; Bodea, L.G.; Martinez-Marmol, R.; Meunier, F.A.; Götz, J. Amyloid-β and tau complexity-towards improved biomarkers and targeted therapies. Nat. Rev. Neurol. 2018, 14, 22–39. [Google Scholar] [CrossRef]
- Gutierrez, A.; Vitorica, J. Toward a New Concept of Alzheimer’s Disease Models: A Perspective from Neuroinflammation. J. Alzheimers Dis. 2018, 64, S329–S338. [Google Scholar] [CrossRef]
- Olabarria, M.; Noristani, H.N.; Verkhratsky, A.; Rodríguez, J.J. Concomitant astroglial atrophy and astrogliosis in a triple transgenic animal model of Alzheimer’s disease. Glia 2010, 58, 831–838. [Google Scholar] [CrossRef] [PubMed]
- Taylor, J.P.; Brown, R.H.; Cleveland, D.W. Decoding ALS: From genes to mechanism. Nature 2016, 539, 197–206. [Google Scholar] [CrossRef] [PubMed]
- Chua, J.P.; De Calbiac, H.; Kabashi, E.; Barmada, S.J. Autophagy and ALS: Mechanistic insights and therapeutic implications. Autophagy 2022, 18, 254–282. [Google Scholar] [CrossRef] [PubMed]
- Yamout, B.I.; Alroughani, R. Multiple Sclerosis. Semin. Neurol. 2018, 38, 212–225. [Google Scholar] [CrossRef] [PubMed]
- Stoker, T.B.; Mason, S.L.; Greenland, J.C.; Holden, S.T.; Santini, H.; Barker, R.A. Huntington’s disease: Diagnosis and management. Pract. Neurol. 2022, 22, 32–41. [Google Scholar] [CrossRef] [PubMed]
- Zuccato, C.; Valenza, M.; Cattaneo, E. Molecular mechanisms and potential therapeutical targets in Huntington’s disease. Physiol. Rev. 2010, 90, 905–981. [Google Scholar] [CrossRef] [PubMed]
- Antonov, S.A.; Novosadova, E.V. Current State-of-the-Art and Unresolved Problems in Using Human Induced Pluripotent Stem Cell-Derived Dopamine Neurons for Parkinson’s Disease Drug Development. Int. J. Mol. Sci. 2021, 22, 3381. [Google Scholar] [CrossRef] [PubMed]
- Costamagna, G.; Comi, G.P.; Corti, S. Advancing Drug Discovery for Neurological Disorders Using iPSC-Derived Neural Organoids. Int. J. Mol. Sci. 2021, 22, 2659. [Google Scholar] [CrossRef]
- Balusu, S.; Praschberger, R.; Lauwers, E.; De Strooper, B.; Verstreken, P. Neurodegeneration cell per cell. Neuron 2023, 111, 767–786. [Google Scholar] [CrossRef]
- Baxi, E.G.; Thompson, T.; Li, J.; Kaye, J.A.; Lim, R.G.; Wu, J.; Ramamoorthy, D.; Lima, L.; Vaibhav, V.; Matlock, A.; et al. Answer ALS, a large-scale resource for sporadic and familial ALS combining clinical and multi-omics data from induced pluripotent cell lines. Nat. Neurosci. 2022, 25, 226–237. [Google Scholar] [CrossRef]
- Chang, C.Y.; Ting, H.C.; Liu, C.A.; Su, H.L.; Chiou, T.W.; Lin, S.Z.; Harn, H.J.; Ho, T.J. Induced Pluripotent Stem Cell (iPSC)-Based Neurodegenerative Disease Models for Phenotype Recapitulation and Drug Screening. Molecules 2020, 25, 2000. [Google Scholar] [CrossRef] [PubMed]
Disease | Gene Mutation | Phenotype | Cell Type | Potential Compound | References |
---|---|---|---|---|---|
PD | Triplication of SNCA | Increased α-synuclein | Dopaminergic neurons | [31] | |
PD | Heterozygous glucocerebrosidase mutation (GBA N370S) | ∼50% glucocerebrosidase enzymatic activity, ∼3-fold elevated α-synuclein protein levels, and a reduced capacity to synthesize and release dopamine | Midbrain dopaminergic neurons | Monoamine oxidase B inhibitors | [32] |
PD | Idiopathic Parkinson’s disease | Midbrain dopaminergic progenitor cells | [33] | ||
AD | Presenilin 1 and presenilin 2 | Increased amyloid β42 secretion | Neurons | γ-secretase inhibitors and modulators | [34] |
AD | Amyloid precursor protein mutation (V717I) | Increased generation of both Aβ42 and Aβ38 and increase in levels of total and phosphorylated tau | Neurons | [35] | |
AD | G384A mutation of the presenilin 1 gene, which encodes presenilin-1 | Increased the production of Aβ42 as a toxic Aβ species, and the Aβ42/40 ratio | Cortical neurons | Three candidates were combined to improve the anti-Aβ effect (bromocriptine, cromolyn, and topiramate) as an anti-Aβ cocktail | [36] |
AD | Aβ1–42 aggregates | Neurons | Several small molecules as effective blockers against Aβ1-42 toxicity, including a Cdk2 inhibitor | [37] | |
AD | Duplication of the amyloid-β precursor protein-encoding gene | Significantly higher levels of the pathological markers amyloid-β, phospho-tau (Thr 231) and active glycogen synthase kinase-3β (aGSK-3β) | Neurons | β-secretase inhibitors | [38] |
AD | MAPT gene | Abnormal tau expression, hyperphosphorylation of tau aggregates, and multiple disease phenotypes | Neurons | [39] | |
AD | Variations in amyloid precursor protein or presenilin 1 genes | Loss of synaptic proteins, increased ratio of intracellular and extracellular Aβ42/Aβ40 peptides, differences in protein aggregation, tau phosphorylation, miRNA pattern, and protein network alterations | Hippocampus neurons | [40] | |
AD | Mislocalization and abnormal expression of mature astrocyte markers, compromised astrocyte heterogeneity and astroglial atrophy. | Neurons and astrocytes | [41] | ||
