Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Eur J Gastroenterol Hepatol ; 33(6): 817-824, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33079783

ABSTRACT

BACKGROUND: Colonic tuft cells are epithelial chemosensory cells involved in barrier integrity, modulation of inflammatory responses and gut homeostasis. Recent evidence indicates an involvement of tuft cells in ulcerative colitis pathogenesis, though mechanisms remain largely unknown.Here, we quantified the colonic tuft cell population in patients with quiescent ulcerative colitis as compared to patients without identified colonic disease (controls). METHODS: In this retrospective study, we obtained endoscopic colonic sigmoid biopsies from 14 patients with quiescent ulcerative colitis and from 17 controls. In a blinded central-reading design, we identified tuft cells by immunohistochemistry using a cyclooxygenase-1 antibody as a marker and performed a simple counting by visual inspection. Poisson regression was employed for statistics and results were adjusted for gender, age and smoking status. RESULTS: Ulcerative colitis patients demonstrated a 55% reduced tuft cell count in colonic mucosa compared with the control group (95% confidence limit: range 31-71%, P = 0.0002). Ulcerative colitis patients had a mean tuft cells count of 46 tuft cells/mm2 (95% CI, 36-59), while controls demonstrated a mean of 104 tuft cells/mm2 (95% CI, 79-136). No interactions of other covariates, such as age, smoking status, total duration of ulcerative colitis disease and duration of clinical remission prior to study inclusion were detected between ulcerative colitis patients and controls. CONCLUSION: Quiescent ulcerative colitis patients have a relatively low number of colonic tuft cells. Further studies are warranted to explore the potential involvement of tuft cells in ulcerative colitis pathogenesis.


Subject(s)
Colitis, Ulcerative , Colitis , Colitis, Ulcerative/diagnosis , Colon , Humans , Intestinal Mucosa , Retrospective Studies
2.
Int J Mol Sci ; 21(5)2020 Mar 10.
Article in English | MEDLINE | ID: mdl-32164249

ABSTRACT

Mucosal healing determined by endoscopy is currently the remission standard for ulcerative colitis (UC). However, new criteria for remission are emerging, such as histologic normalization, which appears to correlate better to the risk of relapse. Here, we study mucosal healing on a molecular and functional level in quiescent UC. We obtained endoscopic biopsies from 33 quiescent UC patients and from 17 controls. Histology was assessed using Geboes score. Protein and mRNA levels were evaluated for the tight junction proteins claudin-2, claudin-4, occludin, and tricellulin, as well as Cl-/HCO3- exchanger DRA, and cyclo-oxygenase enzymes (COX-1, COX-2). The mucosal activity of COX-1 and COX-2 enzymes was assessed in modified Ussing chambers, measuring electrogenic ion transport (short-circuit current, SCC). Chronic inflammation was present in most UC patients. The protein level of claudin-4 was reduced, while mRNA-levels of claudin-2 and claudin-4 were upregulated in UC patients. Surprisingly, the mRNA level of COX-1 was downregulated, but was unaltered for COX-2. Basal ion transport was not affected, while COX-2 inhibition induced a two-fold larger decrease in SCC in UC patients. Despite being in clinical and endoscopic remission, quiescent UC patients demonstrated abnormal mucosal barrier properties at the molecular and functional level. Further exploration of mucosal molecular signature for revision of current remission standards should be considered.


Subject(s)
Claudin-1/genetics , Claudins/genetics , Colitis, Ulcerative/pathology , Cyclooxygenase 1/genetics , Cyclooxygenase 2/genetics , MARVEL Domain Containing 2 Protein/genetics , Adult , Aged , Biopsy , Case-Control Studies , Claudin-1/metabolism , Claudins/metabolism , Colitis, Ulcerative/genetics , Colitis, Ulcerative/metabolism , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Female , Gene Expression Regulation , Gene Regulatory Networks , Humans , MARVEL Domain Containing 2 Protein/metabolism , Male , Middle Aged , Young Adult
3.
Mol Brain ; 10(1): 43, 2017 09 04.
Article in English | MEDLINE | ID: mdl-28870203

