Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Curr Biol ; 30(11): 2068-2077.e4, 2020 06 08.
Article in English | MEDLINE | ID: mdl-32359429

ABSTRACT

African naked mole-rats were likely the first mammals to evolve eusociality, and thus required adaptations to conserve energy and tolerate the low oxygen (O2) and high carbon dioxide (CO2) of a densely populated fossorial nest. As hypercapnia is known to suppress neuronal activity, we studied whether naked mole-rats might demonstrate energy savings in GABAergic inhibition. Using whole-colony behavioral monitoring of captive naked mole-rats, we found a durable nest, characterized by high CO2 levels, where all colony members spent the majority of their time. Analysis of the naked mole-rat genome revealed, uniquely among mammals, a histidine point variation in the neuronal potassium-chloride cotransporter 2 (KCC2). A histidine missense substitution mutation at this locus in the human ortholog of KCC2, found previously in patients with febrile seizures and epilepsy, has been demonstrated to diminish neuronal Cl- extrusion capacity, and thus impairs GABAergic inhibition. Seizures were observed, without pharmacological intervention, in adult naked mole-rats exposed to a simulated hyperthermic surface environment, causing systemic hypocapnic alkalosis. Consistent with the diminished function of KCC2, adult naked mole-rats demonstrate a reduced efficacy of inhibition that manifests as triggering of seizures at room temperature by the GABAA receptor (GABAAR) positive allosteric modulator diazepam. These seizures are blocked in the presence of nest-like levels of CO2 and likely to be mediated through GABAAR activity, based on in vitro recordings. Thus, altered GABAergic inhibition adds to a growing list of adaptations in the naked mole-rat and provides a plausible proximate mechanism for nesting behavior, where a return to the colony nest restores GABA-mediated inhibition.


Subject(s)
Carbon Dioxide/metabolism , Disease Susceptibility/veterinary , Mole Rats , Receptors, GABA-A/metabolism , Rodent Diseases/physiopathology , Seizures/veterinary , Animals , Disease Susceptibility/etiology , Disease Susceptibility/metabolism , Female , Male , Rodent Diseases/genetics , Seizures/genetics , Seizures/physiopathology
2.
Acta Physiol (Oxf) ; 229(3): e13467, 2020 07.
Article in English | MEDLINE | ID: mdl-32174009

ABSTRACT

AIM: To study brain-sparing physiological responses in a rodent model of birth asphyxia which reproduces the asphyxia-defining systemic hypoxia and hypercapnia. METHODS: Steady or intermittent asphyxia was induced for 15-45 minutes in anaesthetized 6- and 11-days old rats and neonatal guinea pigs using gases containing 5% or 9% O2 plus 20% CO2 (in N2 ). Hypoxia and hypercapnia were induced with low O2 and high CO2 respectively. Oxygen partial pressure (PO2 ) and pH were measured with microsensors within the brain and subcutaneous ("body") tissue. Blood lactate was measured after asphyxia. RESULTS: Brain and body PO2 fell to apparent zero with little recovery during 5% O2 asphyxia and 5% or 9% O2 hypoxia, and increased more than twofold during 20% CO2 hypercapnia. Unlike body PO2 , brain PO2 recovered rapidly to control after a transient fall (rat), or was slightly higher than control (guinea pig) during 9% O2 asphyxia. Asphyxia (5% O2 ) induced a respiratory acidosis paralleled by a progressive metabolic (lact)acidosis that was much smaller within than outside the brain. Hypoxia (5% O2 ) produced a brain-confined alkalosis. Hypercapnia outlasting asphyxia suppressed pH recovery and prolonged the post-asphyxia PO2 overshoot. All pH changes were accompanied by consistent shifts in the blood-brain barrier potential. CONCLUSION: Regardless of brain maturation stage, hypercapnia can restore brain PO2 and protect the brain against metabolic acidosis despite compromised oxygen availability during asphyxia. This effect extends to the recovery phase if normocapnia is restored slowly, and it is absent during hypoxia, demonstrating that exposure to hypoxia does not mimic asphyxia.


Subject(s)
Asphyxia Neonatorum , Brain/physiology , Oxygen/physiology , Animals , Disease Models, Animal , Guinea Pigs , Humans , Hydrogen-Ion Concentration , Hypercapnia , Hypoxia , Infant, Newborn , Rats
3.
EMBO Rep ; 21(4): e48880, 2020 04 03.
Article in English | MEDLINE | ID: mdl-32064760

ABSTRACT

KCC2, encoded in humans by the SLC12A5 gene, is a multifunctional neuron-specific protein initially identified as the chloride (Cl- ) extruder critical for hyperpolarizing GABAA receptor currents. Independently of its canonical function as a K-Cl cotransporter, KCC2 regulates the actin cytoskeleton via molecular interactions mediated through its large intracellular C-terminal domain (CTD). Contrary to the common assumption that embryonic neocortical projection neurons express KCC2 at non-significant levels, here we show that loss of KCC2 enhances apoptosis of late-born upper-layer cortical projection neurons in the embryonic brain. In utero electroporation of plasmids encoding truncated, transport-dead KCC2 constructs retaining the CTD was as efficient as of that encoding full-length KCC2 in preventing elimination of migrating projection neurons upon conditional deletion of KCC2. This was in contrast to the effect of a full-length KCC2 construct bearing a CTD missense mutation (KCC2R952H ), which disrupts cytoskeletal interactions and has been found in patients with neurological and psychiatric disorders, notably seizures and epilepsy. Together, our findings indicate ion transport-independent, CTD-mediated regulation of developmental apoptosis by KCC2 in migrating cortical projection neurons.


Subject(s)
Apoptosis , Epilepsy , Neurons/pathology , Symporters/genetics , Apoptosis/genetics , Chlorides/metabolism , Humans , Neurons/metabolism
4.
PLoS One ; 14(2): e0211151, 2019.
Article in English | MEDLINE | ID: mdl-30759095

ABSTRACT

The 30-amino acid peptide Y-P30 corresponds to the N-terminus of the primate-specific, sweat gland-derived dermcidin prepropeptide. Previous work has revealed that Y-P30 enhances the interaction of pleiotrophin and syndecans-2/3, and thus represents a natural ligand to study this signaling pathway. In immature neurons, Y-P30 activates the c-Src and p42/44 ERK kinase pathway, increases the amount of F-actin in axonal growth cones, and promotes neuronal survival, cell migration and axonal elongation. The action of Y-P30 on axonal growth requires syndecan-3 and heparan sulfate side chains. Whether Y-P30 has the potential to influence dendrites and dendritic protrusions has not been explored. The latter is suggested by the observations that syndecan-2 expression increases during postnatal development, that syndecan-2 becomes enriched in dendritic spines, and that overexpression of syndecan-2 in immature neurons results in a premature morphological maturation of dendritic spines. Here, analysing rat cortical pyramidal and non-pyramidal neurons in organotypic cultures, we show that Y-P30 does not alter the development of the dendritic arborization patterns. However, Y-P30 treatment decreases the density of apical, but not basal dendritic protrusions at the expense of the filopodia. Analysis of spine morphology revealed an unchanged mushroom/stubby-to-thin spine ratio and a shortening of the longest decile of dendritic protrusions. Whole-cell recordings from cortical principal neurons in dissociated cultures grown in the presence of Y-P30 demonstrated a decrease in the frequency of glutamatergic mEPSCs. Despite these differences in protrusion morphology and synaptic transmission, the latter likely attributable to presynaptic effects, calcium event rate and amplitude recorded in pyramidal neurons in organotypic cultures were not altered by Y-P30 treatment. Together, our data suggest that Y-P30 has the capacity to decelerate spinogenesis and to promote morphological, but not synaptic, maturation of dendritic protrusions.


Subject(s)
Dendritic Spines/metabolism , Neocortex/cytology , Peptides/metabolism , Animals , Calcium/metabolism , Cell Differentiation , Cells, Cultured , Neocortex/metabolism , Organ Culture Techniques , Patch-Clamp Techniques , Rats , Syndecan-2/metabolism
5.
Cell Rep ; 26(5): 1073-1081.e3, 2019 01 29.
Article in English | MEDLINE | ID: mdl-30699338

ABSTRACT

It is generally thought that hippocampal neurons of perinatal rats and mice lack transport-functional K-Cl cotransporter KCC2, and that Cl- regulation is dominated by Cl- uptake via the Na-K-2Cl cotransporter NKCC1. Here, we demonstrate a robust enhancement of spontaneous hippocampal network events (giant depolarizing potentials [GDPs]) by the KCC2 inhibitor VU0463271 in neonatal rats and late-gestation, wild-type mouse embryos, but not in their KCC2-null littermates. VU0463271 increased the depolarizing GABAergic synaptic drive onto neonatal CA3 pyramidal neurons, increasing their spiking probability and synchrony during the rising phase of a GDP. Our data indicate that Cl- extrusion by KCC2 is involved in modulation of GDPs already at their developmental onset during the perinatal period in mice and rats.


Subject(s)
Chlorides/metabolism , Hippocampus/metabolism , Symporters/metabolism , Action Potentials/drug effects , Animals , Animals, Newborn , Mice, Inbred ICR , Pyramidal Cells/metabolism , Rats, Wistar , gamma-Aminobutyric Acid/pharmacology , K Cl- Cotransporters
7.
Neuropharmacology ; 143: 186-204, 2018 12.
Article in English | MEDLINE | ID: mdl-30248303

ABSTRACT

Based on the potential role of Na-K-Cl cotransporters (NKCCs) in epileptic seizures, the loop diuretic bumetanide, which blocks the NKCC1 isoforms NKCC1 and NKCC2, has been tested as an adjunct with phenobarbital to suppress seizures. However, because of its physicochemical properties, bumetanide only poorly penetrates through the blood-brain barrier. Thus, concentrations needed to inhibit NKCC1 in hippocampal and neocortical neurons are not reached when using doses (0.1-0.5 mg/kg) in the range of those approved for use as a diuretic in humans. This prompted us to search for a bumetanide derivative that more easily penetrates into the brain. Here we show that bumepamine, a lipophilic benzylamine derivative of bumetanide, exhibits much higher brain penetration than bumetanide and is more potent than the parent drug to potentiate phenobarbital's anticonvulsant effect in two rodent models of chronic difficult-to-treat epilepsy, amygdala kindling in rats and the pilocarpine model in mice. However, bumepamine suppressed NKCC1-dependent giant depolarizing potentials (GDPs) in neonatal rat hippocampal slices much less effectively than bumetanide and did not inhibit GABA-induced Ca2+ transients in the slices, indicating that bumepamine does not inhibit NKCC1. This was substantiated by an oocyte assay, in which bumepamine did not block NKCC1a and NKCC1b after either extra- or intracellular application, whereas bumetanide potently blocked both variants of NKCC1. Experiments with equilibrium dialysis showed high unspecific tissue binding of bumetanide in the brain, which, in addition to its poor brain penetration, further reduces functionally relevant brain concentrations of this drug. These data show that CNS effects of bumetanide previously thought to be mediated by NKCC1 inhibition can also be achieved by a close derivative that does not share this mechanism. Bumepamine has several advantages over bumetanide for CNS targeting, including lower diuretic potency, much higher brain permeability, and higher efficacy to potentiate the anti-seizure effect of phenobarbital.


Subject(s)
Anticonvulsants/pharmacology , Benzylamines/pharmacology , Bumetanide/pharmacology , Phenobarbital/pharmacology , Animals , Anticonvulsants/chemical synthesis , Anticonvulsants/chemistry , Anticonvulsants/pharmacokinetics , Benzylamines/chemical synthesis , Benzylamines/chemistry , Benzylamines/pharmacokinetics , Brain/drug effects , Brain/metabolism , Bumetanide/analogs & derivatives , Bumetanide/chemistry , Bumetanide/pharmacokinetics , Drug Evaluation, Preclinical , Drug Synergism , Epilepsy/drug therapy , Epilepsy/metabolism , Female , Mice , Oocytes , Phenobarbital/pharmacokinetics , Rats, Wistar , Seizures/drug therapy , Seizures/metabolism , Sodium Potassium Chloride Symporter Inhibitors/chemistry , Sodium Potassium Chloride Symporter Inhibitors/pharmacokinetics , Sodium Potassium Chloride Symporter Inhibitors/pharmacology , Solute Carrier Family 12, Member 2/metabolism , Tissue Culture Techniques , Xenopus laevis
8.
Proc Natl Acad Sci U S A ; 114(50): E10819-E10828, 2017 12 12.
Article in English | MEDLINE | ID: mdl-29183979

ABSTRACT

During birth in mammals, a pronounced surge of fetal peripheral stress hormones takes place to promote survival in the transition to the extrauterine environment. However, it is not known whether the hormonal signaling involves central pathways with direct protective effects on the perinatal brain. Here, we show that arginine vasopressin specifically activates interneurons to suppress spontaneous network events in the perinatal hippocampus. Experiments done on the altricial rat and precocial guinea pig neonate demonstrated that the effect of vasopressin is not dependent on the level of maturation (depolarizing vs. hyperpolarizing) of postsynaptic GABAA receptor actions. Thus, the fetal mammalian brain is equipped with an evolutionarily conserved mechanism well-suited to suppress energetically expensive correlated network events under conditions of reduced oxygen supply at birth.


Subject(s)
Brain/embryology , Interneurons/physiology , Vasopressins/physiology , Animals , Brain/growth & development , Evoked Potentials , Female , Guinea Pigs , Hippocampus/embryology , Hippocampus/growth & development , Hippocampus/physiology , Male , Nerve Net/physiology , Parturition , Rats , Rats, Wistar , gamma-Aminobutyric Acid/metabolism
9.
Anesthesiology ; 126(5): 855-867, 2017 05.
Article in English | MEDLINE | ID: mdl-28301408

ABSTRACT

BACKGROUND: General anesthetics potentiating γ-aminobutyric acid (GABA)-mediated signaling are known to induce a persistent decrement in excitatory synapse number in the cerebral cortex when applied during early postnatal development, while an opposite action is produced at later stages. Here, the authors test the hypothesis that the effect of general anesthetics on synaptogenesis depends upon the efficacy of GABA receptor type A (GABAA)-mediated inhibition controlled by the developmental up-regulation of the potassium-chloride (K-Cl) cotransporter 2 (KCC2). METHODS: In utero electroporation of KCC2 was used to prematurely increase the efficacy of (GABAA)-mediated inhibition in layer 2/3 pyramidal neurons in the immature rat somatosensory cortex. Parallel experiments with expression of the inward-rectifier potassium channel Kir2.1 were done to reduce intrinsic neuronal excitability. The effects of these genetic manipulations (n = 3 to 4 animals per experimental group) were evaluated using iontophoretic injection of Lucifer Yellow (n = 8 to 12 cells per animal). The total number of spines analyzed per group ranged between 907 and 3,371. RESULTS: The authors found a robust effect of the developmental up-regulation of KCC2-mediated Cl transport on the age-dependent action of propofol on dendritic spines. Premature expression of KCC2, unlike expression of a transport-inactive KCC2 variant, prevented a propofol-induced decrease in spine density. In line with a reduction in neuronal excitability, the above result was qualitatively replicated by overexpression of Kir2.1. CONCLUSIONS: The KCC2-dependent developmental increase in the efficacy of GABAA-mediated inhibition is a major determinant of the age-dependent actions of propofol on dendritic spinogenesis.


Subject(s)
Dendritic Spines/drug effects , Dendritic Spines/metabolism , Propofol/pharmacology , Symporters/drug effects , Symporters/metabolism , Up-Regulation/drug effects , Animals , Electroporation , Female , Hypnotics and Sedatives/pharmacology , Neurons/drug effects , Neurons/metabolism , Pregnancy , Rats , Rats, Wistar , Receptors, GABA/drug effects , Somatosensory Cortex/drug effects , Somatosensory Cortex/embryology , Somatosensory Cortex/metabolism , K Cl- Cotransporters
10.
Epilepsia ; 57(1): e1-6, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26547277

ABSTRACT

Febrile seizures are the most common type of convulsive events in children. It is generally assumed that the generalization of these seizures is a result of brainstem invasion by the initial limbic seizure activity. Using precollicular transection in 13-day-old rats to isolate the forebrain from the brainstem, we demonstrate that the forebrain is not required for generation of tonic-clonic convulsions induced by hyperthermia or kainate. Compared with sham-operated littermate controls, latency to onset of convulsions in both models was significantly shorter in pups that had undergone precollicular transection, indicating suppression of the brainstem seizure network by the forebrain in the intact animal. We have shown previously that febrile seizures are precipitated by hyperthermia-induced respiratory alkalosis. Here, we show that triggering of hyperthermia-induced hyperventilation and consequent convulsions in transected animals are blocked by diazepam. The present data suggest that the role of endogenous brainstem activity in triggering tonic-clonic seizures should be re-evaluated in standard experimental models of limbic seizures. Our work sheds new light on the mechanisms that generate febrile seizures in children and, therefore, on how they might be treated.


Subject(s)
Hyperthermia, Induced/adverse effects , Prosencephalon/physiology , Seizures/etiology , Animals , Animals, Newborn , Anticonvulsants/therapeutic use , Body Temperature/physiology , Denervation , Diazepam/therapeutic use , Disease Models, Animal , Excitatory Amino Acid Agonists/toxicity , Kainic Acid/toxicity , Male , Rats , Rats, Wistar , Seizures/drug therapy , Time Factors
11.
Cereb Cortex ; 26(12): 4574-4589, 2016 12.
Article in English | MEDLINE | ID: mdl-26428952

ABSTRACT

Work on rodents demonstrated that steep upregulation of KCC2, a neuron-specific Cl- extruder of cation-chloride cotransporter (CCC) family, commences in supraspinal structures at around birth, leading to establishment of hyperpolarizing GABAergic responses. We describe spatiotemporal expression profiles of the entire CCC family in human brain. KCC2 mRNA was observed already at 10th postconceptional week (PCW) in amygdala, cerebellum, and thalamus. KCC2-immunoreactive (KCC2-ir) neurons were abundant in subplate at 18 PCW. By 25 PCW, numerous subplate and cortical plate neurons became KCC2-ir. The mRNA expression profiles of α- and ß-isoforms of Na-K ATPase, which fuels cation-chloride cotransport, as well of tropomyosin receptor kinase B (TrkB), which promotes developmental upregulation of KCC2, were consistent with data from studies on rodents about their interactions with KCC2. Thus, in human brain, expression of KCC2 and its functionally associated proteins begins in early fetal period. Our work facilitates translation of results on CCC functions from animal studies to human and refutes the view that poor efficacy of anticonvulsants in the term human neonate is attributable to the lack of KCC2. We propose that perinatally low threshold for activation of Ca2+-dependent protease calpain renders neonates susceptible to downregulation of KCC2 by traumatic events, such as perinatal hypoxia ischemia.


Subject(s)
Brain/growth & development , Brain/metabolism , Symporters/metabolism , Adult , Aged, 80 and over , Brain/cytology , Child , Gene Expression Regulation, Developmental , Humans , Immunohistochemistry , Infant , Infant, Newborn , Membrane Glycoproteins/metabolism , Microarray Analysis , Middle Aged , RNA, Messenger/metabolism , Receptor, trkB/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Solute Carrier Family 12, Member 2/metabolism , Young Adult , K Cl- Cotransporters
12.
Neuropharmacology ; 88: 103-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25229715

ABSTRACT

A robust increase in the functional expression of the neuronal K-Cl cotransporter KCC2 during CNS development is necessary for the emergence of hyperpolarizing ionotropic GABAergic transmission. BDNF-TrkB signaling has been implicated in the developmental up-regulation of KCC2 and, in mature animals, in fast activity-dependent down-regulation of KCC2 function following seizures and trauma. In contrast to the decrease in KCC2 expression observed in the adult hippocampus following trauma, seizures in the neonate trigger a TrkB-dependent up-regulation of neuronal Cl(-) extrusion capacity associated with enhanced surface expression of KCC2. Here, we show that this effect is transient, and impaired in the hippocampus of Bdnf(-/-) mice. Notably, however, a complete absence of BDNF does not compromise the increase in KCC2 protein or K-Cl transport functionality during neuronal development. Furthermore, we present data indicating that the functional up-regulation of KCC2 by neonatal seizures is temporally limited by calpain activity.


Subject(s)
Brain-Derived Neurotrophic Factor/deficiency , Brain-Derived Neurotrophic Factor/physiology , Hippocampus/physiopathology , Seizures/physiopathology , Symporters/metabolism , Animals , Animals, Newborn , Blotting, Western , Brain-Derived Neurotrophic Factor/genetics , Calpain/metabolism , Chlorides/metabolism , Disease Models, Animal , Hippocampus/drug effects , Kainic Acid , Mice, Knockout , Neurons/drug effects , Neurons/physiology , Status Epilepticus/physiopathology , Tissue Culture Techniques , Up-Regulation , K Cl- Cotransporters
13.
Nat Rev Neurosci ; 15(10): 637-54, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25234263

ABSTRACT

Electrical activity in neurons requires a seamless functional coupling between plasmalemmal ion channels and ion transporters. Although ion channels have been studied intensively for several decades, research on ion transporters is in its infancy. In recent years, it has become evident that one family of ion transporters, cation-chloride cotransporters (CCCs), and in particular K(+)-Cl(-) cotransporter 2 (KCC2), have seminal roles in shaping GABAergic signalling and neuronal connectivity. Studying the functions of these transporters may lead to major paradigm shifts in our understanding of the mechanisms underlying brain development and plasticity in health and disease.


Subject(s)
Brain , Central Nervous System Diseases , Neuronal Plasticity/physiology , Neurons/physiology , Sodium-Potassium-Chloride Symporters/metabolism , Animals , Brain/cytology , Brain/growth & development , Central Nervous System Diseases/genetics , Central Nervous System Diseases/metabolism , Central Nervous System Diseases/pathology , Humans , Models, Molecular , Sodium-Potassium-Chloride Symporters/genetics
14.
Epilepsia ; 55(6): 806-18, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24802699

ABSTRACT

Seizures are a common manifestation of acute neurologic insults in neonates and are often resistant to the standard antiepileptic drugs that are efficacious in children and adults. The paucity of evidence-based treatment guidelines, coupled with a rudimentary understanding of disease pathogenesis, has made the current treatment of neonatal seizures empiric and often ineffective, highlighting the need for novel therapies. Key developmental differences in γ-aminobutyric acid (GABA)ergic neurotransmission between the immature and mature brain, and trauma-induced alterations in the function of the cation-chloride cotransporters (CCCs) NKCC1 and KCC2, probably contribute to the poor efficacy of standard antiepileptic drugs used in the treatment of neonatal seizures. Although CCCs are attractive drug targets, bumetanide and other existing CCC inhibitors are suboptimal because of pharmacokinetic constraints and lack of target specificity. Newer approaches including isoform-specific NKCC1 inhibitors with increased central nervous system penetration, and direct and indirect strategies to enhance KCC2-mediated neuronal chloride extrusion, might allow therapeutic modulation of the GABAergic system for neonatal seizure treatment. A PowerPoint slide summarizing this article is available for download in the Supporting Information section here.


Subject(s)
Anticonvulsants/therapeutic use , Infant, Newborn, Diseases/drug therapy , Seizures/drug therapy , Sodium-Potassium-Chloride Symporters/drug effects , Brain/drug effects , Bumetanide/therapeutic use , Humans , Infant, Newborn , Solute Carrier Family 12, Member 2/drug effects , Symporters/drug effects , K Cl- Cotransporters
15.
Curr Opin Neurobiol ; 26: 34-41, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24650502

ABSTRACT

Concepts of epilepsy, based on a simple change in neuronal excitation/inhibition balance, have subsided in face of recent insights into the large diversity and context-dependence of signaling mechanisms at the molecular, cellular and neuronal network level. GABAergic transmission exerts both seizure-suppressing and seizure-promoting actions. These two roles are prone to short-term and long-term alterations, evident both during epileptogenesis and during individual epileptiform events. The driving force of GABAergic currents is controlled by ion-regulatory molecules such as the neuronal K-Cl cotransporter KCC2 and cytosolic carbonic anhydrases. Accumulating evidence suggests that neuronal ion regulation is highly plastic, thereby contributing to the multiple roles ascribed to GABAergic signaling during epileptogenesis and epilepsy.


Subject(s)
Brain/pathology , Epilepsy/pathology , Nerve Net/physiopathology , Neuronal Plasticity/physiology , Symporters/metabolism , gamma-Aminobutyric Acid/metabolism , Adaptation, Physiological/physiology , Animals , Brain/physiopathology , Humans , Neural Inhibition , Receptors, GABA-A/physiology , Signal Transduction , Synaptic Transmission/physiology , K Cl- Cotransporters
16.
EMBO Rep ; 15(6): 723-9, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24668262

ABSTRACT

Genetic variation in SLC12A5 which encodes KCC2, the neuron-specific cation-chloride cotransporter that is essential for hyperpolarizing GABAergic signaling and formation of cortical dendritic spines, has not been reported in human disease. Screening of SLC12A5 revealed a co-segregating variant (KCC2-R952H) in an Australian family with febrile seizures. We show that KCC2-R952H reduces neuronal Cl(-) extrusion and has a compromised ability to induce dendritic spines in vivo and in vitro. Biochemical analyses indicate a reduced surface expression of KCC2-R952H which likely contributes to the functional deficits. Our data suggest that KCC2-R952H is a bona fide susceptibility variant for febrile seizures.


Subject(s)
Dendritic Spines/pathology , Genetic Predisposition to Disease/genetics , Models, Molecular , Mutation, Missense/genetics , Neurons/metabolism , Seizures, Febrile/genetics , Symporters/genetics , Amino Acid Sequence , Animals , Australia , Blotting, Western , Chlorides/metabolism , Dendritic Spines/genetics , Humans , Mice , Mice, Inbred ICR , Microscopy, Fluorescence , Molecular Sequence Data , Pedigree , Protein Conformation , Rats , Rats, Wistar , Statistics, Nonparametric , Symporters/metabolism , K Cl- Cotransporters
17.
Trends Neurosci ; 36(12): 726-737, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24139641

ABSTRACT

The K-Cl cotransporter KCC2 establishes the low intraneuronal Cl- levels required for the hyperpolarizing inhibitory postsynaptic potentials mediated by ionotropic γ-aminobutyric acid receptors (GABAARs) and glycine receptors (GlyRs). Decreased KCC2-mediated Cl- extrusion and impaired hyperpolarizing GABAAR- and/or GlyR-mediated currents have been implicated in epilepsy, neuropathic pain, and spasticity. Recent evidence suggests that the intrinsic ion transport rate, cell surface stability, and plasmalemmal trafficking of KCC2 are rapidly and reversibly modulated by the (de)phosphorylation of critical serine, threonine, and tyrosine residues in the C terminus of this protein. Alterations in KCC2 phosphorylation have been associated with impaired KCC2 function in several neurological diseases. Targeting KCC2 phosphorylation directly or indirectly via upstream regulatory kinases might be a novel strategy to modulate GABA- and/or glycinergic signaling for therapeutic benefit.


Subject(s)
Neurons/metabolism , Symporters/metabolism , Animals , Humans , Models, Biological , Nervous System Diseases/metabolism , Nervous System Diseases/pathology , Phosphorylation , Receptors, GABA , Receptors, Glycine , Signal Transduction/physiology , K Cl- Cotransporters
18.
Neuropharmacology ; 69: 62-74, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22705273

ABSTRACT

In cortical and hippocampal neurons, cation-chloride cotransporters (CCCs) control the reversal potential (EGABA) of GABAA receptor-mediated current and voltage responses and, consequently, they modulate the efficacy of GABAergic inhibition. Two members of the CCC family, KCC2 (the major neuron-specific K-Cl cotransporter; KCC isoform 2) and NKCC1 (the Na-K-2Cl cotransporter isoform 1 which is expressed in both neurons and glial cells) have attracted much interest in studies on GABAergic signaling under both normal and pathophysiological conditions, such as epilepsy. There is tentative evidence that loop diuretic compounds such as furosemide and bumetanide may have clinically relevant antiepileptic actions, especially when administered in combination with conventional GABA-mimetic drugs such as phenobarbital. Furosemide is a non-selective inhibitor of CCCs while at low concentrations bumetanide is selective for NKCCs. Search for novel antiepileptic drugs (AEDs) is highly motivated especially for the treatment of neonatal seizures which are often resistant to, or even aggravated by conventional AEDs. This review shows that the antiepileptic effects of loop diuretics described in the pertinent literature are based on widely heterogeneous mechanisms ranging from actions on both neuronal NKCC1 and KCC2 to modulation of the brain extracellular volume fraction. A promising strategy for the development of novel CCC-blocking AEDs is based on prodrugs that are activated following their passage across the blood-brain barrier. This article is part of the Special Issue entitled 'New Targets and Approaches to the Treatment of Epilepsy'.


Subject(s)
Anticonvulsants/pharmacology , Epilepsy/drug therapy , Epilepsy/prevention & control , Sodium-Potassium-Chloride Symporters/drug effects , Symporters/drug effects , Adult , Animals , Brain/drug effects , Brain/physiopathology , Bumetanide/pharmacology , Diuretics/pharmacology , Epilepsy/physiopathology , Humans , Infant, Newborn , Receptors, GABA-A/drug effects , Receptors, GABA-A/physiology , Solute Carrier Family 12, Member 2 , gamma-Aminobutyric Acid/physiology , K Cl- Cotransporters
19.
Cereb Cortex ; 23(2): 378-88, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22345354

ABSTRACT

The neuron-specific K-Cl cotransporter, KCC2, is highly expressed in the vicinity of excitatory synapses in pyramidal neurons, and recent in vitro data suggest that this protein plays a role in the development of dendritic spines. The in vivo relevance of these observations is, however, unknown. Using in utero electroporation combined with post hoc iontophoretic injection of Lucifer Yellow, we show that premature expression of KCC2 induces a highly significant and permanent increase in dendritic spine density of layer 2/3 pyramidal neurons in the somatosensory cortex. Whole-cell recordings revealed that this increased spine density is correlated with an enhanced spontaneous excitatory activity in KCC2-transfected neurons. Precocious expression of the N-terminal deleted form of KCC2, which lacks the chloride transporter function, also increased spine density. In contrast, no effect on spine density was observed following in utero electroporation of a point mutant of KCC2 (KCC2-C568A) where both the cotransporter function and the interaction with the cytoskeleton are disrupted. Transfection of the C-terminal domain of KCC2, a region involved in the interaction with the dendritic cytoskeleton, also increased spine density. Collectively, these results demonstrate a role for KCC2 in excitatory synaptogenesis in vivo through a mechanism that is independent of its ion transport function.


Subject(s)
Dendritic Spines/metabolism , Neurogenesis/physiology , Pyramidal Cells/growth & development , Pyramidal Cells/metabolism , Symporters/metabolism , Animals , Electroporation , Immunohistochemistry , Patch-Clamp Techniques , Rats , Rats, Wistar , Somatosensory Cortex/growth & development , Somatosensory Cortex/metabolism , Transfection , K Cl- Cotransporters
20.
J Neurosci ; 32(33): 11356-64, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22895718

ABSTRACT

The K-Cl cotransporter KCC2 plays a crucial role in neuronal chloride regulation. In mature central neurons, KCC2 is responsible for the low intracellular Cl(-) concentration ([Cl(-)](i)) that forms the basis for hyperpolarizing GABA(A) receptor-mediated responses. Fast changes in KCC2 function and expression have been observed under various physiological and pathophysiological conditions. Here, we show that the application of protein synthesis inhibitors cycloheximide and emetine to acute rat hippocampal slices have no effect on total KCC2 protein level and K-Cl cotransporter function. Furthermore, blocking constitutive lysosomal degradation with leupeptin did not induce significant changes in KCC2 protein levels. These findings indicate a low basal turnover rate of the total KCC2 protein pool. In the presence of the glutamate receptor agonist NMDA, the total KCC2 protein level decreased to about 30% within 4 h, and this effect was blocked by calpeptin and MDL-28170, inhibitors of the calcium-activated protease calpain. Interictal-like activity induced by incubation of hippocampal slices in an Mg(2+)-free solution led to a fast reduction in KCC2-mediated Cl(-) transport efficacy in CA1 pyramidal neurons, which was paralleled by a decrease in both total and plasmalemmal KCC2 protein. These effects were blocked by the calpain inhibitor MDL-28170. Taken together, these findings show that calpain activation leads to cleavage of KCC2, thereby modulating GABAergic signaling.


Subject(s)
Calpain/metabolism , Gene Expression Regulation/physiology , Pyramidal Cells/physiology , Symporters/metabolism , Action Potentials/drug effects , Analysis of Variance , Animals , Animals, Newborn , Calcium/metabolism , Calcium Ionophores/pharmacology , Calpain/pharmacology , Cycloheximide/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Dipeptides/pharmacology , Dizocilpine Maleate/pharmacology , Dose-Response Relationship, Drug , Emetine/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Regulation/drug effects , Hippocampus/cytology , In Vitro Techniques , Ionomycin/pharmacology , Leupeptins/pharmacology , Magnesium/metabolism , Male , Membrane Potentials/drug effects , N-Methylaspartate/pharmacology , Patch-Clamp Techniques , Protein Synthesis Inhibitors/pharmacology , Pyramidal Cells/drug effects , Rats , Rats, Wistar , Statistics, Nonparametric , Valine/analogs & derivatives , Valine/pharmacology , K Cl- Cotransporters
SELECTION OF CITATIONS
SEARCH DETAIL
...