APP下载

Cell replacement therapy for central nervous system diseases

2015-01-21DanjuTsoRandallMcKinnon

Danju Tso, Randall D. McKinnon

Department of Surgery (Neurosurgery), Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA

Cell replacement therapy for central nervous system diseases

Danju Tso#, Randall D. McKinnon*,#

Department of Surgery (Neurosurgery), Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA

The brain and spinal cord can not replace neurons or supporting glia that are lost through traumatic injury or disease. In pre-clinical studies, however, neural stem and progenitor cell transplants can promote functional recovery. Thus the central nervous system is repair competent but lacks endogenous stem cell resources. To make transplants clinically feasible, this fi eld needs a source of histocompatible, ethically acceptable and non-tumorgenic cells. One strategy to generate patient-specifi c replacement cells is to reprogram autologous cells such as fi broblasts into pluripotent stem cells which can then be diff erentiated into the required cell grafts. However, the utility of pluripotent cell derived grafts is limited since they can retain founder cells with intrinsic neoplastic potential. A recent extension of this technology directly reprograms fi broblasts into the fi nal graftable cells without an induced pluripotent stem cell intermediate, avoiding the pluripotent caveat. For both types of reprogramming the conversion effi ciency is very low resulting in the need to amplify the cells in culture which can lead to chromosomal instability and neoplasia. Thus to make reprogramming biology clinically feasible, we must improve the effi ciency. The ultimate source of replacement cells may reside in directly reprogramming accessible cells within the brain.

in vivo direct reprogramming; spinal cord injury; trauma; personalized medicine; induced pluripotent stem cell; embryonic stem cells

Funding: This study was supported by grants from the New Jersey Commission on Spinal Cord Research (11-0015-SCR).

Tso D, McKinnon RD (2015) Cell replacement therapy for central nervous system diseases. Neural Regen Res 10(9):1356-1358.

Introduction

The adult central nervous system (CNS) has limited de novo neurogenesis (Blight, 2002; Bechmann, 2005) and only minimal capacity to replace cells lost due to tissue insult, injury or disease. This is seen in acquired and inherited brain disease (Alzheimer’s disease (AD), Parkinson’s disease (PD), multiple sclerosis (MS)) and in CNS injury induced by trauma, vascular insult or surgery. Trauma carries the added burden of a breakdown in the blood-brain barrier which introduces systemic immunity that exacerbates parenchymal destruction. Immune suppression can slow wound progression, but this does not promote repair. Damaged axons also do not regenerate through gliotic scars or inhibitors in myelin (Filbin, 2003). Thus we rely on redundancy and rewiring of surviving circuits for partial recovery of lost function. This failure to repair likely refl ects insuffi -cient stem cells, rather than repair competence, since exogenous cells are eff ective in many preclinical models of genetic, chemical and traumatic brain and spinal cord injury (SCI) (Chen et al., 2007). Indeed we (Kiel et al., 2008) and others (Windrem et al., 2008) have used cell grafts to completely rescue a lethal cell autonomous neurodegenerative mouse model. Thus at present the objectives for clinical intervention in brain repair are to arrest wound progression and promote cell replacement therapy. Here we examine potential sources of replacement cells.

Replacement Cells

For pre-clinical milestones to be translated in clinical practice, we must fi rst identify an appropriate graft resource. At present, the three sources include allografts of fetal brain tissue, allografts derived from pluripotent embryonic stem cells (ESCs) (Murry and Keller, 2008), and autologous neural cells generated in vitro by reprogramming patient-specifi c somatic cells such as dermal fi broblasts. Fetal brain allografts were used for PD, the fi rst placebo-controlled neurosurgical trial in the U.S., and the cell source, fetal dopaminergic neurons, proved both ineff ective and diffi cult to standardize (Freed et al., 2001). ESC-derived glial progenitor cells were used in a trial for acute SCI sponsored by Geron Inc. This trial was based on initially promising pre-clinical studies with CNS myelin forming oligodendroglial progenitor cell (OPC) transplants (Keirstead et al., 2005). Both pre-clinical and clinical trials with such cells have generated sometimes sensational but often controversial results (Burke et al., 2013), and the Geron trial was halted early due to cost, immune complications and apparently cystic nodule formations. Both the fetal tissue and embryonic stem (ES) derived cells have ethical limitations, both represent allografts that require immune suppression with serious side eff ects, and cell grafts derived from pluripotent ESCs can also carry an unacceptable risk for neoplasia. We demonstrated that therapeutic engraftment requires substantial cell numbers (Kiel et al., 2008), well within the neoplastic load limit of ESC-derived cultures (Sadowski et al., 2010). Thus grafts derived from pluripotent cells may never be considered therapeutically safe and appropriate for organ repair.

A third potential source of replacement brain cells are autol-

ogous somatic cells genetically engineered to trans-diff erentiate into neural cells. The reprogramming fi eld has now given us this novel and exciting strategy to generate ethically neutral, patient-specifi c replacement cells from induced pluripotent stem (iPS) cells. Yamanaka identifi ed four factors that reprogram fi broblasts into iPS cells (Takahashi and Yamanaka, 2006), extending early work in amphibian (Gurdon and Melton, 2008) showing that cell fate can be plastic (Yamanaka, 2009). The process appears to work on any cell type (Park et al., 2008b; Soldner et al., 2009; Yu et al., 2009) including readily accessible cells such as adipose-derived mesenchymal stromal cells (MSCs), and iPS lines have now been established from many sources (Park et al., 2008a; Soldner et al., 2009). The process requires pioneer factors Oct4 and Sox2 (Takahashi et al., 2007; Smale, 2010) but can also work with Oct4 (Kim et al., 2009; Zhu et al., 2010) combined with small molecules that promote chromatin remodeling (Huangfu et al., 2008; Lin et al., 2009). Patient derived iPS cells now have potential to generate disease specifi c cell types that can replace animals for drug screens. However the iPS cell reprogramming is not complete (Kim et al., 2010; Lister et al., 2011) raising the concern they may not generate valid replacement cells, and since they are immune protected autologous cells their neoplastic potential will be even greater than ESC-derived allografts.

An alternative to generate autologous cells for graft therapy is to fi nd some combination of factors that directly reprogram fi broblasts into the desired cell types and avoid the pluripotent intermediates. Trans-diff erentiation has been controversial and early reports were either disproved (Bertani et al., 2005; Woodbury et al., 2000), misinterpretations due to cell fusion (Lagasse et al., 2000; Wang et al., 2003) or remain unconfi rmed (Jiang et al., 2002). However, Yamanaka has rejuvenated the fi eld and recent studies now demonstrate direct reprogramming of pancreatic exocrine cells into β-cells (Zhou et al., 2008) and somatic fi broblasts into hepatocytes, cardiomyocytes, blood progenitors and neurons (Zhou et al., 2008; Ieda et al., 2010; Szabo et al., 2010; Vierbuchen et al., 2010; Efe et al., 2011; Pfi sterer et al., 2011; Sekiya and Suzuki, 2011; Son et al., 2011; Karow et al., 2012). The process is not fully understood and to date reprogramming to generate some cell types has resulted in only partial phenotypes (Najm et al., 2013; Yang et al., 2013).

Reprogramming Effi ciency

For SCI repair three conclusions are quite clear and none are unique to SCI research. First we need a standardized source of autologous cells. Second we need rigid and objective approaches to resolve graft outcome and evaluate effi cacy (Burke et al., 2013). Third, in order to generate a safe graft reagent it is imperative that we improve the effi ciency of the reprogramming process. The low effi ciency of reprogramming generates very few graftable cells, and the necessary mitogen amplifi cation in vitro can compromises the quality control and result in karyotype abnormalities and neoplasia concerns (Miura et al., 2006; Tolar et al., 2007).

All forms of cell reprogramming suff er from low effi ciencies. For iPS cell reprogramming the effi ciency is commonly 0.1%. This can be improved under experimental settings by starting with fi broblasts that contain drug inducible reprogramming factor transgenes, although for these the rate remains less than 10%. Chromatin remodeling is also an early event (Simonsson and Gurdon, 2004) and a limiting factor (Luna-Zurita and Bruneau, 2013) for cell reprogramming. Small molecules that block DNA and histone methylation, which remodel the epigenome by removing transcription repressive marks, also enhance iPS cell reprogramming (Huangfu et al., 2008; Lin et al., 2009). We recently identifi ed a histone H1 chaperone that also promotes reprogramming (Tso et al., in preparation). This factor appears to modulate chromatin structure by relaxing condensed chromatin, as seen with other H1 modifying enzymes (Christophorou et al., 2014) and binding proteins (Philpott et al., 1991; Martic et al., 2005; Hayakawa et al., 2012). Thus chromatin remodeling factors may provide the key to optimizing the reprogramming process.

In vivo Reprogramming

Finally, an emerging strategy to avoid long term culture of graft cells is to deliver the exogenous reprogramming factors directly into target cells in vivo. A graft-free reprogramming strategy would expand our paradigm for cell replacement for clinical therapies. Gene delivery methods such as viral vectors are feasible, and direct gene transduction using episomal plasmids would avoid the safety concerns and oncogenic potential of viral vectors. For in vivo reprogramming to work eff ectively it will be necessary to identify target cell populations that can serve as a resource for cell reprogramming. For neuronal cell replacement, both pericytes (Karow et al., 2012) and astrocytes (Niu et al., 2013) can be reprogrammed into induced neurons, and elevated levels of the transcription factor Sox10 can convert peripheral satellite glia into CNS-like myelinating glia (Weider et al., 2015). Another target population in the adult brain are NG2 cells (Nishiyama et al., 1996; Nishiyama et al., 2009). NG2, a transmembrane proteoglycan expressed by OPCs in vitro, identifi es presumed myelin lineage glial progenitors in vivo. The adult NG2 population includes 5% of the cells in the adult brain (Nishiyama, 2007), and at least some of these may generate other cell types (Nishiyama et al., 2009). NG2 cells can generate protoplasmic astrocytes in grey matter. They can respond to extrinsic stimuli and injury. They can also respond to neurotransmitters and thus may have a role in neural transmission. While it is not clear why NG2 cells do not promote myelin replacement in injury or disease, it is clear that at least a subset of these cells are good targets for transgene delivery to regenerate myelinating oligodendrocytes in vivo. To achieve this we will need to identify relevant transcription factors to eff ectively reprogram these cells in vivo.

Confl icts of interest: The authors declare no competing fi nancial interests.

Bechmann I (2005) Failed central nervous system regeneration - A downside of immune privilege? Neuromol Med 7:217-228.

Bertani N, Malatesta P, Volpi G, Sonego P, Perris R (2005) Neurogenic potential of human mesenchymal stem cells revisited: analysis by immunostaining, time-lapse video and microarray. J Cell Sci 118:3925-3936.

Blight AR (2002) Miracles and molecules - progress in spinal cord repair. Nat Neurosci 5:1051-1054.

Burke DA, Whittemore SR, Magnuson DS (2013) Consequences of common data analysis inaccuracies in CNS trauma injury basic research. J Neurotrauma 30:797-805.

Chen CP, Kiel ME, Sadowski D, McKinnon RD (2007) From stem cells to oligodendrocytes: Prospects for brain therapy. Stem Cell Rev 3:280-288.

Christophorou MA, Castelo-Branco G, Halley-Stott RP, Oliveira CS, Loos R, Radzisheuskaya A, Mowen KA, Bertone P, Silva JC, Zernicka-Goetz M, Nielsen ML, Gurdon JB, Kouzarides T (2014) Citrullination regulates pluripotency and histone H1 binding to chromatin. Nature 507:104-108.

Efe JA, Hilcove S, Kim J, Zhou H, Ouyang K, Wang G, Chen J, Ding S (2011) Conversion of mouse fi broblasts into cardiomyocytes using a direct reprogramming strategy. Nat Cell Biol 13:215-222.

Filbin MT (2003) Myelin-associated inhibitors of axonal regeneration in the adult mammalian CNS. Nat Rev Neurosci 4:703-713.

Freed CR, Breeze RE, Greene P, Fahn S, Tsai WY, Trojanowski JQ, Eidelberg D (2001) Transplanted dopaminergic neurons: More or less? Nat Med 7:512-513.

Gurdon JB, Melton DA (2008) Nuclear reprogramming in cells. Science 322:1811-1815.

Hayakawa K, Ohgane J, Tanaka S, Yagi S, Shiota K (2012) Oocyte-specifi c linker histone H1foo is an epigenomic modulator that decondenses chromatin and impairs pluripotency. Epigenetics 7:1029-1036.

Huangfu DW, Maehr R, Guo WJ, Eijkelenboom A, Snitow M, Chen AE, Melton DA (2008) Induction of pluripotent stem cells by defi ned factors is greatly improved by small-molecule compounds. Nat Biotechnol 26:795-797.

Ieda M, Fu JD, Delgado-Olguin P, Vedantham V, Hayashi Y, Bruneau BG, Srivastava D (2010) Direct reprogramming of fi broblasts into functional cardiomyocytes by defi ned factors. Cell 142:375-386.

Jiang YH, Jahagirdar BN, Reinhardt RL, Schwartz RE, Keene CD, Ortiz-Gonzalez XR, Reyes M, Lenvik T, Lund T, Blackstad M, Du JB, Aldrich S, Lisberg A, Low WC, Largaespada DA, Verfaillie CM (2002) Pluripotency of mesenchymal stem cells derived from adult marrow. Nature 418:41-49.

Karow M, Sanchez R, Schichor C, Masserdotti G, Ortega F, Heinrich C, Gascon S, Khan MA, Lie DC, Dellavalle A, Cossu G, Goldbrunner R, Gotz M, Berninger B (2012) Reprogramming of pericyte-derived cells of the adult human brain into induced neuronal cells. Cell Stem Cell 11:471-476.

Keirstead HS, Nistor G, Bernal G, Totoiu M, Cloutier F, Sharp K, Steward O (2005) Human embryonic stem cell-derived oligodendrocyte progenitor cell transplants remyelinate and restore locomotion after spinal cord injury. J Neurosci 25:4694-4705.

Kiel ME, Chen CP, Sadowski D, McKinnon RD (2008) Stem cell-derived therapeutic myelin repair requires 7% cell replacement. Stem Cells 26:2229-2236.

Kim JB, Greber B, Arauzo-Bravo MJ, Meyer J, Park KI, Zaehres H, Scholer HR (2009) Direct reprogramming of human neural stem cells by OCT4. Nature 461:649-653.

Kim K, Doi A, Wen B, Ng K, Zhao R, Cahan P, Kim J, Aryee MJ, Ji H, Ehrlich LIR, Yabuuchi A, Takeuchi A, Cunniff KC, Hongguang H, Mckinney-Freeman S, Naveiras O, Yoon TJ, Irizarry RA, Jung N, Seita J, et al. (2010) Epigenetic memory in induced pluripotent stem cells. Nature 467:285-290.

Lagasse E, Connors H, Al Dhalimy M, Reitsma M, Dohse M, Osborne L, Wang X, Finegold M, Weissman IL, Grompe M (2000) Purified hematopoietic stem cells can diff erentiate into hepatocytes in vivo. Nat Med 6:1229-1234.

Lin TX, Ambasudhan R, Yuan X, Li WL, Hilcove S, Abujarour R, Lin XY, Hahm HS, Hao E, Hayek A, Ding S (2009) A chemical platform for improved induction of human iPSCs. Nat Methods 6:805-808.

Lister R, Pelizzola M, Kida YS, Hawkins RD, Nery JR, Hon G, Antosiewicz-Bourget J, O’Malley R, Castanon R, Klugman S, Downes M, Yu R, Stewart R, Ren B, Thomson JA, Evans RM, Ecker JR (2011) Hotspots of aberrant epigenomic reprogramming in human induced pluripotent stem cells. Nature 471:68-73.

Luna-Zurita L, Bruneau BG (2013) Chromatin modulators as facilitating factors in cellular reprogramming. Curr Opin Genet Dev 23:556-561.

Martic G, Karetsou Z, Kefala K, Politou AS, Clapier CR, Straub T, Papamarcaki T (2005) Parathymosin aff ects the binding of linker histone H1 to nucleosomes and remodels chromatin structure. J Biol Chem 280:16143-16150.

Miura M, Miura Y, Padilla-Nash HM, Molinolo AA, Fu B, Patel V, Seo BM, Sonoyama W, Zheng JJ, Baker CC, Chen W, Ried T, Shi S (2006) Accumulated chromosomal instability in murine bone marrow mesenchymal stem cells leads to malignant transformation. Stem Cells 24:1095-1103. Murry CE, Keller G (2008) Diff erentiation of embryonic stem cells to clinically relevant populations: Lessons from embryonic development. Cell 132:661-680.

Najm FJ, Lager AM, Zaremba A, Wyatt K, Caprariello AV, Factor DC, Karl RT, Maeda T, Miller RH, Tesar PJ (2013) Transcription factor-mediated reprogramming of fi broblasts to expandable, myelinogenic oligodendrocyte progenitor cells. Nat Biotechnol 31:426-433.

Nishiyama A (2007) Polydendrocytes: NG2 cells with many roles in development and repair of the CNS. Neuroscientist 13:62-76.

Nishiyama A, Komitova M, Suzuki R, Zhu X (2009) Polydendrocytes (NG2 cells): multifunctional cells with lineage plasticity. Nat Rev Neurosci 10:9-22.

Nishiyama A, Lin XH, Giese N, Heldin CH, Stallcup WB (1996) Co-localization of NG2 proteoglycan and PDGF alpha-receptor on O2A progenitor cells in the developing rat brain. J Neurosci Res 43:299-314.

Niu W, Zang T, Zou Y, Fang S, Smith DK, Bachoo R, Zhang CL (2013) In vivo reprogramming of astrocytes to neuroblasts in the adult brain. Nat Cell Biol 15:1164-1175.

Park IH, Arora N, Huo H, Maherali N, Ahfeldt T, Shimamura A, Lensch MW, Cowan C, Hochedlinger K, Daley GQ (2008a) Disease-specifi c induced pluripotent stem cells. Cell 134:877-886.

Park IH, Lerou PH, Zhao R, Huo HG, Daley GQ (2008b) Generation of human-induced pluripotent stem cells. Nat Protocols 3:1180-1186.

Pfi sterer U, Kirkeby A, Torper O, Wood J, Nelander J, Dufour A, Bjorklund A, Lindvall O, Jakobsson J, Parmar M (2011) Direct conversion of human fi broblasts to dopaminergic neurons. Proc Natl Acad Sci U S A 108:10343-10348. Philpott A, Leno GH, Laskey RA (1991) Sperm decondensation in Xenopus egg cytoplasm is mediated by nucleoplasmin. Cell 65:569-578.

Sadowski D, Kiel ME, Apicella M, Arriola AG, Chen CP, McKinnon RD (2010) Teratogenic potential in cultures optimized for oligodendrocyte development from mouse embryonic stem cells. Stem Cells Dev 19:1343-1353.

Sekiya S, Suzuki A (2011) Direct conversion of mouse fi broblasts to hepatocyte-like cells by defi ned factors. Nature 475:390-393.

Simonsson S, Gurdon J (2004) DNA demethylation is necessary for the epigenetic reprogramming of somatic cell nuclei. Nat Cell Biol 6:984-990. Smale ST (2010) Pioneer factors in embryonic stem cells and diff erentiation. Curr Opin Genet Dev 20:519-526.

Soldner F, Hockemeyer D, Beard C, Gao Q, Bell GW, Cook EG, Hargus G, Blak A, Cooper O, Mitalipova M, Isacson O, Jaenisch R (2009) Parkinson’s disease patient-derived induced pluripotent stem cells free of viral reprogramming factors. Cell 136:964-977.

Son EY, Ichida JK, Wainger BJ, Toma JS, Rafuse VF, Woolf CJ, Eggan K (2011) Conversion of mouse and human fi broblasts into functional spinal motor neurons. Cell Stem Cell 9:205-218.

Szabo E, Rampalli S, Risueno RM, Schnerch A, Mitchell R, Fiebig-Comyn A, Levadoux-Martin M, Bhatia M (2010) Direct conversion of human fi broblasts to multilineage blood progenitors. Nature 468:521-526.

Takahashi K, Yamanaka S (2006) Induction of pluripotent stem cells from mouse embryonic and adult fi broblast cultures by defi ned factors. Cell 126:663-676.

Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, Yamanaka S (2007) Induction of pluripotent stem cells from adult human fi broblasts by defi ned factors. Cell 131:861-872.

Tolar J, Nauta AJ, Osborn MJ, Panoskaltsis MA, McElmurry RT, Bell S, Xia L, Zhou N, Riddle M, Schroeder TM, Westendorf JJ, McIvor RS, Hogendoorn PC, Szuhai K, Oseth L, Hirsch B, Yant SR, Kay MA, Peister A, Prockop DJ, et al. (2007) Sarcoma derived from cultured mesenchymal stem cells. Stem Cells 25:371-379.

Vierbuchen T, Ostermeier A, Pang ZP, Kokubu Y, Sudhof TC, Wernig M (2010) Direct conversion of fi broblasts to functional neurons by defi ned factors. Nature 463:1035-1041.

Wang X, Willenbring H, Akkari Y, Torimaru Y, Foster M, Al Dhalimy M, Lagasse E, Finegold M, Olson S, Grompe M (2003) Cell fusion is the principal source of bone-marrow-derived hepatocytes. Nature 422:897-901. Weider M, Wegener A, Schmitt C, Kuspert M, Hillgartner S, Bosl MR, Hermans-Borgmeyer I, Nait-Oumesmar B, Wegner M (2015) Elevated in vivo levels of a single transcription factor directly convert satellite glia into oligodendrocyte-like cells. PLoS Genet 11:e1005008.

Windrem MS, Schanz SJ, Guo M, Tian GF, Washco V, Stanwood N, Rasband M, Roy NS, Nedergaard M, Havton LA, Wang S, Goldman SA (2008) Neonatal chimerization with human glial progenitor cells can both remyelinate and rescue the otherwise lethally hypomyelinated shiverer mouse. Cell Stem Cell 2:553-565.

Woodbury D, Schwarz EJ, Prockop DJ, Black IB (2000) Adult rat and human bone marrow stromal cells diff erentiate into neurons. J Neurosci Res 61:364-370.

Yamanaka S (2009) Elite and stochastic models for induced pluripotent stem cell generation. Nature 460:49-52.

Yang N, Zuchero JB, Ahlenius H, Marro S, Ng YH, Vierbuchen T, Hawkins JS, Geissler R, Barres BA, Wernig M (2013) Generation of oligodendroglial cells by direct lineage conversion. Nat Biotechnol 31:434-439.

Yu JY, Hu KJ, Smuga-Otto K, Tian SL, Stewart R, Slukvin II, Thomson JA (2009) Human induced pluripotent stem cells free of vector and transgene sequences. Science 324:797-801.

Zhou Q, Brown J, Kanarek A, Rajagopal J, Melton DA (2008) In vivo reprogramming of adult pancreatic exocrine cells to beta-cells. Nature 455:627-U30.

Zhu SY, Li WL, Zhou HY, Wei WG, Ambasudhan R, Lin TX, Kim J, Zhang K, Ding S (2010) Reprogramming of human primary somatic cells by OCT4 and chemical compounds. Cell Stem Cell 7:651-655.

*Correspondence to:

Randall D. McKinnon, Ph.D.,

mckinnon@rwjms.rutgers.edu.

# These authors contributed equally to this work.

10.4103/1673-5374.165209

http://www.nrronline.org/

Accepted: 2015-06-19