Elsevier

Progress in Neurobiology

Volume 134, November 2015, Pages 161-177
Progress in Neurobiology

Human induced pluripotent stem cells in Parkinson's disease: A novel cell source of cell therapy and disease modeling

https://doi.org/10.1016/j.pneurobio.2015.09.009Get rights and content

Highlights

  • iPSCs are novel cell sources for cell therapy, drug screening and disease modeling.

  • Virus-free approaches combined with small molecules promote reprogramming of iPSCs.

  • Dopaminergic differentiation is improved by modulation of signaling pathways.

Abstract

Human induced pluripotent stem cells (hiPSCs) and human embryonic stem cells (hESCs) are two novel cell sources for studying neurodegenerative diseases. Dopaminergic neurons derived from hiPSCs/hESCs have been implicated to be very useful in Parkinson's disease (PD) research, including cell replacement therapy, disease modeling and drug screening. Recently, great efforts have been made to improve the application of hiPSCs/hESCs in PD research. Considerable advances have been made in recent years, including advanced reprogramming strategies without the use of viruses or using fewer transcriptional factors, optimized methods for generating highly homogeneous neural progenitors with a larger proportion of mature dopaminergic neurons and better survival and integration after transplantation. Here we outline the progress that has been made in these aspects in recent years, particularly during the last year, and also discuss existing issues that need to be addressed.

Introduction

Parkinson's disease (PD) is the most common movement disorder in man. Loss of dopaminergic neurons in the substantia nigra pars compacta is the pathological hallmark of the disease. Therapies for PD have been mainly restricted to the relief of symptoms and there is still no effective and reliable model for studying the mechanisms of the disease.

Since the 1980s, several open-labeled trials have been performed to transplant human fetal dopaminergic neurons into the putamen and/or caudate nucleus as a replacement therapy to restore dopaminergic transmission (Lindvall et al., 1989, Sawle et al., 1992). Concrete evidence shows that neural transplants survive and become functional and integrated with the host brain neurons (Lindvall et al., 1994, Wenning et al., 1997). Fluoro-Dopa PET scans demonstrate that grafted neurons can actively take up dopamine, reflecting functional neurons in the transplant; the patients show benefits in general performance, reduced rigidity and increased speed of movement (Piccini et al., 1999, Piccini et al., 2000). Post-mortem studies of the brain tissues that received neural transplantation demonstrate the long-term survival of dopaminergic neurons, even longer than two decades post-operation (Fig. 1). These open-labeled clinical trials show that, when successful, dopaminergic neuron transplantation can be beneficial and promising to PD patients. However, two NIH-sponsored double-blind placebo trials failed to reach the primary outcome, i.e. a significant benefit to the patients (Freed et al., 2001, Olanow et al., 2003). Furthermore, a considerable number of transplanted patients developed clear adverse effects – graft-induced dyskinesia. After intensive debates on clinical trial designs of cell replacement therapy, including patient selection, graft tissue preparation and processing and optimization of surgical procedures, a new clinical trial (TRANSEURO) was established and is currently ongoing cross-continentally in multi-research and clinical centers (Barker et al., 2013). Success of this trial will have a major impact on neural transplantation with fetal dopaminergic tissues, and also with human embryonic stem cell (hESC)- and human induced pluripotent stem cell (hiPSC)-derived dopaminergic neurons in future.

Embryonic stem cells (ESCs) provide hope for regenerative medicine, and have been proposed as a source of donor cells for replacement therapy in PD. ESCs are pluripotent; they have a wide differentiation potential to generate tissues and cells derived from all three embryonic germ layers. Mouse ESCs can be differentiated into dopaminergic neurons (Roy et al., 2006, Kawasaki et al., 2000) with efficient survival rates and can give rise to functional recovery after transplantation into the brains of rodent models of PD (Bjorklund et al., 2002, Kim et al., 2006). However, it has been difficult and complicated to generate high yields of dopaminergic neurons from hESCs with various differentiation protocols. Furthermore, following intra-cerebral transplantation, the survival of transplants and functional effects have not been satisfactory, even with reports of tumor or teratoma formation (Roy et al., 2006, Brederlau et al., 2006). HiPSCs (Park et al., 2008a) are promising potential cell sources for studying neurodegenerative diseases and may, in the future, be used for cell therapy as well. From the perspective of the differentiation potential, hiPSCs encounter similar problems to hESCs. The application of hiPSCs/hESCs in PD research has been substantially limited by the lack of effective protocols for differentiation and transplantation. A series of studies have been undertaken to optimize protocols of hiPSC/hESC differentiation and transplantation; although improvements have been made in the last few years, many problems still exist. Recently, a breakthrough of differentiating hESCs in vitro broke the deadlock (Fasano et al., 2010, Kirkeby et al., 2012). Here we will discuss advances that have been made in terms of dopaminergic conversion from hiPSCs/hESCs and transplantation studies in recent years and summarize the state-of-the-art development and prospects for effective and safe use of hiPSCs/hESCs for PD therapy in the future. We will keep to the following outline: hiPSCs/hESCs  neural progenitors  mature dopaminergic neurons  transplantation, and specify the progress that has been made in each of these aspects, including advanced reprogramming strategies without the use of viruses or using fewer transcriptional factors (Hiratsuka et al., 2011, Kim et al., 2009a, Rhee et al., 2011, Tsai et al., 2011), the optimal methods for generating highly homogeneous neural progenitors and a greater proportion of mature dopaminergic neurons (Kirkeby et al., 2012, Cooper et al., 2010, Chambers et al., 2009, Kriks et al., 2011, Morizane et al., 2010, Pang et al., 2011). Furthermore, we will focus on the survival, integration and safety issues regarding teratoma/tumor formation after intra-cerebral transplantation. This review will provide a timely highlight of recent advances and a better understanding of the use of hiPSCs/hESCs in cell therapy and in disease mechanism studies of PD for both clinical neurologists and researchers.

Section snippets

The original protocol – viral vector approach

Traditionally, reprogramming of somatic cells to pluripotency has been achieved by two different methods. Firstly, nuclear transfer – transplanting nuclei from differentiated somatic cells into oocytes. Secondly, cell fusion – involving fusion of two or more cells into one, which reveals the fact that silent genes in differentiated cells can be activated by certain regulators (Yamanaka and Blau, 2010).

In 2006, Yamanaka and co-workers showed that somatic cells can be reprogrammed into an

Sufficient dopaminergic differentiation

Degeneration of dopaminergic neurons in the substantia nigra is the pathological hallmark of PD. Using iPSCs for cell replacement therapy has been considered as a promising approach for the treatment of PD. However, the realization of this potential has been largely hindered by several problems, especially the low efficiency of generating a large quantity and well-defined population of dopaminergic neurons from iPSCs (Li et al., 2008). Previous discussions regarding differentiation efficiency

Survival of transplanted cells versus phenotypic stability

Together with advanced differentiation protocols, the result of research into transplantation has been greatly improved. Dopaminergic neurons, or dopaminergic neural progenitors, can be transplanted at a very early stage (for example, day 10 (Kirkeby et al., 2012)) with an improved survival rate and lower tumor formation. Also, the grafts after transplantation show promising integration with the host brain (Lee et al., 2000, Wernig et al., 2008). Below, we summarize the cell lines,

Unique models to study disease mechanisms

A number of cell and animal models have been used in PD research. Among these models, more and more genetically modified models, either knockout or transgenic, expressing or silencing different PD related genes, have been generated. Use of these models substantially deepens our understanding of PD pathogenesis and helps us to search for disease modifiers and novel targets for possible therapeutic intervention. These models have played important roles in the progress of PD research. However,

Perspective

Here, we have summarized recent developments regarding the application of iPSCs and ESCs in PD research, following the line of: hiPSCs/hESCs  neural progenitors  neural progenitor with dopaminergic identity  transplantation. Many remarkable achievements have been made. We are currently at a stage of generating dopaminergic neurons from PD derived-iPSCs using relatively rapid and feeder cell-free, and most importantly, more highly efficient protocols, which are based on a better understanding of

Acknowledgements

We would like to acknowledge financial support by the National Key Basic Research Program of China (973 Program – no. 2010CB945203, 2011CB504104) (SD Chen) and the National Natural Science Foundation (81371407, SD Chen; 81430025 JY Li). Acknowledgements are also to the supports of the Swedish Research Council, BAGADILICO-Excellence in Parkinson and Huntington Research, Swedish Parkinson Foundation, Swedish Brain Foundation, MJ Fox Foundation for Parkinson's Research and ERA-Net Neuron Program

References (212)

  • S. Chung et al.

    The homeodomain transcription factor Pitx3 facilitates differentiation of mouse embryonic stem cells into AHD2-expressing dopaminergic neurons

    Mol. Cell. Neurosci.

    (2005)
  • O. Cooper et al.

    Differentiation of human ES and Parkinson's disease iPS cells into ventral midbrain dopaminergic neurons requires a high activity form of SHH, FGF8a and specific regionalization by retinoic acid

    Mol. Cell. Neurosci.

    (2010)
  • D.D. De Carvalho et al.

    DNA methylation and cellular reprogramming

    Trends Cell Biol.

    (2010)
  • C.A. Fasano et al.

    Efficient derivation of functional floor plate tissue from human embryonic stem cells

    Cell Stem Cell

    (2010)
  • B. Feng et al.

    Molecules that promote or enhance reprogramming of somatic cells to induced pluripotent stem cells

    Cell Stem Cell

    (2009)
  • P. Guha et al.

    Lack of immune response to differentiated cells derived from syngeneic induced pluripotent stem cells

    Cell Stem Cell

    (2013)
  • A. Hemmati-Brivanlou et al.

    Follistatin, an antagonist of activin, is expressed in the Spemann organizer and displays direct neuralizing activity

    Cell

    (1994)
  • W. Herzog et al.

    Adenohypophysis formation in the zebrafish and its dependence on sonic hedgehog

    Dev. Biol.

    (2003)
  • K. Hochedlinger et al.

    Ectopic expression of Oct-4 blocks progenitor-cell differentiation and causes dysplasia in epithelial tissues

    Cell

    (2005)
  • W. Hu et al.

    Activation of aryl hydrocarbon receptor (ahr) by tranilast, an anti-allergy drug, promotes miR-302 expression and cell reprogramming

    J. Biol. Chem.

    (2013)
  • M. Hynes et al.

    Induction of midbrain dopaminergic neurons by Sonic hedgehog

    Neuron

    (1995)
  • J.K. Ichida et al.

    A small-molecule inhibitor of tgf-Beta signaling replaces sox2 in reprogramming by inducing nanog

    Cell Stem Cell

    (2009)
  • S. Kanda et al.

    Over-expression of bHLH genes facilitate neural formation of mouse embryonic stem (ES) cells in vitro

    Int. J. Dev. Neurosci.

    (2004)
  • H. Kawasaki et al.

    Induction of midbrain dopaminergic neurons from ES cells by stromal cell-derived inducing activity

    Neuron

    (2000)
  • T.E. Kim et al.

    Sonic hedgehog and FGF8 collaborate to induce dopaminergic phenotypes in the Nurr1-overexpressing neural stem cell

    Biochem. Biophys. Res. Commun.

    (2003)
  • J.B. Kim et al.

    Oct4-induced pluripotency in adult neural stem cells

    Cell

    (2009)
  • D. Kim et al.

    Generation of human induced pluripotent stem cells by direct delivery of reprogramming proteins

    Cell Stem Cell

    (2009)
  • A. Kirkeby et al.

    Generation of regionally specified neural progenitors and functional neurons from human embryonic stem cells under defined conditions

    Cell Rep.

    (2012)
  • J.Y. Ko et al.

    Conditions for tumor-free and dopamine neuron-enriched grafts after transplanting human ES cell-derived neural precursor cells

    Mol. Ther.

    (2009)
  • T. Kondo et al.

    Modeling Alzheimer's disease with iPSCs reveals stress phenotypes associated with intracellular Abeta and differential drug responsiveness

    Cell Stem Cell

    (2013)
  • N.J. Larsen et al.

    DJ-1 knock-down impairs astrocyte mitochondrial function

    Neuroscience

    (2011)
  • L.C. Laurent et al.

    Dynamic changes in the copy number of pluripotency and cell proliferation genes in human ESCs and iPSCs during reprogramming and time in culture

    Cell Stem Cell

    (2011)
  • H.J. Lee et al.

    Direct transfer of alpha-synuclein from neuron to astroglia causes inflammatory responses in synucleinopathies

    J. Biol. Chem.

    (2010)
  • J.Y. Li et al.

    Critical issues of clinical human embryonic stem cell therapy for brain repair

    Trends Neurosci.

    (2008)
  • W. Li et al.

    Generation of rat and human induced pluripotent stem cells by combining genetic reprogramming and chemical inhibitors

    Cell Stem Cell

    (2009)
  • G. Liang et al.

    Butyrate promotes induced pluripotent stem cell generation

    J. Biol. Chem.

    (2010)
  • S. Appel-Cresswell et al.

    Alpha-synuclein p.H50Q, a novel pathogenic mutation for Parkinson's disease

    Mov. Disord.

    (2013)
  • V. Bagga et al.

    Ascorbic acid increases the number of dopamine neurons in vitro and in transplants to the 6-OHDA-lesioned rat brain

    Cell Transplant.

    (2008)
  • J. Beers et al.

    A cost-effective and efficient reprogramming platform for large-scale production of integration-free human induced pluripotent stem cells in chemically defined culture

    Sci. Rep.

    (2015)
  • J.E. Beevers et al.

    Induced pluripotent stem cell (iPSC)-derived dopaminergic models of Parkinson's disease

    Biochem. Soc. Trans.

    (2013)
  • N. Bhutani et al.

    Reprogramming towards pluripotency requires AID-dependent DNA demethylation

    Nature

    (2010)
  • L.M. Bjorklund et al.

    Embryonic stem cells develop into functional dopaminergic neurons after transplantation in a Parkinson rat model

    Proc. Natl. Acad. Sci. U. S. A.

    (2002)
  • V. Bonifati et al.

    Mutations in the DJ-1 gene associated with autosomal recessive early-onset parkinsonism

    Science

    (2003)
  • A. Brederlau et al.

    Transplantation of human embryonic stem cell-derived cells to a rat model of Parkinson's disease: effect of in vitro differentiation on graft survival and teratoma formation

    Stem Cells

    (2006)
  • K.A. Buytaert-Hoefen et al.

    Generation of tyrosine hydroxylase positive neurons from human embryonic stem cells after coculture with cellular substrates and exposure to GDNF

    Stem Cells

    (2004)
  • B. Byers et al.

    SNCA triplication Parkinson's patient's iPSC-derived DA neurons accumulate alpha-synuclein and are susceptible to oxidative stress

    PLoS ONE

    (2011)
  • G. Castelo-Branco et al.

    Differential regulation of midbrain dopaminergic neuron development by Wnt-1, Wnt-3a, and Wnt-5a

    Proc. Natl. Acad. Sci. U. S. A.

    (2003)
  • G. Castelo-Branco et al.

    GSK-3beta inhibition/beta-catenin stabilization in ventral midbrain precursors increases differentiation into dopamine neurons

    J. Cell Sci.

    (2004)
  • S.M. Chambers et al.

    Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling

    Nat. Biotechnol.

    (2009)
  • Y.L. Chang et al.

    Docosahexaenoic acid promotes dopaminergic differentiation in induced pluripotent stem cells and inhibits teratoma formation in rats with Parkinson-like pathology

    Cell Transplant.

    (2012)
  • Cited by (28)

    • Pluripotent stem cell-based therapy for Parkinson's disease: Current status and future prospects

      2018, Progress in Neurobiology
      Citation Excerpt :

      It opens the door to the plausible use of iPSCs for personalized as well as generalized cell therapy. The enormous progress in iPSC research over the last decade has been extensively discussed in a series of recent reviews (Hockemeyer and Jaenisch, 2016; Karagiannis and Eto, 2016; Li and Izpisua Belmonte, 2016; Li et al., 2015; Mertens et al., 2016; Scudellari, 2016; Takahashi and Yamanaka, 2016; Tapia and Scholer, 2016). However, as pointed out in a recent commentary (Scudellari, 2016), the major goals of most iPSC research have switched away from personalized cell therapy, and instead focus on mechanistic studies of human disease and development.

    • Combining NT3-overexpressing MSCs and PLGA microcarriers for brain tissue engineering: A potential tool for treatment of Parkinson's disease

      2017, Materials Science and Engineering C
      Citation Excerpt :

      The steady motor improvement of hemiparkinsonian rat in vivo with no significant adverse effects after MSCs transplantation has been also reported [19]. Hence, many researchers have recently utilized MSCs as cell source for treatment of PD since they can be differentiated into neuron like cells before in vivo transplantation [12,20–22]. The use of chemical cues [23], cytokines [24], co-culture with nerve cells [25], and transfection of genes responsible for differentiation of MSCs to DA neurons [26] have been recently reported.

    • RNAi-mediated silencing of HLA A2 suppressed acute rejection against human fibroblast xenografts in the striatum of 6-OHDA lesioned rats

      2016, Journal of Neuroimmunology
      Citation Excerpt :

      However, xenografts of dopaminergic neurons derived from human embryonic stem cells in cyclosporine (CsA)-suppressed rhesus brain were mostly rejected (Emborg et al., 2013). Human iPS cells converted from patient-specific somatic cells compared to allogeneic cells were found to result in a much lower immune response and free of ethical issues of human embryonic stem cells (Li et al., 2015). iPS cells derived from monkey skin fibroblasts (Hallett et al., 2015) or human skin fibroblasts (Han et al., 2015) can be further induced to differentiate into neurons and dopaminergic neurons, and subsequent autologous or xenogeneic transplantation into the striatum improved Parkinsonian symptoms of PD animals.

    • Blood-brain barrier dysfunction and recovery after ischemic stroke

      2018, Progress in Neurobiology
      Citation Excerpt :

      It is, therefore, important that human in vitro BBB models be developed. Recently, Shusta and colleagues have used human induced pluripotent stem cells (iPSCs) to generate brain endothelial cells and other NVU cells (Li et al., 2015b; Lippmann et al., 2014; Lippmann et al., 2012). They have been used to create humanized in vitro BBB models with TEERs close to those in vivo (Lippmann et al., 2014).

    View all citing articles on Scopus
    View full text