Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 982
Filter
1.
Sci Rep ; 11(1): 19115, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34580355

ABSTRACT

Amyloid precursor protein (APP) is expressed in many tissues in human, mice and in zebrafish. In zebrafish, there are two orthologues, Appa and Appb. Interestingly, some cellular processes associated with APP overlap with cilia-mediated functions. Whereas the localization of APP to primary cilia of in vitro-cultured cells has been reported, we addressed the presence of APP in motile and in non-motile sensory cilia and its potential implication for ciliogenesis using zebrafish, mouse, and human samples. We report that Appa and Appb are expressed by ciliated cells and become localized at the membrane of cilia in the olfactory epithelium, otic vesicle and in the brain ventricles of zebrafish embryos. App in ependymal cilia persisted in adult zebrafish and was also detected in mouse and human brain. Finally, we found morphologically abnormal ependymal cilia and smaller brain ventricles in appa-/-appb-/- mutant zebrafish. Our findings demonstrate an evolutionary conserved localisation of APP to cilia and suggest a role of App in ciliogenesis and cilia-related functions.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Amyloidogenic Proteins/metabolism , Cerebral Ventricles/metabolism , Zebrafish Proteins/metabolism , Amyloid beta-Protein Precursor/analysis , Amyloid beta-Protein Precursor/genetics , Amyloidogenic Proteins/analysis , Amyloidogenic Proteins/genetics , Animals , Animals, Genetically Modified , Cerebral Ventricles/cytology , Cilia/metabolism , Embryo, Nonmammalian , Ependyma/cytology , Ependyma/metabolism , Humans , Mice , Models, Animal , Mutation , Olfactory Mucosa/cytology , Olfactory Mucosa/metabolism , Zebrafish , Zebrafish Proteins/analysis , Zebrafish Proteins/genetics
2.
Fluids Barriers CNS ; 18(1): 31, 2021 Jul 07.
Article in English | MEDLINE | ID: mdl-34233705

ABSTRACT

BACKGROUND: Cerebrospinal fluid (CSF) is an ultra-filtrated colorless brain fluid that circulates within brain spaces like the ventricular cavities, subarachnoid space, and the spine. Its continuous flow serves many primary functions, including nourishment, brain protection, and waste removal. MAIN BODY: The abnormal accumulation of CSF in brain cavities triggers severe hydrocephalus. Accumulating evidence had indicated that synchronized beats of motile cilia (cilia from multiciliated cells or the ependymal lining in brain ventricles) provide forceful pressure to generate and restrain CSF flow and maintain overall CSF circulation within brain spaces. In humans, the disorders caused by defective primary and/or motile cilia are generally referred to as ciliopathies. The key role of CSF circulation in brain development and its functioning has not been fully elucidated. CONCLUSIONS: In this review, we briefly discuss the underlying role of motile cilia in CSF circulation and hydrocephalus. We have reviewed cilia and ciliated cells in the brain and the existing evidence for the regulatory role of functional cilia in CSF circulation in the brain. We further discuss the findings obtained for defective cilia and their potential involvement in hydrocephalus. Furthermore, this review will reinforce the idea of motile cilia as master regulators of CSF movements, brain development, and neuronal diseases.


Subject(s)
Brain/physiology , Cerebrospinal Fluid/physiology , Cilia/physiology , Hydrocephalus/cerebrospinal fluid , Hydrocephalus/physiopathology , Animals , Brain/cytology , Cerebral Ventricles/cytology , Cerebral Ventricles/physiology , Humans
3.
Science ; 372(6547): 1205-1209, 2021 06 11.
Article in English | MEDLINE | ID: mdl-34112692

ABSTRACT

Quiescent neural stem cells (NSCs) in the adult mouse ventricular-subventricular zone (V-SVZ) undergo activation to generate neurons and some glia. Here we show that platelet-derived growth factor receptor beta (PDGFRß) is expressed by adult V-SVZ NSCs that generate olfactory bulb interneurons and glia. Selective deletion of PDGFRß in adult V-SVZ NSCs leads to their release from quiescence, uncovering gliogenic domains for different glial cell types. These domains are also recruited upon injury. We identify an intraventricular oligodendrocyte progenitor derived from NSCs inside the brain ventricles that contacts supraependymal axons. Together, our findings reveal that the adult V-SVZ contains spatial domains for gliogenesis, in addition to those for neurogenesis. These gliogenic NSC domains tend to be quiescent under homeostasis and may contribute to brain plasticity.


