Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Epilepsia ; 64(9): 2515-2527, 2023 09.
Article in English | MEDLINE | ID: mdl-37329181

ABSTRACT

OBJECTIVE: Duplication of the maternal chromosome 15q11.2-q13.1 region causes Dup15q syndrome, a highly penetrant neurodevelopmental disorder characterized by severe autism and refractory seizures. Although UBE3A, the gene encoding the ubiquitin ligase E3A, is thought to be the main driver of disease phenotypes, the cellular and molecular mechanisms that contribute to the development of the syndrome are yet to be determined. We previously established the necessity of UBE3A overexpression for the development of cellular phenotypes in human Dup15q neurons, including increased action potential firing and increased inward current density, which prompted us to further investigate sodium channel kinetics. METHODS: We used a Dup15q patient-derived induced pluripotent stem cell line that was CRISPR-edited to remove the supernumerary chromosome and create an isogenic control line. We performed whole cell patch clamp electrophysiology on Dup15q and corrected control neurons at two time points of in vitro development. RESULTS: Compared to corrected neurons, Dup15q neurons showed increased sodium current density and a depolarizing shift in steady-state inactivation. Moreover, onset of slow inactivation was delayed, and a faster recovery from both fast and slow inactivation processes was observed in Dup15q neurons. A fraction of sodium current in Dup15q neurons (~15%) appeared to be resistant to slow inactivation. Not unexpectedly, a higher fraction of persistent sodium current was also observed in Dup15q neurons. These phenotypes were modulated by the anticonvulsant drug rufinamide. SIGNIFICANCE: Sodium channels play a crucial role in the generation of action potentials, and sodium channelopathies have been uncovered in multiple forms of epilepsy. For the first time, our work identifies in Dup15q neurons dysfunctional inactivation kinetics, which have been previously linked to multiple forms of epilepsy. Our work can also guide therapeutic approaches to epileptic seizures in Dup15q patients and emphasize the role of drugs that modulate inactivation kinetics, such as rufinamide.


Subject(s)
Epilepsy , Humans , Kinetics , Epilepsy/genetics , Sodium Channels , Syndrome , Sodium
2.
Stem Cell Reports ; 18(4): 884-898, 2023 04 11.
Article in English | MEDLINE | ID: mdl-36898382

ABSTRACT

Chromosome 15q11-q13 duplication syndrome (Dup15q) is a neurodevelopmental disorder caused by maternal duplications of this region. Autism and epilepsy are key features of Dup15q. UBE3A, which encodes an E3 ubiquitin ligase, is likely a major driver of Dup15q because UBE3A is the only imprinted gene expressed solely from the maternal allele. Nevertheless, the exact role of UBE3A has not been determined. To establish whether UBE3A overexpression is required for Dup15q neuronal deficits, we generated an isogenic control line for a Dup15q patient-derived induced pluripotent stem cell line. Dup15q neurons exhibited hyperexcitability compared with control neurons, and this phenotype was generally prevented by normalizing UBE3A levels using antisense oligonucleotides. Overexpression of UBE3A resulted in a profile similar to that of Dup15q neurons except for synaptic phenotypes. These results indicate that UBE3A overexpression is necessary for most Dup15q cellular phenotypes but also suggest a role for other genes in the duplicated region.


Subject(s)
Autistic Disorder , Chromosome Aberrations , Chromosomes, Human, Pair 15 , Ubiquitin-Protein Ligases , Humans , Autistic Disorder/genetics , Autistic Disorder/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats , Intellectual Disability/genetics , Intellectual Disability/metabolism , Neurons/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
3.
Biogerontology ; 24(2): 163-181, 2023 04.
Article in English | MEDLINE | ID: mdl-36626035

