Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
J Neurosci ; 39(50): 9967-9988, 2019 12 11.
Article in English | MEDLINE | ID: mdl-31685650

ABSTRACT

New neurons, referred to as neuroblasts, are continuously generated in the ventricular-subventricular zone of the brain throughout an animal's life. These neuroblasts are characterized by their unique potential for proliferation, formation of chain-like cell aggregates, and long-distance and high-speed migration through the rostral migratory stream (RMS) toward the olfactory bulb (OB), where they decelerate and differentiate into mature interneurons. The dynamic changes of ultrastructural features in postnatal-born neuroblasts during migration are not yet fully understood. Here we report the presence of a primary cilium, and its ultrastructural morphology and spatiotemporal dynamics, in migrating neuroblasts in the postnatal RMS and OB. The primary cilium was observed in migrating neuroblasts in the postnatal RMS and OB in male and female mice and zebrafish, and a male rhesus monkey. Inhibition of intraflagellar transport molecules in migrating neuroblasts impaired their ciliogenesis and rostral migration toward the OB. Serial section transmission electron microscopy revealed that each migrating neuroblast possesses either a pair of centrioles or a basal body with an immature or mature primary cilium. Using immunohistochemistry, live imaging, and serial block-face scanning electron microscopy, we demonstrate that the localization and orientation of the primary cilium are altered depending on the mitotic state, saltatory migration, and deceleration of neuroblasts. Together, our results highlight a close mutual relationship between spatiotemporal regulation of the primary cilium and efficient chain migration of neuroblasts in the postnatal brain.SIGNIFICANCE STATEMENT Immature neurons (neuroblasts) generated in the postnatal brain have a mitotic potential and migrate in chain-like cell aggregates toward the olfactory bulb. Here we report that migrating neuroblasts possess a tiny cellular protrusion called a primary cilium. Immunohistochemical studies with zebrafish, mouse, and monkey brains suggest that the presence of the primary cilium in migrating neuroblasts is evolutionarily conserved. Ciliogenesis in migrating neuroblasts in the rostral migratory stream is suppressed during mitosis and promoted after cell cycle exit. Moreover, live imaging and 3D electron microscopy revealed that ciliary localization and orientation change during saltatory movement of neuroblasts. Our results reveal highly organized dynamics in maturation and positioning of the primary cilium during neuroblast migration that underlie saltatory movement of postnatal-born neuroblasts.


Subject(s)
Cell Movement/physiology , Cilia/ultrastructure , Lateral Ventricles/ultrastructure , Neural Stem Cells/ultrastructure , Neurons/ultrastructure , Olfactory Bulb/ultrastructure , Animals , Female , Macaca mulatta , Male , Mice , Zebrafish
2.
Transl Stroke Res ; 8(4): 362-373, 2017 08.
Article in English | MEDLINE | ID: mdl-28102508

ABSTRACT

We previously found that hematoma worsens hydrocephalus after intraventricular hemorrhage (IVH) via increasing iron deposition and aggravating ependymal cilia injury; therefore, promoting hematoma absorption may be a promising strategy for IVH. Recently, some investigations imply that simvastatin has the ability of accelerating hematoma absorption. Thus, this study was designed to examine the efficacy of simvastatin for IVH in rats. Intracerebral hemorrhage with ventricular extension was induced in adult male Sprague-Dawley rats after autologous blood injection. Simvastatin or vehicle was administered orally at 1 day after IVH and then daily for 1 week. MRI studies were performed to measure the volumes of intracranial hematoma and lateral ventricle at days 1, 3, 7, 14, and 28 after IVH. Motor and neurocognitive functions were assessed at days 1 to 7 and 23 to 28, respectively. Iron deposition, iron-related protein expression, ependymal damage, and histology were detected at day 28. Expression of CD36 scavenger receptor (facilitating phagocytosis) was examined at day 3 after IVH using western blotting and immunofluorescence. Simvastatin significantly increased hematoma absorption ratio, reduced ventricular volume, and attenuated neurological dysfunction post-IVH. In addition, less iron accumulation and more cilia survival was observed in the simvastatin group when compared with the control. What's more, higher expression of CD36 was detected around the hematoma after simvastatin administration. Simvastatin significantly enhanced brain hematoma absorption, alleviated hydrocephalus, and improved neurological recovery after experimental IVH, which may in part by upregulating CD36 expression. Our data suggest that early simvastatin use may be a novel therapy for IVH patients.


Subject(s)
CD36 Antigens/metabolism , Hematoma/drug therapy , Hydrocephalus/drug therapy , Hypolipidemic Agents/therapeutic use , Simvastatin/therapeutic use , Up-Regulation/drug effects , Animals , Brain/pathology , Brain/ultrastructure , CD11b Antigen/metabolism , Cell Count , Cerebral Hemorrhage/complications , Cerebral Hemorrhage/diagnostic imaging , Disease Models, Animal , Ependyma/metabolism , Ependyma/pathology , Ependyma/ultrastructure , Ferritins/metabolism , Ferritins/ultrastructure , Follow-Up Studies , Hematoma/diagnostic imaging , Hematoma/etiology , Hydrocephalus/diagnostic imaging , Hydrocephalus/etiology , Lateral Ventricles/diagnostic imaging , Lateral Ventricles/pathology , Lateral Ventricles/ultrastructure , Magnetic Resonance Imaging , Male , Maze Learning/drug effects , Microscopy, Electron, Transmission , Neurologic Examination , Neurons/metabolism , Neurons/pathology , Neurons/ultrastructure , Rats , Rats, Sprague-Dawley
3.
Redox Biol ; 11: 231-239, 2017 04.
Article in English | MEDLINE | ID: mdl-28012438

ABSTRACT

Fulfilling a bevy of biological roles, copper is an essential metal for healthy brain function. Cu dyshomeostasis has been demonstrated to be involved in some neurological conditions including Menkes and Alzheimer's diseases. We have previously reported localized Cu-rich aggregates in astrocytes of the subventricular zone (SVZ) in rodent brains with Cu concentrations in the hundreds of millimolar. Metallothionein, a cysteine-rich protein critical to metal homeostasis and known to participate in a variety of neuroprotective and neuroregenerative processes, was proposed as a binding protein. Here, we present an analysis of metallothionein(1,2) knockout (MTKO) mice and age-matched controls using X-ray fluorescence microscopy. In large structures such as the corpus callosum, cortex, and striatum, there is no significant difference in Cu, Fe, or Zn concentrations in MTKO mice compared to age-matched controls. In the astrocyte-rich subventricular zone where Cu-rich aggregates reside, approximately 1/3 as many Cu-rich aggregates persist in MTKO mice resulting in a decrease in periventricular Cu concentration. Aggregates in both wild-type and MTKO mice show XANES spectra characteristic of CuxSy multimetallic clusters and have similar [S]/[Cu] ratios. Consistent with assignment as a CuxSy multimetallic cluster, the astrocyte-rich SVZ of both MTKO and wild-type mice exhibit autofluorescent bodies, though MTKO mice exhibit fewer. Furthermore, XRF imaging of Au-labeled lysosomes and ubiquitin demonstrates a lack of co-localization with Cu-rich aggregates suggesting they are not involved in a degradation pathway. Overall, these data suggest that Cu in aggregates is bound by either metallothionein-3 or a yet unknown protein similar to metallothionein.


Subject(s)
Astrocytes/metabolism , Copper/chemistry , Lateral Ventricles/metabolism , Metallothionein/deficiency , Animals , Astrocytes/ultrastructure , Biomarkers/metabolism , Cations, Divalent , Cerebral Cortex/metabolism , Cerebral Cortex/ultrastructure , Chemical Precipitation , Corpus Callosum/metabolism , Corpus Callosum/ultrastructure , Corpus Striatum/metabolism , Corpus Striatum/ultrastructure , Gene Deletion , Gene Expression , Iron/chemistry , Lateral Ventricles/ultrastructure , Lysosomal Membrane Proteins/genetics , Lysosomal Membrane Proteins/metabolism , Lysosomes/metabolism , Lysosomes/ultrastructure , Metallothionein/genetics , Mice , Mice, Knockout , Optical Imaging , Rats , Rats, Sprague-Dawley , Spectrometry, X-Ray Emission , Ubiquitin/genetics , Ubiquitin/metabolism , Zinc/chemistry
4.
Adv Exp Med Biol ; 913: 397-402, 2016.
Article in English | MEDLINE | ID: mdl-27796901

ABSTRACT

It is well proved already that neurogenesis does take place in mammals' brain, including human brain. However, neurogenesis by itself is not able to compensate for brain tissue loss in serious neurological diseases, such as stroke, brain trauma or neurodegenerative disorders. Recent evidences show that neural stem cell niches are present not only in classical locations, such as subventricularor subgranular zones, but in other areas as well, including tissues contiguous to the brain (meninges and choroid plexus).In this chapter we revise the relationship of neural stem cells with interstitial cells (mainly telocytes), which we think is significant, and we describe what is known about the juxtacerebral tissue neurogenesis potential.


Subject(s)
Choroid Plexus/physiology , Meninges/physiology , Nerve Regeneration/physiology , Neural Stem Cells/physiology , Stem Cell Niche/physiology , Telocytes/physiology , Animals , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/rehabilitation , Choroid Plexus/ultrastructure , Hippocampus/physiology , Hippocampus/ultrastructure , Humans , Lateral Ventricles/physiology , Lateral Ventricles/ultrastructure , Meninges/ultrastructure , Microscopy, Electron, Transmission , Neural Stem Cells/ultrastructure , Neurogenesis/physiology , Rats , Stroke/pathology , Stroke Rehabilitation , Telocytes/ultrastructure
5.
Stem Cells ; 34(10): 2574-2586, 2016 10.
Article in English | MEDLINE | ID: mdl-27299504

ABSTRACT

In the mouse brain, neuroblasts generated in the subventricular zone (SVZ) migrate to the olfactory bulb (OB) through the rostral migratory stream (RMS). Although the RMS is not present in the human brain, a migratory pathway that is organized around a ventricular cavity that reaches the OB has been reported. A similar cavity, the lateral ventricle extension (LVE), is found in the adult guinea pig brain. Therefore, we analyzed cytoarchitecture, proliferative activity and precursor cell migration in the SVZ and LVE of 1-, 6- and 12-month-old guinea pigs. In young animals, we used confocal spectral and transmission electron microscopy to identify neuroblasts, astrocytes, and progenitor cells in the SVZ and LVE. Analysis of peroxidase diffusion demonstrated that the LVE was a continuous cavity lined by ependymal cells and surrounded by neuroblasts. Precursor cells were mostly located in the SVZ and migrated from the SVZ to the OB through the LVE. Finally, analysis of 6- and 12-month-old guinea pigs revealed that the LVE was preserved in older animals; however, the number of neurogenic cells was significantly reduced. Consequently, we propose that the guinea pig brain may be used as a new neurogenic model with increased similarity to humans, given that the LVE connects the LV with the OB, as has been described in humans, and that the LVE works a migratory pathway. Stem Cells 2016;34:2574-2586.


Subject(s)
Aging/physiology , Cell Movement , Lateral Ventricles/cytology , Neurons/cytology , Animals , Cell Proliferation , Cell Shape , Guinea Pigs , Lateral Ventricles/ultrastructure , Male
6.
Cell Death Dis ; 7: e2223, 2016 05 12.
Article in English | MEDLINE | ID: mdl-27171265

ABSTRACT

Current treatments for demyelinating diseases are generally only capable of ameliorating the symptoms, with little to no effect in decreasing myelin loss nor promoting functional recovery. Mesenchymal stem cells (MSCs) have been shown by many researchers to be a potential therapeutic tool in treating various neurodegenerative diseases, including demyelinating disorders. However, in the majority of the cases, the effect was only observed locally, in the area surrounding the graft. Thus, in order to achieve general remyelination in various brain structures simultaneously, bone marrow-derived MSCs were transplanted into the lateral ventricles (LVs) of the cuprizone murine model. In this manner, the cells may secrete soluble factors into the cerebrospinal fluid (CSF) and boost the endogenous oligodendrogenic potential of the subventricular zone (SVZ). As a result, oligodendrocyte progenitor cells (OPCs) were recruited within the corpus callosum (CC) over time, correlating with an increased myelin content. Electrophysiological studies, together with electron microscopy (EM) analysis, indicated that the newly formed myelin correctly enveloped the demyelinated axons and increased signal transduction through the CC. Moreover, increased neural stem progenitor cell (NSPC) proliferation was observed in the SVZ, possibly due to the tropic factors released by the MSCs. In conclusion, the findings of this study revealed that intraventricular injections of MSCs is a feasible method to elicit a paracrine effect in the oligodendrogenic niche of the SVZ, which is prone to respond to the factors secreted into the CSF and therefore promoting oligodendrogenesis and functional remyelination.


Subject(s)
Demyelinating Diseases/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Neural Stem Cells/cytology , Oligodendroglia/cytology , Paracrine Communication/physiology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Cell Differentiation , Cell- and Tissue-Based Therapy/methods , Corpus Callosum/metabolism , Corpus Callosum/ultrastructure , Cuprizone , Demyelinating Diseases/chemically induced , Demyelinating Diseases/genetics , Demyelinating Diseases/pathology , Disease Models, Animal , Gene Expression , Humans , Injections, Intraventricular , Intercellular Signaling Peptides and Proteins/cerebrospinal fluid , Intercellular Signaling Peptides and Proteins/genetics , Lateral Ventricles/metabolism , Lateral Ventricles/ultrastructure , Mesenchymal Stem Cells/physiology , Mice , Myelin Sheath/metabolism , Myelin Sheath/ultrastructure , Neural Stem Cells/physiology , Oligodendroglia/physiology , Signal Transduction
7.
Brain Struct Funct ; 221(1): 239-60, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25300351

ABSTRACT

New subventricular zone (SVZ)-derived neuroblasts that migrate via the rostral migratory stream are continuously added to the olfactory bulb (OB) of the adult rodent brain. Anosmin-1 (A1) is an extracellular matrix protein that binds to FGF receptor 1 (FGFR1) to exert its biological effects. When mutated as in Kallmann syndrome patients, A1 is associated with severe OB morphogenesis defects leading to anosmia and hypogonadotropic hypogonadism. Here, we show that A1 over-expression in adult mice strongly increases proliferation in the SVZ, mainly with symmetrical divisions, and produces substantial morphological changes in the normal SVZ architecture, where we also report the presence of FGFR1 in almost all SVZ cells. Interestingly, for the first time we show FGFR1 expression in the basal body of primary cilia in neural progenitor cells. Additionally, we have found that A1 over-expression also enhances neuroblast motility, mainly through FGFR1 activity. Together, these changes lead to a selective increase in several GABAergic interneuron populations in different OB layers. These specific alterations in the OB would be sufficient to disrupt the normal processing of sensory information and consequently alter olfactory memory. In summary, this work shows that FGFR1-mediated A1 activity plays a crucial role in the continuous remodelling of the adult OB.


Subject(s)
Extracellular Matrix Proteins/metabolism , Extracellular Matrix Proteins/physiology , Lateral Ventricles/physiology , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/physiology , Neurogenesis , Olfactory Bulb/physiology , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Animals , Cell Division , Cell Movement , Cells, Cultured , Extracellular Matrix Proteins/genetics , Humans , Interneurons/metabolism , Interneurons/physiology , Lateral Ventricles/metabolism , Lateral Ventricles/ultrastructure , Memory, Short-Term/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neural Pathways/metabolism , Neural Pathways/physiology , Neural Pathways/ultrastructure , Odorants , Olfactory Bulb/metabolism , Olfactory Perception/physiology
8.
Neurotoxicology ; 50: 46-55, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26215120

ABSTRACT

It is still being debated whether neurogenesis in the subventricular zone (SVZ) is enhanced in response to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) injury in the adult mouse brain. Our previous studies provided evidence that MPTP induces apoptosis of migrating neuroblasts (neural progenitor cells, A cells) in the SVZ and rostral migratory stream (RMS). We investigated cellular kinetics in the adult SVZ and olfactory bulb (OB) after MPTP damage. Cells were labeled with bromodeoxyuridine (BrdU), and the effects of MPTP on the survival and fate of migrating and residing neuroblasts were evaluated. Two days after BrdU labeling and MPTP treatment, the number of BrdU-positive cells in the SVZ and OB of MPTP-treated mice was significantly lower than in the SVZ and OB of saline controls. Additionally, fewer BrdU-positive cells migrated to the OB of treated mice than to that of saline controls, and the cells that did migrate diffused radially into the granule cell layer (GCL) when observed at 7, 14, and 28 days. In the OB GCL, the differentiation of BrdU-positive cells into mature neurons significantly attenuated 14 and 28 days after MPTP injury. Moreover, the impaired neurogenesis was followed by a recovery of A cells in the SVZ and OB, suggesting activation of the self-repair process as a result of MPTP-induced depletion of BrdU-positive cells. Our findings clarify the kinetics underlying neurogenesis in MPTP-treated mice and may contribute to the development of an animal model of Parkinson's disease, and the demonstration of cellular kinetics in SVZ may also provide a new insight into assessing neurogenesis in MPTP-treated mouse.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Lateral Ventricles/drug effects , MPTP Poisoning/chemically induced , MPTP Poisoning/pathology , Neurogenesis/drug effects , Olfactory Bulb/drug effects , Animals , Apoptosis/drug effects , Bromodeoxyuridine/metabolism , Calcium-Binding Proteins/metabolism , Cell Differentiation/drug effects , Cell Movement/drug effects , Disease Models, Animal , ErbB Receptors/metabolism , Lateral Ventricles/physiology , Lateral Ventricles/ultrastructure , Male , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Microscopy, Electron, Transmission , Nerve Tissue Proteins/metabolism , Olfactory Bulb/physiology , Olfactory Bulb/ultrastructure , Proliferating Cell Nuclear Antigen/metabolism , Time Factors
9.
J Vis Exp ; (100): e52853, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-26067390

ABSTRACT

Multiciliated ependymal cells line the ventricles in the adult brain. Abnormal function or structure of ependymal cilia is associated with various neurological deficits. The current ex vivo live imaging of motile ependymal cilia technique allows for a detailed study of ciliary dynamics following several steps. These steps include: mice euthanasia with carbon dioxide according to protocols of The University of Toledo's Institutional Animal Care and Use Committee (IACUC); craniectomy followed by brain removal and sagittal brain dissection with a vibratome or sharp blade to obtain very thin sections through the brain lateral ventricles, where the ependymal cilia can be visualized. Incubation of the brain's slices in a customized glass-bottom plate containing Dulbecco's Modified Eagle's Medium (DMEM)/High-Glucose at 37 °C in the presence of 95%/5% O2/CO2 mixture is essential to keep the tissue alive during the experiment. A video of the cilia beating is then recorded using a high-resolution differential interference contrast microscope. The video is then analyzed frame by frame to calculate the ciliary beating frequency. This allows distinct classification of the ependymal cells into three categories or types based on their ciliary beating frequency and angle. Furthermore, this technique allows the use of high-speed fluorescence imaging analysis to characterize the unique intracellular calcium oscillation properties of ependymal cells as well as the effect of pharmacological agents on the calcium oscillations and the ciliary beating frequency. In addition, this technique is suitable for immunofluorescence imaging for ciliary structure and ciliary protein localization studies. This is particularly important in disease diagnosis and phenotype studies. The main limitation of the technique is attributed to the decrease in live motile cilia movement as the brain tissue starts to die.


Subject(s)
Cilia/physiology , Ependyma/physiology , Ependyma/ultrastructure , Lateral Ventricles/physiology , Lateral Ventricles/ultrastructure , Microscopy, Interference/methods , Animals , Mice , Mice, Inbred C57BL
10.
J Appl Toxicol ; 35(7): 737-51, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25256750

ABSTRACT

Ochratoxin A (OTA), a mycotoxin that was discovered as a secondary metabolite of the fungal species Aspergillus and Penicillium, is a common contaminant in food and animal feed. This mycotoxin has been described as teratogenic, carcinogenic, genotoxic, immunotoxic and has been proven a potent neurotoxin. Other authors have previously reported the effects of OTA in different structures of the central nervous system as well as in some neurogenic regions. However, the impact of OTA exposure in the subventricular zone (SVZ) has not been assessed yet. To elucidate whether OTA affects neural precursors of the mouse SVZ we investigated, in vitro and in vivo, the effects of OTA exposure on the SVZ and on the neural precursors obtained from this neurogenic niche. In this work, we prove the cumulative effect of OTA exposure on proliferation, differentiation and depletion of neural stem cells cultured from the SVZ. In addition, we corroborated these results in vivo by immunohistochemistry and electron microscopy. As a result, we found a significant alteration in the proliferation process, which was evidenced by a decrease in the number of 5-bromo-2-deoxyuridine-positive cells and glial cells, as well as, a significant decrease in the number of neuroblasts in the SVZ. To summarize, in this study we demonstrate how OTA could be a threat to the developing and the adult SVZ through its impact in cell viability, proliferation and differentiation in a dose-dependent manner.


Subject(s)
Lateral Ventricles/drug effects , Mycotoxins/toxicity , Ochratoxins/toxicity , Animals , Astrocytes/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Lateral Ventricles/pathology , Lateral Ventricles/ultrastructure , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron , Neural Stem Cells/drug effects , Neuroglia/drug effects
11.
Exp Neurol ; 261: 236-44, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24858805

ABSTRACT

Hydrocephalus can develop secondarily to a disturbance in production, flow and/or absorption of cerebrospinal fluid. Experimental models of hydrocephalus, especially subacute and chronic hydrocephalus, are few and limited, and the effects of hydrocephalus on the subventricular zone are unclear. The aim of this study was to analyze the effects of long-term obstructive hydrocephalus on the subventricular zone, which is the neurogenic niche lining the lateral ventricles. We developed a new method to induce hydrocephalus by obstructing the aqueduct of Sylvius in the mouse brain, thus simulating aqueductal stenosis in humans. In 120-day-old rodents (n=18 per group), the degree of ventricular dilatation and cellular composition of the subventricular zone were studied by immunofluorescence and transmission electron microscopy. In adult patients (age>18years), the sizes of the subventricular zone, corpus callosum, and internal capsule were analyzed by magnetic resonance images obtained from patients with and without aqueductal stenosis (n=25 per group). Mice with 60-day hydrocephalus had a reduced number of Ki67+ and doublecortin+cells on immunofluorescence, as well as decreased number of neural progenitors and neuroblasts in the subventricular zone on electron microscopy analysis as compared to non-hydrocephalic mice. Remarkably, a number of extracellular matrix structures (fractones) contacting the ventricular lumen and blood vessels were also observed around the subventricular zone in mice with hydrocephalus. In humans, the widths of the subventricular zone, corpus callosum, and internal capsule in patients with aqueductal stenosis were significantly smaller than age and gender-matched patients without aqueductal stenosis. In summary, supratentorial hydrocephalus reduces the proliferation rate of neural progenitors and modifies the cytoarchitecture and extracellular matrix compounds of the subventricular zone. In humans, this similar process reduces the subventricular niche as well as the width of corpus callosum and internal capsule.


Subject(s)
Hydrocephalus/pathology , Lateral Ventricles/metabolism , Lateral Ventricles/pathology , Adult , Animals , Cohort Studies , Corpus Callosum/metabolism , Corpus Callosum/pathology , Disease Models, Animal , Doublecortin Domain Proteins , Female , Gene Expression Regulation , Glial Fibrillary Acidic Protein/metabolism , Humans , Hydrocephalus/physiopathology , Internal Capsule/metabolism , Internal Capsule/pathology , Ki-67 Antigen/metabolism , Lateral Ventricles/ultrastructure , Magnetic Resonance Imaging , Male , Maze Learning/physiology , Mice , Mice, Inbred BALB C , Microtubule-Associated Proteins/metabolism , Neuropeptides/metabolism , Time Factors , Young Adult
12.
Glia ; 62(5): 790-803, 2014 May.
Article in English | MEDLINE | ID: mdl-24677590

ABSTRACT

Neurogenesis persists in the adult subventricular zone (SVZ) of the mammalian brain. During aging, the SVZ neurogenic capacity undergoes a progressive decline, which is attributed to a decrease in the population of neural stem cells (NSCs). However, the behavior of the NSCs that remain in the aged brain is not fully understood. Here we performed a comparative ultrastructural study of the SVZ niche of 2-month-old and 24-month-old male C57BL/6 mice, focusing on the NSC population. Using thymidine-labeling, we showed that residual NSCs in the aged SVZ divide less frequently than those in young mice. We also provided evidence that ependymal cells are not newly generated during senescence, as others studies suggest. Remarkably, both astrocytes and ependymal cells accumulated a high number of intermediate filaments and dense bodies during aging, resembling reactive cells. A better understanding of the changes occurring in the neurogenic niche during aging will allow us to develop new strategies for fighting neurological disorders linked to senescence.


Subject(s)
Aging/physiology , Astrocytes/physiology , Ependyma/cytology , Ependyma/physiology , Lateral Ventricles/cytology , Lateral Ventricles/physiology , Animals , Astrocytes/ultrastructure , Cell Differentiation/physiology , Cell Proliferation , Ependyma/ultrastructure , Lateral Ventricles/ultrastructure , Male , Mice , Mice, Inbred C57BL , Neural Stem Cells/physiology , Neural Stem Cells/ultrastructure , Neurogenesis/physiology
13.
J Neurosci ; 32(34): 11511-23, 2012 Aug 22.
Article in English | MEDLINE | ID: mdl-22915098

ABSTRACT

Hydrocephalus formation is a frequent complication of neuropathological insults associated with neuroinflammation. However, the mechanistic role of neuroinflammation in hydrocephalus development is unclear. We have investigated the function of the proinflammatory acting inhibitor of κB kinase (IKK)/nuclear factor κB (NF-κB) signaling system in neuroinflammatory processes and generated a novel mouse model that allows conditional activation of the IKK/NF-κB system in astrocytes. Remarkably, NF-κB activation in astrocytes during early postnatal life results in hydrocephalus formation and additional defects in brain development. NF-κB activation causes global neuroinflammation characterized by a strong, astrocyte-specific expression of proinflammatory NF-κB target genes as well as a massive infiltration and activation of macrophages. In this animal model, hydrocephalus formation is specifically induced during a critical time period of early postnatal development, in which IKK/NF-κB-induced neuroinflammation interferes with ependymal ciliogenesis. Our findings demonstrate for the first time that IKK/NF-κB activation is sufficient to induce hydrocephalus formation and provides a potential mechanistic explanation for the frequent association of neuroinflammation and hydrocephalus formation during brain development, namely impairment of ependymal cilia formation. Therefore, our study might open up new perspectives for the treatment of certain types of neonatal and childhood hydrocephalus associated with hemorrhages and infections.


Subject(s)
Encephalitis/etiology , Gene Expression Regulation, Developmental/physiology , Glioma, Subependymal/etiology , Hydrocephalus , I-kappa B Kinase/metabolism , Age Factors , Animals , Animals, Newborn , Astrocytes/drug effects , Astrocytes/enzymology , Brain/enzymology , Brain/growth & development , Brain/pathology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Chemokines/genetics , Chemokines/metabolism , Complement System Proteins/genetics , Complement System Proteins/metabolism , Disease Models, Animal , Doxycycline/administration & dosage , Enzyme Activation/drug effects , Enzyme Activation/genetics , Gene Expression Regulation, Developmental/drug effects , Glial Fibrillary Acidic Protein/genetics , Glioma, Subependymal/pathology , Humans , Hydrocephalus/complications , Hydrocephalus/enzymology , Hydrocephalus/pathology , I-kappa B Kinase/genetics , I-kappa B Proteins/metabolism , Lateral Ventricles/growth & development , Lateral Ventricles/pathology , Lateral Ventricles/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microarray Analysis , Microscopy, Electron, Scanning , NF-KappaB Inhibitor alpha , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Statistics, Nonparametric , Transcription Factor RelA/metabolism , NF-kappaB-Inducing Kinase
14.
Dev Neurosci ; 34(4): 299-309, 2012.
Article in English | MEDLINE | ID: mdl-22831917

ABSTRACT

Much work has focused on the possible contribution of adult hippocampal neurogenesis to neuropsychiatric diseases. The hippocampal subgranular zone and the other stem cell-containing neurogenic niche, the subventricular zone (SVZ), share several cytological features and are regulated by some of the same molecular mechanisms. However, very little is known about the SVZ in neuropsychiatric disorders. This is important since it surrounds the lateral ventricles and in schizophrenia ventricular enlargement frequently follows forebrain nuclei shrinkage. Also, adult neurogenesis has been implicated in pharmacotherapy for affective disorders and many of the molecules associated with neuropsychiatric disorders affect SVZ biology. To assess the neurogenic niche, we examined material from 60 humans (Stanley Collection) and characterized the cytoarchitecture of the SVZ and ependymal layer in age-, sex- and post mortem interval-matched controls, and patients diagnosed with schizophrenia, bipolar illness, and depression (n = 15 each). There is a paucity of post mortem brains available for study in these diseases, so to maximize the number of possible parameters examined here, we quantified individual sections rather than a large series. Previous work showed that multiple sclerosis is associated with increased width of the hypocellular gap, a cell-sparse region that typifies the human SVZ. Statistically there were no differences between disease groups and controls in the width of the hypocellular gap or in the density of cells in the hypocellular gap. Because ventricular enlargement in schizophrenia may disrupt ependymal cells, we quantified them, but observed no difference between diagnostic groups and controls. There are significant differences in the prevalence of neuropsychiatric illness between the sexes. Therefore, we looked for male versus female differences, but did not observe any in the parameters quantified. We next turned to a finer spatial resolution and asked if there were differences amongst the disease groups in dorsal ventral subdivisions of the SVZ. Similar to when we treated the SVZ as a whole, we did not find such differences. However, compared to the dorsal SVZ, the ventral SVZ had a wider hypocellular gap and more ependymal cells in all four groups. In contrast, cell density was similar in dorsal ventral subregions of the SVZ hypocellular gap. These results show that though there are regional differences in the SVZ in humans, neuropsychiatric disorders do not seem to alter several fundamental histological features of this adult neurogenic zone.


Subject(s)
Bipolar Disorder/pathology , Caudate Nucleus/ultrastructure , Depressive Disorder/pathology , Ependyma/ultrastructure , Hippocampus/ultrastructure , Lateral Ventricles/ultrastructure , Putamen/ultrastructure , Schizophrenia/pathology , Adult , Astrocytes/ultrastructure , Female , Humans , Male , Middle Aged , Neurogenesis , Neurons/ultrastructure , Organ Specificity , Stem Cells/ultrastructure , Young Adult
15.
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
16.
J Neurosci Methods ; 197(1): 65-81, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21310178

ABSTRACT

Mammalian neurogenic sites are good models for physiological neural cell renewal in the perspective of brain repair. Yet, investigating their stem cell niches is not easy since they are small areas deeply hidden in the brain hemispheres. Organotypic slices could be a useful tool since they substantially retain the three-dimensional tissue organization. The postnatal forebrain subventricular zone (SVZ), as a dynamic structure endowed with proliferation and migration, might undergo striking cellular changes in culture. Literature concerning this ex vivo approach applied to SVZ neurogenic activity and response to damage is scarce and heterogeneous, not considering the fine cellular composition of the slice and not taking into account the modifications occurring as a consequence of the culture conditions. Our aim was to describe in detail what happens in the SVZ when establishing an ex vivo model. We addressed the changes occurring in five day-old, postnatal mice forebrain organotypic slices cultured for several days in vitro, by using confocal and ultrastructural analyses. We found that during the first two days in vitro the slices undergo progressive structural disaggregation accompanied by remarkable increase in cell proliferation and death with respect to basal levels. In addition, these facts occur in parallel with strong activation of astrocytic cells and microglia. Our results highlight technical limits in the use of forebrain organotypic slices for studying the activity of SVZ neurogenic niche, indicating that they can be reliable for a very short time (1-2 days) and could be misleading when addressing lesion-induced responses.


Subject(s)
Lateral Ventricles/growth & development , Neural Stem Cells/physiology , Prosencephalon/growth & development , Animals , Animals, Newborn , Astrocytes/physiology , Astrocytes/ultrastructure , Cell Death/physiology , Cell Survival/physiology , Lateral Ventricles/ultrastructure , Mice , Mice, Inbred ICR , Microglia/physiology , Microglia/ultrastructure , Microscopy, Confocal/methods , Microscopy, Electron, Transmission/methods , Neural Stem Cells/ultrastructure , Organ Culture Techniques/methods , Prosencephalon/ultrastructure
17.
Nat Neurosci ; 13(6): 700-7, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20473291

ABSTRACT

Ependymal cells form the epithelial lining of cerebral ventricles. Their apical surface is covered by cilia that beat in a coordinated fashion to facilitate circulation of the cerebrospinal fluid (CSF). The genetic factors that govern the development and function of ependymal cilia remain poorly understood. We found that the planar cell polarity cadherins Celsr2 and Celsr3 control these processes. In Celsr2-deficient mice, the development and planar organization of ependymal cilia are compromised, leading to defective CSF dynamics and hydrocephalus. In Celsr2 and Celsr3 double mutant ependyma, ciliogenesis is markedly impaired, resulting in lethal hydrocephalus. The membrane distribution of Vangl2 and Fzd3, two key planar cell polarity proteins, was disturbed in Celsr2 mutants, and even more so in Celsr2 and Celsr3 double mutants. Our findings suggest that planar cell polarity signaling is involved in ependymal cilia development and in the pathophysiology of hydrocephalus, with possible implications in other ciliopathies.


Subject(s)
Cadherins/metabolism , Cilia/physiology , Ependyma/physiology , Hydrocephalus/metabolism , Receptors, Cell Surface/metabolism , Animals , Blotting, Western , Cadherins/deficiency , Cadherins/genetics , Cell Polarity/physiology , Choroid Plexus/growth & development , Choroid Plexus/physiology , Cilia/ultrastructure , Ependyma/growth & development , Ependyma/ultrastructure , Hydrocephalus/genetics , Immunohistochemistry , In Vitro Techniques , Lateral Ventricles/growth & development , Lateral Ventricles/physiology , Lateral Ventricles/ultrastructure , Mice , Mice, Transgenic , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Motion , Mutation , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Reverse Transcriptase Polymerase Chain Reaction , Video Recording
18.
Neuroscience ; 168(1): 314-22, 2010 Jun 16.
Article in English | MEDLINE | ID: mdl-20226234

ABSTRACT

Shc(s) family of adaptor molecules has been implicated in several physiological functions. In particular, our previous studies have shown major roles in the mechanisms that control the transition from proliferating neural stem cells (NSCs) to postmitotic neurons in the mammalian brain. In the adult brain, ShcA expression is mainly restricted to a subpopulation of cells in the subventricular zone (SVZ) neurogenic area, enlightening a potential role for this molecule in the establishment/maintenance of this adult NSC niche. In order to investigate this matter, here we took advantage of Cre/lox technology with the purpose of interfering with (or delete) ShcA function in nestin-expressing neural progenitors in vivo. Our analyses revealed signs of anatomical disorganization in the adult brain at the boundary between the striatum and the corpus callosum and reduced thickness both at the ventricular level and through the rostral migratory stream. Analysis of cell proliferation and cell death unveiled a prominent reduction of the former and no substantial alterations of the latter. Ultrastructural studies showed SVZ anatomical disarray and manifest variation in the SVZ cell type composition. In conclusion, these results provide evidence for a role of ShcA in the assembly and/or maintenance of the SVZ NSC niche in the adult brain.


Subject(s)
Lateral Ventricles/anatomy & histology , Shc Signaling Adaptor Proteins/physiology , Stem Cells/cytology , Animals , Axons/ultrastructure , Cell Proliferation , Corpus Callosum/cytology , Corpus Callosum/ultrastructure , Intermediate Filament Proteins/genetics , Lateral Ventricles/cytology , Lateral Ventricles/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Nestin , Neurogenesis , Organ Size , Prosencephalon/cytology , Prosencephalon/ultrastructure , Shc Signaling Adaptor Proteins/genetics , Src Homology 2 Domain-Containing, Transforming Protein 1 , Stem Cells/metabolism
19.
J Comp Neurol ; 514(5): 533-54, 2009 Jun 10.
Article in English | MEDLINE | ID: mdl-19350672

ABSTRACT

Recent publications have shown that the lateral wall of the lateral ventricles in the Macaca fascicularis brain, in particular the subventricular zone (SVZ), contains neural stem cells throughout adulthood that migrate through a migratory pathway (RMS) to the olfactory bulb (OB). To date, a detailed and systematic cytoarchitectural and ultrastructural study of the monkey SVZ and RMS has not been done. We found that the organization of the SVZ was similar to that of humans, with the ependymal layer surrounding the lateral ventricles, a hypocellular GAP layer formed by astrocytic and ependymal expansions, and the astrocyte ribbon, composed of astrocytic bodies. We found no cells corresponding to the type C proliferating precursor of the rodent brain. Instead, proliferating cells, expressed as Ki-67 immunoreactivity, were predominantly young neurons concentrated in the anterior regions, and occasional astrocytes of the ribbon. We observed displaced ependymal cells of still unknown significance. New neurons tended to organize in chain-like structures, which were surrounded by astrocytes. This pattern was highly reminiscent of that observed in rodent RMS, but not in humans. These chains spread from the frontal SVZ along a GAP-like layer, uniquely composed of astrocytic expansions, to the olfactory bulb (OB). The number of neuronal chains and the number of chain-forming cells decreased gradually upon reaching the OB. The purpose of this work is to provide a reference for future studies in the field of adult neurogenesis that may lead to an understanding of the fate and functionality of newborn neurons in primates, and ultimately in humans.


Subject(s)
Lateral Ventricles/physiology , Lateral Ventricles/ultrastructure , Macaca fascicularis/anatomy & histology , Animals , Astrocytes/physiology , Astrocytes/ultrastructure , Cell Movement , Ependyma/cytology , Ependyma/ultrastructure , GTPase-Activating Proteins/metabolism , Immunohistochemistry , Ki-67 Antigen/metabolism , Lateral Ventricles/anatomy & histology , Male , Microscopy, Electron , Neurons/physiology , Neurons/ultrastructure , Olfactory Bulb/anatomy & histology , Olfactory Bulb/physiology
20.
J Neurosci ; 28(48): 12887-900, 2008 Nov 26.
Article in English | MEDLINE | ID: mdl-19036983

ABSTRACT

Primary cilia are important sites of signal transduction involved in a wide range of developmental and postnatal functions. Proteolytic processing of the transcription factor Gli3, for example, occurs in primary cilia, and defects in intraflagellar transport (IFT), which is crucial for the maintenance of primary cilia, can lead to severe developmental defects and diseases. Here we report an essential role of primary cilia in forebrain development. Uncovered by N-ethyl-N-nitrosourea-mutagenesis, cobblestone is a hypomorphic allele of the IFT gene Ift88, in which Ift88 mRNA and protein levels are reduced by 70-80%. cobblestone mutants are distinguished by subpial heterotopias in the forebrain. Mutants show both severe defects in the formation of dorsomedial telencephalic structures, such as the choroid plexus, cortical hem and hippocampus, and also a relaxation of both dorsal-ventral and rostral-caudal compartmental boundaries. These defects phenocopy many of the abnormalities seen in the Gli3 mutant forebrain, and we show that Gli3 proteolytic processing is reduced, leading to an accumulation of the full-length activator isoform. In addition, we observe an upregulation of canonical Wnt signaling in the neocortex and in the caudal forebrain. Interestingly, the ultrastructure and morphology of ventricular cilia in the cobblestone mutants remains intact. Together, these results indicate a critical role for ciliary function in the developing forebrain.


Subject(s)
Cerebral Cortex/abnormalities , Cerebral Cortex/metabolism , Cilia/metabolism , Gene Expression Regulation, Developmental/genetics , Kruppel-Like Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Tumor Suppressor Proteins/genetics , Animals , Cerebral Cortex/ultrastructure , Cilia/ultrastructure , Ependyma/metabolism , Ependyma/ultrastructure , Female , Kruppel-Like Transcription Factors/genetics , Lateral Ventricles/abnormalities , Lateral Ventricles/metabolism , Lateral Ventricles/ultrastructure , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neurons/metabolism , Neurons/ultrastructure , Peptide Hydrolases/metabolism , Prosencephalon/abnormalities , Prosencephalon/metabolism , Prosencephalon/ultrastructure , Tumor Suppressor Proteins/metabolism , Wnt Proteins/metabolism , Zinc Finger Protein Gli3
SELECTION OF CITATIONS
SEARCH DETAIL
...