Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Pharmacol Ther ; 260: 108683, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38950869

ABSTRACT

Parkinson's disease (PD) is diagnosed by its cardinal motor symptoms that are associated with the loss of dopamine neurons in the substantia nigra pars compacta (SNc). However, PD patients suffer from various non-motor symptoms years before diagnosis. These prodromal symptoms are thought to be associated with the appearance of Lewy body pathologies (LBP) in brainstem regions such as the dorsal motor nucleus of the vagus (DMV), the locus coeruleus (LC) and others. The neurons in these regions that are vulnerable to LBP are all slow autonomous pacemaker neurons that exhibit elevated oxidative stress due to their perpetual influx of Ca2+ ions. Aggregation of toxic α-Synuclein (aSyn) - the main constituent of LBP - during the long prodromal period challenges these vulnerable neurons, presumably altering their biophysics and physiology. In contrast to pathophysiology of late stage parkinsonism which is well-documented, little is known about the pathophysiology of the brainstem during prodromal PD. In this review, we discuss ion channel dysregulation associated with aSyn aggregation in brainstem pacemaker neurons and their cellular responses to them. While toxic aSyn elevates oxidative stress in SNc and LC pacemaker neurons and exacerbates their phenotype, DMV neurons mount an adaptive response that mitigates the oxidative stress. Ion channel dysregulation and cellular adaptations may be the drivers of the prodromal symptoms of PD. For example, selective targeting of toxic aSyn to DMV pacemakers, elevates the surface density of K+ channels, which slows their firing rate, resulting in reduced parasympathetic tone to the gastrointestinal tract, which resembles the prodromal PD symptoms of dysphagia and constipation. The divergent responses of SNc & LC vs. DMV pacemaker neurons may explain why the latter outlive the former despite presenting LBPs earlier. Elucidation the brainstem pathophysiology of prodromal PD could pave the way for physiological biomarkers, earlier diagnosis and novel neuroprotective therapies for PD.


Subject(s)
Brain Stem , Ion Channels , Parkinson Disease , alpha-Synuclein , Humans , Animals , Brain Stem/metabolism , alpha-Synuclein/metabolism , Parkinson Disease/metabolism , Parkinson Disease/physiopathology , Ion Channels/metabolism , Oxidative Stress , Lewy Bodies/metabolism
2.
Brain Commun ; 4(4): fcac190, 2022.
Article in English | MEDLINE | ID: mdl-35912135

ABSTRACT

Patients with schizophrenia frequently suffer from motor abnormalities, but underlying alterations in neuroarchitecture remain unclear. Here, we aimed to disentangle dyskinesia from parkinsonism in motor structures of patients with schizophrenia and to assess associated molecular architecture. We measured grey matter of motor regions and correlated volumetric estimates with dyskinesia and parkinsonism severity. Associations with molecular architecture were identified by cross-modal spatial correlations between ensuing maps of abnormality-related volume alterations and neurotransmitter maps from healthy populations. Both phenomena were linked to (specific) striatal and basal forebrain reductions as well as to D1 receptor density. Dyskinesia also manifested in cerebellar decrease, while parkinsonism was associated with less motor cortex volume. The parkinsonism-related brain pattern was additionally associated with 5-HT1A/2A and µ-opioid receptors distribution. Findings suggest the need to develop psychopharmacological compounds that display not only selectivity for receptor subtypes but also anatomical selectivity for alleviating dyskinesia without worsening parkinsonism and vice versa.

3.
Sci Rep ; 12(1): 3180, 2022 02 24.
Article in English | MEDLINE | ID: mdl-35210472

ABSTRACT

Parkinson's disease (PD) is clinically defined by the presence of the cardinal motor symptoms, which are associated with a loss of dopaminergic nigrostriatal neurons in the substantia nigra pars compacta (SNpc). While SNpc neurons serve as the prototypical cell-type to study cellular vulnerability in PD, there is an unmet need to extent our efforts to other neurons at risk. The noradrenergic locus coeruleus (LC) represents one of the first brain structures affected in Parkinson's disease (PD) and plays not only a crucial role for the evolving non-motor symptomatology, but it is also believed to contribute to disease progression by efferent noradrenergic deficiency. Therefore, we sought to characterize the electrophysiological properties of LC neurons in two distinct PD models: (1) in an in vivo mouse model of focal α-synuclein overexpression; and (2) in an in vitro rotenone-induced PD model. Despite the fundamental differences of these two PD models, α-synuclein overexpression as well as rotenone exposure led to an accelerated autonomous pacemaker frequency of LC neurons, accompanied by severe alterations of the afterhyperpolarization amplitude. On the mechanistic side, we suggest that Ca2+-activated K+ (SK) channels are mediators of the increased LC neuronal excitability, as pharmacological activation of these channels is sufficient to prevent increased LC pacemaking and subsequent neuronal loss in the LC following in vitro rotenone exposure. These findings suggest a role of SK channels in PD by linking α-synuclein- and rotenone-induced changes in LC firing rate to SK channel dysfunction.


Subject(s)
Norepinephrine/physiology , Parkinson Disease/physiopathology , Pars Compacta/physiology , Small-Conductance Calcium-Activated Potassium Channels/physiology , alpha-Synuclein/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Locus Coeruleus/physiology , Male , Mice , Mice, Inbred C57BL , Neurons/physiology , Prodromal Symptoms , Rotenone
4.
Sci Adv ; 7(11)2021 03.
Article in English | MEDLINE | ID: mdl-33692101

ABSTRACT

No disease-modifying therapy is currently available for Parkinson's disease (PD), the second most common neurodegenerative disease. The long nonmotor prodromal phase of PD is a window of opportunity for early detection and intervention. However, we lack the pathophysiological understanding to develop selective biomarkers and interventions. By using a mutant α-synuclein selective-overexpression mouse model of prodromal PD, we identified a cell-autonomous selective Kv4 channelopathy in dorsal motor nucleus of the vagus (DMV) neurons. This functional remodeling of intact DMV neurons leads to impaired pacemaker function in vitro and in vivo, which, in turn, reduces gastrointestinal motility, a common early symptom of prodromal PD. We identify a chain of events from α-synuclein via a biophysical dysfunction of a specific neuronal population to a clinically relevant prodromal symptom. These findings will facilitate the rational design of clinical biomarkers to identify people at risk for developing PD.


Subject(s)
Channelopathies , Neurodegenerative Diseases , Parkinson Disease , Animals , Humans , Mice , Motor Neurons , Parkinson Disease/etiology , alpha-Synuclein/genetics
5.
Neuroscience ; 412: 248-256, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31202707

ABSTRACT

Since the landmark discovery that point mutations in the α-synuclein gene (SNCA) cause familial Parkinson's disease (PD) more than 2 decades ago, extensive research has been conducted to unravel the molecular and cellular mechanisms by which α-synuclein drives PD pathogenesis resulting in selective neurodegeneration of vulnerable neuronal populations. Current interest focuses on the identification of relevant toxic α-synuclein conformers and their interaction with basic cellular functions. In this context, seminal advances have been made in defining mechanisms of α-synuclein's toxicity in neurons, but many open questions remain regarding its neuronal subtype pathophysiology, as well as the defining the most disease relevant conformations. Moreover, we still only have a partial understanding of the full spectrum of α-synuclein's physiological functions both in neurons and in other cells. In this short review, we focus on cell-specific responses to α-synuclein with a focus on the toxic conformers of α-synuclein. We will not discuss more general cellular death pathways, which have been comprehensively covered by a number of elegant recent reviews.


Subject(s)
Brain/metabolism , Neurons/metabolism , Parkinson Disease/genetics , Point Mutation , alpha-Synuclein/genetics , Animals , Brain/pathology , Humans , Oxidative Stress/physiology , Parkinson Disease/metabolism , Parkinson Disease/pathology , alpha-Synuclein/metabolism
6.
eNeuro ; 6(1)2019.
Article in English | MEDLINE | ID: mdl-30671536

ABSTRACT

Cholinergic interneurons (CINs) are believed to form synchronous cell assemblies that modulate the striatal microcircuitry and possibly orchestrate local dopamine release. We expressed GCaMP6s, a genetically encoded calcium indicator (GECIs), selectively in CINs, and used microendoscopes to visualize the putative CIN assemblies in the dorsal striatum of freely moving mice. The GECI fluorescence signal from the dorsal striatum was composed of signals from individual CIN somata that were engulfed by a widespread fluorescent neuropil. Bouts of synchronous activation of the cholinergic neuropil revealed patterns of activity that preceded the signal from individual somata. To investigate the nature of the neuropil signal and why it precedes the somatic signal, we target-patched GECI-expressing CINs in acute striatal slices in conjunction with multiphoton imaging or wide-field imaging that emulates the microendoscopes' specifications. The ability to detect fluorescent transients associated with individual action potential was constrained by the long decay constant of GECIs (relative to common inorganic dyes) to slowly firing (<2 spikes/s) CINs. The microendoscopes' resolving power and sampling rate further diminished this ability. Additionally, we found that only back-propagating action potentials but not synchronous optogenetic activation of thalamic inputs elicited observable calcium transients in CIN dendrites. Our data suggest that only bursts of CIN activity (but not their tonic firing) are visible using endoscopic imaging, and that the neuropil patterns are a physiological measure of the collective recurrent CIN network spiking activity.


Subject(s)
Action Potentials , Corpus Striatum/physiology , Interneurons/physiology , Motor Activity/physiology , Neuropil/physiology , Animals , Calcium/metabolism , Calcium Signaling , Corpus Striatum/cytology , Female , Interneurons/cytology , Male , Mice, Transgenic , Microscopy, Confocal , Spatio-Temporal Analysis , Tissue Culture Techniques
7.
Eur J Neurosci ; 49(6): 824-833, 2019 03.
Article in English | MEDLINE | ID: mdl-28922504

ABSTRACT

The widely held view that the pathophysiology of Parkinson's disease arises from an under-activation of the direct pathway striatal spiny neurons (dSPNs) has gained support from a recently described weakening of the glutamatergic projection from the parafascicular nucleus (PfN) to dSPNs in experimental parkinsonism. However, the impact of the remodeling of the thalamostriatal projection cannot be fully appreciated without considering its impact on cholinergic interneurons (ChIs) that themselves preferentially activate indirect pathway spiny neurons (iSPNs). To study this thalamostriatal projection, we virally transfected with Cre-dependent channelrhodopsin-2 (ChR2) the PfN of Vglut2-Cre mice that were dopamine-depleted with 6-hydroxydopamine (6-OHDA). In parallel, we studied the corticostriatal projection to ChIs in 6-OHDA-treated transgenic mice expressing ChR2 under the Thy1 promoter. We found the 6-OHDA lesions failed to affect short-term synaptic plasticity or the size of unitary responses evoked optogenetically in either of these projections. However, we found that NMDA-to-AMPA ratios at PfN synapses-that were significantly larger than NMDA-to-AMPA ratios at cortical synapses-were reduced by 6-OHDA treatment, thereby impairing synaptic integration at PfN synapses onto ChIs. Finally, we found that application of an agonist of the D5 dopamine receptors on ChIs potentiated NMDA currents without affecting AMPA currents or short-term plasticity selectively at PfN synapses. We propose that dopamine depletion leads to an effective de-potentiation of NMDA currents at PfN synapses onto ChIs which degrades synaptic integration. This selective remodeling of NMDA currents at PfN synapses may counter the selective weakening of PfN synapses onto dSPNs in parkinsonism.


Subject(s)
Cholinergic Neurons/metabolism , Dopamine/metabolism , Synapses/physiology , Synaptic Transmission/physiology , Animals , Corpus Striatum/metabolism , Intralaminar Thalamic Nuclei/metabolism , Male , Mice, Transgenic , Neural Pathways/physiology , Neurons/physiology , Parkinson Disease/physiopathology , Parkinsonian Disorders/physiopathology
8.
Acta Neuropathol Commun ; 6(1): 39, 2018 05 10.
Article in English | MEDLINE | ID: mdl-29747690

ABSTRACT

Degeneration of noradrenergic locus coeruleus neurons occurs during the prodromal phase of Parkinson's disease and contributes to a variety of non-motor symptoms, e.g. depression, anxiety and REM sleep behavior disorder. This study was designed to establish the first locus coeruleus α-synucleinopathy mouse model, which should provide sufficient information about the time-course of noradrenergic neurodegeneration, replicate cardinal histopathological features of the human Parkinson's disease neuropathology and finally lead to robust histological markers, which are sufficient to assess the pathological changes in a quantitative and qualitative way. We show that targeted viral vector-mediated overexpression of human mutant A53T-α-synuclein in vivo in locus coeruleus neurons of wild-type mice resulted in progressive noradrenergic neurodegeneration over a time frame of 9 weeks. Observed neuronal cell loss was accompanied by progressive α-synuclein phosphorylation, formation of proteinase K-resistant α-synuclein-aggregates, accumulation of Ubi-1- and p62-positive inclusions in microglia and induction of progressive micro- and astrogliosis. Apart from this local pathology, abundant α-synuclein-positive axons were found in locus coeruleus output regions, indicating rapid anterograde axonal transport of A53T-α-synuclein. Taken together, we present the first model of α-synucleinopathy in the murine locus coeruleus, replicating essential morphological features of human Parkinson's disease pathology. This new model may contribute to the research on prodromal Parkinson's disease, in respect to pathophysiology and the development of disease-modifying therapy.


Subject(s)
Locus Coeruleus/cytology , Mutation/genetics , Neuroglia/metabolism , Neurons/metabolism , Parkinson Disease/pathology , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , Alanine/genetics , Animals , Calcium-Binding Proteins , Disease Models, Animal , Endopeptidase K/pharmacology , Humans , Locus Coeruleus/metabolism , Male , Mice , Mice, Inbred C57BL , Microfilament Proteins , Nerve Tissue Proteins/metabolism , Neuroglia/drug effects , Neuroglia/pathology , Neurons/drug effects , Neurons/pathology , Parkinson Disease/genetics , Protein Aggregates/drug effects , Protein Transport/drug effects , Protein Transport/genetics , Threonine/genetics , Time Factors , Transduction, Genetic , Tyrosine 3-Monooxygenase/metabolism
9.
Sci Rep ; 7(1): 11469, 2017 09 13.
Article in English | MEDLINE | ID: mdl-28904388

ABSTRACT

α-synuclein-induced neurotoxicity is a core pathogenic event in neurodegenerative synucleinopathies such as Parkinson's disease, dementia with Lewy bodies, or multiple system atrophy. There is currently no disease-modifying therapy available for these diseases. We screened 1,600 FDA-approved drugs for their efficacy to protect LUHMES cells from degeneration induced by wild-type α-synuclein and identified dipyridamole, a non-selective phosphodiesterase inhibitor, as top hit. Systematic analysis of other phosphodiesterase inhibitors identified a specific phosphodiesterase 1 inhibitor as most potent to rescue from α-synuclein toxicity. Protection was mediated by an increase of cGMP and associated with the reduction of a specific α-synuclein oligomeric species. RNA interference experiments confirmed PDE1A and to a smaller extent PDE1C as molecular targets accounting for the protective efficacy. PDE1 inhibition also rescued dopaminergic neurons from wild-type α-synuclein induced degeneration in the substantia nigra of mice. In conclusion, this work identifies inhibition of PDE1A in particular as promising target for neuroprotective treatment of synucleinopathies.


Subject(s)
Drug Discovery , Enzyme Inhibitors/pharmacology , Phosphodiesterase I/antagonists & inhibitors , Protein Aggregation, Pathological/metabolism , alpha-Synuclein/metabolism , Animals , Cell Line , Dipyridamole/pharmacology , Drug Evaluation, Preclinical , High-Throughput Screening Assays , Humans , Mice , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Protein Aggregation, Pathological/drug therapy , Vinca Alkaloids/pharmacology , alpha-Synuclein/antagonists & inhibitors
10.
J Neurosci ; 37(1): 47-57, 2017 01 04.
Article in English | MEDLINE | ID: mdl-28053029

ABSTRACT

α-Synuclein overexpression (ASOX) drives the formation of toxic aggregates in neurons vulnerable in Parkinson's disease (PD), including dopaminergic neurons of the substantia nigra (SN) and cholinergic neurons of the dorsal motor nucleus of the vagus (DMV). Just as these populations differ in when they exhibit α-synucleinopathies during PD pathogenesis, they could also differ in their physiological responses to ASOX. An ASOX-mediated hyperactivity of SN dopamine neurons, which was caused by oxidative dysfunction of Kv4.3 potassium channels, was recently identified in transgenic (A53T-SNCA) mice overexpressing mutated human α-synuclein. Noting that DMV neurons display extensive α-synucleinopathies earlier than SN dopamine neurons while exhibiting milder cell loss in PD, we aimed to define the electrophysiological properties of DMV neurons in A53T-SNCA mice. We found that DMV neurons maintain normal firing rates in response to ASOX. Moreover, Kv4.3 channels in DMV neurons exhibit no oxidative dysfunction in the A53T-SNCA mice, which could only be recapitulated in wild-type mice by glutathione dialysis. Two-photon imaging of redox-sensitive GFP corroborated the finding that mitochondrial oxidative stress was diminished in DMV neurons in the A53T-SNCA mice. This reduction in oxidative stress resulted from a transcriptional downregulation of voltage-activated (Cav) calcium channels in DMV neurons, which led to a reduction in activity-dependent calcium influx via Cav channels. Thus, ASOX induces a homeostatic remodeling with improved redox signaling in DMV neurons, which could explain the differential vulnerability of SN dopamine and DMV neurons in PD and could promote neuroprotective strategies that emulate endogenous homeostatic responses to ASOX (e.g., stressless pacemaking) in DMV neurons. SIGNIFICANCE STATEMENT: Overexpression of mutant α-synuclein causes Parkinson's disease, presumably by driving neurodegeneration in vulnerable neuronal target populations. However, the extent of α-synuclein pathology (e.g., Lewy bodies) is not directly related to the degree of neurodegeneration across various vulnerable neuronal populations. Here, we show that, in contrast to dopamine neurons in the substantia nigra, vagal motoneurons do not enhance their excitability and oxidative load in response to chronic mutant α-synuclein overexpression. Rather, by downregulating their voltage-activated calcium channels, vagal motoneurons acquire a stressless form of pacemaking that diminishes mitochondrial and cytosolic oxidative stress. Emulating this endogenous adaptive response to α-synuclein overexpression could lead to novel strategies to protect dopamine neurons and perhaps delay the onset of Parkinson's disease.


Subject(s)
Biological Clocks , Motor Neurons , Parkinson Disease/physiopathology , Vagus Nerve/physiology , alpha-Synuclein/biosynthesis , alpha-Synuclein/genetics , Animals , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium Signaling/genetics , Dopaminergic Neurons/physiology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/metabolism , Oxidative Stress , Shal Potassium Channels/metabolism , Signal Transduction/genetics , Substantia Nigra/cytology , Substantia Nigra/physiology , Vagus Nerve/cytology
11.
Neuroscience ; 331: 120-33, 2016 09 07.
Article in English | MEDLINE | ID: mdl-27320210

ABSTRACT

l-3,4-Dihydroxyphenylalanine (l-DOPA) is the therapeutic gold standard in Parkinson's disease. However, most patients develop debilitating abnormal involuntary movements termed l-DOPA-induced dyskinesia (LID) as therapy-complicating side effects. The underlying mechanisms of LID pathogenesis are still not fully understood. Recent evidence suggests an involvement of striatal tyrosine hydroxylase (TH) protein-expressing neurons, as they are capable of endogenously producing l-DOPA and possibly dopamine. The aim of this study was to elucidate changes of TH transcription in the striatum and nucleus accumbens that occur under experimental conditions of LID. Mice with a unilateral 6-hydroxydopamine-induced lesion of the medial forebrain bundle were treated daily with l-DOPA for 15days to provoke dyskinesia. In situ hybridization analysis revealed a significant numerical decrease of TH mRNA-positive neurons in the striatum and nucleus accumbens of mice not exhibiting LID, whereas dyskinetic animals failed to show this reduction of TH transcription. Interestingly, similar changes were observed in intact non-deafferentiated striata, demonstrating an l-DOPA-responsive transcriptional TH regulation independently from nigrostriatal lesion severity. Consolidation with our previous study on TH protein level (Keber et al., 2015) impressively highlights that LID is associated with both a deficient downregulation of TH transcription and an excessive translation of TH protein in intrastriatal neurons. As TH protein levels in comparison to mRNA levels showed a stronger correlation with development and severity of LID, antidyskinetic treatment strategies should focus on translational and posttranslational modulations of TH as a promising target.


Subject(s)
Antiparkinson Agents/adverse effects , Corpus Striatum/drug effects , Dyskinesia, Drug-Induced/metabolism , Levodopa/adverse effects , Neurons/drug effects , Tyrosine 3-Monooxygenase/metabolism , Animals , Corpus Striatum/metabolism , Corpus Striatum/pathology , Disease Models, Animal , Down-Regulation/drug effects , Dyskinesia, Drug-Induced/pathology , Enkephalins/metabolism , Male , Medial Forebrain Bundle , Mice, Inbred C57BL , Neuronal Plasticity , Neurons/metabolism , Neurons/pathology , Oxidopamine , Protein Precursors/metabolism , RNA, Messenger/metabolism
12.
Neuropharmacology ; 79: 212-21, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24291466

ABSTRACT

Olfactory dysfunction, often preceding the cardinal motor symptoms, such as bradykinesia, rigidity, tremor at rest and postural instability, is frequently reported in Parkinson's disease. This symptom appears to be related to an increased number of dopamine neurons in the periglomerular layer of the olfactory bulb. In animal models of Parkinson's disease, adult neural progenitor cells migrating from the subventricular zone of the lateral ventricle to the olfactory bulb are evidently altered in their survival and progeny. The modulation of neural progenitor cells contributing to the number of dopamine neurons in the periglomerular layer, however, is still poorly understood. In this study, we have investigated the survival and neuronal differentiation of newly generated cells in the olfactory bulb, following treatment with the dopamine precursor l-DOPA and the monoamine oxidase-B inhibitor selegiline in a unilateral, intranigral 6-hydroxydopamine lesion model in mice. Our data show that the number of neural progenitor cells in the subventricular zone is decreased after an intranigral 6-hydroxydopamine lesion, while there is no difference from control in lesioned mice with selegiline or l-DOPA treatment. Selegiline is able to normalize the number of dopamine neurons in the periglomerular layer, while l-DOPA treatment sustains the increased number observed in 6-hydroxydopamine lesioned animals. We conclude that there is a distinct modulation of newly generated dopamine neurons of the olfactory bulb after l-DOPA and selegiline treatment. The differential effects of the two drugs might also play a role in olfactory dysfunction in Parkinson's disease patients.


Subject(s)
Antiparkinson Agents/pharmacology , Dopaminergic Neurons/drug effects , Levodopa/pharmacology , Olfactory Bulb/drug effects , Parkinsonian Disorders/drug therapy , Selegiline/pharmacology , Animals , Cell Count , Cell Survival/drug effects , Dopamine Agents/pharmacology , Dopaminergic Neurons/physiology , Hippocampus/drug effects , Hippocampus/physiopathology , Male , Mice , Mice, Inbred C57BL , Monoamine Oxidase Inhibitors/pharmacology , Neural Stem Cells/drug effects , Neural Stem Cells/physiology , Neurogenesis/drug effects , Neuroprotective Agents/pharmacology , Olfactory Bulb/physiopathology , Oxidopamine , Parkinsonian Disorders/physiopathology
13.
J Neural Transm Suppl ; (73): 279-85, 2009.
Article in English | MEDLINE | ID: mdl-20411786

ABSTRACT

The clinical motor dysfunction in Parkinson's disease is primarily the consequence of a progressive degeneration of dopaminergic neurons in the substantia nigra of the nigrostriatal pathway. The degeneration of this tract provokes a depletion of dopamine in the striatum, where it is required as a permissive factor for normal motor function. Despite intense investigations, no effective therapy is available to prevent the onset or to halt the progression of the neuronal cell loss. Therefore, recent years have seen research into the mechanisms of endogenous repair processes occurring in the adult brain, particularly in the substantia nigra. Neurogenesis occurs in the adult brain in a constitutive manner under physiological circumstances within two regions: the dentate gyrus of the hippocampus and the subventricular zone of the lateral ventricles. In contrast to these two so-called neurogenic areas, the remainder of the brain is considered to be primarily nonneurogenic in nature, implying that no new neurons are produced there under normal conditions. The occurrence of adult neurogenesis in the substantia nigra under the pathological conditions of Parkinson's disease, however, remains controversial. Here, we review the published evidence of whether adult neurogenesis exists or not within the substantia nigra, where dopaminergic neurons are lost in Parkinson's disease.


Subject(s)
Neurogenesis/physiology , Neurons/pathology , Parkinson Disease/pathology , Substantia Nigra/physiopathology , Animals , Humans , Models, Biological
SELECTION OF CITATIONS
SEARCH DETAIL
...