894
Views
2
CrossRef citations to date
0
Altmetric
Basic Research

Stem cell therapy for Parkinson's disease

Terapia de células madre para la enfermedad de Parkinson

Thérapie à base de cellules souches dans la maladie de Parkinson

Pages 303-311 | Published online: 01 Apr 2022

Abstract

Transplantation of human fetal dopamine (DA) neurons to patients with Parkinson's disease (PD) has given proof of the principle that new neurons can survive for at least a decade, and then functionally integrate and provide significant symptomatic relief. Unfortunately, the ethical, technical, and practical limitations of using fetal DA neurons as the source for cell transplantation in PD, in combination with the development of unwanted grafting-related side effects, have put a halt to the spread of this treatment into clinical practice. Hopefully, recent advances in the fields of stem cell biology and adult neurogenesis research will lead totamen in new exciting ways to better understand and control the biological parameters necessary for achieving safe and successful neuronal replacement in PD patients.

El transplante de neuronas dopaminérgicas (DA) fetales humanas a pacientes con enfermedad de Parkinson (EP) ha dado validez al principio según el cual las nuevas neuronas pueden sobrevivir por al menos una década, y después integrarse funcionalmente y aportar un alivio sintomático significativo. Desafortunadamente, las limitaciones éticas, técnicas y prácticas de utilizar neuronas DA fetales como la fuente del transplante celular en la EP, asociadas al desarrollo de efectos secundarios indeseables relacionados con el transplante, ha puesto freno a la extensión de este tratamiento a la práctica clínica. Se espéra que los recientes avances en los campos de la biología de células madre y en la investigación de la neurogénesis en el adulio conducirán a nuevas y desafiantes vías para una mejor comprensión y control de los parámetros biológicos necesarios para realizar un reemplazo neuronal seguro y exiioso en pacientes con EP.

La transplantation de neurones dopaminergiques (DA) humains de fœtus à des patients atteints par la maladie de Parkinson (MP), a apporté la démonstration du principe selon lequel de nouveaux neurones peuvent survivre pendant au moins une décennie, et ensuite s'intégrer fonctionnellement et apporter un soulagement symptomatique significatif. Malheureusement, les restrictions éthiques, techniques et pratiques à l'utilisation des neurones DA de fœtus pour la transplantation cellulaire dans la MP, associées au développement d'effets secondaires indésirables liés aux greffes, ont mis un frein à l'extension de ce traitement en pratique clinique. Heureusement, des avancées récentes dans les domaines de la biologie des cellules souches et de la recherche sur la neurogenèse de l'adulte conduiront à explorer de nouvelles voies passionnantes pour mieux comprendre et contrôler les paramètres biologiques nécessaires pour réaliser un remplacement réussi et sans risque des neurones chez les patients atteints de la MP.

Frustration over the fact that pharmacological treatments for Parkinson's disease (PD) can only provide the patient with symptomatic relief for a limited amount. of time (5-15 years) has stimulated clinicians and basic scientists to seek for alternative treatment, methods. Since the major contributing cause of PD has been found to be the loss or dysfunction of dopamine (DA)-producing neurons in the nigrostriatal pathway, an obvious treatment alternative would be to try to replace or protect the damaged DA neurons. This might, be achieved by transplanting new DA-producing cells and/or by providing the endogenous remaining DA neurons with protective agents such as neurotrophic growth factors.

On the basis of positive results from numerous studies using animal models for PD, the first clinical transplantation studies for PD started in the mid-1980s and involved autologous transplantation of catecholamine-producing adrenal medulla cells.Citation1,Citation2 Previous basic animal research involving cell implantation had convincingly shown encouraging functional effects of intrastriatal grafts of DA-producing cellsCitation3-Citation5 and these effects have since been confirmed in a range of animal behavioral tests.Citation6,Citation7 It was shown that, the observed behavioral effects are dependent on the survival of DA-producing neurons within the striatum, since the removal of transplanted tissueCitation8 or an immune rejection of transplanted neuronsCitation9 reverses the transplant-induced behavioral recovery in animal studies. In addition, intrastriatal grafting in nondopamincrgic tissues produces no behavioral effects.Citation10,Citation11 The results of the first clinical trials using adrenal medulla graft, proved to be quite disappointing because of the absence of any objective reductions in PD signs, which was believed to be partly due to very poor graft, survival. The scientific community, however, responded quickly to this disappointment by adopting the scientifically more sound approach of transplanting PD patients with DA neurons, which were obtained from aborted fetuses.Citation12,Citation13 These transplantation efforts have since continued as small open-label trials. The results from four centers in Sweden, France, USA, and Canada, including 26 patients, have recently been reviewed by Björklund et al,Citation14 and the results of these trials have been reported in numerous publications.Citation12,Citation13,Citation15-Citation30 These open-label studies have shown that, human fetal DA neurons can survive in the recipient brain for more than 10 years without being affected by ongoing disease processes. The neurons show adequate release of DA into the hostCitation18 and, most importantly, they gradually provide substantial clinical improvement, with up to 50% to 60% reductions in the Unified Parkinson's Disease Rating Scale (UPDRS). Moreover, the clinical improvements strongly correlate with recovery of movement-related activation of the host premotor and supplementary motor cortex.Citation14

