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.
Nat Commun ; 14(1): 7248, 2023 11 09.
Article in English | MEDLINE | ID: mdl-37945687

ABSTRACT

Progesterone (≥1 µM) is used in recovery of cerebral ischemia, an effect likely contributed to by cerebrovascular dilation. The targets of this progesterone action are unknown. We report that micromolar (µM) progesterone activates mouse cerebrovascular myocyte BK channels; this action is lost in ß1-/- mice myocytes and in lipid bilayers containing BK α subunit homomeric channels but sustained on ß1/ß4-containing heteromers. Progesterone binds to both regulatory subunits, involving two steroid binding sites conserved in ß1-ß4: high-affinity (sub-µM), which involves Trp87 in ß1 loop, and low-affinity (µM) defined by TM1 Tyr32 and TM2 Trp163. Thus progesterone, but not its oxime, bridges TM1-TM2. Mutation of the high-affinity site blunts channel activation by progesterone underscoring a permissive role of the high-affinity site: progesterone binding to this site enables steroid binding at the low-affinity site, which activates the channel. In support of our model, cerebrovascular dilation evoked by µM progesterone is lost by mutating Tyr32 or Trp163 in ß1 whereas these mutations do not affect alcohol-induced cerebrovascular constriction. Furthermore, this alcohol action is effectively counteracted both in vitro and in vivo by progesterone but not by its oxime.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channels , Progesterone , Mice , Animals , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Progesterone/pharmacology , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/chemistry , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Steroids/pharmacology , Binding Sites , Ethanol/pharmacology , Oximes/pharmacology
4.
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
5.
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
6.
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
7.
J Autism Dev Disord ; 52(10): 4213-4218, 2022 Oct.
Article in English | MEDLINE | ID: mdl-34562210

ABSTRACT

The aim of the study was to perform family-based association analysis of PRKCB1, CBLN1 and KCNMB4 gene polymorphisms and autism disorder. We comprised 206 Caucasian children with autistic spectrum disorder (ASD) and their biological parents. In transmission/disequilibrium test we observed that T-allele of the rs198198 polymorphism of the PRKCB1 gene was more often transmitted to affected children in the male subgroup (p = 0.010). Additionally, the T carrier state was significantly associated with hypotonia (p = 0.048). In the female subgroup, the T-allele carriers more often showed more mobile/vital behavior (p = 0.046). In conclusion, our study showed that the rs198198 of the PRKCB1 gene may be associated with ASD in men and with some features characteristic for the disorder.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Nerve Tissue Proteins/genetics , Protein Kinase C beta/genetics , Protein Precursors/genetics , Autism Spectrum Disorder/genetics , Autistic Disorder/genetics , Child , Cohort Studies , Female , Genetic Predisposition to Disease , Humans , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Male , Nerve Tissue Proteins/metabolism , Poland , Polymorphism, Single Nucleotide
8.
Acta Neuropathol Commun ; 9(1): 152, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34526147

ABSTRACT

Limbic-predominant age-related TDP-43 encephalopathy neuropathologic change (LATE-NC) is the most prevalent subtype of TDP-43 proteinopathy, affecting up to 1/3rd of aged persons. LATE-NC often co-occurs with hippocampal sclerosis (HS) pathology. It is currently unknown why some individuals with LATE-NC develop HS while others do not, but genetics may play a role. Previous studies found associations between LATE-NC phenotypes and specific genes: TMEM106B, GRN, ABCC9, KCNMB2, and APOE. Data from research participants with genomic and autopsy measures from the National Alzheimer's Coordinating Center (NACC; n = 631 subjects included) and the Religious Orders Study and Memory and the Rush Aging Project (ROSMAP; n = 780 included) were analyzed in the current study. Our goals were to reevaluate disease-associated genetic variants using newly collected data and to query whether the specific genotype/phenotype associations could provide new insights into disease-driving pathways. Research subjects included in prior LATE/HS genome-wide association studies (GWAS) were excluded. Single nucleotide variants (SNVs) within 10 kb of TMEM106B, GRN, ABCC9, KCNMB2, and APOE were tested for association with HS and LATE-NC, and separately for Alzheimer's pathologies, i.e. amyloid plaques and neurofibrillary tangles. Significantly associated SNVs were identified. When results were meta-analyzed, TMEM106B, GRN, and APOE had significant gene-based associations with both LATE and HS, whereas ABCC9 had significant associations with HS only. In a sensitivity analysis limited to LATE-NC + cases, ABCC9 variants were again associated with HS. By contrast, the associations of TMEM106B, GRN, and APOE with HS were attenuated when adjusting for TDP-43 proteinopathy, indicating that these genes may be associated primarily with TDP-43 proteinopathy. None of these genes except APOE appeared to be associated with Alzheimer's-type pathology. In summary, using data not included in prior studies of LATE or HS genomics, we replicated several previously reported gene-based associations and found novel evidence that specific risk alleles can differentially affect LATE-NC and HS.


