Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
J Biol Chem ; 300(8): 107526, 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38960041

RESUMO

The human ether-a-go-go-related gene (hERG) encodes the Kv11.1 (or hERG) channel that conducts the rapidly activating delayed rectifier potassium current (IKr). Naturally occurring mutations in hERG impair the channel function and cause long QT syndrome type 2. Many missense hERG mutations lead to a lack of channel expression on the cell surface, representing a major mechanism for the loss-of-function of mutant channels. While it is generally thought that a trafficking defect underlies the lack of channel expression on the cell surface, in the present study, we demonstrate that the trafficking defective mutant hERG G601S can reach the plasma membrane but is unstable and quickly degrades, which is akin to WT hERG channels under low K+ conditions. We previously showed that serine (S) residue at 624 in the innermost position of the selectivity filter of hERG is involved in hERG membrane stability such that substitution of serine 624 with threonine (S624T) enhances hERG stability and renders hERG insensitive to low K+ culture. Here, we report that the intragenic addition of S624T substitution to trafficking defective hERG mutants G601S, N470D, and P596R led to a complete rescue of the function of these otherwise loss-of-function mutant channels to a level similar to the WT channel, representing the most effective rescue means for the function of mutant hERG channels. These findings not only provide novel insights into hERG mutation-mediated channel dysfunction but also point to the critical role of S624 in hERG stability on the plasma membrane.

2.
J Biol Chem ; 300(7): 107483, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38897569

RESUMO

The voltage-gated Kv1.5 potassium channel, conducting the ultra-rapid delayed rectifier K+ current (IKur) in human cells, plays important roles in the repolarization of atrial action potentials and regulation of the vascular tone. We previously reported that activation of protein kinase C (PKC) by phorbol 12-myristate 13-acetate (PMA) induces endocytic degradation of cell-surface Kv1.5 channels, and a point mutation removing the phosphorylation site, T15A, in the N terminus of Kv1.5 abolished the PMA-effect. In the present study, using mutagenesis, patch clamp recording, Western blot analysis, and immunocytochemical staining, we demonstrate that ubiquitination is involved in the PMA-mediated degradation of mature Kv1.5 channels. Since the expression of the Kv1.4 channel is unaffected by PMA treatment, we swapped the N- and/or C-termini between Kv1.5 and Kv1.4. We found that the N-terminus alone did not but both N- and C-termini of Kv1.5 did confer PMA sensitivity to mature Kv1.4 channels, suggesting the involvement of Kv1.5 C-terminus in the channel ubiquitination. Removal of each of the potential ubiquitination residue Lysine at position 536, 565, and 591 by Arginine substitution (K536R, K565R, and K591R) had little effect, but removal of all three Lysine residues with Arginine substitution (3K-R) partially reduced PMA-mediated Kv1.5 degradation. Furthermore, removing the cysteine residue at position 604 by Serine substitution (C604S) drastically reduced PMA-induced channel degradation. Removal of the three Lysines and Cys604 with a quadruple mutation (3K-R/C604S) or a truncation mutation (Δ536) completely abolished the PKC activation-mediated degradation of Kv1.5 channels. These results provide mechanistic insight into PKC activation-mediated Kv1.5 degradation.


Assuntos
Canal de Potássio Kv1.5 , Proteína Quinase C , Proteólise , Acetato de Tetradecanoilforbol , Ubiquitinação , Canal de Potássio Kv1.5/metabolismo , Canal de Potássio Kv1.5/genética , Humanos , Proteína Quinase C/metabolismo , Proteína Quinase C/genética , Acetato de Tetradecanoilforbol/farmacologia , Células HEK293 , Animais , Fosforilação , Membrana Celular/metabolismo , Canal de Potássio Kv1.4/metabolismo , Canal de Potássio Kv1.4/genética
3.
Perfusion ; : 2676591241239279, 2024 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-38486379

RESUMO

BACKGROUND: Reduced oxygen delivery (DO2) during cardiopulmonary bypass (CPB) was proposed as a risk factor for the development of postoperative neurological complications (PONCs), including cerebrovascular accidents (CVA), delirium, and postoperative cognitive dysfunction (POCD). We aimed to review the current evidence on the association between intraoperative DO2 and the incidence of PONCs. METHODS: MEDLINE, Embase, the Cochrane Library, and Web of Science were electronically searched to identify comparative studies from inception until July 2023 that reported the association between intraoperative DO2 levels and the incidence of PONCs (as defined by the scales and diagnostic tools utilized by the studies' authors) in adults patients undergoing cardiac surgery using CPB. RESULTS: Of the 2513 papers identified, 10 studies, including 21,875 participants, were included. Of these, three studies reported on delirium, two on POCD, and five on CVA. Eight studies reported reduced intraoperative DO2 in patients who developed delirium and CVA. There was a lack of consensus on the cut-off of DO2 levels or the correlation between the period below these threshold values and the development of PONC. CONCLUSIONS: Limited data suggest that maintaining intraoperative DO2 above the critical threshold levels and ensuring adequate intraoperative cerebral perfusion may play a role in minimizing the incidence of neurological events in adult patients undergoing cardiac surgery on cardiopulmonary bypass.

4.
Mol Pharmacol ; 104(4): 164-173, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37419691

RESUMO

The human ether-a-go-go-related gene (hERG) encodes for the pore-forming subunit of the channel that conducts the rapidly activating delayed K+ current (IKr) in the heart. The hERG channel is important for cardiac repolarization, and reduction of its expression in the plasma membrane due to mutations causes long QT syndrome type 2 (LQT2). As such, promoting hERG membrane expression is a strategy to rescue mutant channel function. In the present study, we applied patch clamp, western blots, immunocytochemistry, and quantitative reverse transcription polymerase chain reaction techniques to investigate the rescue effects of two drugs, remdesivir and lumacaftor, on trafficking-defective mutant hERG channels. As our group has recently reported that the antiviral drug remdesivir increases wild-type (WT) hERG current and surface expression, we studied the effects of remdesivir on trafficking-defective LQT2-causing hERG mutants G601S and R582C expressed in HEK293 cells. We also investigated the effects of lumacaftor, a drug used to treat cystic fibrosis, that promotes CFTR protein trafficking and has been shown to rescue membrane expression of some hERG mutations. Our results show that neither remdesivir nor lumacaftor rescued the current or cell-surface expression of homomeric mutants G601S and R582C. However, remdesivir decreased while lumacaftor increased the current and cell-surface expression of heteromeric channels formed by WT hERG and mutant G601S or R582C hERG. We concluded that drugs can differentially affect homomeric WT and heteromeric WT+G601S (or WT+R582C) hERG channels. These findings extend our understanding of drug-channel interaction and may have clinical implications for patients with hERG mutations. SIGNIFICANCE STATEMENT: Various naturally occurring mutations in a cardiac potassium channel called hERG can impair channel function by decreasing cell-surface channel expression, resulting in cardiac electrical disturbances and even sudden cardiac death. Promotion of cell-surface expression of mutant hERG channels represents a strategy to rescue channel function. This work demonstrates that drugs such as remdesivir and lumacaftor can differently affect homomeric and heteromeric mutant hERG channels, which have biological and clinical implications.


Assuntos
Canais de Potássio Éter-A-Go-Go , Síndrome do QT Longo , Humanos , Canais de Potássio Éter-A-Go-Go/metabolismo , Canal de Potássio ERG1/genética , Células HEK293 , Síndrome do QT Longo/tratamento farmacológico , Síndrome do QT Longo/genética , Síndrome do QT Longo/metabolismo
5.
J Biol Chem ; 296: 100514, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33676894

RESUMO

The voltage-gated potassium channel Kv1.5 plays important roles in the repolarization of atrial action potentials and regulation of the vascular tone. While the modulation of Kv1.5 function has been well studied, less is known about how the protein levels of Kv1.5 on the cell membrane are regulated. Here, through electrophysiological and biochemical analyses of Kv1.5 channels heterologously expressed in HEK293 cells and neonatal rat ventricular myocytes, as well as native Kv1.5 in human induced pluripotent stem cell (iPSC)-derived atrial cardiomyocytes, we found that activation of protein kinase C (PKC) with phorbol 12-myristate 13-acetate (PMA, 10 nM) diminished Kv1.5 current (IKv1.5) and protein levels of Kv1.5 in the plasma membrane. Mechanistically, PKC activation led to monoubiquitination and degradation of the mature Kv1.5 proteins. Overexpression of Vps24, a protein that sorts transmembrane proteins into lysosomes via the multivesicular body (MVB) pathway, accelerated, whereas the lysosome inhibitor bafilomycin A1 completely prevented PKC-mediated Kv1.5 degradation. Kv1.5, but not Kv1.1, Kv1.2, Kv1.3, or Kv1.4, was uniquely sensitive to PMA treatment. Sequence alignments suggested that residues within the N terminus of Kv1.5 are essential for PKC-mediated Kv1.5 reduction. Using N-terminal truncation as well as site-directed mutagenesis, we identified that Thr15 is the target site for PKC that mediates endocytic degradation of Kv1.5 channels. These findings indicate that alteration of protein levels in the plasma membrane represents an important regulatory mechanism of Kv1.5 channel function under PKC activation conditions.


Assuntos
Endocitose , Células-Tronco Pluripotentes Induzidas/metabolismo , Canal de Potássio Kv1.5/metabolismo , Proteína Quinase C/metabolismo , Ubiquitinação , Animais , Animais Recém-Nascidos , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Canal de Potássio Kv1.5/genética , Fosforilação , Proteína Quinase C/genética , Ratos , Transdução de Sinais
6.
J Card Surg ; 37(12): 5371-5378, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36403267

RESUMO

BACKGROUND: Postoperative atrial fibrillation (POAF) is one of the most common complications following cardiac surgery and is associated with increased morbidity. Intraoperative topical amiodarone application on epicardial tissue has been shown to reduce systemic concentrations while maintaining therapeutic myocardial concentrations, thereby, lowering the risk of extracardiac adverse effects associated with oral and intravenous amiodarone therapy. However, the efficacy and safety of topical amiodarone in preventing POAF is unclear. OBJECTIVES: This study summarizes the clinical studies to-date that have investigated the efficacy and safety of topical amiodarone administration in preventing POAF following cardiac surgery. METHODS: A database search was conducted using Medline, Embase, and Cochrane Library to identify relevant studies. Abstracts were screened and data were extracted from relevant full-text articles that met the inclusion and exclusion criteria. RESULTS: The search returned four studies with variable findings on the effect of topical amiodarone therapy on the incidence of POAF, cardiac effects, extracardiac effects, and hospital length of stay. CONCLUSION: Prophylactic topical application of amiodarone may be effective and safe for preventing post-operative new-onset atrial fibrillation. Further investigation is required to evaluate the efficacy and safety of topical amiodadrone therapy before it can be widely integrated into current practice.


Assuntos
Amiodarona , Fibrilação Atrial , Procedimentos Cirúrgicos Cardíacos , Humanos , Amiodarona/efeitos adversos , Fibrilação Atrial/epidemiologia , Fibrilação Atrial/prevenção & controle , Procedimentos Cirúrgicos Cardíacos/efeitos adversos , Complicações Pós-Operatórias/epidemiologia
7.
J Biol Chem ; 295(14): 4723-4732, 2020 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-32122972

RESUMO

The voltage-gated potassium channel Kv1.5 plays important roles in atrial repolarization and regulation of vascular tone. In the present study, we investigated the effects of mechanical stretch on Kv1.5 channels. We induced mechanical stretch by centrifuging or culturing Kv1.5-expressing HEK 293 cells and neonatal rat ventricular myocytes in low osmolarity (LO) medium and then recorded Kv1.5 current (IKv1.5) in a normal, isotonic solution. We observed that mechanical stretch increased IKv1.5, and this increase required the intact, long, proline-rich extracellular S1-S2 linker of the Kv1.5 channel. The low osmolarity-induced IKv1.5 increase also required an intact intracellular N terminus, which contains the binding motif for endogenous Src tyrosine kinase that constitutively inhibits IKv1.5 Disrupting the Src-binding motif of Kv1.5 through N-terminal truncation or mutagenesis abolished the mechanical stretch-mediated increase in IKv1.5 Our results further showed that the extracellular S1-S2 linker of Kv1.5 communicates with the intracellular N terminus. Although the S1-S2 linker of WT Kv1.5 could be cleaved by extracellularly applied proteinase K (PK), an N-terminal truncation up to amino acid residue 209 altered the conformation of the S1-S2 linker and made it no longer susceptible to proteinase K-mediated cleavage. In summary, the findings of our study indicate that the S1-S2 linker of Kv1.5 represents a mechanosensor that regulates the activity of this channel. By targeting the S1-S2 linker, mechanical stretch may induce a change in the N-terminal conformation of Kv1.5 that relieves Src-mediated tonic channel inhibition and results in an increase in IKv1.5.


Assuntos
Canal de Potássio Kv1.5/metabolismo , Potenciais da Membrana/fisiologia , Estresse Mecânico , Sequência de Aminoácidos , Animais , Sítios de Ligação , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Glicosilação , Células HEK293 , Humanos , Canal de Potássio Kv1.5/química , Canal de Potássio Kv1.5/genética , Células Musculares/citologia , Células Musculares/metabolismo , Pressão Osmótica , Domínios Proteicos , Pirazóis/farmacologia , Pirimidinas/farmacologia , Ratos , Ratos Sprague-Dawley , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo
8.
J Pharmacol Exp Ther ; 377(2): 265-272, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33674391

RESUMO

Drug-induced long QT syndrome (LQTS) is an established cardiac side effect of a wide range of medications and represents a significant concern for drug safety. The rapidly and slowly activating delayed rectifier K+ currents, mediated by channels encoded by the human ether-a-go-go-related gene (hERG) and KCNQ1 + KCNE1, respectively, are two main currents responsible for ventricular repolarization. The common cause for drugs to induce LQTS is through impairing the hERG channel. For the recent emergence of COVID-19, caused by severe acute respiratory syndrome coronavirus 2, several drugs have been investigated as potential therapies; however, there are concerns about their QT prolongation risk. Here, we studied the effects of chloroquine, hydroxychloroquine, azithromycin, and remdesivir on hERG channels. Our results showed that although chloroquine acutely blocked hERG current (IhERG), with an IC50 of 3.0 µM, hydroxychloroquine acutely blocked IhERG 8-fold less potently, with an IC50 of 23.4 µM. Azithromycin and remdesivir did not acutely affect IhERG When these drugs were added at 10 µM to the cell culture medium for 24 hours, remdesivir increased IhERG by 2-fold, which was associated with an increased mature hERG channel expression. In addition, these four drugs did not acutely or chronically affect KCNQ1 + KCNE1 channels. Our data provide insight into COVID-19 drug-associated LQTS and cardiac safety concerns. SIGNIFICANCE STATEMENT: This work demonstrates that, among off-label potential COVID-19 treatment drugs chloroquine, hydroxychloroquine, azithromycin, and remdesivir, the former two drugs block hERG potassium channels, whereas the latter two drugs do not. All four drugs do not affect KCNQ1 + KCNE1. As hERG and KCNQ1 + KCNE1 are two main K+ channels responsible for ventricular repolarization, and most drugs that induce long QT syndrome (LQTS) do so by impairing hERG channels, these data provide insight into COVID-19 drug-associated LQTS and cardiac safety concerns.


Assuntos
Monofosfato de Adenosina/análogos & derivados , Alanina/análogos & derivados , Azitromicina/farmacologia , Tratamento Farmacológico da COVID-19 , Cloroquina/farmacologia , Canal de Potássio ERG1/antagonistas & inibidores , Hidroxicloroquina/farmacologia , Monofosfato de Adenosina/farmacologia , Monofosfato de Adenosina/uso terapêutico , Alanina/farmacologia , Alanina/uso terapêutico , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Antimaláricos/farmacologia , Antimaláricos/uso terapêutico , Antivirais/farmacologia , Antivirais/uso terapêutico , Azitromicina/uso terapêutico , COVID-19/metabolismo , Cloroquina/uso terapêutico , Relação Dose-Resposta a Droga , Canal de Potássio ERG1/metabolismo , Células HEK293 , Humanos , Hidroxicloroquina/uso terapêutico , Bloqueadores dos Canais de Potássio/farmacologia , Bloqueadores dos Canais de Potássio/uso terapêutico
9.
Mol Pharmacol ; 98(4): 508-517, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32321735

RESUMO

Human ether-a-go-go-related gene (hERG) encodes the pore-forming subunit of the rapidly activating delayed rectifier potassium current (IKr) important for repolarization of cardiac action potentials. Drug-induced disruption of hERG channel function is a main cause of acquired long QT syndrome, which can lead to ventricular arrhythmias and sudden death. Illicit fentanyl use is associated with sudden death. We have demonstrated that fentanyl blocks hERG current (IhERG) at concentrations that overlap with the upper range of postmortem blood concentrations in fentanyl-related deaths. Since fentanyl can cause respiratory depression and electrolyte imbalances, in the present study we investigated whether certain pathologic circumstances exacerbate fentanyl-induced block of IhERG Our results show that chronic hypoxia or hypokalemia additively reduced IhERG with fentanyl. As well, high pH potentiated the fentanyl-mediated block of hERG channels, with an IC50 at pH 8.4 being 7-fold lower than that at pH 7.4. Furthermore, although the full-length hERG variant, hERG1a, has been widely used to study hERG channels, coexpression with the short variant, hERG1b (which does not produce current when expressed alone), produces functional hERG1a/1b channels, which gate more closely resembling native IKr Our results showed that fentanyl blocked hERG1a/1b channels with a 3-fold greater potency than hERG1a channels. Thus, in addition to a greater susceptibility due to the presence of hERG1b in the human heart, hERG channel block by fentanyl can be exacerbated by certain conditions, such as hypoxia, hypokalemia, or alkalosis, which may increase the risk of fentanyl-induced ventricular arrhythmias and sudden death. SIGNIFICANCE STATEMENT: This work demonstrates that heterologously expressed human ether a-go-go-related gene (hERG) 1a/1b channels, which more closely resemble rapidly activating delayed rectifier potassium current in the human heart, are blocked by fentanyl with a 3-fold greater potency than the previously studied hERG1a expressed alone. Additionally, chronic hypoxia, hypokalemia, and alkalosis can increase the block of hERG current by fentanyl, potentially increasing the risk of cardiac arrhythmias and sudden death.


Assuntos
Analgésicos Opioides/farmacologia , Canal de Potássio ERG1/genética , Canal de Potássio ERG1/metabolismo , Fentanila/farmacologia , Processamento Alternativo , Hipóxia Celular , Meios de Cultura/química , Canal de Potássio ERG1/antagonistas & inibidores , Células HEK293 , Humanos , Concentração de Íons de Hidrogênio , Concentração Inibidora 50 , Modelos Biológicos , Mutação , Potássio/metabolismo
10.
Mol Pharmacol ; 95(4): 386-397, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30665971

RESUMO

The human ether-a-go-go-related gene (hERG) encodes the pore-forming subunit of the rapidly activating delayed rectifier potassium channel (IKr). Drug-mediated or medical condition-mediated disruption of hERG function is the primary cause of acquired long-QT syndrome, which predisposes affected individuals to ventricular arrhythmias and sudden death. Fentanyl abuse poses a serious health concern, with abuse and death rates rising over recent years. As fentanyl has a propensity to cause sudden death, we investigated its effects on the hERG channel. The effects of norfentanyl, the main metabolite, and naloxone, an antidote used in fentanyl overdose, were also examined. Currents of hERG channels stably expressed in HEK293 cells were recorded using the whole-cell voltage-clamp method. When hERG tail currents were analyzed upon -50 mV repolarization after a 50 mV depolarization, fentanyl and naloxone blocked hERG current (IhERG) with IC50 values of 0.9 and 74.3 µM, respectively, whereas norfentanyl did not block. However, fentanyl-mediated block of IhERG was voltage dependent. When a voltage protocol that mimics a human ventricular action potential (AP) was used, fentanyl blocked IhERG with an IC50 of 0.3 µM. Furthermore, fentanyl (0.5 µM) prolonged AP duration and blocked IKr in ventricular myocytes isolated from neonatal rats. The concentrations of fentanyl used in this study were higher than seen with clinical use but overlap with postmortem overdose concentrations. Although mechanisms of fentanyl-related sudden death need further investigation, blockade of hERG channels may contribute to the death of individuals with high-concentration overdose or compromised cardiac repolarization.


Assuntos
Bloqueadores dos Canais de Potássio/farmacologia , Potenciais de Ação/efeitos dos fármacos , Animais , Arritmias Cardíacas/tratamento farmacológico , Arritmias Cardíacas/metabolismo , Feminino , Fentanila , Células HEK293 , Humanos , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Ratos , Ratos Sprague-Dawley , Regulador Transcricional ERG/antagonistas & inibidores
11.
Mol Pharmacol ; 96(1): 1-12, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31015282

RESUMO

The human ether-à-go-go-related gene (hERG) encodes the channel that conducts the rapidly activating delayed rectifier potassium current (IKr) in the heart. Reduction in IKr causes long QT syndrome, which can lead to fatal arrhythmias triggered by stress. One potential link between stress and hERG function is protein kinase C (PKC) activation; however, seemingly conflicting results regarding PKC regulation of hERG have been reported. We investigated the effects of PKC activation using phorbol 12-myristate 13-acetate (PMA) on hERG channels expressed in human embryonic kidney cell line 293 (HEK293) cells and IKr in isolated neonatal rat ventricular myocytes. Acute activation of PKC by PMA (30 nM, 30 minutes) reduced both hERG current (IhERG) and IKr Chronic activation of PKC by PMA (30 nM, 16 hours) increased IKr in cardiomyocytes and the expression level of hERG proteins; however, chronic (30 nM, 16 hours) PMA treatment decreased IhERG, which became larger than untreated control IhERG after PMA removal for 4 hours. Deletion of amino acid residues 2-354 (Δ2-354 hERG) or 1-136 of the N terminus (ΔN 136 hERG) abolished acute PMA (30 nM, 30 minutes)-mediated IhERG reduction. In contrast to wild-type hERG channels, chronic activation of PKC by PMA (30 nM, 16 hours) increased both Δ2-354 hERG and ΔN136 hERG expression levels and currents. The increase in hERG protein was associated with PKC-induced phosphorylation (inhibition) of Nedd4-2, an E3 ubiquitin ligase that mediates hERG degradation. We conclude that PKC regulates hERG in a balanced manner, increasing expression through inhibiting Nedd4-2 while decreasing current through targeting a site(s) within the N terminus.


Assuntos
Canal de Potássio ERG1/genética , Canal de Potássio ERG1/metabolismo , Miócitos Cardíacos/metabolismo , Proteína Quinase C/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Animais , Animais Recém-Nascidos , Células Cultivadas , Canal de Potássio ERG1/química , Ativação Enzimática/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Miócitos Cardíacos/efeitos dos fármacos , Ubiquitina-Proteína Ligases Nedd4/metabolismo , Fosforilação , Proteólise , Deleção de Sequência
12.
J Biol Chem ; 293(40): 15347-15358, 2018 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-30121572

RESUMO

The voltage-gated potassium channel Kv1.5 belongs to the Shaker superfamily. Kv1.5 is composed of four subunits, each comprising 613 amino acids, which make up the N terminus, six transmembrane segments (S1-S6), and the C terminus. We recently demonstrated that, in HEK cells, extracellularly applied proteinase K (PK) cleaves Kv1.5 channels at a single site in the S1-S2 linker. This cleavage separates Kv1.5 into an N-fragment (N terminus to S1) and a C-fragment (S2 to C terminus). Interestingly, the cleavage does not impair channel function. Here, we investigated the role of the N terminus and S1 in Kv1.5 expression and function by creating plasmids encoding various fragments, including those that mimic PK-cleaved products. Our results disclosed that although expression of the pore-containing fragment (Frag(304-613)) alone could not produce current, coexpression with Frag(1-303) generated a functional channel. Immunofluorescence and biotinylation analyses uncovered that Frag(1-303) was required for Frag(304-613) to traffic to the plasma membrane. Biochemical analysis revealed that the two fragments interacted throughout channel trafficking and maturation. In Frag(1-303)+(304-613)-coassembled channels, which lack a covalent linkage between S1 and S2, amino acid residues 1-209 were important for association with Frag(304-613), and residues 210-303 were necessary for mediating trafficking of coassembled channels to the plasma membrane. We conclude that the N terminus and S1 of Kv1.5 can attract and coassemble with the rest of the channel (i.e. Frag(304-613)) to form a functional channel independently of the S1-S2 linkage.


Assuntos
Canal de Potássio Kv1.5/química , Potenciais da Membrana/fisiologia , Fragmentos de Peptídeos/química , Subunidades Proteicas/química , Endopeptidase K/farmacologia , Expressão Gênica , Células HEK293 , Humanos , Transporte de Íons/efeitos dos fármacos , Canal de Potássio Kv1.5/genética , Canal de Potássio Kv1.5/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Plasmídeos/química , Plasmídeos/metabolismo , Domínios Proteicos , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Transporte Proteico , Relação Estrutura-Atividade , Transformação Genética
13.
FASEB J ; 32(4): 1933-1943, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32172531

RESUMO

The human ether-a-go-go related gene (hERG)-encoded channel hERG undergoes N-linked glycosylation at position 598, which is located in the unusually long S5-pore linker of the channel. In other work we have demonstrated that hERG is uniquely susceptible to proteolytic cleavage at the S5-pore linker by proteinase K (PK) and calpain (CAPN). The scorpion toxin BeKm-1, which binds to the S5-pore linker of hERG, protects hERG from such cleavage. In the present study, our data revealed that, compared with normal glycosylated hERG channels, nonglycosylated hERG channels were significantly more susceptible to cleavage by extracellular PK. Furthermore, the protective effect of BeKm-1 on hERG from PK-cleavage was lost when glycosylation of hERG was inhibited. The inactivation-deficient mutant hERG channels S620T and S631A were resistant to PK cleavage, and inhibition of glycosylation rendered both mutants susceptible to PK cleavage. Compared with normal glycosylated channels, nonglycosylated hERG channels were also more susceptible to cleavage mediated by CAPN, which was present in the medium of human embryonic kidney cells under normal culture conditions. Inhibition of CAPN resulted in an increase of nonglycosylated hERG current. In summary, our results revealed that N-linked glycosylation protects hERG against protease-mediated degradation and thus contributes to hERG channel stability on the plasma membrane.- Lamothe, S. M., Hulbert, M., Guo, J., Li, W., Yang, T., Zhang, S. Glycosylation stabilizes hERG channels on the plasma membrane by decreasing proteolytic susceptibility. FASEB J. 32, 1933-1943 (2018). www.fasebj.org.

14.
FASEB J ; 31(11): 5068-5077, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28784631

RESUMO

Human ether-a-go-go-related gene (hERG) encodes the pore-forming subunit of the rapidly activating delayed rectifier potassium current (IKr) potassium channel, which is important for cardiac repolarization. Impairment of hERG function is the primary cause of acquired long QT syndrome, which predisposes individuals to cardiac arrhythmias and sudden death. Patients with hypoxia due to conditions such as cardiac ischemia or obstructive sleep apnea display increased incidence of cardiac arrhythmias and sudden death. We sought to understand the mechanisms that underlie hypoxia-associated cardiac arrhythmias. Using cell biology and electrophysiologic techniques, we found that hypoxic culture of hERG-expressing human embryonic kidney (HEK) cells and neonatal rat cardiomyocytes reduced hERG current/IKr and mature ERG channel expression with a concomitant increase in calpain expression. Calpain was actively released into the extracellular milieu and degraded cell-surface hERG. In contrast to hERG, the ether-a-go-go (EAG) channel was not reduced by hypoxic culture. By making chimeric channels between hERG and EAG, we identified that hypoxia-induced calpain degraded hERG by targeting its extracellular S5-pore linker. The scorpion toxin BeKm-1, which is known to selectively bind to the S5-pore linker of hERG, prevented hypoxia-induced hERG reduction. Our data provide novel information about hypoxia-mediated hERG dysfunction and may have biological and clinical implications in hypoxia-associated diseases.-Lamothe, S. M., Song, W., Guo, J., Li, W., Yang, T., Baranchuk, A., Graham, C. H., Zhang, S. Hypoxia reduces mature hERG channels through calpain up-regulation.


Assuntos
Calpaína/biossíntese , Canal de Potássio ERG1/metabolismo , Regulação Enzimológica da Expressão Gênica , Síndrome do QT Longo/metabolismo , Miócitos Cardíacos/metabolismo , Regulação para Cima , Calpaína/genética , Hipóxia Celular/efeitos dos fármacos , Hipóxia Celular/genética , Canal de Potássio ERG1/genética , Células HEK293 , Humanos , Síndrome do QT Longo/genética , Venenos de Escorpião/toxicidade
15.
Mol Pharmacol ; 92(2): 162-174, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28495999

RESUMO

The rapidly activating delayed rectifier K+ channel (IKr) is encoded by the human ether-a-go-go-related gene (hERG), which is important for the repolarization of the cardiac action potential. Mutations in hERG or drugs can impair the function or decrease the expression level of hERG channels, leading to long QT syndrome. Thus, it is important to understand hERG channel trafficking and its regulation. For this purpose, G protein-coupled receptors (GPCRs), which regulate a vast array of cellular processes, represent a useful route. The development of designer GPCRs known as designer receptors exclusively activated by designer drugs (DREADDs) has made it possible to dissect specific GPCR signaling pathways in various cellular systems. In the present study, by expressing an arrestin-biased M3 muscarinic receptor-based DREADD (M3D-arr) in stable hERG-expressing human embryonic kidney (HEK) cells, we demonstrate that ß-arrestin signaling plays a role in hERG regulation. By exclusively activating M3D-arr using the otherwise inert compound, clozapine-N-oxide, we found that M3D-arr activation increased mature hERG expression and current. Within this paradigm, M3D-arr recruited ß-arrestin-1 to the plasma membrane, and promoted phosphoinositide 3-kinase-dependent activation of protein kinase B (Akt). The activated Akt acted through phosphatidylinositol 3-phosphate 5-kinase and Rab11 to facilitate hERG recycling to the plasma membrane. Potential ß-arrestin signaling-mediated increases in hERG and IKr were also observed in hERG-HEK cells as well as in neonatal rat ventricular myocytes treated with the muscarinic agonist carbachol. These findings provide novel insight into hERG trafficking and regulation.


Assuntos
Canal de Potássio ERG1/metabolismo , beta-Arrestinas/metabolismo , Animais , Clozapina/análogos & derivados , Clozapina/metabolismo , Clozapina/farmacologia , Relação Dose-Resposta a Droga , Canal de Potássio ERG1/agonistas , Feminino , Células HEK293 , Humanos , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Ratos , Ratos Sprague-Dawley
16.
J Biol Chem ; 291(39): 20387-401, 2016 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-27502273

RESUMO

The human ether-a-go-go-related gene (hERG) encodes the pore-forming subunit of the rapidly activating delayed rectifier potassium channel (IKr), which is important for cardiac repolarization. Dysfunction of hERG causes long QT syndrome and sudden death, which occur in patients with cardiac ischemia. Cardiac ischemia is also associated with activation, up-regulation, and secretion of various proteolytic enzymes. Here, using whole-cell patch clamp and Western blotting analysis, we demonstrate that the hERG/IKr channel was selectively cleaved by the serine protease, proteinase K (PK). Using molecular biology techniques including making a chimeric channel between protease-sensitive hERG and insensitive human ether-a-go-go (hEAG), as well as application of the scorpion toxin BeKm-1, we identified that the S5-pore linker of hERG is the target domain for proteinase K cleavage. To investigate the physiological relevance of the unique susceptibility of hERG to proteases, we show that cardiac ischemia in a rabbit model was associated with a reduction in mature ERG expression and an increase in the expression of several proteases, including calpain. Using cell biology approaches, we found that calpain-1 was actively released into the extracellular milieu and cleaved hERG at the S5-pore linker. Using protease cleavage-predicting software and site-directed mutagenesis, we identified that calpain-1 cleaves hERG at position Gly-603 in the S5-pore linker of hERG. Clarification of protease-mediated damage of hERG extends our understanding of hERG regulation. Damage of hERG mediated by proteases such as calpain may contribute to ischemia-associated QT prolongation and sudden cardiac death.


Assuntos
Canal de Potássio ERG1/metabolismo , Isquemia Miocárdica/metabolismo , Proteólise , Animais , Calpaína/biossíntese , Calpaína/química , Calpaína/genética , Canal de Potássio ERG1/química , Canal de Potássio ERG1/genética , Endopeptidase K/química , Células HEK293 , Humanos , Masculino , Isquemia Miocárdica/genética , Coelhos , Venenos de Escorpião/farmacologia
17.
Biochim Biophys Acta ; 1858(6): 1082-90, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26874203

RESUMO

Kv1.5 channels mediate the ultra-rapidly activating delayed rectifier potassium current (IKur), which is important for atrial repolarization. It has been shown that cell-surface Kv1.5 channels are sensitive to cleavage by the extracellular serine protease, proteinase K (PK). Here, we investigated the effects of extracellular proteolytic digestion on the function of Kv1.5 channels stably expressed in HEK 293 cells. Our data demonstrate that PK treatment cleaved mature membrane-bound (75kDa) Kv1.5 channels at a single locus in the S1-S2 linker, producing 42-kDa N-terminal fragments and 33-kDa C-terminal fragments. Interestingly, such PK treatment did not affect the Kv1.5 current (IKv1.5) recorded using the whole-cell patch clamp technique. Analysis of cell-surface proteins isolated using biotinylation indicated that the PK-generated N- and C-terminal fragments were both present in the plasma membrane. Co-immunoprecipitation (co-IP) experiments indicated that the N- and C-terminal fragments are no longer associated after cleavage. Furthermore, following PK digestion, the N- and C-fragments degraded at different rates. PK is frequently used as a tool to analyze cell-surface localization of membrane proteins, and cleavage of cell-surface channels has been shown to abolish channel function (e.g. hERG). Our data, for the first time, demonstrate that cleavage of cell-surface channels assessed by Western blot analysis does not necessarily correlate with an elimination of the channel activities.


Assuntos
Canal de Potássio Kv1.5/fisiologia , Células HEK293 , Humanos , Canal de Potássio Kv1.5/química , Canal de Potássio Kv1.5/genética , Microscopia de Fluorescência , Técnicas de Patch-Clamp , Proteólise
18.
J Biol Chem ; 290(34): 21101-21113, 2015 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-26152716

RESUMO

The human ether-a-go-go-related gene (hERG) encodes the pore-forming subunit of the rapidly activating delayed rectifier potassium channel (IKr). A reduction in the hERG current causes long QT syndrome, which predisposes affected individuals to ventricular arrhythmias and sudden death. We reported previously that hERG channels in the plasma membrane undergo vigorous internalization under low K(+) conditions. In the present study, we addressed whether hERG internalization occurs under normal K(+) conditions and whether/how internalized channels are recycled back to the plasma membrane. Using patch clamp, Western blot, and confocal imaging analyses, we demonstrated that internalized hERG channels can effectively recycle back to the plasma membrane. Low K(+)-enhanced hERG internalization is accompanied by an increased rate of hERG recovery in the plasma membrane upon reculture following proteinase K-mediated clearance of cell-surface proteins. The increased recovery rate is not due to enhanced protein synthesis, as hERG mRNA expression was not altered by low K(+) exposure, and the increased recovery was observed in the presence of the protein biosynthesis inhibitor cycloheximide. GTPase Rab11, but not Rab4, is involved in the recycling of hERG channels. Interfering with Rab11 function not only delayed hERG recovery in cells after exposure to low K(+) medium but also decreased hERG expression and function in cells under normal culture conditions. We concluded that the recycling pathway plays an important role in the homeostasis of plasma membrane-bound hERG channels.


Assuntos
Membrana Celular/metabolismo , Canais de Potássio Éter-A-Go-Go/metabolismo , Potássio/metabolismo , RNA Mensageiro/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab4 de Ligação ao GTP/metabolismo , Sequência de Aminoácidos , Membrana Celular/química , Membrana Celular/efeitos dos fármacos , Cicloeximida/farmacologia , Canal de Potássio ERG1 , Endopeptidase K/química , Canais de Potássio Éter-A-Go-Go/química , Canais de Potássio Éter-A-Go-Go/genética , Expressão Gênica , Células HEK293 , Humanos , Transporte de Íons , Dados de Sequência Molecular , Técnicas de Patch-Clamp , Transporte Proteico , RNA Mensageiro/genética , Proteínas rab de Ligação ao GTP/genética , Proteínas rab4 de Ligação ao GTP/genética
19.
Biochim Biophys Acta ; 1849(8): 1095-103, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25959059

RESUMO

Splicing factors are often influenced by various signaling pathways, contributing to the dynamic changes of protein isoforms in cells. Heterogeneous ribonucleoproteins (hnRNPs) regulate many steps of RNA metabolism including pre-mRNA splicing but their control by cell signaling particularly through acetylation and ubiquitination pathways remains largely unknown. Here we show that TSA, a deacetylase inhibitor, reduced the ratio of Bcl-x splice variants Bcl-xL/xS in MDA-MB-231 breast cancer cells. This TSA effect was independent of TGFß1; however, only in the presence of TGFß1 was TSA able to change the splicing regulators hnRNP F/H by slightly reducing their mRNA transcripts but strongly preventing protein degradation. The latter was also efficiently prevented by lactacystin, a proteasome inhibitor, suggesting their protein stability control by both acetylation and ubiquitination pathways. Three lysines K87, K98 and K224 of hnRNP F are potential targets of the mutually exclusive acetylation or ubiquitination (K(Ac/Ub)) in the protein modification database PhosphoSitePlus. Mutating each of them but not a control non-K(Ac/Ub) (K68) specifically abolished the TSA enhancement of protein stability. Moreover, mutating K98 (K98R) and K224 (K224R) also abolished the TSA regulation of alternative splicing of a Bcl-x mini-gene. Furthermore, about 86% (30 of 35) of the multi-functional hnRNP proteins in the database contain lysines that are potential sites for acetylation/ubiquitination. We demonstrate that the degradation of three of them (A1, I and L) are also prevented by TSA. Thus, the deacetylase inhibitor TSA enhances hnRNP F stability through the K(Ac/Ub) lysines, with some of them essential for its regulation of alternative splicing. Such a regulation of protein stability is perhaps common for a group of hnRNPs and RNA metabolism.


Assuntos
Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacologia , Sequência de Aminoácidos , Animais , Ribonucleoproteínas Nucleares Heterogêneas/genética , Humanos , Mutação , Células PC12 , Estabilidade Proteica/efeitos dos fármacos , Splicing de RNA/efeitos dos fármacos , Splicing de RNA/genética , Ratos , Fator de Crescimento Transformador beta1/farmacologia , Células Tumorais Cultivadas
20.
Biochem J ; 472(1): 71-82, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26363003

RESUMO

The cardiac electrical disorder long QT syndrome (LQTS) pre-disposes affected individuals to ventricular arrhythmias and sudden death. Dysfunction of the human ether-a-go-go-related gene (hERG)-encoded rapidly activating delayed rectifier K(+) channel (IKr) is a major cause of LQTS. The expression of hERG channels is controlled by anterograde trafficking of newly synthesized channels to and retrograde degradation of existing channels from the plasma membrane. We have previously shown that the E3 ubiquitin (Ub) ligase Nedd4-2 (neural precursor cell expressed developmentally down-regulated protein 4-2) targets the PY motif of hERG channels to initiate channel degradation. Although both immature and mature hERG channels contain the PY motif, Nedd4-2 selectively mediates the degradation of mature hERG channels. In the present study, we demonstrate that Nedd4-2 is directed to specific cellular compartments by the Nedd4 family interacting proteins, Nedd4 family-interacting protein 1 (Ndfip1) and Ndfip2. Ndfip1 is primarily localized in the Golgi apparatus where it recruits Nedd4-2 to mediate the degradation of mature hERG proteins during channel trafficking to the plasma membrane. Although Ndfip2 directs Nedd4-2 to the Golgi apparatus, it also recruits Nedd4-2 to the multivesicular bodies (MVBs), which may impair MVB function and impede the degradation of mature hERG proteins mediated by Nedd4-2. These findings extend our understanding of hERG channel regulation and provide information which may be useful for the rescue of impaired hERG function in LQTS.


Assuntos
Proteínas de Transporte/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Canais de Potássio Éter-A-Go-Go/metabolismo , Proteínas de Membrana/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Animais Recém-Nascidos , Proteínas de Transporte/genética , Células Cultivadas , Canal de Potássio ERG1 , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Canais de Potássio Éter-A-Go-Go/genética , Feminino , Complexo de Golgi/metabolismo , Células HEK293 , Humanos , Immunoblotting , Masculino , Potenciais da Membrana , Proteínas de Membrana/genética , Microscopia de Fluorescência , Corpos Multivesiculares/metabolismo , Mutação , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Ubiquitina-Proteína Ligases Nedd4 , Técnicas de Patch-Clamp , Ligação Proteica , Interferência de RNA , Ratos Sprague-Dawley , Ubiquitina-Proteína Ligases/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA