Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Brain Struct Funct ; 221(7): 3601-15, 2016 09.
Article in English | MEDLINE | ID: mdl-26427846

ABSTRACT

Synaptic reorganization in the epileptic hippocampus involves altered excitatory and inhibitory transmission besides the rearrangement of dendritic spines, resulting in altered excitability, ion homeostasis, and cell swelling. The potassium-chloride cotransporter-2 (KCC2) is the main chloride extruder in neurons and hence will play a prominent role in determining the polarity of GABAA receptor-mediated chloride currents. In addition, KCC2 also interacts with the actin cytoskeleton which is critical for dendritic spine morphogenesis, and for the maintenance of glutamatergic synapses and cell volume. Using immunocytochemistry, we examined the cellular and subcellular levels of KCC2 in surgically removed hippocampi of temporal lobe epilepsy (TLE) patients and compared them to control human tissue. We also studied the distribution of KCC2 in a pilocarpine mouse model of epilepsy. An overall increase in KCC2-expression was found in epilepsy and confirmed by Western blots. The cellular and subcellular distributions in control mouse and human samples were largely similar; moreover, changes affecting KCC2-expression were also alike in chronic epileptic human and mouse hippocampi. At the subcellular level, we determined the neuronal elements exhibiting enhanced KCC2 expression. In epileptic tissue, staining became more intense in the immunopositive elements detected in control tissue, and profiles with subthreshold expression of KCC2 in control samples became labelled. Positive interneuron somata and dendrites were more numerous in epileptic hippocampi, despite severe interneuron loss. Whether the elevation of KCC2-expression is ultimately a pro- or anticonvulsive change, or both-behaving differently during ictal and interictal states in a context-dependent manner-remains to be established.


Subject(s)
Epilepsy, Temporal Lobe/metabolism , Hippocampus/metabolism , Symporters/metabolism , Adult , Aged , Animals , Disease Models, Animal , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/pathology , Female , Hippocampus/pathology , Hippocampus/ultrastructure , Humans , Male , Mice , Middle Aged , Neurons/metabolism , Pilocarpine
2.
Science ; 345(6201): 1130, 2014 Sep 05.
Article in English | MEDLINE | ID: mdl-25190787

ABSTRACT

Glykys et al. (Reports, 7 February 2014, p. 670) conclude that, rather than ion transporters, "local impermeant anions establish the neuronal chloride concentration" and thereby determine "the magnitude and direction of GABAAR currents at individual synapses." If this were possible, perpetual ion-motion machines could be constructed. The authors' conclusions conflict with basic thermodynamic principles.


Subject(s)
Brain/metabolism , Chloride Channels/metabolism , Chlorides/metabolism , Neurons/metabolism , Receptors, GABA-A/metabolism , Animals
3.
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
4.
J Neurochem ; 127(4): 520-30, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24024616

ABSTRACT

The anterior piriform cortex (APC) is activated by, and is the brain area most sensitive to, essential (indispensable) amino acid (IAA) deficiency. The APC is required for the rapid (20 min) behavioral rejection of IAA deficient diets and increased foraging, both crucial adaptive functions supporting IAA homeostasis in omnivores. The biochemical mechanisms signaling IAA deficiency in the APC block initiation of translation in protein synthesis via uncharged tRNA and the general amino acid control kinase, general control nonderepressing kinase 2. Yet, how inhibition of protein synthesis activates the APC is unknown. The neuronal K(+) Cl(-) cotransporter, neural potassium chloride co-transporter (KCC2), and GABAA receptors are essential inhibitory elements in the APC with short plasmalemmal half-lives that maintain control in this highly excitable circuitry. After a single IAA deficient meal both proteins were reduced (vs. basal diet controls) in western blots of APC (but not neocortex or cerebellum) and in immunohistochemistry of APC. Furthermore, electrophysiological analyses support loss of inhibitory elements such as the GABAA receptor in this model. As the crucial inhibitory function of the GABAA receptor depends on KCC2 and the Cl(-) transmembrane gradient it establishes, these results suggest that loss of such inhibitory elements contributes to disinhibition of the APC in IAA deficiency. The circuitry of the anterior piriform cortex (APC) is finely balanced between excitatory (glutamate, +) and inhibitory (GABA, -) transmission. GABAA receptors use Cl(-), requiring the neural potassium chloride co-transporter (KCC2). Both are rapidly turning-over proteins, dependent on protein synthesis for repletion. In IAA (indispensable amino acid) deficiency, within 20 min, blockade of protein synthesis prevents restoration of these inhibitors; they are diminished; disinhibition ensues. GCN2 = general control non-derepressing kinase 2, eIF2α = α-subunit of the eukaryotic initiation factor 2.