Subject(s)
Apolipoproteins E/genetics , Hippocampus/pathology , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Progranulins/genetics , Sulfonylurea Receptors/genetics , Aged, 80 and over , Cohort Studies , Female , Follow-Up Studies , Genetic Predisposition to Disease/epidemiology , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study/methods , Humans , Male , Retrospective Studies , Sclerosis
9.
Cardiovasc Res ; 117(3): 792-804, 2021 02 22.
Article in English | MEDLINE | ID: mdl-32251501

ABSTRACT

AIMS: Our recent study demonstrated that increased Ca2+ sparks and spontaneous transient outward currents (STOCs) played an important role in uterine vascular tone and haemodynamic adaptation to pregnancy. The present study examined the role of ryanodine receptor (RyR) subtypes in regulating Ca2+ sparks/STOCs and myogenic tone in uterine arterial adaptation to pregnancy. METHODS AND RESULTS: Uterine arteries isolated from non-pregnant and near-term pregnant sheep were used in the present study. Pregnancy increased the association of α and ß1 subunits of large-conductance Ca2+-activated K+ (BKCa) channels and enhanced the co-localization of RyR1 and RyR2 with the ß1 subunit in the uterine artery. In contrast, RyR3 was not co-localized with BKCa ß1 subunit. Knockdown of RyR1 or RyR2 in uterine arteries of pregnant sheep downregulated the ß1 but not α subunit of the BKCa channel and decreased the association of α and ß1 subunits. Unlike RyR1 and RyR2, knockdown of RyR3 had no significant effect on either expression or association of BKCa subunits. In addition, knockdown of RyR1 or RyR2 significantly decreased Ca2+ spark frequency, suppressed STOCs frequency and amplitude, and increased pressure-dependent myogenic tone in uterine arteries of pregnant animals. RyR3 knockdown did not affect Ca2+ sparks/STOCs and myogenic tone in the uterine artery. CONCLUSION: Together, the present study demonstrates a novel mechanistic paradigm of RyR subtypes in the regulation of Ca2+ sparks/STOCs and uterine vascular tone, providing new insights into the mechanisms underlying uterine vascular adaptation to pregnancy.


Subject(s)
Calcium Signaling , Calcium/metabolism , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Uterine Artery/metabolism , Vasoconstriction , Adaptation, Physiological , Animals , Arterial Pressure , Female , Gene Expression Regulation , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Pregnancy , Ryanodine Receptor Calcium Release Channel/genetics , Sheep, Domestic , Tissue Culture Techniques
10.
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
11.
Cell Transplant ; 29: 963689720964382, 2020.
Article in English | MEDLINE | ID: mdl-33028109

ABSTRACT

Long noncoding RNA (lncRNA) is emerging as an essential regulator in the development and progression of cancer, including cervical cancer (CC). In this study, we found a CC-related lncRNA, KCNMB2-AS1, which was significantly overexpressed in CC and linked to poor outcomes. Depletion of KCNMB2-AS1 remarkably inhibited CC cell proliferation and induced apoptosis. In vivo xenograft models revealed that knockdown of KCNMB2-AS1 evidently delayed tumor growth. Mechanistically, KCNMB2-AS1 was predominantly located in the cytoplasm and served as a competing endogenous RNA to abundantly sponge miR-130b-5p and miR-4294, resulting in the upregulation of IGF2BP3, a well-documented oncogene in CC. Moreover, IGF2BP3 was able to bind KCNMB2-AS1 by three N6-methyladenosine (m6A) modification sites on KCNMB2-AS1, in which IGF2BP3 acted as an m6A "reader" and stabilized KCNMB2-AS1. Thus, KCNMB2-AS1 and IGF2BP3 formed a positive regulatory circuit that enlarged the tumorigenic effect of KCNMB2-AS1 in CC. Together, our data clearly suggest that KCNMB2-AS1 is a novel oncogenic m6A-modified lncRNA in CC, targeting KCNMB2-AS1 and its related molecules implicate the therapeutic possibility for CC patients.


Subject(s)
Cell Proliferation/physiology , Uterine Cervical Neoplasms/metabolism , Adult , Animals , Blotting, Western , Cell Proliferation/genetics , Female , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Male , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Middle Aged , Prognosis , Real-Time Polymerase Chain Reaction , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology
12.
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
13.
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
14.
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
15.
Invest Ophthalmol Vis Sci ; 61(3): 41, 2020 03 09.
Article in English | MEDLINE | ID: mdl-32203982

ABSTRACT

Purpose: The large-conductance calcium-activated potassium channel KCa1.1 (BKCa, maxi-K) influences aqueous humor outflow facility, but the contribution of auxiliary ß-subunits to KCa1.1 activity in the outflow pathway is unknown. Methods: Using quantitative polymerase chain reaction, we measured expression of ß-subunit genes in anterior segments of C57BL/6J mice (Kcnmb1-4) and in cultured human trabecular meshwork (TM) and Schlemm's canal (SC) cells (KCNMB1-4). We also measured expression of Kcnma1/KCNMA1 that encodes the pore-forming α-subunit. Using confocal immunofluorescence, we visualized the distribution of ß4 in the conventional outflow pathway of mice. Using iPerfusion, we measured outflow facility in enucleated mouse eyes in response to 100 or 500 nM iberiotoxin (IbTX; N = 9) or 100 nM martentoxin (MarTX; N = 12). MarTX selectively blocks ß4-containing KCa1.1 channels, whereas IbTX blocks KCa1.1 channels that lack ß4. Results: Kcnmb4 was the most highly expressed ß-subunit in mouse conventional outflow tissues, expressed at a level comparable to Kcnma1. ß4 was present within the juxtacanalicular TM, appearing to label cellular processes connecting to SC cells. Accordingly, KCNMB4 was the most highly expressed ß-subunit in human TM cells, and the sole ß-subunit in human SC cells. To dissect functional contribution, MarTX decreased outflow facility by 35% (27%, 42%; mean, 95% confidence interval) relative to vehicle-treated contralateral eyes, whereas IbTX reduced outflow facility by 16% (6%, 25%). Conclusions: The ß4-subunit regulates KCa1.1 activity in the conventional outflow pathway, significantly influencing outflow function. Targeting ß4-containing KCa1.1 channels may be a promising approach to lower intraocular pressure to treat glaucoma.


Subject(s)
Aqueous Humor/physiology , Gene Expression Regulation/physiology , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Nerve Tissue Proteins/genetics , Trabecular Meshwork/metabolism , Adult , Animals , Cells, Cultured , Humans , Infant , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/antagonists & inhibitors , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/antagonists & inhibitors , Limbus Corneae/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Middle Aged , Porins/metabolism , Real-Time Polymerase Chain Reaction , Toxins, Biological/pharmacology
16.
Mol Brain ; 13(1): 22, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32070382

ABSTRACT

The anterior cingulate cortex (ACC) serves as a critical hub for the anxiety and pain perception. The large-conductance Ca2+-activated potassium channels, or BKCa channels, are ubiquitously expressed throughout the central nervous system including the cingulate cortex. However, what changes of cortical BKCa channels undergo in the ACC remains unknown in pain-related anxiety. In the present study, a significant upregulation of synaptic and non-synaptic BKCa channel accessory ß4 subunits in the ACC was accompanied with pain-associated anxiety-like behaviors in the chronic compression of multiple dorsal root ganglia (mCCD) of the rat. NS1619, an opener of BKCa channels, significantly rescued the alteration of fAHP and AP duration of ACC pyramidal neurons in mCCD rats. The mRNA expression of BKCa ß4 subunits was extremely upregulated in the ACC after mCCD with the increased amount of both synaptic and non-synaptic BKCa ß4 subunit protein. Meanwhile, NS1619 reversed the enhanced AMPA receptor-mediated spontaneous excitatory postsynaptic current (sEPSC) frequency and the attenuated PPR of ACC neurons in mCCD rats. Local activation of BKCa channels in the ACC reversed mechanical allodynia and anxiety-like behaviors. These results suggest that the upregulation of postsynaptic and presynaptic BKCa ß4 subunit may contribute to neuronal hyperexcitability and the enhanced synaptic transmission in the ACC in neuropathic pain state, and then may result in anxiety-like behavior induced by neuropathic pain.