ABSTRACT

The neurotransmitter glutamate facilitates neuronal signalling at excitatory synapses. Glutamate is released from the presynaptic membrane into the synaptic cleft. Across the synaptic cleft glutamate binds to both ion channels and metabotropic glutamate receptors at the postsynapse, which expedite downstream signalling in the neuron. The postsynaptic density, a highly specialized matrix, which is attached to the postsynaptic membrane, controls this downstream signalling. The postsynaptic density also resets the synapse after each synaptic firing. It is composed of numerous proteins including a family of Discs large associated protein 1, 2, 3 and 4 (DLGAP1-4) that act as scaffold proteins in the postsynaptic density. They link the glutamate receptors in the postsynaptic membrane to other glutamate receptors, to signalling proteins and to components of the cytoskeleton. With the central localisation in the postsynapse, the DLGAP family seems to play a vital role in synaptic scaling by regulating the turnover of both ionotropic and metabotropic glutamate receptors in response to synaptic activity. DLGAP family has been directly linked to a variety of psychological and neurological disorders. In this review we focus on the direct and indirect role of DLGAP family on schizophrenia as well as other brain diseases.


Subject(s)
Brain Diseases/metabolism , Neurons/metabolism , SAP90-PSD95 Associated Proteins/metabolism , Amino Acid Sequence , Animals , Brain/metabolism , Brain/pathology , Humans , Models, Biological , Protein Interaction Mapping , SAP90-PSD95 Associated Proteins/chemistry
4.
Front Neural Circuits ; 11: 111, 2017.
Article in English | MEDLINE | ID: mdl-29375322

ABSTRACT

Motor fatigue occurring during prolonged physical activity has both peripheral and central origins. It was previously demonstrated that the excitability of motoneurons was decreased when a spillover of serotonin could activate extrasynaptic 5-HT1A receptors at the axon initial segment (AIS) of motoneurons. Here we investigated the impact of massive synaptic release of serotonin on motor behavior in an integrated preparation of the adult turtle performing fictive scratching behaviors. We found that a prolonged electrical stimulation of the raphe spinal pathway induced a reversible inhibition of the motor behavior that lasted several tens of seconds. The effect disappeared when the spinal cord was perfused with an antagonist for 5-HT1A receptors. By demonstrating a direct impact of serotonin on motor behavior, we suggest a central role of this monoamine behind central fatigue.


Subject(s)
Fatigue/metabolism , Movement/physiology , Raphe Nuclei/metabolism , Receptor, Serotonin, 5-HT1A/metabolism , Serotonin/metabolism , Spinal Cord/metabolism , Animals , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/metabolism , Electric Stimulation , Fatigue/drug therapy , HEK293 Cells , Humans , In Vitro Techniques , Mice, 129 Strain , Mice, Knockout , Movement/drug effects , Neural Pathways/cytology , Neural Pathways/drug effects , Neural Pathways/metabolism , Peripheral Nerves/physiology , Piperazines/pharmacology , Pyridines/pharmacology , Receptor, Serotonin, 5-HT1A/genetics , Reflex/physiology , Serotonin 5-HT1 Receptor Antagonists , Spinal Cord/cytology , Spinal Cord/drug effects , Turtles
5.
Pharm Res ; 33(3): 716-28, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26563206

ABSTRACT

PURPOSE: Aggregation aspects of therapeutic monoclonal antibodies (mAbs) are of common concern to the pharmaceutical industry. Low pH treatment is applied during affinity purification and to inactivate endogenous retroviruses, directing interest to the mechanisms of acid-induced antibody aggregation. METHODS: We characterized the oligomerization kinetics at pH 3.3, as well as the reversibility upon neutralization, of three model mAbs with identical variable regions, representative of IgG1, IgG2 and IgG4 respectively. We applied size-exclusion high performance liquid chromatography and orthogonal analytical methods, including small-angle X-ray scattering and dynamic light scattering and supplemented the experimental data with crystal structure-based spatial aggregation propensity (SAP) calculations. RESULTS: We revealed distinct solution behaviors between the three mAb models: At acidic pH IgG1 retained monomeric, whereas IgG2 and IgG4 exhibited two-phase oligomerization processes. After neutralization, IgG2 oligomers partially reverted to the monomeric state, while on the contrary, IgG4 oligomers tended to aggregate. Subclass-specific aggregation-prone motifs on the Fc fragments were identified, which may lead to two distinct pathways of reversible and irreversible aggregation, respectively. CONCLUSIONS: We conclude that subtle variations in mAb sequence greatly affect responses towards low-pH incubation and subsequent neutralization, and demonstrate how orthogonal biophysical methods distinguish between reversible and irreversible mAb aggregation pathways at early stages of acidic treatment.


Subject(s)
Acids/chemistry , Antibodies, Monoclonal/chemistry , Chromatography, High Pressure Liquid/methods , Humans , Hydrogen-Ion Concentration , Immunoglobulin G/chemistry , Kinetics , Protein Multimerization , Solutions/chemistry
6.
Am J Physiol Cell Physiol ; 309(10): C693-706, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26405101

ABSTRACT

The potassium channel Kv7.1 plays critical physiological roles in both heart and epithelial tissues. In heart, Kv7.1 and the accessory subunit KCNE1 forms the slowly activating delayed-rectifier potassium current current, which is enhanced by protein kinase A (PKA)-mediated phosphorylation. The observed current increase requires both phosphorylation of Kv7.1 and the presence of KCNE1. However, PKA also stimulates Kv7.1 currents in epithelial tissues, such as colon, where the channel does not coassemble with KCNE1. Here, we demonstrate that PKA activity significantly impacts the subcellular localization of Kv7.1 in Madin-Darby canine kidney cells. While PKA inhibition reduced the fraction of channels at the cell surface, PKA activation increased it. We show that PKA inhibition led to intracellular accumulation of Kv7.1 in late endosomes/lysosomes. By mass spectroscopy we identified eight phosphorylated residues on Kv7.1, however, none appeared to play a role in the observed response. Instead, we found that PKA acted by regulating endocytic trafficking involving the ubiquitin ligase Nedd4-2. We show that a Nedd4-2-resistant Kv7.1-mutant displayed significantly reduced intracellular accumulation upon PKA inhibition. Similar effects were observed upon siRNA knockdown of Nedd4-2. However, although Nedd4-2 is known to regulate Kv7.1 by ubiquitylation, biochemical analyses demonstrated that PKA did not influence the amount of Nedd4-2 bound to Kv7.1 or the ubiquitylation level of the channel. This suggests that PKA influences Nedd4-2-dependent Kv7.1 transport though a different molecular mechanism. In summary, we identify a novel mechanism whereby PKA can increase Kv7.1 current levels, namely by regulating Nedd4-2-dependent Kv7.1 transport.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , KCNQ1 Potassium Channel/metabolism , Protein Transport/physiology , Transport Vesicles/physiology , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Line , Cyclic AMP-Dependent Protein Kinases/genetics , Dogs , Endosomal Sorting Complexes Required for Transport/genetics , Gene Expression Regulation, Enzymologic , Gene Knockdown Techniques , KCNQ1 Potassium Channel/genetics , Mutation , Nedd4 Ubiquitin Protein Ligases , Protein Conformation , Protein Kinase Inhibitors/pharmacology , Ubiquitin-Protein Ligases/genetics
7.
IUCrJ ; 2(Pt 1): 9-18, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25610623

ABSTRACT

IgG subclass-specific differences in biological function and in vitro stability are often referred to variations in the conformational flexibility, while this flexibility has rarely been characterized. Here, small-angle X-ray scattering data from IgG1, IgG2 and IgG4 antibodies, which were designed with identical variable regions, were thoroughly analysed by the ensemble optimization method. The extended analysis of the optimized ensembles through shape clustering reveals distinct subclass-specific conformational preferences, which provide new insights for understanding the variations in physical/chemical stability and biological function of therapeutic antibodies. Importantly, the way that specific differences in the linker region correlate with the solution structure of intact antibodies is revealed, thereby visualizing future potential for the rational design of antibodies with designated physicochemical properties and tailored effector functions. In addition, this advanced computational approach is applicable to other flexible multi-domain systems and extends the potential for investigating flexibility in solutions of macromolecules by small-angle X-ray scattering.

8.
J Pharm Sci ; 103(6): 1701-10, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24700358

ABSTRACT

A crucial step in the development of therapeutic monoclonal antibodies is the selection of robust pharmaceutical candidates and screening of efficacious protein formulations to increase the resistance toward physicochemical degradation and aggregation during processing and storage. Here, we introduce small-angle X-ray scattering (SAXS) to characterize antibody solution behavior, which strongly complements conventional biophysical analysis. First, we apply a variety of conventional biophysical techniques for the evaluation of structural, conformational, and colloidal stability and report a systematic comparison between designed humanized IgG1, IgG2, and IgG4 with identical variable regions. Then, the high information content of SAXS data enables sensitive detection of structural differences between three IgG subclasses at neutral pH and rapid formation of dimers of IgG2 and IgG4 at low pH. We reveal subclass-specific variation in intermolecular repulsion already at low and medium protein concentrations, which explains the observed improved stability of IgG1 with respect to aggregation. We show how excipients dramatically influence such repulsive effects, hence demonstrating the potential application of extensive SAXS screening in antibody selection, eventual engineering, and formulation development.