AD | Presenilin 1 ΔE9 mutation | Increased β-amyloid production, altered cytokine release, and dysregulated Ca2+ homeostasis. | Astrocytes | [42] | |
AD | APOE4 genotype | Impaired phagocytosis, migration, and metabolic activity but exacerbated cytokine secretion | Microglia-like cells | [43] | |
AD | FAD mutations in β-amyloid precursor protein and presenilin 1 |
Robust extracellular deposition of amyloid-β, including amyloid-β plaques. Silver-positive aggregates of phosphorylated tau in the soma and neurites, as well as filamentous tau. | 3D-differentiated neuronal cells | [44] | |
AD | Amyloid precursor protein duplication or presenilin 1 mutation | Amyloid aggregation, hyperphosphorylated tau protein, and endosome abnormalities | Brain organoids | β- and γ-secretase inhibitors | [29] |
ALS | TAR DNA binding protein-43 (TDP-43) | Cytosolic aggregates similar to those seen in postmortem tissue from ALS patients and exhibited shorter neurites as seen in a zebrafish model of ALS. | Motor neurons | Histone acetyltransferase inhibitor, named anacardic acid | [45] |
ALS | TDP-43 aggregation | Motor neurons | FDA-approved small molecule modulators including digoxin | [46] | |
ALS | Mutation in the C9ORF72 gene | Altered expression of genes involved in membrane excitability, including DPP6, demonstrating a diminished capacity to fire continuous spikes upon depolarization compared to control motor neurons | Motor neurons | Antisense oligonucleotides targeting the C9ORF72 transcript suppressed RNA foci formation and reversed gene expression alterations in motor neurons | [47] |
ALS | Mutant SOD1 | Increased oxidative stress, reduced mitochondrial function, altered subcellular transport, and activation of the ER stress and unfolded protein response pathways | Motor neurons | [48] | |
ALS | Mutation SOD1 A4V | Elevated spike rates under weak or no stimulus and greater likelihood of entering depolarization block under strong optogenetic stimulus | Motor neurons | [49] | |
ALS |
Increase in the expression of SOD1, a protein associated with the development of ALS. Astrocytes modulate the autophagy pathway in a noncell autonomous manner. | Astrocytes | [50] | ||
ALS | Oligodendrocytes were mutated for SOD1 | Death of motor neurons induced by oligodendrocytes | Oligodendrocytes | [51] | |
MS | Overexpression of the transcription factor SOX10 | Using in vitro oligodendrocytes-neuron cocultures, myelination of neurons by oligodendrocytes were demonstrated | Oligodendrocytes | [52] | |
MS | Principal source of myelinating oligodendrocytes | Oligodendrocyte progenitor cells | Miconazole and clobetasol | [53] | |
MS | Decrease of proliferation marker Ki67 and a reduction of the SOX2+ stem cell pool associated with increased expression of neuronal markers CTIP2 and TBR1 as well as a strong decrease of oligodendrocyte differentiation | Brain organoids | [54] | ||
HD | The microarray profile distinguished the lines of cells from healthy controls and patients as the gene expression profile showed CAG repeat expansion | Neural cells showed disease-associated differences in electrophysiology, metabolism, cell death, and longer CAG repeat expansions | Neural cells | [55] | |
HD | CAG repeat expansion in huntingtin gene. Genetic correction using CRISPR-Cas9 | Impaired neural rosette formation, increased susceptibility to growth factor withdrawal, and deficits in mitochondrial respiration | Forebrain neurons | [56] | |
HD | Expansion of the CAG repeat in exon 1 of the huntingtin gene | Mutant huntingtin protein aggregation, increased number of lysosomes/autophagosomes, nuclear indentations, and enhanced neuronal death during cell ageing | GABAergic medium spiny neurons | EVP4593 drug, a quinazoline derivative | [57] |
HD | CAG repeat expansion in huntingtin gene | Subtle changes in phenotype, including differences in cell turnover and immune cell adhesion | Brain microvascular endothelial-like cells | [58] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Beghini, D.G.; Kasai-Brunswick, T.H.; Henriques-Pons, A. Induced Pluripotent Stem Cells in Drug Discovery and Neurodegenerative Disease Modelling. Int. J. Mol. Sci. 2024, 25, 2392. https://doi.org/10.3390/ijms25042392
Beghini DG, Kasai-Brunswick TH, Henriques-Pons A. Induced Pluripotent Stem Cells in Drug Discovery and Neurodegenerative Disease Modelling. International Journal of Molecular Sciences. 2024; 25(4):2392. https://doi.org/10.3390/ijms25042392
Chicago/Turabian StyleBeghini, Daniela Gois, Tais Hanae Kasai-Brunswick, and Andrea Henriques-Pons. 2024. "Induced Pluripotent Stem Cells in Drug Discovery and Neurodegenerative Disease Modelling" International Journal of Molecular Sciences 25, no. 4: 2392. https://doi.org/10.3390/ijms25042392
APA StyleBeghini, D. G., Kasai-Brunswick, T. H., & Henriques-Pons, A. (2024). Induced Pluripotent Stem Cells in Drug Discovery and Neurodegenerative Disease Modelling. International Journal of Molecular Sciences, 25(4), 2392. https://doi.org/10.3390/ijms25042392