Subject(s)
Anxiety/metabolism , Gyrus Cinguli/metabolism , Hyperalgesia/metabolism , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Nerve Tissue Proteins/genetics , Up-Regulation , Animals , Anxiety/physiopathology , Behavior, Animal , Benzimidazoles/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Ganglia, Spinal/physiopathology , Gyrus Cinguli/drug effects , Gyrus Cinguli/physiopathology , Hyperalgesia/complications , Hyperalgesia/physiopathology , Indoles/pharmacology , Ion Channel Gating/drug effects , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Male , Nerve Tissue Proteins/metabolism , Neuralgia/complications , Neuralgia/metabolism , Neuralgia/physiopathology , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Receptors, AMPA/metabolism , Synapses/drug effects , Synapses/metabolism , Up-Regulation/drug effects
17.
Cardiovasc Res ; 116(7): 1372-1385, 2020 06 01.
Article in English | MEDLINE | ID: mdl-31738403

ABSTRACT

AIMS: The myogenic reactivity of the middle cerebral arteries (MCA) protects the brain by altering the diameter in response to changes in lumen pressure. Large conductance potassium (BK) channels are known to regulate the myogenic reactivity, yet, it is not clear how aging alters the myogenic reactivity via the BK channel in males and females. Thus, we hypothesize that age-associated changes in BK channel subunits modulate the myogenic reactivity in a sex-specific manner. METHODS AND RESULTS: We used vascular reactivity, patch-clamp, and biochemical methods to measure myogenic reactivity, BK channel function, and expression, respectively in cerebral vessels of adult and aged male and female Sprague Dawley rats. Our results suggest that aging and ovariectomy (OVX) exaggerated the myogenic reactivity of MCA in females but attenuated it in males. Aging induced outward eutrophic remodelling in females but inward hypertrophic remodelling in males. Aging decreased total, Kv, BK channel currents, and spontaneous transient outward currents (STOC) in vascular smooth muscle cells isolated from females, but not in males. Aging increased BKα subunit mRNA and protein both in males and females. However, aging decreased BKß1 subunit protein and mRNA in females only. In males, BKß1 mRNA is increased, but protein is decreased. Iberiotoxin-induced MCA constriction is lower in aged females but higher in aged males. Activation of BKα (10 µM NS1619) and BKß1 (10 µM S-Equol) subunits failed to increase STOCs and were unable to decrease the myogenic reactivity of MCA in aged female but not in aged male rats. OVX decreased, but chronic supplementation of oestradiol restored BK channel expression and function. CONCLUSION: Overall our results suggest that aging or OVX-associated downregulation of the BKß1 expression and function in females results in exaggerated myogenic reactivity of MCA. However, age-associated increase in BK channel function in males attenuated myogenic reactivity of MCA.


Subject(s)
Aging/metabolism , Cerebrovascular Circulation , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Middle Cerebral Artery/metabolism , Vasoconstriction , Age Factors , Animals , Arterial Pressure , Female , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Male , Membrane Potentials , Ovariectomy , Rats, Sprague-Dawley , Sex Factors , Signal Transduction , Vascular Remodeling
18.
Aging (Albany NY) ; 11(16): 6199-6216, 2019 08 21.
Article in English | MEDLINE | ID: mdl-31480021

ABSTRACT

Overactive bladder (OAB) is mostly observed in obese individuals, and is associated with enhanced excitability and contractility of the detrusor smooth muscle (DSM). Large-conductance voltage- and Ca2+-activated K+ (BK) channels reduce the excitability and contractility of the DSM. We tested whether obesity-induced OAB is associated with altered BK channel expression and activity in the DSM. Seven-week-old female Sprague-Dawley rats (N=80) were fed a normal or high-fat diet (HFD) for 12 weeks. HFD-fed rats exhibited a higher average bodyweight and urodynamically established detrusor overactivity. mRNA levels of the Kcnma1 (BKα subunit) and Kcnmb1 (BKß1 subunit) in whole tissues and cells from the DSM were reduced in HFD-fed rats. A selective BK channel opener, NS1619, was then applied to DSM cells from the two groups of rats. Patch-clamp techniques revealed that spontaneous transient outward currents, NS1619-induced activation of spontaneous transient outward currents, and whole-cell BK currents, as well as NS1619-induced membrane hyperpolarization, were attenuated in DSM cells from HFD-fed rats. The relaxation effect of NS1619 on contractility was reduced in DSM strips from HFD-fed rats. Thus, impaired expression of Kcnma1 and Kcnmb1 in the DSM contributes to obesity-induced OAB, suggesting that BK channels could be a useful treatment targets in OAB.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Obesity/chemically induced , Urinary Bladder, Overactive/pathology , Animals , Benzimidazoles/pharmacology , Diet, High-Fat/adverse effects , Electric Stimulation , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Obesity/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Urinary Bladder/drug effects , Urinary Bladder/physiology
19.
Int J Mol Med ; 44(4): 1585-1593, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31364720

