Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Stem Cells Dev ; 21(8): 1356-66, 2012 May 20.
Article in English | MEDLINE | ID: mdl-21740235

ABSTRACT

Epidermal growth factor (EGF) is a mitogen widely used when culturing adult neural stem cells in vitro. Although proliferative effects can also be observed in vivo, intracerebroventricular infusion of EGF has been found to counteract neuronal determination and promote glial differentiation instead. However, EGF receptor activation has different effects on the subventricular zone (SVZ) in mice and rats, possibly because of species differences in SVZ cell composition. Specifically in the rat, EGF stimulation of the SVZ induces the formation of hyperplastic polyps. The present study aims at molecular and morphological characterization of these subventricular polyps. Using immunohistochemistry, electron microscopy, and gene expression analysis, we demonstrate in hyperplastic EGF-induced polyps an upregulation in protein expression of Sox2, Olig2, GFAP, nestin, and vimentin. We found polyp-specific dysplastic changes in the form of coexpression of Sox2 and Olig2. This highly proliferative, Sox2/Olig2 coexpressing dysplastic cell type is >10-fold enriched in the hyperplastic polyps compared with control SVZ and most likely causes the polyp formation. Unique ultrastructural features of the polyps include a lack of ependymal cell lining as well as a large number of cells with large, light, ovoid nuclei and a cytoplasm with abundant ribosomes, whereas other polyp cells contain invaginated nuclei but fewer ribosomes. EGF also induced changes in the expression of Id genes Id1, Id2, and Id4 in the SVZ. Taken together, we here demonstrate dysplastic, structural, and phenotypical changes in the rat SVZ following EGF stimulation, which are specific to hyperplastic polyps.


Subject(s)
Aging/drug effects , Aging/pathology , Epidermal Growth Factor/pharmacology , Lateral Ventricles/drug effects , Lateral Ventricles/pathology , Animals , Biomarkers/metabolism , Cell Proliferation/drug effects , Gene Expression Regulation/drug effects , Humans , Hyperplasia , Inhibitor of Differentiation Proteins/genetics , Inhibitor of Differentiation Proteins/metabolism , Lateral Ventricles/ultrastructure , Male , Mice , Microscopy, Confocal , Models, Biological , Polyps/pathology , Polyps/ultrastructure , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Stem Cells/cytology , Stem Cells/drug effects
2.
J Cereb Blood Flow Metab ; 30(2): 323-35, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19861975

ABSTRACT

Thyroid hormone is critical for the proper development of the central nervous system. However, the specific role of thyroid hormone on brain angiogenesis remains poorly understood. Treatment of rats from birth to postnatal day 21 (P21) with propylthiouracil (PTU), a reversible blocker of triiodothyronine (T3) synthesis, resulted in decreased brain angiogenesis, as indicated by reduced complexity and density of microvessels. However, when PTU was withdrawn at P22, these parameters were fully recovered by P90. These changes were paralleled by an altered expression of vascular endothelial growth factor A (Vegfa) and basic fibroblast growth factor (Fgf2). Physiologic concentrations of T3 and thyroxine (T4) stimulated proliferation and tubulogenesis of rat brain-derived endothelial (RBE4) cells in vitro. Protein and mRNA levels of VEGF-A and FGF-2 increased after T3 stimulation of RBE4 cells. The thyroid hormone receptor blocker NH-3 abolished T3-induced Fgf2 and Vegfa upregulation, indicating a receptor-mediated effect. Thyroid hormone inhibited the apoptosis in RBE4 cells and altered mRNA levels of apoptosis-related genes, namely Bcl2 and Bad. The present results show that thyroid hormone has a substantial impact on vasculature development in the brain. Pathologically altered vascularization could, therefore, be a contributing factor to the neurologic deficits induced by thyroid hormone deficiency.


Subject(s)
Brain/blood supply , Brain/growth & development , Neovascularization, Physiologic/physiology , Thyroxine/metabolism , Triiodothyronine/metabolism , Animals , Antithyroid Agents/toxicity , Fluorescent Antibody Technique , Hypothyroidism/chemically induced , Hypothyroidism/physiopathology , Immunohistochemistry , Microscopy, Electron, Transmission , Microvessels/pathology , Propylthiouracil/toxicity , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
3.
Proc Natl Acad Sci U S A ; 106(49): 20906-11, 2009 Dec 08.
Article in English | MEDLINE | ID: mdl-19948959

ABSTRACT

During early adulthood, a phase in which the central nervous system displays considerable plasticity and in which important cognitive traits are shaped, the effects of exercise on cognition remain poorly understood. We performed a cohort study of all Swedish men born in 1950 through 1976 who were enlisted for military service at age 18 (N = 1,221,727). Of these, 268,496 were full-sibling pairs, 3,147 twin pairs, and 1,432 monozygotic twin pairs. Physical fitness and intelligence performance data were collected during conscription examinations and linked with other national databases for information on school achievement, socioeconomic status, and sibship. Relationships between cardiovascular fitness and intelligence at age 18 were evaluated by linear models in the total cohort and in subgroups of full-sibling pairs and twin pairs. Cardiovascular fitness, as measured by ergometer cycling, positively associated with intelligence after adjusting for relevant confounders (regression coefficient b = 0.172; 95% CI, 0.168-0.176). Similar results were obtained within monozygotic twin pairs. In contrast, muscle strength was not associated with cognitive performance. Cross-twin cross-trait analyses showed that the associations were primarily explained by individual specific, non-shared environmental influences (> or = 80%), whereas heritability explained < 15% of covariation. Cardiovascular fitness changes between age 15 and 18 y predicted cognitive performance at 18 y. Cox proportional-hazards models showed that cardiovascular fitness at age 18 y predicted educational achievements later in life. These data substantiate that physical exercise could be an important instrument for public health initiatives to optimize educational achievements, cognitive performance, as well as disease prevention at the society level.


Subject(s)
Cardiovascular Physiological Phenomena , Cognition/physiology , Physical Fitness/physiology , Adolescent , Cross-Sectional Studies , Education , Humans , Inheritance Patterns/genetics , Intelligence Tests , Longitudinal Studies , Male , Muscle Strength/physiology , Occupations , Proportional Hazards Models , Siblings , Twins , Young Adult
4.
J Endocrinol ; 201(1): 141-50, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19171566

ABSTRACT

IGF-I treatment has been shown to enhance cell genesis in the brains of adult GH- and IGF-I-deficient rodents; however, the influence of GH therapy remains poorly understood. The present study investigated the effects of peripheral recombinant bovine GH (bGH) on cellular proliferation and survival in the neurogenic regions (subventricular zone (SVZ), and dentate gyrus of the hippocampus), as well as the corpus callosum, striatum, parietal cortex, and piriform cortex. Hypopituitarism was induced in female rats by hypophysectomy, and the rats were supplemented with thyroxine and cortisone acetate. Subsequently, the rats received daily s.c. injections of bGH for either 6 or 28 days respectively. Following 5 days of peripheral bGH administration, the number of bromodeoxyuridine (BrdU)-positive cells was increased in the hippocampus, striatum, parietal cortex, and piriform cortex after 6 and 28 days. In the SVZ, however, BrdU-positive cells increased only after 28 days of bGH treatment. No significant change was observed in the corpus callosum. In the hippocampus, after 28 days of bGH treatment, the number of BrdU/NeuN-positive cells was increased proportionally to increase the number of BrdU-positive cells. (3)H-thymidine incorporation in vitro revealed that 24 h of bGH exposure was sufficient to increase cell proliferation in adult hippocampal progenitor cells. This study shows for the first time that 1) peripheral bGH treatment increased the number of newborn cells in the adult brain and 2) bGH exerted a direct proliferative effect on neuronal progenitor cells in vitro.


Subject(s)
Brain/drug effects , Cell Proliferation/drug effects , Growth Hormone/administration & dosage , Administration, Cutaneous , Age Factors , Animals , Brain/physiology , Cells, Cultured , Female , Growth Hormone/pharmacology , Hypophysectomy , Models, Biological , Neurogenesis/drug effects , Rats , Rats, Inbred F344 , Stem Cells/drug effects , Stem Cells/physiology
5.
J Cereb Blood Flow Metab ; 29(2): 342-54, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18985054

ABSTRACT

Regenerative responses after hypoxia-ischemia (HI) were investigated in the immature (P9) and juvenile (P21) mouse striatum and cortex by postischemic 5-bromo-2-deoxyuridine labeling and phenotyping of labeled cells 4 weeks later. HI stimulated the formation of new cells in striatum and cortex in immature, growing brains (P9), but when brain growth was finished (P21) proliferation could be stimulated only in striatum, not in cortex. However, the relative increase was higher in P21 (460%) than P9 striatum (50%), though starting from a lower level at P21. Starting from this lower level, HI-induced proliferation in P21 striatum reached the same level as in P9 striatum, but not higher. Phenotyping revealed that low levels of neurogenesis were still present in nonischemic P9 cortex and striatum, but only in striatum at P21. Ischemia-induced neurogenesis was found only in P9 striatum. Ischemia-induced gliogenesis occurred in P9 and P21 striatum as well as P9 cortex, but not in P21 cortex. Hence, the regenerative response was stronger in striatum than cortex, and stronger in P9 than P21 cortex. The biggest ischemia-induced change was the 49-fold increase in P21 striatal microglia, and this was accompanied by increased inflammation, as judged by the size and numbers of CCL2- and interleukin-18-positive cells.


Subject(s)
Aging/physiology , Cerebrum/pathology , Hypoxia-Ischemia, Brain/pathology , Neurogenesis , Animals , Biomarkers/metabolism , Bromodeoxyuridine , Cell Differentiation , Chemokine CCL2/metabolism , Interleukin-18/metabolism , Male , Mice , Mice, Inbred C57BL , Phenotype
6.
Curr Pharm Biotechnol ; 8(3): 127-31, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17584085

ABSTRACT

Bromodeoxyuridine (BrdU) is widely used for labeling dividing cells to determine their fate. In particular, the analysis of neurogenesis in the adult mammalian brain has made significant progress through the use of this technique. However; when using BrdU for labeling, there are several issues to consider in order to minimalize possible cytotoxicity or false-positive labeling. This current review summarizes methodological and technical aspects of BrdU administration and detection, compares alternative methods and gives recommendations on how to avoid labeling artifacts.


Subject(s)
Artifacts , Bromodeoxyuridine/analysis , Microscopy, Fluorescence/methods , Nerve Regeneration/physiology , Neurons/cytology , Neurons/physiology , Staining and Labeling/methods , Animals , Cell Proliferation , Cells, Cultured , Humans , Image Enhancement/methods , Neurons/drug effects
7.
Stroke ; 38(7): 2165-72, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17510456

ABSTRACT

BACKGROUND AND PURPOSE: The discovery of spontaneous neuronal replacement in the adult brain has shifted experimental stroke therapies toward a combined approach of preventing neuronal cell death and inducing neuronal plasticity. Brain-derived neurotrophic factor (BDNF) was shown to induce antiapoptotic mechanisms after stroke and to reduce infarct size and secondary neuronal cell death. Moreover, in intact animals, BDNF is a potent stimulator of adult neurogenesis. METHODS: The current study analyzed the effects of BDNF on induction of neuronal progenitor cell migration and sensorimotor recovery after cortical photothrombotic stroke. RESULTS: Daily intravenous bolus applications of BDNF during the first 5 days after stroke resulted in significantly improved sensorimotor scores up to 6 weeks. At the structural level, BDNF significantly increased neurogenesis in the dentate gyrus and enhanced migration of subventricular zone progenitor cells to the nearby striatum of the ischemic hemisphere. BDNF treatment could not, however, further stimulate progenitor cell recruitment to the cortex. CONCLUSIONS: These findings consolidate the role of BDNF as a modulator of neurogenesis in the brain and as an enhancer of long-term functional neurological outcome after cerebral ischemia.


Subject(s)
Brain Ischemia/drug therapy , Brain-Derived Neurotrophic Factor/pharmacology , Brain-Derived Neurotrophic Factor/therapeutic use , Neurons , Recovery of Function , Stroke/drug therapy , Animals , Behavior, Animal/physiology , Brain/anatomy & histology , Brain/drug effects , Brain/pathology , Brain/physiology , Brain Ischemia/pathology , Brain Ischemia/rehabilitation , Brain-Derived Neurotrophic Factor/administration & dosage , Cell Differentiation/drug effects , Cell Movement/drug effects , Male , Neurons/drug effects , Neurons/physiology , Random Allocation , Rats , Rats, Wistar , Stem Cells/drug effects , Stroke/pathology , Stroke Rehabilitation , Treatment Outcome
8.
J Cereb Blood Flow Metab ; 27(4): 785-94, 2007 Apr.
Article in English | MEDLINE | ID: mdl-16926844

ABSTRACT

The effects of hypoxia-ischemia (HI) on proliferation and differentiation in the immature (postnatal day 9) and juvenile (postnatal day 21) mouse hippocampus were investigated by injecting bromodeoxyuridine (50 mg/kg) daily for 7 days after the insult and evaluating the labeling 5 weeks after HI. Phenotypic differentiation was evaluated using NeuN, Iba1, APC, and S100beta as markers of neurons, microglia, oligodendrocytes, and astrocytes, respectively. The basal proliferation, in particular neurogenesis, was higher in the immature than in the juvenile hippocampus. Hypoxia-ischemia did not increase neurogenesis significantly in the immature dentate gyrus (DG), but it increased several-fold in the juvenile brain, reaching the same level as in the normal, noninjured immature brain. This suggests that the immature hippocampus is already working at the top of its proliferative capacity and that even though basal neurogenesis decreased with age, the injury-induced generation of new neurons in the juvenile hippocampus could not increase beyond the basal level of the immature brain. Generation of glial cells of all three types after HI was significantly more pronounced in the cornu ammonis of the hippocampus region of the juvenile hippocampus. In the DG, only microglia production was greater in the juvenile brain. Increased microglia proliferation correlated with increased levels of the proinflammatory cytokines MCP-1 and IL-18 3 days after HI, indicating that the inflammatory response is stronger in the juvenile hippocampus. In summary, contrary to what has been generally assumed, our results indicate that the juvenile brain has a greater capacity for neurogenesis after injury than the immature brain.


Subject(s)
Aging/pathology , Hypoxia-Ischemia, Brain/pathology , Inflammation/pathology , Neurons/pathology , Animals , Antimetabolites , Bromodeoxyuridine , Cell Count , Cell Differentiation/drug effects , Cell Proliferation , DNA-Binding Proteins , Dentate Gyrus/pathology , Hippocampus/pathology , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Neuroglia/pathology , Nuclear Proteins/genetics
10.
Brain Pathol ; 14(3): 237-48, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15446578

ABSTRACT

Hypoxia as well as global and focal ischemia are strong activators of neurogenesis in the adult mammalian central nervous system. Here we show that the hypoxia-inducible vascular endothelial growth factor (VEGF) and its receptor VEGFR-2/Flk-1 are expressed in clonally-derived adult rat neural stem cells in vitro. VEGF stimulated the expansion of neural stem cells whereas blockade of VEGFR-2/Flk-1-kinase activity reduced neural stem cell expansion. VEGF was also infused into the lateral ventricle to study changes in neurogenesis in the ventricle wall, olfactory bulb and hippocampus. Using a low dose (2.4 ng/d) to avoid endothelial proliferation and changes in vascular permeability, VEGF stimulated adult neurogenesis in vivo. After VEGF infusion, we observed reduced apoptosis but unaltered proliferation suggesting a survival promoting effect of VEGF in neural progenitor cells. Strong expression of VEGFR-2/Flk-1 was detected in the ventricle wall adjacent to the choroid plexus, a site of significant VEGF production, which suggests a paracrine function of endogenous VEGF on neural stem cells in vivo. We propose that VEGF acts as a trophic factor for neural stem cells in vitro and for sustained neurogenesis in the adult nervous system. These findings may have implications for the pathogenesis and therapy of neurodegenerative diseases.


Subject(s)
Neurons/drug effects , Stem Cells/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Animals , Apoptosis/drug effects , Blood-Brain Barrier/drug effects , Brain/drug effects , Cell Survival/drug effects , Female , Immunohistochemistry , In Situ Nick-End Labeling , Injections, Intraventricular , Rats , Rats, Inbred F344 , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor Receptor-2/biosynthesis , Vascular Endothelial Growth Factor Receptor-2/drug effects
11.
J Neurosci Res ; 77(2): 155-65, 2004 Jul 15.
Article in English | MEDLINE | ID: mdl-15211583

ABSTRACT

Adult neurogenesis has been shown to be regulated by a multitude of extracellular cues, including hormones, growth factors, and neurotransmitters. The cholinergic system of the basal forebrain is one of the key transmitter systems for learning and memory. Because adult neurogenesis has been implicated in cognitive performance, the present work aims at defining the role of cholinergic input for adult neurogenesis by using an immunotoxic lesion approach. The immunotoxin 192IgG-saporin was infused into the lateral ventricle of adult rats to selectively lesion cholinergic neurons of the cholinergic basal forebrain (CBF), which project to the two main regions of adult neurogenesis: the dentate gyrus and the olfactory bulb. Five weeks after lesioning, neurogenesis, defined by the number of cells colocalized for bromodeoxyuridine (BrdU) and the neuronal nuclei marker NeuN, declined significantly in the granule cell layers of the dentate gyrus and olfactory bulb. Furthermore, immunotoxic lesions to the CBF led to increased numbers of apoptotic cells specifically in the subgranular zone, the progenitor region of the dentate gyrus, and within the periglomerular layer of the olfactory bulb. We propose that the cholinergic system plays a survival-promoting role for neuronal progenitors and immature neurons within regions of adult neurogenesis, similar to effects observed previously during brain development. As a working hypothesis, neuronal loss within the CBF system leads not only to cognitive deficits but may also alter on a cellular level the functionality of the dentate gyrus, which in turn may aggravate cognitive deficits.


Subject(s)
Basal Nucleus of Meynert/physiopathology , Brain Injuries/physiopathology , Cell Differentiation/physiology , Cholinergic Fibers/physiology , Nerve Regeneration/physiology , Neuronal Plasticity/physiology , Acetylcholine/metabolism , Animals , Antibodies, Monoclonal , Apoptosis/physiology , Basal Nucleus of Meynert/injuries , Cell Division/physiology , Cholinergic Fibers/ultrastructure , Cognition Disorders/pathology , Cognition Disorders/physiopathology , Dentate Gyrus/cytology , Dentate Gyrus/growth & development , Down-Regulation/physiology , Immunotoxins , Male , N-Glycosyl Hydrolases , Neural Pathways/injuries , Neural Pathways/physiopathology , Olfactory Bulb/cytology , Olfactory Bulb/growth & development , Rats , Rats, Inbred F344 , Ribosome Inactivating Proteins, Type 1 , Saporins , Stem Cells/cytology , Stem Cells/physiology
12.
J Comp Neurol ; 467(1): 1-10, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-14574675

ABSTRACT

During development of the central nervous system, expression of the microtubule binding protein doublecortin (DCX) is associated with migration of neuroblasts. In addition to this developmental role, expression of DCX remains high within certain areas of the adult mammalian brain. These areas, mainly the dentate gyrus and the lateral ventricle wall in conjunction with the rostral migratory stream and olfactory bulb, retain the capacity to generate new neurons into adulthood. Adult neurogenesis is typically detected by incorporation of bromodeoxyuridine (BrdU) into dividing cells and colabeling of BrdU-positive cells with markers for mature neurons. To elucidate whether DCX could act as an alternative indicator for adult neurogenesis, we investigated the temporal expression pattern of DCX in neurogenic regions of the adult brain. Analysis of newly generated cells showed that DCX is transiently expressed in proliferating progenitor cells and newly generated neuroblasts. As the newly generated cells began expressing mature neuronal markers, DCX immunoreactivity decreased sharply below the level of detection and remained undetectable thereafter. The transient expression pattern of DCX in neuronal committed progenitor cells/neuroblasts indicates that DCX could be developed into a suitable marker for adult neurogenesis and may provide an alternative to BrdU labeling. This assumption is further supported by our observation that the number of DCX-expressing cells in the dentate gyrus was decreased with age according to the reduction of neurogenesis in the aging dentate gyrus previously reported.


Subject(s)
Aging/metabolism , Central Nervous System/metabolism , Microtubule-Associated Proteins , Mitosis , Neurons/metabolism , Neuropeptides/metabolism , Animals , Blotting, Western , Bromodeoxyuridine , Cell Differentiation , Cell Movement , Central Nervous System/growth & development , Dentate Gyrus/metabolism , Doublecortin Domain Proteins , Doublecortin Protein , Electrophoresis, Polyacrylamide Gel , Female , Fluorescent Antibody Technique , Hippocampus/metabolism , Lateral Ventricles/metabolism , Olfactory Bulb/metabolism , Rats , Rats, Wistar , Stem Cells/metabolism , Time Factors
13.
Eur J Neurosci ; 17(10): 2042-6, 2003 May.
Article in English | MEDLINE | ID: mdl-12786970

ABSTRACT

Exposure to an enriched environment and physical activity, such as voluntary running, increases neurogenesis of granule cells in the dentate gyrus of adult mice. These stimuli are also known to improve performance in hippocampus-dependent learning tasks, but it is unclear whether their effects on neurogenesis are exclusive to the hippocampal formation. In this study, we housed adult mice under three conditions (enriched environment, voluntary wheel running and standard housing), and analysed proliferation in the lateral ventricle wall and granule cell neurogenesis in the olfactory bulb in comparison to the dentate gyrus. Using bromodeoxyuridine to label dividing cells, we could not detect any difference in the number of newly generated cells in the ventricle wall. When giving the new cells time to migrate and differentiate in the olfactory bulb, we observed no changes in the number of adult-generated olfactory granule cells; however, voluntary running and enrichment produced a doubling in the amount of new hippocampal granule cells. The discrepancy between the olfactory bulb and the dentate gyrus suggests that these living conditions trigger locally through an as yet unidentified mechanism specific to neurogenic signals in the dentate gyrus.


Subject(s)
Environment Design , Hippocampus/physiology , Neurons/cytology , Olfactory Bulb/physiology , Physical Conditioning, Animal/physiology , Age Factors , Animals , Cell Differentiation/physiology , Cell Division/physiology , Female , Hippocampus/cytology , Housing, Animal , Mice , Mice, Inbred C57BL , Neurons/physiology , Olfactory Bulb/cytology
14.
Eur J Neurosci ; 16(9): 1681-9, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12431220

ABSTRACT

In the adult rat olfactory bulb, neurons are continually generated from progenitors that reside in the lateral ventricle wall. This study investigates long-term survival and cell death of newly generated cells within the adult olfactory bulb. After injecting rats at 2 months of age with 5-bromodeoxyuridine (BrdU), the newly generated cells were quantified over a period of 19 months. A peak of BrdU-positive cells was reached in the olfactory bulb 1 month after BrdU injection, when all new cells have finished migrating from the ventricle wall. Thereafter, a reduction of BrdU-positive cells to about 50% was observed and it was confirmed by dUTP-nick end-labelling (TUNEL) that progenitors and young neurons undergo programmed cell death. However, cells that survived the first 3 months after BrdU injection persisted for up to 19 months. The majority of the BrdU-positive cells that reach the olfactory bulb differentiate into granule cells, but a small fraction migrate further into the glomerular layer. These newborn cells differentiate more slowly into periglomerular interneurons, with a delay of more than 1 month when compared to the granule cells. The newly generated periglomerular neurons, among them a significant fraction of dopaminergic cells, showed a similar decline in number compared to the granule cell layer and long-term survival for the remaining new neurons of up to 19 months. Rather than replacing old neurons, this data suggests that adult olfactory bulb neurogenesis utilizes the overproduction and turnover of young neurons, which is reminiscent of the cellular dynamics observed during brain development.


Subject(s)
Cell Differentiation/physiology , Neurons/cytology , Neurons/physiology , Olfactory Bulb/physiology , Animals , Apoptosis , Bromodeoxyuridine , Cell Division , Cell Survival , Female , Immunohistochemistry , In Situ Nick-End Labeling , Rats , Rats, Wistar , Stem Cells , Time Factors
15.
Mol Cell Neurosci ; 21(2): 312-23, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12401450

ABSTRACT

During nervous system development the fate of neural stem cells-whether to undergo proliferation, differentiation, or apoptosis-is controlled by various signals, such as growth factors. Here, we demonstrate that the transcription factor E2F1, which is targeted by several signaling cascades that are activated by growth factors, is involved in neurogenesis in the adult brain. When analyzing the brains of E2F1-deficient mice, we found significantly decreased stem cell and progenitor division in the proliferative zones of the lateral ventricle wall and the hippocampus. As a consequence, the production of newborn neurons in the adult olfactory bulb and dentate gyrus was decreased. Neuronal cell counts of the adult cerebellum revealed a mild but significant cerebellar atrophy, whereas neocortical neurons were unaffected, suggesting that E2F1 deficiency produces a predominantly postnatal phenotype. The results indicate an involvement of E2F1 in controlling proliferation and neuronal cell numbers in the postnatal and adult brain.


Subject(s)
Brain/cytology , Cell Cycle Proteins , DNA-Binding Proteins , Neurons/cytology , Transcription Factors/deficiency , Transcription Factors/genetics , Animals , Animals, Newborn , Brain/growth & development , Brain/metabolism , Cell Death/genetics , Cell Differentiation/genetics , Cell Movement/genetics , E2F Transcription Factors , E2F1 Transcription Factor , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Neurologic Mutants , Neurons/metabolism , Transcription Factors/biosynthesis
16.
Brain Res Dev Brain Res ; 134(1-2): 13-21, 2002 Mar 31.
Article in English | MEDLINE | ID: mdl-11947933

ABSTRACT

Since the early 1960s, in vivo observations have shown the generation of new neurons from dividing precursor cells. Nevertheless, these experiments suffered from skepticism, suggesting that the prevailing labeling method, which incorporates tagged thymidine analogs, such as [3H]-thymidine or bromodeoxyuridine (BrdU), may not be detecting a proliferative event, but could rather mark DNA repair in postmitotic neurons. Even today many scientists outside the field are still skeptical, because the question of specificity for thymidine labeling has not been sufficiently answered. This current paper aims at evaluating the arguments that are used by proponents and skeptics of this method by (i) presenting histological evidence of specificity of BrdU labeling for neural stem cell/progenitor activity in the adult brain; (ii) validating and comparing BrdU labeling with other histological methods; and (iii) combining BrdU and labeling methods for apoptosis to argue against DNA repair being a major contribution of BrdU-positive cells.


Subject(s)
Brain/cytology , DNA Repair , Neurons/cytology , Animals , Apoptosis/physiology , Bromodeoxyuridine , Cell Cycle/physiology , Cell Division , Cellular Senescence/physiology , Female , Fluorescent Antibody Technique , In Situ Nick-End Labeling , Microscopy, Electron , Neurology/methods , Rats , Rats, Wistar , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...