iPSCs in Neurodegenerative Disorders: A Unique Platform for Clinical Research and Personalized Medicine
Abstract
:1. Introduction
2. Brief History of iPSCs
3. iPSCs in Scientific Research
4. Role of iPSCs in Neurodegenerative Diseases
4.1. iPSCs in PD
4.2. iPSCs in AD
4.3. iPSCs in Diabetic Neuropathy
4.4. iPSCs in Stroke
4.5. iPSCs in SCI
5. Role of iPSCs in Personalized Medicine
5.1. Are Clinical-Grade Allogeneic iPSCs Important?
5.2. iPSCs in Personalized Pharmacology
6. Limitations and Challenges
Supplementary Materials
Author Contributions
Funding
Conflicts of Interest
References
- Moradi, S.; Mahdizadeh, H.; Šarić, T.; Kim, J.; Harati, J.; Shahsavarani, H.; Greber, B.; Moore, J.B. Research and therapy with induced pluripotent stem cells (iPSCs): Social, legal, and ethical considerations. Stem Cell Res. Ther. 2019, 10, 341. [Google Scholar] [CrossRef] [PubMed]
- Lin, X.; Tang, J.; Lou, Y.R. Human Pluripotent Stem-Cell-Derived Models as a Missing Link in Drug Discovery and Development. Pharmaceuticals 2021, 14, 525. [Google Scholar] [CrossRef] [PubMed]
- Omole, A.E.; Fakoya, A.O.J. Ten years of progress and promise of induced pluripotent stem cells: Historical origins, characteristics, mechanisms, limitations, and potential applications. PeerJ 2018, 6, e4370. [Google Scholar] [CrossRef] [PubMed]
- Cummings, J.L.; Goldman, D.P.; Simmons-Stern, N.R.; Ponton, E. The costs of developing treatments for Alzheimer’s disease: A retrospective exploration. Alzheimers Dement. 2021, 18, 469–477. [Google Scholar] [CrossRef] [PubMed]
- Till, J.E.; Mc, C.E. A direct measurement of the radiation sensitivity of normal mouse bone marrow cells. Radiat. Res. 1961, 14, 213–222. [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]
- 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]
- Evans, M.J.; Kaufman, M.H. Establishment in culture of pluripotential cells from mouse embryos. Nature 1981, 292, 154–156. [Google Scholar] [CrossRef]
- Tada, M.; Takahama, Y.; Abe, K.; Nakatsuji, N.; Tada, T. Nuclear reprogramming of somatic cells by in vitro hybridization with ES cells. Curr. Biol. 2001, 11, 1553–1558. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Takahashi, K.; Tanabe, K.; Ohnuki, M.; Narita, M.; Ichisaka, T.; Tomoda, K.; Yamanaka, S. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 2007, 131, 861–872. [Google Scholar] [CrossRef] [PubMed]
- Yu, J.; Vodyanik, M.A.; Smuga-Otto, K.; Antosiewicz-Bourget, J.; Frane, J.L.; Tian, S.; Nie, J.; Jonsdottir, G.A.; Ruotti, V.; Stewart, R.; et al. Induced pluripotent stem cell lines derived from human somatic cells. Science 2007, 318, 1917–1920. [Google Scholar] [CrossRef] [PubMed]
- Nakagawa, M.; Koyanagi, M.; Tanabe, K.; Takahashi, K.; Ichisaka, T.; Aoi, T.; Okita, K.; Mochiduki, Y.; Takizawa, N.; Yamanaka, S. Generation of induced pluripotent stem cells without Myc from mouse and human fibroblasts. Nat. Biotechnol. 2008, 26, 101–106. [Google Scholar] [CrossRef]
- Arjmand, B.; Goodarzi, P.; Mohamadi-Jahani, F.; Falahzadeh, K.; Larijani, B. Personalized Regenerative Medicine. Acta Med. Iran. 2017, 55, 144–149. [Google Scholar]
- Chang, E.A.; Jin, S.W.; Nam, M.H.; Kim, S.D. Human Induced Pluripotent Stem Cells: Clinical Significance and Applications in Neurologic Diseases. J. Korean Neurosurg. Soc. 2019, 62, 493–501. [Google Scholar] [CrossRef]
- Shi, Y.; Desponts, C.; Do, J.T.; Hahm, H.S.; Schöler, H.R.; Ding, S. Induction of pluripotent stem cells from mouse embryonic fibroblasts by Oct4 and Klf4 with small-molecule compounds. Cell Stem Cell 2008, 3, 568–574. [Google Scholar] [CrossRef]
- Huangfu, D.; Maehr, R.; Guo, W.; Eijkelenboom, A.; Snitow, M.; Chen, A.E.; Melton, D.A. Induction of pluripotent stem cells by defined factors is greatly improved by small-molecule compounds. Nat. Biotechnol. 2008, 26, 795–797. [Google Scholar] [CrossRef]
- Huangfu, D.; Osafune, K.; Maehr, R.; Guo, W.; Eijkelenboom, A.; Chen, S.; Muhlestein, W.; Melton, D.A. Induction of pluripotent stem cells from primary human fibroblasts with only Oct4 and Sox2. Nat. Biotechnol. 2008, 26, 1269–1275. [Google Scholar] [CrossRef]
- Wakayama, T.; Perry, A.C.; Zuccotti, M.; Johnson, K.R.; Yanagimachi, R. Full-term development of mice from enucleated oocytes injected with cumulus cell nuclei. Nature 1998, 394, 369–374. [Google Scholar] [CrossRef]
- Kato, Y.; Tani, T.; Sotomaru, Y.; Kurokawa, K.; Kato, J.; Doguchi, H.; Yasue, H.; Tsunoda, Y. Eight calves cloned from somatic cells of a single adult. Science 1998, 282, 2095–2098. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Polejaeva, I.A.; Chen, S.H.; Vaught, T.D.; Page, R.L.; Mullins, J.; Ball, S.; Dai, Y.; Boone, J.; Walker, S.; Ayares, D.L.; et al. Cloned pigs produced by nuclear transfer from adult somatic cells. Nature 2000, 407, 86–90. [Google Scholar] [CrossRef] [PubMed]
- Shin, T.; Kraemer, D.; Pryor, J.; Liu, L.; Rugila, J.; Howe, L.; Buck, S.; Murphy, K.; Lyons, L.; Westhusin, M. A cat cloned by nuclear transplantation. Nature 2002, 415, 859. [Google Scholar] [CrossRef]
- Zhou, Q.; Renard, J.P.; Le Friec, G.; Brochard, V.; Beaujean, N.; Cherifi, Y.; Fraichard, A.; Cozzi, J. Generation of fertile cloned rats by regulating oocyte activation. Science 2003, 302, 1179. [Google Scholar] [CrossRef] [PubMed]
- Lee, B.C.; Kim, M.K.; Jang, G.; Oh, H.J.; Yuda, F.; Kim, H.J.; Hossein, M.S.; Kim, J.J.; Kang, S.K.; Schatten, G.; et al. Dogs cloned from adult somatic cells. Nature 2005, 436, 641. [Google Scholar] [CrossRef]
- Rodriguez-Osorio, N.; Urrego, R.; Cibelli, J.B.; Eilertsen, K.; Memili, E. Reprogramming mammalian somatic cells. Theriogenology 2012, 78, 1869–1886. [Google Scholar] [CrossRef]
- Pandey, S.; Chottova Dvorakova, M. Future Perspective of Diabetic Animal Models. Endocr. Metab. Immune Disord. Drug Targets 2019, 20, 25–38. [Google Scholar] [CrossRef]
- Pandey, S.; Malviya, G.; Chottova Dvorakova, M. Role of Peptides in Diagnostics. Int. J. Mol. Sci. 2021, 22, 8828. [Google Scholar] [CrossRef]
- Dawson, T.M.; Golde, T.E.; Lagier-Tourenne, C. Animal models of neurodegenerative diseases. Nat. Neurosci. 2018, 21, 1370–1379. [Google Scholar] [CrossRef]
- Jucker, M. The benefits and limitations of animal models for translational research in neurodegenerative diseases. Nat. Med. 2010, 16, 1210–1214. [Google Scholar] [CrossRef]
- Ribeiro, F.M.; Camargos, E.R.; de Souza, L.C.; Teixeira, A.L. Animal models of neurodegenerative diseases. Braz. J. Psychiatry 2013, 35 (Suppl. 2), S82–S91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cummings, J.L.; Morstorf, T.; Zhong, K. Alzheimer’s disease drug-development pipeline: Few candidates, frequent failures. Alzheimers Res. Ther. 2014, 6, 37. [Google Scholar] [CrossRef]
- Bonifati, V. Genetics of Parkinson’s disease--state of the art, 2013. Park. Relat. Disord. 2014, 20 (Suppl. 1), S23–S28. [Google Scholar] [CrossRef]
- Shi, C.H.; Mao, C.Y.; Zhang, S.Y.; Yang, J.; Song, B.; Wu, P.; Zuo, C.T.; Liu, Y.T.; Ji, Y.; Yang, Z.H.; et al. CHCHD2 gene mutations in familial and sporadic Parkinson’s disease. Neurobiol. Aging 2016, 38, 217.e9–217.e13. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Wang, Z.; Sun, H.; Mao, C.; Yang, J.; Liu, Y.; Liu, H.; Zhang, S.; Zhang, J.; Xu, Y.; et al. Generation of induced pluripotent stem cell line (ZZUi007-A) from a 52-year-old patient with a novel CHCHD2 gene mutation in Parkinson’s disease. Stem Cell Res. 2018, 32, 87–90. [Google Scholar] [CrossRef] [PubMed]
- Tabata, Y.; Imaizumi, Y.; Sugawara, M.; Andoh-Noda, T.; Banno, S.; Chai, M.; Sone, T.; Yamazaki, K.; Ito, M.; Tsukahara, K.; et al. T-type Calcium Channels Determine the Vulnerability of Dopaminergic Neurons to Mitochondrial Stress in Familial Parkinson Disease. Stem Cell Rep. 2018, 11, 1171–1184. [Google Scholar] [CrossRef]
- Imaizumi, Y.; Okada, Y.; Akamatsu, W.; Koike, M.; Kuzumaki, N.; Hayakawa, H.; Nihira, T.; Kobayashi, T.; Ohyama, M.; Sato, S.; et al. Mitochondrial dysfunction associated with increased oxidative stress and α-synuclein accumulation in PARK2 iPSC-derived neurons and postmortem brain tissue. Mol. Brain 2012, 5, 35. [Google Scholar] [CrossRef]
- Suda, Y.; Kuzumaki, N.; Sone, T.; Narita, M.; Tanaka, K.; Hamada, Y.; Iwasawa, C.; Shibasaki, M.; Maekawa, A.; Matsuo, M.; et al. Down-regulation of ghrelin receptors on dopaminergic neurons in the substantia nigra contributes to Parkinson’s disease-like motor dysfunction. Mol. Brain 2018, 11, 6. [Google Scholar] [CrossRef]
- Shiba-Fukushima, K.; Ishikawa, K.I.; Inoshita, T.; Izawa, N.; Takanashi, M.; Sato, S.; Onodera, O.; Akamatsu, W.; Okano, H.; Imai, Y.; et al. Evidence that phosphorylated ubiquitin signaling is involved in the etiology of Parkinson’s disease. Hum. Mol. Genet. 2017, 26, 3172–3185. [Google Scholar] [CrossRef]
- Benkert, J.; Hess, S.; Roy, S.; Beccano-Kelly, D.; Wiederspohn, N.; Duda, J.; Simons, C.; Patil, K.; Gaifullina, A.; Mannal, N.; et al. Cav2.3 channels contribute to dopaminergic neuron loss in a model of Parkinson’s disease. Nat. Commun. 2019, 10, 5094. [Google Scholar] [CrossRef]
- Grigor’eva, E.V.; Drozdova, E.S.; Sorogina, D.A.; Malakhova, A.A.; Pavlova, S.V.; Vyatkin, Y.V.; Khabarova, E.A.; Rzaev, J.A.; Medvedev, S.P.; Zakian, S.M. Generation of induced pluripotent stem cell line, ICGi034-A, by reprogramming peripheral blood mononuclear cells from a patient with Parkinson’s disease associated with GBA mutation. Stem Cell Res. 2021, 59, 102651. [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]
- Chen, Z.T.; Zhao, Z.H.; Chen, L.N.; Fan, F.; Cai, G.E.; Weng, H.D.; Wang, Y.Q.; Liao, L.M.; Chen, X.C.; Huang, E.; et al. Generation of an induced pluripotent stem cell line, FJMUUHi001-A, from a hereditary Parkinson’s disease patient with homozygous mutation of c.189dupA in PARK7. Stem Cell Res. 2021, 51, 102175. [Google Scholar] [CrossRef]
- Peitz, M.; Bechler, T.; Thiele, C.C.; Veltel, M.; Bloschies, M.; Fliessbach, K.; Ramirez, A.; Brüstle, O. Blood-derived integration-free iPS cell line UKBi011-A from a diagnosed male Alzheimer’s disease patient with APOE ε4/ε4 genotype. Stem Cell Res. 2018, 29, 250–253. [Google Scholar] [CrossRef] [PubMed]
- Michaelson, D.M. APOE ε4: The most prevalent yet understudied risk factor for Alzheimer’s disease. Alzheimers Dement. 2014, 10, 861–868. [Google Scholar] [CrossRef] [PubMed]
- Yu, J.T.; Tan, L.; Hardy, J. Apolipoprotein E in Alzheimer’s disease: An update. Annu. Rev. Neurosci. 2014, 37, 79–100. [Google Scholar] [CrossRef] [PubMed]
- Zhang, W.; Di, W.; Zhao, J.; Zhang, B.; Wang, Y. Generation of a SIAISi004-A hiPSC line from PBMCs of a 74 year-old Alzheimer’s disease woman by non-integrating sendai virus mediated reprogramming. Stem Cell Res. 2021, 55, 102501. [Google Scholar] [CrossRef] [PubMed]
- Liu, S.; Zhao, Y.; Su, X.; Zhou, C.; Yang, P.; Lin, Q.; Li, S.; Tan, H.; Wang, Q.; Wang, C.; et al. Reconstruction of Alzheimer’s Disease Cell Model In Vitro via Extracted Peripheral Blood Molecular Cells from a Sporadic Patient. Stem Cells Int. 2020, 2020, 8897494. [Google Scholar] [CrossRef]
- Cusulin, C.; Wells, I.; Badillo, S.; Duran-Pacheco, G.C.; Baumann, K.; Patsch, C. Gamma secretase modulators and BACE inhibitors reduce Aβ production without altering gene expression in Alzheimer’s disease iPSC-derived neurons and mice. Mol. Cell Neurosci. 2019, 100, 103392. [Google Scholar] [CrossRef]
- 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]
- Chang, K.H.; Lee-Chen, G.J.; Huang, C.C.; Lin, J.L.; Chen, Y.J.; Wei, P.C.; Lo, Y.S.; Yao, C.F.; Kuo, M.W.; Chen, C.M. Modeling Alzheimer’s Disease by Induced Pluripotent Stem Cells Carrying APP D678H Mutation. Mol. Neurobiol. 2019, 56, 3972–3983. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, J.; Liu, X.; Du, X.; Ma, Z.; Liu, B.; Guo, R.; Feng, B.; Ma, J.; Cui, H. Induced pluripotent stem cells derived from one 70-years-old male donor with the APOE-ε4/ε4 alleles. Stem Cell Res. 2021, 53, 102395. [Google Scholar] [CrossRef]
- Zhang, L.; Ren, Q.; Liu, X.; Liu, G.; Meng, S.; Xiahou, K.; Zhang, Y.; Jiang, N.; Zhou, W. Generation of induced pluripotent stem cell line (IPTi002-A) from an 87-year old sporadic Alzheimer’s disease patient with APOE3 (ε3/ε3) genotype. Stem Cell Res. 2021, 53, 102282. [Google Scholar] [CrossRef] [PubMed]
- Fujita, Y.; Murakami, T.; Nakamura, A. Recent Advances in Biomarkers and Regenerative Medicine for Diabetic Neuropathy. Int. J. Mol. Sci. 2021, 22, 2301. [Google Scholar] [CrossRef] [PubMed]
- Mittal, K.; Schrenk-Siemens, K. Lessons from iPSC research: Insights on peripheral nerve disease. Neurosci. Lett. 2020, 738, 135358. [Google Scholar] [CrossRef] [PubMed]
- Xu, J.; Zuo, C. The Fate Status of Stem Cells in Diabetes and its Role in the Occurrence of Diabetic Complications. Front. Mol. Biosci. 2021, 8, 745035. [Google Scholar] [CrossRef]
- Kanada, S.; Makino, E.; Nakamura, N.; Miyabe, M.; Ito, M.; Hata, M.; Yamauchi, T.; Sawada, N.; Kondo, S.; Saiki, T.; et al. Direct Comparison of Therapeutic Effects on Diabetic Polyneuropathy between Transplantation of Dental Pulp Stem Cells and Administration of Dental Pulp Stem Cell-Secreted Factors. Int. J. Mol. Sci. 2020, 21, 6064. [Google Scholar] [CrossRef]
- Wang, Q.; Chen, F.Y.; Ling, Z.M.; Su, W.F.; Zhao, Y.Y.; Chen, G.; Wei, Z.Y. The Effect of Schwann Cells/Schwann Cell-Like Cells on Cell Therapy for Peripheral Neuropathy. Front. Cell. Neurosci. 2022, 16, 836931. [Google Scholar] [CrossRef]
- Himeno, T.; Kamiya, H.; Naruse, K.; Cheng, Z.; Ito, S.; Kondo, M.; Okawa, T.; Fujiya, A.; Kato, J.; Suzuki, H.; et al. Mesenchymal stem cell-like cells derived from mouse induced pluripotent stem cells ameliorate diabetic polyneuropathy in mice. Biomed. Res. Int. 2013, 2013, 259187. [Google Scholar] [CrossRef]
- Levi, A.D.; Guénard, V.; Aebischer, P.; Bunge, R.P. The functional characteristics of Schwann cells cultured from human peripheral nerve after transplantation into a gap within the rat sciatic nerve. J. Neurosci. 1994, 14, 1309–1319. [Google Scholar] [CrossRef]
