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










Publication year range
1.
Int J Mol Sci ; 24(4)2023 Feb 13.
Article in English | MEDLINE | ID: mdl-36835161

ABSTRACT

Alzheimer's disease (AD) is the most common neurodegenerative disease in the world. It is classified as familial and sporadic. The dominant familial or autosomal presentation represents 1-5% of the total number of cases. It is categorized as early onset (EOAD; <65 years of age) and presents genetic mutations in presenilin 1 (PSEN1), presenilin 2 (PSEN2), or the Amyloid precursor protein (APP). Sporadic AD represents 95% of the cases and is categorized as late-onset (LOAD), occurring in patients older than 65 years of age. Several risk factors have been identified in sporadic AD; aging is the main one. Nonetheless, multiple genes have been associated with the different neuropathological events involved in LOAD, such as the pathological processing of Amyloid beta (Aß) peptide and Tau protein, as well as synaptic and mitochondrial dysfunctions, neurovascular alterations, oxidative stress, and neuroinflammation, among others. Interestingly, using genome-wide association study (GWAS) technology, many polymorphisms associated with LOAD have been identified. This review aims to analyze the new genetic findings that are closely related to the pathophysiology of AD. Likewise, it analyzes the multiple mutations identified to date through GWAS that are associated with a high or low risk of developing this neurodegeneration. Understanding genetic variability will allow for the identification of early biomarkers and opportune therapeutic targets for AD.


Subject(s)
Alzheimer Disease , Neurodegenerative Diseases , Humans , Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/metabolism , Genome-Wide Association Study , Mutation , Neurodegenerative Diseases/genetics , Presenilin-1/genetics , Presenilin-2/genetics
2.
Int J Mol Sci ; 23(19)2022 Sep 27.
Article in English | MEDLINE | ID: mdl-36232716

ABSTRACT

The α-synucleinopathies constitute a subset of neurodegenerative disorders, of which Parkinson's disease (PD) is the most common worldwide, characterized by the accumulation of misfolded α-synuclein in the cytoplasm of neurons, which spreads in a prion-like manner to anatomically interconnected brain areas. However, it is not clear how α-synucleinopathy triggers neurodegeneration. We recently developed a rat model through a single intranigral administration of the neurotoxic ß-sitosterol ß-D-glucoside (BSSG), which produces α-synucleinopathy. In this model, we aimed to evaluate the temporal pattern of levels in oxidative and nitrosative stress and mitochondrial complex I (CI) dysfunction and how these biochemical parameters are associated with neurodegeneration in different brain areas with α-synucleinopathy (Substantia nigra pars compacta, the striatum, in the hippocampus and the olfactory bulb, where α-syn aggregation spreads). Interestingly, an increase in oxidative stress and mitochondrial CI dysfunction accompanied neurodegeneration in those brain regions. Furthermore, in silico analysis suggests a high-affinity binding site for BSSG with peroxisome proliferator-activated receptors (PPAR) alpha (PPAR-α) and gamma (PPAR-γ). These findings will contribute to elucidating the pathophysiological mechanisms associated with α-synucleinopathies and lead to the identification of new early biomarkers and therapeutic targets.


Subject(s)
Brain , Electron Transport Complex I , Mitochondria , Oxidative Stress , Synucleinopathies , alpha-Synuclein , Animals , Brain/metabolism , Brain/physiopathology , Disease Models, Animal , Electron Transport Complex I/metabolism , Mitochondria/metabolism , Nitrosative Stress , Peroxisome Proliferator-Activated Receptors/metabolism , Rats , Synucleinopathies/metabolism , Synucleinopathies/physiopathology , alpha-Synuclein/chemistry , alpha-Synuclein/metabolism
3.
Cells ; 11(15)2022 07 22.
Article in English | MEDLINE | ID: mdl-35892559

ABSTRACT

Autophagy is a highly conserved lysosomal degradation pathway active at basal levels in all cells. However, under stress conditions, such as a lack of nutrients or trophic factors, it works as a survival mechanism that allows the generation of metabolic precursors for the proper functioning of the cells until the nutrients are available. Neurons, as post-mitotic cells, depend largely on autophagy to maintain cell homeostasis to get rid of damaged and/or old organelles and misfolded or aggregated proteins. Therefore, the dysfunction of this process contributes to the pathologies of many human diseases. Furthermore, autophagy is highly active during differentiation and development. In this review, we describe the current knowledge of the different pathways, molecular mechanisms, factors that induce it, and the regulation of mammalian autophagy. We also discuss its relevant role in development and disease. Finally, here we summarize several investigations demonstrating that autophagic abnormalities have been considered the underlying reasons for many human diseases, including liver disease, cardiovascular, cerebrovascular diseases, neurodegenerative diseases, neoplastic diseases, cancers, and, more recently, infectious diseases, such as SARS-CoV-2 caused COVID-19 disease.


Subject(s)
COVID-19 , Animals , Autophagy/physiology , Homeostasis , Humans , Lysosomes/metabolism , Mammals , SARS-CoV-2
4.
Front Aging Neurosci ; 14: 921573, 2022.
Article in English | MEDLINE | ID: mdl-35847683

ABSTRACT

Alzheimer's disease (AD), a neurodegenerative disorder that can occur in middle or old age, is characterized by memory loss, a continuous decline in thinking, behavioral and social skills that affect the ability of an individual to function independently. It is divided into sporadic and familial subtypes. Early-onset familial AD (FAD) is linked to mutations in genes coding for the amyloid-ß protein precursor (AßPP), presenilin 1 (PS1), and presenilin 2 (PS2), which lead to alterations in AßPP processing, generation of the Amyloid-ß peptide and hyperphosphorylation of tau protein. Identification of early biomarkers for AD diagnosis represents a challenge, and it has been suggested that molecular changes in neurodegenerative pathways identified in the brain of AD patients can be detected in peripheral non-neural cells derived from familial or sporadic AD patients. In the present study, we determined the protein expression, the proteomic and in silico characterization of skin fibroblasts from FAD patients with PS1 mutations (M146L or A246E) or from healthy individuals. Our results shown that fibroblasts from AD patients had increased expression of the autophagy markers LC3II, LAMP2 and Cathepsin D, a significant increase in total GSK3, phosphorylated ERK1/2 (Thr202/Tyr204) and phosphorylated tau (Thr231, Ser396, and Ser404), but no difference in the phosphorylation of Akt (Ser473) or the α (Ser21) and ß (Ser9) GSK3 isoforms, highlighting the relevant role of abnormal protein post-translational modifications in age-related neurodegenerative diseases, such as AD. Both 2-DE gels and mass spectrometry showed significant differences in the expression of the signaling pathways associated with protein folding and the autophagic pathway mediated by chaperones with the expression of HSPA5, HSPE1, HSPD1, HSP90AA1, and HSPE1 and reticular stress in the FAD samples. Furthermore, expression of the heat shock proteins HSP90 and HSP70 was significantly higher in the cells from AD patients as confirmed by Western blot. Taken together our results indicate that fibroblasts from patients with FAD-PS1 present alterations in signaling pathways related to cellular stress, autophagy, lysosomes, and tau phosphorylation. Fibroblasts can therefore be useful in modeling pathways related to neurodegeneration, as well as for the identification of early AD biomarkers.

5.
Diagnostics (Basel) ; 12(5)2022 May 07.
Article in English | MEDLINE | ID: mdl-35626321

ABSTRACT

Alzheimer's disease (AD) is neurodegeneration that accounts for 60-70% of dementia cases. Symptoms begin with mild memory difficulties and evolve towards cognitive impairment. The underlying risk factors remain primarily unclear for this heterogeneous disorder. Bioinformatics is a relevant research tool that allows for identifying several pathways related to AD. Open-access databases of RNA microarrays from the peripheral blood and brain of AD patients were analyzed after background correction and data normalization; the Limma package was used for differential expression analysis (DEA) through statistical R programming language. Data were corrected with the Benjamini and Hochberg approach, and genes with p-values equal to or less than 0.05 were considered to be significant. The direction of the change in gene expression was determined by its variation in the log2-fold change between healthy controls and patients. We performed the functional enrichment analysis of GO using goana and topGO-Limma. The functional enrichment analysis of DEGs showed upregulated (UR) pathways: behavior, nervous systems process, postsynapses, enzyme binding; downregulated (DR) were cellular component organization, RNA metabolic process, and signal transduction. Lastly, the intersection of DEGs in the three databases showed eight shared genes between brain and blood, with potential use as AD biomarkers for blood tests.

6.
Int J Mol Sci ; 22(21)2021 Oct 28.
Article in English | MEDLINE | ID: mdl-34769132

ABSTRACT

Parkinson's disease (PD) is characterized by four pathognomonic hallmarks: (1) motor and non-motor deficits; (2) neuroinflammation and oxidative stress; (3) pathological aggregates of the α-synuclein (α-syn) protein; (4) neurodegeneration of the nigrostriatal system. Recent evidence sustains that the aggregation of pathological α-syn occurs in the early stages of the disease, becoming the first trigger of neuroinflammation and subsequent neurodegeneration. Thus, a therapeutic line aims at striking back α-synucleinopathy and neuroinflammation to impede neurodegeneration. Another therapeutic line is restoring the compromised dopaminergic system using neurotrophic factors, particularly the glial cell-derived neurotrophic factor (GDNF). Preclinical studies with GDNF have provided encouraging results but often lack evaluation of anti-α-syn and anti-inflammatory effects. In contrast, clinical trials have yielded imprecise results and have reported the emergence of severe side effects. Here, we analyze the discrepancy between preclinical and clinical outcomes, review the mechanisms of the aggregation of pathological α-syn, including neuroinflammation, and evaluate the neurorestorative properties of GDNF, emphasizing its anti-α-syn and anti-inflammatory effects in preclinical and clinical trials.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/therapeutic use , Parkinson Disease/metabolism , Protein Aggregation, Pathological , alpha-Synuclein/metabolism , Animals , Clinical Trials as Topic , Disease Models, Animal , Drug Evaluation, Preclinical , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Humans , Neuroinflammatory Diseases/etiology , Parkinson Disease/drug therapy , Parkinson Disease/etiology
7.
PLoS One ; 11(11): e0166370, 2016.
Article in English | MEDLINE | ID: mdl-27832197

ABSTRACT

RhoGDI proteins have been implicated in several human cancers; changes in their expression levels have shown pro- or anti-tumorigenic effects. Pancreatic Ductal Adenocarcinoma (PDAC) is a complex pathology, with poor prognosis, and most patients die shortly after diagnosis. Efforts have been focused on understanding the role of RhoGDI's in PDAC, specially, RhoGDI1 and RhoGDI2. However, the role of RhoGDI3 has not been studied in relation to cancer or to PDAC. Here, we characterized the expression and functionality of RhoGDI3 and its target GTPases, RhoG and RhoB in pancreatic cell lines from both normal pancreatic tissue and tissue in late stages of PDAC, and compared them to human biopsies. Through immunofluorescences, pulldown assays and subcellular fractionation, we found a reduction in RhoGDI3 expression in the late stages of PDAC, and this reduction correlates with tumor progression and aggressiveness. Despite the reduction in the expression of RhoGDI3 in PDAC, we found that RhoB was underexpressed while RhoG was overexpressed, suggesting that cancerous cells preserve their capacity to activate this pathway, thus these cells may be more eager to response to the stimuli needed to proliferate and become invasive unlike normal cells. Surprisingly, we found nuclear localization of RhoGDI3 in non-cancerous pancreatic cell line and normal pancreatic tissue biopsies, which could open the possibility of novel nuclear functions for this protein, impacting gene expression regulation and cellular homeostasis.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Pancreatic Ducts/pathology , Pancreatic Neoplasms/pathology , rho GTP-Binding Proteins/metabolism , rho Guanine Nucleotide Dissociation Inhibitor gamma/metabolism , rhoB GTP-Binding Protein/metabolism , Cell Line, Tumor , Enzyme Activation , Fluorescent Antibody Technique , Humans , rho GTP-Binding Proteins/analysis , rho Guanine Nucleotide Dissociation Inhibitor gamma/analysis , rhoB GTP-Binding Protein/analysis
9.
PLoS One ; 10(9): e0137328, 2015.
Article in English | MEDLINE | ID: mdl-26378780

ABSTRACT

The precise functional role of the dystrophin 71 in neurons is still elusive. Previously, we reported that dystrophin 71d and dystrophin 71f are present in nuclei from cultured neurons. In the present work, we performed a detailed analysis of the intranuclear distribution of dystrophin 71 isoforms (Dp71d and Dp71f), during the temporal course of 7-day postnatal rats hippocampal neurons culture for 1h, 2, 4, 10, 15 and 21 days in vitro (DIV). By immunofluorescence assays, we detected the highest level of nuclear expression of both dystrophin Dp71 isoforms at 10 DIV, during the temporal course of primary culture. Dp71d and Dp71f were detected mainly in bipolar GABAergic (≥60%) and multipolar Glutamatergic (≤40%) neurons, respectively. We also characterized the existence of two nuclear dystrophin-associated protein complexes (DAPC): dystrophin 71d or dystrophin 71f bound to ß-dystroglycan, α1-, ß-, α2-dystrobrevins, α-syntrophin, and syntrophin-associated protein nNOS (Dp71d-DAPC or Dp71f-DAPC, respectively), in the hippocampal neurons. Furthermore, both complexes were localized in interchromatin granule cluster structures (nuclear speckles) of neuronal nucleoskeleton preparations. The present study evinces that each Dp71's complexes differ slightly in dystrobrevins composition. The results demonstrated that Dp71d-DAPC was mainly localized in bipolar GABAergic and Dp71f-DAPC in multipolar Glutamatergic hippocampal neurons. Taken together, our results show that dystrophin 71d, dystrophin 71f and DAP integrate protein complexes, and both complexes were associated to nuclear speckles structures.


Subject(s)
Cell Nucleus/metabolism , Dystrophin-Associated Protein Complex/genetics , Dystrophin/genetics , GABAergic Neurons/cytology , Hippocampus/cytology , Animals , Calcium-Binding Proteins/metabolism , Cells, Cultured , Dystroglycans/metabolism , Dystrophin-Associated Protein Complex/metabolism , Female , Fluorescent Antibody Technique , Membrane Proteins/metabolism , Muscle Proteins/metabolism , Protein Isoforms/genetics , Rats , Rats, Wistar
10.
PLoS One ; 10(3): e0118627, 2015.
Article in English | MEDLINE | ID: mdl-25769033

ABSTRACT

Autism is a neurodevelopmental disorder characterized clinically by impairments in social interaction and verbal and non-verbal communication skills as well as restricted interests and repetitive behavior. It has been hypothesized that altered brain environment including an imbalance in neurotrophic support during early development contributes to the pathophysiology of autism. Here we report that sera from children with autism which exhibited abnormal levels of various neurotrophic factors induced cell death and oxidative stress in mouse primary cultured cortical neurons. The effects of sera from autistic children were rescued by pre-treatment with a ciliary neurotrophic factor (CNTF) small peptide mimetic, Peptide 6 (P6), which was previously shown to exert its neuroprotective effect by modulating CNTF/JAK/STAT pathway and LIF signaling and by enhancing brain derived neurotrophic factor (BDNF) expression. Similar neurotoxic effects and neuroinflammation were observed in young Wistar rats injected intracerebroventricularly with autism sera within hours after birth. The autism sera injected rats demonstrated developmental delay and deficits in social communication, interaction, and novelty. Both the neurobiological changes and the behavioral autistic phenotype were ameliorated by P6 treatment. These findings implicate the involvement of neurotrophic imbalance during early brain development in the pathophysiology of autism and a proof of principle of P6 as a potential therapeutic strategy for autism.


Subject(s)
Autistic Disorder/blood , Autistic Disorder/drug therapy , Ciliary Neurotrophic Factor/chemistry , Neuroprotective Agents/pharmacology , Peptidomimetics/pharmacology , Animals , Autistic Disorder/pathology , Autistic Disorder/physiopathology , Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/genetics , Cell Death/drug effects , Child, Preschool , Developmental Disabilities/drug therapy , Developmental Disabilities/etiology , Female , Gene Expression Regulation/drug effects , Humans , Male , Mice , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Peptidomimetics/therapeutic use , Phenotype , Rats , Social Behavior , Vocalization, Animal/drug effects
11.
ACS Chem Neurosci ; 5(12): 1178-91, 2014 Dec 17.
Article in English | MEDLINE | ID: mdl-25268947

ABSTRACT

Most neurodegenerative diseases are characterized by the presence of protein aggregates. Alzheimer's disease (AD) is the most common cause of dementia in people over age 60. One of the histopathological hallmarks of AD is the presence of tau protein aggregates. Historically, it has been thought that paired helical filaments (PHFs) were the toxic form of tau that assembled to form neurofibrillary tangles (NFTs), but recently there has been evidence that tau oligomers, which form before PHFs and NFTs, could be the structures mediating neurodegeneration even before the fibrillary tau is deposited. Here, we discuss the recent advances in tau oligomer research, their implications on AD and other tauopathies, the mechanisms of tau turnover by the principal protein clearance systems (the proteasome and autophagy), and the potential use of tau oligomers as drug targets for the development of new therapeutic approaches.


Subject(s)
Alzheimer Disease/pathology , Brain/metabolism , tau Proteins/chemistry , tau Proteins/metabolism , Animals , Humans , Models, Biological
12.
PLoS One ; 8(1): e53596, 2013.
Article in English | MEDLINE | ID: mdl-23320097

ABSTRACT

The level of brain-derived neurotrophic factor (BDNF), a member of the neurotrophin family, is down regulated in Alzheimer's disease (AD), Parkinson's disease (PD), depression, stress, and anxiety; conversely the level of this neurotrophin is increased in autism spectrum disorders. Thus, modulating the level of BDNF can be a potential therapeutic approach for nervous system pathologies. In the present study, we designed five different tetra peptides (peptides B-1 to B-5) corresponding to different active regions of BDNF. These tetra peptides were found to be non-toxic, and they induced the expression of neuronal markers in mouse embryonic day 18 (E18) primary hippocampal neuronal cultures. Additionally, peptide B-5 induced the expression of BDNF and its receptor, TrkB, suggesting a positive feedback mechanism. The BDNF peptides induced only a moderate activation (phosphorylation at Tyr 706) of the TrkB receptor, which could be blocked by the Trk's inhibitor, K252a. Peptide B-3, when combined with BDNF, potentiated the survival effect of this neurotrophin on H(2)O(2)-treated E18 hippocampal cells. Peptides B-3 and B-5 were found to work as partial agonists and as partial antagonists competing with BDNF to activate the TrkB receptor in a dose-dependent manner. Taken together, these results suggest that the described BDNF tetra peptides are neurotrophic, can modulate BDNF signaling in a partial agonist/antagonist way, and offer a novel therapeutic approach to neural pathologies where BDNF levels are dysregulated.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Hippocampus/cytology , Hippocampus/metabolism , Nerve Growth Factors/physiology , Neurogenesis/physiology , Peptide Fragments/physiology , Amino Acid Sequence , Animals , Biomarkers/metabolism , Brain-Derived Neurotrophic Factor/genetics , Cells, Cultured , Hippocampus/drug effects , Mice , Molecular Sequence Data , Nerve Growth Factors/metabolism , Neurogenesis/drug effects , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Oligopeptides/genetics , Oligopeptides/metabolism , Oligopeptides/pharmacology , Peptide Fragments/genetics , Receptor, trkB/agonists , Receptor, trkB/antagonists & inhibitors , Receptor, trkB/metabolism
13.
Acta Neuropathol ; 120(5): 605-21, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20697724

ABSTRACT

In addition to the occurrence of numerous neurofibrillary tangles and Aß plaques, neurogenesis and neuronal plasticity are markedly altered in Alzheimer disease (AD). Although the most popular therapeutic approach has been to inhibit neurodegeneration, another is to promote neurogenesis and neuronal plasticity by utilizing the regenerative capacity of the brain. Here we show that, in a transgenic mouse model of AD, 3xTg-AD mice, there was a marked deficit in neurogenesis and neuroplasticity, which occurred before the formation of any neurofibrillary tangles or Aß plaques and was associated with cognitive impairment. Furthermore, peripheral administration of Peptide 6, an 11-mer, which makes an active region of ciliary neurotrophic factor (CNTF, amino acid residues 146-156), restored cognition by enhancing neurogenesis and neuronal plasticity in these mice. Although this treatment had no detectable effect on Aß and tau pathologies in 9-month animals, it enhanced neurogenesis in dentate gyrus, reduced ectopic birth in the granular cell layer, and increased neuronal plasticity in the hippocampus and cerebral cortex. These findings, for the first time, demonstrate the possibility of therapeutic treatment of AD and related disorders by peripheral administration of a peptide corresponding to a biologically active region of CNTF.


Subject(s)
Cognition Disorders/prevention & control , Neurofibrillary Tangles/drug effects , Neurogenesis/drug effects , Neuronal Plasticity/drug effects , Neuroprotective Agents/pharmacology , Plaque, Amyloid/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/prevention & control , Amyloid beta-Peptides/metabolism , Animals , Blotting, Western , Brain/drug effects , Brain/metabolism , Brain/pathology , Chromatography, High Pressure Liquid , Ciliary Neurotrophic Factor/chemistry , Cognition Disorders/metabolism , Cognition Disorders/pathology , Disease Models, Animal , Immunohistochemistry , Maze Learning/drug effects , Memory/drug effects , Mice , Mice, Transgenic , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Peptides/chemical synthesis , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Protein Structure, Secondary , tau Proteins/metabolism
14.
J Hematother Stem Cell Res ; 12(6): 735-48, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14977482

ABSTRACT

Although apoptosis has been considered the typical mechanism for physiological cell death, presently alternative mechanisms need to be considered. We previously showed that fibroblast growth factor-2 (FGF2) could act as a survival factor for neural precursor cells. To study the death mechanism activated by the absence of this growth factor, we followed the changes in cell morphology and determined cell viability by staining with several dyes after FGF2 removal from mesencephalic neural-progenitor-cell cultures. The changes observed did not correspond to those associated with apoptosis. After 48 h in the absence of FGF2, cells began to develop vacuoles in their cytoplasm, a phenotype that became very obvious 3-5 days later. Double-membrane vacuoles containing cell debris were observed. Vacuolated cells did not stain with either ethidium bromide or trypan Blue, and did not show chromatin condensations. Nonetheless, during the course of culture, vacuolated cells formed aggregates with highly condensed chromatin and detached from the plate. Neural progenitor cells grown in the presence of FGF2 did not display any of those characteristics. The vacuolated phenotype could be reversed by the addition of FGF2. Typical autophagy inhibitors such as 3-MA and LY294002 inhibited vacuole development, whereas a broad-spectrum caspase inhibitor did not. Interestingly, Bcl-2 overexpression retarded vacuole development. In conclusion, we identified a death autophagy-like mechanism activated by the lack of a specific survival factor that can be inhibited by Bcl2. We propose that anti-apoptotic Bcl2 family members are key molecules controlling death activation independently of the cell degeneration mechanism used.


Subject(s)
Adenine/analogs & derivatives , Fibroblast Growth Factor 2/physiology , Neurons/cytology , Proto-Oncogene Proteins/physiology , Stem Cells/cytology , Adenine/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Apoptosis Regulatory Proteins , Autophagy/drug effects , Autophagy/physiology , Beclin-1 , Caspase Inhibitors , Cell Death/drug effects , Cell Death/physiology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Survival/drug effects , Cell Survival/physiology , Chromones/pharmacology , Cycloheximide/pharmacology , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/pharmacology , Fibroblast Growth Factor 2/deficiency , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation/genetics , Glial Fibrillary Acidic Protein/analysis , Green Fluorescent Proteins , Immunohistochemistry , Intermediate Filament Proteins/analysis , Luminescent Proteins/genetics , Mesencephalon/cytology , Mice , Microscopy, Electron , Morpholines/pharmacology , Nerve Tissue Proteins/analysis , Nestin , Neurons/drug effects , Neurons/physiology , Proteins/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2 , Stem Cells/drug effects , Stem Cells/physiology , Transformation, Genetic/genetics , Tubulin/analysis , Vacuoles/drug effects , Vacuoles/physiology , Vacuoles/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...