Most of the early transplantation efforts for PD were carried out as open-label trials. These trials gave similar results and suggested the potential benefits of cell transplantation, but concerns were raised about their validity because of the relative limited number of patients, the variable inclusion criteria, and the lack of adequate control groups. In 1992, to circumvent these issues, the National Institutes of Health (NIH) agreed to sponsor two larger controlled clinical trials. These were designed as doubleblind clinical trials and even included highly controversial sham surgeries as placebo controls. The results of the first trial were published in 2001Citation31 and the results of the second trial have recently been reported.Citation32 To transplantation enthusiasts, the results were rather disappointing - even troubling. The first study showed no overall improvement on a subjective global rating scale; however, some reductions in UPDRS score were found in patients who had responded well to L-dopa treatment prior to surgery.Citation14,Citation31,Citation33 The most troubling result was that 15% of the grafted patients showed severe dyskinesias as a side effect of treatment. The second study also failed to show any significant improvements after grafting and, in this study, more than 50% of the patients developed dyskinesias.Citation32 In spite of the disappointing and troubling results of these recent NIH trials, most of the scientists involved seem to agree that more basic research and clinical trials are needed to be fully able to evaluate the benefits from this highly novel and still experimental treatment. A more detailed discussion of these issues can be found in Bjôrklund et al.Citation14

One issue that, becomes very clear from the discussion about cell transplantation for PD is that, the current method of using fetal DA neurons has major technical and practical limitations, including the limited and ethically controversial availability of human fetal DA neurons, and the potential immunological and virological complications of using nonhuman species as fetal cell sources. Therefore, most, of the scientific community agree that this approach now requires a better source of transplantable DA neurons if cell therapy is ever to become a realistic and accessible treatment modality for PD. This review will focus on the various types of stem or progenitor cells currently under investigation as potential sources for cell replacement, in PD. For additional, reading on this subject, I would like to refer the reader to the two excellent review articles by ArenasCitation34 and Isacson.Citation35

ES cells

Embryonic stem (ES) cells, which were first, isolated from mouse blastocysts in 1981 ,Citation36,Citation37 have been shown to proliferate indefinitely in vitro in an undifferentiated state, and to differentiate into various lineages in response to different cell culture conditions. Current, extensive knowledge of cell biology, genetic manipulation, and in vitro culture methods make mouse ES cells an optimal system for potential development, of unlimited transplantable cell source with reproducible genetic modification and cell biological methods.Citation38 It has been known for several years that mouse blastocyst-dcrived cell lines could differentiate into teratomas containing cells of neuroectodermal lineage after transplantation of undifferentiated cells into syngeneic mice.Citation39 Using retinoic acid (RA) treatment, Bain et al described the first, in vitro protocol for efficient generation of neurons from ES cells.Citation40 However, the Bain protocol was not suitable to generate DA neurons, most probably due to the fact, that RA primes the neural cells towards more “dorsal” phenotypes. Recently, Barberi et al described several protocols for the generation of several kinds of neurons from mouse ES cells.Citation41 Interestingly, some reports suggest that neural differentiation from ES cells may even be a “default” option occurring unless other cell fates are

actively induced.Citation42,Citation43 This review will focus on the successful derivation of DA neurons from ES cells.

In vivo differentiation of DA neurons from ES cells

The first demonstration of ES cell-derived DA cells after transplantation came from Deacon et al,Citation44 when they showed that ES cells could spontaneously differentiate into DA neurons when grafted to either the brain or the kidney capsule. In this study, high numbers of cells (>50 000) were used and the grafts often became very large teratoma-like grafts that outgrew the target area, thus making any functional effects impossible to study. On the basis of the encouraging findings of DA cells in these large grafts, the protocol used by Deacon et al was primarily modified by decreasing the number of cells grafted. This led to smaller primarily neural grafts with numerous DA neurons, which showed beneficial functional integration in a rat model of PD.Citation45 Importantly, this study also highlighted the dangers of using dividing, undifferentiated ES cells for grafting, since about a quarter of the grafts still developed into teratomas, even when as few as 1000 ES cells were grafted.

In vitro differentiation of DA neurons from ES eels

Mouse ES cells

The in vitro derivation of DA neurons from mouse ES cells was first, described by McKay and colleagues at the NIH.Citation46 They used a five-step protocol in which approximately 30% (percentage of DA neurons/total neurons) DA differentiation was obtained using treatment with fibroblast growth factor 2 (FGF2), sonic hedgehog (SHH), FGF8, and ascorbic acid. This method for derivation of DA neurons was then further refined to about. 80% DA differentiation through transgenic expression of Nurr-1 in combination with FG.F2, SHH, FGF8, and ascorbic acid treatment.Citation47 Using a similar Nurr-1 transgenic approach, the McKay group later showed functional effects of such in vitro ES cell-derived DA neurons in a rat. model of PD.Citation48 By differentiating the ES cells from DA neurons prctransplantation, these authors claimed that, they could avoid the teratoma issue seen using undifferentiated ES cells.Citation45 Unfortunately, teratomas can still develop even when cells are prediffercntiated in vitro,Citation49 probably due to contamination of remaining undifferentiated ES cells within the cultures. Other protocols for the in vitro derivation of DA neurons from ES cells have been established. Kawasaki et al showed that, yet unknown soluble factors (named stromal cell-derived inducing activity [SDIA]) from the PA6 stromal cell line could facilitate DA differentiation in approximately 30% to 35% of the neurons derived from ES cells; unfortunately, these DA neurons survived very poorly after grafting into the brain.Citation50 Barbieri et al used MS5 stromal feeder cells in combination with SHH, FGF8, ascorbic acid, and brain-derived neurotrophic factor (BDNF) treatment to obtain approximately 50% DA differentiation from normal mouse ES cellsCitation41; similar results have also been obtained from nuclear transfer-derived ES cells.Citation41,Citation51 Furthermore, Ying et al described “significant.” DA differentiation using monolayer ES cell cultures in combination with SHH, FGF8, and FGF2 treatment.Citation52 Thus, many recent, reports have now made it clear that efficient generation (30% -80%) of DA neurons can be achieved from mouse ES cells and that such cells can survive, integrate, and show functional effects in rodent, models of PD.Citation45,Citation48

Primate (nonhuman and human) ES cells

On the basis of the encouraging results from mouse ES cells and Thomson's successful generation of nonhuman primateCitation53,Citation54 and human ES cell lines,Citation55 several labs started to investigate the possibilities of making DA neurons from primate ES cells. Kawasaki et al created DA neurons from nonhuman primate ES cells using PA6 cells and SDIA,Citation56 and Vrana et al showed DA differentiation from nonhuman primate parthenogenetic stem cells (Cyno-1 cells).Citation57 The in vitro derivation of a smaller number of DA neurons from human ES cells was described by three different groups in 2001.Citation58-Citation60 We are now eagerly awaiting the first convincing demonstration of human ES cell-derived functional DA neurons in rodent, or primate models of PD.

EG cells

Embryonic germ (EG) cell lines are pluripotent, selfrenewing stem cells with many similarities to ES cells. The establishment of mouse EG cell lines was first described by Matsui et alCitation61 when they showed that the addition of basic FGF (bFGF) to primordial germ cell (PGC) cultures in the presence of membrane-associated steel factor (SE) and leukemia inhibitory factor (LIF) enhances the growth of PGC beyond that occurring normally. In 1998, Shamblott et al created human EG cell lines after culturing gonadal ridges and mesenteries containing primordial germ cells derived from 5- to 9-week postfertilization embryos.Citation62 Although it has been shown that EG cells can differentiate into neurons in vivo,Citation63 no studies on DA differentiation have been presented so far.

Unspecified NPCs

Neural progenitor cells (NPCs) are multipotent, selfrenewing cells that can differentiate into neurons, astrocytes, and oligodendrocytes. NPCs can be derived from several regions of the fetal or the adult, brainCitation64,Citation65 and are usually propagated as free floating clumps of cells, socalled “neurospheres” in which cells are kept dividing through stimulation via epidermal growth factor (EGF) and/or FGF2. A smaller proportion of the NPCs have been shown to differentiate into DA neurons (defined by their expression of tyrosine hydroxylase [TH], which is the rate-limiting enzyme in the DA synthetic pathway) when replated on extracellular matrix protein-coated dishes and stimulated to differentiation via conditioned mediaCitation66 or through stimulation with growth factors, such as interleukins (ILs) and glial cell line-derived neurotrophic factor (GDNF).Citation67 Unfortunately, although human NPCs can survive transplantation, they show no significant behavioral effects in a rat model of PD.Citation68

Genetically modified NPCs and neural cell lines

Using an immortalized cerebellar neuronal cell line (C17.2), Yang et al showed that, such cells could spontaneously achieve some DA features after being grafted into the DA-depleted rat striatumCitation69; however, others have shown that most. C17.2 cells remain undifferentiated after transplantation and many downregulate TH expression, suggesting that positive functional effects are primarily due to other mechanisms.Citation70 Previously, using the same C17.2 cell line in combination with transgenic overexpression of Nurr-1, a transcription factor known to be of importance for the normal development of nigral DA neurons,Citation71 Wagner ct al had shown that such C17.2 cells could start to express TH when stimulated by condi tioned media from midbrain type 1 astrocytes.Citation72 Another cell line that has been used in animal models for PD is the human embryonic carcinoma-derived NTN2/hNT cell line.Citation73 These cells differentiate into neurons upon treatment with RA and can display DA properties in vitro,Citation74,Citation75 as well as in vivo, after graftingCitation76,Citation77; however, survival after grafting is usually poor and grafted animals display no significant behavioral recovery.Citation77

Growth factor-producing nondopaminergic stem cells

One additional option for stem cell treatment of PD is to use stem cells as biological “pumps” for growth factors or other protective agents. Stem cells can quite easily be genetically modified to produce high amounts of such agents and could then be grafted to either the putamen or the substantia nigra compacta (SNc), where they could help by protecting the remaining endogenous DA neurons. It has been shown that stem cells producing glial cell line-derived neurotrophic factor (GDNF) can increase the survival of cocultured DA neuronsCitation78 or cotransplanted DA neurons.Citation79 In addition, C17.2 cells producing GDNFCitation80 or the GDNF family member persephinCitation81 can protect, the remaining DA neurons in a mouse model of PD. Since chronic injections of GDNF have shown positive effects on parkinsonian symptoms in a small clinical trial,Citation82 the delivery of GDNF using stem cells could become an interesting treatment, alternative for PD.

Fetal midbrain dopaminergic progenitors

A possible way to compensate for the limitations in obtaining fetal DA neurons for grafting is to try to expand the numbers of fetal DA neurons via in vitro expansion of mesencephalic precursor cells. Studer et al showed that treatment of primary cultures of fetal DA neurons with FGF2 resulted in a 30-fold increase in the number of DA neurons in the cultures, and such neurons could reduce rotational asymmetry after grafting in a rat model of PD.Citation83 In another study, Studer et al showed that the expansion of mesencephalic precursor cells could be further increased by culturing the neurons in low (3%) oxygen concentrationCitation84 or by adding ascorbic acid to the cultures.Citation85 Using a similar approach, the same group later described the expansion and differentiation of human mesencephalic precursor cells into DA neurons that survived grafting to the rat. brain.Citation86 One problem with this method is that the expanded mesencephalic precursor cells show such poor survival after grafting that most, of the benefits of the expansion step are lost.Citation87 Another disadvantage is that the mesencephalic precursor cells seems to lose their ability to become DA neurons after prolonged expansion for more than 2 to 3 weeks. A different research group led by Carvey have used cytokines, such as IL-1, IL-11, LIF, and GDNF, to increase DA differentiation from ratCitation88-Citation90 or humanCitation91 mesencephalic precursor cells. Other protocols for expansion and DA differentiation of human fetal mesencephalic progenitors have also been described,Citation67,Citation92 but no significant, functional effects have been yet shown for such human DA neurons.

Adult neural stem cells

For many people, the use of any kind of embryonic cells is highly controversial and therefore the use of stem cells derived from adult individuals has become an attractive option. The traditional view of the nervous system used to be that, no new neurons were born in adults. This concept was first challenged by Altaian,Citation93 and later it was shown that several regions of the adult nervous system could give rise to new neurons, astrocytes, and oligodendrocytes in vitro.Citation65,Citation94-Citation96 In vivo, however, neurogenesis has so far been considered to be restricted to the subventricular zone and its projection through the rostral migratory stream to the olfactory bulb and to dentate gyrus of the hippocampus.Citation97 One interesting possibility for the treatment of PD would be the occurrence of endogenous neural stem cells residing within the vicinity of the adult SNc or the striatal target area, which could be stimulated to repair some of the damaged nigrostriatal circuit found in PD. With this aim, Fallon et al described how infusion of transforming growth factor α (TGFα) into the striatal parenchyma resulted in an “in vivo induction of massive proliferation, directed migration, and differentiation of neural cells” from the subventricular zone, with positive functional effects in a rat model of PD.Citation98 This potentially very interesting observation now awaits confirmation by other independent research groups. Moreover, it was recently suggested that, there is a turnover of DA neurons in the SNc of the adult, mouse and that, this turnover increases when the DA neurons are toxically injured by MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) treatment.Citation99 Other workers have failed to find such a turnover normally ongoing in the adult SNc, but it has been shown that adult neural stem cells isolated from the SNc region have the potential to differentiate into neurons when grafted into neurogenic regions such as the hippocampus.Citation100 Additional research will have to show whether or not similar results can be found in humans.

Conclusion

The optimal source for transplantation is one of cells that can be efficiently and reproducibly produced at a reasonable cost in combination with showing predictable therapeutic efficacy after grafting. The cells should require minimal genetic manipulation or modification by signaling molecules in culture media to properly differentiate into the required cell types. Furthermore, they should be nonproliferative after grafting, free of infectious elements, and immunologically compatible with the host.

As reviewed in this article, research to develop transplant procedures is trying to fulfill these criteria. In contrast to the limitations of fetal cell sources, and to the cellularmolecular complexities of diverse NPCs, advances in the biology of blastula-derived ES cells suggest that this source may have important advantages over the others. However, to keep the expectations of stem cells from becoming unrealistic, it should probably be emphasized to clinicians and to patients and their families not to expect the clinical outcome using stem cell-derived DA neurons to be fundamentally “better” than what has already been achieved in the best cases using fetal DA neurons. After all, stem cells are basically just a way of obtaining a more practical and reliable source of the same type of neuron that has already been tried in clinics.

However, one potentially important benefit of using stem cell-derived DA neurons, which is separate from all logistical advantages, is that this is a much purer source of DA neurons than the currently used fetal midbrain preparations, where only about 10% of the neurons are actually DA neurons, the other 90% being primarily GABAergic (GABA, γ-aminobutyric acid). Although fetal midbrain cell preparations have been used extensively, very little is known to what extent such GABAergic cells might actually be counteracting some of the positive effects generated by the DA neurons. In addition, the 10% DA neurons will consist of both SNc (A9) and ventral tegmental midbrain (VTA, A10) DA neurons. There is selective degeneration of A9 neurons and a relative sparing of A10 neurons in PD.Citation101-Citation104 These two subpopulations of DA neurons within the SNc serve different functions and project to different brain areas (even within the SNc through dendritic release). The midline-positioned A10 DA neuronsCitation105 project primarily to limbic and cortical regions,Citation106 while the neighboring A9 DA neurons (which dysfunction in PD) innervate putamen motor areas.Citation107 Thus, the differences between DA A10 and A9Citation108,Citation109 are significant, and it might be possible to increase the functional effects of DA neuronal transplants by increasing the proportion of A9 neurons compared with A10 neurons.Citation110-Citation112

Another limiting aspect of cell therapy for PD is the fact that, in most studies, cells have been placed in the ectopic target, area and not in the SNc where the actual degeneration takes place. Such an ectopic placement is necessary due to the very limited success of getting DA neurons grafted on the SNc to exhibit long-distance growth and show reestablishment, of the nigrostriatal pathway. The use of stem cells for generating DA neurons for transplantation could allow for genetic or epigenetic manipulations that facilitate target finding and long-distance growth. Another option that is currently under investigation is grafting to multiple target areas within the basal ganglia circuit.Citation113 Thus, besides finding the optimal cell source, there are several other areas such as patient selection, study design, transplantation techniques, target selection, and combination therapies, where considerable improvements can be madeCitation14,Citation35,Citation111 before making the final judgment of whether cell transplantation is a useful treatment, for PD.

Selected abbreviations and acronyms

DA=

dopamine

EG=

embryonic germ (cell)

ES=

embryonic stem (cell)

FGF=

fibroblast growth factor

GDNF=

glial cell line-derived neurotrophic factor

LIF=

leukemia inhibitory factor

NPC=

neural progenitor cell

PD=

Parkinson's disease

RA=

retinoic acid

SHH=

sonic hedgehog

SNc=

substantia nigra compacta

I acknowledge financial support from the Swedish Research Council.

REFERENCES

  • MadrazoI.LeonV.TorresC.et al.Transplantation of fetal substantia nigra and adrenal medulla to the caudade putamen in two patients with Parkinson's disease.N Engl J Med.1988318513336384
  • BacklundE.GranbergP.HambergerB.et al.Transplantation of adrenal medullary tissue to striatum in parkinsonism.J Neurosurg.1985621691732578558
  • FreedWJ.MolikisaJM.SpoolE.et al.Transplanted adrenal chromaffin cells in rat brain reduce lesion-induced rotational behaviour.Nature.19812923513527254334
  • BjorklundA.SteneviU.Reconstruction of the nigrostriatal dopamine pathway by intracerebral nigral transplants.Brain Res.1979177555560574053
  • PerlowMJ.FreedWJ.HofferBJ.et al.Brain grafts reduce motor abnormalities produced by destruction of nigrostriatal dopamine system.Science.1979204643647571147
  • BrundinP.DuanW.SauerH.Functional effects of mesencephalic dopamine neurons and adrenal chromaffin cells grafted to the rodent striatum. In: Dunnett S, Bjorklund A, eds.Functional Neural Transplantation. Vol 2. New York, NY: Raven Press;1994946
  • AnnettLE.MartelFL.RogersDC.et al.Behavioral assessment of the effects of embryonic nigral grafts in marmosets with unilateral 6-OHDA lesions of the nigrostriatal pathway.Exp Neurol.19941252282467906227
  • BjorklundA.DunnettSB.SteneviV.LewisME.IversenSD.Reinnervation of the denervated striatum by substantia nigra transplants: functional consequences as revealed by pharmacological and sensorimotor testing.Brain Res.19801993073337417786
  • CarderRK.Snyder-KellerAM.LundRD.Behavioral and anatomical correlates of immunologically induced rejection of nigral xenografts.J Comp Neurol.19882773914022904453
  • DunnettSB.HernandezTD.SummerfieldA.JonesGH.ArbuthnottG.Graft-derived recovery from 6-OHDA lesions: specificity of ventral mesencephalic graft tissue.Exp Brain Res.1988714114242901978
  • BrownellAL.LivniE.GalpernW.IsacsonO.In vivo PET imaging in rat of dopamine terminals reveals functional neural transplants.Ann Neurol.1998433873909506557
  • FreedCR.BreezeRE.RosenbergNL.et al.Transplantation of human fetal dopamine cells for Parkinson's disease. Results at 1 year.Arch Neurol.1990475055122334298
  • LindvallO.RehncronaS.GustaviiB.et al.Fetal dopamine-rich mesencephalic grafts in Parkinson's disease.Lancet.19882148314842904587
  • BjorklundA.DunnettSB.BrundinP.et al.Neural transplantation for the treatment of Parkinson's disease.Lancet Neurol.2003243744512849125
  • HagellP.PicciniP.BjorklundA.et al.Dyskinesias following neural transplantation in Parkinson's disease.Nat Neurosci.2002562762812042822
  • PicciniP.LindvallO.BjorklundA.et al.Delayed recovery of movementrelated cortical function in Parkinson's disease after striatal dopaminergic grafts.Ann Neurol.20004868969511079531
  • BrundinP.PogarellO.HagellP.et al.Bilateral caudate and putamen grafts of embryonic mesencephalic tissue treated with lazaroids in Parkinson's disease.Brain.2000123(Pt 7)1380139010869050
  • PicciniP.BrooksDJ.BjorklundA.et al.Dopamine release from nigral transplants visualized in vivo in a Parkinson's patient.Nat Neurosci.199921137114010570493
  • HagellP.SchragA.PicciniP.et al.Sequential bilateral transplantation in Parkinson's disease: effects of the second graft.Brain.1999122(Pt 6)1121113210356064
  • WenningGK.OdinP.MorrishP.et al.Short- and long-term survival and function of unilateral intrastriatal dopaminergic grafts in Parkinson's disease.Ann Neurol.199742951079225690
  • WidnerH.TertrudJ.RehncronaS.et al.Fifteen months' follow-up on bilateral embryonic mesencephalic grafts in two cases of severe MPTP-induced parkinsonism.Adv Neurol.1993607297338420218
  • WidnerH.TertrudJ.RehncronaS.et al.Bilateral fetal mesencephalic grafting in two patients with severe parkinsonism induced by MPTP.N Engl J Med.1992327155615631435882
  • SawleGV.BloomfieldPM.BjorklundA.Transplantation of fetal dopamine neurons in Parkinson's disease: positron emission tomography [18F]-6-L-fluorodopa studies in two patients with putamina implants.Ann Neurol.1992311661731575455
  • LindvallO.WidnerH.RehncronaS.et al.Transplantation of fetal dopamine neurons in Parkinson's disease: 1-year clinical and neurophysiological observations in two patients with putaminal implants.Ann Neurol.1992311551651575454
  • LindvallO.BrundinP.WidnerH.et al.Grafts of fetal dopamine neurons survive and improve motor function in Parkinson's disease.Science.19902475745772105529
  • LindvallO.RehncronaS.BrundinP.et al.Neural transplantation in Parkinson's disease: the Swedish experience.Prog Brain Res.1990827297342290977
  • LindvallO.BrundinP.RehncronaS.et al.Human fetal dopamine neurons grafted into the striatum in two patients with severe Parkinson's disease: a detailed account of methodology and a 6-month follow-up.Arch Neurol.1989466156312786405
  • BreezeRE.WellsTH Jr.FreedCR.Implantation of fetal tissue for the management of Parkinson's disease: a technical note.Neurosurgery.199536104410477791972
  • FreedCR.BreezeRE.RosenbergNL.et al.Survival of implanted fetal dopamine cells and neurologic improvement 12 to 46 months after transplantation for Parkinson's disease.N Engl J Med.1992327154915551435881
  • FreedCR.RosenbergNL.SchneckSA.BreezeRE.Improved drug responsiveness following fetal tissue implant for Parkinson's disease.Neurochem Int.199220 (suppl)321S327S1365449
  • FreedCR.GreenePE.BreezeRE.et al.Transplantation of embryonic dopamine neurons for severe Parkinson's disease.N Engl J Med.200134471071911236774
  • OlanowCW.GoetzCG.KoldowerJH.et al.A double-blind controlled trial of bilateral fetal nigral transplantation in Parkinson's disease.Ann Neurol.20035440341412953276
  • FreedCR.Will embryonic stem cells be a useful source of dopamine neurons for transplant into patients with Parkinson's disease?Proc Natl Acad Sci U S A.2002991755175711854478
  • ArenasE.Stem cells in the treatment of Parkinson's disease.Brain Res Bull.20025779580812031276
  • IsacsonO.The production and use of cells as therapeutic agents in neurodegenerative diseases.Lancet Neurol.2003241742412849120
  • EvansMJ.KaufmanMH.Establishment in culture of pluripotential cells from mouse embryos.Nature.19812921541567242681
  • MartinGR.Isolation of a pleuripotent cell line from early mouse embryos cultured in medium conditioned by teratocarcinoma stem cells.Proc Natl Acad Sci U S A.198178763476386950406
  • PedersonRA.Embryonic stem cells for medicine.Sci Am.1999April6873
  • ShermanMl.MillerRA.RichterCB.Histologie analyses of experimental tumors from mouse blastocyst-derived cell lines.J Natl Cancer Inst.1977589931002846000
  • BainG.KitchensD.YaoM.HuettnerJE.GottliebDl.Embryonic stem cells express neuronal properties in vitro.Dev Biol.19951683423577729574
  • BarberiT.KlivenyiP.CalingasanNY.et al.Neural subtype specification of fertilization and nuclear transfer embryonic stem cells and application in parkinsonian mice.Nat Biotechnol.2003211200120714502203
  • TropepeV.HitoshiS.SirardC.et al.Direct neural fate specification from embryonic stem cells. A primitive mammalian neural stem cell stage acquired through a default mechanism.Neuron.200130657811343645
  • Munoz-SanjuanI.BrivanlouAH.Neural induction, the default model and embryonic stem cells.Nat Rev Neurosci.2002327128011967557
  • DeaconT.DinsmouJ.CostantiniLC.RatliffJ.IsacsonO.Blastula-derived stem cells can differentiate into dopaminergic and serotonergic neurons after transplantation.Exp Neurol.199814928419454612
  • BjorklundLM.Sanchez-PernauteR.ChungS.et al.Embryonic stem cells develop into functional dopaminergic neurons after transplantation in a Parkinson rat model.Proc Natl Acad Sci U S A.2002992344234911782534
  • LeeSH.LumelskyN.StuderL.AverbackJM.McKayRD.Efficient generation of midbrain and hindbrain neurons from mouse embryonic stem cells.Nat Biotechnol.20001867567910835609
  • ChungS.SonntagKC.AnderssonT.et al.Genetic engineering of mouse embryonic stem cells by Nurr 1 enhances differentiation and maturation into dopaminergic neurons.Eur J Neurosci.2002161829183812453046
  • KimJH.AverbackJM.Rodriguez-GomezJA.et al.Dopamine neurons derived from embryonic stem cells function in an animal model of Parkinson's disease.Nature.2002418505612077607
  • NishimuraF.YoshikawaM.KandaS.et al.Potential use of embryonic stem cells for the treatment of mouse parkinsonian models: improved behavior by transplantation of in vitro differentiated dopaminergic neurons from embryonic stem cells.Stem Cells.20032117118012634413
  • KawasakiH.MizusekiK.NishikawaS.et al.Induction of midbrain dopaminergic neurons from ES cells by stromal cell-derived inducing activity.Neuron.200028314011086981
  • WakayamaT.TabarV.RodriguezI.et al.Differentiation of embryonic stem cell lines generated from adult somatic cells by nuclear transfer.Science.200129274074311326103
  • YingQL.StavridisM.GriffithsD.LiM.SmithA.Conversion of embryonic stem cells into neuroectodermal precursors in adherent monoculture.Nat Biotechnol.20032118318612524553
  • ThomsonJA.KalishmanJ.GolosTG.et al.Isolation of a primate embryonic stem cell line.Proc Natl Acad Sci USA.199592784478487544005
  • ThomsonJA.KalishmanJ.GolosTG.et al.Pluripotent cell lines derived from common marmoset (Callithrix jacchus) blastocysts.Biol Reprod.1996552542598828827
  • ThomsonJA.Itskovitz-EldorJ.ShapiroSS.et al.Embryonic stem cells derived from human blastocysts.Science.1998282114511479804556
  • KawasakiH.SuemoriH.MizusekiK.et al.Generation of dopaminergic neurons and pigmented epithelia from primate ES cells by stromal cellderived inducing activity.Proc Natl Acad Sci USA.2002991580158511818560
  • VranaKE.HippJD.GossAM.et al.Nonhuman primate parthenogenetic stem cells.Proc Natl Acad Sci US A.2003100(suppl 1)1191111916
  • ZhangSC.WernigM.DuncanID.BrustleO.ThomsonJA.In vitro differentiation of transplantable neural precursors from human embryonic stem cells.Nat Biotechnol.2001191129113311731781
  • CarpenterMK.InokumaMS.DenhamJ.et al.Enrichment of neurons and neural precursors from human embryonic stem cells.Exp Neurol.200117238339711716562
  • ReubinoffBE.ItsyksonP.TuretskyT.et al.Neural progenitors from human embryonic stem cells.Nat Biotechnol.2001191134114011731782
  • MatsuiY.ZseboK.HoganBL.Derivation of pluripotential embryonic stem cells from murine primordial germ cells in culture.Cell.1992708418471381289
  • ShamblottMJ.AxelmanJ.WangS.et al.Derivation of pluripotent stem cells from cultured human primordial germ cells.Proc Natl Acad Sci U S A.19989513726137319811868
  • KerrDA.LladoJ.ShamblottMJ.et al.Human embryonic germ cell derivatives facilitate motor recovery of rats with diffuse motor neuron injury.J Neurosci.2003235131514012832537
  • JoheKK.HazelTG.MullerT.Dugish-DiordjevicMM.McKayRD.Single factors direct the differentiation of stem cells from the fetal and adult central nervous system. Functions of basic fibroblast growth factor and neurotrophins in the differentiation of hippocampal neurons.Genes Dev.199610312931408985182
  • ReynoldsBA.WeissS.Generation of neurons and astrocytes from isolated cells of the adult mammalian central nervous system.Science.1992255170717101553558
  • DaadiMM.WeissS.Generation of tyrosine hydroxylase-producing neurons from precursors of the embryonic and adult forebrain.J Neurosci.1999194484449710341249
  • CarpenterMK.CuiX.HuZY.et al.In vitro expansion of a multipotent population of human neural progenitor cells.Exp Neurol.199915826527810415135
  • SvendsenCN.CaldwellMA.ShenJ.et al.Long-term survival of human central nervous system progenitor cells transplanted into a rat model of Parkinson's disease.Exp Neurol.19971481351469398456
  • YangM.StullND.BerkMA.SnyderEY.lacovittiL.Neural stem cells spontaneously express dopaminergic traits after transplantation into the intact or 6-hydroxydopamine-lesioned rat.Exp Neurol.2002177506012429210
  • OurednikJ.OurednikV.LynchWP.SchachnerM.SnyderEY.Neural stem cells display an inherent mechanism for rescuing dysfunctional neurons.Nat Biotechnol.20022011031012379867
  • ZetterstromRH.SolomlnL.JanssonL.et al.Dopamine neuron agenesis in Nurrl-deficient mice.Science.19972762482509092472
  • WagnerJ.AkerudP.CastroDS.et al.Induction of a midbrain dopaminergic phenotype in Nurr1-overexpressing neural stem cells by type 1 astrocytes.Nat Biotechnol.19991765365910404157
  • LeeVMY.AndrewsPW.Differentiation of NTERA-2 clonal human embryonal carcinoma cells into neurons involves the induction of all three neurofilament proteins.J Neurosci.198665145212419526
  • IacovittiL.StullND.Expression of tyrosine hydroxylase in newly differentiated neurons from a human cell line (hNT).Neuroreport.19978147114749172156
  • ZigovaT.BarrosoLF.WillingAE.et al.Dopaminergic phenotype of hNT cells in vitro.Brain Res Dev Brain Res.20001228790
  • SaportaS.WillingAE.ColinaLO.et al.In vitro and in vivo characterization of hNT neuron neurotransmitter phenotypes.Brain Res Bull.20005326326811113579
  • BakerKA.HongM.SadiD.MendezI.Intrastriatal and intranigral grafting of hNT neurons in the 6-OHDA rat model of Parkinson's disease.Exp Neurol.200016235036010739641
  • MeyerM.JohansenJ.GramsbergenJB.JohansenTE.ZirnmerJ.Improved survival of embryonic porcine dopaminergic neurons in coculture with a conditionally immortalized GDNF-producing hippocampal cell line.Exp Neurol.2000164829310877918
  • OstenfeldT.TaiYT.MartinP.et al.Neurospheres modified to produce glial cell line-derived neurotrophic factor increase the survival of transplanted dopamine neurons.J Neurosci Res.20026995596512205689
  • AkerudP.CAnalsJM.SnyderEY.ArenasE.Neuroprotection through delivery of glial cell line-derived neurotrophic factor by neural stem cells in a mouse model of Parkinson's disease.J Neurosci.2001218108811811588183
  • AkerudP.HOmPC.Castelo-BrancoG.et al.Persephin-overexpressing neural stem cells regulate the function of nigral dopaminergic neurons and prevent their degeneration in a model of Parkinson's disease.Mol Cell Neurosci.20022120522212401443
  • GillSS.PatelNK.HottonGR.et al.Direct brain infusion of glial cell linederived neurotrophic factor in Parkinson disease.Nat Med.2003958959512669033
  • StuderL.TabarV.McKayRD.Transplantation of expanded mesencephalic precursors leads to recovery in parkinsonian rats.Nat Neurosci.1998129029510195162
  • StuderL.CseteM.LeeSH.et al.Enhanced proliferation, survival, and dopaminergic differentiation of CNS precursors in lowered oxygen.J Neurosci.2000207377738311007896
