Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Circ Res ; 114(6): 982-92, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24508725

ABSTRACT

RATIONALE: Kv1.5 (KCNA5) mediates the ultra-rapid delayed rectifier current that controls atrial action potential duration. Given its atrial-specific expression and alterations in human atrial fibrillation, Kv1.5 has emerged as a promising target for the treatment of atrial fibrillation. A necessary step in the development of novel agents that selectively modulate trafficking pathways is the identification of the cellular machinery controlling Kv1.5 surface density, of which little is yet known. OBJECTIVE: To investigate the role of the unconventional myosin-V (MYO5A and MYO5B) motors in determining the cell surface density of Kv1.5. METHODS AND RESULTS: Western blot analysis showed MYO5A and MYO5B expression in the heart, whereas disruption of endogenous motors selectively reduced IKur current in adult rat cardiomyocytes. Dominant negative constructs and short hairpin RNA silencing demonstrated a role for MYO5A and MYO5B in the surface trafficking of Kv1.5 and connexin-43 but not potassium voltage-gated channel, subfamily H (eag-related), member 2 (KCNH2). Live-cell imaging of Kv1.5-GFP and retrospective labeling of phalloidin demonstrated motility of Kv1.5 vesicles on actin tracts. MYO5A participated in anterograde trafficking, whereas MYO5B regulated postendocytic recycling. Overexpression of mutant motors revealed a selective role for Rab11 in coupling MYO5B to Kv1.5 recycling. CONCLUSIONS: MYO5A and MYO5B control functionally distinct steps in the surface trafficking of Kv1.5. These isoform-specific trafficking pathways determine Kv1.5-encoded IKur in myocytes to regulate repolarizing current and, consequently, cardiac excitability. Therapeutic strategies that manipulate Kv1.5 selective trafficking pathways may prove useful in the treatment of arrhythmias.


Subject(s)
Cell Membrane/metabolism , Kv1.5 Potassium Channel/metabolism , Myocytes, Cardiac/metabolism , Myosin Heavy Chains/physiology , Myosin Type V/physiology , Myosins/physiology , Protein Transport/physiology , Actin Cytoskeleton/physiology , Animals , Arrhythmias, Cardiac/physiopathology , Cell Line , Connexin 43/analysis , ERG1 Potassium Channel , Endocytosis , Ether-A-Go-Go Potassium Channels/analysis , Gap Junctions , Genes, Reporter , Heart Conduction System/physiopathology , Ion Transport , Kv1.5 Potassium Channel/genetics , Male , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Models, Cardiovascular , Myosin Heavy Chains/deficiency , Myosin Heavy Chains/genetics , Myosin Type V/deficiency , Myosin Type V/genetics , Myosins/deficiency , Myosins/genetics , Potassium/metabolism , Protein Isoforms/metabolism , RNA Interference , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/metabolism , rab GTP-Binding Proteins/physiology
2.
Circ Res ; 111(7): 842-53, 2012 Sep 14.
Article in English | MEDLINE | ID: mdl-22843785

ABSTRACT

RATIONALE: Kv1.5 (KCNA5) is expressed in the heart, where it underlies the I(Kur) current that controls atrial repolarization, and in the pulmonary vasculature, where it regulates vessel contractility in response to changes in oxygen tension. Atrial fibrillation and hypoxic pulmonary hypertension are characterized by downregulation of Kv1.5 protein expression, as well as with oxidative stress. Formation of sulfenic acid on cysteine residues of proteins is an important, dynamic mechanism for protein regulation under oxidative stress. Kv1.5 is widely reported to be redox-sensitive, and the channel possesses 6 potentially redox-sensitive intracellular cysteines. We therefore hypothesized that sulfenic acid modification of the channel itself may regulate Kv1.5 in response to oxidative stress. OBJECTIVE: To investigate how oxidative stress, via redox-sensitive modification of the channel with sulfenic acid, regulates trafficking and expression of Kv1.5. METHODS AND RESULTS: Labeling studies with the sulfenic acid-specific probe DAz and horseradish peroxidase-streptavidin Western blotting demonstrated a global increase in sulfenic acid-modified proteins in human patients with atrial fibrillation, as well as sulfenic acid modification to Kv1.5 in the heart. Further studies showed that Kv1.5 is modified with sulfenic acid on a single COOH-terminal cysteine (C581), and the level of sulfenic acid increases in response to oxidant exposure. Using live-cell immunofluorescence and whole-cell voltage-clamping, we found that modification of this cysteine is necessary and sufficient to reduce channel surface expression, promote its internalization, and block channel recycling back to the cell surface. Moreover, Western blotting demonstrated that sulfenic acid modification is a trigger for channel degradation under prolonged oxidative stress. CONCLUSIONS: Sulfenic acid modification to proteins, which is elevated in diseased human heart, regulates Kv1.5 channel surface expression and stability under oxidative stress and diverts channel from a recycling pathway to degradation. This provides a molecular mechanism linking oxidative stress and downregulation of channel expression observed in cardiovascular diseases.


Subject(s)
Atrial Fibrillation/metabolism , Kv1.5 Potassium Channel/chemistry , Kv1.5 Potassium Channel/metabolism , Myocardium/metabolism , Sulfenic Acids/metabolism , Amino Acid Sequence , Animals , Atrial Fibrillation/pathology , Case-Control Studies , Cell Line , Cells, Cultured , Humans , Mice , Models, Animal , Molecular Sequence Data , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oxidation-Reduction , Oxidative Stress/physiology , Rats , Reactive Oxygen Species , Signal Transduction/physiology
3.
J Neurosci ; 31(37): 13224-35, 2011 Sep 14.
Article in English | MEDLINE | ID: mdl-21917805

ABSTRACT

Voltage-gated potassium (Kv) channels are critical for neuronal excitability and are targeted to specific subcellular compartments to carry out their unique functions. While it is widely believed that Kv channels exist as heteromeric complexes in neurons, direct tests of the hypothesis that specific heteromeric channel populations display divergent spatial and temporal dynamics are limited. Using a bimolecular fluorescence complementation approach, we monitored the assembly and localization of cell surface channel complexes in living cells. While PSD95-mediated clustering was subunit independent, selective visualization of heteromeric Kv complexes in rat hippocampal neurons revealed subunit-dependent localization that was not predicted by analyzing individual subunits. Assembly of Kv1.1 with Kv1.4 prevented axonal localization but not surface expression, while inclusion of Kv1.2 imparted clustering at presynaptic sites and decreased channel mobility within the axon. This mechanism by which specific Kv channel subunits can act in a dominant manner to impose unique trafficking properties to heteromeric complexes extended to Shab-related family of Kv channels. When coexpressed, Kv2.1 and Kv2.2 heteromultimers did not aggregate in somatodendritic clusters observed with expression of Kv2.1 alone. These studies demonstrate selective axonal trafficking and surface localization of distinct Kv channels based on their subunit composition.


Subject(s)
Axonal Transport/physiology , Protein Subunits/metabolism , Protein Transport/physiology , Shaker Superfamily of Potassium Channels/metabolism , Animals , COS Cells , Cell Membrane/metabolism , Cells, Cultured , Chlorocebus aethiops , Female , Hippocampus/metabolism , Hippocampus/physiology , Male , Membrane Potentials , Neurons/metabolism , Neurons/physiology , Patch-Clamp Techniques/methods , Rats
4.
Am J Physiol Cell Physiol ; 297(3): C759-65, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19587217

ABSTRACT

Enhanced expression of the facilitative glucose transporter, GLUT1, has been shown to inhibit apoptosis in several cell systems including vascular smooth muscle cells (VSMCs). A decrease in apoptosis could lead to increased VSMC numbers in neointimal and medial arterial layers under several pathologic conditions. The hypothesis underlying these studies is that GLUT1 induces expression of antiapoptotic and prosurvival genes that increase VSMC survival. Transcriptomic analysis of A7r5 VSMCs, in which GLUT1 was acutely overexpressed, showed a 2.14-fold increase in c-FLICE inhibitory protein (cFLIP), which promotes cellular growth and prevents apoptosis through caspase 8 binding. We confirmed that overexpression of GLUT1 induced mRNA and protein expression of both the long and short isoforms of cFLIP (cFLIP(L) and cFLIP(S)) in primary and stable immortalized VSMC lines as well as in aortas from GLUT1 transgenic mice. Increased GLUT1 reduced VSMC death by more than twofold after serum withdrawal, as evidenced by decreased caspase 3 activity and Trypan blue exclusion studies. GLUT1 overexpression resulted in a greater than twofold increase in proliferating cell nuclear antigen expression and live cell numbers, consistent with augmented VSMC proliferation. Lentiviral knockdown of cFLIP(L) showed that cFLIP(L) was necessary for the proproliferative and antiapoptotic effects of GLUT1 overexpression. Taken together, these data suggest that GLUT1 induction of cFLIP(L) expression augments proliferation and prevents apoptosis in VSMCs.


Subject(s)
Apoptosis/physiology , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Glucose Transporter Type 1/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Animals , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Cell Line , Cell Proliferation/drug effects , Gene Expression Regulation/physiology , Glucose Transporter Type 1/genetics , Mice , Mice, Transgenic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...