Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Clin Chem Lab Med ; 61(7): 1245-1254, 2023 06 27.
Article in English | MEDLINE | ID: mdl-36709509

ABSTRACT

OBJECTIVES: Neurofilament light chain (NfL) concentration in blood is a biomarker of neuro-axonal injury in the nervous system and there now exist several assays with high enough sensitivity to measure NfL in serum and plasma. There is a need for harmonization with the goal of creating a certified reference material (CRM) for NfL and an early step in such an effort is to determine the best matrix for the CRM. This is done in a commutability study and here the results of the first one for NfL in blood is presented. METHODS: Forty paired individual serum and plasma samples were analyzed for NfL on four different analytical platforms. Neat and differently spiked serum and plasma were evaluated for their suitability as a CRM using the difference in bias approach. RESULTS: The correlation between the different platforms with regards to measured NfL concentrations were very high (Spearman's ρ≥0.96). Samples spiked with cerebrospinal fluid (CSF) showed higher commutability compared to samples spiked with recombinant human NfL protein and serum seems to be a better choice than plasma as the matrix for a CRM. CONCLUSIONS: The results from this first commutability study on NfL in serum/plasma showed that it is feasible to create a CRM for NfL in blood and that spiking should be done using CSF rather than with recombinant human NfL protein.


Subject(s)
Intermediate Filaments , Neurofilament Proteins , Humans , Serum , Plasma , Reference Standards , Biomarkers , Recombinant Proteins
2.
Elife ; 102021 10 18.
Article in English | MEDLINE | ID: mdl-34658339

ABSTRACT

Ionotropic neurotransmitter receptors at postsynapses mediate fast synaptic transmission upon binding of the neurotransmitter. Post- and trans-synaptic mechanisms through cytosolic, membrane, and secreted proteins have been proposed to localize neurotransmitter receptors at postsynapses. However, it remains unknown which mechanism is crucial to maintain neurotransmitter receptors at postsynapses. In this study, we ablated excitatory or inhibitory neurons in adult mouse brains in a cell-autonomous manner. Unexpectedly, we found that excitatory AMPA receptors remain at the postsynaptic density upon ablation of excitatory presynaptic terminals. In contrast, inhibitory GABAA receptors required inhibitory presynaptic terminals for their postsynaptic localization. Consistent with this finding, ectopic expression at excitatory presynapses of neurexin-3 alpha, a putative trans-synaptic interactor with the native GABAA receptor complex, could recruit GABAA receptors to contacted postsynaptic sites. These results establish distinct mechanisms for the maintenance of excitatory and inhibitory postsynaptic receptors in the mature mammalian brain.


Subject(s)
Receptors, AMPA/metabolism , Receptors, GABA-A/metabolism , Synapses/physiology , Synaptic Transmission/physiology , Animals , Mice , Mice, Transgenic , Post-Synaptic Density/metabolism , Presynaptic Terminals/metabolism
3.
Cell Rep ; 33(11): 108511, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33326786

ABSTRACT

Early-life adversity (ELA) is associated with lifelong memory deficits, yet the responsible mechanisms remain unclear. We impose ELA by rearing rat pups in simulated poverty, assess hippocampal memory, and probe changes in gene expression, their transcriptional regulation, and the consequent changes in hippocampal neuronal structure. ELA rats have poor hippocampal memory and stunted hippocampal pyramidal neurons associated with ~140 differentially expressed genes. Upstream regulators of the altered genes include glucocorticoid receptor and, unexpectedly, the transcription factor neuron-restrictive silencer factor (NRSF/REST). NRSF contributes critically to the memory deficits because blocking its function transiently following ELA rescues spatial memory and restores the dendritic arborization of hippocampal pyramidal neurons in ELA rats. Blocking NRSF function in vitro augments dendritic complexity of developing hippocampal neurons, suggesting that NRSF represses genes involved in neuronal maturation. These findings establish important, surprising contributions of NRSF to ELA-induced transcriptional programming that disrupts hippocampal maturation and memory function.


Subject(s)
Hippocampus/immunology , Memory Disorders/immunology , Neurons/metabolism , Transcription Factors/immunology , Animals , Disease Models, Animal , Humans , Rats
4.
Neuron ; 97(3): 479-481, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29420928

ABSTRACT

There are significant challenges in identifying receptor-specific functional interactors in vivo. In this issue of Neuron, Ge et al. (2018) identify a novel GABAA receptor (GABAAR)-interacting protein, Clptm1, that regulates forward trafficking of GABAARs and inhibitory transmission.


Subject(s)
Proteomics , Receptors, GABA-A , Neurons , Protein Transport , gamma-Aminobutyric Acid
5.
J Neurosci ; 37(14): 3799-3812, 2017 04 05.
Article in English | MEDLINE | ID: mdl-28275159

ABSTRACT

In a subset of children experiencing prolonged febrile seizures (FSs), the most common type of childhood seizures, cognitive outcomes are compromised. However, the underlying mechanisms are unknown. Here we identified significant, enduring spatial memory problems in male rats following experimental prolonged FS (febrile status epilepticus; eFSE). Remarkably, these deficits were abolished by transient, post hoc interference with the chromatin binding of the transcriptional repressor neuron restrictive silencing factor (NRSF or REST). This transcriptional regulator is known to contribute to neuronal differentiation during development and to programmed gene expression in mature neurons. The mechanisms of the eFSE-provoked memory problems involved complex disruption of memory-related hippocampal oscillations recorded from CA1, likely resulting in part from impairments of dendritic filtering of cortical inputs as well as abnormal synaptic function. Accordingly, eFSE provoked region-specific dendritic loss in the hippocampus, and aberrant generation of excitatory synapses in dentate gyrus granule cells. Blocking NRSF transiently after eFSE prevented granule cell dysmaturation, restored a functional balance of γ-band network oscillations, and allowed treated eFSE rats to encode and retrieve spatial memories. Together, these studies provide novel insights into developing networks that underlie memory, the mechanisms by which early-life seizures influence them, and the means to abrogate the ensuing cognitive problems.SIGNIFICANCE STATEMENT Whereas seizures have been the central focus of epilepsy research, they are commonly accompanied by cognitive problems, including memory impairments that contribute to poor quality of life. These deficits often arise before the onset of spontaneous seizures, or independent from them, yet the mechanisms involved are unclear. Here, using a rodent model of common developmental seizures that provoke epilepsy in a subset of individuals, we identify serious consequent memory problems. We uncover molecular, cellular, and circuit-level mechanisms that underlie these deficits and successfully abolish them by targeted therapeutic interventions. These findings may be important for understanding and preventing cognitive problems in individuals suffering long febrile seizures.


Subject(s)
Memory Disorders/metabolism , Memory Disorders/physiopathology , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/metabolism , Seizures, Febrile/metabolism , Seizures, Febrile/physiopathology , Animals , Animals, Newborn , Hippocampus/growth & development , Hippocampus/metabolism , Hippocampus/physiopathology , Male , Memory Disorders/etiology , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Seizures, Febrile/complications
6.
Cell Rep ; 16(2): 531-544, 2016 07 12.
Article in English | MEDLINE | ID: mdl-27346345

ABSTRACT

Synaptic communication between neurons requires the precise localization of neurotransmitter receptors to the correct synapse type. Kainate-type glutamate receptors restrict synaptic localization that is determined by the afferent presynaptic connection. The mechanisms that govern this input-specific synaptic localization remain unclear. Here, we examine how subunit composition and specific subunit domains contribute to synaptic localization of kainate receptors. The cytoplasmic domain of the GluK2 low-affinity subunit stabilizes kainate receptors at synapses. In contrast, the extracellular domain of the GluK4/5 high-affinity subunit synergistically controls the synaptic specificity of kainate receptors through interaction with C1q-like proteins. Thus, the input-specific synaptic localization of the native kainate receptor complex involves two mechanisms that underlie specificity and stabilization of the receptor at synapses.


Subject(s)
Protein Subunits/physiology , Receptors, Kainic Acid/physiology , Synapses/metabolism , Animals , Cerebellum/cytology , Cerebellum/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Mice, Transgenic , Protein Domains , Protein Stability , Protein Subunits/chemistry , Protein Transport , Receptors, Kainic Acid/chemistry
7.
J Biol Chem ; 289(9): 5889-903, 2014 Feb 28.
Article in English | MEDLINE | ID: mdl-24403084

ABSTRACT

The actin-binding protein filamin A (FLNa) regulates neuronal migration during development, yet its roles in the mature brain remain largely obscure. Here, we probed the effects of FLNa on the regulation of ion channels that influence neuronal properties. We focused on the HCN1 channels that conduct Ih, a hyperpolarization-activated current crucial for shaping intrinsic neuronal properties. Whereas regulation of HCN1 channels by FLNa has been observed in melanoma cell lines, its physiological relevance to neuronal function and the underlying cellular pathways that govern this regulation remain unknown. Using a combination of mutational, pharmacological, and imaging approaches, we find here that FLNa facilitates a selective and reversible dynamin-dependent internalization of HCN1 channels in HEK293 cells. This internalization is accompanied by a redistribution of HCN1 channels on the cell surface, by accumulation of the channels in endosomal compartments, and by reduced Ih density. In hippocampal neurons, expression of a truncated dominant-negative FLNa enhances the expression of native HCN1. Furthermore, acute abrogation of HCN1-FLNa interaction in neurons, with the use of decoy peptides that mimic the FLNa-binding domain of HCN1, abolishes the punctate distribution of HCN1 channels in neuronal cell bodies, augments endogenous Ih, and enhances the rebound-response ("voltage-sag") of the neuronal membrane to transient hyperpolarizing events. Together, these results support a major function of FLNa in modulating ion channel abundance and membrane trafficking in neurons, thereby shaping their biophysical properties and function.


Subject(s)
Dynamins/metabolism , Filamins/metabolism , Hippocampus/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Neurons/metabolism , Potassium Channels/metabolism , Animals , Dynamins/genetics , Filamins/genetics , Hippocampus/cytology , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Membrane Potentials/physiology , Mice , Neurons/cytology , Potassium Channels/genetics , Rats , Rats, Sprague-Dawley
8.
Epilepsy Behav ; 26(3): 253-60, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23219411

ABSTRACT

The highest incidence of seizures in humans occurs during the first year of life. The high susceptibility to seizures in neonates and infants is paralleled by animal studies showing a high propensity to seizures during early life. The immature brain is highly susceptible to seizures because of an imbalance of excitation and inhibition. While the primary outcome determinant of early-life seizures is etiology, there is evidence that seizures which are frequent or prolonged can result in long-term adverse consequences, and there is a consensus that recurrent early-life seizures should be treated. Unfortunately, seizures in many neonates and children remain refractory to therapy. There is therefore a pressing need for new seizure drugs as well as antiepileptic targets in children. In this review, we focus on mechanisms of early-life seizures, such as hypoxia-ischemia, and novel molecular targets, including the hyperpolarization-activated cyclic nucleotide-gated channels.


Subject(s)
Biomedical Research , Epilepsy/metabolism , Epilepsy/therapy , Pediatrics , Brain/drug effects , Brain/growth & development , Brain/metabolism , Brain/physiopathology , Cyclic Nucleotide-Gated Cation Channels/metabolism , Humans , Potassium Channels/metabolism
9.
J Comp Neurol ; 520(13): 3013-34, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22434607

ABSTRACT

Filamin A (FLNa) is an actin-binding protein that regulates cell motility, adhesion, and elasticity by cross-linking filamentous actin. Additional roles of FLNa include regulation of protein trafficking and surface expression. Although the functions of FLNa during brain development are well studied, little is known on its expression, distribution, and function in the adult brain. Here we characterize in detail the neuroanatomical distribution and subcellular localization of FLNa in the mature rat brain, by using two antisera directed against epitopes at either the N' or the C' terminus of the protein, further validated by mRNA expression. FLNa was widely and selectively expressed throughout the brain, and the intensity of immunoreactivity was region dependent. The most intensely FLNa-labeled neurons were found in discrete neuronal systems, including basal forebrain structures, anterior nuclear group of thalamus, and hypothalamic parvocellular neurons. Pyramidal neurons in neocortex and hippocampus and magnocellular cells in basolateral amygdaloid nucleus were also intensely FLNa immunoreactive, and strong FLNa labeling was evident in the pontine and medullary raphe nuclei and in sensory and spinal trigeminal nuclei. The subcellular localization of FLNa was evaluated in situ as well as in primary hippocampal neurons. Punctate expression was found in somata and along the dendritic shaft, but FLNa was not detected in dendritic spines. These subcellular distribution patterns were recapitulated in hippocampal and neocortical pyramidal neurons in vivo. The characterization of the expression and subcellular localization of FLNa may provide new clues to the functional roles of this cytoskeletal protein in the adult brain.


Subject(s)
Brain/metabolism , Contractile Proteins/biosynthesis , Microfilament Proteins/biosynthesis , Neurons/metabolism , Animals , Blotting, Western , Filamins , Immunohistochemistry , In Situ Hybridization , Rats , Rats, Sprague-Dawley
10.
Curr Opin Neurobiol ; 21(6): 873-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21782415

ABSTRACT

Epilepsy is the third most common brain disorder and affects millions of people. Epilepsy is characterized by the occurrence of spontaneous seizures, that is, bursts of synchronous firing of large populations of neurons. These are believed to result from abnormal regulation of neuronal excitability that favors hypersynchrony. Among the intrinsic conductances that govern neuronal excitability, the hyperpolarization-activated current (I(h)) plays complex and important roles in the fine-tuning of both cellular and network activity. Not surprisingly, dysregulation of I(h) and/or of its conducting ion-channels (HCN) has been strongly implicated in various experimental models of epilepsy, as well as in human epilepsy. Here we provide an overview of recent findings on the distinct physiological roles played by I(h) in specific contexts, and the cellular mechanisms that underlie these functions, including the subunit make-up of the channels. We further discuss current knowledge of dysregulation of I(h) and HCN channels in epilepsy in light of the multifaceted functions of I(h) in the brain.


Subject(s)
Brain/metabolism , Brain/physiopathology , Cyclic Nucleotide-Gated Cation Channels/metabolism , Epilepsy/metabolism , Epilepsy/physiopathology , Potassium Channels/metabolism , Animals , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels
11.
J Biol Chem ; 286(23): 20823-34, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21504900

ABSTRACT

Ion channel trafficking and gating are often influenced by interactions with auxiliary subunits. Tetratricopeptide repeat-containing Rab8b-interacting protein (TRIP8b) is an auxiliary subunit for neuronal hyperpolarization-activated cyclic nucleotide-gated (HCN) channels. TRIP8b interacts directly with two distinct sites of HCN channel pore-forming subunits to control channel trafficking and gating. Here we use mutagenesis combined with electrophysiological studies to define and distinguish the functional importance of the HCN/TRIP8b interaction sites. Interaction with the last three amino acids of the HCN1 C terminus governed the effect of TRIP8b on channel trafficking, whereas TRIP8b interaction with the HCN1 cyclic nucleotide binding domain (CNBD) affected trafficking and gating. Biochemical studies revealed that direct interaction between TRIP8b and the HCN1 CNBD was disrupted by cAMP and that TRIP8b binding to the CNBD required an arginine residue also necessary for cAMP binding. In accord, increasing cAMP levels in cells antagonized the up-regulation of HCN1 channels mediated by a TRIP8b construct binding the CNBD exclusively. These data illustrate the distinct roles of the two TRIP8b-HCN interaction domains and suggest that TRIP8b and cAMP may directly compete for binding the HCN CNBD to control HCN channel gating, kinetics, and trafficking.


Subject(s)
Cyclic AMP/metabolism , Cyclic Nucleotide-Gated Cation Channels/metabolism , Ion Channel Gating/physiology , Membrane Proteins/metabolism , Potassium Channels/metabolism , Animals , Cyclic AMP/genetics , Cyclic Nucleotide-Gated Cation Channels/genetics , HEK293 Cells , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Membrane Proteins/genetics , Mice , Peroxins , Potassium Channels/genetics , Protein Binding , Protein Transport/physiology , Rats , Up-Regulation/physiology
12.
J Biol Chem ; 285(19): 14724-36, 2010 May 07.
Article in English | MEDLINE | ID: mdl-20215108

ABSTRACT

Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels mediate the hyperpolarization-activated current I(h) and thus play important roles in the regulation of brain excitability. The subcellular distribution pattern of the HCN channels influences the effects that they exert on the properties and activity of neurons. However, little is known about the mechanisms that control HCN channel trafficking to subcellular compartments or that regulate their surface expression. Here we studied the dynamics of HCN channel trafficking in hippocampal neurons using dissociated cultures coupled with time lapse imaging of fluorophore-fused HCN channels. HCN1-green fluorescence protein (HCN1-GFP) channels resided in vesicle-like organelles that moved in distinct patterns along neuronal dendrites, and these properties were isoform-specific. HCN1 trafficking required intact actin and tubulin and was rapidly inhibited by activation of either NMDA or AMPA-type ionotropic glutamate receptors in a calcium-dependent manner. Glutamate-induced inhibition of the movement of HCN1-GFP-expressing puncta was associated with increased surface expression of both native and transfected HCN1 channels, and this surface expression was accompanied by augmented I(h). Taken together, the results reveal the highly dynamic nature of HCN1 channel trafficking in hippocampal neurons and provide a novel potential mechanism for rapid regulation of I(h), and hence of neuronal properties, via alterations of HCN1 trafficking and surface expression.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels/metabolism , Dendrites/metabolism , Hippocampus/metabolism , Neurons/metabolism , Potassium Channels/metabolism , Actins/metabolism , Animals , Animals, Newborn , Biotinylation , Cells, Cultured , Cyclic Nucleotide-Gated Cation Channels/genetics , Electrophysiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hippocampus/cytology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Immunoenzyme Techniques , Ion Channel Gating , Microtubules/metabolism , Neurons/cytology , Potassium Channels/genetics , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
14.
J Neurosci ; 29(19): 6250-65, 2009 May 13.
Article in English | MEDLINE | ID: mdl-19439603

ABSTRACT

Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels (h channels) are the molecular basis for the current, I(h), which contributes crucially to intrinsic neuronal excitability. The subcellular localization and biophysical properties of h channels govern their function, but the mechanisms controlling these characteristics, and especially the potential role of auxiliary subunits or other binding proteins, remain unclear. We focused on TRIP8b, an h channel-interacting protein that colocalizes with HCN1 in cortical and hippocampal pyramidal neuron dendrites, and found that it exists in multiple alternative splice variants with distinct effects on h channel trafficking and function. The developmentally regulated splice variants of TRIP8b all shared dual, C terminus-located interaction sites with HCN1. When coexpressed with HCN1 in heterologous cells individual TRIP8b isoforms similarly modulated gating of I(h), causing a hyperpolarizing shift in voltage dependence of channel activation, but differentially upregulated or downregulated I(h) current density and HCN1 surface expression. In hippocampal neurons, coexpression of TRIP8b isoforms with HCN1 produced isoform-specific changes of HCN1 localization. Interestingly, the TRIP8b isoforms most abundant in the brain are those predicted to enhance h channel surface expression. Indeed, shRNA knockdown of TRIP8b in hippocampal neurons significantly reduced native I(h). Thus, although TRIP8b exists in multiple splice isoforms, our data suggest that the predominant role of this protein in brain is to promote h channel surface expression and enhance I(h). Because I(h) expression is altered in models of several diseases, including temporal lobe epilepsy, TRIP8b may play a role in both normal neuronal function and in aberrant neuronal excitability associated with neurological disease.


Subject(s)
Brain/physiology , Cyclic Nucleotide-Gated Cation Channels/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neurons/physiology , Potassium Channels/metabolism , Alternative Splicing , Animals , Base Sequence , Cell Line , Cells, Cultured , Gene Knockdown Techniques , Hippocampus/physiology , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Membrane Potentials/physiology , Molecular Sequence Data , Protein Isoforms , Rats , Rats, Sprague-Dawley
15.
J Physiol ; 586(15): 3629-38, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18566001

ABSTRACT

Natively expressed serotonin 5-HT(3) receptors typically possess a negative-slope conductance region in their I-V curve, due to a voltage-dependent block by external Ca(2+) ions. However, in almost all studies performed with heterologously expressed 5-HT(3) receptors, this feature was not observed. Here we show that mere addition of ATP to the pipette solution is sufficient to reliably observe a voltage-dependent block in homomeric (h5-HT(3A)) and heteromeric (h5-HT(3AB)) receptors expressed in HEK293 cells. A similar block was observed with a plethora of molecules containing a phosphate moiety, thus excluding a role of phosphorylation. A substitution of three arginines in the intracellular vestibule of 5-HT(3A) with their counterpart residues from the 5-HT(3B) subunit (RRR-QDA) was previously shown to dramatically increase single channel conductance. We find this mutant to have a linear I-V curve that is unaffected by the presence of ATP, with a fractional Ca(2+) current (Pf%) that is reduced (1.8 +/- 0.2%) compared to that of the homomeric receptor (4.1 +/- 0.2%), and similar to that of the heteromeric form (2.0 +/- 0.3%). Moreover, whereas ATP decreased the Pf% of the homomeric receptor, this was not observed with the RRR-QDA mutant. Finally, ATP was found to be critical for voltage-dependent channel block also in hippocampal interneurons that natively express 5-HT(3) receptors. Taken together, our results indicate a novel mechanism by which ATP, and similar molecules, modulate 5-HT(3) receptors via interactions with the intracellular vestibule of the receptor.


Subject(s)
Calcium/pharmacology , Phosphates/metabolism , Serotonin 5-HT3 Receptor Antagonists , Adenosine Triphosphate/pharmacology , Animals , Cell Line , Gene Expression Regulation , Hippocampus/cytology , Humans , Interneurons/metabolism , Protein Subunits , Rats , Receptors, Serotonin, 5-HT3/genetics , Receptors, Serotonin, 5-HT3/metabolism
16.
Epilepsy Curr ; 7(5): 136-7, 2007.
Article in English | MEDLINE | ID: mdl-17998975
SELECTION OF CITATIONS
SEARCH DETAIL
...