Subject(s)
Amino Acids, Essential/deficiency , Olfactory Pathways/metabolism , Receptors, GABA-A/biosynthesis , Symporters/biosynthesis , Animals , Down-Regulation , Excitatory Postsynaptic Potentials , Male , Rats , K Cl- Cotransporters
5.
Curr Top Membr ; 70: 215-37, 2012.
Article in English | MEDLINE | ID: mdl-23177987

ABSTRACT

The cation chloride cotransporters (CCCs) represent an important family of transporters that plays key roles in vectorial electrolyte movement across epithelia and in intracellular chloride homeostasis of neurons and muscle cells. The CCCs are composed of three broad groups, two of which include multiple isoforms: Na-Cl cotransporter (NCC; SLC12A3), Na-K-2Cl cotransporter (NKCC; SLC12A1-2), and K-Cl cotransporter (KCC; SLC12A4-7). The CCCs are inhibited by clinically relevant drugs, including loop diuretics that inhibit NKCC2 in the renal thick ascending limb and thiazide diuretics that inhibit NCC in the renal distal tubule. For many years, much research on this gene family has centered on understanding ion binding and inhibitor interaction which represent important features of the molecular operation of these transporters. Recently, high resolution structures of bacterial transport proteins related to the CCCs have become available, thus permitting structural context in which to evaluate previous ion and inhibitor studies of the CCCs. In this article, I review past molecular and structure-function studies that have provided key pieces of information about ion binding and inhibitor interaction primarily of NKCC for which we have the most information. I then place these findings into the structural context of recent homology models of NKCC based on the outward-facing open and occluded conformations of the related bacterial transporters. These homology models provide our first glimpse into the fine details of the molecular operation of the CCCs.


Subject(s)
Chlorides/metabolism , Sodium Potassium Chloride Symporter Inhibitors/pharmacology , Sodium-Potassium-Chloride Symporters/chemistry , Sodium-Potassium-Chloride Symporters/metabolism , Animals , Binding Sites , Cations , Humans , Kidney/metabolism , Models, Molecular , Muscle Cells/metabolism , Neurons/metabolism , Protein Conformation , Sodium Chloride Symporter Inhibitors/metabolism , Structure-Activity Relationship
6.
Am J Physiol Cell Physiol ; 303(11): C1180-91, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23034386

ABSTRACT

Using antibodies prepared against a unique region (exon 22-24) of rat K(+)-Cl(-) cotransporter-2 (KCC2), we confirmed that the ~140-kDa KCC2 protein is exclusively expressed in rat brain, but in chicken, we observed strong reactivity not only with the ~140-kDa KCC2 protein in brain but also a slightly larger ~145-kDa protein in heart. In silico analysis showed that while exon 22 of KCC2 is unique to this isoform in therian mammals, it is retained in KCC2's closest paralog, KCC4, of lower vertebrates, including chicken. To eliminate potential cross-reactivity with chicken KCC4, the antibodies were preadsorbed with blocking peptides prepared over the only two regions showing significant sequence identity to chicken KCC4. This completely eliminated antibody recognition of exogenously expressed chicken KCC4 but not of the ~145-kDa protein in chicken heart, indicating that chicken heart expresses KCC2. Real-time PCR confirmed robust KCC2 transcript expression in both chicken brain and heart. Chicken heart expressed predominantly the longer KCC2a splice variant consistent with the larger ~145-kDa protein in chicken heart. Immunofluorescence microscopy revealed prominent plasma membrane KCC2 labeling in chicken ventricular cardiomyocytes. We hypothesize that KCC2 is an important Cl(-) extrusion pathway in avian cardiomyocytes that counters channel-mediated Cl(-) loading during high heart rates with ß-adrenergic stimulation. While KCC2 is absent from mammalian cardiomyocytes, understanding the role that the other KCC isoforms play in Cl(-) homeostasis of these cells represents a nascent area of research.


Subject(s)
Brain/metabolism , Chickens/metabolism , Myocardium/metabolism , Symporters/metabolism , Animals , Antibodies, Neutralizing/immunology , HEK293 Cells , Humans , Protein Isoforms/biosynthesis , Rats , Symporters/genetics , Symporters/immunology , K Cl- Cotransporters
7.
J Biol Chem ; 282(41): 29777-84, 2007 Oct 12.
Article in English | MEDLINE | ID: mdl-17693402

ABSTRACT

The potassium chloride cotransporter KCC2 plays a major role in the maintenance of transmembrane chloride potential in mature neurons; thus KCC2 activity is critical for hyperpolarizing membrane currents generated upon the activation of gamma-aminobutyric acid type A and glycine (Gly) receptors that underlie fast synaptic inhibition in the adult central nervous system. However, to date an understanding of the cellular mechanism that neurons use to modulate the functional expression of KCC2 remains rudimentary. Using Escherichia coli expression coupled with in vitro kinase assays, we first established that protein kinase C (PKC) can directly phosphorylate serine 940 (Ser(940)) within the C-terminal cytoplasmic domain of KCC2. We further demonstrated that Ser(940) is the major site for PKC-dependent phosphorylation for full-length KCC2 molecules when expressed in HEK-293 cells. Phosphorylation of Ser(940) increased the cell surface stability of KCC2 in this system by decreasing its rate of internalization from the plasma membrane. Coincident phosphorylation of Ser(940) increased the rate of ion transport by KCC2. It was further evident that phosphorylation of endogenous KCC2 in cultured hippocampal neurons is regulated by PKC-dependent activity. Moreover, in keeping with our recombinant studies, enhancing PKC-dependent phosphorylation increased the targeting of KCC2 to the neuronal cell surface. Our studies thus suggest that PKC-dependent phosphorylation of KCC2 may play a central role in modulating both the functional expression of this critical transporter in the brain and the strength of synaptic inhibition.


Subject(s)
Cell Membrane/metabolism , Potassium Chloride/chemistry , Protein Kinase C/metabolism , Symporters/chemistry , Binding Sites , Cell Line , Endocytosis , Escherichia coli/metabolism , Hippocampus/metabolism , Humans , Models, Biological , Neurons/metabolism , Phosphorylation , Protein Structure, Tertiary , Receptors, GABA-A/chemistry , K Cl- Cotransporters
8.
Gene Expr Patterns ; 6(8): 1000-6, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16814616

ABSTRACT

We examined the expression of Slc12a2 (NKCC1) transcripts in the developing mouse by Northern blot analysis and in situ hybridization (ISH) using riboprobes transcribed from a cDNA encoding the transmembrane domain of human Slc12a2. In developing kidney, the 7.5-kb Slc12a2 transcript was expressed at all stages examined (13.5 d.p.c. to adult) but was more abundant in immature metanephroi. ISH revealed that NKCC1 was expressed in both mesenchymal cells and early nephric structures, but not branching ureteric buds, of developing metanephroi. A marked increase in expression was observed in the endocapillary cells of capillary loop stage glomeruli, and high expression was observed in the glomeruli of more mature nephrons. This was in contrast to Slc12a1 (NKCC2), where expression was excluded from the glomerulus. Extra-renal expression of Slc12a2 was examined in 13.5, 15.5, and 16.5 d.p.c. mouse embryos. Slc12a2 was highly expressed in the developing lung, gut, submandibular gland, tooth bud, and nasal epithelium. Slc12a2 expression was also observed in the developing central and peripheral nervous systems, including choroid plexus and trigeminal and dorsal root ganglia.


Subject(s)
Embryonic Development/physiology , Gene Expression , Kidney/embryology , Organogenesis/physiology , Sodium-Potassium-Chloride Symporters/metabolism , Animals , Basement Membrane/metabolism , Brain/embryology , Brain/metabolism , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Intestinal Mucosa/metabolism , Intestines/embryology , Kidney/metabolism , Lung/embryology , Lung/metabolism , Mice , Solute Carrier Family 12, Member 2 , Submandibular Gland/embryology , Submandibular Gland/metabolism , Tissue Distribution
9.
Neurobiol Dis ; 17(1): 62-9, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15350966

ABSTRACT

Cation chloride cotransporters have been proposed to play a role in the modulation of neuronal responses to gamma-aminobutyric acid (GABA). In conditions of neuronal damage, where neuronal excitability is increased, the expression of the KCC2 transporter is decreased. This is also seen in spinal cord in models of neuropathic pain. We have investigated the expression of the Na-K-Cl, and K-Cl cotransporters NKCC1 and KCC2, in dorsal root ganglion (DRG) and spinal sensory neurons during arthritis, a condition in which neuronal excitability is also increased. NKCC1 was expressed in control DRG neurons, and its expression was decreased in arthritis. Both NKCC1 and KCC2 were expressed in sensory neurons in the spinal cord. In acute arthritis, both NKCC1 and KCC2 mRNA increased in superficial but not deep dorsal horn, and this was accompanied by an increase in protein expression. In chronic arthritis, NKCC1 expression remained raised, but KCC2 mRNA and protein expression returned to control levels. Altered KCC2 and NKCC1 expression in arthritis may contribute to the control of spinal cord excitability and may represent novel therapeutic targets in the treatment of inflammatory pain.


Subject(s)
Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Neurons, Afferent/metabolism , Neurons, Afferent/pathology , Sodium-Potassium-Chloride Symporters/biosynthesis , Symporters/biosynthesis , Animals , Arthritis, Experimental/genetics , Ganglia, Spinal/metabolism , Gene Expression Regulation/physiology , Inflammation/genetics , Inflammation/metabolism , RNA, Messenger/biosynthesis , Rats , Sodium-Potassium-Chloride Symporters/genetics , Solute Carrier Family 12, Member 2 , Spinal Cord/metabolism , Spinal Cord/pathology , Symporters/genetics , K Cl- Cotransporters
10.
Am J Physiol Cell Physiol ; 287(4): C919-31, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15175220

ABSTRACT

Both Cs(+) and NH(4)(+) alter neuronal Cl(-) homeostasis, yet the mechanisms have not been clearly elucidated. We hypothesized that these two cations altered the operation of the neuronal K(+)-Cl(-) cotransporter (KCC2). Using exogenously expressed KCC2 protein, we first examined the interaction of cations at the transport site of KCC2 by monitoring furosemide-sensitive (86)Rb(+) influx as a function of external Rb(+) concentration at different fixed external cation concentrations (Na(+), Li(+), K(+), Cs(+), and NH(4)(+)). Neither Na(+) nor Li(+) affected furosemide-sensitive (86)Rb(+) influx, indicating their inability to interact at the cation translocation site of KCC2. As expected for an enzyme that accepts Rb(+) and K(+) as alternate substrates, K(+) was a competitive inhibitor of Rb(+) transport by KCC2. Like K(+), both Cs(+) and NH(4)(+) behaved as competitive inhibitors of Rb(+) transport by KCC2, indicating their potential as transport substrates. Using ion chromatography to measure unidirectional Rb(+) and Cs(+) influxes, we determined that although KCC2 was capable of transporting Cs(+), it did so with a lower apparent affinity and maximal velocity compared with Rb(+). To assess NH(4)(+) transport by KCC2, we monitored intracellular pH (pH(i)) with a pH-sensitive fluorescent dye after an NH(4)(+)-induced alkaline load. Cells expressing KCC2 protein recovered pH(i) much more rapidly than untransfected cells, indicating that KCC2 can mediate net NH(4)(+) uptake. Consistent with KCC2-mediated NH(4)(+) transport, pH(i) recovery in KCC2-expressing cells could be inhibited by furosemide (200 microM) or removal of external [Cl(-)]. Thermodynamic and kinetic considerations of KCC2 operating in alternate transport modes can explain altered neuronal Cl(-) homeostasis in the presence of Cs(+) and NH(4)(+).


Subject(s)
Cations/metabolism , Ion Transport/physiology , Kinetics , Symporters/metabolism , Thermodynamics , Animals , Cations/chemistry , Cell Line , Cesium/chemistry , Cesium/metabolism , Chlorine/chemistry , Chlorine/metabolism , Dogs , Hydrogen-Ion Concentration , Intracellular Fluid/chemistry , Quaternary Ammonium Compounds/chemistry , Quaternary Ammonium Compounds/metabolism , Rats , Rubidium Radioisotopes/chemistry , Rubidium Radioisotopes/metabolism , Symporters/chemistry , K Cl- Cotransporters
11.
J Neurosci ; 24(19): 4683-91, 2004 May 12.
Article in English | MEDLINE | ID: mdl-15140939

ABSTRACT

GABA-mediated fast-hyperpolarizing inhibition depends on extrusion of chloride by the neuron-specific K-Cl cotransporter, KCC2. Here we show that sustained interictal-like activity in hippocampal slices downregulates KCC2 mRNA and protein expression in CA1 pyramidal neurons, which leads to a reduced capacity for neuronal Cl- extrusion. This effect is mediated by endogenous BDNF acting on tyrosine receptor kinase B (TrkB), with down-stream cascades involving both Shc/FRS-2 (src homology 2 domain containing transforming protein/FGF receptor substrate 2) and PLCgamma (phospholipase Cgamma)-cAMP response element-binding protein signaling. The plasmalemmal KCC2 has a very high rate of turnover, with a time frame that suggests a novel role for changes in KCC2 expression in diverse manifestations of neuronal plasticity. A downregulation of KCC2 may be a general early response involved in various kinds of neuronal trauma.


Subject(s)
Adaptor Proteins, Signal Transducing , Down-Regulation/physiology , Neurons/metabolism , Symporters/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Binding Sites/physiology , Biotinylation , Brain-Derived Neurotrophic Factor/pharmacology , Brain-Derived Neurotrophic Factor/physiology , Cell Membrane/metabolism , Chlorides/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Down-Regulation/drug effects , Hippocampus/cytology , Hippocampus/physiology , In Vitro Techniques , Magnesium/pharmacology , Mice , Mice, Mutant Strains , Neurons/drug effects , Phospholipase C gamma , Phosphorylation/drug effects , Pyramidal Cells/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptor, trkB/drug effects , Receptor, trkB/genetics , Receptor, trkB/metabolism , Shc Signaling Adaptor Proteins , Signal Transduction/physiology , Src Homology 2 Domain-Containing, Transforming Protein 1 , Symporters/genetics , Synaptic Transmission/physiology , Type C Phospholipases/metabolism , K Cl- Cotransporters
12.
Trends Neurosci ; 26(4): 199-206, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12689771

ABSTRACT

Electrical signaling in neurons is based on the operation of plasmalemmal ion pumps and carriers that establish transmembrane ion gradients, and on the operation of ion channels that generate current and voltage responses by dissipating these gradients. Although both voltage- and ligand-gated channels are being extensively studied, the central role of ion pumps and carriers is largely ignored in current neuroscience. Such an information gap is particularly evident with regard to neuronal Cl- regulation, despite its immense importance in the generation of inhibitory synaptic responses by GABA- and glycine-gated anion channels. The cation-chloride co-transporters (CCCs) have been identified as important regulators of neuronal Cl- concentration, and recent work indicates that CCCs play a key role in shaping GABA- and glycine-mediated signaling, influencing not only fast cell-to-cell communication but also various aspects of neuronal development, plasticity and trauma.


Subject(s)
Ion Transport/physiology , Neurons/metabolism , Symporters/metabolism , Wounds and Injuries/metabolism , Animals , Cells, Cultured , Circadian Rhythm , Epilepsy/etiology , Epilepsy/metabolism , Hippocampus/growth & development , Hippocampus/metabolism , Humans , Models, Biological , Tissue Distribution , gamma-Aminobutyric Acid/metabolism , K Cl- Cotransporters
SELECTION OF CITATIONS
SEARCH DETAIL
...