Subject(s)
Adult Stem Cells/physiology , Cerebral Ventricles/physiology , Lateral Ventricles/physiology , Neural Stem Cells/physiology , Neuroglia/physiology , Receptor, Platelet-Derived Growth Factor beta/metabolism , Animals , Astrocytes/cytology , Astrocytes/physiology , Axons/physiology , Cell Differentiation , Cell Division , Cerebral Ventricles/cytology , Ependyma/cytology , Ependyma/physiology , Female , Gene Expression Profiling , Homeostasis , Lateral Ventricles/cytology , Male , Mice , Neurogenesis , Olfactory Bulb/cytology , Olfactory Bulb/physiology , Oligodendroglia/cytology , Oligodendroglia/physiology , Receptor, Platelet-Derived Growth Factor beta/genetics
4.
J Neurosci ; 41(15): 3301-3306, 2021 04 14.
Article in English | MEDLINE | ID: mdl-33597270

ABSTRACT

Heterogeneity is defined as the quality or state of being diverse in character or content. This article summarizes the natural progression from my studies, reported in the first issue of the Journal of Neuroscience, that identified molecular heterogeneity in precursor cells of the developing primate cerebral cortex to the current state in which differences defined at the molecular, cellular, circuit, and systems levels are building data encyclopedias. The emphasis on heterogeneity has impacted many contributors in the field of developmental neuroscience, who have led a quest to determine the extent to which there is diversity, when it appears developmentally, and what heritable and nonheritable factors mediate nervous system assembly and function. Since the appearance of the article on progenitor cell heterogeneity in the inaugural issue of the Journal of Neuroscience, there have been continuous advances in technologies and data analytics that are contributing to a much better understanding of the origins of neurobiological and behavioral heterogeneity.


Subject(s)
Cerebral Ventricles/cytology , Neural Stem Cells/physiology , Neurogenesis , Neuroglia/physiology , Animals , Cerebral Ventricles/growth & development , Cerebral Ventricles/physiology , Humans , Neural Stem Cells/cytology , Neuroglia/cytology
6.
Cereb Cortex ; 31(4): 2139-2155, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33279961

ABSTRACT

Microglial cells make extensive contacts with neural precursor cells (NPCs) and affiliate with vasculature in the developing cerebral cortex. But how vasculature contributes to cortical histogenesis is not yet fully understood. To better understand functional roles of developing vasculature in the embryonic rat cerebral cortex, we investigated the temporal and spatial relationships between vessels, microglia, and NPCs in the ventricular zone. Our results show that endothelial cells in developing cortical vessels extend numerous fine processes that directly contact mitotic NPCs and microglia; that these processes protrude from vessel walls and are distinct from tip cell processes; and that microglia, NPCs, and vessels are highly interconnected near the ventricle. These findings demonstrate the complex environment in which NPCs are embedded in cortical proliferative zones and suggest that developing vasculature represents a source of signaling with the potential to broadly influence cortical development. In summary, cortical histogenesis arises from the interplay among NPCs, microglia, and developing vasculature. Thus, factors that impinge on any single component have the potential to change the trajectory of cortical development and increase susceptibility for altered neurodevelopmental outcomes.


Subject(s)
Cerebral Ventricles/blood supply , Cerebral Ventricles/embryology , Neocortex/blood supply , Neocortex/embryology , Neurogenesis/physiology , Neuroimmunomodulation/physiology , Animals , Cerebral Ventricles/cytology , Embryonic Development/physiology , Female , Microglia/physiology , Neocortex/cytology , Neural Stem Cells/physiology , Pregnancy , Rats
7.
Mol Brain ; 13(1): 169, 2020 12 14.
Article in English | MEDLINE | ID: mdl-33317583

ABSTRACT

The brain is composed of cells having distinct genomic DNA sequences that arise post-zygotically, known as somatic genomic mosaicism (SGM). One form of SGM is aneuploidy-the gain and/or loss of chromosomes-which is associated with mitotic spindle defects. The mitotic spindle orientation determines cleavage plane positioning and, therefore, neural progenitor cell (NPC) fate during cerebral cortical development. Here we report receptor-mediated signaling by lysophosphatidic acid (LPA) as a novel extracellular signal that influences cleavage plane orientation and produces alterations in SGM by inducing aneuploidy during murine cortical neurogenesis. LPA is a bioactive lipid whose actions are mediated by six G protein-coupled receptors, LPA1-LPA6. RNAscope and qPCR assessment of all six LPA receptor genes, and exogenous LPA exposure in LPA receptor (Lpar)-null mice, revealed involvement of Lpar1 and Lpar2 in the orientation of the mitotic spindle. Lpar1 signaling increased non-vertical cleavage in vivo by disrupting cell-cell adhesion, leading to breakdown of the ependymal cell layer. In addition, genomic alterations were significantly increased after LPA exposure, through production of chromosomal aneuploidy in NPCs. These results identify LPA as a receptor-mediated signal that alters both NPC fate and genomes during cortical neurogenesis, thus representing an extracellular signaling mechanism that can produce stable genomic changes in NPCs and their progeny. Normal LPA signaling in early life could therefore influence both the developing and adult brain, whereas its pathological disruption could contribute to a range of neurological and psychiatric diseases, via long-lasting somatic genomic alterations.


Subject(s)
Aneuploidy , Cerebral Cortex/cytology , Genome , Neural Stem Cells/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Signal Transduction , Adherens Junctions/metabolism , Animals , Cell Adhesion , Cell Division , Cell Polarity , Cell Proliferation , Cells, Cultured , Cerebral Cortex/embryology , Cerebral Ventricles/cytology , Lysophospholipids/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mosaicism , Neural Stem Cells/cytology , Neurogenesis
8.
Bioessays ; 42(3): e1900186, 2020 03.
Article in English | MEDLINE | ID: mdl-32078177

ABSTRACT

The brain ventricular system is a series of connected cavities, filled with cerebrospinal fluid (CSF), that forms within the vertebrate central nervous system (CNS). The hollow neural tube is a hallmark of the chordate CNS, and a closed neural tube is essential for normal development. Development and function of the ventricular system is examined, emphasizing three interdigitating components that form a functional system: ventricle walls, CSF fluid properties, and activity of CSF constituent factors. The cellular lining of the ventricle both can produce and is responsive to CSF. Fluid properties and conserved CSF components contribute to normal CNS development. Anomalies of the CSF/ventricular system serve as diagnostics and may cause CNS disorders, further highlighting their importance. This review focuses on the evolution and development of the brain ventricular system, associated function, and connected pathologies. It is geared as an introduction for scholars with little background in the field.


Subject(s)
Cerebral Ventricles/growth & development , Cerebral Ventricles/metabolism , Cerebrospinal Fluid/metabolism , Animals , Biological Evolution , Brain Diseases/metabolism , Cerebral Ventricles/cytology , Cerebrospinal Fluid Pressure/physiology , Cerebrospinal Fluid Proteins/metabolism , Cilia/metabolism , Epithelium/growth & development , Epithelium/metabolism , Humans , Kinetics , Neural Tube/cytology , Neural Tube/growth & development , Neural Tube/metabolism , Signal Transduction
9.
Cereb Cortex ; 30(7): 4092-4109, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32108222

ABSTRACT

Even after birth, neuronal production continues in the ventricular-subventricular zone (V-SVZ) and hippocampus in many mammals. The immature new neurons ("neuroblasts") migrate and then mature at their final destination. In humans, neuroblast production and migration toward the neocortex and the olfactory bulb (OB) occur actively only for a few months after birth and then sharply decline with age. However, the precise spatiotemporal profiles and fates of postnatally born neurons remain unclear due to methodological limitations. We previously found that common marmosets, small nonhuman primates, share many features of V-SVZ organization with humans. Here, using marmosets injected with thymidine analogue(s) during various postnatal periods, we demonstrated spatiotemporal changes in neurogenesis during development. V-SVZ progenitor proliferation and neuroblast migration toward the OB and neocortex sharply decreased by 4 months, most strikingly in a V-SVZ subregion from which neuroblasts migrated toward the neocortex. Postnatally born neurons matured within a few months in the OB and hippocampus but remained immature until 6 months in the neocortex. While neurogenic activity was sustained for a month after birth, the distribution and/or differentiation diversity was more restricted in 1-month-born cells than in the neonatal-born population. These findings shed light on distinctive features of postnatal neurogenesis in primates.


Subject(s)
Cell Proliferation , Hippocampus/growth & development , Lateral Ventricles/growth & development , Neocortex/growth & development , Neural Stem Cells/cytology , Neurogenesis , Olfactory Bulb/growth & development , Animals , Brain/cytology , Brain/growth & development , Callithrix , Cell Movement , Cerebral Ventricles/cytology , Cerebral Ventricles/growth & development , Hippocampus/cytology , Lateral Ventricles/cytology , Neocortex/cytology , Olfactory Bulb/cytology , Spatio-Temporal Analysis
10.
J Anat ; 236(2): 334-350, 2020 02.
Article in English | MEDLINE | ID: mdl-31670387

ABSTRACT

The ventricular layer of the spinal cord is remodelled during embryonic development and ultimately forms the ependymal cell lining of the adult central canal, which retains neural stem cell potential. This anatomical transformation involves the process of dorsal collapse; however, accompanying changes in tissue organisation and cell behaviour as well as the precise origin of cells contributing to the central canal are not well understood. Here, we describe sequential localised cell rearrangements which accompany the gradual attrition of the spinal cord ventricular layer during development. This includes local breakdown of the pseudostratified organisation of the dorsal ventricular layer prefiguring dorsal collapse and evidence for a new phenomenon, ventral dissociation, during which the ventral-most floor plate cells separate from a subset that are retained around the central canal. Using cell proliferation markers and cell-cycle reporter mice, we further show that following dorsal collapse, ventricular layer attrition involves an overall reduction in cell proliferation, characterised by an intriguing increase in the percentage of cells in G1/S. In contrast, programmed cell death does not contribute to ventricular layer remodelling. By analysing transcript and protein expression patterns associated with key signalling pathways, we provide evidence for a gradual decline in ventral sonic hedgehog activity and an accompanying ventral expansion of initial dorsal bone morphogenetic protein signalling, which comes to dominate the forming the central canal lining. This study identifies multiple steps that may contribute to spinal cord ventricular layer attrition and adds to increasing evidence for the heterogeneous origin of the spinal cord ependymal cell population, which includes cells from the floor plate and the roof plate as well as ventral progenitor domains.


Subject(s)
Cell Proliferation/physiology , Cerebral Ventricles/cytology , Spinal Cord/cytology , Animals , Apoptosis/physiology , Bone Morphogenetic Proteins/metabolism , Cell Cycle/physiology , Cerebral Ventricles/metabolism , Ependyma/cytology , Ependyma/metabolism , Hedgehog Proteins/metabolism , Mice , SOXB1 Transcription Factors/metabolism , Signal Transduction/physiology , Spinal Cord/metabolism
11.
Glia ; 68(2): 435-450, 2020 02.
Article in English | MEDLINE | ID: mdl-31626379

ABSTRACT

Postnatal subventricular zone (SVZ) neural stem cells generate forebrain glia, namely astrocytes and oligodendrocytes. The cues necessary for this process are unclear, despite this phase of brain development being pivotal in forebrain gliogenesis. Galectin-3 (Gal-3) is increased in multiple brain pathologies and thereby regulates astrocyte proliferation and inflammation in injury. To study the function of Gal-3 in inflammation and gliogenesis, we carried out functional studies in mouse. We overexpressed Gal-3 with electroporation and using immunohistochemistry surprisingly found no inflammation in the healthy postnatal SVZ. This allowed investigation of inflammation-independent effects of Gal-3 on gliogenesis. Loss of Gal-3 function via knockdown or conditional knockout reduced gliogenesis, whereas Gal-3 overexpression increased it. Gal-3 overexpression also increased the percentage of striatal astrocytes generated by the SVZ but decreased the percentage of oligodendrocytes. These novel findings were further elaborated with multiple analyses demonstrating that Gal-3 binds to the bone morphogenetic protein receptor one alpha (BMPR1α) and increases bone morphogenetic protein (BMP) signaling. Conditional knockout of BMPR1α abolished the effect of Gal-3 overexpression on gliogenesis. Gain-of-function of Gal-3 is relevant in pathological conditions involving the human forebrain, which is particularly vulnerable to hypoxia/ischemia during perinatal gliogenesis. Hypoxic/ischemic injury induces astrogliosis, inflammation and cell death. We show that Gal-3 immunoreactivity was increased in the perinatal human SVZ and striatum after hypoxia/ischemia. Our findings thus show a novel inflammation-independent function for Gal-3; it is necessary for gliogenesis and when increased in expression can induce astrogenesis via BMP signaling.


Subject(s)
Astrocytes/metabolism , Galectin 3/metabolism , Lateral Ventricles/cytology , Neuroglia/metabolism , Animals , Cell Differentiation/physiology , Cell Movement/physiology , Cerebral Ventricles/cytology , Gene Expression Regulation , Ischemia/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Neural Stem Cells/metabolism , Neurogenesis/physiology , Oligodendroglia/metabolism
12.
J Vis Exp ; (147)2019 05 11.
Article in English | MEDLINE | ID: mdl-31132045

ABSTRACT

Gene control of neuronal cytoarchitecture is currently the subject of intensive investigation. Described here is a simple method developed to study in vivo gene control of neocortical projection neuron morphology. This method is based on (1) in vitro lentiviral engineering of neuronal precursors as "test" and "control" cells, (2) their co-transplantation into wild-type brains, and (3) paired morphometric evaluation of their neuronal derivatives. Specifically, E12.5 pallial precursors from panneuronal, genetically labeled donors, are employed for this purpose. They are engineered to take advantage of selected promoters and tetON/OFF technology, and they are free-hand transplanted into neonatal lateral ventricles. Later, upon immunofluorescence profiling of recipient brains, silhouettes of transplanted neurons are fed into NeurphologyJ open source software, their morphometric parameters are extracted, and average length and branching index are calculated. Compared to other methods, this one offers three main advantages: it permits achieving of fine control of transgene expression at affordable costs, it only requires basic surgical skills, and it provides statistically reliable results upon analysis of a limited number of animals. Because of its design, however, it is not adequate to address non cell-autonomous control of neuroarchitecture. Moreover, it should be preferably used to investigate neurite morphology control after completion of neuronal migration. In its present formulation, this method is exquisitely tuned to investigate gene control of glutamatergic neocortical neuron architecture. Taking advantage of transgenic lines expressing EGFP in other specific neural cell types, it can be re-purposed to address gene control of their architecture.


Subject(s)
Cerebral Ventricles/cytology , Neural Stem Cells/transplantation , Animals , Cell Differentiation/genetics , Cell Movement , Female , Male , Mice , Neurites , Neurons/physiology , Tissue Engineering
13.
Elife ; 82019 02 18.
Article in English | MEDLINE | ID: mdl-30775968

ABSTRACT

We use the transparency of zebrafish embryos to reveal the de novo generation of a simple squamous epithelium and identify the cellular architecture in the epithelial transition zone that ties this squamous epithelium to the columnar neuroepithelium within the embryo's brain. The simple squamous epithelium of the rhombencephalic roof plate is pioneered by distinct mesenchymal cells at the dorsal midline of the neural tube. Subsequently, a progenitor zone is established at the interface between columnar epithelium of the rhombic lip and the expanding squamous epithelium of the roof plate. Surprisingly, this interface consists of a single progenitor cell type that we have named the veil cell. Veil cells express gdf6a and constitute a lineage restricted stem zone that generates the squamous roof plate by direct transformation and asymmetrically fated divisions. Experimental restriction of roof plate expansion leads to extrusion of veil cell daughters and squamous cells, suggesting veil cell fate is regulated by the space available for roof plate growth.


Subject(s)
Cerebral Ventricles/anatomy & histology , Epithelium/anatomy & histology , Zebrafish/anatomy & histology , Animals , Asymmetric Cell Division , Cell Proliferation , Cell Self Renewal , Cerebral Ventricles/cytology , Embryo, Nonmammalian/cytology , Epithelium/embryology , Growth Differentiation Factor 6/metabolism , Mesoderm/embryology , Rhombencephalon/anatomy & histology , Rhombencephalon/embryology , Zebrafish/embryology , Zebrafish Proteins/metabolism
14.
Stem Cells Dev ; 28(8): 515-527, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30760110

ABSTRACT

White matter damage persists in hypoxic-ischemic newborns even when treated with hypothermia. We have previously shown that intraventricular delivery of human glial progenitors (GPs) at the neonatal stage is capable of replacing abnormal host glia and rescuing the lifespan of dysmyelinated mice. However, such transplantation in the human brain poses significant challenges as related to high-volume ventricles and long cell migration distances. These challenges can only be studied in large animal model systems. In this study, we developed a three dimensional (3D)-printed model of the ventricular system sized to a newborn pig to investigate the parameters that can maximize a global biodistribution of injected GPs within the ventricular system, while minimizing outflow to the subarachnoid space. Bioluminescent imaging and magnetic resonance imaging were used to image the biodistribution of luciferase-transduced GPs in simple fluid containers and a custom-designed, 3D-printed model of the piglet ventricular system. Seven independent variables were investigated. The results demonstrated that a low volume (0.1 mL) of cell suspension is essential to keep cells within the ventricular system. If higher volumes (1 mL) are needed, a very slow infusion speed (0.01 mL/min) is necessary. Real-time magnetic resonance imaging demonstrated that superparamagnetic iron oxide (SPIO) labeling significantly alters the rheological properties of the GP suspension, such that, even at high speeds and high volumes, the outflow to the subarachnoid space is reduced. Several other factors, including GP species (human vs. mouse), type of catheter tip (end hole vs. side hole), catheter length (0.3 vs. 7.62 m), and cell concentration, had less effect on the overall distribution of GPs. We conclude that the use of a 3D-printed phantom model represents a robust, reproducible, and cost-saving alternative to in vivo large animal studies for determining optimal injection parameters.


Subject(s)
Cerebral Ventricles , Models, Anatomic , Neural Stem Cells/cytology , Neuroglia/cytology , Printing, Three-Dimensional , Animals , Animals, Newborn , Brain/growth & development , Brain/metabolism , Brain/pathology , Cells, Cultured , Cerebral Ventricles/cytology , Cerebral Ventricles/growth & development , Cerebral Ventricles/metabolism , Fluorescent Dyes/pharmacokinetics , Humans , Magnetite Nanoparticles/analysis , Mice , Neural Stem Cells/physiology , Neuroglia/physiology , Swine , Tissue Distribution
15.
Neuron ; 101(4): 625-634.e3, 2019 02 20.
Article in English | MEDLINE | ID: mdl-30661739

ABSTRACT

In vertebrates, commissural axons extend ventrally toward the floor plate in the spinal cord and hindbrain. Netrin-1, secreted by floor plate cells, was proposed to attract commissural axons at a distance. However, recent genetic studies in mice have shown that netrin-1 is also produced by ventricular zone (VZ) progenitors and that in the hindbrain, it represents the main source of netrin-1 for commissural axons. Here, we show that genetically deleting netrin-1 either from the VZ or the floor plate does not prevent midline crossing in the spinal cord, although axon pathfinding and fasciculation are perturbed. Strikingly, the VZ and floor plate act synergistically, as the simultaneous ablation of netrin-1 from these two sources severely impedes crossing. These results suggest that floor-plate-derived netrin-1 has a distinct impact on commissural axons in the spinal cord and hindbrain.


Subject(s)
Axon Guidance , Cerebral Ventricles/embryology , Netrin-1/metabolism , Neurons/metabolism , Rhombencephalon/embryology , Spinal Cord/embryology , Animals , Cerebral Ventricles/cytology , Cerebral Ventricles/metabolism , Female , Male , Mice , Netrin-1/genetics , Neurons/cytology , Rhombencephalon/cytology , Rhombencephalon/metabolism , Spinal Cord/cytology , Spinal Cord/metabolism
16.
Neuron ; 101(4): 635-647.e4, 2019 02 20.
Article in English | MEDLINE | ID: mdl-30661738

ABSTRACT

An important model for axon pathfinding is provided by guidance of embryonic commissural axons from dorsal spinal cord to ventral midline floor plate (FP). FP cells produce a chemoattractive activity, comprised largely of netrin1 (FP-netrin1) and Sonic hedgehog (Shh), that can attract the axons at a distance in vitro. netrin1 is also produced by ventricular zone (VZ) progenitors along the axons' route (VZ-netrin1). Recent studies using region-specific netrin1 deletion suggested that FP-netrin1 is dispensable and VZ-netrin1 sufficient for netrin guidance activity in vivo. We show that removing FP-netrin1 actually causes guidance defects in spinal cord consistent with long-range action (i.e., over hundreds of micrometers), and double mutant analysis supports that FP-netrin1 and Shh collaborate to attract at long range. We further provide evidence that netrin1 may guide via chemotaxis or haptotaxis. These results support the model that netrin1 signals at both short and long range to guide commissural axons in spinal cord.


Subject(s)
Axon Guidance , Cerebral Ventricles/embryology , Hedgehog Proteins/metabolism , Netrin-1/metabolism , Neurons/metabolism , Spinal Cord/embryology , Animals , Cells, Cultured , Cerebral Ventricles/cytology , Cerebral Ventricles/metabolism , Female , Hedgehog Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Netrin-1/genetics , Neurons/cytology , Rats , Rats, Sprague-Dawley , Rhombencephalon/cytology , Rhombencephalon/embryology , Rhombencephalon/metabolism , Spinal Cord/cytology , Spinal Cord/metabolism
17.
Sci Rep ; 8(1): 16855, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30442908

ABSTRACT

The neuroplastic mechanisms in the fish brain that underlie sex reversal remain unknown. Gonadotropin-releasing hormone 3 (GnRH3) neurons control male reproductive behaviours in Mozambique tilapia and show sexual dimorphism, with males having a greater number of GnRH3 neurons. Treatment with androgens such as 11-ketotestosterone (KT), but not 17ß-estradiol, increases the number of GnRH3 neurons in mature females to a level similar to that observed in mature males. Compared with oestrogen, the effect of androgen on neurogenesis remains less clear. The present study examined the effects of 11-KT, a non-aromatizable androgen, on cellular proliferation, neurogenesis, generation of GnRH3 neurons and expression of cell cycle-related genes in mature females. The number of proliferating cell nuclear antigen-positive cells was increased by 11-KT. Simultaneous injection of bromodeoxyuridine and 11-KT significantly increased the number of newly-generated (newly-proliferated) neurons, but did not affect radial glial cells, and also resulted in newly-generated GnRH3 neurons. Transcriptome analysis showed that 11-KT modulates the expression of genes related to the cell cycle process. These findings suggest that tilapia could serve as a good animal model to elucidate the effects of androgen on adult neurogenesis and the mechanisms for sex reversal in the fish brain.


Subject(s)
Androgens/pharmacology , Brain/cytology , Brain/metabolism , Gonadotropin-Releasing Hormone/metabolism , Neurogenesis/drug effects , Neurons/metabolism , Tilapia/metabolism , Animals , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Proliferation/drug effects , Cerebral Ventricles/cytology , Female , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , Image Processing, Computer-Assisted , Neuroglia/cytology , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/drug effects , Proliferating Cell Nuclear Antigen/metabolism , Testosterone/analogs & derivatives , Testosterone/pharmacology
18.
Glia ; 66(12): 2659-2672, 2018 12.
Article in English | MEDLINE | ID: mdl-30338559

ABSTRACT

Nuclear factor-kappaB (NF-κB) is activated in neural progenitor cells in the developing murine cerebral cortex during the neurogenic phase, when it acts to prevent premature neuronal differentiation. Here we show that NF-κB activation continues in mouse neocortical neural progenitor cells during the neurogenic-to-gliogenic switch. Blockade of endogenous NF-κB activity during neocortical gliogenesis leads to the formation of supernumerary committed gliogenic progenitors and premature glial cell differentiation. Conversely, forced NF-κB activation during the neocortical neurogenic-to-gliogenic transition causes delayed gliogenic commitment and decreased astroglial gene expression. NF-κB activation continues in neocortical gliogenic progenitors following commitment and is important to inhibit the differentiation of oligodendrocyte precursor cells and to maintain persistent expression of glial fibrillary acidic protein in maturing astrocytes. These results reveal a number of previously uncharacterized roles for NF-κB during different phases of neocortical gliogenesis and identify NF-κB as an inhibitor of early oligodendrocyte development in the cerebral cortex.


Subject(s)
Cerebral Cortex , Gene Expression Regulation, Developmental/genetics , NF-kappa B/metabolism , Neurogenesis/genetics , Neuroglia/physiology , Animals , Animals, Newborn , Cell Differentiation/physiology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Cerebral Cortex/growth & development , Cerebral Ventricles/cytology , Cerebral Ventricles/embryology , Cerebral Ventricles/growth & development , Ciliary Neurotrophic Factor/pharmacology , Embryo, Mammalian , Gene Expression Regulation, Developmental/physiology , Glial Fibrillary Acidic Protein/metabolism , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Ki-67 Antigen/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/physiology
19.
J Mol Neurosci ; 65(1): 60-73, 2018 May.
Article in English | MEDLINE | ID: mdl-29705933

ABSTRACT

Mesenchymal stem cells (MSCs) are regarded as an immune privileged cell type with numerous regeneration-promoting effects. The in vivo behavior of MSC and underlying mechanisms leading to their regenerative effects are largely unknown. The aims of this study were to comparatively investigate the in vivo behavior of canine (cMSC), human (hMSC), and murine MSC (mMSC) following intra-cerebroventricular transplantation. At 7 days post transplantation (dpt), clusters of cMSC, hMSC, and mMSC were detected within the ventricular system. At 49 dpt, cMSC-transplanted mice showed clusters mostly consisting of extracellular matrix lacking transplanted MSC. Similarly, hMSC-transplanted mice lacked MSC clusters at 49 dpt. Xenogeneic MSC transplantation was associated with a local T lymphocyte-dominated immune reaction at both time points. Interestingly, no associated inflammation was observed following syngeneic mMSC transplantation. In conclusion, transplanted MSC formed intraventricular cell clusters and exhibited a short life span in vivo. Xenogeneically in contrast to syngeneically transplanted MSC triggered a T cell-mediated graft rejection indicating that MSCs are not as immune privileged as previously assumed. However, MSC may mediate their effects by a "hit and run" mechanism and future studies will show whether syngeneically or xenogeneically transplanted MSCs exert better therapeutic effects in animals with CNS disease.


Subject(s)
Cerebral Ventricles/surgery , Heterografts/cytology , Mesenchymal Stem Cell Transplantation/adverse effects , Mesenchymal Stem Cells/cytology , Animals , Cells, Cultured , Cerebral Ventricles/cytology , Dogs , Female , Graft Rejection/immunology , Heterografts/immunology , Humans , Male , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/immunology , Mice , Mice, Inbred C57BL , T-Lymphocytes/immunology
20.
Cell Mol Life Sci ; 75(6): 1027-1041, 2018 03.
Article in English | MEDLINE | ID: mdl-29018869

ABSTRACT

Originating from ectodermal epithelium, radial glial cells (RGCs) retain apico-basolateral polarity and comprise a pseudostratified epithelial layer in the developing cerebral cortex. The apical endfeet of the RGCs faces the fluid-filled ventricles, while the basal processes extend across the entire cortical span towards the pial surface. RGC functions are largely dependent on this polarized structure and the molecular components that define it. In this review, we will dissect existing molecular evidence on RGC polarity establishment and during cerebral cortex development and provide our perspective on the remaining key questions.


Subject(s)
Cell Polarity , Cerebral Cortex/metabolism , Ectoderm/metabolism , Gene Expression Regulation, Developmental , Nerve Tissue Proteins/genetics , Neuroglia/metabolism , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Adherens Junctions/metabolism , Adherens Junctions/ultrastructure , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Cerebral Ventricles/cytology , Cerebral Ventricles/growth & development , Cerebral Ventricles/metabolism , Ectoderm/cytology , Ectoderm/growth & development , Embryo, Mammalian , Epithelium/growth & development , Epithelium/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix/ultrastructure , Humans , Nerve Tissue Proteins/metabolism , Neuroglia/cytology , Pia Mater/cytology , Pia Mater/growth & development , Pia Mater/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...