Subject(s)
Antibodies, Monoclonal/chemistry , Immunoglobulin G/chemistry , Antibodies, Monoclonal/classification , Biophysics , Chromatography, Gel , Chromatography, High Pressure Liquid , Humans , Immunoglobulin G/classification , Protein Conformation , Scattering, Small Angle
9.
Cardiovasc Res ; 101(1): 175-84, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24148898

ABSTRACT

AIMS: The purpose of this study was to investigate the functional role of G-protein-coupled inward rectifier potassium (GIRK) channels in the cardiac ventricle. METHODS AND RESULTS: Immunofluorescence experiments demonstrated that GIRK4 was localized in outer sarcolemmas and t-tubules in GIRK1 knockout (KO) mice, whereas GIRK4 labelling was not detected in GIRK4 KO mice. GIRK4 was localized in intercalated discs in rat ventricle, whereas it was expressed in intercalated discs and outer sarcolemmas in rat atrium. GIRK4 was localized in t-tubules and intercalated discs in human ventricular endocardium and epicardium, but absent in mid-myocardium. Electrophysiological recordings in rat ventricular tissue ex vivo showed that the adenosine A1 receptor agonist N6-cyclopentyladenosine (CPA) and acetylcholine (ACh) shortened action potential duration (APD), and that the APD shortening was reversed by either the GIRK channel blocker tertiapin-Q, the adenosine A1 receptor antagonist DPCPX or by the muscarinic M2 receptor antagonist AF-DX 116. Tertiapin-Q prolonged APD in the absence of the exogenous receptor activation. Furthermore, CPA and ACh decreased the effective refractory period and the effect was reversed by either tertiapin-Q, DPCPX or AF-DX 116. Receptor activation also hyperpolarized the resting membrane potential, an effect that was reversed by tertiapin-Q. In contrast, tertiapin-Q depolarized the resting membrane potential in the absence of the exogenous receptor activation. CONCLUSION: Confocal microscopy shows that among species GIRK4 is differentially localized in the cardiac ventricle, and that it is heterogeneously expressed across human ventricular wall. Electrophysiological recordings reveal that GIRK current may contribute significantly to ventricular repolarization and thereby to cardiac electrical stability.


Subject(s)
G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Heart Ventricles/metabolism , Animals , Humans , Male , Membrane Potentials , Mice , Mice, Knockout , Potassium/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley
10.
Front Genet ; 4: 54, 2013.
Article in English | MEDLINE | ID: mdl-23596459

ABSTRACT

Heterozygous mutations in the KCNQ3 gene on chromosome 8q24 encoding the voltage-gated potassium channel KV7.3 subunit have previously been associated with rolandic epilepsy and idiopathic generalized epilepsy (IGE) including benign neonatal convulsions. We identified a de novo t(3;8) (q21;q24) translocation truncating KCNQ3 in a boy with childhood autism. In addition, we identified a c.1720C > T [p.P574S] nucleotide change in three unrelated individuals with childhood autism and no history of convulsions. This nucleotide change was previously reported in patients with rolandic epilepsy or IGE and has now been annotated as a very rare SNP (rs74582884) in dbSNP. The p.P574S KV7.3 variant significantly reduced potassium current amplitude in Xenopus laevis oocytes when co-expressed with KV7.5 but not with KV7.2 or KV7.4. The nucleotide change did not affect trafficking of heteromeric mutant KV7.3/2, KV7.3/4, or KV7.3/5 channels in HEK 293 cells or primary rat hippocampal neurons. Our results suggest that dysfunction of the heteromeric KV7.3/5 channel is implicated in the pathogenesis of some forms of autism spectrum disorders, epilepsy, and possibly other psychiatric disorders and therefore, KCNQ3 and KCNQ5 are suggested as candidate genes for these disorders.

11.
Traffic ; 14(4): 399-411, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23324056

ABSTRACT

The voltage-gated potassium channel KV 7.1 is regulated by non-pore forming regulatory KCNE ß-subunits. Together with KCNE1, it forms the slowly activating delayed rectifier potassium current IKs . However, where the subunits assemble and which of the subunits determines localization of the IKs -complex has not been unequivocally resolved yet. We employed trafficking-deficient KV 7.1 and KCNE1 mutants to investigate IKs trafficking using the polarized Madin-Darby Canine Kidney cell line. We find that the assembly happens early in the secretory pathway but provide three lines of evidence that it takes place in a post-endoplasmic reticulum compartment. We demonstrate that KV 7.1 targets the IKs -complex to the basolateral membrane, but that KCNE1 can redirect the complex to the apical membrane upon mutation of critical KV 7.1 basolateral targeting signals. Our data provide a possible explanation to the fact that KV 7.1 can be localized apically or basolaterally in different epithelial tissues and offer a solution to divergent literature results regarding the effect of KCNE subunits on the subcellular localization of KV 7.1/KCNE complexes.


Subject(s)
KCNQ1 Potassium Channel/metabolism , Potassium Channels, Voltage-Gated/metabolism , Protein Multimerization , Protein Subunits/metabolism , Animals , Cell Membrane/metabolism , Dogs , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , KCNQ1 Potassium Channel/chemistry , KCNQ1 Potassium Channel/genetics , Madin Darby Canine Kidney Cells , Mutation, Missense , Potassium Channels, Voltage-Gated/chemistry , Potassium Channels, Voltage-Gated/genetics , Protein Interaction Domains and Motifs , Protein Sorting Signals/genetics , Protein Subunits/genetics , Protein Transport , Secretory Pathway
12.
Commun Integr Biol ; 5(5): 480-4, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-23181165

ABSTRACT

Ion transport processes are highly energy consuming. It is therefore critical to couple ion transport processes to the metabolic state of the cell. An important player in this coupling appears to be the AMP-activated protein kinase (AMPK). This kinase becomes activated during conditions of cellular metabolic stress and is well-known for its role in promoting ATP-generating catabolic pathways while turning off ATP-utilizing anabolic pathways. Over the past decade AMPK has also emerged as a key regulator of ion channel activity as an increasing number of ion channels are reported to be either directly or indirectly regulated by the kinase. AMPK therefore provides a necessary link between cellular energy levels and ion channel activity.

13.
Front Pharmacol ; 3: 11, 2012.
Article in English | MEDLINE | ID: mdl-22347859

ABSTRACT

Dopamine (DA) containing midbrain neurons play critical roles in several psychiatric and neurological diseases, including schizophrenia and attention deficit hyperactivity disorder, and the substantia nigra pars compacta neurons selectively degenerate in Parkinson's disease. Pharmacological modulation of DA receptors and transporters are well established approaches for treatment of DA-related disorders. Direct modulation of the DA system by influencing the discharge pattern of these autonomously firing neurons has yet to be exploited as a potential therapeutic strategy. Small conductance Ca(2+)-activated K(+) channels (SK channels), in particular the SK3 subtype, are important in the physiology of DA neurons, and agents modifying SK channel activity could potentially affect DA signaling and DA-related behaviors. Here we show that cyclohexyl-[2-(3,5-dimethyl-pyrazol-1-yl)-6-methyl-pyrimidin-4-yl]-amine (CyPPA), a subtype-selective positive modulator of SK channels (SK3 > SK2 > > > SK1, IK), decreased spontaneous firing rate, increased the duration of the apamin-sensitive afterhyperpolarization, and caused an activity-dependent inhibition of current-evoked action potentials in DA neurons from both mouse and rat midbrain slices. Using an immunocytochemically and pharmacologically validated DA release assay employing cultured DA neurons from rats, we show that CyPPA repressed DA release in a concentration-dependent manner with a maximal effect equal to the D2 receptor agonist quinpirole. In vivo studies revealed that systemic administration of CyPPA attenuated methylphenidate-induced hyperactivity and stereotypic behaviors in mice. Taken together, the data accentuate the important role played by SK3 channels in the physiology of DA neurons, and indicate that their facilitation by CyPPA profoundly influences physiological as well as pharmacologically induced hyperdopaminergic behavior.

14.
Traffic ; 13(1): 143-56, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21957902

ABSTRACT

The potassium channel Kv7.1 is expressed in the heart, where it contributes to the repolarization of the cardiac action potential. Additionally, Kv7.1 is expressed in epithelial tissues playing a role in salt and water transport. We recently demonstrated that surface-expressed Kv7.1 is internalized in response to polarization of the epithelial Madin-Darby canine kidney (MDCK) cell line and that this was mediated by activation of protein kinase C (PKC). In this study, the pathway downstream of PKC, which leads to internalization of Kv7.1 upon cell polarization, is elucidated. We show by confocal microscopy that Kv7.1 is endocytosed upon initiation of the polarization process and sent for degradation by the lysosomal pathway. The internalization could be mimicked by pharmacological activation of the AMP-activated protein kinase (AMPK) using three different AMPK activators. We demonstrate that the downstream effector of AMPK is the E3 ubiquitin ligase Nedd4-2. Additionally, we show that AMPK activation results in a downregulation of Kv7.1 currents in Xenopus oocytes through a Nedd4-2-dependent mechanism. In summary, surface-expressed Kv7.1 channels are endocytosed and sent for degradation in lysosomes by an AMPK-mediated activation of Nedd4-2 during the initial phase of the MDCK cell polarization process.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cell Polarity , Endocytosis , KCNQ1 Potassium Channel/biosynthesis , Action Potentials , Animals , Blotting, Western , Calcium/metabolism , Cell Line , Dogs , Down-Regulation , Endosomal Sorting Complexes Required for Transport/metabolism , Fluorescent Antibody Technique , Humans , KCNQ1 Potassium Channel/genetics , Lysosomes/metabolism , Microscopy, Confocal , Nedd4 Ubiquitin Protein Ligases , Oocytes/metabolism , Protein Kinase C/metabolism , Protein Transport , Transfection , Ubiquitin-Protein Ligases/metabolism , Xenopus Proteins , Xenopus laevis
15.
Mol Cell Neurosci ; 48(4): 288-97, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21627990

ABSTRACT

The computational ability of CNS neurons depends critically on the specific localization of ion channels in the somatodendritic and axonal membranes. Neuronal dendrites receive synaptic inputs at numerous spines and integrate them in time and space. The integration of synaptic potentials is regulated by voltage-gated potassium (Kv) channels, such as Kv4.2, which are specifically localized in the dendritic membrane. The synaptic potentials eventually depolarize the membrane of the axon initial segment, thereby activating voltage-gated sodium channels to generate action potentials. Specific Kv channels localized in the axon initial segment, such as Kv1 and Kv7 channels, determine the shape and the rate of action potentials. Kv1 and Kv7 channels present at or near nodes of Ranvier and in presynaptic terminals also influence the propagation of action potentials and neurotransmitter release. The physiological significance of proper Kv channel localization is emphasized by the fact that defects in the trafficking of Kv channels are observed in several neurological disorders including epilepsy. In this review, we will summarize the current understanding of the mechanisms of Kv channel trafficking and discuss how they contribute to the establishment and maintenance of the specific localization of Kv channels in neurons.


Subject(s)
Neurons/physiology , Potassium Channels, Voltage-Gated/metabolism , Animals , Protein Transport/physiology , Synaptic Transmission/physiology
16.
Am J Physiol Cell Physiol ; 300(4): C814-24, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21228319

ABSTRACT

The potassium channel K(V)7.1 is expressed in the heart where it contributes to the repolarization of the cardiac action potential. In addition, K(V)7.1 is expressed in epithelial tissues where it plays a role in salt and water transport. Mutations in the kcnq1 gene can lead to long QT syndrome and deafness, and several mutations have been described as trafficking mutations. To learn more about the basic mechanisms that regulate K(V)7.1 surface expression, we have investigated the trafficking of K(V)7.1 during the polarization process of the epithelial cell line Madin-Darby Canine Kidney (MDCK) using a modified version of the classical calcium switch. We discovered that K(V)7.1 exhibits a very dynamic localization pattern during the calcium switch. When MDCK cells are kept in low calcium medium, K(V)7.1 is mainly observed at the plasma membrane. During the first hours of the switch, K(V)7.1 is removed from the plasma membrane and an intracellular accumulation in the endoplasmic reticulum (ER) is observed. The channel is retained in the ER until the establishment of the lateral membranes at which point K(V)7.1 is released from the ER and moves to the plasma membrane. Our data furthermore suggest that while the removal of K(V)7.1 from the cell surface and its accumulation in the ER could involve activation of protein kinase C, the subsequent release of K(V)7.1 from the ER depends on phosphoinositide 3-kinase (PI3K) activation. In conclusion, our results demonstrate that K(V)7.1 surface expression is regulated by signaling mechanisms involved in epithelial cell polarization in particular signaling cascades involving protein kinase C and PI3K.


Subject(s)
Cell Polarity/physiology , Epithelial Cells/cytology , Epithelial Cells/metabolism , KCNQ1 Potassium Channel/metabolism , Adherens Junctions/metabolism , Animals , Calcium/metabolism , Cell Line , Cell Membrane/metabolism , Desmosomes/metabolism , Dogs , Endoplasmic Reticulum/metabolism , Enzyme Activation , Humans , KCNQ1 Potassium Channel/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/metabolism , Protein Transport , Signal Transduction/physiology , Tight Junctions/metabolism
17.
Am J Hum Genet ; 86(6): 872-80, 2010 Jun 11.
Article in English | MEDLINE | ID: mdl-20560207

ABSTRACT

Congenital long QT syndrome (LQTS) is a hereditary disorder that leads to sudden cardiac death secondary to fatal cardiac arrhythmias. Although many genes for LQTS have been described, the etiology remains unknown in 30%-40% of cases. In the present study, a large Chinese family (four generations, 49 individuals) with autosomal-dominant LQTS was clinically evaluated. Genome-wide linkage analysis was performed by using polymorphic microsatellite markers to map the genetic locus, and positional candidate genes were screened by sequencing for mutations. The expression pattern and functional characteristics of the mutated protein were investigated by western blotting and patch-clamp electrophysiology. The genetic locus of the LQTS-associated gene was mapped to chromosome 11q23.3-24.3. A heterozygous mutation (Kir3.4-Gly387Arg) was identified in the G protein-coupled, inwardly rectifying potassium channel subunit Kir3.4, encoded by the KCNJ5 gene. The Kir3.4-Gly387Arg mutation was present in all nine affected family members and absent in 528 ethnically matched controls. Western blotting of human cardiac tissue demonstrated significant Kir3.4 expression levels in the cardiac ventricles. Heterologous expression studies with Kir3.4-Gly387Arg revealed a loss-of-function electrophysiological phenotype resulting from reduced plasma membrane expression. Our findings suggest a role for Kir3.4 in the etiology of LQTS.


Subject(s)
G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , Adolescent , Adult , Aged , Aminophylline , Atropine , Chromosome Mapping , Drug Combinations , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Genetic Linkage , Humans , Infant, Newborn , Long QT Syndrome/genetics , Male , Middle Aged , Mutation , Myocardium/metabolism , Nitroglycerin , Papaverine , Pedigree , Phenobarbital
18.
Biochem J ; 413(2): 251-9, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18402552

ABSTRACT

HAI-1 [HGF (hepatocyte growth factor) activator inhibitor-1] is a Kunitz-type transmembrane serine protease inhibitor that forms inhibitor complexes with the trypsin-like serine protease, matriptase. HAI-1 is essential for mouse placental development and embryo survival and together with matriptase it is a key regulator of carcinogenesis. HAI-1 is expressed in polarized epithelial cells, which have the plasma membrane divided by tight junctions into an apical and a basolateral domain. In the present study we show that HAI-1 at steady-state is mainly located on the basolateral membrane of both Madin-Darby canine kidney cells and mammary gland epithelial cells. After biosynthesis, HAI-1 is exocytosed mainly to the basolateral plasma membrane from where 15% of the HAI-1 molecules are proteolytically cleaved and released into the basolateral medium. The remaining membrane-associated HAI-1 is endocytosed and then recycles between the basolateral plasma membrane and endosomes for hours until it is transcytosed to the apical plasma membrane. Minor amounts of HAI-1 present at the apical plasma membrane are proteolytically cleaved and released into the apical medium. Full-length membrane-bound HAI-1 has a half-life of 1.5 h and is eventually degraded in the lysosomes, whereas proteolytically released HAI-1 is more stable. HAI-1 is co-localized with its cognate protease, matriptase, at the basolateral plasma membrane. We suggest that HAI-1, in addition to its protease inhibitory function, plays a role in transporting matriptase as a matriptase-HAI-1 complex from the basolateral plama membrane to the apical plasma membrane, as matriptase is known to interact with prostasin, located at the apical plasma membrane.


Subject(s)
Gene Expression Regulation , Membrane Glycoproteins/physiology , Animals , Cell Line , Cell Membrane/metabolism , Dogs , Endocytosis , Exocytosis , Lysosomes/metabolism , Mammary Glands, Animal/metabolism , Membrane Glycoproteins/metabolism , Mice , Models, Biological , Protein Transport , Proteinase Inhibitory Proteins, Secretory , Serine Endopeptidases/metabolism
19.
Biochem J ; 405(3): 429-38, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17456045

ABSTRACT

The remarkably high specificity of the coagulation proteases towards macromolecular substrates is provided by numerous interactions involving the catalytic groove and remote exosites. For FVIIa [activated FVII (Factor VII)], the principal initiator of coagulation via the extrinsic pathway, several exosites have been identified, whereas only little is known about the specificity dictated by the active-site architecture. In the present study, we have profiled the primary P4-P1 substrate specificity of FVIIa using positional scanning substrate combinatorial libraries and evaluated the role of the selective active site in defining specificity. Being a trypsin-like serine protease, FVIIa had P1 specificity exclusively towards arginine and lysine residues. In the S2 pocket, threonine, leucine, phenylalanine and valine residues were the most preferred amino acids. Both S3 and S4 appeared to be rather promiscuous, however, with some preference for aromatic amino acids at both positions. Interestingly, a significant degree of interdependence between the S3 and S4 was observed and, as a consequence, the optimal substrate for FVIIa could not be derived directly from a subsite-directed specificity screen. To evaluate the role of the active-site residues in defining specificity, a series of mutants of FVIIa were prepared at position 239 (position 99 in chymotrypsin), which is considered to be one of the most important residues for determining P2 specificity of the trypsin family members. This was confirmed for FVIIa by marked changes in primary substrate specificity and decreased rates of antithrombin III inhibition. Interestingly, these changes do not necessarily coincide with an altered ability to activate Factor X, demonstrating that inhibitor and macromolecular substrate selectivity may be engineered separately.


Subject(s)
Factor VIIa/antagonists & inhibitors , Factor VIIa/metabolism , Protein Engineering/methods , Amino Acid Sequence , Humans , Kinetics , Models, Molecular , Protein Conformation , Substrate Specificity
20.
J Cell Sci ; 120(Pt 6): 953-63, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17311847

ABSTRACT

The potassium channel subunits KCNQ2 and KCNQ3 are believed to underlie the M current of hippocampal neurons. The M-type potassium current plays a key role in the regulation of neuronal excitability; however, the subcellular location of the ion channels underlying this regulation has been controversial. We report here that KCNQ2 and KCNQ3 subunits are localized to the axon initial segment of pyramidal neurons of adult rat hippocampus and in cultured hippocampal neurons. We demonstrate that the localization of the KCNQ2/3 channel complex to the axon initial segment is favored by co-expression of the two channel subunits. Deletion of the ankyrin-G-binding motif in both the KCNQ2 and KCNQ3 C-terminals leads to the disappearance of the complex from the axon initial segment, albeit the channel complex remains functional and still reaches the plasma membrane. We further show that although heteromeric assembly of the channel complex favours localization to the axon initial segment, deletion of the ankyrin-G-binding motif in KCNQ2 alone does not alter the subcellular localization of KCNQ2/3 heteromers. By contrast, deletion of the ankyrin-G-binding motif in KCNQ3 significantly reduces AIS enrichment of the complex, implicating KCNQ3 as a major determinant of M channel localization to the AIS.


Subject(s)
Ankyrins/metabolism , Axons/physiology , KCNQ2 Potassium Channel/metabolism , KCNQ3 Potassium Channel/metabolism , Pyramidal Cells/physiology , Amino Acid Motifs , Animals , Binding Sites , COS Cells , Cell Membrane/physiology , Cells, Cultured , Chlorocebus aethiops , Female , Hippocampus/physiology , Ion Channel Gating/physiology , KCNQ2 Potassium Channel/genetics , KCNQ3 Potassium Channel/genetics , Mutation , Neurons/physiology , Pregnancy , Protein Binding , Protein Subunits/genetics , Protein Subunits/metabolism , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL