Jia-Hua Hu's research while affiliated with Eunice Kennedy Shriver National Institute of Child Health and Human Development and other places

What is this page?


This page lists the scientific contributions of an author, who either does not have a ResearchGate profile, or has not yet added these contributions to their profile.

It was automatically created by ResearchGate to create a record of this author's body of work. We create such pages to advance our goal of creating and maintaining the most comprehensive scientific repository possible. In doing so, we process publicly available (personal) data relating to the author as a member of the scientific community.

If you're a ResearchGate member, you can follow this page to keep up with this author's work.

If you are this author, and you don't want us to display this page anymore, please let us know.

Publications (25)


FIGURE 1
FIGURE 2 Purification of Kv4.2 protein complex using TAP in culture hippocampal neurons. (A) Schematic diagram of Kv4.2 protein complex purification using TAP in cultured hippocampal neurons. (B) Lentivirus expression of GFP or TAP-Kv4.2 plus GFP in HEK293FT cells (DIV 5). (C) Lentivirus expression of GFP or TAP-Kv4.2 plus GFP in hippocampal neurons (DIV10 after infection). (D) Western blot showing TAP-Kv4.2 expression in DIV14 hippocampal neurons. (E) Silver staining showing purified TAP-Kv4.2 protein complex in DIV14 hippocampal neurons. Note that there are plenty of non-specific proteins in control TAP purification after streptavidin resin pulldown (1st elution), while non-specific proteins are not visible after calmodulin resin pulldown (2nd elution), demonstrating the effectiveness of TAP in cultured hippocampal neurons. E, empty; M, marker.
FIGURE 4 Identification of Kv4.2 phosphorylation sites in hippocampal neurons by TAP. (A) Coomassie staining showing TAP-Kv4.2 is purified by TAP using stronger lysis buffer (1% Triton X-100 and 0.5% DOC) in the control and AMPA treatment conditions. (B) Phospho-peptides of Kv4.2 that contain multiple phosphorylation sites identified by TAP-MS. (C) Number of phospho-sites in Kv4.2 were identified by TAP-MS in control and AMPA (50µM for 15 min) treatment in culture hippocampal neurons. Note that T607 phospho-peptides but not T602 and S616 phospho-peptides were increased in AMPA treatment compared to control.
Candidate proteins that were identified by TAP-MS in hippocampal neurons.
Continued)
Identification of Kv4.2 protein complex and modifications by tandem affinity purification-mass spectrometry in primary neurons
  • Article
  • Full-text available

December 2022

·

27 Reads

·

3 Citations

Frontiers in Cellular Neuroscience

Jia-Hua Hu

·

·

Proteins usually form complexes to fulfill variable physiological functions. In neurons, communication relies on synapses where receptors, channels, and anchoring proteins form complexes to precisely control signal transduction, synaptic integration, and action potential firing. Although there are many published protocols to isolate protein complexes in cell lines, isolation in neurons has not been well established. Here we introduce a method that combines lentiviral protein expression with tandem affinity purification followed by mass-spectrometry (TAP-MS) to identify protein complexes in neurons. This protocol can also be used to identify post-translational modifications (PTMs) of synaptic proteins. We used the A-type voltage-gated K+ channel subunit Kv4.2 as the target protein. Kv4.2 is highly expressed in the hippocampus where it contributes to learning and memory through its regulation of neuronal excitability and synaptic plasticity. We tagged Kv4.2 with the calmodulin-binding-peptide (CBP) and streptavidin-binding-peptide (SBP) at its C-terminus and expressed it in neurons via lentivirus. Kv4.2 was purified by two-step TAP and samples were analyzed by MS. MS identified two prominently known Kv4.2 interacting proteins [dipeptidyl peptidase like (DPPs) and Kv channel-interacting proteins (KChIPs)] in addition to novel synaptic proteins including glutamate receptors, a calcium channel, and anchoring proteins. Co-immunoprecipitation and colocalization experiments validated the association of Kv4.2 with glutamate receptors. In addition to protein complex identification, we used TAP-MS to identify Kv4.2 phosphorylation sites. Several known and unknown phosphorylation sites were identified. These findings provide a novel path to identify protein-protein interactions and PTMs in neurons and shed light on mechanisms of neuronal signaling potentially involved in the pathology of neurological diseases.

Download
Share

Passive membrane properties and single action potential (AP) parameters (mean ± SEM).
P38 Regulates Kainic Acid-Induced Seizure and Neuronal Firing via Kv4.2 Phosphorylation

August 2020

·

50 Reads

·

8 Citations

International Journal of Molecular Sciences

The subthreshold, transient A-type K+ current is a vital regulator of the excitability of neurons throughout the brain. In mammalian hippocampal pyramidal neurons, this current is carried primarily by ion channels comprising Kv4.2 α-subunits. These channels occupy the somatodendritic domains of these principle excitatory neurons and thus regulate membrane voltage relevant to the input–output efficacy of these cells. Owing to their robust control of membrane excitability and ubiquitous expression in the hippocampus, their dysfunction can alter network stability in a manner that manifests in recurrent seizures. Indeed, growing evidence implicates these channels in intractable epilepsies of the temporal lobe, which underscores the importance of determining the molecular mechanisms underlying their regulation and contribution to pathologies. Here, we describe the role of p38 kinase phosphorylation of a C-terminal motif in Kv4.2 in modulating hippocampal neuronal excitability and behavioral seizure strength. Using a combination of biochemical, single-cell electrophysiology, and in vivo seizure techniques, we show that kainic acid-induced seizure induces p38-mediated phosphorylation of Thr607 in Kv4.2 in a time-dependent manner. The pharmacological and genetic disruption of this process reduces neuronal excitability and dampens seizure intensity, illuminating a cellular cascade that may be targeted for therapeutic intervention to mitigate seizure intensity and progression.



Activity-dependent isomerization of Kv4.2 by Pin1 regulates cognitive flexibility

March 2020

·

250 Reads

·

34 Citations

Nature Communications

Voltage-gated K⁺ channels function in macromolecular complexes with accessory subunits to regulate brain function. Here, we describe a peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (Pin1)-dependent mechanism that regulates the association of the A-type K⁺ channel subunit Kv4.2 with its auxiliary subunit dipeptidyl peptidase 6 (DPP6), and thereby modulates neuronal excitability and cognitive flexibility. We show that activity-induced Kv4.2 phosphorylation triggers Pin1 binding to, and isomerization of, Kv4.2 at the pThr⁶⁰⁷-Pro motif, leading to the dissociation of the Kv4.2-DPP6 complex. We generated a novel mouse line harboring a knock-in Thr607 to Ala (Kv4.2TA) mutation that abolished dynamic Pin1 binding to Kv4.2. CA1 pyramidal neurons of the hippocampus from these mice exhibited altered Kv4.2-DPP6 interaction, increased A-type K⁺ current, and reduced neuronal excitability. Behaviorally, Kv4.2TA mice displayed normal initial learning but improved reversal learning in both Morris water maze and lever press paradigms. These findings reveal a Pin1-mediated mechanism regulating reversal learning and provide potential targets for the treatment of neuropsychiatric disorders characterized by cognitive inflexibility.



Fig. 1. Expression of BBS-Kv4.2 in HEK 293T cells. (A) Design of the BBS-Kv4.2 construct. A BBS tag (red text) flanked by dual glycines was inserted into the 2nd extracellular loop of an existing wild-type human Kv4.2 construct (Kv4.2) containing C-terminal myc and DDK (FLAG) tags. (B and C) Western blots showing BBS-Kv4.2 expression in HEK 293T cells. Both the Kv4.2 and BBS-Kv4.2 constructs were detected with anti-Myc (B) and anti-Kv4.2 (C) antibodies. Actin is shown as a loading control. (D) Surface labeling of cells expressing Kv4.2 with or without the S3-S4 linker region BBS insert. Cells were stained with rhodamine-BTX (Rh-BTX) before permeabilization (to label surface BBS) and anti-Myc antibody after permeabilization (to label total Kv4.2). Both Kv4.2 and BBS-Kv4.2 constructs were visualized with the anti-Myc antibody, while only the BBS-Kv4.2 construct was labeled with Rh-BTX at the cell surface. Intensity plots along the white line were shown on the right. Scale bar: 10 μm. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)
Fig. 3. Detection of BBS-Kv4.2 internalization in HEK 293T cells. (A) Cells transfected with BBS-Kv4.2 were labeled with Rh-BTX, washed, and incubated at 37 °C to allow internalization of the channel. Control cells were incubated at 4 °C for 30 min after BTX-labeling. Cells were fixed at the indicated times and imaged with confocal microscopy. Kv4.2 surface fraction signal decreased over time when the cells were incubated at 37 °C and remained strong when the cells were incubated at 4 °C. (B) Graphical representation of (A), n = 15 for each group. (C) Cells transfected with BBS-Kv4.2 were labeled with biotin-BTX, washed, and incubated at 37 °C to allow internalization of the channel. Control cells were incubated at 4 °C for 30 min after BTX-biotin labeling. Cells were lysed at the indicated times, and the labeled Kv4.2 was pulled down with streptavidin beads. Western blots showed that the levels of surface labeled BBS-Kv4.2 decreased throughout the incubation period at 37 °C. (D) Graphical representation of (C), n = 4 for each group. Ctl: Control.
Fig. 4. Detection of BBS-Kv4.2 insertion in HEK 293T cells. (A) Cells transfected with BBS-Kv4.2 were preincubated with unlabeled BTX at 17 °C followed by incubation with Rh-BTX at 37 °C for the indicated times to visualize the insertion of new channels into the plasma membrane. Control cells were incubated with Rh-BTX at 4 °C for 30 min. Rapid channel insertion within 5 min could be visualized with this method. Scale bar: 20 μm. (B) Graphical representation of (A), n = 15 for each group. (C) Cells transfected with BBS-Kv4.2 were preincubated with unlabeled BTX at 17 °C followed by incubation with biotin-BTX at 37 °C for the indicated times to visualize exocytosis. Control cells were incubated with biotin-BTX at 4 °C for 30 min. Cells were lysed at the indicated times and the labeled BBS-Kv4.2 was pulled down with streptavidin beads. Western blots showed that the levels of surface labeled BBS-Kv4.2 increased throughout the incubation period at 37 °C. (D) Graphical representation of (C), n = 3 for each group. Ctl: Control.
Fig. 5. Colocalization of internalized BBS-Kv4.2 with endosome markers in HEK 293T cells. Cells transfected with BBS-Kv4.2 and the indicated endosome markers were incubated with Rh-BTX at 37 °C for 1.5 h. (A, B) Internalized Kv4.2 strongly colocalized with the early endosome marker Rab5. Scale bar: 20 μm. (B) High magnification of Kv4.2 colocalization with Rab5. Scale bar: 5 μm. (C, D) Some internalized Kv4.2 colocalized with the late endosome marker Rab7. (D) High magnification of Kv4.2 colocalization with Rab7. (E, F) A large proportion of internalized Kv4.2 colocalized with the recycling endosome marker Rab11. (F) High magnification of Kv4.2 colocalization with Rab11. (G, H) A portion of internalized Kv4.2 colocalized with the lysosome marker Lamp1. (H) High magnification of Kv4.2 colocalization with Lamp1. Intensity plots along the white line were shown on the right. The origin of the intensity plots coincides with the top end of each line in the fluorescent images.
Biophysical properties of Kv4.2 and BBS-Kv4.2 voltage gated K + currents. VDA: voltage dependence of activation; VDI: voltage dependence of inactivation; RFI: recovery from inactivation; All entries describe the mean ± SEM. Statistical comparisons were made between WT Kv4.2 alone vs. BBS-Kv4.2 alone, WT Kv4.2 + KChiP2 vs. BBS-Kv4.2 + KChIP2, or WT Kv4.2 + DPP6 vs. BBS-Kv4.2 + DPP6.
A novel bungarotoxin binding site-tagged construct reveals MAPK-dependent Kv4.2 trafficking

June 2019

·

276 Reads

·

2 Citations

Molecular and Cellular Neuroscience

Kv4.2 voltage-gated K+ channel subunits, the primary source of the somatodendritic A-type K+ current in CA1 pyramidal neurons of the hippocampus, play important roles in regulating dendritic excitability and plasticity. To better study the trafficking and subcellular distribution of Kv4.2, we created and characterized a novel Kv4.2 construct encoding a bungarotoxin binding site in the extracellular S3–S4 linker region of the α-subunit. When expressed, this construct can be visualized in living cells after staining with rhodamine-conjugated bungarotoxin. We validated the utility of this construct by visualizing the spontaneous internalization and insertion of Kv4.2 in HEK 293T cells. We further report that Kv4.2 colocalized with several endosome markers in HEK 293T cells. In addition, Kv4.2 internalization is significantly impaired by mitogen-activated protein kinase (MAPK) inhibitors in transfected primary hippocampal neurons. Therefore, this newly developed BBS-Kv4.2 construct provides a novel and powerful tool for studying surface Kv4.2 channel localization and trafficking.


Figure 1. Pedigree, photographs, and brain MRIs of Affected Males with ID and FRMPD4 p.Cys618ValfsX8 in an XLID Family. (A) Pedigree shows 5 affected males from a multigenerational family. Filled square: affected males; circle with a center dot, carrier females; unfilled square: unaffected males; unfilled circle: unaffected females. Symbols with a line crossed: deceased. (B) Note absence of distinct dysmorphic features except for bifid tip of the nose in P1 (II: 1). (C) Note absence of distinct dysmorphic features and a white matter hyper-signal in the left occipital horn on brain MRI (axial T2) in P2 (III: 1). (D) Note absence of distinct dysmorphic features and a left temporal arachnoid cyst (axial T1) in P5 (III: 8).
Figure 2. Additional FRMPD4 Mutations in Affected Individuals with XLID. (A) Established domain structure of FRMPD4 and nature and locations of FRMPD4 mutations found in patients with ID. WW (domain with two conserved Trp residues): PDZ (PSD-95/Dlg/ZO-1): RA; FERM (four-point-one, ezrin, radixin, and moesin). (B) Genomic structure of FRMPD4 and genomic location of family 3 microdeletion. (C) Pedigrees and segregation analysis of FRMPD4 mutations identified in families 2, 3 and 4. Filled square, affected males; open square, unaffected males; circle with a center dot, unaffected carrier female; filled circle with a center dot, mildly disabled carrier female. (D) Brain MRI scans of P6 (at 12 years) and P7 (at 17 years) from Family 2. Note diffuse atrophy of white matter principally in the periventricular region, delayed myelination with periventricular white matter atrophy of P6 (top panel, axial T1), and compare to cortical atrophy and enlarged ventricles of P7 (bottom panel, axial T2). (E) Photographs of family 3 probands and their mildly disabled sister. (F) Proband of family 4 at different ages. Note frontal upsweep, trigonocephaly and broad nasal bridge.
Figure 3. Binding of FRMPD4 p.Cys618ValfsX8 with known interacting proteins. (A) Domain structure of human FRMPD4 and its mutant protein. (B) Various FRMPD4 and Homer1c constructs were transfected into HEK293T cells. Anti-myc antibody immunoprecipitated FRMPD4-WT and FRMPD4 p.Cys618ValfsX8 . Homer1C was co-immunoprecipitated by FRMPD4-WT, but not FRMPD4 p.Cys618ValfsX8 . (C) Various FRMPD4 and PSD-95 constructs were transfected into HEK293T cells. Anti-myc antibody immunoprecipitated FRMPD4-WT and FRMPD4 p.Cys618ValfsX8 . PSD-95 was co-immunoprecipitated by FRMPD4-WT while this binding was significantly reduced between
Figure 5. Frmpd4-KO mice show normal spatial working memory but a defective spatial reference memory. The Y-maze tests for spatial working memory include Spontaneous Alternation and Blocked Arm tests. (A) Spontaneous Alternation: the test mouse was placed at the end of one arm and remained in the maze for 5 min.
FRMPD4 Mutations Cause X-linked Intellectual Disability and Disrupt Dendritic Spine Morphogenesis

December 2017

·

779 Reads

·

23 Citations

Human Molecular Genetics

FRMPD4 (FERM and PDZ Domain Containing 4) is a neural scaffolding protein that interacts with PSD-95 to positively regulate dendritic spine morphogenesis, and with mGluR1/5 and Homer to regulate mGluR1/5 signaling. We report the genetic and functional characterization of 4 FRMPD4 deleterious mutations that cause a new X-linked intellectual disability (ID) syndrome. These mutations were found to be associated with ID in ten affected male patients from four unrelated families, following an apparent X-linked mode of inheritance. Mutations include deletion of an entire coding exon, a nonsense mutation, a frame-shift mutation resulting in premature termination of translation, and a missense mutation involving a highly conserved amino acid residue neighboring FRMPD4-FERM domain. Clinical features of these patients consisted of moderate to severe ID, language delay and seizures alongside with behavioral and/or psychiatric disturbances. In-depth functional studies showed that a frame-shift mutation, FRMPD4p.Cys618ValfsX, result in a disruption of FRMPD4 binding with PSD-95 and HOMER1, and a failure to increase spine density in transfected hippocampal neurons. Behavioral studies of frmpd4-KO mice identified hippocampus-dependent spatial learning and memory deficits in Morris Water Maze test. These findings point to an important role of FRMPD4 in normal cognitive development and function in humans and mice, and support the hypothesis that FRMPD4 mutations cause ID by disrupting dendritic spine morphogenesis in glutamatergic neurons.


Behavioral and Neurochemical Phenotyping of Mice Incapable of Homer1a Induction

November 2017

·

134 Reads

·

13 Citations

Frontiers in Behavioral Neuroscience

Frontiers in Behavioral Neuroscience

Immediate early and constitutively expressed products of the Homer1 gene regulate the functional assembly of post-synaptic density proteins at glutamatergic synapses to influence excitatory neurotransmission and synaptic plasticity. Earlier studies of Homer1 gene knock-out (KO) mice indicated active, but distinct, roles for IEG and constitutively expressed Homer1 gene products in regulating cognitive, emotional, motivational and sensorimotor processing, as well as behavioral and neurochemical sensitivity to cocaine. More recent characterization of transgenic mice engineered to prevent generation of the IEG form (a.k.a Homer1a KO) pose a critical role for Homer1a in cocaine-induced behavioral and neurochemical sensitization of relevance to drug addiction and related neuropsychiatric disorders. Here, we extend our characterization of the Homer1a KO mouse and report a modest pro-depressant phenotype, but no deleterious effects of the KO upon spatial learning/memory, prepulse inhibition, or cocaine-induced place-conditioning. As we reported previously, Homer1a KO mice did not develop cocaine-induced behavioral or neurochemical sensitization within the nucleus accumbens; however, virus-mediated Homer1a over-expression within the nucleus accumbens reversed the sensitization phenotype of KO mice. We also report several neurochemical abnormalities within the nucleus accumbens of Homer1a KO mice that include: elevated basal dopamine and reduced basal glutamate content, Group1 mGluR agonist-induced glutamate release and high K+-stimulated release of dopamine and glutamate within this region. Many of the neurochemical anomalies exhibited by Homer1a KO mice are recapitulated upon deletion of the entire Homer1 gene; however, Homer1 deletion did not affect NAC dopamine or alter K+-stimulated neurotransmitter release within this region. These data show that the selective deletion of Homer1a produces a behavioral and neurochemical phenotype that is distinguishable from that produced by deletion of the entire Homer1 gene. Moreover, the data indicate a specific role for Homer1a in regulating cocaine-induced behavioral and neurochemical sensitization of potential relevance to the psychotogenic properties of this drug.


Dynamic regulation of Homer binding to group I mGluRs by Preso1 and converging kinase cascades

February 2017

·

15 Reads

·

4 Citations

Journal of Pharmacology and Experimental Therapeutics

In rat sympathetic neurons from the superior cervical ganglia (SCG) expressing metabotropic glutamate receptors (mGluRs) 1 or 5, overexpression of scaffolding Homer proteins, which bind to a Homer ligand in their C-termini, cause receptor clustering and uncoupling from ion channel modulation. In the absence of recombinant Homer protein overexpression, uncoupling of mGluRs from voltage dependent channels can be induced by expression of Preso1, an adaptor of proline directed kinases that phosphorylates the Homer ligand and recruits binding of endogenous Homer proteins. Here we show that in SCG neurons expressing mGluR1 or 5 and the receptor tyrosine kinase TrkB, treatment with BDNF produces a similar uncoupling of the receptors from calcium channels. We investigated the pathways that mediate this uncoupling and compared it with uncoupling observed with Preso1 expression. Both BDNF- and Preso1-induced uncoupling requires residues T1151 and S1154 in the mGluR1 Homer ligand (TPPSPF). Uncoupling via Preso1 but not BDNF was prevented by expression of a dominant negative Cdk5 suggesting endogenous Cdk5 mediates Preso1-dependent phosphorylation of mGluR. dnCDK5 did not block the BDNF effect but this was sensitive to inhibitors of the MEK-ERK kinase cascade. Interestingly, the BDNF pathway appeared to require native Preso1 binding to mGluR, as over-expression of the Preso1 FERM domain, which mediates the Preso1-mGluR interaction, prevented BDNF-induced uncoupling. These data suggest that BDNF-TrkB and CDK5 pathways converge at the level of mGluR to similarly induce Homer ligand phosphorylation, recruit Homer binding, and uncouple mGluRs from channel regulation.


Protein Kinase C Epsilon Activity in the Nucleus Accumbens and Central Nucleus of the Amygdala Mediates Binge Alcohol Consumption

March 2015

·

72 Reads

·

34 Citations

Biological Psychiatry

Protein kinase C epsilon (PKCε) is emerging as a potential target for the development of pharmacotherapies to treat alcohol use disorders, yet little is known regarding how a history of a highly prevalent form of drinking, binge alcohol intake, influences enzyme priming or the functional relevance of kinase activity for excessive alcohol intake. Immunoblotting was employed on tissue from subregions of the nucleus accumbens (NAc) and the amygdala to examine both idiopathic and binge drinking-induced changes in constitutive PKCε priming. The functional relevance of PKCε translocation for binge drinking and determination of potential upstream signaling pathways involved were investigated using neuropharmacologic approaches within the context of two distinct binge drinking procedures, drinking in the dark and scheduled high alcohol consumption. Binge alcohol drinking elevated p(Ser729)-PKCε levels in both the NAc and the central nucleus of the amygdala (CeA). Moreover, immunoblotting studies of selectively bred and transgenic mouse lines revealed a positive correlation between the propensity to binge drink alcohol and constitutive p(Ser729)-PKCε levels in the NAc and CeA. Finally, neuropharmacologic inhibition of PKCε translocation within both regions reduced binge alcohol consumption in a manner requiring intact group 1 metabotropic glutamate receptors, Homer2, phospholipase C, and/or phosphotidylinositide-3 kinase function. Taken together, these data indicate that PKCε signaling in both the NAc and CeA is a major contributor to binge alcohol drinking and to the genetic propensity to consume excessive amounts of alcohol. Copyright © 2015 Society of Biological Psychiatry. Published by Elsevier Inc. All rights reserved.


Citations (19)


... This specific neuronal distribution allows I A not only to regulate the integration and the propagation of the excitatory synaptic potentials, but also to modulate the back-propagating potentials in dendritic branches [60,61]. Among the five A-channels in mammals, recently, several authors have focused their attention on two specific subunits: Kv4.2 and Kv4.3 [43,55,62,63]. The latter is often associated with different β subunits and multiple K + channel interacting proteins, such as KChIPs, which can modulate Kv4 current properties, neuronal trafficking and, in turn, its functions [63,64]. ...

Reference:

Ion Channel and Transporter Involvement in Chemotherapy-Induced Peripheral Neurotoxicity
Identification of Kv4.2 protein complex and modifications by tandem affinity purification-mass spectrometry in primary neurons

Frontiers in Cellular Neuroscience

... The presumed mechanism of this increase in I A is the prevention of p38-Pin1-mediated dissociation of Kv4.2 and DPP6. Similar blockade of Kv4.2-DPP6 dissociation was also shown to reduce kainic acid induced seizure intensity in mice [123]. ...

P38 Regulates Kainic Acid-Induced Seizure and Neuronal Firing via Kv4.2 Phosphorylation

International Journal of Molecular Sciences

... When PIN1 becomes activated, it binds dynamically to the Thr-607 phosphorylated form of Kv4.2, leading to the isomerization of the proline involved at the related pSer/Thr-Pro motif. This causes the dissociation of Kv4.2 from its dipeptidyl peptidase 6 (DPP6) subunit, inhibiting the Kv4.2 function for protection from excess neuron excitability through depolarization [256]. The isomerization induced by PIN1 in Kv4.2 is important for proper neuronal and cognitive function [256]. ...

Activity-dependent isomerization of Kv4.2 by Pin1 regulates cognitive flexibility

Nature Communications

... The importance of tagged α-bungarotoxin is nevertheless substantiated by the fact that the α-bungarotoxin binding site (BS), an optimized small peptide of about 13 amino acids derived from a combinatorial phage-display peptide library, fully preserves the unique properties of high affinity binding of the toxin [16,17]. Hence, in addition to the fact that it may serve as a lead compound for the development of antidotes [18], this BBS peptide can also be used as a tag onto poorly immunogenic proteins to visualize their cell behaviour with tagged α-bungarotoxin [19][20][21][22][23][24][25][26][27][28]. ...

A novel bungarotoxin binding site-tagged construct reveals MAPK-dependent Kv4.2 trafficking

Molecular and Cellular Neuroscience

... In 2016, Hu et al. conducted genetic examinations on five male patients with ID in a large family and revealed, for the first time, that FRMPD4 gene defects were related to ID. Subsequently, Piard et al. (2018) discovered the FRMPD4 gene in six patients with ID and further enriched the spectrum of genetic variation. Since then, ID caused by FRMPD4 gene defects have been reported in many families. ...

FRMPD4 Mutations Cause X-linked Intellectual Disability and Disrupt Dendritic Spine Morphogenesis

Human Molecular Genetics

... However, subsequent research into Homer1a function has cast some doubt on classifying Homer1a as a PRP. Knock-out studies of Homer1a in mice have produced varied results, ranging from moderate effects on memory retention [61] to no observable effects at all [62]. In addition, Klugmann and colleagues compared the overexpression of all individual isoforms of Homer1 in the rat hippocampus [63]. ...

Behavioral and Neurochemical Phenotyping of Mice Incapable of Homer1a Induction
Frontiers in Behavioral Neuroscience

Frontiers in Behavioral Neuroscience

... Through these interactions, Preso coordinates the coupling of mGluR5 with Homer1 and thus promotes mGluR5-mediated pain signaling [18]. Similarly, Preso modulates the interaction between mGluR1 and Homer1 via a dynamic mechanism [19]. Considering the important role of the mGluR1-Homer1 complex in excitotoxicity, it is possible that the Preso-mediated mGluR1-Homer1 interaction is also involved in regulating neuronal injury after TBI. ...

Dynamic regulation of Homer binding to group I mGluRs by Preso1 and converging kinase cascades
  • Citing Article
  • February 2017

Journal of Pharmacology and Experimental Therapeutics

... The high-drinking-in-the-dark (HDID) replicate lines of mice (HDID-1 and HDID-2) have been selectively bred to achieve high blood alcohol levels (BALs) in the drinking-in-the-dark (DID) task 8 and now represent two unique genetic models of risk for binge-like ethanol intake. 9,10 These mice have since been used as animal models to probe the genetic, molecular and neural underpinnings of AUD [11][12][13][14][15][16][17][18] and to screen potential pharmacotherapies for AUD. 11,[19][20][21][22] However, little is known about the extent to which the HDID lines of mice consume other addictive drugs, which would provide information about shared genetic influences for risk of comorbid drug use. ...

Protein Kinase C Epsilon Activity in the Nucleus Accumbens and Central Nucleus of the Amygdala Mediates Binge Alcohol Consumption
  • Citing Article
  • March 2015

Biological Psychiatry

... In addition to the regulatory effect of endogenous Homer1a, phosphorylation of interacting binding sites is an alternative mechanism underlying the regulation of the mGluR-Homer complex structure and function [20]. Phosphorylation of the Homer-binding site of group I mGluRs enhances the mGluR-Homer interaction [21,43]. In contrast, phosphorylation of Homer The extent of brain edema was reduced in Preso -/mice after TBI. ...

A Prolyl-Isomerase Mediates Dopamine-Dependent Plasticity and Cocaine Motor Sensitization
  • Citing Article
  • August 2013

Cell

... The medial prefrontal cortex (mPFC), which is primarily known for its prominent role in attention and goal-directed behavior 9 , provides top-down regulation of sensory and affective processes 10 , including inhibition of both sensory and affective nociceptive signals by descending projections to various brain and spinal cord regions [11][12][13] . In both human subjects and rodent models, the mPFC undergoes structural as well as functional changes in chronic pain states [14][15][16][17][18][19][20][21] , which are reflected by cognitive deficits and decreased attention (for review see 22 ). In line with this, attention directed to painful stimuli increases reported pain intensities 23 and attention distraction reduces subjective pain intensities in human healthy volunteers 24,25 , thereby suggesting a strong impact of attention on the chronification of pain. ...

Homers at the Interface between Reward and Pain