Skip to main content
Therapeutic Advances in Neurological Disorders logoLink to Therapeutic Advances in Neurological Disorders
. 2009 Jan;2(1):51–60. doi: 10.1177/1756285608100324

Cellular Repair Strategies in Parkinson's Disease

Beate Winner 1, Daniela M Vogt-Weisenhorn 2, Chichung D Lie 3, Ingmar Blümcke 4,, Jürgen Winkler 5
PMCID: PMC3002617  PMID: 21180641

Abstract

Parkinson's disease (PD) is the second most common neurodegenerative disorder after Alzheimer's disease, affecting 0.7% of the elderly population (defined as over 65 years of age). PD is clinically characterized by resting tremor, muscular rigidity, hypokinesia and postural instability. These motor symptoms result largely from the deficiency or dysfunction of dopaminergic neurons in the substantia nigra. Histopathological analysis reveals depletion of dopaminergic neurons as well as eosinophilic intracytoplasmic inclusions (Lewy bodies) in surviving neurons of the substantia nigra and other brain regions. The molecular pathogenesis is linked to protein misfolding by compromised alpha-synuclein and/or related proteins (synucleinopathy). Therefore, successful therapy of motor symptoms aims for the restoration of dopaminergic neurotransmission. Pharmacological drug treatment is usually effective only at an early stage of the disease but cannot halt progressive neuronal degeneration. With recent developments in stem cell technology, cell repair or replacement approaches came into focus. Here, we review new therapeutic strategies resulting from the innate propensity of the adult brain to generate new neurons, either by pharmacological stimulation of endogenous adult stem cell population or exogenous cell transplantation modalities.

Keywords: stem cells, neurogenesis, transplantation, neurodegeneration, alpha-synuclein

Introduction

Parkinson's disease (PD) is an adult-onset, progressive, neurodegenerative movement disorder affecting the aged (senescent) population in the Western world [Thomas and Beal, 2007; Trenkwalder et al. 1995]. Both sporadic and genetic forms of PD are characterized by progressive degeneration of neurons in the striato-nigral system. In addition to the dopaminergic cell population, other groups of neurons degenerate, including cholinergic cells in the nucleus basalis, the serotonergic system of the raphe nuclei, noradrenergic neurons in the locus coeruleus, autonomic ganglia, amygdala, hippocampus, olfactory bulb, temporal cortex and cingulate cortex [Harding and Halliday, 2001]. The neu-ropathological hallmark is the formation of Lewy bodies (Figure 1) and Lewy neurites [Shults 2006; Takeda et al. 1998; Trojanowski et al. 1998; Spillantini et al. 1997; Wakabayashi et al. 1997]. These inclusions contain alpha-synuclein, ubiquitin and synaptic as well as cytoskeletal components. Clinically, the striatal dopaminergic deficit leads to resting tremor, muscular rigidity and bradykinesia. Current pharmacological treatment is symptomatic and enhances dopaminergic neurotransmission. However, drug treatment is most effective only during the early course of the disease without halting the progressive loss of pigmented dopaminergic neurons in the substantia nigra pars compacta or improving other deficits related to compromised, nondopa-minergic neuronal populations. Furthermore, life quality of PD patients is often hampered by nonmotor symptoms such as olfactory deficits, cognitive impairments, depression and auto-nomic dysfunctions. To accurately diagnose PD and effectively treat it, the understanding of its molecular pathogenesis is mandatory.

Figure 1.

Figure 1.

Neuropathological findings in Parkinson's disease. (a) Pigmented neurons within the pars compacta of the substantia nigra. This neuronal subpopulation constitutes the major reservoir for dopamine in the human brain. Extracellular pigment deposits (arrowheads) indicate substantial neuronal cell loss in PD. Hematoxylin–eosin staining. Scale bar¼100 mm. (b) An eosinophilic intracytoplasmic inclusion body (Lewy) is visible in this pigmented neuron (arrowhead). Hematoxylin-eosin staining. Scale bar ¼20mm, applies also for C. (c) Alpha-synuclein immunoreactivity specifically detects intracytoplasmic Lewy bodies in this neuron (arrowhead).

The etiology of PD is thought to be caused by a combination of environmental (toxins, lifestyle and aging) and genetic factors [for reviews, see Meredith et al. 2008; Terzioglu and Galter, 2008; Sulzer, 2007]. In recent years, several genes have been linked to familial forms of PD [for reviews, see Belin and Westerlund, 2008; Gasser, 2007]. The identification of these genes and the evaluation of their molecular functions were a major step forward in understanding the disease patho-genesis. In fact, the analysis of genetic but also of toxin-induced PD animal models revealed common intersecting molecular pathways associated with mitochondrial dysfunction, oxidative damage, abnormal protein accumulation, cytos-keletal dysfunction and neuroinflammation.

Interestingly, and despite the major contribution of these animal models to the understanding of PD, they also confront us with the issue that no single animal model for PD created to date mimics comprehensively the clinical and structural phenotype of the disease: nonmotor symptoms, slowly progressing motor deficits, loss of dopaminergic neurons in the substantia nigra and the formation of Lewy bodies. This also indicates the lack of our knowledge towards the molecular pathogenesis, which will be required to allow specific ‘targeted’ molecular therapeutic interventions, and results in the fact that, at present, it remains impossible to stop the disease progression or to restore lost functions. However, animal models were instrumental in the development of current symptomatic as well as neuron replacement therapies in Parkinson's disease. Indeed, the fascinating progress in stem cell biology during the last decade in combination with the development of appropriate animal models has opened new avenues for cell repair and replacement strategies.

Dopaminergic neurons and the molecular basis of their development

Notwithstanding, experimental or therapeutic engineering of dopaminergic cells, either obtained from adult, fetal or embryonic stem cells, or by ‘pharmacological’ stimulation of the endogenous regenerative capacity of a given brain area (i.e. striatum; see Figure 2), will substantially rely on the precise understanding of developmental signaling mechanisms involved in the induction, specification and maintenance of dopaminergic neurons in the substantia nigra, the cell population degenerating and in need of replacement in PD patients. Recently, substantial advances have been made to unravel the genetic network regulating these processes by using different animal models [reviewed in Smidt and Burbach, 2007; Prakash and Wurst, 2007, 2006; Castelo-Branco and Arenas, 2006].

Figure 2.

Figure 2.

Cell repair strategies in Parkinson's disease. Striatal connections are received from the substantia nigra (SN) in normal brain (a). Cell death of dopaminergic neurons in the substantia nigra (as induced by a unilateral 6-OHDA lesion) leads to a dopaminergic deficit in the striatum as well as in the subventricular zone (b). Growth factor treatment in this animal model induces migration of neural progenitor cells residing in the subventricular zone towards the striatum (c).

Initially, the development of the ventral midbrain is regulated by the isthmus, a signaling center located at the junction between the prospective mid- and hindbrain and determined by the opposing location of two transcription factors: ortho-denticle homeobox 2 (Otx2) and gastrulation and brain-specific homeobox protein 2 (Gbx2). Major signaling molecules at this center are fibroblast growth factor 8 (Fgf 8), sonic hedgehog (Shh) and wingless type 1 (Wnt1). Furthermore, the initial establishment of the dopaminergic progenitor domain in this region in vivo is dependent on a genetic network regulated by Wnt1. Later in development, Wnt1 also appears to initiate the differentiation of the postmitotic progeny of the dopaminergic precursors by regulating the expression of midbrain dopaminergic-specific transcription factors. In parallel to this Wnt1-regulated genetic network, a second genetic cascade regulated by Shh is involved in the induction and specification of the dopaminergic phenotype. In addition to an involvement in the establishment of the dopaminergic progenitor domain, the Shh-controlled cascade is apparently responsible for the induction of proneural transcription factors required for the acquisition of generic neuronal properties by the midbrain dopaminergic progeny. Further molecular pathways thought to play a role in the initial development of dopaminergic neurons involve the transcription factors pentraxin-related protein 3 (PITX3, also known as PTX3), engrailed 1 (EN1), engrailed 2 (EN2), nuclear receptor related 1 protein (NURR1) and the signaling molecules transforming growth factor ß (TGFyß) and retinoic acid. Interestingly, many of the transcription factors described above have also been identified in being essential for the maintenance of the dopaminergic phenotype in adults. Further trophic factors involved in dopaminergic neuron maintenance constitute glial cell line-derived neurotrophic factor (GDNF), brain-derived growth factor (BDNF), fibroblast growth factor 2 (FGF2), fibroblast growth factor 20 (FGF20) and neurotrophins. The underlying intracellular mechanisms leading to their neuro-protective function remain, however, to be specified.

Taken together, during the last decade many molecular mechanisms underlying dopaminergic induction, specification and maintenance have been identified. The next step is now to understand the molecular codes of the different subsets of adult dopaminergic neurons. The knowledge of these data is instrumental for the development of new drugs to treat PD and to provide a functional cell-replacement strategy for this disease, be it exogenous or endogenous cell replacement.

Exogenous ‘stem cell’ repair strategies in Parkinson's disease – 14 years of expertise

As motor dysfunction is considered the most prominent and debilitating symptom and clearly linked to the dopaminergic deficit in the striatum, the major therapeutic focus aimed for many years was to replace the loss of dopaminergic neurons. As these dopaminergic cells in the substantia nigra project to the striatum, transplantation therapies using either dopaminergic or fetal dopaminergic neurons were considered an ideal approach since the 1980s.

Stem cells are defined as immature cells with the ability to self-renew and differentiate into multiple cell types [Gage, 2000]. Different cell populations have been described as potential substrates for an exogenous cell repair strategy in neurological disorders, among them fetal dopaminergic neurons, neurons derived from embryonic stem cells (ESC), induced pluripotent stem cells (iPS), adult neural stem cells, and mesenchymal and umbilical blood stem cells transdifferentiated into dopaminergic neurons.

Dopaminergic neurons propagated and differentiated from ESC are a highly challenging option to treat parkinsonian symptoms in rodent animal models [Takahashi, 2007; Kawasaki et al. 2000; Lee et al. 2000]. Transplantation of ESC-derived dopaminergic neurons into primate models showed indeed amelioration of motor symptoms [Takagi et al. 2005]. However, reduced survival of cell transplants and, more importantly with respect to clinical safety, the oncogenic potential of undifferentiated ESC contaminated within the transplanted cell suspension remains an unmet challenge.

The ability to derive pluripotent cells from adult human tissues opens important opportunities in research for synucleinopathies like PD. Recently, Yamanaka and colleagues [Takahashi et al. 2007] and Yu and colleagues [Yu et al. 2007] demonstrated that expression of specific transcription factors induces adult human fibroblasts to express many of the characteristics of human embryonic stem cells. In a recent study another milestone was achieved: human iPS cells were induced to differentiate into dopamine neurons of midbrain character and were transplanted into a 6-OHDA rat model of PD. The cells showed characteristics of midbrain neurons 4 weeks after surgery. Histological analysis also showed teratoma formation, indicating that this current method still needs refinement [Wernig et al. 2008].

Adult neural stem/precursor cells reside within the subventricular zone of the lateral ventricle (SVZ) and the subgranular layer of the hippo-campal dentate gyrus, which give raise to both neurons and glia even in adult stages [Doetsch, 2003; Gage, 2000]. Human hippocampal specimens can be obtained from epilepsy surgery and thus 'stem/precursor’ cells have become increasingly available for experimental studies. There are different protocols proving that adult stem cells can be expanded in vitro and differentiated into neuronal cell populations [Walton et al. 2006]. Cell transplantation into animal models further support their ability to anatomically and functionally integrate within the adult brain [Pluchino et al. 2003]. The oncogenic potential is generally assumed to be low in the adult neural stem cell population although recent findings indicate a tumorigenicity associated with rapid propagation cycles in vitro [Siebzehnrubl and Blumcke, 2008]. However, the study of adult human stem cells and their ability to differentiate into dopaminergic neurons will be mandatory not only for ‘exogeneous’ therapy strategies [Siebzehnrubl and Blumcke 2008]. These experiments are also important to confirm basic neurodevelopmental mechanisms of dopaminergic differentiation (obtained from animal models) in the human brain when targeting this cell population for an ‘endoge-neous’ regenerative approach (Figure 3; see also below).

Figure 3.

Figure 3.

A dopaminergic cell engineered from human adult stem cells. Triple-immunofluorescence (d) of a tyrosine hydroxylase (TH) immunoreactive cell (b) co-labelled with antibodies against the neuronal marker MAP2 (a). Blue Hoechst labeling of a cell nucleus (c). This stem cell culture was isolated and expanded from a human hippocampal specimen obtained from epilepsy surgery (courtesy of Dr R. Coras; Dept of Neuropathology, Univ. Hospital Erlangen).

As a third option, neural precursor cells can also be isolated from human fetal midbrain. Recent data prove their potential to differentiate into dopaminergic neurons, thus being a valuable alternative source for cell replacement strategies [Maciaczyk et al. 2008]. Indeed, fetal nerve cell grafts were implanted into the striatum of PD patients and were the first breakthrough of cell-based therapeutic strategies in the CNS. These cell-based approaches with transplantation of dopamine-producing fetal cells provided the central proof that transplanted neurons can survive, innervate the host brain and elicit clinical beneficial effects as demonstrated by motor improvements [Hagell and Brundin, 2001; Kordower et al. 1995; Lindvall et al. 1994]. In addition, PET-analysis even showed an increased fluoro-dopa uptake in the putamen [Hauser et al. 1999; Wenning et al. 1997] indicating that implanted cells were functionally integrated [Olanow et al. 1996; Kordower et al. 1995]. In a double blind trial published in 2001, however, only a subpopulation (younger patients) benefited from transplantation [Freed et al. 2001]. The second control study showed improvement in those PD patients with milder disease [Olanow et al. 2003] indicating that the time point for cell transplantation in PD patients will be another crucial selection criteria for the success of transplantation approaches. After initial improvements in the first year, dystonia and dys-kinesias recurred in 15% of patients who received transplants, even after reduction or discontinuation of levodopa substitution. This led to a complete halt of transplantation programs aiming to improve the neurosurgical transplantation techniques as well as the preparation methods for fetal cells [Freed et al. 2001]. In addition, limited access to suitable donor tissue, variability in the outcome and adverse side effects (graft-related dyskinesias) in some patients discouraged the continuation of this therapeutic option [Bjorklund et al. 2003]. While these trials were disappointing and led to a halt of transplantation trials in the US, open-label trials have continued with good success in individual patients [e.g. Mendez et al. 2005]).

Seven years later, three post-mortem studies examined long-term fetal transplant in a total of eight subjects with advanced PD. In these studies, most individuals who survived long-term after surgery showed only limited clinical benefits after transplantation [Kordower et al. 2008; Li et al. 2008; Mendez et al. 2008]. Strikingly, neu-ropathological analysis revealed alpha-synuclein inclusions within the transplanted cells in the striatum in two of the three groups. This is reminiscent of the work by Meyer-Luehmann and colleagues in AD mouse models, which demonstrated that deposition of aggregates can be induced in grafted cells [Meyer-Luehmann et al. 2008, 2006]. These reports raise several important questions: How was alpha-synuclein pathology transmitted to the grafted cells? What induced the misfolding and aggregation of alpha-synuclein within the initially healthy fetal donor cell [Braak and Del Tredici, 2008]?

Several factors may have contributed to the spread of alpha-synuclein to the grafted cells, including lack of immunosuppression, cell preparations and surgical techniques. One alternative explanation may be linked to the endogenous spread of alpha-synuclein via striatal projections from the cerebral cortex and thalamic nuclei, affected by Lewy pathology in late-stage PD, as suggested by Braak and colleagues [Braak and Del Tredici 2008].

Did alpha-synuclein pathology affect the function of the graft? This is unlikely, as more than 90–99% of grafted neurons did not reveal any pathology [Li et al. 2008]. Instead, host PD progresses and the grafts may not continue to provide long-term benefits in some of these patients. To examine PD pathology in grafted neurons is an interesting option, although functionality and benefits to patients are even more important, and so far these studies do not suggest that the PD graft pathology should compromise functionality of the grafts. Taking these considerations together at present exogenous approaches need a deeper understanding of underlying graft–host interactions, integration mechanisms and immunogenicity of exogenously added cells, due to the limitations in cell sources or preparations of cells.

Endogenous approaches – future options for the treatment of PD

Despite several open-label and controlled trials of exogenous cell repair treatment, long-term evaluation of exogeneous cell repair treatment is still pending and controversial. There are two important facts to be addressed that also consider nondopaminergic regions to be affected in PD: (1) there are several clinical deficits before the onset of motor symptoms that support early non-dopaminergic involvement: REM sleep behavior, subtle cognitive deficits, depression, olfactory dysfunction and constipation [for a review see Ahlskog, 2007]; (2) some brain areas that are associated with a subset of these functions, in particular the hippocampus and the olfactory bulb, contain stem- and progenitor cell populations and are regions of continuous generation of new neurons and represent, therefore, areas of long-living self-maintenance. As mentioned above, neurogenesis occurs in the subgranular (SGZ) zone of the hippocampal dentate gyrus and the (SVZ) adjacent to the lateral ventricles. Adult neurogenesis involves three crucial steps: (1) asymmetric cell division of a stem cell, resulting in one daughter stem cell and one with the potential to develop into a neuron; (2) the second step is the migration of the newborn cell to its final and appropriate destination in the brain; (3) the final step involves the maturation of the cells into a neuron that forms both efferent and afferent connections within the brain. Importantly, disease processes may interfere at different levels during the generation and maturation of newly generated cells. In the dentate gyrus the newly generated cells differentiate into neurons in the granule cell layer and extend dendritic processes into the molecular layer and axonal processes towards their target area in CA3 [Zhao et al. 2006]. In the SVZ/olfactory bulb system, newborn cells arising in the SVZ normally migrate along the rostral migratory stream to the olfactory bulb where they differentiate into GABAergic and dopaminergic neurons [Ming and Song, 2005]. This pool of endogenous neural stem cells of the adult brain provides an alternative and attractive cell source for cell repair strategies. Furthermore, recent data encourage research efforts to target endogenous neural stem and progenitor cells for the treatment of early nonmotor symptoms in PD.

Neurotoxin-induced models of Parkinson's disease challenge adult neurogenesis

Several neurotoxin-induced animal models of PD have revealed severe alterations in adult neuro-genesis, in particular decreased proliferation and/or survival of newly generated neurons. In 6-hydroxydopamine (6-OHDA) lesion models, which result in an acute cell death in the substan-tia nigra, a decreased SVZ proliferation was reported by several groups [Winner et al. 2006; Baker et al. 2004; Hoglinger et al. 2004]. Interestingly, as supported by a recent study, the proliferation rate is also affected in patients with PD, as is suggested by the decrease in the number of mitotic cells in the SVZ as well as in the hippocampus [Hoglinger et al. 2004]. Moreover, toxin-induced models show an increase in dopaminergic neurogenesis in the olfactory bulb glomerular layer, which has been described for the MPTP [Yamada et al. 2004] as well as for the 6-OHDA model [Winner et al. 2006]. This finding is of particular interest, as an increase in dopaminergic olfactory neurons has been observed in the olfactory bulb of PD patients [Huisman et al. 2004] and parallels these experimental findings.

Does neurogenesis in the substantia nigra provide new dopaminergic neurons for Parkinson's disease?

This idea has stimulated several research groups. The fundamental question: does neurogenesis normally take place in the adult substantia nigra, or is such a process stimulated by damage or disease? Undoubtedly cell division takes place in the adult substantia nigra, but the majority of dividing cells appear to differentiate into different types of glial cells [Steiner et al. 2006; Chen et al. 2005; Lie et al. 2002; Mao et al. 2001]. One explanation may be that the microenvironment does not provide a suitable neurogenic niche to enable neuronal differentiation [Lie et al. 2002]. After all, there is still no unequivocal evidence that neurogenesis takes place in the human substantia nigra pars compacta of humans. If it does at all, the basal rate is not sufficient to compensate for the loss of dopaminergic neurons that occurs in PD [Yoshimi et al. 2005]. Three recent studies have once again suggested that neurogenesis may occur in the adult substantia nigra and, in one study, even restore lesion-induced behavioral deficits [Van Kampen et al. 2006, 2005; Shan et al. 2006]. A comprehensive review presents evidence that there are technical shortcomings in each of these three studies [Borta and Hoglinger, 2007].

Is the striatum the target region for endogenous replacement?

Another important focus of stem cell research has been looking to repopulate the dopamine depleted striatum. This approach seems compelling due to the close proximity of the SVZ containing the pool of endogenous stem- and progenitor cells. Several groups studied the effects of different growth factors or a combination of them; for example, GDNF, TGFß, platelet-derived growth factor (PDGF), epidermal growth factor (EGF), FGF2, liver growth factor and BDNF on striatal neurogenesis in rats with nigral lesions [Winner et al. 2008]. Interestingly, many of these factors are capable of inducing neuroblast migration to the dopamine-depleted striatum, but newly generated dopaminergic neurons have not been identified [Reimers et al. 2006; Chen et al. 2005; Mohapel et al. 2005]. In conclusion, there seem to be direct dopaminergic effects on the endogenous stem- and progenitor cell population. Future studies are warranted to determine the interplay between dopamine-related mechanisms on the brain's regenerative potential and alpha-synuclein pathology.

Acknowledgments

This work was financially supported by the Bavarian State Ministry of Sciences, Research and the Arts, For NeuroCell (Regensburg, Germany), the Adalbert Raps Foundation (Kulmbach, Germany), and a PD fellowship of Glaxo-Smith-Kline (Munich, Germany). BW is a fellow of the Alexander von Humboldt Foundation.

Conflict of interest statement

None declared.

Contributor Information

Beate Winner, Department of Neurology, University of Regensburg, Regensburg, Germany; and Salk Institute of Biological Studies, La Jolla, CA, USA.

Daniela M. Vogt-Weisenhorn, Institute for Developmental Genetics, Helmholtz Zentrum München German Research Center for Environmental Health, Neuherberg, Germany

Chichung D. Lie, Institute for Developmental Genetics, Helmholtz Zentrum München German Research Center for Environmental Health, Neuherberg, Germany

Ingmar Blümcke, Department of Neuropathology, University Hospital Erlangen, Schwabachanalge 6 DE - 91054 Erlangen, Germany ingmar.bluemcke@uk-erlangen.de.

Jürgen Winkler, Department of Neurosciences, University of California San Diego, La Jolla, CA, USA; and Division of Molecular Neurology, University Hospital Erlangen, Erlangen, Germany.

References

  1. Ahlskog J.E.(2007)Beating a dead horse: dopamine and Parkinson disease,Neurology 69:1701–1711 [DOI] [PubMed] [Google Scholar]
  2. Baker S.A., Baker K.A., Hagg T.(2004)Dopaminergic nigrostriatal projections regulate neural precursor proliferation in the adult mouse subventricular zone,Eur J Neurosci 20:575–579 [DOI] [PubMed] [Google Scholar]
  3. Belin A.C., Westerlund M.(2008)Parkinson's disease: a genetic perspective,FEBS J 275:1377–1383 [DOI] [PubMed] [Google Scholar]
  4. Bjorklund A., Dunnett S.B., Brundin P., Stoessl A.J., Freed C.R., Breeze R.E.et al. (2003)Neural transplantation for the treatment of Parkinson's disease,Lancet Neurol 2:437–445 [DOI] [PubMed] [Google Scholar]
  5. Borta A., Hoglinger G.U.(2007)Dopamine and adult neurogenesis,J Neurochem 100:587–595 [DOI] [PubMed] [Google Scholar]
  6. Braak H., Del Tredici K.(2008)Assessing fetal nerve cell grafts in Parkinson's disease,Nat Med 14:483–485 [DOI] [PubMed] [Google Scholar]
  7. Castelo-Branco G., Arenas E.(2006)Function of Wnts in dopaminergic neuron development,Neurodegener Dis 3:5–11 [DOI] [PubMed] [Google Scholar]
  8. Chen Y., Ai Y., Slevin J.R., Maley B.E., Gash D.M.(2005)Progenitor proliferation in the adult hippocampus and substantia nigra induced by glial cell line-derived neurotrophic factor,Exp Neurol 196:87–95 [DOI] [PubMed] [Google Scholar]
  9. Doetsch F.(2003)A niche for adult neural stem cells,Curr Opin Genet Dev 13:543–550 [DOI] [PubMed] [Google Scholar]
  10. Freed C.R., Greene P.E., Breeze R.E., Tsai W.Y., Dumouchel W., Kao R.et al. (2001)Transplantation of embryonic dopamine neurons for severe Parkinson's disease,N Engl J Med 344:710–719 [DOI] [PubMed] [Google Scholar]
  11. Gage F.H.(2000)Mammalian neural stem cells,Science 287:1433–1438 [DOI] [PubMed] [Google Scholar]
  12. Gasser T.(2007)Update on the genetics of Parkinson's disease,Mov Disord 22:S343–S350 [DOI] [PubMed] [Google Scholar]
  13. Hagell P., Brundin P.(2001)Cell survival and clinical outcome following intrastriatal transplantation in Parkinson disease,J Neuropathol Exp Neurol 60:741–752 [DOI] [PubMed] [Google Scholar]
  14. Harding A.J., Halliday G.M.(2001)Cortical Lewy body pathology in the diagnosis of dementia,Acta Neuropathol (Berl) 102:355–363 [DOI] [PubMed] [Google Scholar]
  15. Hauser R.A., Freeman T.B., Snow B.J., Nauert M., Gauger L., Kordower J.H.et al. (1999)Long-term evaluation of bilateral fetal nigral transplantation in Parkinson disease,Arch Neurol 56:179–187 [DOI] [PubMed] [Google Scholar]
  16. Hoglinger G.U., Rizk P., Muriel M.P., Duyckaerts C., Oertel W.H., Caille I.et al. (2004)Dopamine depletion impairs precursor cell proliferation in Parkinson disease,Nat Neurosci 7:726–735 [DOI] [PubMed] [Google Scholar]
  17. Huisman E., Uylings H.B., Hoogland P.V.(2004)A 100% increase of dopaminergic cells in the olfactory bulb may explain hyposmia in Parkinson's disease,Mov Disord 19:687–692 [DOI] [PubMed] [Google Scholar]
  18. Kawasaki H., Mizuseki K., Nishikawa S., Kaneko S., Kuwana Y., Nakanishi S.et al. (2000)Induction of midbrain dopaminergic neurons from ES cells by stromal cell-derived inducing activity,Neuron 28:31–40 [DOI] [PubMed] [Google Scholar]
  19. Kordower J.H., Chu Y., Hauser R.A., Freeman T.B., Olanow C.W.(2008)Lewy body-like pathology in long-term embryonic nigral transplants in Parkinson's disease,Nat Med 14:504–506 [DOI] [PubMed] [Google Scholar]
  20. Kordower J.H., Freeman T.B., Snow B.J., Vingerhoets F.J., Mufson E.J., Sanberg P.R.et al. (1995)Neuropathological evidence of graft survival and striatal reinnervation after the transplantation of fetal mesencephalic tissue in a patient with Parkinson's disease,N Engl J Med 332:1118–1124 [DOI] [PubMed] [Google Scholar]
  21. Lee S.H., Lumelsky N., Studer L., Auerbach J.M., McKay R.D.(2000)Efficient generation of midbrain and hindbrain neurons from mouse embryonic stem cell,Nat Biotechnol 18:675–679 [DOI] [PubMed] [Google Scholar]
  22. Li J.Y., Englund E., Holton J.L., Soulet D., Hagell P., Lees A.J.et al. (2008)Lewy bodies in grafted neurons in subjects with Parkinson's disease suggest host-to-graft disease propagation,Nat Med 14:501–503 [DOI] [PubMed] [Google Scholar]
  23. Lie D.C., Dziewczapolski G., Willhoite A.R., Kaspar B.K., Shults C.W., Gage F.H.(2002)The adult substantia nigra contains progenitor cells with neurogenic potential,J Neurosci 22:6639–6649 [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Lindvall O., Sawle G., Widner H., Rothwell J.C., Bjorklund A., Brooks D.et al. (1994)Evidence for long-term survival and function of dopaminergic grafts in progressive Parkinson's disease,Ann Neurol 35:172–180 [DOI] [PubMed] [Google Scholar]
  25. Maciaczyk J., Singec I., Maciaczyk D., Nikkhah G.(2008)Combined use of BDNF, ascorbic acid, low oxygen, and prolonged differentiation time generates tyrosine hydroxylase-expressing neurons after long-term in vitro expansion of human fetal mid-brain precursor cells,Exp Neurol 213:354–362 [DOI] [PubMed] [Google Scholar]
  26. Mao L., Lau Y.S., Petroske E., Wang J.Q.(2001)Profound astrogenesis in the striatum of adult mice following nigrostriatal dopaminergic lesion by repeated MPTP administration,Brain Res Dev Brain Res 131:57–65 [DOI] [PubMed] [Google Scholar]
  27. Mendez I., Sanchez-Pernaute R., Cooper O., Vinuela A., Ferrari D., Bjorklund L.et al. (2005)Cell type analysis of functional fetal dopamine cell suspension transplants in the striatum and substantia nigra of patients with parkinson's disease,Brain 128:1498–1510 [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Mendez I., Vinuela A., Astradsson A., Mukhida K., Hallett P., Robertson H.et al. (2008)Dopamine neurons implanted into people with Parkinson's disease survive without pathology for 14 years,Nat Med 14:507–509 [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Meredith G.E., Sonsalla P.K., Chesselet M.F.(2008)Animal models of Parkinson's disease progression,Acta Neuropathol 115:385–398 [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Meyer-Luehmann M., Coomaraswamy J., Bolmont T., Kaeser S., Schaefer C., Kilger E.et al. (2006)Exogenous induction of cerebral beta-amyloidogenesis is governed by agent and host,Science 313:1781–1784 [DOI] [PubMed] [Google Scholar]
  31. Meyer-Luehmann M., Spires-Jones T.L., Prada C., Garcia-Alloza M., De Calignon A., Rozkalne A.et al. (2008)Rapid appearance and local toxicity of amyloid-beta plaques in a mouse model of Alzheimer's disease,Nature 451:720–724 [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Ming G.L., Song H.(2005)Adult neurogenesis in the mammalian central nervous system,Annu Rev Neurosci 28:223–250 [DOI] [PubMed] [Google Scholar]
  33. Mohapel P., Frielingsdorf H., Haggblad J., Zachrisson O., Brundin P.(2005)Platelet-derived growth factor (PDGF-BB) and brain-derived neurotrophic factor (BDNF) induce striatal neurogenesis in adult rats with 6-hydroxydopamine lesions,Neuroscience 132:767–776 [DOI] [PubMed] [Google Scholar]
  34. Olanow C.W., Goetz C.G., Kordower J.H., Stoessl A.J., Sossi V., Brin M.F.et al. (2003)A double-blind controlled trial of bilateral fetal nigral transplantation in Parkinson's disease,Ann Neurol 54:403–414 [DOI] [PubMed] [Google Scholar]
  35. Olanow C.W., Kordower J.H., Freeman T.B.(1996)Fetal nigral transplantation as a therapy for Parkinson's disease,Trends Neurosci 19:102–109 [DOI] [PubMed] [Google Scholar]
  36. Pluchino S., Quattrini A., Brambilla E., Gritti A., Salani G., Dina G.et al. (2003)Injection of adult neurospheres induces recovery in a chronic model of multiple sclerosis,Nature 422:688–694 [DOI] [PubMed] [Google Scholar]
  37. Prakash N., Wurst W.(2006)Genetic networks controlling the development of midbrain dopaminergic neurons,J Physiol 575:403–410 [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Prakash N., Wurst W.(2007)A Wnt signal regulates stem cell fate and differentiation in vivo,Neurodegener Dis 4:333–338 [DOI] [PubMed] [Google Scholar]
  39. Reimers D., Herranz A.S., Diaz-Gil J.J., Lobo M.V., Paino C.L., Alonso R.et al. (2006)Intrastriatal infusion of liver growth factor stimulates dopamine terminal sprouting and partially restores motor function in 6-hydroxydopamine-lesioned rats,J Histochem Cytochem 54:457–465 [DOI] [PubMed] [Google Scholar]
  40. Shan X., Chi L., Bishop M., Luo C., Lien L., Zhang Z.et al. (2006)Enhanced de novo neurogen-esis and dopaminergic neurogenesis in the substantia nigra of MPTP-induced Parkinson's disease-like mice,Stem Cells 24:1280–1287 [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Shults C.W.(2006)Lewy bodies,Proc Natl Acad Sci U S A 103:1661–1668 [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Siebzehnrubl F., Blumcke I.(2008)Neurogenesis in the human hippocampus and its relevance to temporal lobe epilepsies,Epilepsia 49:55–65 [DOI] [PubMed] [Google Scholar]
  43. Siebzehnrubl F.A., Jeske I., Muller D., Buslei R., Coras R., Hahnen E.et al. (2008)Spontaneous in vitro transformation of adult neural precursors into stem-like cancer cells,Brain Pathol2008 Jul 10 [Epub ahead of print] [DOI] [PMC free article] [PubMed]
  44. Smidt M.P., Burbach J.P.(2007)How to make a mesodiencephalic dopaminergic neuron,Nat Rev Neurosci 8:21–32 [DOI] [PubMed] [Google Scholar]
  45. Spillantini M., Schmidt M., Lee V.-Y., Trojanowski J., Jakes R., Goedert M.(1997)A-synuclein in Lewy bodies,Nature 388:839–840 [DOI] [PubMed] [Google Scholar]
  46. Steiner B., Winter C., Hosman K., Siebert E., Kempermann G., Petrus D.S.et al. (2006)Enriched environment induces cellular plasticity in the adult substantia nigra and improves motor behavior function in the 6-OHDA rat model of Parkinson's disease,Exp Neurol 199:291–300 [DOI] [PubMed] [Google Scholar]
  47. Sulzer D.(2007)Multiple hit hypotheses for dopamine neuron loss in Parkinson's disease,Trends in Neurosciences 30:244–250 [DOI] [PubMed] [Google Scholar]
  48. Takagi Y., Takahashi J., Saiki H., Morizane A., Hayashi T., Kishi Y.et al. (2005)Dopaminergic neurons generated from monkey embryonic stem cells function in a Parkinson primate model,J Clin Invest 115:102–109 [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Takahashi J.(2007)Stem cell therapy for Parkinson's disease,Expert Rev Neurother 7:667–675 [DOI] [PubMed] [Google Scholar]
  50. Takahashi K., Tanabe K., Ohnuki M., Narita M., Ichisaka T., Tomoda K.et al. (2007)Induction of pluripotent stem cells from adult human fibroblasts by defined factors,Cell 131:861–872 [DOI] [PubMed] [Google Scholar]
  51. Takeda A., Mallory M., Sundsmo M., Honer W, Hansen L., Masliah E.(1998)Abnormal accumulation of Nacp/a-Synuclein in neurodegenerative disorders,Am J Pathol 152:367–372 [PMC free article] [PubMed] [Google Scholar]
  52. Terzioglu M., Galter D.(2008)Parkinson's disease: genetic versus toxin-induced rodent models,FEBS J 275:1384–1391 [DOI] [PubMed] [Google Scholar]
  53. Thomas B., Beal M.F.(2007)Parkinson's disease,Hum Mol Genet 16Spec No. 2:R183–194 [DOI] [PubMed] [Google Scholar]
  54. Trenkwalder C, Schwarz J., Gebhard J., Ruland D., Trenkwalder P., Hense H.W.et al. (1995)Starnberg trial on epidemiology of parkinsonism and hypertension in the elderly. Prevalence of Parkinson's disease and related disorders assessed by a door-to-door survey of inhabitants older than 65 years, Arch Neurol 52:1017–1022 [DOI] [PubMed] [Google Scholar]
  55. Trojanowski J.Q., Lee V.M.(1998)Aggregation of neurofilament and alpha-synuclein proteins in Lewy bodies: implications for the pathogenesis of Parkinson disease and Lewy body dementia,Arch Neurol 55:151–152 [DOI] [PubMed] [Google Scholar]
  56. Van Kampen J.M., Eckman C.B.(2006)Dopamine D3 receptor agonist delivery to a model of Parkinson's disease restores the nigrostriatal pathway and improves locomotor behavior,J Neurosci 26:7272–7280 [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Van Kampen J.M., Robertson H.A.(2005)A possible role for dopamine D3 receptor stimulation in the induction of neurogenesis in the adult rat substantia nigra,Neuroscience 136:381–386 [DOI] [PubMed] [Google Scholar]
  58. Wakabayashi K, Matsumoto K, Takayama K, Yoshimoto M., Takahashi H.(1997)NACP, a presynaptic protein, immunoreactivity in Lewy bodies in Parkinson's disease,Neurosci Lett 239:45–48 [DOI] [PubMed] [Google Scholar]
  59. Walton N.M., Sutter B.M., Chen H.X., Chang L.J., Roper S.N., Scheffler B.et al. (2006)Derivation and large-scale expansion of multipotent astroglial neural progenitors from adult human brain,Development 133:3671–3681 [DOI] [PubMed] [Google Scholar]
  60. Wenning G.K., Odin P., Morrish P., Rehncrona S., Widner H., Brundin P.et al. (1997)Short- and long-term survival and function of unilateral intrastriatal dopaminergic grafts in Parkinson's disease,Ann Neurol 42:95–107 [DOI] [PubMed] [Google Scholar]
  61. Wernig M., Zhao J.P., Pruszak J., Hedlund E., Fu D., Soldner F.et al. (2008)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 105:5856–5861 [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Winner B., Couillard-Despres S., Geyer M., Aigner R., Bogdahn U., Aigner L.et al. (2008)Dopaminergic lesion enhances growth factor-induced striatal neuroblast migration,J Neuropathol Exp Neurol 67:105–116 [DOI] [PubMed] [Google Scholar]
  63. Winner B., Geyer M., Couillard-Despres S., Aigner R., Bogdahn U., Aigner L.et al. (2006)Striatal deafferentation increases dopaminergic neurogenesis in the adult olfactory bulb,Exp Neurol 197:113–121 [DOI] [PubMed] [Google Scholar]
  64. Yamada M., Onodera M., Mizuno Y., Mochizuki H.(2004)Neurogenesis in olfactory bulb identified by retroviral labeling in normal and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated adult mice,Neuroscience 124:173–181 [DOI] [PubMed] [Google Scholar]
  65. Yoshimi K., Ren Y.R., Seki T., Yamada M., Ooizumi H., Onodera M.et al. (2005)Possibility for neurogenesis in substantia nigra of Parkinsonian brain,Ann Neurol 58:31–40 [DOI] [PubMed] [Google Scholar]
  66. Yu J., Vodyanik M.A., Smuga-Otto K., Antosiewicz-Bourget J., Frane J.L., Tian S.et al. (2007)Induced pluripotent stem cell lines derived from human somatic cells,Science 318:1917–1920 [DOI] [PubMed] [Google Scholar]
  67. Zhao C., Teng E.M., Summers R.G., Jr., Ming G.L., Gage F.H.(2006)Distinct morphological stages of dentate granule neuron maturation in the adult mouse hippocampus,J Neurosci 26:3–11 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Therapeutic Advances in Neurological Disorders are provided here courtesy of SAGE Publications

RESOURCES