ABSTRACT

Neuropathic pain is one of most intense types of chronic pain. Numerous studies investigating neuropathic pain have described the critical involvement of microglia in the spinal cord. Previous studies have indicated that activation of large conductance Ca2+­activated K+ (BK) channels contributes to microglial activation in the spinal dorsal horn (SDH) and the generation of neuropathic pain. However, the specific role of BK channels in spinal microglia in neuropathic pain has not been fully addressed in previous studies, as BK channel inhibitors were used to inhibit microglial BK channel based on their inhibitory kinetics. We previously identified that Ca2+­activated K+ channel ß3 auxiliary subunit (KCNMB3), which is an auxiliary subunit of BK channels and regulates gating properties of the channel, is exclusively expressed in microglia in the spinal cord. The present study analyzed the role of BK channels in spinal microglia in neuropathic pain using a spinal microglia­specific BK channel knockdown method, with intrathecal injection of KCNMB3 small interfering RNA. Neuropathic pain was significantly attenuated in KCNMB3 knockdown mice. Increases in the number of microglia in the SDH following nerve injury were attenuated by KCNMB3 knockdown. Furthermore, increased levels of pain­associated molecules in the SDH were attenuated in KCNMB3 knockdown mice. Attempts were also made to analyze the effects of KCNMB3 knockdown on chronic pain. KCNMB3 knockdown ameliorated chronic pain and inhibited the expression levels of pain­associated molecules in the SDH. The results from the present study suggested that BK channels modulated the activation state of spinal microglia, and that KCNMB3 is a potential therapeutic target for neuropathic pain.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Microglia/metabolism , Neuralgia/etiology , Neuralgia/metabolism , Spinal Cord/cytology , Spinal Cord/metabolism , Animals , Disease Models, Animal , Gene Silencing , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Male , Mice , Mice, Knockout
20.
Medicine (Baltimore) ; 98(23): e15828, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31169684

ABSTRACT

BACKGROUND: The K channel, subfamily J, member-11 (KCNJ11) E23K and ß1 subunit of large-conductance Ca-activated K channel (KCNMB1) E65K polymorphisms were shown to be associated with the risk of essential hypertension (EH). However, the results were inconclusive with relatively small sample size. Thus, we carried out a meta-analysis to investigate the genetic association between KCNJ11 E23K and KCNMB1 E65K polymorphisms and essential hypertension risk. METHODS: Relative studies were collected using PubMed, Web of Science, the Cochrane Library databases, Chinese National Knowledge Infrastructure and Embase databases. Pooled odds ratios with 95% confidence intervals were used to assess the strength of associations. RESULTS: The dominant models of KCNJ11 E23K (P = .006, OR [95%CI] = 0.45 [0.25, 0.79]) and KCNMB1 E65K (P = .04, OR [95%CI] = 0.91 [0.83, 1.00]) were significantly associated with essential hypertension risk. No significant association was detected between the allelic and recessive models of KCNJ11 E23K and KCNMB1 E65K and the susceptibility of EH. Subgroup analysis stratified by ethnicity showed that the dominant model of KCNMB1 E65K was associated with EH risk in Asian population (P = .003, OR [95%CI] = 0.83 [0.74, 0.94]), but not in Caucasian (P = .74, OR [95%CI] = 1.02 [0.89, 1.18]). CONCLUSIONS: The dominant model of KCNJ11 E23K and KCNMB1 E65K might be susceptible factors for essential hypertension. To confirm this result, large-scale case-control studies with more subjects are necessary.


Subject(s)
Asian People/genetics , Essential Hypertension/genetics , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Potassium Channels, Inwardly Rectifying/genetics , Adult , Aged , Female , Genetic Predisposition to Disease/genetics , Humans , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...