Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Brain Commun ; 5(2): fcad060, 2023.
Article in English | MEDLINE | ID: mdl-36993946

ABSTRACT

Inflammation contributes to Parkinson's disease pathogenesis. We hypothesized that B lymphocytes are involved in Parkinson's disease progression. We measured antibodies to alpha-synuclein and tau in serum from patients with rapid eye movement sleep behaviour disorder (n = 79), early Parkinson's disease (n = 50) and matched controls (n = 50). Rapid eye movement sleep behaviour disorder cases were stratified by risk of progression to Parkinson's disease (low risk = 30, high risk = 49). We also measured B-cell activating factor of the tumour necrosis factor receptor family, C-reactive protein and total immunoglobulin G. We found elevated levels of antibodies to alpha-synuclein fibrils in rapid eye movement sleep behaviour disorder patients at high risk of Parkinson's disease conversion (ANOVA, P < 0.001) and lower S129D peptide-specific antibodies in those at low risk (ANOVA, P < 0.001). An early humoral response to alpha-synuclein is therefore detectable prior to the development of Parkinson's disease. Peripheral B lymphocyte phenotyping using flow cytometry in early Parkinson's disease patients and matched controls (n = 41 per group) revealed reduced B cells in Parkinson's disease, particularly in those at higher risk of developing an early dementia [t(3) = 2.87, P = 0.01]. Patients with a greater proportion of regulatory B cells had better motor scores [F(4,24) = 3.612, P = 0.019], suggesting they have a protective role in Parkinson's disease. In contrast, B cells isolated from Parkinson's disease patients at higher risk of dementia had greater cytokine (interleukin 6 and interleukin 10) responses following in vitro stimulation. We assessed peripheral blood lymphocytes in alpha-synuclein transgenic mouse models of Parkinson's disease: they also had reduced B cells, suggesting this is related to alpha-synuclein pathology. In a toxin-based mouse model of Parkinson's disease, B-cell deficiency or depletion resulted in worse pathological and behavioural outcomes, supporting the conclusion that B cells play an early protective role in dopaminergic cell loss. In conclusion, we found changes in the B-cell compartment associated with risk of disease progression in rapid eye movement sleep behaviour disorder (higher alpha-synuclein antibodies) and early Parkinson's disease (lower levels of B lymphocytes that were more reactive to stimulation). Regulatory B cells play a protective role in a mouse model, potentially by attenuating inflammation and dopaminergic cell loss. B cells are therefore likely to be involved in the pathogenesis of Parkinson's disease, albeit in a complex way, and thus warrant consideration as a therapeutic target.

2.
Sci Rep ; 7: 44899, 2017 03 21.
Article in English | MEDLINE | ID: mdl-28322325

ABSTRACT

Haemangioblastoma is a rare malignancy of the CNS where vascular proliferation causes lesions due to endothelial propagation. We found that conditionally expressing mutant Kras, using Rag1-Cre, gave rise to CNS haemangioblastoma in the cortex and cerebellum in mice that present with highly vascular tumours with stromal cells similar to human haemangioblastomas. The aberrant haemangioblastoma endothelial cells do not express mutant Kras but rather the mutant oncogene is expressed in CNS interstitial cells, including neuronal cells and progeny. This demonstrates a non-cell autonomous origin of this disease that is unexpectedly induced via Rag1-Cre expression in CNS interstitial cells. This is the first time that mutant RAS has been shown to stimulate non-cell autonomous proliferation in malignancy and suggests that mutant RAS can control endothelial cell proliferation in neo-vascularisation when expressed in certain cells.


Subject(s)
Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Genes, ras , Hemangioblastoma/genetics , Hemangioblastoma/pathology , Mutation , Animals , Cerebellar Neoplasms/mortality , Disease Models, Animal , Gene Expression , Genes, Reporter , Hemangioblastoma/mortality , Humans , Incidence , Mice , Mice, Transgenic
3.
Dis Model Mech ; 7(10): 1193-203, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25085990

ABSTRACT

The endogenous reparative capacity of the adult human brain is low, and chronic neurodegenerative disorders of the central nervous system represent one of the greatest areas of unmet clinical need in the developing world. Novel therapeutic strategies to treat them include: (i) growth factor delivery to boost endogenous repair and (ii) replacement cell therapy, including replacing dopaminergic neurons to treat Parkinson's disease (PD). However, these approaches are restricted not only by rapid degradation of growth factors, but also by the limited availability of cells for transplant and the poor survival of implanted cells that lack the necessary stromal support. We therefore hypothesised that provision of a transient artificial stroma for paracrine delivery of pro-survival factors could overcome both of these issues. Using leukaemia inhibitory factor (LIF) - a proneural, reparative cytokine - formulated as target-specific poly(lactic-co-glycolic acid) (PLGA) nano-particles (LIF-nano-stroma), we discovered that attachment of LIF-nano-stroma to freshly isolated fetal dopaminergic cells improved their survival fourfold: furthermore, in vivo, the number of surviving human fetal dopaminergic cells tended to be higher at 3 months after grafting into the striatum of nude rats, compared with controls treated with empty nanoparticles. In addition, we also analysed the effect of a novel nano-stroma incorporating XAV939 (XAV), a potent inhibitor of the developmentally important Wnt-ß-catenin signalling pathway, to investigate whether it could also promote the survival and differentiation of human fetal dopaminergic precursors; we found that the numbers of both tyrosine-hydroxylase-positive neurons (a marker of dopaminergic neurons) and total neurons were increased. This is the first demonstration that LIF-nano-stroma and XAV-nano-stroma each have pro-survival effects on human dopaminergic neurons, with potential value for target-specific modulation of neurogenic fate in cell-based therapies for PD.


Subject(s)
Drug Carriers , Heterocyclic Compounds, 3-Ring/administration & dosage , Leukemia Inhibitory Factor/administration & dosage , Nanoparticles , Parkinson Disease/therapy , Signal Transduction , Wnt Proteins/metabolism , beta Catenin/metabolism , Dopamine/administration & dosage , Humans , Microscopy, Electron, Scanning
4.
J Neurol Neurosurg Psychiatry ; 84(6): 657-65, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23345280

ABSTRACT

Huntington's disease (HD) is a fatal autosomal dominant neurodegenerative disease involving progressive motor, cognitive and behavioural decline, leading to death approximately 20 years after motor onset. The disease is characterised pathologically by an early and progressive striatal neuronal cell loss and atrophy, which has provided the rationale for first clinical trials of neural repair using fetal striatal cell transplantation. Between 2000 and 2003, the 'NEST-UK' consortium carried out bilateral striatal transplants of human fetal striatal tissue in five HD patients. This paper describes the long-term follow up over a 3-10-year postoperative period of the patients, grafted and non-grafted, recruited to this cohort using the 'Core assessment program for intracerebral transplantations-HD' assessment protocol. No significant differences were found over time between the patients, grafted and non-grafted, on any subscore of the Unified Huntington's Disease Rating Scale, nor on the Mini Mental State Examination. There was a trend towards a slowing of progression on some timed motor tasks in four of the five patients with transplants, but overall, the trial showed no significant benefit of striatal allografts in comparison with a reference cohort of patients without grafts. Importantly, no significant adverse or placebo effects were seen. Notably, the raclopride positron emission tomography (PET) signal in individuals with transplants, indicated that there was no obvious surviving striatal graft tissue. This study concludes that fetal striatal allografting in HD is safe. While no sustained functional benefit was seen, we conclude that this may relate to the small amount of tissue that was grafted in this safety study compared with other reports of more successful transplants in patients with HD.


Subject(s)
Brain Tissue Transplantation , Corpus Striatum/transplantation , Fetal Tissue Transplantation , Huntington Disease/surgery , Adult , Brain Tissue Transplantation/adverse effects , Brain Tissue Transplantation/methods , Corpus Striatum/embryology , Female , Fetal Tissue Transplantation/adverse effects , Fetal Tissue Transplantation/methods , Humans , Male , Middle Aged , Neuropsychological Tests , Treatment Outcome
5.
J Exp Med ; 209(1): 1-10, 2012 Jan 16.
Article in English | MEDLINE | ID: mdl-22184634

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder that results in the loss of nigrostriatal dopamine neurons. The etiology of this cell loss is unknown, but it involves abnormalities in mitochondrial function. In this study, we have demonstrated that the administration of a novel noncoding p137 RNA, derived from the human cytomegaloviral ß2.7 transcript, can prevent and rescue dopaminergic cell death in vitro and in animal models of PD by protecting mitochondrial Complex I activity. Furthermore, as this p137 RNA is fused to a rabies virus glycoprotein peptide that facilitates delivery of RNA across the blood-brain barrier, such protection can be achieved through a peripheral intravenous administration of this agent after the initiation of a dopaminergic lesion. This approach has major implications for the potential treatment of PD, especially given that this novel agent could have the same protective effect on all diseased neurons affected as part of this disease process, not just the dopaminergic nigrostriatal pathway.


Subject(s)
Electron Transport Complex I/metabolism , Mitochondria/metabolism , Neuroprotective Agents/administration & dosage , Parkinson Disease/therapy , RNA, Untranslated/administration & dosage , RNA, Viral/administration & dosage , Animals , Cell Death , Cell Line , Cytomegalovirus/genetics , Disease Models, Animal , Dopaminergic Neurons/metabolism , Enzyme Activation , HEK293 Cells , Humans , Injections, Intravenous , Male , Neuroprotective Agents/metabolism , Neurotoxins/metabolism , Parkinson Disease/metabolism , Peptides/administration & dosage , Peptides/chemistry , Peptides/metabolism , RNA Transport , RNA, Untranslated/metabolism , RNA, Viral/metabolism , Rats , Rats, Sprague-Dawley , Substantia Nigra/metabolism
6.
Hippocampus ; 21(3): 233-8, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20095007

ABSTRACT

Adult neurogenesis mainly occurs in two brain regions, the subventricular zone and the dentate gyrus (DG) of the hippocampus. Neuropeptide Y (NPY) is widely expressed throughout the brain and is known to enhance in vitro hippocampal cell proliferation. Mice lacking either NPY or the Y1 receptor display lower levels of cell proliferation, thereby suggesting a role for NPY in basal in vivo neurogenesis. Here, we investigated whether exogenous NPY stimulates DG progenitors proliferation in vivo. We show that intracerebroventricular administration of NPY increases DG cell proliferation and promotes neuronal differentiation in C57BL/6 adult mice. In these mice, the proliferative effect of NPY is mediated by the Y1 and not the Y2 receptor, as a Y1 ([Leu(31) ,Pro(34) ]), but not a Y2 (NPY(3-36) ), receptor agonist enhanced proliferation. In addition, no NPY-induced DG cellular proliferation is observed following NPY injection when coadministered with a Y1 antagonist or in the Y1 receptor knockout mouse. These results are in line with data obtained in Y1(-/-) mice, demonstrating that NPY regulates in vivo hippocampal neurogenesis. © 2010 Wiley-Liss, Inc.


Subject(s)
Dentate Gyrus/metabolism , Neural Stem Cells/metabolism , Neurogenesis , Neuropeptide Y/administration & dosage , Receptors, Neuropeptide Y/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Proliferation/drug effects , Dentate Gyrus/cytology , Dentate Gyrus/drug effects , Injections, Intraventricular , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurogenesis/drug effects , Neurogenesis/physiology , Neurons/drug effects , Neurons/metabolism , Neuropeptide Y/chemistry , Neuropeptide Y/metabolism , Receptors, Neuropeptide Y/agonists , Receptors, Neuropeptide Y/antagonists & inhibitors , Receptors, Neuropeptide Y/deficiency
7.
Neuroreport ; 21(5): 349-53, 2010 Mar 31.
Article in English | MEDLINE | ID: mdl-20147861

ABSTRACT

The major histocompatibility complex (MHC) class I has a role in the regulation of immune responses and has been implicated recently in neural plasticity and neurogenesis. We therefore sought to investigate in functional MHC class I knockout mice, transporter associated with antigen processing 1 (TAP1) KO, whether there are alterations in adult neurogenesis. We found no significant differences in cell proliferation or neurogenesis in either the dentate gyrus or subventricular zone, in TAP1 KO versus wild-type mice at several different time points. Our results do not support a role for MHC class I in adult neurogenesis, although it may still have a role in the maturation and integration of newborn neurons.


Subject(s)
ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Genes, MHC Class I , Neurogenesis/genetics , Neurogenesis/physiology , ATP Binding Cassette Transporter, Subfamily B, Member 2 , Adult Stem Cells/physiology , Aging , Animals , Anxiety/genetics , Anxiety/metabolism , Brain/growth & development , Brain/physiology , Cell Proliferation , Dentate Gyrus/growth & development , Dentate Gyrus/physiology , Maze Learning/physiology , Memory/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/physiology , Space Perception/physiology , Stem Cell Niche/growth & development , Stem Cell Niche/physiology
8.
Proc Natl Acad Sci U S A ; 106(21): 8754-9, 2009 May 26.
Article in English | MEDLINE | ID: mdl-19433789

ABSTRACT

A reduction in dopaminergic innervation of the subventricular zone (SVZ) is responsible for the impaired proliferation of its resident precursor cells in this region in Parkinson's disease (PD). Here, we show that this effect involves EGF, but not FGF2. In particular, we demonstrate that dopamine increases the proliferation of SVZ-derived cells by releasing EGF in a PKC-dependent manner in vitro and that activation of the EGF receptor (EGFR) is required for this effect. We also show that dopamine selectively expands the GFAP(+) multipotent stem cell population in vitro by promoting their self-renewal. Furthermore, in vivo dopamine depletion leads to a decrease in precursor cell proliferation in the SVZ concomitant with a reduction in local EGF production, which is reversed through the administration of the dopamine precursor levodopa (L-DOPA). Finally, we show that EGFR(+) cells are depleted in the SVZ of human PD patients compared with age-matched controls. We have therefore demonstrated a unique role for EGF as a mediator of dopamine-induced precursor cell proliferation in the SVZ, which has potential implications for future therapies in PD.


Subject(s)
Aging/physiology , Cell Differentiation , Dopamine/pharmacology , Epidermal Growth Factor/metabolism , Neurons/cytology , Neurons/metabolism , Animals , Cell Proliferation/drug effects , Enzyme Activation , ErbB Receptors/metabolism , Female , Parkinson Disease/metabolism , Parkinson Disease/pathology , Rats , Rats, Sprague-Dawley
9.
J Anat ; 214(3): 396-405, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19245506

ABSTRACT

Cell replacement therapies offer promise in the treatment of neurotrauma and neurodegenerative disorders and have concentrated on the use of primary fetal brain tissue. However, there is a growing promise of using neural stem cells, in which case other factors may be important in their successful engraftment. We therefore investigated whether the co-expression of the major developmental transcription factor (Pax7 in this study) of donor tissue to graft site influences transplant survival and differentiation in the rat midbrain. Neural progenitor cells were prepared from either the Pax7-expressing dorsal (DM) or non-Pax7-expressing ventral mesencephalon (VM) of embryonic EGFP(+/+) rats. Cells were dissociated and grafted into the adult rat superior colliculus (SC) lesioned with quinolinic acid 3 days previously, a time shown to be associated with the up-regulation of Pax7. Grafts were then examined 4 weeks later. Our results suggest the origin of the graft tissue did not alter graft survival in the SC; however, dorsal grafts appear to have a higher incidence of neuronal survival, whereas ventral grafts have a higher incidence of astrocytic survivors.


Subject(s)
Brain Tissue Transplantation/methods , Fetal Stem Cells/transplantation , Graft Survival , Mesencephalon/transplantation , Paired Box Transcription Factors/metabolism , Animals , Astrocytes/cytology , Cell Survival , Cells, Cultured , Fetal Stem Cells/metabolism , Fetal Tissue Transplantation/methods , Mesencephalon/embryology , Mesencephalon/metabolism , Rats , Rats, Sprague-Dawley , Rats, Transgenic
10.
Brain ; 131(Pt 3): 630-41, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18202103

ABSTRACT

Neural stem cells (NSCs) are widely endorsed as a cell source for replacement strategies in neurodegenerative disease. However, their usefulness is currently limited by the inability to induce specific neurotransmitter phenotypes in these cells. In order to direct dopaminergic neuronal fate, we overexpressed Pitx3 in NSCs that were then exposed to E11 developing ventral mesencephalon (VM) in explant culture. This resulted in a significant potentiation of dopaminergic differentiation of the cells. When transplanted into the 6-hydroxydopamine lesioned Parkinsonian rats, these cografts of VM and Pitx3 overexpressing NSCs resulted in a significant restitution of motor function. In addition, there were greater numbers of Girk2 positive A9 neurons in the periphery of the transplants that were NSC derived. This demonstrates that given the correct signals, NSCs can be induced to become dopaminergic neurons that can differentiate into the correct nigrastriatal phenotype required for the treatment of Parkinson's disease.


Subject(s)
Brain Tissue Transplantation/methods , Dopamine/biosynthesis , Parkinson Disease/therapy , Stem Cell Transplantation/methods , Animals , Cell Differentiation , Cell Survival , Coculture Techniques , Disease Models, Animal , Female , Gene Transfer Techniques , Genetic Vectors , Homeodomain Proteins/metabolism , Lentivirus/genetics , Mesencephalon/transplantation , Motor Activity , Neurons/pathology , Neurons/transplantation , Parkinson Disease/metabolism , Parkinson Disease/physiopathology , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction/methods , Tissue Culture Techniques , Transcription Factors/metabolism
11.
Neurobiol Dis ; 25(3): 594-608, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17188499

ABSTRACT

Intrastriatal transplantation of fetal ventral mesencephalon (VM) tissue provides the potential to alleviate motor symptoms of Parkinson's disease (PD) and levodopa-induced dyskinesia (LID). However, the degree of recovery varies among individuals with an incidence of "off-phase", graft-induced dyskinesia (GID) in some patients. We hypothesised that this variability is due to the heterogeneous nature of dopaminergic neurons in the transplant. We therefore investigated this in the unilateral 6-hydroxydopamine-lesioned rat model of PD. These animals were primed to develop LID and then transplanted with fetal VM into the caudal aspects of the striatum. No GID was observed but in a significant number of animals the transplants ameliorated LID. There was a correlation between the degree of behavioural and LID recovery with the number of A9 dopaminergic neurons in the transplant, based on their expression of a G-protein-coupled inward rectifying current potassium channel (Girk2). Furthermore, we showed that LID development is related to an abnormal expression profile of cyclin-dependent kinase 5 (Cdk5) and dopamine- and cAMP-regulated phosphoprotein of 32 kDa (DARPP-32) in the striatum and that intrastriatal VM transplants normalised both Cdk5 expression and DARPP-32 phosphorylation in animals exhibiting functional improvement. These results suggest that an A9 dopaminergic neuron-enriched transplant may be the key to an effective PD cell replacement therapy through normalisation of the altered striatal expression of Cdk5/DARPP-32.


Subject(s)
Dopamine Agents/adverse effects , Dyskinesia, Drug-Induced/surgery , Levodopa/adverse effects , Mesencephalon/transplantation , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/surgery , Animals , Behavior, Animal , Brain Tissue Transplantation , Combined Modality Therapy , Corpus Striatum/cytology , Corpus Striatum/metabolism , Corpus Striatum/surgery , Dopamine/physiology , Female , Fetal Tissue Transplantation , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Mesencephalon/cytology , Motor Skills , Neurons/metabolism , Oxidopamine , Rats , Rats, Sprague-Dawley , Recovery of Function , Severity of Illness Index , Sympatholytics
12.
Brain Res Bull ; 68(1-2): 83-94, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16325008

ABSTRACT

EGF and FGF-2 induce the proliferation of embryonic neural precursors (ENPs) in vitro from a number of different species. In this study, we demonstrate that embryonic age is a crucial determinant of the number and differentiation potential of rat embryonic neural precursor cells responding to either EGF and/or FGF-2, in that (i) there is a differential response to the two growth factors (both alone and in combination) according to the gestational age of isolation and (ii) when allowed to differentiate, there are temporal changes in the ability of these cells to produce neurons. Furthermore, for cultures of all gestational ages, there is a defined pattern of senescence, with cultures expanding longest when cells are isolated earlier in gestation. The suggestion is that rat ENPs in this study consist predominantly of neural progenitor cells with limited division potential rather than self-renewing multipotential neural stem cells. In contrast, mouse ENPs appeared to expand indefinitely and thus allow for longer studies to be carried out looking at the effects of growth factor concentrations. The effect of varying the concentration of EGF was assessed using mouse ENPs.


Subject(s)
Cell Culture Techniques/methods , Epidermal Growth Factor/pharmacology , Fibroblast Growth Factor 2/pharmacology , Neurons/cytology , Stem Cells/drug effects , Animals , Anticoagulants/pharmacology , Brain Stem/cytology , Cell Differentiation/drug effects , Cell Division/drug effects , Corpus Striatum/cytology , Female , Fetus/cytology , Gestational Age , Heparin/pharmacology , Mice , Pregnancy , Rats , Stem Cells/cytology
13.
Cell Transplant ; 12(7): 687-95, 2003.
Article in English | MEDLINE | ID: mdl-14653616

ABSTRACT

Transplantation of human fetal CNS tissue is a promising therapy for neurodegenerative conditions such as Huntington's disease (HD), but its widespread adoption is limited by restricted tissue availability. One method of overcoming this problem would be to store the tissue in hibernation medium, an approach that we reported previously for human fetal striatal tissue stored for up to 24 h. We now demonstrate the feasibility of storing such tissue for up to 8 days in hibernation medium. When either fresh or 8-day hibernated striatal cells were cultured under standard conditions for 4 days, the proportion of DARPP-32-positive neurons did not differ significantly, although the total number of cells was significantly less from tissue that had been hibernated. Six weeks after transplantation into cyclosporin A-immunosuppressed unilateral quinolinic acid-lesioned rats, there was no significant difference between fresh and hibernated grafts, both in terms of graft volume and extent of striatal phenotypic markers. This study therefore clearly demonstrates that hibernation of human fetal striatal tissue for up to 8 days is not deleterious to its differentiation in culture or survival following transplantation, and is therefore an appropriate method of storage for this tissue.


Subject(s)
Brain Tissue Transplantation/methods , Fetal Tissue Transplantation/methods , Neostriatum/transplantation , Tissue Preservation/methods , Animals , Cell Differentiation , Cells, Cultured , Clinical Trials as Topic , Female , Graft Survival , Humans , Huntington Disease/surgery , Neostriatum/embryology , Neurons/ultrastructure , Pregnancy , Rats
14.
Exp Neurol ; 175(1): 98-111, 2002 May.
Article in English | MEDLINE | ID: mdl-12009763

ABSTRACT

Neural precursors with the properties of neural stem cells can be isolated from the developing brain, can be expanded in culture, and have been suggested as a potential source of cells for neuronal replacement therapies in degenerative disorders such as Parkinson's disease (PD). Under such conditions an improved spectrum of functional benefit may be obtained through homotypic reconstruction of degenerated neural circuitry, and to this end we have investigated the potential of expanded neural precursor cells (ENPs) to form long axonal projections following transplantation in the 6-hydroxydopamine-lesioned rat model of PD. ENPs have been isolated from the embryonic pig, since implantation in a xenograft environment is thought to favor axonal growth. These porcine ENPs possessed similar properties in vitro to those described in other species: they proliferated in response to epidermal and fibroblast growth factor-2, expressed the neuroepithelial marker nestin, and differentiated into neurons, astrocytes, and occasional oligodendrocytes on mitogen withdrawal. The use of pig-specific markers following xenotransplantion into cyclosporin A-immunosuppressed rats revealed that many cells differentiated into neurons and displayed extensive axogenesis, such that when placed in the region of the substantia nigra fibers projected throughout the striatal neuropil. These neurons were not restricted in the targets to which they could project since following intrastriatal grafting fibers were seen in the normal striatal targets of the pallidum and substantia nigra. Staining for a pig-specific synaptic marker suggested that synapses were formed in these distant sites. A small number of these cells differentiated spontaneously to express a catecholaminergic phenotype, but were insufficient to mediate behavioral recovery. Our results suggest that when the efficiency of neurochemical phenotype induction is increased, ENP-derived neurons have the potential to be a uniquely flexible source of cells for therapeutic cell replacement where anatomical reconstruction is advantageous.


Subject(s)
Nerve Net/cytology , Neurons/cytology , Parkinson Disease, Secondary/therapy , Stem Cell Transplantation , Amphetamines/pharmacology , Animals , Antigens, Differentiation/biosynthesis , Axons/physiology , Behavior, Animal/drug effects , Brain Tissue Transplantation/methods , Cell Count , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Division/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Corpus Striatum/cytology , Disease Models, Animal , Female , Fetal Tissue Transplantation , Fibroblast Growth Factor 2/pharmacology , Graft Survival , Neurons/metabolism , Oxidopamine , Parkinson Disease, Secondary/chemically induced , Phenotype , Rats , Rats, Sprague-Dawley , Stem Cells/cytology , Stem Cells/drug effects , Swine , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...