  • YanJ.StuderL.McKayRD.Ascorbic acid increases the yield of dopaminergic neurons derived from basic fibroblast growth factor expanded mesencephalic precursors.J Neurochem.20017630731111146004
  • Sanchez-PernauteR.StuderL.BankiewiczKS.MajorEO.McKayRD.In vitro generation and transplantation of precursor-derived human dopamine neurons.J Neurosci Res.20016528428811494363
  • BrundinP.BjorklundA.Survival of expanded dopaminergic precursors is critical for clinical trials.Nat Neurosci.1998153710196555
  • LingZD.PotterED.LiptonJW.CarveyPM.Differentiation of mesencephalic progenitor cells into dopaminergic neurons by cytokines.Exp Neurol.19981494114239500954
  • PotterED.LingZD.CarveyPM.Cytokine-induced conversion of mesencephalic-derived progenitor cells into dopamine neurons.Cell Tiss Res.1999296235246
  • CarveyPM.LingZD.SortwellCE.et al.A clonal line of mesencephalic progenitor cells converted to dopamine neurons by hematopoietic cytokines: a source of cells for transplantation in Parkinson's disease.Exp Neurol.20011719810811520124
  • StorchA.PaulG.CseteM.et al.Long-term proliferation and dopaminergic differentiation of human mesencephalic neural precursor cells.Exp Neurol.200117031732511476598
  • RiazSS.JauniauxE.SternGM.BradfordHF.The controlled conversion of human neural progenitor cells derived from foetal ventral mesencephalon into dopaminergic neurons in vitro.Brain Res Dev Brain Res.20021362734
  • AltmanJ.Are new neurons formed in the brains of adult mammals?Science.19621351127112813860748
  • RichardsLJ.KilpatrickTJ.BartlettPF.De novo generation of neuronal cells from the adult mouse brain.Proc Natl Acad Sci U S A.199289859185951528866
  • GageFH.CoatesPW.PAImerTD.Survival and differentiation of adult neuronal progenitor cells transplanted to the adult brainProc Natl Acad Sci USA.19959211879118838524867
  • PalmerTD.TakahashiJ.GageFH.The adult rat hippocampus contains primordial neural stem cells.Mol Cell Neurosci.199783894049143557
  • KempermannG.WiskottL.GageFH.Functional significance of adult neurogenesis.Curr Opin Neurobiol.2004141869115082323
  • FallonJ.reidS.KinyamuR.et al.In vivo induction of massive proliferation, directed migration, and differentiation of neural cells in the adult mammalian brain.Proc Natl Acad Sci U S A.200097146861469111121069
  • ZhaoM.MommaS.DelfaniK.et al.Evidence for neurogenesis in the adult mammalian substantia nigra.Proc Natl Acad Sci U S A.20031007925793012792021
  • LieDC.DziewczapolskiG.WillhoiteAR.et al.The adult substantia nigra contains progenitor cells with neurogenic potential.J Neurosci.2002226639664912151543
  • IacopinoAM.ChristakosS.Specific reduction of calcium-binding protein (28-kilodalton calbindin-D) gene expression in aging and neurodegenerative diseases.Proc Natl Acad Sci U S A.199087407840822140897
  • YamadaT.McGeerPL.BaimbridgeKG.McGeerEG.Relative sparing in Parkinson's disease of substantia nigra dopamine neurons containing calbindin-D28K.Brain Res.19905263033072257487
  • ItoH.GotoS.SakamotoS.HiranoA.Calbindin-D28k in the basal ganglia of patients with parkinsonism.Ann Neurol.1992325435501456738
  • GibbWR.Melanin, tyrosine hydroxylase, calbindin and substance P in the human midbrain and substantia nigra in relation to nigrostriatal projections and differential neuronal susceptibility in Parkinson's disease.Brain Res.19925812832911382801
  • DahlstromA.FuxeK.Localization of monoamines in the lower brain stem.Experientia.1964203983995856530
  • GerfenCR.HerkenhamM.ThibaultJ.The neostriatal mosaic: II. Patchand matrix-directed mesostriatal dopaminergic and non-dopaminergic systems.J Neurosci.19877391539342891799
  • DamierP.HirschEC.AgidY.GraybielAM.The substantia nigra of the human brain. I. Nigrosomes and the nigral matrix, a compartmental organization based on calbindin D(28K) immunohistochemistry [see comments].Brain.1999122 (Pt 8)1421143610430829
  • CostantiniLC.LinL.IsacsonO.Medial fetal ventral mesencephalon: a preferred source for dopamine neuron grafts.Neuroreport.19978225322579243621
  • HaqueN.LeBlancC.IsacsonO.Differential dissection of the rat E16 ventral mesencephalon and survival and reinnervation of the 6-OHDAlesioned striatum by a subset of aldehyde dehydrogenase-positive TH neurons.Cell Transplant.199762392489171157
  • BjorklundL.HerlihyD.IsacsonO.Cell and synaptic replacement therapy for Parkinson's disease: current status and future directions.Neurosci News.20003612
  • BjorklundLM.IsacsonO.Regulation of dopamine cell type and transmitter function in fetal and stem cell transplantation for Parkinson's disease.Prog Brain Res.200213841142012432781
  • IsacsonO.BjorklundLM.SchumacherJM.Toward full restoration of synaptic and terminal function of the dopaminergic system in Parkinson's disease by stem cells.Ann Neurol.200353(3 suppl 1)S135S14812666105
  • RamachandranAC.BartlettLE.MendezIM.A multiple target neural transplantation strategy for Parkinson's disease.Rev Neurosci.20021324325612405227