Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Proc Natl Acad Sci U S A ; 120(50): e2304074120, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38051767

ABSTRACT

Severity of neurobehavioral deficits in children born from adverse pregnancies, such as maternal alcohol consumption and diabetes, does not always correlate with the adversity's duration and intensity. Therefore, biological signatures for accurate prediction of the severity of neurobehavioral deficits, and robust tools for reliable identification of such biomarkers, have an urgent clinical need. Here, we demonstrate that significant changes in the alternative splicing (AS) pattern of offspring lymphocyte RNA can function as accurate peripheral biomarkers for motor learning deficits in mouse models of prenatal alcohol exposure (PAE) and offspring of mother with diabetes (OMD). An aptly trained deep-learning model identified 29 AS events common to PAE and OMD as superior predictors of motor learning deficits than AS events specific to PAE or OMD. Shapley-value analysis, a game-theory algorithm, deciphered the trained deep-learning model's learnt associations between its input, AS events, and output, motor learning performance. Shapley values of the deep-learning model's input identified the relative contribution of the 29 common AS events to the motor learning deficit. Gene ontology and predictive structure-function analyses, using Alphafold2 algorithm, supported existing evidence on the critical roles of these molecules in early brain development and function. The direction of most AS events was opposite in PAE and OMD, potentially from differential expression of RNA binding proteins in PAE and OMD. Altogether, this study posits that AS of lymphocyte RNA is a rich resource, and deep-learning is an effective tool, for discovery of peripheral biomarkers of neurobehavioral deficits in children of diverse adverse pregnancies.


Subject(s)
Diabetes Mellitus , Fetal Alcohol Spectrum Disorders , Prenatal Exposure Delayed Effects , Mice , Animals , Child , Humans , Pregnancy , Female , Alternative Splicing , Prenatal Exposure Delayed Effects/chemically induced , Ethanol , Diabetes Mellitus/chemically induced , Biomarkers/metabolism , RNA/metabolism , Fetal Alcohol Spectrum Disorders/genetics
2.
Mol Psychiatry ; 2024 May 11.
Article in English | MEDLINE | ID: mdl-38734844

ABSTRACT

A hallmark of fetal alcohol spectrum disorders (FASD) is neurobehavioral deficits that still do not have effective treatment. Here, we present that reduction of Apolipoprotein E (APOE) is critically involved in neurobehavioral deficits in FASD. We show that prenatal alcohol exposure (PAE) changes chromatin accessibility of Apoe locus, and causes reduction of APOE levels in both the brain and peripheral blood in postnatal mice. Of note, postnatal administration of an APOE receptor agonist (APOE-RA) mitigates motor learning deficits and anxiety in those mice. Several molecular and electrophysiological properties essential for learning, which are altered by PAE, are restored by APOE-RA. Our human genome-wide association study further reveals that the interaction of PAE and a single nucleotide polymorphism in the APOE enhancer which chromatin is closed by PAE in mice is associated with lower scores in the delayed matching-to-sample task in children. APOE in the plasma is also reduced in PAE children, and the reduced level is associated with their lower cognitive performance. These findings suggest that controlling the APOE level can serve as an effective treatment for neurobehavioral deficits in FASD.

3.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Article in English | MEDLINE | ID: mdl-33443207

ABSTRACT

The developing brain is under the risk of exposure to a multitude of environmental stressors. While perinatal exposure to excessive levels of environmental stress is responsible for a wide spectrum of neurological and psychiatric conditions, the developing brain is equipped with intrinsic cell protection, the mechanisms of which remain unknown. Here we show, using neonatal mouse as a model system, that primary cilia, hair-like protrusions from the neuronal cell body, play an essential role in protecting immature neurons from the negative impacts of exposure to environmental stress. More specifically, we found that primary cilia prevent the degeneration of dendritic arbors upon exposure to alcohol and ketamine, two major cell stressors, by activating cilia-localized insulin-like growth factor 1 receptor and downstream Akt signaling. We also found that activation of this pathway inhibits Caspase-3 activation and caspase-mediated cleavage/fragmentation of cytoskeletal proteins in stress-exposed neurons. These results indicate that primary cilia play an integral role in mitigating adverse impacts of environmental stressors such as drugs on perinatal brain development.


Subject(s)
Cilia/metabolism , Neural Stem Cells/metabolism , Prosencephalon/embryology , Animals , Animals, Newborn/metabolism , Brain/metabolism , Dendrites/metabolism , Female , Insulin-Like Growth Factor I/metabolism , Mice/embryology , Mice, Inbred C57BL , Neurons/metabolism , Pregnancy , Prenatal Exposure Delayed Effects , Prosencephalon/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
4.
Dev Neurosci ; 45(2): 66-75, 2023.
Article in English | MEDLINE | ID: mdl-36642064

ABSTRACT

Throughout our lives, we are exposed to a variety of hazards, such as environmental pollutants and chemical substances that affect our health, and viruses and bacteria that cause infectious diseases. These external factors that are undesirable to an organism are called environmental stress. During the perinatal period, when neural networks are drastically reorganized and refined, the tolerance of the developing brain to various environmental stresses is lower than in adulthood. Thus, exposure to environmental stress during this vulnerable period is strongly associated with cognitive and behavioral deficits in later life. Recent studies have uncovered various mechanisms underlying the adverse impacts of environmental stress during the perinatal period on brain development. In this mini-review, we will present the findings from these studies, focusing on caspase-mediated apoptotic and nonapoptotic effects of environmental stress, and discuss several compounds that mitigate these caspase-mediated effects as examples of potential therapeutic approaches.


Subject(s)
Brain , Ethanol , Pregnancy , Female , Humans , Caspase 3/pharmacology , Caspases/pharmacology
5.
Cereb Cortex ; 30(12): 6444-6457, 2020 11 03.
Article in English | MEDLINE | ID: mdl-32780108

ABSTRACT

The development of the cerebral cortex is directed by a series of methodically precise events, including progenitor cell proliferation, neural differentiation, and cell positioning. Over the past decade, many studies have demonstrated the critical contributions of Notch signaling in neurogenesis, including that in the developing telencephalon. However, in vivo evidence for the role of Notch signaling in cortical development still remains limited partly due to the redundant functions of four mammalian Notch paralogues and embryonic lethality of the knockout mice. Here, we utilized the conditional deletion and in vivo gene manipulation of Rbpj, a transcription factor that mediates signaling by all four Notch receptors, to overcome these challenges and examined the specific roles of Rbpj in cortical development. We report severe structural abnormalities in the embryonic and postnatal cerebral cortex in Rbpj conditional knockout mice, which provide strong in vivo corroboration of previously reported functions of Notch signaling in neural development. Our results also provide evidence for a novel dual role of Rbpj in cell type-specific regulation of two key developmental events in the cerebral cortex: the maintenance of the undifferentiated state of neural progenitor cells, and the radial and tangential allocation of neurons, possibly through stage-dependent differential regulation of Ngn1.


Subject(s)
Cell Movement , Cerebral Cortex/growth & development , Immunoglobulin J Recombination Signal Sequence-Binding Protein/physiology , Neural Stem Cells/physiology , Neurons/physiology , Animals , Cell Differentiation , Cerebral Cortex/cytology , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Mice, Inbred C57BL , Mice, Knockout , Neural Stem Cells/cytology
6.
Proc Natl Acad Sci U S A ; 114(9): 2367-2372, 2017 02 28.
Article in English | MEDLINE | ID: mdl-28123061

ABSTRACT

Development of prognostic biomarkers for the detection of prenatally damaged neurons before manifestations of postnatal disorders is an essential step for prevention and treatment of susceptible individuals. We have developed a versatile fluorescence reporter system in mice enabling detection of Heat Shock Factor 1 activation in response to prenatal cellular damage caused by exposure to various harmful chemical or physical agents. Using an intrautero electroporation-mediated reporter assay and transgenic reporter mice, we are able to identify neurons that survive prenatal exposure to harmful agents but remain vulnerable in postnatal life. This system may provide a powerful tool for exploring the pathogenesis and treatment of multiple disorders caused by exposure to environmental stress before symptoms become manifested, exacerbated, and/or irreversible.


Subject(s)
Cerebral Cortex/drug effects , Heat Shock Transcription Factors/genetics , Neurons/drug effects , Prenatal Exposure Delayed Effects/diagnosis , Prenatal Exposure Delayed Effects/genetics , Response Elements , Animals , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Electroporation , Embryo, Mammalian , Ethanol/toxicity , Female , Flow Cytometry , Gene Expression Regulation , Genes, Reporter , Heat Shock Transcription Factors/metabolism , Heat-Shock Response/genetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Transgenic , Neurons/metabolism , Neurons/pathology , Nicotine/toxicity , Plasmids/chemistry , Plasmids/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/pathology , Suramin/toxicity
7.
Ann Neurol ; 82(1): 121-127, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28556287

ABSTRACT

To characterize the mechanism of Zika virus (ZIKV)-associated microcephaly, we performed immunolabeling on brain tissue from a 20-week fetus with intrauterine ZIKV infection. Although ZIKV demonstrated a wide range of neuronal and non-neuronal tropism, the infection rate was highest in intermediate progenitor cells and immature neurons. Apoptosis was observed in both infected and uninfected bystander cortical neurons, suggesting a role for paracrine factors in induction of neuronal apoptosis. Our results highlight differential neuronal susceptibility and neuronal apoptosis as potential mechanisms in the development of ZIKV-associated microcephaly, and may provide insights into the design and best timing of future therapy. Ann Neurol 2017;82:121-127.


Subject(s)
Fetus/pathology , Fetus/virology , Neurons/pathology , Neurons/virology , Zika Virus Infection/pathology , Apoptosis , Brain/pathology , Brain/virology , Disease Susceptibility , Humans , Zika Virus Infection/virology
8.
Nature ; 461(7263): 524-8, 2009 Sep 24.
Article in English | MEDLINE | ID: mdl-19759535

ABSTRACT

The cerebral cortex is a laminated sheet of neurons composed of the arrays of intersecting radial columns. During development, excitatory projection neurons originating from the proliferative units at the ventricular surface of the embryonic cerebral vesicles migrate along elongated radial glial fibres to form a cellular infrastructure of radial (vertical) ontogenetic columns in the overlaying cortical plate. However, a subpopulation of these clonally related neurons also undergoes a short lateral shift and transfers from their parental to the neighbouring radial glial fibres, and intermixes with neurons originating from neighbouring proliferative units. This columnar organization acts as the primary information processing unit in the cortex. The molecular mechanisms, role and significance of this lateral dispersion for cortical development are not understood. Here we show that an Eph receptor A (EphA) and ephrin A (Efna) signalling-dependent shift in the allocation of clonally related neurons is essential for the proper assembly of cortical columns. In contrast to the relatively uniform labelling of the developing cortical plate by various molecular markers and retrograde tracers in wild-type mice, we found alternating labelling of columnar compartments in Efna knockout mice that are caused by impaired lateral dispersion of migrating neurons rather than by altered cell production or death. Furthermore, in utero electroporation showed that lateral dispersion depends on the expression levels of EphAs and ephrin-As during neuronal migration. This so far unrecognized mechanism for lateral neuronal dispersion seems to be essential for the proper intermixing of neuronal types in the cortical columns, which, when disrupted, might contribute to neuropsychiatric disorders associated with abnormal columnar organization.


Subject(s)
Cell Movement , Cerebral Cortex/embryology , Ephrins/metabolism , Neurons/cytology , Neurons/metabolism , Receptors, Eph Family/metabolism , Signal Transduction , Animals , Cerebral Cortex/anatomy & histology , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Ephrins/deficiency , Ephrins/genetics , Mice , Mice, Knockout , Neocortex/cytology , Neocortex/metabolism , Organogenesis , Rats , Receptors, Eph Family/deficiency , Receptors, Eph Family/genetics
9.
Proc Natl Acad Sci U S A ; 109(21): 8280-5, 2012 May 22.
Article in English | MEDLINE | ID: mdl-22566616

ABSTRACT

The prospective pyramidal neurons, migrating from the proliferative ventricular zone to the overlaying cortical plate, assume multipolar morphology while passing through the transient subventricular zone. Here, we show that this morphogenetic transformation, from the bipolar to the mutipolar and then back to bipolar again, is associated with expression of connexin 43 (Cx43) and, that knockdown of Cx43 retards, whereas its overexpression enhances, this morphogenetic process. In addition, we have observed that knockdown of Cx43 reduces expression of p27, whereas overexpression of p27 rescues the effect of Cx43 knockdown in the multipolar neurons. Furthermore, functional gap junction/hemichannel domain, and the C-terminal domain of Cx43, independently enhance the expression of p27 and promote the morphological transformation and migration of the multipolar neurons in the SVZ/IZ. Collectively, these results indicate that Cx43 regulates the passage of migrating neurons through their multipolar stage via p27 signaling and that interference with this process, by either genetic and/or environmental factors, may cause cortical malformations.


Subject(s)
Cell Movement/physiology , Cerebral Cortex , Connexin 43/physiology , Cyclin-Dependent Kinase Inhibitor p27/physiology , Pyramidal Cells/cytology , Animals , Calcium/metabolism , Cell Membrane/physiology , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Cerebral Cortex/physiology , Connexin 43/chemistry , Connexin 43/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , Female , Gap Junctions/physiology , Gene Knockdown Techniques , Ion Channels/physiology , Mice , Mice, Inbred Strains , Pregnancy , Protein Structure, Tertiary , Pyramidal Cells/physiology
10.
Cereb Cortex ; 23(4): 775-85, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22490549

ABSTRACT

Auditory stimulus representations are dynamically maintained by ascending and descending projections linking the auditory cortex (Actx), medial geniculate body (MGB), and inferior colliculus. Although the extent and topographic specificity of descending auditory corticofugal projections can equal or surpass that of ascending corticopetal projections, little is known about the molecular mechanisms that guide their development. Here, we used in utero gene electroporation to examine the role of EphA receptor signaling in the development of corticothalamic (CT) and corticocollicular connections. Early in postnatal development, CT axons were restricted to a deep dorsal zone (DDZ) within the MGB that expressed low levels of the ephrin-A ligand. By hearing onset, CT axons had innervated surrounding regions of MGB in control-electroporated mice but remained fixed within the DDZ in mice overexpressing EphA7. In vivo neurophysiological recordings demonstrated a corresponding reduction in spontaneous firing rate, but no changes in sound-evoked responsiveness within MGB regions deprived of CT innervation. Structural and functional CT disruption occurred without gross alterations in thalamocortical connectivity. These data demonstrate a potential role for EphA/ephrin-A signaling in the initial guidance of corticofugal axons and suggest that "genetic rewiring" may represent a useful functional tool to alter cortical feedback without silencing Actx.


Subject(s)
Auditory Cortex , Auditory Pathways/physiology , Brain Mapping , Geniculate Bodies/physiology , Receptor, EphA7/metabolism , Signal Transduction/physiology , Acoustic Stimulation , Age Factors , Amino Acids , Animals , Animals, Newborn , Auditory Cortex/embryology , Auditory Cortex/growth & development , Auditory Cortex/metabolism , Axons/physiology , Electroencephalography , Electroporation , Embryo, Mammalian , Evoked Potentials, Auditory/genetics , Female , Gene Expression Regulation, Developmental/genetics , Green Fluorescent Proteins/genetics , Male , Mice , Mice, Transgenic , RNA, Messenger/metabolism , Receptor, EphA7/genetics , Vesicular Glutamate Transport Protein 1/metabolism
11.
J Neurosci ; 30(12): 4197-209, 2010 Mar 24.
Article in English | MEDLINE | ID: mdl-20335455

ABSTRACT

During mitotic division in the telencephalic proliferative ventricular zone (VZ), the nuclei of the neural precursors move basally away from the ventricular surface for DNA synthesis, and apically return to the surface for mitotic division; a process known as interkinetic migration or "to-and-fro" nuclear translocation. The cell, which remains attached to the ventricular surface, either continues cycling, or exits the cycle and migrates to the subventricular zone or the developing cortical plate. Although gap junctions/hemichannels are known to modulate DNA synthesis via Ca(2+) waves, the role of Ca(+) oscillations and the mechanism of nuclear translocation in the VZ precursors are unclear. Here, we provide evidence that, during apical nuclear migration, VZ precursors display dynamic spontaneous Ca(2+) transients, which depend on functional gap junctions/hemichannels via ATP release and Ca(2+)-mobilizing messenger diffusion. Furthermore, we found that blocking gap junctions/hemichannels or short hairpin RNA-mediated knockdown of Cx43 (connexin 43) retards the apically directed interkinetic nuclear migration accompanied with changes in the nuclear length/width ratio. In addition, we demonstrated that blocking functional gap junctions/hemichannels induces phosphorylation of small GTPase cdc42 in the VZ precursors. The basal phase of interkinetic migration is much slower and appears to be mediated passively by mechanical forces after cell division. Our findings indicate that functional interference with gap junctions/hemichannels during embryonic development may lead to abnormal corticogenesis and dysfunction of the cerebral cortex in adult organisms.


Subject(s)
Cell Movement/physiology , Cell Nucleus/metabolism , Gap Junctions/physiology , Stem Cells/cytology , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Animals , Boron Compounds/pharmacology , Bromodeoxyuridine/metabolism , Calcium/metabolism , Calcium Signaling/drug effects , Carbenoxolone/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Nucleus/drug effects , Cerebral Ventricles/cytology , Chelating Agents/pharmacology , Connexin 43/genetics , Connexin 43/metabolism , Cyclooxygenase Inhibitors/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Embryo, Mammalian , Female , Gap Junctions/drug effects , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Ki-67 Antigen/metabolism , Meclofenamic Acid/pharmacology , Mice , Neuroblastoma , Neurons/drug effects , Neurons/physiology , Organ Culture Techniques , Patch-Clamp Techniques/methods , Platelet Aggregation Inhibitors/pharmacology , Pregnancy , Prosencephalon/cytology , Pyridoxal Phosphate/analogs & derivatives , Pyridoxal Phosphate/pharmacology
12.
J Neurosci ; 30(42): 13992-4007, 2010 Oct 20.
Article in English | MEDLINE | ID: mdl-20962221

ABSTRACT

Endocannabinoids, particularly 2-arachidonoyl glycerol (2-AG), impact the directional turning and motility of a developing axon by activating CB(1) cannabinoid receptors (CB(1)Rs) in its growth cone. Recent findings posit that sn-1-diacylglycerol lipases (DAGLα/ß) synthesize 2-AG in the motile axon segment of developing pyramidal cells. Coincident axonal targeting of CB(1)Rs and DAGLs prompts the hypothesis that autocrine 2-AG signaling facilitates axonal outgrowth. However, DAGLs alone are insufficient to account for the spatial specificity and dynamics of 2-AG signaling. Therefore, we hypothesized that local 2-AG degradation by monoacylglycerol lipase (MGL) must play a role. We determined how subcellular recruitment of MGL is temporally and spatially restricted to establish the signaling competence of 2-AG during axonal growth. MGL is expressed in central and peripheral axons of the fetal nervous system by embryonic day 12.5. MGL coexists with DAGLα and CB(1)Rs in corticofugal axons of pyramidal cells. Here, MGL and DAGLα undergo differential axonal targeting with MGL being excluded from the motile neurite tip. Thus, spatially confined MGL activity generates a 2-AG-sensing microdomain and configures 2-AG signaling to promote axonal growth. Once synaptogenesis commences, MGL disperses in stationary growth cones. The axonal polarity of MGL is maintained by differential proteasomal degradation because inhibiting the ubiquitin proteasome system also induces axonal MGL redistribution. Because MGL inactivation drives a CB(1)R-dependent axonal growth response, we conclude that 2-AG may act as a focal protrusive signal for developing neurons and whose regulated metabolism is critical for attaining correct axonal complexity.


Subject(s)
Arachidonic Acids/physiology , Axons/enzymology , Cannabinoid Receptor Modulators/physiology , Glycerides/physiology , Monoacylglycerol Lipases/metabolism , Signal Transduction/physiology , Subcellular Fractions/enzymology , Animals , Axons/ultrastructure , Blotting, Western , Cells, Cultured , Chromatography, High Pressure Liquid , Endocannabinoids , Glutamate Decarboxylase/genetics , Immunohistochemistry , Lipoprotein Lipase/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Electron , Monoacylglycerol Lipases/genetics , Neural Pathways/cytology , Neural Pathways/physiology , Neurons/enzymology , Neurons/ultrastructure , Pyramidal Cells/enzymology , Pyramidal Cells/metabolism , Receptor, Cannabinoid, CB1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Subcellular Fractions/ultrastructure , Tandem Mass Spectrometry
13.
Proc Natl Acad Sci U S A ; 105(33): 11802-7, 2008 Aug 19.
Article in English | MEDLINE | ID: mdl-18689674

ABSTRACT

Most neurons of the cerebral cortex are generated in the germinal zones near the embryonic cerebral ventricle and migrate radially to the overlying cortical plate. Initially, all dividing cells are attached to the surface of the embryonic ventricle (ventricular zone) until a subset of dividing cells (basal or intermediate neuronal progenitors, INPs), recognized by their immunoreactivity to Tbr2, detach from the ventricular surface and migrate a short distance to establish a secondary proliferative compartment (the subventricular zone). The mechanism that regulates migration of the Tbr2(+) INPs from the ventricular to the subventricular zones is unknown. Here, we show that INPs, unlike the postmitotic neurons that tend to lose the ATP response, continue to express the purinergic P2Y1 receptor. Furthermore, blocking ATP signaling by the P2Y1 blockers, MRS2176, suramin, and apyrase, reduces Ca(2+) transients and retards INP migration to the subventricular zone. In addition, genetic knockdown of the P2Y1 receptor by in vivo application of short hairpin RNA selectively impairs the migration of INPs to the subventricular zone. Together, these results suggest that intercellular ATP signaling is essential for the migration of INPs and the proper formation of the subventricular zone. Interference of ATP signaling or abnormal Ca(2+) fluctuations in INPs may play a significant role in variety of genetic or acquired cortical malformations.


Subject(s)
Adenosine Triphosphate/metabolism , Cerebral Ventricles/cytology , Cerebral Ventricles/metabolism , Neurons/cytology , Neurons/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Calcium/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Gene Expression Regulation, Developmental , Mice , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2/metabolism , Signal Transduction
14.
Nat Neurosci ; 10(5): 588-97, 2007 May.
Article in English | MEDLINE | ID: mdl-17450135

ABSTRACT

Modifying serotonin (5-HT) abundance in the embryonic mouse brain disrupts the precision of sensory maps formed by thalamocortical axons (TCAs), suggesting that 5-HT influences their growth. We investigated the mechanism by which 5-HT influences TCAs during development. 5-HT(1B) and 5-HT(1D) receptor expression in the fetal forebrain overlaps with that of the axon guidance receptors DCC and Unc5c. In coculture assays, axons originating from anterior and posterior halves of the embryonic day 14.5 dorsal thalamus responded differently to netrin-1, reflecting the patterns of DCC and Unc5c expression. 5-HT converts the attraction exerted by netrin-1 on posterior TCAs to repulsion. Pharmacological manipulation of 5-HT(1B/1D) receptors and intracellular cAMP showed the signaling cascade through which this modulation occurs. An in vivo correlate of altered TCA pathfinding was obtained by transient manipulation of 5-HT(1B/1D) receptor expression abundance in the dorsal thalamus by in utero electroporation. These data demonstrate that serotonergic signaling has a previously unrecognized role in the modulation of axonal responsiveness to a classic guidance cue.


Subject(s)
Axons/metabolism , Cerebral Cortex , Gene Expression Regulation, Developmental/physiology , Nerve Growth Factors/metabolism , Serotonin/metabolism , Thalamus , Tumor Suppressor Proteins/metabolism , Afferent Pathways/cytology , Afferent Pathways/embryology , Age Factors , Animals , Cell Line, Transformed , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Electroporation/methods , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental/drug effects , Humans , In Situ Hybridization/methods , In Vitro Techniques , Luminescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Netrin-1 , Pregnancy , RNA, Small Interfering/pharmacology , Receptors, Serotonin/genetics , Receptors, Serotonin/metabolism , Thalamus/cytology , Thalamus/embryology , Thalamus/metabolism
15.
Neuron ; 52(5): 789-801, 2006 Dec 07.
Article in English | MEDLINE | ID: mdl-17145501

ABSTRACT

Periventricular heterotopia (PVH) is a congenital malformation of human cerebral cortex frequently associated with Filamin-A (FLN-A) mutations but the pathogenetic mechanisms remain unclear. Here, we show that the MEKK4 (MAP3K4) pathway is involved in Fln-A regulation and PVH formation. MEKK4(-/-) mice developed PVH associated with breaches in the neuroependymal lining which were largely comprised of neurons that failed to reach the cortical plate. RNA interference (RNAi) targeting MEKK4 also impaired neuronal migration. Expression of Fln was elevated in MEKK4(-/-) forebrain, most notably near sites of failed neuronal migration. Importantly, recombinant MKK4 protein precipitated a complex containing MEKK4 and Fln-A, and MKK4 mediated signaling between MEKK4 and Fln-A, suggesting that MKK4 may bridge these molecules during development. Finally, we showed that wild-type FLN-A overexpression inhibited neuronal migration. Collectively, our results demonstrate a link between MEKK4 and Fln-A that impacts neuronal migration initiation and provides insight into the pathogenesis of human PVH.


Subject(s)
Cell Movement/physiology , Contractile Proteins/biosynthesis , Gene Expression Regulation, Developmental/physiology , MAP Kinase Kinase Kinase 4/physiology , Microfilament Proteins/biosynthesis , Neurons/physiology , Signal Transduction/physiology , Animals , Antibodies, Monoclonal , Antimetabolites , Blotting, Western , Bromodeoxyuridine , Cell Differentiation/physiology , Cell Movement/genetics , Contractile Proteins/genetics , Contractile Proteins/physiology , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Electroporation , Female , Filamins , Gene Expression Regulation, Developmental/genetics , Humans , Immunohistochemistry , MAP Kinase Kinase Kinase 4/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/genetics , Microfilament Proteins/physiology , Neurons/ultrastructure , Paraventricular Hypothalamic Nucleus/cytology , Paraventricular Hypothalamic Nucleus/physiology , Phosphorylation , Pregnancy , Prosencephalon/growth & development , Prosencephalon/metabolism , RNA, Small Interfering/genetics , Stem Cells/physiology
16.
Cereb Cortex ; 19 Suppl 1: i78-89, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19346272

ABSTRACT

It is now well established that inhibitory interneurons of the cerebral cortex display large diversity, but where each subclass originates and how they acquire final position and physiological characteristics is only begin to be elucidated. Recent studies indicate that the phenotypes of many forebrain interneurons are specified in the ganglionic eminence (GE) at the time of their origin. However, developmental history of cannabinoid type 1 receptor (CB(1)) positive (+) interneurons is not known. Here, we focus on the origin and migratory routs of prospective CB(1)/cholecystokinin (CCK)+ and CB(1)/reelin/calretinin+ gamma-aminobutyric acid (GABA)-ergic hippocampal interneurons. We have used variety of markers and a combination of methods, including immunocytochemistry at light and electron microscopic level, and in utero electroporation, to identify a subpopulation of CB(1)+ cells at the time of their origin in the caudal GE and pallial-subpallial boundary at the 11th-12th embryonic days. We have followed their migration, first radially to the marginal zone, then tangentially in the lateral-to-medial direction within the dorsal telencephalon, before they reach their final destination in the hippocampus proper and the dentate gyrus where they differentiate into CB(1)/CCK+ or CB(1)/reelin/calretinin+ GABAergic interneurons. Thus, the specific subclasses of CB(1)+ inhibitory interneurons, similar to the projection neurons, are determined at the time and place of last cell division and follow their own complex migratory pattern to the final positions.


Subject(s)
Brain/embryology , Brain/physiology , Interneurons/physiology , Neural Inhibition/physiology , Neurogenesis/physiology , Receptor, Cannabinoid, CB1/metabolism , Animals , Brain/cytology , Cell Movement/physiology , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Reelin Protein
17.
Cells ; 10(1)2020 12 26.
Article in English | MEDLINE | ID: mdl-33375263

ABSTRACT

Cortical development in humans is a long and ongoing process that continuously modifies the neural circuitry into adolescence. This is well represented by the dynamic maturation of the corpus callosum, the largest white matter tract in the brain. Callosal projection neurons whose long-range axons form the main component of the corpus callosum are evolved relatively recently with a substantial, disproportionate increase in numbers in humans. Though the anatomy of the corpus callosum and cellular processes in its development have been intensively studied by experts in a variety of fields over several decades, the whole picture of its development, in particular, the molecular controls over the development of callosal projections, still has many missing pieces. This review highlights the most recent progress on the understanding of corpus callosum formation with a special emphasis on the novel molecular players in the development of axonal projections in the corpus callosum.


Subject(s)
Axons/metabolism , Corpus Callosum , Neurons/metabolism , Animals , Corpus Callosum/embryology , Corpus Callosum/growth & development , Corpus Callosum/metabolism , Humans , Neurons/cytology , Proteins/metabolism
18.
J Neurodev Disord ; 12(1): 38, 2020 12 16.
Article in English | MEDLINE | ID: mdl-33327938

ABSTRACT

BACKGROUND: Harsh environments surrounding fetuses and children can induce cellular damage in the developing brain, increasing the risk of intellectual disability and other neurodevelopmental disorders such as schizophrenia. However, the mechanisms by which early damage leads to disease manifestation in later life remain largely unknown. Previously, we demonstrated that the activation of heat shock (HS) signaling can be utilized as a unique reporter to label the cells that undergo specific molecular/cellular changes upon exposure to environmental insults throughout the body. Since the activation of HS signaling is an acute and transient event, this approach was not intended for long-term tracing of affected cells after the activation has diminished. In the present study, we generated new reporter transgenic mouse lines as a novel tool to achieve systemic and long-term tracking of affected cells and their progeny. METHODS: The reporter transgenic mouse system was designed so that the activation of HS signaling through HS response element (HSE) drives flippase (FLPo)-flippase recognition target (FRT) recombination-mediated permanent expression of the red fluorescent protein (RFP), tdTomato. With a priority on consistent and efficient assessment of the reporter system, we focused on intraperitoneal (i.p.) injection models of high-dose, short prenatal exposure to alcohol (ethanol) and sodium arsenite (ethanol at 4.0 g/kg/day and sodium arsenite at 5.0 mg/kg/day, at embryonic day (E) 12 and 13). Long-term reporter expression was examined in the brain of reporter mice that were prenatally exposed to these insults. Electrophysiological properties were compared between RFP+ and RFP- cortical neurons in animals prenatally exposed to arsenite. RESULTS: We detected RFP+ neurons and glia in the brains of postnatal mice that had been prenatally exposed to alcohol or sodium arsenite. In animals prenatally exposed to sodium arsenite, we also detected reduced excitability in RFP+ cortical neurons. CONCLUSION: The reporter transgenic mice allowed us to trace the cells that once responded to prenatal environmental stress and the progeny derived from these cells long after the exposure in postnatal animals. Tracing of these cells indicates that the impact of prenatal exposure on neural progenitor cells can lead to functional abnormalities in their progeny cells in the postnatal brain. Further studies using more clinically relevant exposure models are warranted to explore this mechanism.


Subject(s)
Brain , Environment , Neurons , Animals , Brain/embryology , Brain/growth & development , Female , Mice , Mice, Transgenic , Pregnancy , Prenatal Exposure Delayed Effects
19.
Nat Neurosci ; 23(4): 533-543, 2020 04.
Article in English | MEDLINE | ID: mdl-32203497

ABSTRACT

Learning disabilities are hallmarks of congenital conditions caused by prenatal exposure to harmful agents. These include fetal alcohol spectrum disorders (FASDs) with a wide range of cognitive deficiencies, including impaired motor skill development. Although these effects have been well characterized, the molecular effects that bring about these behavioral consequences remain to be determined. We previously found that the acute molecular responses to alcohol in the embryonic brain are stochastic, varying among neural progenitor cells. However, the pathophysiological consequences stemming from these heterogeneous responses remain unknown. Here we show that acute responses to alcohol in progenitor cells altered gene expression in their descendant neurons. Among the altered genes, an increase of the calcium-activated potassium channel Kcnn2 in the motor cortex correlated with motor learning deficits in a mouse model of FASD. Pharmacologic blockade of Kcnn2 improves these learning deficits, suggesting Kcnn2 blockers as a new intervention for learning disabilities in FASD.


Subject(s)
Behavior, Animal/drug effects , Fetal Alcohol Spectrum Disorders/drug therapy , Learning Disabilities/drug therapy , Learning/drug effects , Motor Cortex/drug effects , Scorpion Venoms/pharmacology , Small-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Animals , Cell Shape/drug effects , Dendrites/drug effects , Dendrites/metabolism , Disease Models, Animal , Learning Disabilities/metabolism , Mice , Motor Activity/drug effects , Motor Cortex/metabolism , Neurons/drug effects , Neurons/metabolism , Scorpion Venoms/therapeutic use , Small-Conductance Calcium-Activated Potassium Channels/metabolism
20.
Neuron ; 48(4): 563-75, 2005 Nov 23.
Article in English | MEDLINE | ID: mdl-16301174

ABSTRACT

Molecular mechanisms generating the topographic organization of corticothalamic (CT) circuits, which comprise more than three-quarters of the synaptic inputs onto sensory relay neurons, and their interdependence with thalamocortical (TC) axon development are unknown. Using in utero electroporation-mediated gene transfer, we show that EphA7-mediated signaling on neocortical axons controls the within-nucleus topography of CT projections in the thalamus. Notably, CT axons that mis-express EphA7 do not shift the relative positioning of their pathway within the subcortical telencephalon (ST), indicating that they do not depend upon EphA7/ephrin-A signaling in the ST for establishing this topography. Moreover, mis-expression of cortical EphA7 results in disrupted topography of CT projections, but unchanged inter- and intra-areal topography of TC projections. Our results support a model in which EphA/ephrin-A signaling controls independently the precision with which CT and TC projections develop, yet is essential for establishing their topographic reciprocity.


Subject(s)
Axons/physiology , Cerebral Cortex/physiology , Receptor, EphA7/physiology , Thalamus/physiology , Animals , Axons/metabolism , Brain Mapping , Cerebral Cortex/metabolism , Mice , Mice, Inbred Strains , Neocortex/anatomy & histology , Neocortex/metabolism , Neural Pathways/physiology , Receptor, EphA5/metabolism , Receptor, EphA7/metabolism , Synaptic Transmission/physiology , Telencephalon/physiology , Thalamus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL