Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 131
Filter
1.
Circ Res ; 134(7): 858-871, 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38362769

ABSTRACT

BACKGROUND: Vascular large conductance Ca2+-activated K+ (BK) channel, composed of the α-subunit (BK-α) and the ß1-subunit (BK-ß1), is a key determinant of coronary vasorelaxation and its function is impaired in diabetic vessels. However, our knowledge of diabetic BK channel dysregulation is incomplete. The Sorbs2 (Sorbin homology [SoHo] and Src homology 3 [SH3] domains-containing protein 2), is ubiquitously expressed in arteries, but its role in vascular pathophysiology is unknown. METHODS: The role of Sorbs2 in regulating vascular BK channel activity was determined using patch-clamp recordings, molecular biological techniques, and in silico analysis. RESULTS: Sorbs2 is not only a cytoskeletal protein but also an RNA-binding protein that binds to BK channel proteins and BK-α mRNA, regulating BK channel expression and function in coronary smooth muscle cells. Molecular biological studies reveal that the SH3 domain of Sorbs2 is necessary for Sorbs2 interaction with BK-α subunits, while both the SH3 and SoHo domains of Sorbs2 interact with BK-ß1 subunits. Deletion of the SH3 or SoHo domains abolishes the Sorbs2 effect on the BK-α/BK-ß1 channel current density. Additionally, Sorbs2 is a target gene of the Nrf2 (nuclear factor erythroid-2-related factor 2), which binds to the promoter of Sorbs2 and regulates Sorbs2 expression in coronary smooth muscle cells. In vivo studies demonstrate that Sorbs2 knockout mice at 4 months of age display a significant decrease in BK channel expression and function, accompanied by impaired BK channel Ca2+-sensitivity and BK channel-mediated vasodilation in coronary arteries, without altering their body weights and blood glucose levels. Importantly, Sorbs2 expression is significantly downregulated in the coronary arteries of db/db type 2 diabetic mice. CONCLUSIONS: Sorbs2, a downstream target of Nrf2, plays an important role in regulating BK channel expression and function in vascular smooth muscle cells. Vascular Sorbs2 is downregulated in diabetes. Genetic knockout of Sorbs2 manifests coronary BK channelopathy and vasculopathy observed in diabetic mice, independent of obesity and glucotoxicity.


Subject(s)
Channelopathies , Diabetes Mellitus, Experimental , Mice , Animals , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , NF-E2-Related Factor 2/metabolism , Channelopathies/metabolism , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Muscle, Smooth, Vascular/metabolism , Large-Conductance Calcium-Activated Potassium Channels/genetics , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Coronary Vessels/metabolism , RNA-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism
2.
Am J Physiol Regul Integr Comp Physiol ; 324(4): R480-R496, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36717168

ABSTRACT

Alcohol intake leading to blood ethanol concentrations (BEC) ≥ legal intoxication modifies brain blood flow with increases in some regions and decreases in others. Brain regions receive blood from the Willis' circle branches: anterior, middle (MCA) and posterior cerebral (PCA), and basilar (BA) arteries. Rats and mice have been used to identify the targets mediating ethanol-induced effects on cerebral arteries, with conclusions being freely interchanged, albeit data were obtained in different species/arterial branches. We tested whether ethanol action on cerebral arteries differed between male rat and mouse and/or across different brain regions and identified the targets of alcohol action. In both species and all Willis' circle branches, ethanol evoked reversible and concentration-dependent constriction (EC50s ≈ 37-86 mM; below lethal BEC in alcohol-naïve humans). Although showing similar constriction to depolarization, both species displayed differential responses to ethanol: in mice, MCA constriction was highly sensitive to the presence/absence of the endothelium, whereas in rat PCA was significantly more sensitive to ethanol than its mouse counterpart. In the rat, but not the mouse, BA was more ethanol sensitive than other branches. Both interspecies and regional variability were ameliorated by endothelium. Selective large conductance (BK) channel block in de-endothelialized vessels demonstrated that these channels were the effectors of alcohol-induced cerebral artery constriction across regions and species. Variabilities in alcohol actions did not fully matched KCNMB1 expression across vessels. However, immunofluorescence data from KCNMB1-/- mouse arteries electroporated with KCNMB1-coding cDNA demonstrate that KCNMB1 proteins, which regulate smooth muscle (SM) BK channel function and vasodilation, regulate interspecies and regional variability of brain artery responses to alcohol.


Subject(s)
Cerebral Arteries , Ethanol , Animals , Male , Mice , Rats , Ethanol/pharmacology , Ethanol/metabolism , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Muscle, Smooth, Vascular/metabolism , Rats, Sprague-Dawley
3.
Cell Mol Life Sci ; 79(2): 75, 2022 Jan 17.
Article in English | MEDLINE | ID: mdl-35039944

ABSTRACT

Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is a new member of the Betacoronaviridae family, responsible for the recent pandemic outbreak of COVID-19. To start exploring the molecular events that follow host cell infection, we queried VirusCircBase and identified a circular RNA (circRNA) predicted to be synthesized by SARS-CoV-2, circ_3205, which we used to probe: (i) a training cohort comprised of two pools of cells from three nasopharyngeal swabs of SARS-CoV-2 infected (positive) or uninfected (negative, UCs) individuals; (ii) a validation cohort made up of 12 positive and 3 negative samples. The expression of circRNAs, miRNAs and miRNA targets was assayed through real-time PCR. CircRNA-miRNA interactions were predicted by TarpMiR, Analysis of Common Targets for circular RNAs (ACT), and STarMir tools. Enrichment of the biological processes and the list of predicted miRNA targets were retrieved from DIANA miRPath v3.0. Our results showed that the predicted SARS-CoV-2 circ_3205 was expressed only in positive samples and its amount positively correlated with that of SARS-CoV-2 Spike (S) mRNA and the viral load (r values = 0.80952 and 0.84867, Spearman's correlation test, respectively). Human (hsa) miR-298 was predicted to interact with circ_3205 by all three predictive tools. KCNMB4 and PRKCE were predicted as hsa-miR-298 targets. Interestingly, the function of both is correlated with blood coagulation and immune response. KCNMB4 and PRKCE mRNAs were upregulated in positive samples as compared to UCs (6 and 8.1-fold, p values = 0.049 and 0.02, Student's t test, respectively) and their expression positively correlated with that of circ_3205 (r values = 0.6 and 0.25, Spearman's correlation test, respectively). We propose that our results convincingly suggest that circ_3205 is a circRNA synthesized by SARS-CoV-2 upon host cell infection and that it may behave as a competitive endogenous RNA (ceRNA), sponging hsa-miR-298 and contributing to the upregulation of KCNMB4 and PRKCE mRNAs.


Subject(s)
COVID-19/genetics , COVID-19/metabolism , RNA, Circular/genetics , RNA, Viral , SARS-CoV-2/genetics , Computational Biology , Gene Expression Regulation, Viral , Gene Regulatory Networks , Humans , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Nasopharynx/virology , Nerve Tissue Proteins/genetics , Protein Interaction Mapping , Protein Kinase C-epsilon/genetics , Reproducibility of Results
4.
Proc Natl Acad Sci U S A ; 117(47): 29937-29947, 2020 11 24.
Article in English | MEDLINE | ID: mdl-33168717

ABSTRACT

Analysis of the presynaptic action potential's (APsyn) role in synaptic facilitation in hippocampal pyramidal neurons has been difficult due to size limitations of axons. We overcame these size barriers by combining high-resolution optical recordings of membrane potential, exocytosis, and Ca2+ in cultured hippocampal neurons. These recordings revealed a critical and selective role for Kv1 channel inactivation in synaptic facilitation of excitatory hippocampal neurons. Presynaptic Kv1 channel inactivation was mediated by the Kvß1 subunit and had a surprisingly rapid onset that was readily apparent even in brief physiological stimulation paradigms including paired-pulse stimulation. Genetic depletion of Kvß1 blocked all broadening of the APsyn during high-frequency stimulation and eliminated synaptic facilitation without altering the initial probability of vesicle release. Thus, using all quantitative optical measurements of presynaptic physiology, we reveal a critical role for presynaptic Kv channels in synaptic facilitation at presynaptic terminals of the hippocampus upstream of the exocytic machinery.


Subject(s)
Hippocampus/metabolism , Kv1.3 Potassium Channel/metabolism , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Pyramidal Cells/metabolism , Synaptic Potentials/physiology , Animals , Calcium/metabolism , Cells, Cultured , Elapid Venoms/pharmacology , Exocytosis/drug effects , Exocytosis/physiology , Female , Gene Knockdown Techniques , Hippocampus/cytology , Intravital Microscopy , Kv1.3 Potassium Channel/genetics , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/antagonists & inhibitors , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Male , Mice , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Optical Imaging , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Primary Cell Culture , Pyramidal Cells/drug effects , Rats , Synaptic Potentials/drug effects
5.
Int J Mol Sci ; 24(10)2023 May 12.
Article in English | MEDLINE | ID: mdl-37240049

ABSTRACT

Calcium/voltage-activated potassium channels (BK) control smooth muscle (SM) tone and cerebral artery diameter. They include channel-forming α and regulatory ß1 subunits, the latter being highly expressed in SM. Both subunits participate in steroid-induced modification of BK activity: ß1 provides recognition for estradiol and cholanes, resulting in BK potentiation, whereas α suffices for BK inhibition by cholesterol or pregnenolone. Aldosterone can modify cerebral artery function independently of its effects outside the brain, yet BK involvement in aldosterone's cerebrovascular action and identification of channel subunits, possibly involved in steroid action, remains uninvestigated. Using microscale thermophoresis, we demonstrated that each subunit type presents two recognition sites for aldosterone: at 0.3 and ≥10 µM for α and at 0.3-1 µM and ≥100 µM for ß1. Next, we probed aldosterone on SM BK activity and diameter of middle cerebral artery (MCA) isolated from ß1-/- vs. wt mice. Data showed that ß1 leftward-shifted aldosterone-induced BK activation, rendering EC50~3 µM and ECMAX ≥ 10 µM, at which BK activity increased by 20%. At similar concentrations, aldosterone mildly yet significantly dilated MCA independently of circulating and endothelial factors. Lastly, aldosterone-induced MCA dilation was lost in ß1-/- mice. Therefore, ß1 enables BK activation and MCA dilation by low µM aldosterone.


Subject(s)
Aldosterone , Large-Conductance Calcium-Activated Potassium Channels , Mice , Animals , Aldosterone/pharmacology , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Muscle, Smooth, Vascular , Dilatation , Steroids/pharmacology , Cerebral Arteries
6.
Arterioscler Thromb Vasc Biol ; 40(11): e284-e295, 2020 11.
Article in English | MEDLINE | ID: mdl-32967457

ABSTRACT

OBJECTIVE: Antenatal exposure to glucocorticoids increases cardiovascular risks related to vascular dysfunctions in offspring, although underlying mechanisms are still unknown. As an important vascular mediator, high-conductance Ca2+-activated K+ channels (BK) plays an essential role in determining vascular tone. Long-term effects of antenatal glucocorticoids on BK in offspring are largely unknown. This study examined the effects and mechanisms of antenatal exposure to clinically relevant doses of glucocorticoids on vascular BK in offspring. Approach and Results: Pregnant Sprague-Dawley rats received synthetic glucocorticoids dexamethasone or vehicle during the last week of pregnancy. Vascular functions, cellular electrophysiology, target gene expression, and promoter methylation were examined in mesenteric arteries of male offspring (gestational day 21 [fetus] and postnatal day 120 [adult offspring]). Antenatal dexamethasone exposure impaired BK activators-mediated relaxation and reduced whole-cell BK currents in mesenteric arteries. Antenatal dexamethasone exposure did not alter Ca2+/voltage-sensitivity of BK but downregulated the expressions of BK α and ß1 subunits in both fetal and adult mesenteric arteries. In addition, increased promoter methylations within BKα and BKß1 were compatible with reduced expressions of the 2 genes. CONCLUSIONS: Our findings showed a profound and long-term impact of antenatal dexamethasone exposure on vascular BK via an altered epigenetic pattern from fetal stage to adulthood, advancing understanding of prolonged adverse effects and mechanisms of antenatal glucocorticoids exposure on vascular health in offspring.


Subject(s)
DNA Methylation , Dexamethasone/toxicity , Epigenesis, Genetic , Glucocorticoids/toxicity , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Mesenteric Arteries/metabolism , Prenatal Exposure Delayed Effects , Promoter Regions, Genetic , Action Potentials , Animals , Dexamethasone/administration & dosage , Female , Glucocorticoids/administration & dosage , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Male , Maternal Exposure , Mesenteric Arteries/physiopathology , Pregnancy , Rats, Sprague-Dawley , Vasodilation
7.
J Cell Mol Med ; 24(8): 4716-4725, 2020 04.
Article in English | MEDLINE | ID: mdl-32163656

ABSTRACT

Vascular dysfunction resulting from diabetes is an important factor in arteriosclerosis. Previous studies have shown that during hyperglycaemia and diabetes, AKAP150 promotes vascular tone enhancement by intensifying the remodelling of the BK channel. However, the interaction between AKAP150 and the BK channel remains open to discussion. In this study, we investigated the regulation of impaired BK channel-mediated vascular dysfunction in diabetes mellitus. Using AKAP150 null mice (AKAP150-/- ) and wild-type (WT) control mice (C57BL/6J), diabetes was induced by intraperitoneal injection of streptozotocin. We found that knockout of AKAP150 reversed vascular remodelling and fibrosis in mice with diabetes and in AKAP150-/- diabetic mice. Impaired Akt/GSK3ß signalling contributed to decreased BK-ß1 expression in aortas from diabetic mice, and the silencing of AKAP150 increased Akt phosphorylation and BK-ß1 expression in MOVAS cells treated with HG medium. The inhibition of Akt activity caused a decrease in BK-ß1 expression, and treatment with AKAP150 siRNA suppressed GSK3ß expression in the nuclei of MOVAS cells treated with HG. Knockout of AKAP150 reverses impaired BK channel-mediated vascular dysfunction through the Akt/GSK3ß signalling pathway in diabetes mellitus.


Subject(s)
A Kinase Anchor Proteins/genetics , Diabetes Complications/genetics , Diabetes Mellitus, Experimental/genetics , Glycogen Synthase Kinase 3 beta/genetics , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Animals , Arteriosclerosis/complications , Arteriosclerosis/genetics , Arteriosclerosis/pathology , Arteriosclerosis/therapy , Diabetes Complications/pathology , Diabetes Complications/therapy , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/therapy , Disease Models, Animal , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Humans , Hyperglycemia/complications , Hyperglycemia/genetics , Hyperglycemia/pathology , Hyperglycemia/therapy , Large-Conductance Calcium-Activated Potassium Channels/genetics , Mice , Mice, Knockout , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology
8.
J Biol Chem ; 294(32): 12066-12076, 2019 08 09.
Article in English | MEDLINE | ID: mdl-31213527

ABSTRACT

The properties and physiological function of pore-forming α-subunits of large conductance calcium- and voltage-activated potassium (BK) channels are potently modified by their functional coupling with regulatory subunits in many tissues. However, mechanisms that might control functional coupling are very poorly understood. Here we show that S-acylation, a dynamic post-translational lipid modification of proteins, of the intracellular S0-S1 loop of the BK channel pore-forming α-subunit controls functional coupling to regulatory ß1-subunits. In HEK293 cells, α-subunits that cannot be S-acylated show attenuated cell surface expression, but expression was restored by co-expression with the ß1-subunit. However, we also found that nonacylation of the S0-S1 loop reduces functional coupling between α- and ß1-subunits by attenuating the ß1-subunit-induced left shift in the voltage for half-maximal activation. In mouse vascular smooth muscle cells expressing both α- and ß1-subunits, BK channel α-subunits were endogenously S-acylated. We further noted that S-acylation is significantly reduced in mice with a genetic deletion of the palmitoyl acyltransferase (Zdhhc23) that controls S-acylation of the S0-S1 loop. Genetic deletion of Zdhhc23 or broad-spectrum pharmacological inhibition of S-acylation attenuated endogenous BK channel currents independently of changes in cell surface expression of the α-subunit. We conclude that functional effects of S-acylation on BK channels depend on the presence of ß1-subunits. In the absence of ß1-subunits, S-acylation promotes cell surface expression, whereas in its presence, S-acylation controls functional coupling. S-Acylation thus provides a mechanism that dynamically regulates the functional coupling with ß1-subunits, enabling an additional level of conditional, cell-specific control of ion-channel physiology.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channels/metabolism , Acylation , Animals , Cells, Cultured , HEK293 Cells , Humans , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Large-Conductance Calcium-Activated Potassium Channels/genetics , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Patch-Clamp Techniques , Sulfur/metabolism
9.
Pharmacogenet Genomics ; 30(6): 124-130, 2020 08.
Article in English | MEDLINE | ID: mdl-32371615

ABSTRACT

OBJECTIVE: The present prospective follow-up study aimed to evaluate the effects of KCNMB2 gene polymorphisms on ritodrine efficacy and adverse drug events (ADEs) in patients with preterm labor. METHODS: A total of 163 preterm labor patients were included in this single-center study. Nine single nucleotide polymorphisms (SNPs) in the KCNMB2 gene (rs10936979, rs7624046, rs7429015, rs7625907, rs6443559, rs9839376, rs9637454, rs11918114, and rs1382045) were assessed. The primary endpoint was time to delivery, and the secondary endpoint was ritodrine-induced ADEs. RESULTS: Patients with variant homozygotes of two SNPs (rs7624046 and rs9839376), which were in linkage disequilibrium, showed 2.06 [95% confidence interval (CI), 1.14-3.73] and 2.68 (95% CI, 1.16-6.20) times the hazard of time to delivery compared to wild-type allele carriers, respectively. Among demographic characteristics, gestational age at start of drug therapy and modified Bishop score were significant factors for time to delivery. Regarding safety outcomes, patients with variant homozygotes of rs7625907 had fewer ADEs compared to those with other genotypes (odds ratio, 0.32; 95% CI, 0.13-0.83). CONCLUSION: This pharmacogenomic study suggests that ritodrine efficacy and ADEs are associated with KCNMB2 gene polymorphisms in patients with preterm labor.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Obstetric Labor, Premature/drug therapy , Polymorphism, Single Nucleotide , Ritodrine/administration & dosage , Tocolytic Agents/administration & dosage , Adult , Female , Gestational Age , Humans , Linkage Disequilibrium , Logistic Models , Maternal Age , Obstetric Labor, Premature/genetics , Pregnancy , Pregnancy Trimester, Second/genetics , Pregnancy Trimester, Third/genetics , Prospective Studies , Ritodrine/adverse effects , Tocolytic Agents/adverse effects
10.
J Physiol ; 597(15): 3817-3832, 2019 08.
Article in English | MEDLINE | ID: mdl-31173379

ABSTRACT

KEY POINTS: Association of plasma membrane BKCa channels with BK-ß subunits shapes their biophysical properties and physiological roles; however, functional modulation of the mitochondrial BKCa channel (mitoBKCa ) by BK-ß subunits is not established. MitoBKCa -α and the regulatory BK-ß1 subunit associate in mouse cardiac mitochondria. A large fraction of mitoBKCa display properties similar to that of plasma membrane BKCa when associated with BK-ß1 (left-shifted voltage dependence of activation, V1/2  = -55 mV, 12 µm matrix Ca2+ ). In BK-ß1 knockout mice, cardiac mitoBKCa displayed a low Po and a depolarized V1/2 of activation (+47 mV at 12 µm matrix Ca2+ ) Co-expression of BKCa with the BK-ß1 subunit in HeLa cells doubled the density of BKCa in mitochondria. The present study supports the view that the cardiac mitoBKCa channel is functionally modulated by the BK-ß1 subunit; proper targeting and activation of mitoBKCa shapes mitochondrial Ca2+ handling. ABSTRACT: Association of the plasma membrane BKCa channel with auxiliary BK-ß1-4 subunits profoundly affects the regulatory mechanisms and physiological processes in which this channel participates. However, functional association of mitochondrial BK (mitoBKCa ) with regulatory subunits is unknown. We report that mitoBKCa functionally associates with its regulatory subunit BK-ß1 in adult rodent cardiomyocytes. Cardiac mitoBKCa is a calcium- and voltage-activated channel that is sensitive to paxilline with a large conductance for K+ of 300 pS. Additionally, mitoBKCa displays a high open probability (Po ) and voltage half-activation (V1/2  = -55 mV, n = 7) resembling that of plasma membrane BKCa when associated with its regulatory BK-ß1 subunit. Immunochemistry assays demonstrated an interaction between mitochondrial BKCa -α and its BK-ß1 subunit. Mitochondria from the BK-ß1 knockout (KO) mice showed sparse mitoBKCa currents (five patches with mitoBKCa activity out of 28 total patches from n = 5 different hearts), displaying a depolarized V1/2 of activation (+47 mV in 12 µm matrix Ca2+ ). The reduced activity of mitoBKCa was accompanied by a high expression of BKCa transcript in the BK-ß1 KO, suggesting a lower abundance of mitoBKCa channels in this genotype. Accordingly, BK-ß1subunit increased the localization of BKDEC (i.e. the splice variant of BKCa that specifically targets mitochondria) into mitochondria by two-fold. Importantly, both paxilline-treated and BK-ß1 KO mitochondria displayed a more rapid Ca2+ overload, featuring an early opening of the mitochondrial transition pore. We provide strong evidence that mitoBKCa associates with its regulatory BK-ß1 subunit in cardiac mitochondria, ensuring proper targeting and activation of the mitoBKCa channel that helps to maintain mitochondrial Ca2+ homeostasis.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , Action Potentials , Animals , Cells, Cultured , Ion Channel Gating , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Male , Myocytes, Cardiac/physiology , Protein Binding , Rats , Rats, Sprague-Dawley
11.
Am J Physiol Renal Physiol ; 316(2): F341-F350, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30484346

ABSTRACT

Special high-K diets have cardioprotective effects and are often warranted in conjunction with diuretics such as furosemide for treating hypertension. However, it is not understood how a high-K diet (HK) influences the actions of diuretics on renal K+ handling. Furosemide acidifies the urine by increasing acid secretion via the Na+-H+ exchanger 3 (NHE3) in TAL and vacuolar H+-ATPase (V-ATPase) in the distal nephron. We previously found that an alkaline urine is required for large conductance Ca2+-activated K+ (BK)-αß4-mediated K+ secretion in mice on HK. We therefore hypothesized that furosemide could reduce BK-αß4-mediated K+ secretion by acidifying the urine. Treating with furosemide (drinking water) for 11 days led to decreased urine pH in both wild-type (WT) and BK-ß4-knockout mice (BK-ß4-KO) with increased V-ATPase expression and elevated plasma aldosterone levels. However, furosemide decreased renal K+ clearance and elevated plasma [K+] in WT but not BK-ß4-KO. Western blotting and immunofluorescence staining showed that furosemide treatment decreased cortical expression of BK-ß4 and reduced apical localization of BK-α in connecting tubules. Addition of the carbonic anhydrase inhibitor, acetazolamide, to furosemide water restored urine pH along with renal K+ clearance and plasma [K+] to control levels. Acetazolamide plus furosemide also restored the cortical expression of BK-ß4 and BK-α in connecting tubules. These results indicate that in mice adapted to HK, furosemide reduces BK-αß4-mediated K+ secretion by acidifying the urine.


Subject(s)
Diet , Furosemide/pharmacology , Kidney/drug effects , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Potassium/metabolism , Renal Elimination/drug effects , Sodium Potassium Chloride Symporter Inhibitors/pharmacology , Acid-Base Equilibrium , Animals , Female , Hydrogen-Ion Concentration , Kidney/metabolism , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/deficiency , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Potassium/blood , Potassium/urine , Vacuolar Proton-Translocating ATPases/metabolism
12.
Int J Cancer ; 145(10): 2850-2860, 2019 11 15.
Article in English | MEDLINE | ID: mdl-30977117

ABSTRACT

Lung metastasis is one of the leading causes of death for triple-negative breast cancer (TNBC). We sought to characterize the genetic alterations underlying TNBC lung metastases by integrating whole-genome sequencing and functional screening. Furthermore, we aimed to develop a metastasis-related gene signature for TNBC patients to improve risk stratification. In this prospective observational study, we first conducted whole-genome sequencing of paired primary tumor and lung metastasis from one TNBC patient to identify potential genetic driver alterations. An in vivo gain-of-function screening using an amplified open reading frame library was then employed to screen candidate genes promoting lung metastasis. Finally, we applied Cox proportional hazard regression modeling to develop a prognostic gene signature from 14 candidate genes in TNBC. Compared to the primary tumor, copy number amplifications of chromosomes 3q and 8q were identified in the lung metastasis. We discovered an enrichment of 14 genes from chromosomes 3q and 8q in mouse lung metastases model. We further developed and validated a four-gene signature (ENY2, KCNK9, TNFRSF11B and KCNMB2) that predicts recurrence-free survival and lung metastasis in TNBC. Our data also demonstrated that upregulated expression of ENY2 could promote invasion and lung metastasis of TNBC cells both in vitro and in vivo. In conclusion, our study reveals functional genes with copy number amplifications among chromosome 3q and 8q in lung metastasis of TNBC. And we develop a functional gene signature that can effectively stratify patients into low- and high-risk subgroups of recurrence, helping frame personalized treatments for TNBC.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/diagnosis , Neoplasm Recurrence, Local/diagnosis , Triple Negative Breast Neoplasms/genetics , Animals , Cell Line, Tumor , Chromosomes, Human, Pair 3/genetics , Chromosomes, Human, Pair 8/genetics , DNA Copy Number Variations , Disease-Free Survival , Female , Follow-Up Studies , Gain of Function Mutation , Gene Expression Profiling , Gene Library , HEK293 Cells , Humans , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Middle Aged , Neoplasm Recurrence, Local/genetics , Osteoprotegerin/genetics , Potassium Channels, Tandem Pore Domain/genetics , Prognosis , Prospective Studies , Transcription Factors/genetics , Triple Negative Breast Neoplasms/pathology , Up-Regulation , Whole Genome Sequencing , Xenograft Model Antitumor Assays
13.
Proc Natl Acad Sci U S A ; 113(23): E3231-9, 2016 Jun 07.
Article in English | MEDLINE | ID: mdl-27217576

ABSTRACT

Large-conductance Ca(2+)- and voltage-activated K(+) (BK) channels are involved in a large variety of physiological processes. Regulatory ß-subunits are one of the mechanisms responsible for creating BK channel diversity fundamental to the adequate function of many tissues. However, little is known about the structure of its voltage sensor domain. Here, we present the external architectural details of BK channels using lanthanide-based resonance energy transfer (LRET). We used a genetically encoded lanthanide-binding tag (LBT) to bind terbium as a LRET donor and a fluorophore-labeled iberiotoxin as the LRET acceptor for measurements of distances within the BK channel structure in a living cell. By introducing LBTs in the extracellular region of the α- or ß1-subunit, we determined (i) a basic extracellular map of the BK channel, (ii) ß1-subunit-induced rearrangements of the voltage sensor in α-subunits, and (iii) the relative position of the ß1-subunit within the α/ß1-subunit complex.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channel beta Subunits/chemistry , Animals , Energy Transfer , Female , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/chemistry , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/physiology , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/physiology , Models, Molecular , Oocytes , Protein Conformation , Protein Domains , Xenopus laevis
14.
J Biol Chem ; 292(21): 8694-8704, 2017 05 26.
Article in English | MEDLINE | ID: mdl-28373283

ABSTRACT

The properties and function of large-conductance calcium- and voltage-activated potassium (BK) channels are modified by the tissue-specific expression of regulatory ß1-subunits. Although the short cytosolic N-terminal domain of the ß1-subunit is important for controlling both BK channel trafficking and function, whether the same, or different, regions of the N terminus control these distinct processes remains unknown. Here we demonstrate that the first six N-terminal residues including Lys-3, Lys-4, and Leu-5 are critical for controlling functional regulation, but not trafficking, of BK channels. This membrane-distal region has features of an amphipathic helix that is predicted to control the orientation of the first transmembrane-spanning domain (TM1) of the ß1-subunit. In contrast, a membrane-proximal leucine residue (Leu-17) controls trafficking without affecting functional coupling, an effect that is in part dependent on controlling efficient endoplasmic reticulum exit of the pore-forming α-subunit. Thus cell surface trafficking and functional coupling with BK channels are controlled by distinct domains of the ß1-subunit N terminus.


Subject(s)
Endoplasmic Reticulum/metabolism , Gene Expression Regulation/physiology , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/biosynthesis , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/biosynthesis , Endoplasmic Reticulum/genetics , HEK293 Cells , Humans , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Protein Domains , Protein Transport/physiology
15.
Am J Physiol Lung Cell Mol Physiol ; 315(2): L265-L275, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29644895

ABSTRACT

Accessory subunits associated with the calcium-sensitive potassium channel (BKCa), a major determinant of vascular tone, confer functional and anatomical diversity. The ß1 subunit increases Ca2+ and voltagesensitivity of the BKCa channel and is expressed exclusively in smooth muscle cells. Evidence supporting the physiological significance of the ß1 subunit includes the observations that murine models with deletion of the ß1 subunit are hypertensive and that humans with a gain-of-function ß1 mutation are at a decreased risk of diastolic hypertension. However, whether the ß1 subunit of the BKCa channel contributes to the low tone that characterizes the normal pulmonary circulation or modulates the pulmonary vascular response to hypoxia remains unknown. To determine the role of the BKCa channel ß1 subunit in the regulation of pulmonary vascular tone and the response to acute and chronic hypoxia, mice with deletion of the Kcnmb1 gene that encodes for the ß1 subunit ( Kcnmb1-/-) were placed in chronic hypoxia (10% O2) for 21-24 days. In normoxia, right ventricular systolic pressure (RVSP) did not differ between Kcnmb1+/+ (controls) and Kcnmb1-/- mice. After exposure to either acute or chronic hypoxia, RVSP was higher in Kcnmb1-/- mice compared with Kcnmb1+/+ mice, without increased vascular remodeling. ß1 subunit expression was predominantly confined to pulmonary artery smooth muscle cells (PASMCs) from vessels ≤ 150 µm. Peripheral PASMCs contracted collagen gels irrespective of ß1 expression. Focal adhesion expression and Rho kinase activity were greater in Kcnmb1-/- compared with Kcnmb1+/+ PASMCs. Compromised PASMC ß1 function may contribute to the heightened microvascular vasoconstriction that characterizes pulmonary hypertension.


Subject(s)
Hypoxia/metabolism , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Lung/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/metabolism , Acute Disease , Animals , Chronic Disease , Focal Adhesions/genetics , Focal Adhesions/metabolism , Focal Adhesions/pathology , Gene Deletion , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Hypoxia/genetics , Hypoxia/pathology , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Lung/blood supply , Lung/pathology , Mice , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Pulmonary Artery/pathology , Vasoconstriction
17.
Proc Natl Acad Sci U S A ; 112(15): 4809-14, 2015 Apr 14.
Article in English | MEDLINE | ID: mdl-25825713

ABSTRACT

Being activated by depolarizing voltages and increases in cytoplasmic Ca(2+), voltage- and calcium-activated potassium (BK) channels and their modulatory ß-subunits are able to dampen or stop excitatory stimuli in a wide range of cellular types, including both neuronal and nonneuronal tissues. Minimal alterations in BK channel function may contribute to the pathophysiology of several diseases, including hypertension, asthma, cancer, epilepsy, and diabetes. Several gating processes, allosterically coupled to each other, control BK channel activity and are potential targets for regulation by auxiliary ß-subunits that are expressed together with the α (BK)-subunit in almost every tissue type where they are found. By measuring gating currents in BK channels coexpressed with chimeras between ß1 and ß3 or ß2 auxiliary subunits, we were able to identify that the cytoplasmic regions of ß1 are responsible for the modulation of the voltage sensors. In addition, we narrowed down the structural determinants to the N terminus of ß1, which contains two lysine residues (i.e., K3 and K4), which upon substitution virtually abolished the effects of ß1 on charge movement. The mechanism by which K3 and K4 stabilize the voltage sensor is not electrostatic but specific, and the α (BK)-residues involved remain to be identified. This is the first report, to our knowledge, where the regulatory effects of the ß1-subunit have been clearly assigned to a particular segment, with two pivotal amino acids being responsible for this modulation.


Subject(s)
Calcium/metabolism , Ion Channel Gating/physiology , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/physiology , Potassium/metabolism , Animals , Binding Sites/genetics , Female , Humans , Ion Channel Gating/genetics , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/chemistry , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Lysine/chemistry , Lysine/genetics , Lysine/physiology , Membrane Potentials/genetics , Membrane Potentials/physiology , Models, Molecular , Mutation , Oocytes/metabolism , Oocytes/physiology , Protein Structure, Tertiary , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/physiology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Xenopus laevis
18.
J Cell Physiol ; 232(11): 3088-3102, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28019664

ABSTRACT

Modulation from contractile to synthetic phenotype of vascular smooth muscle cells is a central process in disorders involving compromised integrity of the vascular wall. Phenotype modulation has been shown to include transition from voltage-dependent toward voltage-independent regulation of the intracellular calcium level, and inhibition of non-voltage dependent calcium influx contributes to maintenance of the contractile phenotype. One possible mediator of calcium-dependent signaling is the FAK-family non-receptor protein kinase Pyk2, which is activated by a number of stimuli in a calcium-dependent manner. We used the Pyk2 inhibitor PF-4594755 and Pyk2 siRNA to investigate the role of Pyk2 in phenotype modulation in rat carotid artery smooth muscle cells and in cultured intact arteries. Pyk2 inhibition promoted the expression of smooth muscle markers at the mRNA and protein levels under stimulation by FBS or PDGF-BB and counteracted phenotype shift in cultured intact carotid arteries and balloon injury ex vivo. During long-term (24-96 hr) treatment with PF-4594755, smooth muscle markers increased before cell proliferation was inhibited, correlating with decreased KLF4 expression and differing from effects of MEK inhibition. The Pyk2 inhibitor reduced Orai1 and preserved SERCA2a expression in carotid artery segments in organ culture, and eliminated the inhibitory effect of PDGF stimulation on L-type calcium channel and large-conductance calcium-activated potassium channel expression in carotid cells. Basal intracellular calcium level, calcium wave activity, and store-operated calcium influx were reduced after Pyk2 inhibition of growth-stimulated cells. Pyk2 inhibition may provide an interesting approach for preserving vascular smooth muscle differentiation under pathophysiological conditions.


Subject(s)
Carotid Artery Injuries/enzymology , Cell Differentiation/drug effects , Focal Adhesion Kinase 2/antagonists & inhibitors , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Protein Kinase Inhibitors/pharmacology , Vasoconstriction/drug effects , Animals , Becaplermin , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Calcium Signaling/drug effects , Carotid Artery Injuries/genetics , Carotid Artery Injuries/physiopathology , Carotid Artery, Common/drug effects , Carotid Artery, Common/enzymology , Carotid Artery, Common/physiopathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Focal Adhesion Kinase 2/genetics , Focal Adhesion Kinase 2/metabolism , Gene Expression Regulation , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/enzymology , ORAI1 Protein/genetics , ORAI1 Protein/metabolism , Organ Culture Techniques , Phenotype , Proto-Oncogene Proteins c-sis/pharmacology , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Time Factors , Transfection
19.
J Vasc Res ; 54(6): 329-343, 2017.
Article in English | MEDLINE | ID: mdl-29040972

ABSTRACT

AIM: The objective of this study was to examine the effects of n-3 polyunsaturated fatty acids (n-3 PUFAs) on coronary arterial large conductance Ca2+-activated K+ (BK) channel function in coronary smooth muscle cells (SMCs) of streptozotocin-induced diabetic rats. METHODS: The effects of docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) on coronary BK channel open probabilities were determined using the patch clamp technique. The mRNA and protein expressions of BK channel subunits were measured using qRT-PCR and Western blots. The coronary artery tension and coronary SMC Ca2+ concentrations were measured using a myograph system and fluorescence Ca2+ indicator. RESULTS: Compared to nondiabetic control rats, the BK channel function was impaired with a reduced response to EPA and DHA in freshly isolated SMCs of diabetic rats. Oral administration of n-3 PUFAs had no effects on protein expressions of BK channel subunits in nondiabetic rats, but significantly enhanced those of BK-ß1 in diabetic rats without altering BK-α protein levels. Moreover, coronary ring tension induced by iberiotoxin (a specific BK channel blocker) was increased and cytosolic Ca2+ concentrations in coronary SMCs were decreased in diabetic rats, but no changes were found in nondiabetic rats. CONCLUSIONS: n-3 PUFAs protect the coronary BK channel function and coronary vasoreactivity in diabetic rats as a result of not only increasing BK-ß1 protein expressions, but also decreasing coronary artery tension and coronary smooth muscle cytosolic Ca2+ concentrations.


Subject(s)
Coronary Artery Disease/prevention & control , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/drug therapy , Diabetic Angiopathies/prevention & control , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/drug effects , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Vasoconstriction/drug effects , Animals , Calcium Signaling/drug effects , Coronary Artery Disease/genetics , Coronary Artery Disease/metabolism , Coronary Artery Disease/physiopathology , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Coronary Vessels/physiopathology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/physiopathology , Diabetic Angiopathies/genetics , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/physiopathology , Dose-Response Relationship, Drug , Drug Combinations , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/drug effects , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Membrane Potentials , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/metabolism , Potassium Channel Blockers/pharmacology , Rats, Sprague-Dawley , Time Factors
20.
J Physiol ; 594(1): 83-97, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26427907

ABSTRACT

KEY POINTS: Single-channel recordings in CA3 pyramidal neurons revealed that large-conductance calcium-activated K(+) (BK) channel open probability was reduced by loss of fragile X mental retardation protein (FMRP) and that FMRP acts on BK channels by modulating the channel's gating kinetics. Fmr1/BKß4 double knockout mice were generated to genetically upregulate BK channel activity in the absence of FMRP. Deletion of the BKß4 subunit alleviated reduced BK channel open probability via increasing BK channel open frequency, but not through prolonging its open duration. Genetic upregulation of BK channel activity via deletion of BKß4 normalized action potential duration, excessive glutamate release and short-term synaptic plasticity during naturalistic stimulus trains in excitatory hippocampal neurons in the absence of FMRP. Genetic upregulation of BK channel activity via deletion of BKß4 was sufficient to normalize excessive epileptiform activity in an in vitro model of seizure activity in the hippocampal circuit in the absence of FMRP. Loss of fragile X mental retardation protein (FMRP) causes fragile X syndrome (FXS), yet the mechanisms underlying the pathophysiology of FXS are incompletely understood. Recent studies identified important new functions of FMRP in regulating neural excitability and synaptic transmission via both translation-dependent mechanisms and direct interactions of FMRP with a number of ion channels in the axons and presynaptic terminals. Among these presynaptic FMRP functions, FMRP interaction with large-conductance calcium-activated K(+) (BK) channels, specifically their auxiliary ß4 subunit, regulates action potential waveform and glutamate release in hippocampal and cortical pyramidal neurons. Given the multitude of ion channels and mechanisms that mediate presynaptic FMRP actions, it remains unclear, however, to what extent FMRP-BK channel interactions contribute to synaptic and circuit defects in FXS. To examine this question, we generated Fmr1/ß4 double knockout (dKO) mice to genetically upregulate BK channel activity in the absence of FMRP and determine its ability to normalize multilevel defects caused by FMRP loss. Single-channel analyses revealed that FMRP loss reduced BK channel open probability, and this defect was compensated in dKO mice. Furthermore, dKO mice exhibited normalized action potential duration, glutamate release and short-term dynamics during naturalistic stimulus trains in hippocampal pyramidal neurons. BK channel upregulation was also sufficient to correct excessive seizure susceptibility in an in vitro model of seizure activity in hippocampal slices. Our studies thus suggest that upregulation of BK channel activity normalizes multi-level deficits caused by FMRP loss.


Subject(s)
Action Potentials , Excitatory Postsynaptic Potentials , Fragile X Syndrome/metabolism , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Pyramidal Cells/metabolism , Up-Regulation , Animals , CA3 Region, Hippocampal/cytology , CA3 Region, Hippocampal/metabolism , CA3 Region, Hippocampal/physiology , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Mice , Mice, Inbred C57BL , Pyramidal Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL