Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Mar 13.
Published in final edited form as: Int J High Throughput Screen. 2011 Jun;2011(2):15–25. doi: 10.2147/IJHTS.S8681

Drug discovery in Parkinson’s disease—Update and developments in the use of cellular models

Gaia Skibinski 1,2, Steven Finkbeiner 1,2,3,4
PMCID: PMC3596173  NIHMSID: NIHMS435741  PMID: 23505333

Abstract

Parkinson’s disease (PD) is the second most common neurodegenerative disorder and is characterized by the degeneration of dopaminergic (DA) neurons within the substantia nigra. Dopamine replacement drugs remain the most effective PD treatment but only provide temporary symptomatic relief. New therapies are urgently needed, but the search for a disease-modifying treatment and a definitive understanding of the underlying mechanisms of PD has been limited by the lack of physiologically relevant models that recapitulate the disease phenotype. The use of immortalized cell lines as in vitro model systems for drug discovery has met with limited success, since efficacy and safety too often fail to translate successfully in human clinical trials. Drug discoverers are shifting their focus to more physiologically relevant cellular models, including primary neurons and stem cells. The recent discovery of induced pluripotent stem (iPS) cell technology presents an exciting opportunity to derive human DA neurons from patients with sporadic and familial forms of PD. We anticipate that these human DA models will recapitulate key features of the PD phenotype. In parallel, high-content screening platforms, which extract information on multiple cellular features within individual neurons, provide a network-based approach that can resolve temporal and spatial relationships underlying mechanisms of neurodegeneration and drug perturbations. These emerging technologies have the potential to establish highly predictive cellular models that could bring about a desperately needed revolution in PD drug discovery.

Drug discovery in Parkinson’s disease

Parkinson’s disease (PD), the second most common neurodegenerative disorder, is characterized by motor symptoms, including rigidity, bradykinesia and tremor. These symptoms are the direct result of a nigrostriatal dopamine deficit, due to the selective loss of dopaminergic (DA) neurons in the substantia nigra. Other prominent neuropathological features include dystrophic neurites and intracellular proteinaceous inclusions called Lewy bodies (LB)[1]. Approximately 1–2% of those over the age of 65 suffer from PD, and the incidence increases with age[2]. The present annual cost of healthcare for PD patients is estimated to exceed US $5.6 billion in the United States (National Institute of Neurological Disorders and Stroke). With the rapid increase in worldwide life expectancy, the prevalence of PD is expected to double by 2030[3].

The selective loss of DA neurons in the substantia nigra has focused treatments towards dopamine-replacement drugs, such as Levodopa (L-dopa). Although it was identified almost 45 years ago[4], L-dopa remains the principle and most effective treatment for PD. However, as the disease progresses and more DA neurons are lost, the efficacy of L-dopa diminishes, and patients experience increasing fluctuations in motor symptoms, including dyskinesias. Furthermore, dopamine-replacement drugs fail to address the degeneration observed in other brain areas, such as the locus coeruleus and cerebral cortex[5, 6], and the wide range of autonomic symptoms noted in patients with PD[7]. Ultimately, disease-modifying treatments are needed that address both the motor and non-motor symptoms of PD.

Despite increasing investment in biomedical research by industry and government[8], the success rate of new drugs in clinical trials is dropping[9]. Drug discovery in PD is no exception, and promising new compounds validated at the preclinical stage often fail during development and expensive clinical trials[10]. For example, Sarizotan, an anti-dyskinesia drug, was successful in preclinical and phase II trials, yet failed in phase III trials[11]. These are costly failures: discovering and developing a new drug is now estimated at US $800 million[12]. Attrition rates of late-stage drug candidates could be modestly reduced by improving the design of clinical trials[13]. However, academic and industry scientists acknowledge that, ultimately, to increase the success rate and reduce the financial burden of drug discovery, improvements should be focused on the drug discovery pipeline itself. In particular, they should be focused on identifying and validating relevant targets and characterizing candidate drugs[14].

During the last 20 years, cellular models of neurodegenerative diseases have undergone many evolutionary changes. Conventional drug discovery strategies utilized genetically engineered and immortalized cell lines to develop cell-based assays. In these systems, the cell type is subservient to the limitations of the assay technology. The focus is now shifting towards developing cell-based assays that more faithfully recapitulate characteristics of the disease phenotype, leading to better target identification and predictions of drug efficacy and toxicity. Consequently, primary neurons and stem cells (SCs) are increasingly recognized as powerful tools for drug discovery[14]. Parallel developments in high through-put screening (HTS) and high-content screening (HCS) platforms, which capture multiparameteric features within single cells, have enabled cellular signatures to be defined for disease states and toxic activities of compounds that are screened[15].

Modeling PD for drug discovery: Too many targets

Drugs against novel targets are estimated to have a 50% greater failure rate and create less value than those developed against validated targets[16]. As a result, much of the drug discovery in PD has focused on enhancing the efficacy of L-dopa and minimizing its side effects[17]. However L-dopa provides only symptomatic improvement and has little effect on the multisystem neuronal dysfunction and deterioration that occur in PD. Novel drug targets are needed if we are to develop effective disease modifying treatments for PD. To date, no single mechanism or cause of sporadic PD, which makes up the vast majority of PD, has been identified. However, insights into mechanisms of PD pathogenesis have emerged from mutations in several genes that associate with familial forms of PD. They encode leucine-rich repeat kinase 2 (LRRK2)[18, 19], α-synuclein[20], parkin[21], DJ-1[22], (PTEN)-induced kinase 1 (PINK1)[23] and ATP13A2[24] (Table 1). These genes implicate several cellular processes as potentially involved with familial and sporadic PD, including oxidative stress, mitochondrial dysfunction, pertubations in proteostasis, inflammation and protein phosphorylation[25, 26]. It is unknown which mechanism or risk factor, if any, plays the dominant role in PD. As a result, the validation and exploration of these targets remain a significant bottle-neck in the PD drug discovery pipeline.

Table 1.

Genetic loci associated with PD

PARK loci Gene Types of PD Mutations Function
PARK1/PARK4 SNCA AD, early onset and sporadic A53T, A30P, E46K, duplications and triplications
~2% of familial parkinsonism[109]
Pre-synaptic protein implicated in neurotransmitter release[122]
PARK2 Parkin AR, juvenile and early onset and sporadic >100 mutations
almost 50% of early onset AR PD, ~20% isolated juvenile PD[123]
Ubiquitin ligase, targets protein to the proteasome for degradation[124] and recruited to depolarized mitochondria to aid mitophagy[125]
PARK6 PINK1 AR, early onset >40 point mutations, rare large deletions
1–9% early onset [126]
Preserve mitochondria integrity[127] and required for Parkin mediated mitophagy[128]
PARK7 DJ1 AR, early onset >10 point mutations and large deletion
<1% early onset
Redox-sensitive molecular chaperone preventing aggregation of α-synuclein[38]
PARK8 LRRK2 AD, late onset and sporadic >40 missense mutations, at least 7 are pathogenic
10% AD familial cases and 3.6% sporadic PD [129]
Kinase and GTPase activity. Implicated in signaling pathways including apoptosis, regulation of cytoskeleton, MAPK signaling and protein translation[130]
PARK9 ATP13A2 AR, juvenile Kufor-Rakeb syndrome and early onset PD >5 point mutations Lysosomal P type ATPase[24]

AD, autosomal dominant; AR, autosomal recessive

Defining the mechanisms of neurodegeneration in PD has been hampered by our limited access to the specific neuronal populations most affected by the disease. Pathological studies on postmortem tissue from PD patients provided hints about the cellular events underlying neurodegeneration in PD, such as LB formation[27] and mitochondrial dysfunction[28]. However, these are only snapshots of cellular events at the end of the disease progression, when most of the DA neurons have already been destroyed. Furthermore, there is no way of knowing which of these cellular features are primary or secondary to the initial pathogenic insult, and which are incidental events or homeostatic responses exhibited by injured neurons[29].