- Gersey, Z.C.; Burks, S.S.; Anderson, K.D.; Dididze, M.; Khan, A.; Dietrich, W.D.; Levi, A.D. First human experience with autologous Schwann cells to supplement sciatic nerve repair: Report of 2 cases with long-term follow-up. Neurosurg. Focus 2017, 42, E2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Levi, A.D.; Burks, S.S.; Anderson, K.D.; Dididze, M.; Khan, A.; Dietrich, W.D. The Use of Autologous Schwann Cells to Supplement Sciatic Nerve Repair With a Large Gap: First in Human Experience. Cell Transplant. 2016, 25, 1395–1403. [Google Scholar] [CrossRef] [PubMed]
- Stratton, J.A.; Kumar, R.; Sinha, S.; Shah, P.; Stykel, M.; Shapira, Y.; Midha, R.; Biernaskie, J. Purification and Characterization of Schwann Cells from Adult Human Skin and Nerve. eNeuro 2017, 4, ENEURO.0307-16.2017. [Google Scholar] [CrossRef] [PubMed]
- Namer, B.; Schmidt, D.; Eberhardt, E.; Maroni, M.; Dorfmeister, E.; Kleggetveit, I.P.; Kaluza, L.; Meents, J.; Gerlach, A.; Lin, Z.; et al. Pain relief in a neuropathy patient by lacosamide: Proof of principle of clinical translation from patient-specific iPS cell-derived nociceptors. EBioMedicine 2019, 39, 401–408. [Google Scholar] [CrossRef]
- Berlet, R.; Galang Cabantan, D.A.; Gonzales-Portillo, D.; Borlongan, C.V. Enriched Environment and Exercise Enhance Stem Cell Therapy for Stroke, Parkinson’s Disease, and Huntington’s Disease. Front. Cell Dev. Biol. 2022, 10, 798826. [Google Scholar] [CrossRef]
- Duan, R.; Gao, Y.; He, R.; Jing, L.; Li, Y.; Gong, Z.; Yao, Y.; Luan, T.; Zhang, C.; Li, L.; et al. Induced Pluripotent Stem Cells for Ischemic Stroke Treatment. Front. Neurosci. 2021, 15, 628663. [Google Scholar] [CrossRef]
- Eckert, A.; Huang, L.; Gonzalez, R.; Kim, H.S.; Hamblin, M.H.; Lee, J.P. Bystander Effect Fuels Human Induced Pluripotent Stem Cell-Derived Neural Stem Cells to Quickly Attenuate Early Stage Neurological Deficits After Stroke. Stem Cells Transl. Med. 2015, 4, 841–851. [Google Scholar] [CrossRef]
- Chen, Y.; Song, F.; Tu, M.; Wu, S.; He, X.; Liu, H.; Xu, C.; Zhang, K.; Zhu, Y.; Zhou, R.; et al. Quantitative proteomics revealed extensive microenvironmental changes after stem cell transplantation in ischemic stroke. Front. Med. 2022, 16, 429–441. [Google Scholar] [CrossRef]
- Kirabali, T.; Rust, R. iPS-derived pericytes for neurovascular regeneration. Eur. J. Clin. Investig. 2021, 51, e13601. [Google Scholar] [CrossRef]
- Sun, J.; Huang, Y.; Gong, J.; Wang, J.; Fan, Y.; Cai, J.; Wang, Y.; Qiu, Y.; Wei, Y.; Xiong, C.; et al. Transplantation of hPSC-derived pericyte-like cells promotes functional recovery in ischemic stroke mice. Nat. Commun. 2020, 11, 5196. [Google Scholar] [CrossRef]
- Salikhova, D.; Bukharova, T.; Cherkashova, E.; Namestnikova, D.; Leonov, G.; Nikitina, M.; Gubskiy, I.; Akopyan, G.; Elchaninov, A.; Midiber, K.; et al. Therapeutic Effects of hiPSC-Derived Glial and Neuronal Progenitor Cells-Conditioned Medium in Experimental Ischemic Stroke in Rats. Int. J. Mol. Sci. 2021, 22, 4694. [Google Scholar] [CrossRef]
- Li, X.; Sundström, E. Stem Cell Therapies for Central Nervous System Trauma: The 4 Ws-What, When, Where, and Why. Stem Cells Transl. Med. 2022, 11, 14–25. [Google Scholar] [CrossRef] [PubMed]
- Huang, L.; Fu, C.; Xiong, F.; He, C.; Wei, Q. Stem Cell Therapy for Spinal Cord Injury. Cell Transplant. 2021, 30, 963689721989266. [Google Scholar] [CrossRef] [PubMed]
- Kajikawa, K.; Imaizumi, K.; Shinozaki, M.; Shibata, S.; Shindo, T.; Kitagawa, T.; Shibata, R.; Kamata, Y.; Kojima, K.; Nagoshi, N.; et al. Cell therapy for spinal cord injury by using human iPSC-derived region-specific neural progenitor cells. Mol. Brain 2020, 13, 120. [Google Scholar] [CrossRef] [PubMed]
- Imaizumi, K.; Sone, T.; Ibata, K.; Fujimori, K.; Yuzaki, M.; Akamatsu, W.; Okano, H. Controlling the Regional Identity of hPSC-Derived Neurons to Uncover Neuronal Subtype Specificity of Neurological Disease Phenotypes. Stem Cell Rep. 2015, 5, 1010–1022. [Google Scholar] [CrossRef] [PubMed]
- Gu, J.; Cromer, B.; Sumer, H. Forward Programming of Pluripotent Stem Cells to Neurons. Curr. Mol. Med. 2021, 21, 5–14. [Google Scholar] [CrossRef]
- 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]
- Capuz, A.; Karnoub, M.A.; Osien, S.; Rose, M.; Mériaux, C.; Fournier, I.; Devos, D.; Vanden Abeele, F.; Rodet, F.; Cizkova, D.; et al. The Antibody Dependant Neurite Outgrowth Modulation Response Involvement in Spinal Cord Injury. Front. Immunol. 2022, 13, 882830. [Google Scholar] [CrossRef]
- Kong, D.; Feng, B.; Amponsah, A.E.; He, J.; Guo, R.; Liu, B.; Du, X.; Liu, X.; Zhang, S.; Lv, F.; et al. hiPSC-derived NSCs effectively promote the functional recovery of acute spinal cord injury in mice. Stem Cell Res. Ther. 2021, 12, 172. [Google Scholar] [CrossRef]
- Csobonyeiova, M.; Polak, S.; Zamborsky, R.; Danisovic, L. Recent Progress in the Regeneration of Spinal Cord Injuries by Induced Pluripotent Stem Cells. Int. J. Mol. Sci. 2019, 20, 3838. [Google Scholar] [CrossRef]
- Hamazaki, T.; El Rouby, N.; Fredette, N.C.; Santostefano, K.E.; Terada, N. Concise Review: Induced Pluripotent Stem Cell Research in the Era of Precision Medicine. Stem Cells 2017, 35, 545–550. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hankowski, K.E.; Hamazaki, T.; Umezawa, A.; Terada, N. Induced pluripotent stem cells as a next-generation biomedical interface. Lab. Investig. 2011, 91, 972–977. [Google Scholar] [CrossRef] [PubMed]
- Gowing, G.; Svendsen, S.; Svendsen, C.N. Ex vivo gene therapy for the treatment of neurological disorders. Prog. Brain Res. 2017, 230, 99–132. [Google Scholar] [CrossRef] [PubMed]
- Savić, N.; Schwank, G. Advances in therapeutic CRISPR/Cas9 genome editing. Transl. Res. 2016, 168, 15–21. [Google Scholar] [CrossRef]
- Mingozzi, F.; High, K.A. Therapeutic in vivo gene transfer for genetic disease using AAV: Progress and challenges. Nat. Rev. Genet. 2011, 12, 341–355. [Google Scholar] [CrossRef]
- Liu, Y.; Wang, D.A. Viral vector-mediated transgenic cell therapy in regenerative medicine: Safety of the process. Expert Opin. Biol. Ther. 2015, 15, 559–567. [Google Scholar] [CrossRef]
- Barrett, R.; Ornelas, L.; Yeager, N.; Mandefro, B.; Sahabian, A.; Lenaeus, L.; Targan, S.R.; Svendsen, C.N.; Sareen, D. Reliable generation of induced pluripotent stem cells from human lymphoblastoid cell lines. Stem Cells Transl. Med. 2014, 3, 1429–1434. [Google Scholar] [CrossRef]
- Vierbuchen, T.; Ostermeier, A.; Pang, Z.P.; Kokubu, Y.; Südhof, T.C.; Wernig, M. Direct conversion of fibroblasts to functional neurons by defined factors. Nature 2010, 463, 1035–1041. [Google Scholar] [CrossRef]
- Mandai, M.; Watanabe, A.; Kurimoto, Y.; Hirami, Y.; Morinaga, C.; Daimon, T.; Fujihara, M.; Akimaru, H.; Sakai, N.; Shibata, Y.; et al. Autologous Induced Stem-Cell-Derived Retinal Cells for Macular Degeneration. N. Engl. J. Med. 2017, 376, 1038–1046. [Google Scholar] [CrossRef]
- Madrid, M.; Sumen, C.; Aivio, S.; Saklayen, N. Autologous Induced Pluripotent Stem Cell-Based Cell Therapies: Promise, Progress, and Challenges. Curr. Protoc. 2021, 1, e88. [Google Scholar] [CrossRef]
- Karagiannis, P.; Nakauchi, A.; Yamanaka, S. Bringing Induced Pluripotent Stem Cell Technology to the Bedside. JMA J. 2018, 1, 6–14. [Google Scholar] [CrossRef] [PubMed]
- Perestrelo, A.R.; Águas, A.C.; Rainer, A.; Forte, G. Microfluidic Organ/Body-on-a-Chip Devices at the Convergence of Biology and Microengineering. Sensors 2015, 15, 31142–31170. [Google Scholar] [CrossRef] [PubMed]
- Imamura, Y.; Mukohara, T.; Shimono, Y.; Funakoshi, Y.; Chayahara, N.; Toyoda, M.; Kiyota, N.; Takao, S.; Kono, S.; Nakatsura, T.; et al. Comparison of 2D- and 3D-culture models as drug-testing platforms in breast cancer. Oncol. Rep. 2015, 33, 1837–1843. [Google Scholar] [CrossRef] [PubMed]
- Workman, M.J.; Gleeson, J.P.; Troisi, E.J.; Estrada, H.Q.; Kerns, S.J.; Hinojosa, C.D.; Hamilton, G.A.; Targan, S.R.; Svendsen, C.N.; Barrett, R.J. Enhanced Utilization of Induced Pluripotent Stem Cell-Derived Human Intestinal Organoids Using Microengineered Chips. Cell Mol. Gastroenterol. Hepatol. 2018, 5, 669–677.e662. [Google Scholar] [CrossRef] [PubMed]
- Bircsak, K.M.; DeBiasio, R.; Miedel, M.; Alsebahi, A.; Reddinger, R.; Saleh, A.; Shun, T.; Vernetti, L.A.; Gough, A. A 3D microfluidic liver model for high throughput compound toxicity screening in the OrganoPlate®. Toxicology 2021, 450, 152667. [Google Scholar] [CrossRef]
- Wang, Y.; Wang, H.; Deng, P.; Chen, W.; Guo, Y.; Tao, T.; Qin, J. In situ differentiation and generation of functional liver organoids from human iPSCs in a 3D perfusable chip system. Lab Chip 2018, 18, 3606–3616. [Google Scholar] [CrossRef]
- Musah, S.; Mammoto, A.; Ferrante, T.C.; Jeanty, S.S.F.; Hirano-Kobayashi, M.; Mammoto, T.; Roberts, K.; Chung, S.; Novak, R.; Ingram, M.; et al. Mature induced-pluripotent-stem-cell-derived human podocytes reconstitute kidney glomerular-capillary-wall function on a chip. Nat. Biomed. Eng. 2017, 1, 0069. [Google Scholar] [CrossRef]
- Kujala, V.J.; Pasqualini, F.S.; Goss, J.A.; Nawroth, J.C.; Parker, K.K. Laminar ventricular myocardium on a microelectrode array-based chip. J. Mater. Chem. B 2016, 4, 3534–3543. [Google Scholar] [CrossRef]
- Zhang, Y.S.; Arneri, A.; Bersini, S.; Shin, S.R.; Zhu, K.; Goli-Malekabadi, Z.; Aleman, J.; Colosi, C.; Busignani, F.; Dell’Erba, V.; et al. Bioprinting 3D microfibrous scaffolds for engineering endothelialized myocardium and heart-on-a-chip. Biomaterials 2016, 110, 45–59. [Google Scholar] [CrossRef]
- Marsano, A.; Conficconi, C.; Lemme, M.; Occhetta, P.; Gaudiello, E.; Votta, E.; Cerino, G.; Redaelli, A.; Rasponi, M. Beating heart on a chip: A novel microfluidic platform to generate functional 3D cardiac microtissues. Lab Chip 2016, 16, 599–610. [Google Scholar] [CrossRef]
- Schneider, O.; Zeifang, L.; Fuchs, S.; Sailer, C.; Loskill, P. User-Friendly and Parallelized Generation of Human Induced Pluripotent Stem Cell-Derived Microtissues in a Centrifugal Heart-on-a-Chip. Tissue Eng. Part A 2019, 25, 786–798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brown, J.A.; Codreanu, S.G.; Shi, M.; Sherrod, S.D.; Markov, D.A.; Neely, M.D.; Britt, C.M.; Hoilett, O.S.; Reiserer, R.S.; Samson, P.C.; et al. Metabolic consequences of inflammatory disruption of the blood-brain barrier in an organ-on-chip model of the human neurovascular unit. J. Neuroinflamm. 2016, 13, 306. [Google Scholar] [CrossRef] [PubMed]
- Brown, J.A.; Pensabene, V.; Markov, D.A.; Allwardt, V.; Neely, M.D.; Shi, M.; Britt, C.M.; Hoilett, O.S.; Yang, Q.; Brewer, B.M.; et al. Recreating blood-brain barrier physiology and structure on chip: A novel neurovascular microfluidic bioreactor. Biomicrofluidics 2015, 9, 054124. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Wang, L.; Guo, Y.; Zhu, Y.; Qin, J. Engineering stem cell-derived 3D brain organoids in a perfusable organ-on-a-chip system. RSC Adv. 2018, 8, 1677–1685. [Google Scholar] [CrossRef] [PubMed]
- Sakolish, C.; Reese, C.E.; Luo, Y.S.; Valdiviezo, A.; Schurdak, M.E.; Gough, A.; Taylor, D.L.; Chiu, W.A.; Vernetti, L.A.; Rusyn, I. Analysis of reproducibility and robustness of a human microfluidic four-cell liver acinus microphysiology system (LAMPS). Toxicology 2021, 448, 152651. [Google Scholar] [CrossRef]
- Fanizza, F.; Campanile, M.; Forloni, G.; Giordano, C.; Albani, D. Induced pluripotent stem cell-based organ-on-a-chip as personalized drug screening tools: A focus on neurodegenerative disorders. J. Tissue Eng. 2022, 13, 20417314221095339. [Google Scholar] [CrossRef]
- Baghbaderani, B.A.; Tian, X.; Neo, B.H.; Burkall, A.; Dimezzo, T.; Sierra, G.; Zeng, X.; Warren, K.; Kovarcik, D.P.; Fellner, T.; et al. cGMP-Manufactured Human Induced Pluripotent Stem Cells Are Available for Pre-clinical and Clinical Applications. Stem Cell Rep. 2015, 5, 647–659. [Google Scholar] [CrossRef]
- Rivera, T.; Zhao, Y.; Ni, Y.; Wang, J. Human-Induced Pluripotent Stem Cell Culture Methods Under cGMP Conditions. Curr. Protoc. Stem Cell Biol. 2020, 54, e117. [Google Scholar] [CrossRef]
- Denker, H.W. Ethical concerns over use of new cloning technique in humans. Nature 2009, 461, 341. [Google Scholar] [CrossRef]
- Denker, H.W. Induced pluripotent stem cells: How to deal with the developmental potential. Reprod. Biomed. Online 2009, 19 (Suppl. 1), 34–37. [Google Scholar] [CrossRef]
- Lo, B.; Parham, L.; Alvarez-Buylla, A.; Cedars, M.; Conklin, B.; Fisher, S.; Gates, E.; Giudice, L.; Halme, D.G.; Hershon, W.; et al. Cloning mice and men: Prohibiting the use of iPS cells for human reproductive cloning. Cell Stem Cell 2010, 6, 16–20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 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
Pandey, S.; Jirásko, M.; Lochman, J.; Chvátal, A.; Chottova Dvorakova, M.; Kučera, R. iPSCs in Neurodegenerative Disorders: A Unique Platform for Clinical Research and Personalized Medicine. J. Pers. Med. 2022, 12, 1485. https://doi.org/10.3390/jpm12091485
Pandey S, Jirásko M, Lochman J, Chvátal A, Chottova Dvorakova M, Kučera R. iPSCs in Neurodegenerative Disorders: A Unique Platform for Clinical Research and Personalized Medicine. Journal of Personalized Medicine. 2022; 12(9):1485. https://doi.org/10.3390/jpm12091485
Chicago/Turabian StylePandey, Shashank, Michal Jirásko, Jan Lochman, Alexandr Chvátal, Magdalena Chottova Dvorakova, and Radek Kučera. 2022. "iPSCs in Neurodegenerative Disorders: A Unique Platform for Clinical Research and Personalized Medicine" Journal of Personalized Medicine 12, no. 9: 1485. https://doi.org/10.3390/jpm12091485
APA StylePandey, S., Jirásko, M., Lochman, J., Chvátal, A., Chottova Dvorakova, M., & Kučera, R. (2022). iPSCs in Neurodegenerative Disorders: A Unique Platform for Clinical Research and Personalized Medicine. Journal of Personalized Medicine, 12(9), 1485. https://doi.org/10.3390/jpm12091485