ABSTRACT

Volume hyposensitivity resulting from impaired sympathetic detrusor relaxation during bladder filling contributes to detrusor underactivity (DU) associated with aging. Detrusor tension regulation provides an adaptive sensory input of bladder volume to the brainstem and is challenged by physiological stressors superimposed upon biological aging. We recently showed that HCN channels have a stabilizing role in detrusor sympathetic relaxation. While mature mice maintain homeostasis in the face of stressors, old mice are not always capable. In old mice, there is a dichotomous phenotype, in which resilient mice adapt and maintain homeostasis, while non-resilient mice fail to maintain physiologic homeostasis. In this DU model, we used cystometry as a stressor to categorize mice as old-responders (old-R, develop a filling/voiding cycle) or old-non-responders (old-NR, fail to develop a filling/voiding cycle; fluctuating high pressures and continuous leaking), while also assessing functional and molecular differences. Lamotrigine (HCN activator)-induced bladder relaxation is diminished in old-NR mice following HCN-blockade. Relaxation responses to NS 1619 were reduced in old-NR mice, with the effect lost following HCN-blockade. However, RNA-sequencing revealed no differences in HCN gene expression and electrophysiology studies showed similar percentage of detrusor myocytes expressing HCN (Ih) current between old-R and old-NR mice. Our murine model of DU further defines a role for HCN, with failure of adaptive recalibration of HCN participation and intensity of HCN-mediated stabilization, while genomic studies show upregulated myofibroblast and fibrosis pathways and downregulated neurotransmitter-degradation pathways in old-NR mice. Thus, the DU phenotype is multifactorial and represents the accumulation of age-associated loss in homeostatic mechanisms.


Subject(s)
Urinary Bladder, Underactive , Mice , Animals , Urinary Bladder , Aging/physiology
4.
Front Cell Neurosci ; 16: 1023541, 2022.
Article in English | MEDLINE | ID: mdl-36212685

ABSTRACT

It is widely accepted that exogenous cannabinoids can impair short-term memory and cognition in humans and other animals. This is likely related to the inhibition of long-term potentiation (LTP), a form of synaptic plasticity, by the global and sustained activation of CB1 cannabinoid receptors in the presence of exogenous agonists. Conversely, the temporally and spatially restricted release of endogenous cannabinoid (eCB) ligands may enhance synaptic plasticity in a synapse-specific manner. We examined the role of eCB signaling in LTP by recording field excitatory postsynaptic potentials (fEPSPs) in the CA1 stratum radiatum in hippocampal slices from juvenile mice. LTP was induced either electrically, by theta burst stimulation (TBS), or pharmacologically, by treatment for 15 min with a solution designed to increase intracellular cAMP (chem-LTP). A stable and long-lasting potentiation in fEPSP slope following TBS was significantly reduced by blocking cannabinoid receptor activation with CB1 receptor antagonists. Chem-LTP caused a sustained 2-fold increase in fEPSP slope and was also blocked by CB1 receptor antagonists. TBS-LTP was partially reduced by inhibiting the synthesis of the endogenous ligands 2-arachidonylglycerol (2-AG) and anandamide. A similar effect was observed with chem-LTP. Blocking inhibitory synapses completely prevented the effect of CB1 receptor antagonists or inhibition of eCB synthesis on TBS-LTP and chem-LTP. These results indicate that simultaneous activation of CB1 receptors by 2-AG and anandamide enhances TBS-induced and pharmacologically-induced LTP, and this effect is mediated by the suppression of inhibition at GABAergic synapses.

5.
Am J Physiol Regul Integr Comp Physiol ; 323(1): R110-R122, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35503519

ABSTRACT

Control of urinary continence is predicated on sensory signaling about bladder volume. Bladder sensory nerve activity is dependent on tension, implicating autonomic control over detrusor myocyte activity during bladder filling. Hyperpolarization-activated cyclic nucleotide-gated (HCN) ion channels are known contributors to bladder control, but their mechanism of action is not well understood. The lack of a definitive identification of cell type(s) expressing HCN in the bladder presents a significant knowledge gap. We recently reported a complete transcriptomic atlas of the C57BL/6 mouse bladder showing the dominant HCN paralog in mouse bladder, Hcn1, is limited to a subpopulation of detrusor smooth myocytes (DSMs). Here, we report details of these findings, along with results of patch-clamp experiments, immunohistochemistry, and functional myobath/tension experiments in bladder strips. With the use of a transgenic mouse expressing fluorescence-tagged α-smooth muscle actin, our data confirmed location and function of DSM HCN channels. Despite previous associations of HCN with postulated bladder interstitial cells, neither evidence of specific interstitial cell types nor an association of nonmyocytes with HCN was discovered. We confirm that HCN activation participates in reducing sustained (tonic) detrusor tension via cAMP, with no effect on intermittent (phasic) detrusor activity. In contrast, blockade of HCN increases phasic activity induced by a protein kinase A (PKA) blocker or a large-conductance Ca2+-activated K+ (BK) channel opener. Our findings, therefore, suggest a central role for detrusor myocyte HCN in regulating and constraining detrusor myocyte activity during bladder filling.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels , Interstitial Cells of Cajal , Adrenergic Agents , Animals , Cyclic Nucleotide-Gated Cation Channels/genetics , Cyclic Nucleotide-Gated Cation Channels/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Interstitial Cells of Cajal/metabolism , Mice , Mice, Inbred C57BL , Nucleotides, Cyclic/metabolism
6.
Biol Psychiatry ; 90(11): 756-765, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34538422

ABSTRACT

BACKGROUND: Chromosome 15q11-q13 duplication syndrome (Dup15q) is a neurogenetic disorder caused by duplications of the maternal copy of this region. In addition to hypotonia, motor deficits, and language impairments, patients with Dup15q commonly meet the criteria for autism spectrum disorder and have a high prevalence of seizures. It is known from mouse models that synaptic impairments are a strong component of Dup15q pathophysiology; however, cellular phenotypes that relate to seizures are less clear. The development of patient-derived induced pluripotent stem cells provides a unique opportunity to study human neurons with the exact genetic disruptions that cause Dup15q. METHODS: Here, we explored electrophysiological phenotypes in induced pluripotent stem cell-derived neurons from 4 patients with Dup15q compared with 6 unaffected control subjects, 1 patient with a 15q11-q13 paternal duplication, and 3 patients with Angelman syndrome. RESULTS: We identified several properties of Dup15q neurons that could contribute to neuronal hyperexcitability and seizure susceptibility. Compared with control neurons, Dup15q neurons had increased excitatory synaptic event frequency and amplitude, increased density of dendritic protrusions, increased action potential firing, and decreased inhibitory synaptic transmission. Dup15q neurons also showed impairments in activity-dependent synaptic plasticity and homeostatic synaptic scaling. Finally, Dup15q neurons showed an increased frequency of spontaneous action potential firing compared with control neurons, in part due to disruption of KCNQ2 potassium channels. CONCLUSIONS: Together, these data point to multiple electrophysiological mechanisms of hyperexcitability that may provide new targets for the treatment of seizures and other phenotypes associated with Dup15q.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Induced Pluripotent Stem Cells , Animals , Autism Spectrum Disorder/genetics , Humans , Mice , Neurons , Phenotype
7.
Hum Mol Genet ; 29(18): 3021-3031, 2020 11 04.
Article in English | MEDLINE | ID: mdl-32833011

ABSTRACT

Loss of UBE3A expression, a gene regulated by genomic imprinting, causes Angelman syndrome (AS), a rare neurodevelopmental disorder. The UBE3A gene encodes an E3 ubiquitin ligase with three known protein isoforms in humans. Studies in mouse suggest that the human isoforms may have differences in localization and neuronal function. A recent case study reported mild AS phenotypes in individuals lacking one specific isoform. Here we have used CRISPR/Cas9 to generate isogenic human embryonic stem cells (hESCs) that lack the individual protein isoforms. We demonstrate that isoform 1 accounts for the majority of UBE3A protein in hESCs and neurons. We also show that UBE3A predominantly localizes to the cytoplasm in both wild type and isoform-null cells. Finally, we show that neurons lacking isoform 1 display a less severe electrophysiological AS phenotype.


Subject(s)
Angelman Syndrome/genetics , Genetic Predisposition to Disease , Ubiquitin-Protein Ligases/genetics , Angelman Syndrome/pathology , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Electrophysiological Phenomena/genetics , Genomic Imprinting/genetics , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/pathology , Humans , Mice , Neurons/metabolism , Neurons/pathology , Protein Isoforms/genetics
8.
Adv Neurobiol ; 25: 55-77, 2020.
Article in English | MEDLINE | ID: mdl-32578144

ABSTRACT

The chromosome 15q11-q13 region of the human genome is regulated by genomic imprinting, an epigenetic phenomenon in which genes are expressed exclusively from one parental allele. Several genes within the 15q11-q13 region are expressed exclusively from the paternally inherited chromosome 15. At least one gene UBE3A, shows exclusive expression of the maternal allele, but this allele-specific expression is restricted to neurons. The appropriate regulation of imprinted gene expression across chromosome 15q11-q13 has important implications for human disease. Three different neurodevelopmental disorders result from aberrant expression of imprinted genes in this region: Prader-Willi syndrome (PWS), Angelman syndrome (AS), and 15q duplication syndrome.


Subject(s)
Angelman Syndrome , Prader-Willi Syndrome , Angelman Syndrome/genetics , Chromosomes , Genomic Imprinting/genetics , Humans , Prader-Willi Syndrome/genetics
9.
Alcohol Clin Exp Res ; 44(8): 1561-1570, 2020 08.
Article in English | MEDLINE | ID: mdl-32574382

ABSTRACT

BACKGROUND: There is growing evidence that the anticonvulsant topiramate is efficacious in reducing alcohol consumption. Further, an intronic single nucleotide polymorphism (rs2832407, C A) in the GRIK1 gene, which encodes the GluK1 subunit of the excitatory kainate receptor, predicted topiramate's effectiveness in reducing heavy drinking in a clinical trial. The molecular correlates of GRIK1 genotype that may relate to topiramate's ability to reduce drinking remain unknown. METHODS: We differentiated induced pluripotent stem cells (iPSCs) characterized by GRIK1 rs2832407 genotype from 8 A/A and 8 C/C donors into forebrain-lineage neural cultures. Our differentiation protocol yielded mixed neural cultures enriched for glutamatergic neurons. Basal mRNA expression of the GRIK1 locus was examined via quantitative polymerase chain reaction (qPCR). The effects of acute topiramate exposure on excitatory spontaneous synaptic activity were examined via whole-cell patch-clamp electrophysiology. Results were compared and contrasted between iPSC donor genotypes. RESULTS: Although characterization of the GRIK1 locus revealed no effect of rs2832407 genotype on GRIK1 isoform mRNA expression, a significant difference was observed on GRIK1 antisense-2 expression, which was greater in C/C neural cultures. Differential effects of acute exposure to 5 µM topiramate were observed on spontaneous synaptic activity in A/A versus C/C neurons, with a smaller reduction in excitatory event frequency observed in C/C donor neurons. CONCLUSIONS: This work highlights the use of iPSC technologies to study pharmacogenetic treatment effects in psychiatric disorders and furthers our understanding of the molecular effects of topiramate exposure in human neural cells.


Subject(s)
Anticonvulsants/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Neurons/drug effects , RNA, Antisense/metabolism , RNA, Messenger/metabolism , Receptors, Kainic Acid/genetics , Topiramate/pharmacology , Excitatory Postsynaptic Potentials/genetics , Genotype , Humans , Neurons/metabolism , Patch-Clamp Techniques , Pharmacogenomic Variants , Pluripotent Stem Cells , Polymorphism, Single Nucleotide , Receptors, Kainic Acid/metabolism
10.
Synapse ; : e22075, 2018 Oct 17.
Article in English | MEDLINE | ID: mdl-30334291

ABSTRACT

Brain-derived neurotrophic factor (BDNF), traditionally known for promoting neuronal growth and development, is also a modulator of synaptic transmission. In addition to the well-characterized effects at excitatory synapses, BDNF has been shown to acutely suppress inhibitory neurotransmission; however, the underlying mechanisms are unclear. We have previously shown that at inhibitory synapses in layer 2/3 of the somatosensory cortex, BDNF induces the mobilization of endogenous cannabinoids (eCBs) that act retrogradely to suppress GABA release. Here, we hypothesized that in the hippocampus, BDNF acts similarly via eCB signaling to suppress GABAergic transmission. We found that the acute application of BDNF reduced the spontaneous inhibitory postsynaptic currents (sIPSCs) via postsynaptic TrkB receptor activation. The suppressive effects of BDNF required eCB signaling, as this effect on sIPSCs was prevented by a CB1 receptor antagonist. Further, blocking the postsynaptic eCB release prevented the effect of BDNF, whereas eCB reuptake inhibition enhanced the effect of BDNF. These results suggest that BDNF triggers the postsynaptic release of eCBs. To identify the specific eCB release by BDNF, we tested the effects of disrupting the synthesis or degradation of 2-arachidonoylcglycerol (2-AG). Blocking 2-AG synthesis prevented the effect of BDNF and blocking 2-AG degradation enhanced the effect of BDNF. However, there was no change in the effect of BDNF when anandamide degradation was blocked. Collectively, these results suggest that in the hippocampus, BDNF-TrkB signaling induces the postsynaptic release of the endogenous cannabinoid 2-AG, which acts retrogradely on the presynaptic CB1 receptors to suppress GABA release.

11.
Front Neurol ; 9: 237, 2018.
Article in English | MEDLINE | ID: mdl-29713304

ABSTRACT

Animal models of neurodevelopmental disorders have provided invaluable insights into the molecular-, cellular-, and circuit-level defects associated with a plethora of genetic disruptions. In many cases, these deficits have been linked to changes in disease-relevant behaviors, but very few of these findings have been translated to treatments for human disease. This may be due to significant species differences and the difficulty in modeling disorders that involve deletion or duplication of multiple genes. The identification of primary underlying pathophysiology in these models is confounded by the accumulation of secondary disease phenotypes in the mature nervous system, as well as potential compensatory mechanisms. The discovery of induced pluripotent stem cell technology now provides a tool to accurately model complex genetic neurogenetic disorders. Using this technique, patient-specific cell lines can be generated and differentiated into specific subtypes of neurons that can be used to identify primary cellular and molecular phenotypes. It is clear that impairments in synaptic structure and function are a common pathophysiology across neurodevelopmental disorders, and electrophysiological analysis at the earliest stages of neuronal development is critical for identifying changes in activity and excitability that can contribute to synaptic dysfunction and identify targets for disease-modifying therapies.

12.
Alcohol ; 66: 45-53, 2018 02.
Article in English | MEDLINE | ID: mdl-29156239

ABSTRACT

Factors influencing the development of alcohol-use disorder (AUD) are complex and heterogeneous. While animal models have been crucial to identifying actions of alcohol on neural cells, human-derived in vitro systems that reflect an individual's genetic background hold promise in furthering our understanding of the molecular and functional effects of alcohol exposure and the pathophysiology of AUD. In this report, we utilized induced pluripotent stem cell (iPSCs)-derived neural cell cultures obtained from healthy individuals (CTLs) and those with alcohol dependence (ADs) to 1) examine the effect of 21-day alcohol exposure on mRNA expression of three genes encoding GABAA receptor subunits (GABRA1, GABRG2, and GABRD) using quantitative PCR, and 2) examine the effect of acute and chronic alcohol exposure on GABA-evoked currents using whole-cell patch-clamp electrophysiology. iPSCs from CTLs and ADs were differentiated into neural cultures enriched for forebrain-type excitatory glutamate neurons. Following 21-day alcohol exposure, significant treatment effects were observed in GABRA1, GABRG2, and GABRD mRNA expression. A modestly significant interaction between treatment and donor phenotype was observed for GABRD, which was increased in cell cultures derived from ADs. No effect of acute or chronic alcohol was observed on GABA-evoked currents in neurons from either CTLs or ADs. This work extends findings examining the effects of alcohol on the GABAA receptor in human cell in vitro model systems.


Subject(s)
Alcoholism/metabolism , Ethanol/toxicity , Neural Stem Cells/drug effects , Pluripotent Stem Cells/drug effects , RNA, Messenger/metabolism , Receptors, GABA-A/drug effects , Adult , Alcoholism/genetics , Alcoholism/pathology , Case-Control Studies , Cells, Cultured , Female , Humans , Male , Membrane Potentials , Middle Aged , Neural Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , RNA, Messenger/genetics , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Up-Regulation
13.
Nat Commun ; 8: 15038, 2017 04 24.
Article in English | MEDLINE | ID: mdl-28436452

ABSTRACT

Angelman syndrome (AS) is a neurogenetic disorder caused by deletion of the maternally inherited UBE3A allele and is characterized by developmental delay, intellectual disability, ataxia, seizures and a happy affect. Here, we explored the underlying pathophysiology using induced pluripotent stem cell-derived neurons from AS patients and unaffected controls. AS-derived neurons showed impaired maturation of resting membrane potential and action potential firing, decreased synaptic activity and reduced synaptic plasticity. These patient-specific differences were mimicked by knocking out UBE3A using CRISPR/Cas9 or by knocking down UBE3A using antisense oligonucleotides. Importantly, these phenotypes could be rescued by pharmacologically unsilencing paternal UBE3A expression. Moreover, selective effects of UBE3A disruption at late stages of in vitro development suggest that changes in action potential firing and synaptic activity may be secondary to altered resting membrane potential. Our findings provide a cellular phenotype for investigating pathogenic mechanisms underlying AS and identifying novel therapeutic strategies.


Subject(s)
Action Potentials/physiology , Angelman Syndrome/pathology , Induced Pluripotent Stem Cells/physiology , Neurons/physiology , Action Potentials/genetics , Angelman Syndrome/genetics , Angelman Syndrome/metabolism , Cell Differentiation , Cells, Cultured , Female , Gene Knockout Techniques , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Membrane Potentials/genetics , Membrane Potentials/physiology , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Neurons/metabolism , Synaptic Transmission/genetics , Synaptic Transmission/physiology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
14.
Synapse ; 71(5)2017 05.
Article in English | MEDLINE | ID: mdl-28164368

ABSTRACT

Endocannabinoids (eCBs) and neurotrophins, particularly brain-derived neurotrophic factor (BDNF), are potent neuromodulators found throughout the mammalian neocortex. Both eCBs and BDNF play critical roles in many behavioral and neurophysiological processes and are targets for the development of novel therapeutics. The effects of eCBs and BDNF are primarily mediated by the type 1 cannabinoid (CB1) receptor and the trkB tyrosine kinase receptor, respectively. Our laboratory and others have previously established that BDNF potentiates excitatory transmission by enhancing presynaptic glutamate release and modulating NMDA receptors. In contrast, we have shown that BDNF attenuates inhibitory transmission by inducing postsynaptic release of eCBs that act retrogradely to suppress GABA release in layer 2/3 of somatosensory cortex. Here, we hypothesized that BDNF also induces release of eCBs at excitatory synapses, which could have a mitigating or opposing effect on the direct presynaptic effects of BDNF. We found the highest levels of expression of CB1 and trkB and receptors in layers 2/3 and 5. Surprisingly, BDNF did not increase the frequency of spontaneous miniature excitatory postsynaptic currents (mEPSCs) onto layer 5 pyramidal neurons in somatosensory cortex, in contrast to its effects in the hippocampus and visual cortex. However, the effect of BDNF on mEPSC frequency in somatosensory cortex was unmasked by blocking CB1 receptors or disrupting eCB release. Thus, BDNF-trKB signaling regulates glutamate release in the somatosensory cortex via opposing effects, a direct presynaptic enhancement of release probability, and simultaneous postsynaptically-induced eCB release that decreases release probability via presynaptic CB1 receptors.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Endocannabinoids/metabolism , Excitatory Postsynaptic Potentials , Miniature Postsynaptic Potentials , Neocortex/metabolism , Pyramidal Cells/metabolism , Animals , Cells, Cultured , Glutamic Acid/metabolism , Mice , Mice, Inbred C57BL , Neocortex/cytology , Neocortex/physiology , Pyramidal Cells/physiology , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Receptor, trkB/genetics , Receptor, trkB/metabolism , Somatosensory Cortex/cytology , Somatosensory Cortex/metabolism , Somatosensory Cortex/physiology , Synapses/metabolism , Synapses/physiology
15.
OBM Neurobiol ; 1(2)2017.
Article in English | MEDLINE | ID: mdl-30854511

ABSTRACT

Autism spectrum disorders (ASDs) are diagnosed on the basis of three behavioral features, namely, (1) deficits in social communication, (2) absence or delay in language and (3) stereotypy. The consensus regarding the neurological pathogenesis of ASDs is aberrant synaptogenesis and synapse function. Further, it is now widely accepted that ASD is neurodevelopmental in nature, placing emphasis on derangements occurring at the level of intra- and intercellular signaling during corticogenesis. At present, there is an ever-growing list of mutations in putative susceptibility genes in affected individuals, preventing effective transformation of knowledge gathered from basic science research to the clinic. In response, the focus of ASD biology has shifted toward the identification of cellular signaling pathways that are common to various ASD-related mutations in hopes that these shared pathways may serve as more promising treatment targets than targeting individual genes or proteins. To this end, the endogenous cannabinoid (endocannabinoid, eCB) system has recently emerged as a promising therapeutic target in the field of ASD research. The eCB system is altered in several neurological disorders, but the role of these bioactive lipids in ASD etiology remains poorly understood. In this perspective, we review current evidence linking eCB signaling to ASDs and put forth the notion that continued focus on eCBs in autism research may provide valuable insight into pathophysiology and treatment strategies. In addition to its role in modulating transmitter release at mature synapses, the eCB signaling system plays important roles in many aspects of cortical development, and disruption of these effects of eCBs may also be related to ASD pathophysiology.

16.
Psychiatry Res ; 247: 172-181, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27915167

ABSTRACT

In peripheral blood leukocytes, FKBP5 mRNA expression is upregulated following glucocorticoid receptor activation. The single nucleotide polymorphism rs1360780 in FKBP5 is associated with psychiatric illness and has functional molecular effects. However, examination of FKBP5 regulation has largely been limited to peripheral cells, which may not reflect regulation in neural cells. We used 27 human induced pluripotent stem cell lines (iPSCs) derived from 20 subjects to examine FKBP5 mRNA expression following GR activation. Following differentiation into forebrain-lineage neural cultures, cells were exposed to 1µM dexamethasone and mRNA expression of FKBP5 and NR3C1 analyzed. Results from the iPSC-derived neural cells were compared with those from 15 donor matched fibroblast lines. Following dexamethasone treatment, there was a 670% increase in FKBP5 expression in fibroblasts, mimicking findings in peripheral blood-derived cells, but only a 23% increase in iPSC-derived neural cultures. FKBP5 rs1360780 genotype did not affect the induction of FKBP5 mRNA in either fibroblasts or neural cells. These results suggest that iPSC-derived forebrain-lineage neurons may not be an optimal neural cell type in which to examine relationships between GR activation, FKBP5 expression, and genetic variation in human subjects. Further, FKBP5 induction following GR activation may differ between cell types derived from the same individual.


Subject(s)
Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Neural Stem Cells/drug effects , RNA, Messenger/metabolism , Tacrolimus Binding Proteins/genetics , Genotype , Humans , Induced Pluripotent Stem Cells/physiology , Polymorphism, Single Nucleotide , Receptors, Glucocorticoid/drug effects
17.
J Dent ; 43(12): 1539-46, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26404407

ABSTRACT

OBJECTIVES: Biofilm acids contribute to secondary caries which is a reason for restoration failure. Previous studies synthesized nanoparticles of amorphous calcium phosphate (NACP) and dimethylaminohexadecyl methacrylate (DMAHDM). The objectives of this study were to develop DMAHMD-NACP nanocomposite for double benefits of antibacterial and remineralization capabilities, and investigate the DMAHMD mass fraction effects on fracture toughness and biofilm response of NACP nanocomposite for the first time. METHODS: DMAHDM was incorporated into NACP nanocomposite at mass fractions of 0% (control), 0.75%, 1.5%, 2.25% and 3%. A single edge V-notched beam method was used to measure fracture toughness K(IC). A dental plaque microcosm biofilm model using human saliva as inoculum was used to measure the antibacterial properties of composites. RESULTS: K(IC) was about 1 MPa×m(1/2) for all composite (mean±sd; n=6). Adding DMAHDM from 0% to 3% did not affect K(IC) (p>0.1). Lactic acid production by biofilms on composite containing 3% DMAHDM was reduced to less than 1% of that on composite control. Metabolic activity of adherent biofilms on composite containing 3% DMAHDM was reduced to 4% of that on composite control. Biofilm colony-forming unit (CFU) counts were reduced by three orders of magnitude on NACP nanocomposite containing 3% DMAHDM. CONCLUSIONS: DMAHDM-NACP nanocomposite had good fracture resistance, strong antibacterial potency, and NACP for remineralization (shown in previous studies). The DMAHDM-NACP nanocomposite may be promising for caries-inhibiting dental restorations, and the method of using double agents (DMAHDM and NACP) may have a wide applicability to other dental materials including bonding agents and cements.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Methacrylates/chemistry , Methacrylates/pharmacology , Nanocomposites/chemistry , Tooth Remineralization/methods , Bacteria/drug effects , Calcium Phosphates/chemistry , Calcium Phosphates/pharmacology , Composite Resins/chemistry , Composite Resins/pharmacology , Dental Caries/prevention & control , Dental Materials/pharmacology , Dental Plaque/drug therapy , Dental Plaque/microbiology , Humans , Lactic Acid/metabolism , Microbial Viability/drug effects , Nanoparticles/chemistry , Saliva/microbiology , Streptococcus mutans/drug effects , Streptococcus mutans/physiology
18.
eNeuro ; 2(2)2015 02 28.
Article in English | MEDLINE | ID: mdl-25938134

ABSTRACT

The endogenous cannabinoid (endocannabinoid) system is an important regulator of synaptic function. Endocannabinoids acutely modulate inhibitory and excitatory transmission, and also mediate long-term depression at GABAergic and glutamatergic synapses. Typically, endocannabinoid synthesis and release is stimulated by depolarization-induced calcium influx and/or activation of phospholipase-C (PLC) signaling triggered by mGluR activation. Recently it has been shown that brain-derived neurotrophic factor (BDNF) can also induce endocannabinoid release. Although there is growing evidence for cross-talk between BDNF and endocannabinoid signaling, little is known about the functional relevance of these interactions. In the present studies, we examined BDNF - endocannabinoid interactions in regulating activity-dependent long-term depression at inhibitory synapses (iLTD). We found that theta burst stimulation (TBS) in layer 2/3 of mouse somatosensory cortical slices can induce a form of endocannabinoid-mediated iLTD that is independent of metabotropic glutamate receptor (mGluR) activation. This endocannabinoid-dependent iLTD, however, requires endogenous BDNF-trkB signaling, as it is blocked by a trk tyrosine kinase inhibitor and by a trkB receptor antagonist, and also requires activation of diacylglycerol lipase (DAG-lipase, DGL). In addition, endocannabinoid-mediated iLTD can be induced by combining a subthreshold concentration of exogenous BDNF with weak TBS stimulation that by itself is insufficient to induce iLTD. Taken together, our results suggest that TBS can induce the release of endogenous BDNF, which triggers DGL-dependent endocannabinoid mobilization and cannabinoid receptor-dependent iLTD at layer 2/3 cortical synapses.

19.
J Dent Educ ; 78(12): 1673-80, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25480283

ABSTRACT

The Commission on Dental Accreditation (CODA)'s accreditation standards for dental schools state that "graduates must demonstrate the ability to self-assess." Therefore, dental schools have developed preclinical and clinical self-assessment (SA) protocols aimed at fostering a reflective process. This study comparing students' visual SA with students' digital SA and with faculty assessment was designed to test the hypothesis that higher agreement would occur when utilizing a digital evaluation. Twenty-five first-year dental students at one dental school participated by preparing a mesial occlusal preparation on tooth #30 and performing both types of SAs. A faculty evaluation was then performed both visually and digitally using the same evaluation criteria. The Kappa statistic was used to measure agreement between evaluators. The results showed statistically significant moderate agreement between the faculty visual and faculty digital modes of evaluation for occlusal shape (K=0.507, p=0.002), proximal shape (K=0.564, p=0.001), orientation (K=0.425, p=0.001), and definition (K=0.480, p=0.001). There was slight to poor agreement between the student visual and faculty visual assessment, except for preparation orientation occlusal shape (K=0.164, p=0.022), proximal shape (K=-0.227, p=0.032), orientation (K=0.253, p=0.041), and definition (K=-0.027, p=0.824). This study showed that the students had challenges in self-assessing even when using CAD/CAM and the digital assessment did not improve the amount of student/faculty agreement.


Subject(s)
Computer-Aided Design , Dentistry, Operative/education , Education, Dental , Educational Measurement/methods , Self-Assessment , Students, Dental/psychology , Tooth Preparation/methods , Clinical Competence , Dental Cavity Preparation/classification , Dental Cavity Preparation/standards , Dental Prosthesis Retention , Faculty, Dental , Humans , Image Processing, Computer-Assisted/methods , Motor Skills , Tooth Preparation/standards , Visual Perception
20.
J Neurophysiol ; 111(5): 1008-15, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24335212

ABSTRACT

Endogenous cannabinoids (endocannabinoids) and neurotrophins, particularly brain-derived neurotrophic factor (BDNF), are potent synaptic modulators that are expressed throughout the forebrain and play critical roles in many behavioral processes. Although the effects of BDNF at excitatory synapses have been well characterized, the mechanisms of action of BDNF at inhibitory synapses are not well understood. Previously we have found that BDNF suppresses presynaptic GABA release in layer 2/3 of the neocortex via postsynaptic tropomyosin-related kinase receptor B (trkB) receptor-induced release of endocannabinoids. To examine the intracellular signaling pathways that underlie this effect, we used pharmacological approaches and whole cell patch-clamp techniques in layer 2/3 pyramidal neurons of somatosensory cortex in brain slices from juvenile Swiss CD1 mice. Our results indicated that phospholipase Cγ (PLCγ) is involved in the CB1 receptor-mediated synaptic effect of BDNF, because the BDNF effect was blocked in the presence of the broad-spectrum PLC inhibitors U-73122 and edelfosine, whereas the inactive analog U-73343 did not alter the suppressive effect of BDNF at inhibitory synapses. Endocannabinoid release can also be triggered by metabotropic glutamate receptor (mGluR)-mediated activation of PLCß, and BDNF has been shown to enhance spontaneous glutamate release. An mGluR antagonist, E4CPG, however, did not block the BDNF effect. In addition, the effect of BDNF was independent of other signaling pathways downstream of trkB receptor activation, namely, mitogen-activated protein kinase and phosphoinositide 3-kinase pathways, as well as protein kinase C signaling.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Inhibitory Postsynaptic Potentials , Neurons/metabolism , Phospholipase C gamma/metabolism , Receptor, Cannabinoid, CB1/metabolism , Somatosensory Cortex/metabolism , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Cannabinoid Receptor Antagonists/pharmacology , Inhibitory Postsynaptic Potentials/drug effects , Mice , Neurons/drug effects , Phospholipase C beta/metabolism , Piperidines/pharmacology , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Somatosensory Cortex/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...