Animal and cellular models attempt to recapitulate various aspects of the PD phenotype, with mixed success. Animal models of PD have been generated by injecting neurotoxins and by transgenic expression of PD-associated mutations[30]. Toxin models induce nigro-striatal cell loss, one of the key pathological traits of PD. They have been criticized because the rate of neurodegeneration is far greater than that of humans with PD[31]. However, continuous low-level administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mimics more progressive behavioral changes and inclusion formation characteristic of PD[32]. In the past, animal models harboring genetic mutations that mimic inherited forms of PD have been limited in their ability to recapitulate the classical features of PD[30]. More recently, transgenic mouse models involving the expression of α-synuclein and LRRK2 developed distinct behavioral changes, DA cell loss and pathological inclusions reminiscent of PD[33, 34]. Although animal models are often used to validate the efficacy and safety of novel drugs, they are of limited use in unraveling the complexities of cellular mechanisms and impractical for HTS and other drug development stages, such as toxicological dose-response studies.

Cell Models for Drug Development

This review describes current cell models that have been developed to recapitulate traits of PD pathogenesis and their applicability to HTS. We will also describe emerging cellular technologies that have the potential to significantly improve drug discovery.

Immortalized cell lines

Traditionally, recombinant immortalized cells have been a popular choice for drug discovery in cell-based assays[35]. These cells can be grown in virtually unlimited quantities, are not labor intensive and can be stored frozen to limit replicative senescence[36]. They provide a relatively homogenous target expression system that leads to less variable screening and can be genetically manipulated to express or knock down a target or express a reporter. Several cell lines display characteristics of DA neurons (Table 2) and have provided valuable insights into cellular mechanisms of PD.

Table 2.

Sources and examples of immortalized cell lines used in PD research

Source Examples Differentiation Dopaminergic like properties
Mouse CNS (catecholaminergic) CAD Serum deprivation Displays neurites and expresses TH
Mouse Fusion of embryonic mid brain and neuroblastoma cell line MN9D N-butyric acid Expresses TH and synthesizes, stores and releases DA
Rat Mid brain N27 Db-cAMP and DHEA Displays neurites and expresses TH and DAT
Rat Pheochromocytoma PC12 Nerve growth factor Neuronal processes and produces DA
Human Neuroblastoma SHSY5Y, SK-N-SH, SK-N-MC Retinoic acid or TPA Display neurites and expresses TH, DBH and DAT
Human Carcinoma Ntera2 Retinoic acid Expresses TH, DAT and dopamine D2 receptor
Human Embryonic ventral mid brain MESC2.10 Retracycline, db-cAMP and GDNF Exhibits neurites, electrically active, expresses TH, DAT and produces DA
Human Fetal ventral mid brain ReNcell VM Db-cAMP and GDNF Expresses TH and displays electrically active potentials.

Central nervous system (CNS), Cath.A derived (CAD), dopamine (DA), dopamine transporter (DAT), dibutyryl cyclic AMP (db-cAMP), dopamine-beta-hydroxylase (DBH), dehydroepiandrosterone (DHEA), glial cell line-derived neurotrophic factor (GDNF), 12-O-tetradecanoylphorbol-13-acetate (TPA), tyrosine hydroxylase (TH).

Human neuroblastoma cell lines, including SHSY5Y, SK-N-SH and SK-N-MC, have often been used to model cellular traits of PD, including the formation of LBs[37]. Many studies have taken advantage of non-human cell lines to probe the mechanisms in PD. For example, a murine central nervous system (CNS) catecholaminergic-derived cell line was used to show that DJ-1, a protein involved in early onset parkinsonism[22], inhibits formation of α-synuclein aggregates[38]. A rat DA cell line, N27, was used to investigate the role of dimer formation in α-synuclein aggregation[39] and human mutant α-synuclein-mediated toxicity[40]. PC12 cells derived from a rat pheochromocytoma were used to investigate the role of autophagy and the proteasome degradation pathway during α-synuclein-mediated apoptosis[41].

Ultimately, however, these immortalized cell lines are not authentic DA neurons. A renewable source of human DA neurons is of particular interest for drug discovery and cell-transplantion therapy in PD. Attempts have been made to develop human midbrain cell lines. For example, MESC2.10 was derived from the ventral mescencephalon of an 8-week-old human embryo and immortalized by ectopic myc expression. Upon differentiation, these cells exhibit neurites, express markers of mature neurons and are electrically active. A subpopulation of these cells also expresses tyrosine hydroxylase (TH) and dopamine transporters (DATs), produces dopamine[42] and has been used to investigate the role of α-synuclein in the impairment of vesicular dopamine storage[42].

Despite the scalability and genetic malleability of these immortalized cell lines, their genetic and molecular phenotypes are significantly different from native DA neurons in vivo[43, 44]. Importantly, the effects of potential drugs or therapies on recombinant molecular targets expressed in immortalized cells may not be fully analogous to physiologic effects in vivo [45]. In fact, the success rate of compounds developed against CNS disorders is estimated to be only 8%, with lack of efficacy and safety being major reasons for this high attrition rate[46]. As a result, there is a growing need for the development of novel predictive cellular models.

Primary cells

Primary neurons are post-mitotic, differentiated cells and are functionally and morphologically more similar to those found in patients. Cellular phenotype is critical in the pharmacokinetics and pharmacodynamics of a drug compound, and this is particularly relevant to neurons, where the complex interplay among ion channels, intracellular signaling pathways and the regulation of gene expression might be essential for accurately identifying novel compounds. Consequently, primary cell systems are more effective at predicting human responses to novel drugs [47].

Primary DA neurons can be dissociated from the mid-brain of embryonic and post-natal rats or mice[48]. However, studies with embryonic cultures have been limited: DA neurons make up less than 1% of the total culture[49] and are immature. The characteristics they display in vitro vary significantly with the culture conditions[50]. In mid-brain cultures derived from post-natal animals, up to 50% of the cells are DA neurons, and these cells faithfully recapitulate electrophysiological and morphological properties of DA neurons in vivo[51]. Rodent primary DA neurons have been invaluable in studies of PD-specific toxicity and LB formation[52, 53]. Similar to immortalized cell lines, primary cells can be use to evaluate endogenous targets or be genetically modified to express recombinant targets or reporters. Primary mid-brain DA neurons have also been obtained from human fetal tissue. However, the limited availability and ethical implications of obtaining and using these cells makes them less attractive. Although most studies with human DA neurons have involved cell-replacement therapies as a treatment for PD patients[54, 55], these cells have been used to model the DA specificity of α-synuclein-mediated toxicity and LB formation[56].

The use of primary neurons for drug discovery permits the simultaneous analysis of multiple cell types. These cultures are significantly more physiologically relevant than immortalized cells[14], but primary DA cells have shortcomings, including their significant handling demands, the inability to freeze the cells, and greater expense as they are derived directly from animals. Furthermore, genetic manipulation is more difficult in primary neurons than in immortalized cell lines, and the variability is greater when cultures are prepared on different days and by different investigators. Of particular relevance to HTS, only small numbers of cells can be derived from each dissection. To some extent, these limitations have been outweighed by the development of automated fluorescence microscopy and high-content screening (HCS), which require fewer cells for each data point (described in more detail below), but logistical challenges remain.

Stem cells

Stem cells (SCs) have been used for drug discovery since the 1970s[57]. The use of SCs avoids several of the drawbacks of primary neurons[58]: they can be propagated indefinitely and grown to the required scale, similar to immortalized cell lines. Yet they retain pluripotency and can differentiate into neurons that are genetically and functionally analogous to those in vivo [59]. PD patients display a well-characterized selective loss of DA neurons in the mid-brain[60], and DA neurons derived from embryonic SCs (ESCs) and neural SCs (NSCs) are a renewable source for potential cell-replacement therapies. Furthermore, DA neurons derived from SCs could lead to more relevant and accurate models for studies on disease mechanisms and drug discovery.

Embryonic stem cells

ESCs are derived from the inner mass of the blastocyst. DA neurons have been differentiated from mouse ESCs by several protocols, including co-culturing with stromal PA-6 cells[61, 62] and embryoid body-based lineage selection[63]. DA neurons derived from mouse ESCs have pharmacological phenotypes similar to primary DA neurons[64]. Furthermore, transplantation of ES-derived neural precursors or DA neurons rescues behavioral deficits in animal PD models[6567]. Human ES cells (hESCs) have also been successfully differentiated into DA neurons in vitro [6870]. Compared to mouse ESCs, hESCs are more difficult to grow and expand, and their differentiation into DA specific neurons is less robust. For example, in hESC-derived DA neurons, TH expression decreases in vitro, and after transplantation, no TH-positive cells were detected, despite good survival [68]. However, recent progress has been made in developing more robust protocols for large-scale generation of DA neurons from hES cells[71] and differentiation of specific subtypes of DA neurons, including the substantia nigra A9 type (SN-A9), that appear to be more vulnerable to degeneration in PD[72].

Neural stem cells

DA neurons can also be derived from NSCs isolated from neuronal tissues in more advanced developmental stages than ESCs. NSCs have been cultured from embryonic, fetal and adult mammals, including humans [73, 74]. The limited accessibility to human tissue, as well as the difficulty in growing these cells and maintaining a stable phenotype across passages[75], has made the use of human-derived NSCs challenging. However, the development of immortalized human NSC lines has significantly facilitated long-term culturing of these cells in an undifferentiated state. When needed, immortalized NSCs can be expanded and differentiated into the appropriate type of neuron (reviewed in [76]). One such cell line, ReNcell VM, is an immortalized human fetal NSC line derived from the ventral mesencephalon by transduction with v-myc. Differentiation leads to neurons that express TH and generate action potentials[77]. A recent study with ReNcell VM cells demonstrated that loss of PINK1 leads to mitochondrial dysfunction, increased oxidative stress and lysosomal pathology, which subsequently led to increased cell death[78].

Genetic manipulation of stem cells

In addition to self-renewal and pluripotency, ESCs are amendable to genetic manipulation. Inserting selectable genes under the control of cell-type-specific promoters allows the enrichment of specific cell populations. This is particularly helpful: the differentiation of ES cells is asynchronous, and cells undergoing differentiation are heterogeneous[79]. For example, selection for neuronal cells have been accomplished by placing a neomycin gene under the control of the Soxl promoter [80]. Another group inserted GFP into the DAT locus in mouse ESCs; in this way, live DA neurons can be selected based on GFP fluorescence[81]. Furthermore, expression of specific genes, such as Nurr1 in ES cell lines, enables a greater enrichment of DA neurons during differentiation[65].

ES cells have also been modified to dissect mechanisms of genetic models of PD, such as mutant α-synuclein-mediated toxicity [82]. In addition, disease-causing mutations can be introduced into ESCs or ESCs can be isolated from animal models of the disease. For instance, ESCs from knock-out DJ-1 mouse embryos were differentiated into DA neurons. DJ-1-deficient cells had a lower survival rate and were more sensitive to oxidative stress, suggesting that DJ-1 is an essential component of the oxidative stress response[83].

Applying stem cells to HTS

SCs are an attractive alternative for HTS. ES cell-derived neurons have been used in HTS to assay more than 2.4 million small molecules for AMPA potentiation and toxicity [8486]. Importantly, these neurons demonstrate pharmacological responses similar to those of primary rat neurons[84]. The ability of SCs to differentiate into clinically relevant neuron-specific populations makes them ideal for identifying drugs that are specific for targets and also potentially for cell types. Although mouse SCs have advantages over primary cells as a drug discovery tool[58], especially in terms of scalability, questions remain about how well drug evaluations in mouse cells translate to efficacy and safety in humans. Growing human SCs to the scale required for drug discovery is difficult and has been hindered by availability and ethical and political constraints.

Induced Pluripotent Stem Cells

In 2006, the Yamanaka group discovered a group of genes that directly reprogram fibroblast cells to ESC-like cells, known as induced pluripotent stem (iPS) cells[87, 88]. The discovery of human iPS cells opened the door to a new generation of cell-based models for human neurological diseases, including PD. Established protocols that promote differentiation of hESCs have been adapted for iPS cells, allowing the induction of iPS cells into DA neurons[89, 90]. Progress has been made in characterizing the DA phenotype and function of these cells[91].

iPS cells from individual patients

One of the biggest advantages of iPS cells is that they can be easily derived from patients with sporadic and familial forms of the disease. In the past, obtaining patient tissues to model neurodegenerative disease was very difficult. hESC lines had been derived from affected in vitro fertilization (IVF) embryos of patients with Huntington’s disease, an incurable neurodegenerative disorder[92]. However these cells are collected before symptoms manifest and cannot be expanded into new ESC lines. In PD research, skin biopsies from patients with mutations in PINK1 were used to generate primary human fibroblasts. Although these cells provided insight into mechanisms of PINK1-specific degeneration[93, 94], it is unclear how results in fibroblasts mimic the processes of degeneration in DA neurons. The ability to grow iPS cell–derived neurons from patients provides a direct link between the human tissues and the disease model in vitro. Several groups have already derived iPS cells from PD patients and differentiated them into DA neurons[90, 95]. Upon transplantation into the striatum of a PD rodent model, these cells were functional and developed dendritic arborization expected of mature DA neurons. Furthermore, the transplantation of these cells effectively rescue motor deficits in these animals[96].

Future possibilities with iPS cells

It is unknown whether patient-derived iPS cell–based models of PD will recapitulate the characteristic pathology found in PD, including the formation of LBs. Due to the late onset of PD symptoms, the brief lifetime of the reprogrammed cells may be insufficient to observe any pathogenic process. Also the subtype of DA neuron, specifically the SN-A9 neurons, is critical for reestablishing new DA terminals and restoring motor function in midbrain transplantations[97]. Therefore, the iPS cells might need to be pushed towards differentiating SN-A9 type DA neurons[72], before a PD-specific phenotype is observed. On the other hand, PD patients do not display motor deficits until they have lost 50–60% of their DA neurons and 70–80% of their striatal DA terminals[98100]. Thus, cellular dysfunction might occur well before symptoms are visible. So far DA neurons derived from iPS cells of five patients with idiopathic PD have showed no obvious differences with control cells[90]. However, a recent encouraging paper showed that DA neurons derived from iPS cells of a patient with the most common LRRK2 mutation, G2019S, had enhanced sensitivity to an array of cellular stresses and accumulated α-synuclein[101]. Validation of disease-associated phenotypes may involve knocking down the underlying genetic mutations in an attempt to rescue the disease phenotypes. An exciting new technology involving homologous recombination combined with zinc finger nucleases may allow for targeted genetic modification of iPS cells[102, 103]. The ability to specifically disrupt genes and precisely insert transgenes might allow gain or loss of function experiments to facilitate the validation of specific targets and pathways in these cells.

iPS cells have the potential to resolve many unanswered issues. For example, ethnicity is important for the penetrance of PD-associated mutations. The frequency of the G2019S LRRK2 mutation in northern European PD cases is less than 5%. However, in North African Arab and Ashkenazi Jews, it is 40% and 18%, respectively (reviewed in [104]). Unlike available human-derived ES lines, which represent a limited spectrum of ethnicity [105], iPSc cells can be isolated easily from individuals from a broad range of ethnic backgrounds, enabling investigation into differences in ethnic susceptibility to disease-associated mutations and accelerating progress towards individualized medicine.

iPS cells will also allow us to examine more subtypes of PD. Existing cellular models of PD are based on inherited forms of the disease. However, only 5–10% of PD patients suffer from monogenic forms of the disease. Most develop idiopathic or sporadic PD[106]. PD is a multifaceted and complex disorder[107], but disease subtypes can be defined based on age of onset, dominant clinical features and progression rate[108]. Multiple genetic risk factors likely contribute to the development of sporadic PD and disease subtypes[109]. Routinely deriving iPS cells from sporadic patients potentially will enable the engineering of cellular models that faithfully incorporate all these risk factors.

Like ES cell lines, iPS cells from different patients vary in their propensity to differentiated[96, 110]. ES and iPS cells maintain their genomic information, but they might lose critical epigenetic signals[88]. In the future, this variation might be harnessed to distinguish between genetic and environmental variables that contribute to sporadic PD. However, variation has been observed in different iPS cell clones derived from the same individual[111]. The viral vectors used to transduce reprogramming factors could be one source of this variation[90]. In a recent study, removing the reprogramming genes from an iPS cell line derived from a PD patient led to expression patterns that were more similar to hESCs than the parental iPS line[90]. For iPS cells to become “mainstream” as a tool in drug discovery, they must be generated without the genomic integration of reprogramming vectors.

Currently, the protocols for differentiating these cells are costly and inefficient, and the resulting cellular subtypes have considerable variability. However, ongoing research seeks new ways to differentiate iPS cells into specific neuronal subtypes and to characterize these neurons at the molecular and functional levels. Most importantly, neuronal phenotypes found in these cells must recapitulate patient phenotypes, and these phenotypes must be validated in cells derived from multiple patients.

Modeling PD Cellular Complexity

A successful cell-based model for drug discovery would facilitate identification of relevant therapeutic targets and compounds to modulate those targets. An ideal model would accurately predict the efficacy and safety of these drugs in humans, with high predictive value for clinical trials. More effective preclinical evaluation would significantly lower the costs of drug development by reducing the number of animal validation studies needed in later testing and would result in better drug success rates in expensive clinical trials.

Most cell-based HTS involve assays that are conducted on all the cells in a particular sample well, quantifying relatively simple readouts that focus on the target or single endpoints. For example, using systems, such as the Flash Cytometer (Trophos), automated fluorescence measurements can be used to quantify toxicity or other cellular parameters. This type of system is applicable to the rapid screening of thousands of compounds. A recent study screened approximately 40,000 compounds in primary motor neurons for candidates that promote neuronal survival and neurite outgrowth[112]. However, conventional well-based HTS approaches are poorly suited to deal with the increased heterogeneity and well-to-well variability of primary neurons and SCs.

The emergence of high-content screening (HCS), which measures multiple neuronal features within single cells, for thousands of cells[113, 114], provides a new level of sensitivity in screening. These systems combine high-resolution fluorescence and confocal imaging platforms, with powerful image analysis algorithms (for example Harmony™ software from PerkinElmer or Cellomics Array Scan® from Thermo Scientific) that select and analyze single cells, according to pre-defined morphology or intensity parameters, including neurite extension, cell shape, organelle morphology or translocation of fluorescently labeled proteins. However, despite significant progress, more vigorous and detailed image analysis algorithms with less manual input are still required[115, 116].

The ability to track individual neurons over the course of the disease process, combined with longitudinal measurements of multiple cellular features within each of these cells, leads to a remarkable new level of sensitivity and the ability to determine the predictive relationships between cellular changes and the ultimate fate of the cell [117119]. Conventional screening systems that take static snap shots of the disease state are poorly suited to capture the complexity and heterogeneity of cellular systems. In addition to the cell-to-cell variability of primary neurons and SCs, the biology of neurodegeneration and the differentiation of SCs unfold asynchronously within individual cells to produce additional heterogeneity.

We applied this new approach to resolve a long-standing controversy over the role of inclusion bodies in the pathogenesis of Huntington’s disease[118] and to reveal temporal relationships between the ubiquitin proteasome system and inclusion body formation[120]. In addition to determining qualitative relationships, this approach also simultaneously quantifies the contribution of one or more cellular changes on the ultimate fate of the cell[119]. We showed that cytoplasmic mislocalization of TDP43, a major protein component of neuronal aggregates characteristic of amyotrophic lateral sclerosis and frontotemporal dementia is critical for disease pathogenesis[121].

This new technology also offers powerful new approaches for HTS [115]. The added sensitivity (> 100-fold compared to snap-shot-based conventional HTS[117]) makes it much more feasible to use primary neurons, including those differentiated from human iPS cells, as a screening platform because many fewer cells are needed to detect significant effects. The ability of the system to follow thousands of individual neurons longitudinally allows each cell to serve as its own control, effectively managing the cell-to-cell and well-to-well variability that plagues cell-based HTS. These advances make this technology well suited to pursue genome-wide RNAi screening in primary neurons to identify relevant therapeutic targets and to conduct screens of moderately sized small-molecule libraries to do drug discovery. Finally, the measures of fate that the system provides are sufficiently quantitative that structure-activity relationships among chemical series can be delineated, and some lead optimization can occur even if the target of the small molecule remains to be identified.

Conclusions

A wide range of cellular systems has been used to model and investigate the underlying molecular mechanisms of PD. Each system has its own advantages and limitations for drug discovery. Improving drug discovery will require more relevant cellular models and advanced screening platforms that analyze neurons at the level of single cells, enabling multivariable modeling of disease-associated mechanisms. However, a question remains whether one model system can model the multifaceted phenotype of PD. Now, iPS cells from familial and sporadic PD patients that are differentiated into DA neurons hold great promise for faithful human cellular models that recapitulate essential features of PD. Even so, further development and characterization of these cells is required before their full potential for drug development can be realized. Ultimately, more physiologically relevant models that capture the complexity of PD can only lead to better decisions and translation at all levels of drug discovery.

Acknowledgments

We thank the members of the Finkbeiner Lab for the generous support and advice. We thank G. Howard for editorial assistance and K. Nelson for administrative assistance. This work was supported by the Hillblom Foundation (to G.S.), the NIH - National Institute on Aging grant 2P01 AG022074, Taube-Koret Center for Huntington's Disease Research, the NIH - National Institute of Neurological Disorders and Stroke grants 2R01 NS039074 and 2R01 NS045091 and by the Keck Foundation (to S.F).

References

  • 1.Wakabayashi K, Mori F, Takahashi H. Progression patterns of neuronal loss and Lewy body pathology in the substantia nigra in Parkinson's disease. Parkinsonism Relat Disord. 2006;12(Suppl 2):S92–S98. [Google Scholar]
  • 2.de Lau LM, et al. Incidence of parkinsonism and Parkinson disease in a general population: the Rotterdam Study. Neurology. 2004;63(7):1240–1244. doi: 10.1212/01.wnl.0000140706.52798.be. [DOI] [PubMed] [Google Scholar]
  • 3.Dorsey ER, et al. Projected number of people with Parkinson disease in the most populous nations, 2005 through 2030. Neurology. 2007;68(5):384–386. doi: 10.1212/01.wnl.0000247740.47667.03. [DOI] [PubMed] [Google Scholar]
  • 4.Cotzias MH, Van Woert, Schiffer LM. Aromatic amino acids and modification of parkinsonism. N. Engl. J. Med. 1967;276:374–379. doi: 10.1056/NEJM196702162760703. [DOI] [PubMed] [Google Scholar]
  • 5.Braak H, et al. Staging of brain pathology related to sporadic Parkinson's disease. Neurobiol Aging. 2003;24(2):197–211. doi: 10.1016/s0197-4580(02)00065-9. [DOI] [PubMed] [Google Scholar]
  • 6.Zarow C, et al. Neuronal loss is greater in the locus coeruleus than nucleus basalis and substantia nigra in Alzheimer and Parkinson diseases. Arch Neurol. 2003;60(3):337–341. doi: 10.1001/archneur.60.3.337. [DOI] [PubMed] [Google Scholar]
  • 7.Hawkes CH, Del Tredici K, Braak H. A timeline for Parkinson's disease. Parkinsonism Relat Disord. 2010;16(2):79–84. doi: 10.1016/j.parkreldis.2009.08.007. [DOI] [PubMed] [Google Scholar]
  • 8.Booth B, Zemmel R. Prospects for productivity. Nat Rev Drug Discov. 2004;3(5):451–456. doi: 10.1038/nrd1384. [DOI] [PubMed] [Google Scholar]
  • 9.Lou K, de Rond M. The 'not invented here' myth. Nat Rev Drug Discov. 2006;5(6):451–452. doi: 10.1038/nrd2063. [DOI] [PubMed] [Google Scholar]
  • 10.Muller T. New small molecules for the treatment of Parkinson's disease. Expert Opin Investig Drugs. 2010;19(9):1077–1086. doi: 10.1517/13543784.2010.504711. [DOI] [PubMed] [Google Scholar]
  • 11.Goetz CG, et al. Sarizotan as a treatment for dyskinesias in Parkinson's disease: a double-blind placebo-controlled trial. Mov Disord. 2007;22(2):179–186. doi: 10.1002/mds.21226. [DOI] [PubMed] [Google Scholar]
  • 12.DiMasi JA, Hansen RW, Grabowski HG. The price of innovation: new estimates of drug development costs. J Health Econ. 2003;22(2):151–185. doi: 10.1016/S0167-6296(02)00126-1. [DOI] [PubMed] [Google Scholar]
  • 13.de Vries HE, et al. Nrf2-induced antioxidant protection: A promising target to counteract ROS-mediated damage in neurodegenerative disease? Free Radic Biol Med. 2008 doi: 10.1016/j.freeradbiomed.2008.09.001. [DOI] [PubMed] [Google Scholar]
  • 14.Nolan GP. What's wrong with drug screening today. Nat Chem Biol. 2007;3(4):187–191. doi: 10.1038/nchembio0407-187. [DOI] [PubMed] [Google Scholar]
  • 15.Young DW, et al. Integrating high-content screening and ligand-target prediction to identify mechanism of action. Nat Chem Biol. 2008;4(1):59–68. doi: 10.1038/nchembio.2007.53. [DOI] [PubMed] [Google Scholar]
  • 16.Ma P, Zemmel R. Value of novelty? Nat Rev Drug Discov. 2002;1(8):571–572. doi: 10.1038/nrd884. [DOI] [PubMed] [Google Scholar]
  • 17.Rezak M. Current pharmacotherapeutic treatment options in Parkinson's disease. Dis Mon. 2007;53(4):214–222. doi: 10.1016/j.disamonth.2007.05.002. [DOI] [PubMed] [Google Scholar]
  • 18.Zimprich A, et al. Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology. Neuron. 2004;44(4):601–607. doi: 10.1016/j.neuron.2004.11.005. [DOI] [PubMed] [Google Scholar]
  • 19.Paisan-Ruiz C, et al. Cloning of the gene containing mutations that cause PARK8-linked Parkinson's disease. Neuron. 2004;44(4):595–600. doi: 10.1016/j.neuron.2004.10.023. [DOI] [PubMed] [Google Scholar]
  • 20.Polymeropoulos MH, et al. Mutation in the alpha-synuclein gene identified in families with Parkinson's disease. Science. 1997;276(5321):2045–2047. doi: 10.1126/science.276.5321.2045. [DOI] [PubMed] [Google Scholar]
  • 21.Kitada T, et al. Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism. Nature. 1998;392(6676):605–608. doi: 10.1038/33416. [DOI] [PubMed] [Google Scholar]
  • 22.Bonifati V, et al. Mutations in the DJ-1 gene associated with autosomal recessive early-onset parkinsonism. Science. 2003;299(5604):256–259. doi: 10.1126/science.1077209. [DOI] [PubMed] [Google Scholar]
  • 23.Valente EM, et al. Hereditary early-onset Parkinson's disease caused by mutations in PINK1. Science. 2004;304(5674):1158–1160. doi: 10.1126/science.1096284. [DOI] [PubMed] [Google Scholar]
  • 24.Ramirez A, et al. Hereditary parkinsonism with dementia is caused by mutations in ATP13A2, encoding a lysosomal type 5 P-type ATPase. Nat Genet. 2006;38(10):1184–1191. doi: 10.1038/ng1884. [DOI] [PubMed] [Google Scholar]
  • 25.Wood-Kaczmar A, Gandhi S, Wood NW. Understanding the molecular causes of Parkinson's disease. Trends Mol Med. 2006;12(11):521–528. doi: 10.1016/j.molmed.2006.09.007. [DOI] [PubMed] [Google Scholar]
  • 26.Malkus KA, Tsika E, Ischiropoulos H. Oxidative modifications, mitochondrial dysfunction, and impaired protein degradation in Parkinson's disease: how neurons are lost in the Bermuda triangle. Mol Neurodegener. 2009;4:24. doi: 10.1186/1750-1326-4-24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Lewy FH. Paralysis agitans. I, Pathologische anatomie. Handbuch der neurologie. 1912:920–933. [Google Scholar]
  • 28.Schapira AH, et al. Mitochondrial complex I deficiency in Parkinson's disease. J Neurochem. 1990;54(3):823–827. doi: 10.1111/j.1471-4159.1990.tb02325.x. [DOI] [PubMed] [Google Scholar]
  • 29.Finkbeiner S, et al. Disease-modifying pathways in neurodegeneration. J Neurosci. 2006;26(41):10349–10357. doi: 10.1523/JNEUROSCI.3829-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Lim KL, Ng CH. Genetic models of Parkinson disease. Biochim Biophys Acta. 2009;1792(7):604–615. doi: 10.1016/j.bbadis.2008.10.005. [DOI] [PubMed] [Google Scholar]
  • 31.Bove J, et al. Toxin-induced models of Parkinson's disease. NeuroRx. 2005;2(3):484–494. doi: 10.1602/neurorx.2.3.484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Fornai F, et al. Parkinson-like syndrome induced by continuous MPTP infusion: convergent roles of the ubiquitin-proteasome system and alpha-synuclein. Proc Natl Acad Sci USA. 2005;102(9):3413–3418. doi: 10.1073/pnas.0409713102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Lee BD, et al. Inhibitors of leucine-rich repeat kinase-2 protect against models of Parkinson's disease. Nat Med. 2010;16(9):998–1000. doi: 10.1038/nm.2199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Lin X, et al. Leucine-rich repeat kinase 2 regulates the progression of neuropathology induced by Parkinson's-disease-related mutant alpha-synuclein. Neuron. 2009;64(6):807–827. doi: 10.1016/j.neuron.2009.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Yoon IS, et al. Development of a high-throughput screening assay for cytoprotective agents in rotenone-induced cell death. Analytical Biochemistry. 2010;407(2):205–210. doi: 10.1016/j.ab.2010.08.011. [DOI] [PubMed] [Google Scholar]
  • 36.Falkenburger BH, Schulz JB. Limitations of cellular models in Parkinson's disease research. J Neural Transm Suppl. 2006;(70):261–268. doi: 10.1007/978-3-211-45295-0_40. [DOI] [PubMed] [Google Scholar]
  • 37.Marx FP, et al. Identification and functional characterization of a novel R621C mutation in the synphilin-1 gene in Parkinson's disease. Hum Mol Genet. 2003;12(11):1223–1231. doi: 10.1093/hmg/ddg134. [DOI] [PubMed] [Google Scholar]
  • 38.Shendelman S, et al. DJ-1 is a redox-dependent molecular chaperone that inhibits alpha-synuclein aggregate formation. PLoS Biol. 2004;2(11):e362. doi: 10.1371/journal.pbio.0020362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Zhou W, Freed CR. Tyrosine-to-cysteine modification of human alpha-synuclein enhances protein aggregation and cellular toxicity. J Biol Chem. 2004;279(11):10128–10135. doi: 10.1074/jbc.M307563200. [DOI] [PubMed] [Google Scholar]
  • 40.Zhou W, et al. Overexpression of human alpha-synuclein causes dopamine neuron death in rat primary culture and immortalized mesencephalon-derived cells. Brain Res. 2000;866(1–2):33–43. doi: 10.1016/s0006-8993(00)02215-0. [DOI] [PubMed] [Google Scholar]
  • 41.Yang F, et al. Role of autophagy and proteasome degradation pathways in apoptosis of PC12 cells overexpressing human alpha-synuclein. Neurosci Lett. 2009;454(3):203–208. doi: 10.1016/j.neulet.2009.03.027. [DOI] [PubMed] [Google Scholar]
  • 42.Lotharius J, et al. Effect of mutant alpha-synuclein on dopamine homeostasis in a new human mesencephalic cell line. J Biol Chem. 2002;277(41):38884–38894. doi: 10.1074/jbc.M205518200. [DOI] [PubMed] [Google Scholar]
  • 43.Eglen RM, Gilchrist A, Reisine T. The use of immortalized cell lines in GPCR screening: the good, bad and ugly. Comb Chem High Throughput Screen. 2008;11(7):560–565. doi: 10.2174/138620708785204144. [DOI] [PubMed] [Google Scholar]
  • 44.Do JH, et al. Genome-wide examination of chromosomal aberrations in neuroblastoma SH-SY5Y cells by array-based comparative genomic hybridization. Mol Cells. 2007;24(1):105–112. [PubMed] [Google Scholar]
  • 45.Kenakin TP. New eyes to see texture in ligand efficacy. Nat Methods. 2005;2(3):163–164. doi: 10.1038/nmeth0305-163. [DOI] [PubMed] [Google Scholar]
  • 46.Kola I, Landis J. Can the pharmaceutical industry reduce attrition rates? Nat Rev Drug Discov. 2004;3(8):711–715. doi: 10.1038/nrd1470. [DOI] [PubMed] [Google Scholar]
  • 47.Eglen RM, Gilchrist A, Reisine T. An overview of drug screening using primary and embryonic stem cells. Comb Chem High Throughput Screen. 2008;11(7):566–572. doi: 10.2174/138620708785204108. [DOI] [PubMed] [Google Scholar]
  • 48.Mena MA, Davila V, Sulzer D. Neurotrophic effects of L-DOPA in postnatal midbrain dopamine neuron/cortical astrocyte cocultures. J Neurochem. 1997;69(4):1398–1408. doi: 10.1046/j.1471-4159.1997.69041398.x. [DOI] [PubMed] [Google Scholar]
  • 49.Heyer EJ. Electrophysiological study of mammalian neurons from ventral mesencephalon grown in primary dissociated cell culture. Brain Res. 1984;310(1):142–148. doi: 10.1016/0006-8993(84)90018-0. [DOI] [PubMed] [Google Scholar]
  • 50.Chiodo LA, Bunney BS. Population response of midbrain dopaminergic neurons to neuroleptics: further studies on time course and nondopaminergic neuronal influences. J Neurosci. 1987;7(3):629–633. doi: 10.1523/JNEUROSCI.07-03-00629.1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Rayport S, et al. Identified postnatal mesolimbic dopamine neurons in culture: morphology and electrophysiology. J Neurosci. 1992;12(11):4264–4280. doi: 10.1523/JNEUROSCI.12-11-04264.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.McNaught KS, et al. Aggresome-related biogenesis of Lewy bodies. Eur J Neurosci. 2002;16(11):2136–2148. doi: 10.1046/j.1460-9568.2002.02301.x. [DOI] [PubMed] [Google Scholar]
  • 53.Staropoli JF, et al. Parkin is a component of an SCF-like ubiquitin ligase complex and protects postmitotic neurons from kainate excitotoxicity. Neuron. 2003;37(5):735–749. doi: 10.1016/s0896-6273(03)00084-9. [DOI] [PubMed] [Google Scholar]
  • 54.Freed CR, et al. Transplantation of embryonic dopamine neurons for severe Parkinson's disease. N Engl J Med. 2001;344(10):710–719. doi: 10.1056/NEJM200103083441002. [DOI] [PubMed] [Google Scholar]
  • 55.Li JY, et al. Lewy bodies in grafted neurons in subjects with Parkinson's disease suggest host-to-graft disease propagation. Nat Med. 2008;14(5):501–503. doi: 10.1038/nm1746. [DOI] [PubMed] [Google Scholar]
  • 56.Zhou W, et al. Overexpression of human alpha-synuclein causes dopamine neuron death in primary human mesencephalic culture. Brain Res. 2002;926(1–2):42–50. doi: 10.1016/s0006-8993(01)03292-9. [DOI] [PubMed] [Google Scholar]
  • 57.McNeish JD. Stem cells as screening tools in drug discovery. Curr Opin Pharmacol. 2007;7(5):515–520. doi: 10.1016/j.coph.2007.06.005. [DOI] [PubMed] [Google Scholar]
  • 58.Pouton CW, Haynes JM. Embryonic stem cells as a source of models for drug discovery. Nat Rev Drug Discov. 2007;6(8):605–616. doi: 10.1038/nrd2194. [DOI] [PubMed] [Google Scholar]
  • 59.McNeish J. Embryonic stem cells in drug discovery. Nat Rev Drug Discov. 2004;3(1):70–80. doi: 10.1038/nrd1281. [DOI] [PubMed] [Google Scholar]
  • 60.Lazic SE, Barker RA. The future of cell-based transplantation therapies for neurodegenerative disorders. J Hematother Stem Cell Res. 2003;12(6):635–642. doi: 10.1089/15258160360732669. [DOI] [PubMed] [Google Scholar]
  • 61.Kawasaki H, et al. Generation of dopaminergic neurons and pigmented epithelia from primate ES cells by stromal cell-derived inducing activity. Proc Natl Acad Sci USA. 2002;99(3):1580–1585. doi: 10.1073/pnas.032662199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Kawasaki H, et al. Induction of midbrain dopaminergic neurons from ES cells by stromal cell-derived inducing activity. Neuron. 2000;28(1):31–40. doi: 10.1016/s0896-6273(00)00083-0. [DOI] [PubMed] [Google Scholar]
  • 63.Lee SH, et al. Efficient generation of midbrain and hindbrain neurons from mouse embryonic stem cells. Nat Biotechnol. 2000;18(6):675–679. doi: 10.1038/76536. [DOI] [PubMed] [Google Scholar]
  • 64.Raye WS, et al. Heterogeneous population of dopaminergic neurons derived from mouse embryonic stem cells: preliminary phenotyping based on receptor expression and function. Eur J Neurosci. 2007;25(7):1961–1970. doi: 10.1111/j.1460-9568.2007.05489.x. [DOI] [PubMed] [Google Scholar]
  • 65.Kim JH, et al. Dopamine neurons derived from embryonic stem cells function in an animal model of Parkinson's disease. Nature. 2002;418(6893):50–56. doi: 10.1038/nature00900. [DOI] [PubMed] [Google Scholar]
  • 66.Ben-Hur T, et al. Transplantation of human embryonic stem cell-derived neural progenitors improves behavioral deficit in Parkinsonian rats. Stem Cells. 2004;22(7):1246–1255. doi: 10.1634/stemcells.2004-0094. [DOI] [PubMed] [Google Scholar]
  • 67.Bjorklund LM, et al. Embryonic stem cells develop into functional dopaminergic neurons after transplantation in a Parkinson rat model. Proc Natl Acad Sci USA. 2002;99(4):2344–2349. doi: 10.1073/pnas.022438099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Park CH, et al. In vitro and in vivo analyses of human embryonic stem cell-derived dopamine neurons. J Neurochem. 2005;92(5):1265–1276. doi: 10.1111/j.1471-4159.2004.03006.x. [DOI] [PubMed] [Google Scholar]
  • 69.Perrier AL, et al. Derivation of midbrain dopamine neurons from human embryonic stem cells. Proc Natl Acad Sci USA. 2004;101(34):12543–12548. doi: 10.1073/pnas.0404700101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Zeng X, et al. Dopaminergic differentiation of human embryonic stem cells. Stem Cells. 2004;22(6):925–940. doi: 10.1634/stemcells.22-6-925. [DOI] [PubMed] [Google Scholar]
  • 71.Morizane A, et al. A simple method for large-scale generation of dopamine neurons from human embryonic stem cells. J Neurosci Res. 2010;88(16):3467–3478. doi: 10.1002/jnr.22515. [DOI] [PubMed] [Google Scholar]
  • 72.Cooper O, 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;45(3):258–266. doi: 10.1016/j.mcn.2010.06.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Nunes MC, et al. Identification and isolation of multipotential neural progenitor cells from the subcortical white matter of the adult human brain. Nat Med. 2003;9(4):439–447. doi: 10.1038/nm837. [DOI] [PubMed] [Google Scholar]
  • 74.Reynolds BA, Weiss S. Generation of neurons and astrocytes from isolated cells of the adult mammalian central nervous system. Science. 1992;255(5052):1707–1710. doi: 10.1126/science.1553558. [DOI] [PubMed] [Google Scholar]
  • 75.Wright LS, et al. Human progenitor cells isolated from the developing cortex undergo decreased neurogenesis and eventual senescence following expansion in vitro. Exp Cell Res. 2006;312(11):2107–2120. doi: 10.1016/j.yexcr.2006.03.012. [DOI] [PubMed] [Google Scholar]
  • 76.Kim SU. Human neural stem cells genetically modified for brain repair in neurological disorders. Neuropathology. 2004;24(3):159–171. doi: 10.1111/j.1440-1789.2004.00552.x. [DOI] [PubMed] [Google Scholar]
  • 77.Donato R, et al. Differential development of neuronal physiological responsiveness in two human neural stem cell lines. BMC Neurosci. 2007;8:36. doi: 10.1186/1471-2202-8-36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Wood-Kaczmar A, et al. PINK1 is necessary for long term survival and mitochondrial function in human dopaminergic neurons. PLoS One. 2008;3(6):e2455. doi: 10.1371/journal.pone.0002455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Pruszak J, et al. Markers and methods for cell sorting of human embryonic stem cell-derived neural cell populations. Stem Cells. 2007;25(9):2257–2268. doi: 10.1634/stemcells.2006-0744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Li M, et al. Generation of purified neural precursors from embryonic stem cells by lineage selection. Curr Biol. 1998;8(17):971–974. doi: 10.1016/s0960-9822(98)70399-9. [DOI] [PubMed] [Google Scholar]
  • 81.Zhou W, et al. Embryonic stem cells with GFP knocked into the dopamine transporter yield purified dopamine neurons in vitro and from knock-in mice. Stem Cells. 2009;27(12):2952–2961. doi: 10.1002/stem.216. [DOI] [PubMed] [Google Scholar]
  • 82.Schneider BL, et al. Over-expression of alpha-synuclein in human neural progenitors leads to specific changes in fate and differentiation. Hum Mol Genet. 2007;16(6):651–666. doi: 10.1093/hmg/ddm008. [DOI] [PubMed] [Google Scholar]
  • 83.Martinat C, et al. Sensitivity to oxidative stress in DJ-1-deficient dopamine neurons: an ES-derived cell model of primary Parkinsonism. PLoS Biol. 2004;2(11):e327. doi: 10.1371/journal.pbio.0020327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.McNeish J, et al. High-throughput screening in embryonic stem cell-derived neurons identifies potentiators of alpha-amino-3-hydroxyl-5-methyl-4-isoxazolepropionate-type glutamate receptors. J Biol Chem. 2010;285(22):17209–17217. doi: 10.1074/jbc.M109.098814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Paquette JA, et al. Assessment of the Embryonic Stem Cell Test and application and use in the pharmaceutical industry. Birth Defects Res B Dev Reprod Toxicol. 2008;83(2):104–111. doi: 10.1002/bdrb.20148. [DOI] [PubMed] [Google Scholar]
  • 86.Seiler A, et al. Improvement of an in vitro stem cell assay for developmental toxicity: the use of molecular endpoints in the embryonic stem cell test. Reprod Toxicol. 2004;18(2):231–240. doi: 10.1016/j.reprotox.2003.10.015. [DOI] [PubMed] [Google Scholar]
  • 87.Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126(4):663–676. doi: 10.1016/j.cell.2006.07.024. [DOI] [PubMed] [Google Scholar]
  • 88.Okita K, Ichisaka T, Yamanaka S. Generation of germline-competent induced pluripotent stem cells. Nature. 2007;448(7151):313–317. doi: 10.1038/nature05934. [DOI] [PubMed] [Google Scholar]
  • 89.Swistowski A, et al. Efficient generation of functional dopaminergic neurons from human induced pluripotent stem cells under defined conditions. Stem Cells. 2010;28(10):1893–1904. doi: 10.1002/stem.499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Soldner F, et al. Parkinson's disease patient-derived induced pluripotent stem cells free of viral reprogramming factors. Cell. 2009;136(5):964–977. doi: 10.1016/j.cell.2009.02.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Wernig M, et al. Neurons derived from reprogrammed fibroblasts functionally integrate into the fetal brain and improve symptoms of rats with Parkinson's disease. Proc Natl Acad Sci USA. 2008;105(15):5856–5861. doi: 10.1073/pnas.0801677105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Niclis JC, et al. Human embryonic stem cell models of Huntington disease. Reprod Biomed Online. 2009;19(1):106–113. doi: 10.1016/s1472-6483(10)60053-3. [DOI] [PubMed] [Google Scholar]
  • 93.Maj MC, et al. Oxidative stress alters the regulatory control of p66Shc and Akt in PINK1 deficient cells. Biochem Biophys Res Commun. 2010;399(3):331–335. doi: 10.1016/j.bbrc.2010.07.033. [DOI] [PubMed] [Google Scholar]
  • 94.Rakovic A, et al. Effect of endogenous mutant and wild-type PINK1 on Parkin in fibroblasts from Parkinson disease patients. Hum Mol Genet. 2010;19(16):3124–3137. doi: 10.1093/hmg/ddq215. [DOI] [PubMed] [Google Scholar]
  • 95.Park IH, et al. Disease-specific induced pluripotent stem cells. Cell. 2008;134(5):877–886. doi: 10.1016/j.cell.2008.07.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Hargus G, et al. Differentiated Parkinson patient-derived induced pluripotent stem cells grow in the adult rodent brain and reduce motor asymmetry in Parkinsonian rats. Proc Natl Acad Sci USA. 2010;107(36):15921–15926. doi: 10.1073/pnas.1010209107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Grealish S, et al. The A9 dopamine neuron component in grafts of ventral mesencephalon is an important determinant for recovery of motor function in a rat model of Parkinson's disease. Brain. 2010;133(Pt 2):482–495. doi: 10.1093/brain/awp328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Elsworth JD, et al. MPTP-induced parkinsonism: relative changes in dopamine concentration in subregions of substantia nigra, ventral tegmental area and retrorubral field of symptomatic and asymptomatic vervet monkeys. Brain Res. 1990;513(2):320–324. doi: 10.1016/0006-8993(90)90474-p. [DOI] [PubMed] [Google Scholar]
  • 99.Zigmond MJ, et al. Compensations after lesions of central dopaminergic neurons: some clinical and basic implications. Trends Neurosci. 1990;13(7):290–296. doi: 10.1016/0166-2236(90)90112-n. [DOI] [PubMed] [Google Scholar]
  • 100.Zigmond MJ, et al. Neurochemical compensation after nigrostriatal bundle injury in an animal model of preclinical parkinsonism. Arch Neurol. 1984;41(8):856–861. doi: 10.1001/archneur.1984.04050190062015. [DOI] [PubMed] [Google Scholar]
  • 101.Nguyen HN, et al. LRRK2 Mutant iPSC-Derived DA Neurons Demonstrate Increased Susceptibility to Oxidative Stress. Cell Stem Cell. 2011;8(3):267–280. doi: 10.1016/j.stem.2011.01.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Hockemeyer D, et al. Efficient targeting of expressed and silent genes in human ESCs and iPSCs using zinc-finger nucleases. Nat Biotechnol. 2009;27(9):851–857. doi: 10.1038/nbt.1562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Zou J, et al. Gene targeting of a disease-related gene in human induced pluripotent stem and embryonic stem cells. Cell Stem Cell. 2009;5(1):97–110. doi: 10.1016/j.stem.2009.05.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Change N, Mercier G, Lucotte G. Genetic screening of the G2019S mutation of the LRRK2 gene in Southwest European, North African, and Sephardic Jewish subjects. Genet Test. 2008;12(3):333–339. doi: 10.1089/gte.2007.0098. [DOI] [PubMed] [Google Scholar]
  • 105.Laurent LC, et al. Restricted ethnic diversity in human embryonic stem cell lines. Nat Methods. 2010;7(1):6–7. doi: 10.1038/nmeth0110-06. [DOI] [PubMed] [Google Scholar]
  • 106.Carr J, et al. Familial and sporadic Parkinson's disease usually display the same clinical features. Parkinsonism Relat Disord. 2003;9(4):201–204. doi: 10.1016/s1353-8020(02)00048-2. [DOI] [PubMed] [Google Scholar]
  • 107.Langston JW. The Parkinson's complex: parkinsonism is just the tip of the iceberg. Ann Neurol. 2006;59(4):591–596. doi: 10.1002/ana.20834. [DOI] [PubMed] [Google Scholar]
  • 108.Selikhova M, et al. A clinico-pathological study of subtypes in Parkinson's disease. Brain. 2009;132(Pt 11):2947–2957. doi: 10.1093/brain/awp234. [DOI] [PubMed] [Google Scholar]
  • 109.Lesage S, Brice A. Parkinson's disease: from monogenic forms to genetic susceptibility factors. Hum Mol Genet. 2009;18(R1):R48–R59. doi: 10.1093/hmg/ddp012. [DOI] [PubMed] [Google Scholar]
  • 110.Osafune K, et al. Marked differences in differentiation propensity among human embryonic stem cell lines. Nat Biotechnol. 2008;26(3):313–315. doi: 10.1038/nbt1383. [DOI] [PubMed] [Google Scholar]
  • 111.Choi KD, et al. Hematopoietic and endothelial differentiation of human induced pluripotent stem cells. Stem Cells. 2009;27(3):559–567. doi: 10.1634/stemcells.2008-0922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Bordet T, et al. Identification and characterization of cholest-4-en-3-one, oxime (TRO19622), a novel drug candidate for amyotrophic lateral sclerosis. J Pharmacol Exp Ther. 2007;322(2):709–720. doi: 10.1124/jpet.107.123000. [DOI] [PubMed] [Google Scholar]
  • 113.Barbaric I, Gokhale PJ, Andrews PW. High-content screening of small compounds on human embryonic stem cells. Biochem Soc Trans. 2010;38(4):1046–1050. doi: 10.1042/BST0381046. [DOI] [PubMed] [Google Scholar]
  • 114.Jain S, Heutink P. From single genes to gene networks: high-throughput-high-content screening for neurological disease. Neuron. 2010;68(2):207–217. doi: 10.1016/j.neuron.2010.10.010. [DOI] [PubMed] [Google Scholar]
  • 115.Daub A, Sharma P, Finkbeiner S. High-content screening of primary neurons: ready for prime time. Curr Opin Neurobiol. 2009;19(5):537–543. doi: 10.1016/j.conb.2009.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Jones TR, et al. Scoring diverse cellular morphologies in image-based screens with iterative feedback and machine learning. Proc Natl Acad Sci USA. 2009;106(6):1826–1831. doi: 10.1073/pnas.0808843106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Arrasate M, Finkbeiner S. Automated microscope system for determining factors that predict neuronal fate. Proc Natl Acad Sci USA. 2005;102(10):3840–3845. doi: 10.1073/pnas.0409777102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Arrasate M, et al. Inclusion body formation reduces levels of mutant huntingtin and the risk of neuronal death. Nature. 2004;431(7010):805–810. doi: 10.1038/nature02998. [DOI] [PubMed] [Google Scholar]
  • 119.Miller J, et al. Quantitative relationships between huntingtin levels, polyglutamine length, inclusion body formation, and neuronal death provide novel insight into huntington's disease molecular pathogenesis. J Neurosci. 2010;30(31):10541–10550. doi: 10.1523/JNEUROSCI.0146-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Mitra S, Tsvetkov AS, Finkbeiner S. Single neuron ubiquitin-proteasome dynamics accompanying inclusion body formation in huntington disease. J Biol Chem. 2009;284(7):4398–4403. doi: 10.1074/jbc.M806269200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Barmada SJ, et al. Cytoplasmic mislocalization of TDP-43 is toxic to neurons and enhanced by a mutation associated with familial amyotrophic lateral sclerosis. J Neurosci. 2010;30(2):639–649. doi: 10.1523/JNEUROSCI.4988-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Cookson MR. alpha-Synuclein and neuronal cell death. Mol Neurodegener. 2009;4:9. doi: 10.1186/1750-1326-4-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Lucking CB, et al. Association between early-onset Parkinson's disease and mutations in the parkin gene. N Engl J Med. 2000;342(21):1560–1567. doi: 10.1056/NEJM200005253422103. [DOI] [PubMed] [Google Scholar]
  • 124.Shimura H, et al. Familial Parkinson disease gene product, parkin, is a ubiquitin-protein ligase. Nat Genet. 2000;25(3):302–305. doi: 10.1038/77060. [DOI] [PubMed] [Google Scholar]
  • 125.Narendra D, et al. Parkin is recruited selectively to impaired mitochondria and promotes their autophagy. J Cell Biol. 2008;183(5):795–803. doi: 10.1083/jcb.200809125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Klein C, Grunewald A, Hedrich K. Early-onset parkinsonism associated with PINK1 mutations: frequency, genotypes, andphenotypes. Neurology. 2006;66(7):1129–1130. doi: 10.1212/01.wnl.0000220157.81513.85. author reply 1129-30. [DOI] [PubMed] [Google Scholar]
  • 127.Yao Z, Wood NW. Cell death pathways in Parkinson's disease: role of mitochondria. Antioxid Redox Signal. 2009;11(9):2135–2149. doi: 10.1089/ars.2009.2624. [DOI] [PubMed] [Google Scholar]
  • 128.Narendra DP, et al. PINK1 is selectively stabilized on impaired mitochondria to activate Parkin. PLoS Biol. 2010;8(1):e1000298. doi: 10.1371/journal.pbio.1000298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Paisan-Ruiz C, et al. Comprehensive analysis of LRRK2 in publicly available Parkinson's disease cases and neurologically normal controls. Hum Mutat. 2008;29(4):485–490. doi: 10.1002/humu.20668. [DOI] [PubMed] [Google Scholar]
  • 130.Daniels V, Baekelandt V, Taymans JM. On the Road to Leucine-Rich Repeat Kinase 2 Signalling: Evidence from Cellular and in vivo Studies. Neurosignals. 2011 doi: 10.1159/000324488. [DOI] [PubMed] [Google Scholar]

RESOURCES