Marys Medicine

 

Doi:10.1016/j.bbrc.2007.12.168

Available online at www.sciencedirect.com Biochemical and Biophysical Research Communications 367 (2008) 687–692 Lysosome mediated Kir2.1 breakdown directly influences inward rectifier current density John A. Jansen, Teun P. de Boer, Rianne Wolswinkel, Toon A.B. van Veen, Marc A. Vos, Harold V.M. van Rijen, Marcel A.G. van der Heyden * Department of Medical Physiology, Division Heart & Lungs, University Medical Center Utrecht, Yalelaan 50, 3584 CM Utrecht, The Netherlands Received 18 December 2007 Available online 7 January 2008 The inward rectifier current generated by Kir2.1 ion channel proteins is primarily responsible for the stable resting membrane poten- tial in various excitable cell types, like neurons and myocytes. Tight regulation of Kir2.1 functioning prevents premature action potentialformation and ensures optimal repolarization times. While Kir2.1 forward trafficking has been addressed in a number of studies, its deg-radation pathways are thus far unknown. Using three different lysosomal inhibitors, NH4Cl, chloroquine and leupeptin, we now dem-onstrate involvement of the lysosomal degradation pathway in Kir2.1 breakdown. Upon application of the inhibitors, increased steadystate protein levels are detectable within few hours coinciding with intracellular granular Kir2.1 accumulation. Treatment for 24 h witheither chloroquine or leupeptin results in increased plasmamembrane originating inward rectifier current densities, while current–voltagecharacteristics remain unaltered. We conclude that the lysosomal degradation pathway contributes to Kir2.1 mediated inward rectifiercurrent regulation.
Ó 2007 Elsevier Inc. All rights reserved.
Keywords: Inward rectifier; Kir2.1; Lysosome; Chloroquine; Degradation The formation of an action potential (AP) stands at the izing currents) and fine tuning of final repolarization basis of many physiological processes. A number of dis- (directly, contributing repolarizing current) of the AP tinct diseases of the cardiovascular system (atrial or ven- In mammals, several different but closely related ion chan- nel proteins constitute the cardiac ventricular IK1 channel.
migraine) and the motor system (myotonia) can be related Of these, the KCNJ2 and KCNJ12 gene products Kir2.1 to ion channel malfunctioning The AP of excitable and Kir2.2 are the main determinants. To function as an cells, such as neurons and myocytes, is the resultant of ion channel, Kir2.x proteins form either homotypic or het- sequential and coordinated activity of a number of ion erotypic tetramers defined by specific sequence domains channels. Depolarizing currents are generally carried by Several studies indicate that manipulating IK1 by means of sodium and calcium ions, while repolarizing currents result null mutation overexpression or dominant nega- mainly from potassium fluxes. The inward rectifier potas- tive expression of Kir2.1 elucidates the importance sium current (IK1) is one of the few that operates between of Kir2.1 mediated IK1 for normal AP formation and con- subsequent APs and is primarily responsible for generating trol of sinus rhythm. The ultimate expression level of and stabilizing the resting membrane potential at a rather Kir2.1 may influence the eventual arrhythmogenic out- negative level between 90 mV, and secondly come. In humans, loss-of-function mutations in Kir2.1 lead for the initial depolarization (indirectly, opposing depolar- to Andersen–Tawil syndrome which is characterized bypotentially lethal ventricular arrhythmias, periodic paraly- sis and dysmorphic features, further emphasizing the pleio- Corresponding author. Fax: +31 30 2539036.
tropic action of Kir2.1 in development and adult E-mail address: (M.A.G. van der physiology .
0006-291X/$ - see front matter Ó 2007 Elsevier Inc. All rights reserved.
doi:10.1016/j.bbrc.2007.12.168 J.A. Jansen et al. / Biochemical and Biophysical Research Communications 367 (2008) 687–692 Trafficking of potassium channels involves a large num- PAGE and blotted onto nitrocellulose membrane (Bio-Rad, Veenendaal, ber of subsequent steps, all of which are subject to tight reg- The Netherlands). Blots were incubated with GFP (cat. no. Sc-9996; SantaCruz Biotechnology, Santa Cruz, CA, USA) and Pan-Cadherin (cat. no.
ulation . Several elegant studies have addressed C1821; Sigma) primary antibodies and peroxidase-conjugated secondary molecular mechanisms of Kir2.x trafficking towards and antibody (Jackson ImmunoResearch, West Grove, PA, USA). Standard anchoring at the plasma membrane. This disclosed several ECL procedure was used as final detection (Amersham Bioscience, Kir2.x intracellular N- and C-terminal domains and inter- acting proteins involved in endoplasmic reticulum retention, Immunofluorescence microscopy. HEK-KWGF or HEK-HsKir2.1 cells cultured on Ø 11 mm glass cover slips (Smethwick, Warley, UK) were fixed forward trafficking and plasma membrane targeting using 4% paraformaldehyde. After permeabilization with 0.2% Triton X- Several human KCNJ2 mutations that display trafficking 100 (BDH) in PBS, cells were pre-incubated with 2% BSA. Next, cells were defects have been identified , but interestingly these incubated overnight with either anti-GFP or anti-Kir2.1 (cat. no. Sc-18708; mutations are not in the previously mentioned export signal Santa Cruz Biotechnology, Santa Cruz, CA, USA) primary antibody for sequences. Finally, celastrol, a bioactive compound with HEK-KWGF and HEK-HsKir2.1 cells, respectively, followed by incuba-tion with anti-mouse or anti-goat FITC conjugated secondary antibody strong antioxidant characteristics, inhibits Kir2.1 trafficking (Jackson ImmunoResearch) for 2 h. Cover slips were mounted with Vec- towards the plasmamembrane in HEK293 cells As tashield (Vector Laboratories Inc., Burlingame, CA, USA) and imaged demonstrated by Tong et al., tyrosine242 of Kir2.1 is using a Nikon Optiphot-2 microscope equipped for epifluorescence.
involved in clathrin mediated endocytosis, by a tyrosine Electrophysiology. IK1-currents were recorded using the whole cell kinase dependent mechanism . However, beyond endo- voltage clamp configuration in randomly chosen single HEK-KWGF cellsusing an Axopatch 200B amplifier (Molecular Devices, Toronto, Canada).
cytosis no experimental evidence has been described on the Currents were low-pass filtered at 2 kHz and recorded at 4 kHz using a pathways involved in Kir2.x degradation thus far. In this Apple PowerMac fitted with A/D card (National Instruments, Austin, study we focused on the lysosomal degradation pathway TX, USA). From a holding potential of 40 mV, 750 ms long square test by using three different inhibitors of lysosomal breakdown pulses to potentials ranging between 100 and +50 mV were applied to . The lysosomal degradation pathway starts in endo- elicit membrane currents. Steady state currents at the end of the test pulsewere normalized to membrane capacitance and plotted versus test somes containing trapped membrane proteins, followed by potential. To obtain membrane conductances, the slope of I–V plot fusion with early lysosomes which results in mature lyso- 80 mV was determined using linear regression.
somes (for reviews see ). The acid environment in Experiments were done at 20 °C using an extracellular solution con- the lysosomes is required for protein digestion by acidic taining (mmol/L) 140 NaCl, 17.5 NaCO3, 15 Hepes, 6 glucose, 5.4 KCl, hydrolases. We used the lysosomal protease inhibitors chlo- 1.8 CaCl2 and 1 MgCl2 in H2O (pH 7.20, NaOH). Patch pipettes werefilled with an internal solution containing (mmol/L) 125 potassium glu- roquine, ammoniumchloride (NH4Cl) and leupeptin. Chlo- conate, 10 KCl, 5 Hepes, 5 EGTA, 4 Na2ATP, 2 MgCl2 and 0.6 CaCl2 in roquine, an antimalarial drug, and NH4Cl are weak bases, H2O (pH 7.20, KOH) and had resistances ranging between 2 and 5 MX.
increasing the lysosomal pH and thereby prohibiting the Liquid junction potential was calculated using pClamp (Molecular Devi- breakdown of proteins by hydrolases. Furthermore, both ces) and used for offline correction.
chemicals inhibit the transport of hydrolases to the lyso- Statistics. All data are presented as mean ± SEM. Differences among groups were evaluated using one-way ANOVA and a post-hoc Holm– somes. Leupeptin, on the other hand, acts directly as an Sidak test, significance was assumed if p < 0.05.
inhibitor of the hydrolases. Interestingly, leupeptin appearsto affect only the lysosomes, whereas NH4Cl and chloro-quine influence both the lysosomes and the endosomes. In this study we demonstrate that inhibition of lysosomal pro-tein breakdown results in increased steady state level and Inhibition of lysosomal breakdown pathways increases Kir2.1 intracellular accumulation of Kir2.1 protein, and elevated steady state levels IK1 current densities.
HEK-KWGF cells were incubated with either 1 mM NH4Cl, 10 lM chloroquine or 5 lg/mL leupeptin for 6 Materials and methods and 24 h, respectively. Subsequently, expression level ofKir2.1–GFP fusion protein was analyzed by Western blot- Cell culture and pharmacological treatment. HEK-KWGF cells stably ting. As depicted in A, chloroquine rapidly increased expressing wildtype murine Kir2.1–GFP fusion protein were generatedand cultured as described previously . HEK-HsKir2.1 cells stably Kir2.1–GFP expression levels within 6 h which was even express non-tagged human Kir2.1 from a pcDNA3 (InVitrogen, Breda, more pronounced after 24 h. Leupeptin resulted in The Netherlands) based expression vector. For lysosomal degradation enhanced Kir2.1–GFP following 6 and 24 h of incubation.
pathway inhibition, NH4Cl (BDH, Poole, UK), chloroquine (Sigma, St.
Only modest increased Kir2.1–GFP levels were observed Louis, MO, USA) and leupeptin (Sigma) were added to the culture medium to obtain final concentrations of 1 mM, 10 lM and 5 lg/mL, 4Cl application. Following the strong upregulation respectively for the indicated time at the end of the experiment until due to chloroquine or leupeptin treatment, an additional harvesting or electrophysiological recording.
signal is observed at 70 kDa. Since the fusion protein is Western blotting. HEK-KWGF cells were lysed in RIPA buffer detected by an antibody directed against the C-terminal (20 mM Tris–HCl, pH 7.4, 150 mM NaCl, 10 mM Na2HPO4, 1% (v/v) fused GFP, this product most likely represents a protein Triton X-100, 1% (w/v) Na-deoxycholate, 0.1% (w/v) SDS, 1mM EDTA, product that results from N-terminal Kir2.1 cleavage. To 50 mM NaF, 2 mM PMSF and 14 lg/ml aprotinin). Lysates were clarifiedby centrifugation at 14,000 rpm for 10 min at 4 °C and mixed with loading further elucidate the kinetics of Kir2.1 upregulation, buffer. Twenty micrograms of proteins were separated by 10% SDS– HEK-KWGF cells were treated with 10 lM chloroquine






J.A. Jansen et al. / Biochemical and Biophysical Research Communications 367 (2008) 687–692 for 1, 2, 3, 4, 6 and 24 h, respectively (B). Increasedexpression levels of full-length Kir2.1–GFP were observedalready after 1 h of treatment. Furthermore, the presumed70 kDa degradation product becomes detectable follow-ing 3 h of chloroquine treatment. These data indicate thatthe lysosomal degradation pathway is involved in Kir2.1–GFP breakdown, and that Kir2.1–GFP turnover takesplace at a time scale of only a few hours.
Lysosomal inhibition results in granular intracellular Kir2.1accumulation Next, we assessed Kir2.1–GFP localization following application of the different inhibitors for 6 and 24 h. Innon-treated cells, Kir2.1–GFP is mainly localized at theplasmamembrane. Chloroquine incubation leads to strongintracellular accumulation A). Similar results wereobtained using non-tagged Kir2.1 Like for chlo-roquine, incubation with leupeptin or NH4Cl results inintracellular accumulation of Kir2.1–GFP and appearedto increase plasmamembrane staining (We con-clude that the strong increase in Kir2.1–GFP levels in chlo-roquine, and to a lesser extend leupeptin and NH4Cl,treated cells substantially results from intracellular granu-lar accumulation, presumably in late endosomes and/orlysosomes .
Fig. 2. Intracellular Kir2.1 accumulation upon treatment of HEK- Hrs 10 μM Chloroquine
KWGF or HEK-HsKir2.1 cells with lysosomal inhibitors. (A) Cellcultures were treated as in . Subsequently, Kir2.1–GFP was detected in fixed cells using anti-GFP as primary and Fitc-labeled secondaryantibody. Scale bar represents 40 lm. (B) Intracellular accumulation ofnon-tagged Kir2.1 (green) upon treatment of HEK-HsKir2.1 cells with 10 lM chloroquine for 2 and 4 h, respectively. Kir2.1 was detected by anti-Kir2.1 antibody, nuclei are stained with DAPI (blue). Scale bar represents Fig. 1. Effect of lysosomal degradation inhibitors on Kir2.1–GFP steady 20 lm. (For interpretation of the references in color in this figure legend, state expression levels. (A) Kir2.1–GFP protein expression levels in HEK- the reader is referred to the web version of this article.) KWGF cells increase upon treatment with different lysosomal inhibitors.
Inhibition of lysosomal mediated Kir2.1 breakdown results in HEK-KWGF cell cultures were treated with 1 mM NH4Cl, 10 lMchloroquine or 5 lg/mL leupeptin for 6 and 24 h, respectively. Kir2.1– increased IK1 densities GFP protein level in total cell lysates was detected using GFP antibody.
(B) Kinetics of Kir2.1–GFP upregulation following treatment with 10 lM To see whether the increased protein levels as observed chloroquine. Cadherin expression levels were identical to total protein by Western blotting and intracellular accumulation would profiles as detected by Ponceau S staining prior to immunodetection (not also result in enhanced functional Kir2.1–GFP expression shown), and are regarded as loading control using a pan-cadherinantibody.
at the plasmamembrane, HEK-KWGF cells treated with J.A. Jansen et al. / Biochemical and Biophysical Research Communications 367 (2008) 687–692 chloroquine or leupeptin were analyzed for IK1 current plasmamembrane. Apparently, our 24 h treatment with densities (As depicted in A and B, chloro- chloroquine and leupeptin results in saturation of the quine significantly increased the inward component of IK1 Kir2.1 degradation pathway which likely affects internali- at membrane potentials between zation capacity of the functional Kir2.1 channels, culmi- both chloroquine and leupeptin significantly increased the nating in increased IK1 densities. Preliminary studies outward component of IK1 between membrane potentials indicated that NH4Cl did not result in a significant increase 65 mV Furthermore, the reversal in IK1 densities, which is in line with the biochemical potential was not changed, which was reflected by a lack results. Furthermore, chloroquine and NH4Cl display the same mechanism of action with respect to lysosomal degra- 74.9 ± 0.6 mV for control, chloroquine dation inhibition although with different efficiencies. In and leupeptin, respectively).
contrast to our results, transiently transfected Kir6.2 chan-nels in COS cells do not display increased cell surface expression upon 6 or 12 h of chloroquine treatment .
This may be due to different methodology between their In the current study we provide biochemical, immuno- (chemiluminescence) and our (patch clamp) study to detect fluorescence and electrophysiological evidence for involve- ion channel expression at the plasmamembrane. On the ment of the lysosomal degradation pathway in Kir2.1 other hand, Kir6.2 channels may be degraded by a different breakdown. Results of Tong et al. suggest that pathway or multiple pathways, or at a different time scale.
Kir2.1 channels enter the degradation pathway via clathrin Finally, Kv1.5 voltage gated channel is degraded via the mediated endocytosis. Whether all Kir2.1 channels are sub- proteasomal pathway instead of the lysosomal pathway sequently degraded via the lysosomal pathway is unknown.
and blocking this degradation pathway results in increased Alternatively, internalized channels may become targeted IKur current densities Obviously, no universal potas- to proteasome mediated degradation or recycle to the sium ion channel protein degradation pathway seems to increased steady state expression levels within 1 h of treat- Chloroquine and leupeptin treatment yields a product ment with lysosomal inhibitors (B). This implicates a that is approximately 10 kDa less in molecular weight relatively rapid turnover, within hours, of the Kir2.1 ion than full-length Kir2.1–GFP. Since GFP is fused to the channel protein in HEK293 cells. Furthermore, lysosomal C-terminus of Kir2.1, we reason that this product is inhibition results in intracellular Kir2.1 protein accumula- the result of N-terminal cleavage. It might have a much tion as depicted in within 2 h, however this does not shorter half-life than full length protein and therefore necessarily imply a retarded internalization from the only becomes detectable following maximal inhibition Fig. 3. Quantification of functional membrane expression of Kir2.1 using the whole cell voltage clamp technique. (A) Diagram depicting the relationbetween steady state IK1 current and test potential. Compared to controls, inward IK1 currents are larger in chloroquine and leupeptin treated HEK-KWGF cell cultures. Asterisk indicates a significant difference between control and chloroquine (p < 0.05). (B) Steady state conductance density negativeto Ek were significantly increased in chloroquine treated cultures, underscoring increased membrane localization of Kir2.1 channels (p < 0.05). (C) Detailof outward currents depicted in (A); legend as in (A). Both chloroquine and leupeptin treated cultures show significantly increased outward current (* and#, p < 0.05).
J.A. Jansen et al. / Biochemical and Biophysical Research Communications 367 (2008) 687–692 of degradation. N-terminal cleavage might be the first [10] D.F. Steele, J. Eldstrom, D. Fedida, Mechanisms of cardiac potas- step in Kir2.1 degradation.
sium channel trafficking, J. Physiol. 582 (2007) 17–26.
[11] D. Ma, N. Zerangue, Y.-F. Lin, A. Collins, M. Yu, Y.N. Jan, L.Y.
Chloroquine has a long clinical history as an antima- Jan, Role of ER export signals in controlling surface potassium larial drug and is associated with cardiac rhythm and channel numbers, Science 291 (2001) 316–319.
conduction disturbances that can evolve in life-threaten- [12] C. Stockklausner, J. Ludwig, J.P. Ruppersberg, N. Klo¨cker, A ing arrhythmias In feline purkinje and ventricular sequence motif responsible for ER export and surface expression of cardiomyocytes, acute application of chloroquine results Kir2.0 inward rectifier K+ channels, FEBS Lett. 493 (2001) 129–133.
in block of various cardiac ion channels including the [13] C. Stockklausner, N. Klo¨cker, Surface expression of inward rectifier IK1 channel. In our studies, chloroquine acts at a potassium is controlled by selective golgi export, J. Biol. Chem. 278 completely different cellular level as a lysosomal inhibitor displaying opposite biological effects, i.e. an increase in [14] A. Hofherr, B. Fakler, N. Klo¨cker, Selective golgi export of Kir2.1 controls the stoichiometry of functional Kir2.x channel heteromers, J.
K1 current densities. Our electrophysiological measure- Cell Sci. 118 (2005) 1935–1943.
ments were performed in the absence of the drug and [15] D. Leonoudakis, L.R. Conti, S. Anderson, C.M. Radeke, L.M.M.
thereby exclude the acute effect of chloroquine on McGuire, M.E. Adams, S.C. Froehner, J.R. Yates III, C.A.
Kir2.1 based ion channels. These data indicate that chlo- Vandenberg, Protein trafficking and anchoring complexes revealed roquine can display a dual effect on inward rectifier cur- by proteomic analysis of inward rectifier potassium channel rents, directly by blocking the channel, and indirectly by (Kir2.x)-associated proteins, J. Biol. Chem. 279 (2004) 22331–22346.
inhibition of channel degradation. The resultant of long [16] A. Grishin, H. Li, E.S. Levitan, E. Zaks-Makhina, Identification of c- term exposure to chloroquine in a complex system as aminobutyric acid receptor-interacting factor 1 (TRAK2) as a the intact cardiomyocyte or heart, however, is difficult trafficking factor for the K+ channel Kir2.1, J. Biol. Chem. 281 to predict and requires a comprehensive analysis of spa- tial and functional expression of Kir2.x isoforms.
[17] L.J. Sampson, M.L. Leyland, C. Dart, Direct interaction between the actin-binding protein filamin-A and the inwardly rectifying potassiumchannel, Kir2.1, J. Biol. Chem. 278 (2003) 41988–41997.
[18] S. Bendahhou, M.R. Donaldson, N.M. Plaster, M. Tristan-Firouzi, Y.-H. Fu, L.J. Pta´cˇek, Defective potassium channel Kir2.1 trafficking This work was supported by the Netherlands Heart underlies Andersen–Tawil syndrome, J. Biol. Chem. 278 (2003) Foundation (2005B170, JJ) and the Prof. Dr. R.L.J. van [19] L.Y. Ballester, D.W. Benson, B. Wong, I.H. Law, K.D. Mathews, Ruyven Foundation (RW).
C.G. Vanoye, A.L. George Jr., Trafficking-competent and trafficking-defective KCNJ2 mutations in Andersen syndrome, Hum. Mutat. 27 [20] H. Sun, X. Liu, Q. Xiong, S. Shikano, M. Li, Chronic inhibition of [1] F.M. Ashcroft, From molecule to malady, Nature 440 (2006) 440– cardiac Kir2.1 and hERG potassium channels by celastrol with dual effects on both ion conductivity and protein trafficking, J. Biol. Chem.
[2] A.S. Dhamoon, J. Jalife, The inward rectifier current (I 281 (2006) 5877–5884.
cardiac excitability and is involved in arrhythmogenesis, Heart [21] M.A.G. van der Heyden, M.E. Smits, M.A. Vos, Drugs and Rhythm 2 (2005) 316–324.
trafficking of ion channels: a new pro-arrhythmic threat on the [3] A. Tinker, Y.N. Jan, L.Y. Jan, Regions responsible for the assembly horizon? Br. J. Pharmacol. doi: 10.1038/sj.bjp.0707618.
of inwardly rectifying potassium channels, Cell 87 (1996) 857–868.
[22] Y. Tong, G.S. Brandt, M. Li, G. Shapovalov, E. Slimko, A. Karschin, [4] J.J. Zarisky, J.B. Redell, B.L. Tempel, T.L. Schwarz, The conse- D.A. Dougherty, H.A. Lester, Tyrosine decaging leads to substantial quences of disrupting cardiac inward rectifying K+ current (I membrane trafficking during modulation of an inward rectifier revealed by the targeted deletion of the murine Kir2.1 and Kir2.2 potassium channel, J. Gen. Physiol. 117 (2001) 103–118.
genes, J. Physiol. 533 (2001) 697–710.
[23] P.O. Seglen, B. Grinde, A.E. Solheim, Inhibition of the lysosomal [5] J. Li, M. McLerie, A.N. Lopatin, Transgenic upregulation of I pathway of protein degradation in isolated rat hepatocytes by the mouse heart leads to multiple abnormalities of cardiac ammonia, methylamine, chloroquine and leupeptin, Eur. J. Biochem.
excitability, Am. J. Physiol. Heart Circ. Physiol. 287 (2004) 95 (1979) 215–225.
[24] A. Hershko, A. Ciechanover, Mechanisms of intracellular protein [6] J. Miake, E. Marba´n, H.B. Nuss, Functional role of inward rectifier breakdown, Ann. Rev. Biochem. 51 (1982) 335–364.
current in heart probed by Kir2.1 overexpression and dominant- [25] C.S. Pillay, E. Elliot, C. Dennison, Endolysosomal proteolysis and its negative suppression, J. Clin. Invest. 111 (2003) 1529–1536.
regulation, Biochem. J. 363 (2002) 417–429.
[7] L. Piao, J. Li, M. McLerie, A.N. Lopatin, Transgenic upregulation of [26] T.P. de Boer, T.A.B. van Veen, M.J.C. Houtman, J.A. Jansen, S.C.M. van Amersfoorth, P.A. Doevendans, M.A. Vos, M.A.G. van K1 in the mouse heart is proarrhythmic, Basic Res. Cardiol. 102 (2007) 416–428.
der Heyden, Inhibition of cardiomyocyte automaticity by electrotonic [8] M. McLerie, A.N. Lopatin, Dominant-negative suppression of I application of inward rectifier current from Kir2.1 expressing cells, the mouse heart leads to altered cardiac excitability, J. Mol. Cell.
Med. Biol. Eng. Comput. 44 (2006) 537–542.
Cardiol. 35 (2003) 367–378.
[27] G. Taschenberger, A. Mougey, S. Shen, L.B. Lester, S. LaFranchi, S.- [9] N.M. Plaster, R. Tawil, M. Tristani-Firouzi, S. Canu´n, S. Bendah- L. Shyng, Identification of a familial hyperinsulinism-causing muta- hou, A. Tsunoda, M.R. Donaldson, S.T. Iannaccone, E. Brunt, R.
tion in the sulfonylurea receptor 1 that prevents normal trafficking Barohn, J. Clark, F. Deymeer, A.L. George Jr., F.A. Fish, A. Hahn, and function of KATP channels, J. Biol. Chem. 277 (2002) 17139– A. Nitu, C. Ozdemir, P. Serdaroglu, S.H. Subramony, G. Wolfe, Y.- H. Fu, L.J. Pta´cˇek, Mutations in Kir2.1 cause the developmental and [28] M. Kato, K. Ogura, J. Miake, N. Sasaki, S. Taniguchi, O. Igawa, A.
episodic electrical phenotypes of Andersen's syndrome, Cell 105 Yoshida, Y. Hoshikawa, M. Murata, E. Nanba, Y. Kurata, Y.
(2001) 511–519.
Kawata, H. Ninomiya, T. Morisaki, M. Kitakaze, I. Hisatome, J.A. Jansen et al. / Biochemical and Biophysical Research Communications 367 (2008) 687–692 Evidence for proteasomal degradation of Kv1.5 channel protein, [30] J. Sa´nchez-Chapula, E. Salinas-Stefanon, J. Torres-Ja´come, D.E.
Biochem. Biophys. Res. Commun. 337 (2005) 343–348.
Benavides-Haro, R.A. Navarro-Polanco, Blockade of currents by the [29] N.J. White, Cardiotoxicity of antimalarial drugs, Lancet Infect. Dis. 7 antimalarial drug chloroquine in feline ventricular myocytes, J.
(2007) 549–558.
Pharmacol. Exp. Ther. 297 (2001) 437–445.

Source: http://www.physiol.med.uu.nl/teundeboer/Site/Publications_files/BBRC2008.pdf

Psychiatrist beats suit blaming him for patient's killing mom

Psychiatrist Beats Suit Blaming Him for Patient's Killing Mom Milton Satcher and Laura Strong defended a psychiatrist accused of breaching the standard of care bycutting a patient's medications.John Disney/Daily Report After 10 years and two appeals, the guardians of a mental y disabled man who kil ed his mother weeks after hisdoctor took him off anti-psychotic medications have walked away with nothing from their medical malpractice suit.

chu-tours.fr

de la Société Française de Neurologie Pédiatrique e de Congrès VINCI-TOURS EVENEMENTS Palais des Congrès S. de la VAISSIERE Membres du Bureau Secrétariat du Congrès - Ant Congrès 154 avenue de Lodève / 34070 Montpellier Tél. : 04 67 10 92 23 / E-mail : [email protected] Conception graphique : www.kom-graphik.pro Bienvenue à Tours Toute l'équipe de Neuropédiatrie est heureuse de vous accueillir à Tours du 21 janvier au 24 janvier 2015 pour ce 25ème congrès de la Société Française de Neurologie Pédiatrique. Cette année, nous reprendrons le déroulement habituel de notre congrès : Troubles des Apprentissages le mercredi 21 janvier avec la session de Neuro-Ophtalmologie et de Neuro-ORL qui est une nouveauté de cette édition, Séances plénières en parallèle le jeudi 22 et le vendredi 23 janvier, Congrès du Personnel Soignant et session recherche en parallèle le vendredi. Et enfin commissions scientifiques le samedi 24 janvier au matin. Les thèmes retenus nous permettront de couvrir les principaux champs de la Neurologie pédiatrique : la neurologie fœtale et périnatale, les affections génétiques et du développement, les affections du système nerveux périphérique, les anomalies de la substance blanche, les maladies métaboliques, inflammatoires, et vasculaires, sans oublier bien sûr l'épileptologie qui prendra une large place cette année.Une large place sera faite également aux Communications orales et aux Posters. Nous comptons sur votre présence, votre participation et votre enthousiasme pour faire de ce congrès un succès.Nous sommes très heureux et très fiers de vous accueillir en Touraine, «Jardin de la France».