Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Hum Mol Genet ; 29(5): 845-858, 2020 03 27.
Article in English | MEDLINE | ID: mdl-31943082

ABSTRACT

SPECC1L mutations have been identified in patients with rare atypical orofacial clefts and with syndromic cleft lip and/or palate (CL/P). These mutations cluster in the second coiled-coil and calponin homology domains of SPECC1L and severely affect the ability of SPECC1L to associate with microtubules. We previously showed that gene-trap knockout of Specc1l in mouse results in early embryonic lethality. We now present a truncation mutant mouse allele, Specc1lΔC510, that results in perinatal lethality. Specc1lΔC510/ΔC510 homozygotes showed abnormal palate rugae but did not show cleft palate. However, when crossed with a gene-trap allele, Specc1lcGT/ΔC510 compound heterozygotes showed a palate elevation delay with incompletely penetrant cleft palate. Specc1lcGT/ΔC510 embryos exhibit transient oral epithelial adhesions at E13.5, which may delay shelf elevation. Consistent with oral adhesions, we show periderm layer abnormalities, including ectopic apical expression of adherens junction markers, similar to Irf6 hypomorphic mutants and Arhgap29 heterozygotes. Indeed, SPECC1L expression is drastically reduced in Irf6 mutant palatal shelves. Finally, we wanted to determine if SPECC1L deficiency also contributed to non-syndromic (ns) CL/P. We sequenced 62 Caucasian, 89 Filipino, 90 Ethiopian, 90 Nigerian and 95 Japanese patients with nsCL/P and identified three rare coding variants (p.Ala86Thr, p.Met91Iso and p.Arg546Gln) in six individuals. These variants reside outside of SPECC1L coiled-coil domains and result in milder functional defects than variants associated with syndromic clefting. Together, our data indicate that palate elevation is sensitive to deficiency of SPECC1L dosage and function and that SPECC1L cytoskeletal protein functions downstream of IRF6 in palatogenesis.


Subject(s)
Cleft Palate/pathology , Interferon Regulatory Factors/metabolism , Mutation , Phosphoproteins/physiology , Animals , Cleft Palate/genetics , Cleft Palate/metabolism , Female , Humans , Interferon Regulatory Factors/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphoproteins/genetics , Phosphoproteins/metabolism
2.
Nature ; 488(7411): 343-8, 2012 Aug 16.
Article in English | MEDLINE | ID: mdl-22878717

ABSTRACT

Brain circuits process information through specialized neuronal subclasses interacting within a network. Revealing their interplay requires activating specific cells while monitoring others in a functioning circuit. Here we use a new platform for two-way light-based circuit interrogation in visual cortex in vivo to show the computational implications of modulating different subclasses of inhibitory neurons during sensory processing. We find that soma-targeting, parvalbumin-expressing (PV) neurons principally divide responses but preserve stimulus selectivity, whereas dendrite-targeting, somatostatin-expressing (SOM) neurons principally subtract from excitatory responses and sharpen selectivity. Visualized in vivo cell-attached recordings show that division by PV neurons alters response gain, whereas subtraction by SOM neurons shifts response levels. Finally, stimulating identified neurons while scanning many target cells reveals that single PV and SOM neurons functionally impact only specific subsets of neurons in their projection fields. These findings provide direct evidence that inhibitory neuronal subclasses have distinct and complementary roles in cortical computations.


Subject(s)
Neural Inhibition/physiology , Neural Pathways/physiology , Neurons/physiology , Visual Cortex/cytology , Visual Cortex/physiology , Animals , Dendrites/metabolism , Electrophysiology , Interneurons/physiology , Mice , Models, Neurological , Parvalbumins/metabolism , Somatostatin/metabolism
3.
J Med Genet ; 52(2): 104-10, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25412741

ABSTRACT

BACKGROUND: Opitz G/BBB syndrome is a heterogeneous disorder characterised by variable expression of midline defects including cleft lip and palate, hypertelorism, laryngealtracheoesophageal anomalies, congenital heart defects, and hypospadias. The X-linked form of the condition has been associated with mutations in the MID1 gene on Xp22. The autosomal dominant form has been linked to chromosome 22q11.2, although the causative gene has yet to be elucidated. METHODS AND RESULTS: In this study, we performed whole exome sequencing on DNA samples from a three-generation family with characteristics of Opitz G/BBB syndrome with negative MID1 sequencing. We identified a heterozygous missense mutation c.1189A>C (p.Thr397Pro) in SPECC1L, located at chromosome 22q11.23. Mutation screening of an additional 19 patients with features of autosomal dominant Opitz G/BBB syndrome identified a c.3247G>A (p.Gly1083Ser) mutation segregating with the phenotype in another three-generation family. CONCLUSIONS: Previously, SPECC1L was shown to be required for proper facial morphogenesis with disruptions identified in two patients with oblique facial clefts. Collectively, these data demonstrate that SPECC1L mutations can cause syndromic forms of facial clefting including some cases of autosomal dominant Opitz G/BBB syndrome and support the original linkage to chromosome 22q11.2.


Subject(s)
Calcium-Binding Proteins/chemistry , Esophagus/abnormalities , Genes, Dominant , Genetic Predisposition to Disease , Hypertelorism/genetics , Hypospadias/genetics , Microfilament Proteins/chemistry , Mutation/genetics , Phosphoproteins/chemistry , Phosphoproteins/genetics , Adult , Base Sequence , DNA Mutational Analysis , Exons/genetics , Family , Female , Genetic Testing , Humans , Infant , Male , Microtubule Proteins/genetics , Molecular Sequence Data , Nuclear Proteins/genetics , Pedigree , Phenotype , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Transcription Factors/genetics , Ubiquitin-Protein Ligases , Calponins
4.
Blood ; 122(20): 3440-9, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24085763

ABSTRACT

We recently identified 2 siblings afflicted with idiopathic, autosomal recessive aplastic anemia. Whole-exome sequencing identified a novel homozygous missense mutation in thrombopoietin (THPO, c.112C>T) in both affected siblings. This mutation encodes an arginine to cysteine substitution at residue 38 or residue 17 excluding the 21-amino acid signal peptide of THPO receptor binding domain (RBD). THPO has 4 conserved cysteines in its RBD that form 2 disulfide bonds. Our in silico modeling predicts that introduction of a fifth cysteine may disrupt normal disulfide bonding to cause poor receptor binding. In functional assays, the mutant-THPO-containing media shows two- to threefold reduced ability to sustain UT7-TPO cells, which require THPO for proliferation. Both parents and a sibling with heterozygous R17C change have reduced platelet counts, whereas a sibling with wild-type sequence has normal platelet count. Thus, the R17C partial loss-of-function allele results in aplastic anemia in the homozygous state and mild thrombocytopenia in the heterozygous state in our family. Together with the recent identification of THPO receptor (MPL) mutations and the effects of THPO agonists in aplastic anemia, our results have clinical implications in the diagnosis and treatment of patients with aplastic anemia and highlight a role for the THPO-MPL pathway in hematopoiesis in vivo.


Subject(s)
Anemia, Aplastic/genetics , Exome/genetics , Thrombopoietin/genetics , Adolescent , Adult , Amino Acid Substitution , Anemia, Aplastic/drug therapy , Base Sequence , Cells, Cultured , Child , Cloning, Molecular , Comparative Genomic Hybridization , Cystine/chemistry , Exons/genetics , Female , Genes, Recessive , Genotype , Humans , Male , Micronesia , Middle Aged , Models, Molecular , Molecular Sequence Data , Molecular Targeted Therapy , Mutation, Missense , Pedigree , Protein Binding , Protein Conformation , Receptors, Thrombopoietin/metabolism , Sequence Alignment , Sequence Homology, Nucleic Acid , Structure-Activity Relationship , Thrombopoietin/chemistry , Thrombopoietin/metabolism , Young Adult
6.
Proc Natl Acad Sci U S A ; 106(6): 2029-34, 2009 Feb 10.
Article in English | MEDLINE | ID: mdl-19208815

ABSTRACT

Rett Syndrome (RTT) is a severe form of X-linked mental retardation caused by mutations in the gene coding for methyl CpG-binding protein 2 (MECP2). Mice deficient in MeCP2 have a range of physiological and neurological abnormalities that mimic the human syndrome. Here we show that systemic treatment of MeCP2 mutant mice with an active peptide fragment of Insulin-like Growth Factor 1 (IGF-1) extends the life span of the mice, improves locomotor function, ameliorates breathing patterns, and reduces irregularity in heart rate. In addition, treatment with IGF-1 peptide increases brain weight of the mutant mice. Multiple measurements support the hypothesis that RTT results from a deficit in synaptic maturation in the brain: MeCP2 mutant mice have sparse dendritic spines and reduced PSD-95 in motor cortex pyramidal neurons, reduced synaptic amplitude in the same neurons, and protracted cortical plasticity in vivo. Treatment with IGF-1 peptide partially restores spine density and synaptic amplitude, increases PSD-95, and stabilizes cortical plasticity to wild-type levels. Our results thus strongly suggest IGF-1 as a candidate for pharmacological treatment of RTT and potentially of other CNS disorders caused by delayed synapse maturation.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , Methyl-CpG-Binding Protein 2/genetics , Rett Syndrome/drug therapy , Action Potentials , Animals , Brain , Disease Models, Animal , Heart Rate , Insulin-Like Growth Factor I/therapeutic use , Mice , Mice, Mutant Strains , Motor Activity , Neurons , Organ Size , Survival Rate , Synaptic Transmission , Treatment Outcome
7.
Front Neural Circuits ; 15: 792228, 2021.
Article in English | MEDLINE | ID: mdl-35069127

ABSTRACT

Here we demonstrate a facile method by which to deliver complex spatiotemporal stimulation to neural networks in fast patterns, to trigger interesting forms of circuit-level plasticity in cortical areas. We present a complete platform by which patterns of electricity can be arbitrarily defined and distributed across a brain circuit, either simultaneously, asynchronously, or in complex patterns that can be easily designed and orchestrated with precise timing. Interfacing with acute slices of mouse cortex, we show that our system can be used to activate neurons at many locations and drive synaptic transmission in distributed patterns, and that this elicits new forms of plasticity that may not be observable via traditional methods, including interesting measurements of associational and sequence plasticity. Finally, we introduce an automated "network assay" for imaging activation and plasticity across a circuit. Spatiotemporal stimulation opens the door for high-throughput explorations of plasticity at the circuit level, and may provide a basis for new types of adaptive neural prosthetics.


Subject(s)
Neurons , Synaptic Transmission , Animals , Brain , Mice , Neural Networks, Computer , Neuronal Plasticity
8.
PLoS One ; 16(2): e0246989, 2021.
Article in English | MEDLINE | ID: mdl-33577554

ABSTRACT

Topiramate is an anti-epileptic drug that is commonly prescribed not just to prevent seizures but also migraine headaches, with over 8 million prescriptions dispensed annually. Topiramate use during pregnancy has been linked to significantly increased risk of babies born with orofacial clefts (OFCs). However, the exact molecular mechanism of topiramate teratogenicity is unknown. In this study, we first used an unbiased antibody array analysis to test the effect of topiramate on human embryonic palatal mesenchyme (HEPM) cells. This analysis identified 40 differentially expressed proteins, showing strong connectivity to known genes associated with orofacial clefts. However, among known OFC genes, only TGFß1 was significantly upregulated in the antibody array analysis. Next, we validated that topiramate could increase expression of TGFß1 and of downstream target phospho-SMAD2 in primary mouse embryonic palatal mesenchyme (MEPM) cells. Furthermore, we showed that topiramate treatment of primary MEPM cells increased expression of SOX9. SOX9 overexpression in chondrocytes is known to cause cleft palate in mouse. We propose that topiramate mediates upregulation of TGFß1 signaling through activation of γ-aminobutyric acid (GABA) receptors in the palate. TGFß1 and SOX9 play critical roles in orofacial morphogenesis, and their abnormal overexpression provides a plausible etiologic molecular mechanism for the teratogenic effects of topiramate.


Subject(s)
Anticonvulsants/pharmacology , Palate/embryology , SOX9 Transcription Factor/genetics , Teratogens/pharmacology , Topiramate/pharmacology , Transforming Growth Factor beta1/genetics , Animals , Cell Line , Cells, Cultured , Cleft Lip/chemically induced , Cleft Lip/genetics , Cleft Palate/chemically induced , Cleft Palate/genetics , Gene Expression Regulation, Developmental/drug effects , Humans , Mice , Palate/cytology , Palate/drug effects , Palate/metabolism , Up-Regulation/drug effects
9.
Sci Rep ; 11(1): 1452, 2021 01 14.
Article in English | MEDLINE | ID: mdl-33446878

ABSTRACT

Cleft lip and/or palate (CL/P) are common anomalies occurring in 1/800 live-births. Pathogenic SPECC1L variants have been identified in patients with CL/P, which signifies a primary role for SPECC1L in craniofacial development. Specc1l mutant mouse embryos exhibit delayed palatal shelf elevation accompanied by epithelial defects. We now posit that the process of palate elevation is itself abnormal in Specc1l mutants, due to defective remodeling of palatal mesenchyme. To characterize the underlying cellular defect, we studied the movement of primary mouse embryonic palatal mesenchyme (MEPM) cells using live-imaging of wound-repair assays. SPECC1L-deficient MEPM cells exhibited delayed wound-repair, however, reduced cell speed only partially accounted for this delay. Interestingly, mutant MEPM cells were also defective in coordinated cell movement. Therefore, we used open-field 2D cultures of wildtype MEPM cells to show that they indeed formed cell streams at high density, which is an important attribute of collective movement. Furthermore, activation of the PI3K-AKT pathway rescued both cell speed and guidance defects in Specc1l mutant MEPM cells. Thus, we show that live-imaging of primary MEPM cells can be used to assess mesenchymal remodeling defects during palatal shelf elevation, and identify a novel role for SPECC1L in collective movement through modulation of PI3K-AKT signaling.


Subject(s)
Cleft Lip/embryology , Cleft Palate/embryology , Embryo, Mammalian/embryology , Gene Expression Regulation, Developmental , Palate/embryology , Phosphoproteins/deficiency , Animals , Cleft Lip/genetics , Cleft Palate/genetics , Mice , Mice, Knockout , Phosphoproteins/metabolism
10.
J Neurosci ; 27(50): 13581-9, 2007 Dec 12.
Article in English | MEDLINE | ID: mdl-18077670

ABSTRACT

Neurons in plastic regions of the brain undergo fundamental changes in the number of cells connecting to them as a result of development, plasticity and disease. Across these same time periods, functional changes in cellular and synaptic physiology are known to occur and are often characterized as developmental features of these periods. However, it remains possible that many such changes are direct consequences of the modified degree of partnering, and that neurons intrinsically scale their physiological parameters with network size. To systematically vary a recurrent network's number of neurons while measuring its synaptic properties, we used microfabricated extracellular matrix adhesive islands created with soft lithography to culture neuronal clusters of precise sizes, and assessed their intrinsic connectivity using intracellular recordings and confocal microscopy. Both large and small clusters supported constant densities of excitatory and inhibitory neurons. However, neurons that were provided with more potential partners (larger clusters) formed more connections per cell via an expanded dendritic surface than cocultured smaller clusters. Electrophysiologically, firing rate was preserved across clusters even as size and synapse number increased, due in part to synapses in larger networks having reduced unitary strengths, and sparser paired connectivity. Larger networks also featured a particular increase in the number of excitatory connections onto inhibitory dendrites. We suggest that these specific homeostatic mechanisms, which match the number, strength, and architecture of connections to the number of total available cellular partners in the network, could account for several known phenomena implicated in the formation, organization and degeneration of neuronal circuits.


Subject(s)
Nerve Net/physiology , Neurons/physiology , Synapses/physiology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Culture Techniques , Cells, Cultured , Dendrites/physiology , Dendrites/ultrastructure , Excitatory Postsynaptic Potentials , Hippocampus/cytology , Nerve Net/growth & development , Neural Inhibition/physiology , Neurons/ultrastructure , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Synaptic Transmission/physiology , gamma-Aminobutyric Acid/metabolism
11.
J Neurosci ; 25(26): 6221-34, 2005 Jun 29.
Article in English | MEDLINE | ID: mdl-15987952

ABSTRACT

A fundamental question in synaptic physiology is whether the unitary strength of a synapse can be regulated by presynaptic characteristics and, if so, what those characteristics might be. Here, we characterize a newly proposed mechanism for altering the strength of glutamatergic synapses based on the recently identified vesicular glutamate transporter VGLUT1. We provide direct evidence that filling in isolated synaptic vesicles is subject to a dynamic equilibrium that is determined by both the concentration of available glutamate and the number of vesicular transporters participating in loading. We observe that changing the number of vesicular transporters expressed at hippocampal excitatory synapses results in enhanced evoked and miniature responses and verify biophysically that these changes correspond to an increase in the amount of glutamate released per vesicle into the synaptic cleft. In addition, we find that this modulation of synaptic strength by vesicular transporter expression is endogenously regulated, both across development to coincide with a maturational increase in vesicle cycling and quantal amplitude and by excitatory and inhibitory receptor activation in mature neurons to provide an activity-dependent scaling of quantal size via a presynaptic mechanism. Together, these findings underscore that vesicular transporter expression is used endogenously to directly regulate the extent of glutamate release, providing a concise presynaptic mechanism for controlling the quantal efficacy of excitatory transmission during synaptic refinement and plasticity.


Subject(s)
Presynaptic Terminals/physiology , Synapses/physiology , Synaptic Transmission/physiology , Synaptic Vesicles/physiology , Vesicular Glutamate Transport Protein 1/physiology , Animals , Base Sequence , Cloning, Molecular , DNA Primers , Evoked Potentials/physiology , Glutamic Acid/metabolism , Homeostasis , Image Processing, Computer-Assisted , PC12 Cells , Patch-Clamp Techniques , Quantum Theory , Rats , Vesicular Glutamate Transport Protein 1/genetics
12.
Sci Rep ; 6: 17735, 2016 Jan 20.
Article in English | MEDLINE | ID: mdl-26787558

ABSTRACT

Cranial neural crest cells (CNCCs) delaminate from embryonic neural folds and migrate to pharyngeal arches, which give rise to most mid-facial structures. CNCC dysfunction plays a prominent role in the etiology of orofacial clefts, a frequent birth malformation. Heterozygous mutations in SPECC1L have been identified in patients with atypical and syndromic clefts. Here, we report that in SPECC1L-knockdown cultured cells, staining of canonical adherens junction (AJ) components, ß-catenin and E-cadherin, was increased, and electron micrographs revealed an apico-basal diffusion of AJs. To understand the role of SPECC1L in craniofacial morphogenesis, we generated a mouse model of Specc1l deficiency. Homozygous mutants were embryonic lethal and showed impaired neural tube closure and CNCC delamination. Staining of AJ proteins was increased in the mutant neural folds. This AJ defect is consistent with impaired CNCC delamination, which requires AJ dissolution. Further, PI3K-AKT signaling was reduced and apoptosis was increased in Specc1l mutants. In vitro, moderate inhibition of PI3K-AKT signaling in wildtype cells was sufficient to cause AJ alterations. Importantly, AJ changes induced by SPECC1L-knockdown were rescued by activating the PI3K-AKT pathway. Together, these data indicate SPECC1L as a novel modulator of PI3K-AKT signaling and AJ biology, required for neural tube closure and CNCC delamination.


Subject(s)
Adherens Junctions/metabolism , Neural Crest/embryology , Neural Crest/metabolism , Phosphoproteins/deficiency , Animals , Apoptosis/genetics , Biomarkers , Cell Adhesion Molecules/metabolism , Cell Lineage/genetics , Gene Expression , Gene Knockout Techniques , Humans , Mice , Models, Biological , Mutation , Neural Tube Defects/genetics , Neural Tube Defects/pathology , Phenotype , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
13.
Nat Protoc ; 8(6): 1184-203, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23702834

ABSTRACT

Neuronal networks process information in a distributed, spatially heterogeneous manner that transcends the layout of electrodes. In contrast, directed and steerable light offers the potential to engage specific cells on demand. We present a unified framework for adapting microscopes to use light for simultaneous in vivo stimulation and recording of cells at fine spatiotemporal resolutions. We use straightforward optics to lock onto networks in vivo, to steer light to activate circuit elements and to simultaneously record from other cells. We then actualize this 'free' augmentation on both an 'open' two-photon microscope and a leading commercial one. By following this protocol, setup of the system takes a few days, and the result is a noninvasive interface to brain dynamics based on directed light, at a network resolution that was not previously possible and which will further improve with the rapid advance in development of optical reporters and effectors. This protocol is for physiologists who are competent with computers and wish to extend hardware and software to interface more fluidly with neuronal networks.


Subject(s)
Light , Nerve Net/physiology , Neurons/radiation effects , Visual Cortex/physiology , Animals , Channelrhodopsins , Mice , Microscopy/methods , Neurons/physiology , Photic Stimulation
14.
Neuron ; 67(5): 847-57, 2010 Sep 09.
Article in English | MEDLINE | ID: mdl-20826315

ABSTRACT

Inhibitory interneurons in the cerebral cortex include a vast array of subtypes, varying in their molecular signatures, electrophysiological properties, and connectivity patterns. This diversity suggests that individual inhibitory classes have unique roles in cortical circuits; however, their characterization to date has been limited to broad classifications including many subtypes. We used the Cre/LoxP system, specifically labeling parvalbumin(PV)-expressing interneurons in visual cortex of PV-Cre mice with red fluorescent protein (RFP), followed by targeted loose-patch recordings and two-photon imaging of calcium responses in vivo to characterize the visual receptive field properties of these cells. Despite their relative molecular and morphological homogeneity, we find that PV+ neurons have a diversity of feature-specific visual responses that include sharp orientation and direction-selectivity, small receptive fields, and band-pass spatial frequency tuning. These results suggest that subsets of parvalbumin interneurons are components of specific cortical networks and that perisomatic inhibition contributes to the generation of precise response properties.


Subject(s)
Interneurons/classification , Interneurons/physiology , Neural Inhibition/physiology , Parvalbumins/metabolism , Visual Cortex/cytology , Animals , Biophysics , Calcium/metabolism , Electric Stimulation/methods , Glutamate Decarboxylase/genetics , Green Fluorescent Proteins/genetics , Luminescent Proteins/genetics , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Patch-Clamp Techniques/methods , Photic Stimulation/methods , Photons , Visual Pathways/physiology , gamma-Aminobutyric Acid/metabolism , Red Fluorescent Protein
SELECTION OF CITATIONS
SEARCH DETAIL