Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 148
Filter
1.
Biochim Biophys Acta Biomembr ; 1863(4): 183555, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33444624

ABSTRACT

Expression, purification, and functional reconstitution of mammalian ion channels are often challenging. Heterologous expression of mammalian channels in bacteria can be advantageous due to unrelated protein environment and the lack of risk of copurification of endogenous proteins, e.g., accessory channel subunits that can influence the channel activity. Also, direct recording of channel activity could be challenging due to their intracellular localization like in the case of mitochondrial channels. The activity of purified channels can be characterized at the single-molecule level by electrophysiological techniques, such as planar lipid bilayers (PLB). In this work, we describe a simple approach to accomplish PLB recording of the activity of single renal outer medullary potassium channels ROMK expressed in E. coli. We focused on the ROMK2 isoform that is present at low levels in the mitochondria and can be responsible for mitoKATP activity. We screened for the best construct to express the codon-optimized ROMK proteins with a 6xHis tag for protein purification. The strategy involved the use of optimal styrene-maleic acid (SMA) copolymer, which forms so-called polymer nanodiscs, to solubilize and purify ROMK-containing SMA lipid particles (SMALPs), which were amenable for fusion with PLB. Reconstituted ROMK channels exhibited ion selectivity, rectification, and pharmacological properties, which are in agreement with previous work on ROMK channels.


Subject(s)
Maleates/chemistry , Nanostructures/chemistry , Potassium Channels, Inwardly Rectifying/chemistry , Styrene/chemistry , Humans , Potassium Channels, Inwardly Rectifying/biosynthesis , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/isolation & purification , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification
2.
Heart Surg Forum ; 23(5): E579-E585, 2020 Aug 12.
Article in English | MEDLINE | ID: mdl-32990585

ABSTRACT

BACKGROUND: Soluble epoxide hydrolase inhibitors (sEHi) have anti-arrhythmic effects, and we previously found that the novel sEHi t-AUCB (trans-4[-4-(3-adamantan-1-yl-ureido)-cyclohexyloxy]-benzoic acid) significantly inhibited ventricular arrhythmias after myocardial infarction (MI). However, the mechanism is unknown. It's known that microRNA-29 (miR-29) participates in the occurrence of arrhythmias. In this study, we investigated whether sEHi t-AUCB was protective against ischemic arrhythmias by modulating miR-29 and its target genes KCNJ12 and KCNIP2. METHODS: Male 8-week-old C57BL/6 mice were divided into five groups and fed distilled water only or distilled water with t-AUCB of different dosages for seven days. Then, the mice underwent MI or sham surgery. The ischemic region of the myocardium was obtained 24 hours after MI to detect miR-29, KCNJ12, and KCNIP2 mRNA expression levels via real-time PCR and KCNJ12 and KCNIP2 protein expression levels via western blotting. RESULTS: MiR-29 expression levels were significantly increased in the ischemic region of MI mouse hearts and the mRNA and protein expression levels of its target genes KCNJ12 and KCNIP2 were significantly decreased. T-AUCB prevented these changes dose-dependently. CONCLUSION: The sEHi t-AUCB regulates the expression levels of miR-29 and its target genes KCNJ12 and KCNIP2, suggesting a possible mechanism for its potential therapeutic application in ischemic arrhythmia.


Subject(s)
Gene Expression Regulation , Kv Channel-Interacting Proteins/genetics , MicroRNAs/genetics , Myocardial Infarction/genetics , Myocardium/metabolism , Potassium Channels, Inwardly Rectifying/genetics , Animals , Blotting, Western , Disease Models, Animal , Down-Regulation , Kv Channel-Interacting Proteins/biosynthesis , Male , Mice , Mice, Inbred C57BL , MicroRNAs/biosynthesis , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/pathology , Potassium Channels, Inwardly Rectifying/biosynthesis , RNA/genetics , RNA/metabolism
3.
Invest Ophthalmol Vis Sci ; 61(6): 46, 2020 06 03.
Article in English | MEDLINE | ID: mdl-32572457

ABSTRACT

Purpose: Diabetic retinopathy (DR) is a leading cause of visual impairment. Müller cells in DR are dysfunctional due to downregulation of the inwardly rectifying potassium channel Kir4.1. Metformin, a commonly used oral antidiabetic drug, is known to elicit its action through 5' adenosine monophosphate-activated protein kinase (AMPK), a cellular metabolic regulator; however, its effect on Kir4.1 channels is unknown. For this study, we hypothesized that metformin treatment would correct circadian rhythm disruption and Kir4.1 channel dysfunction in db/db mice. Methods: Metformin was given orally to db/db mice. Wheel-running activity, retinal levels of Kir4.1, and AMPK phosphorylation were determined at study termination. In parallel, rat retinal Müller cell line (rMC-1) cells were treated using metformin and 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) to assess the effect of AMPK activation on the Kir4.1 channel. Results: The wheel-running activity of the db/db mice was improved following the metformin treatment. The Kir4.1 level in Müller cells was corrected after metformin treatment. Metformin treatment led to an upregulation of clock regulatory genes such as melanopsin (Opn4) and aralkylamine N-acetyltransferase (Aanat). In rMC-1 cells, AMPK activation via AICAR and metformin resulted in increased Kir4.1 and intermediate core clock component Bmal-1 protein expression. The silencing of Prkaa1 (gene for AMPKα1) led to decreased Kir4.1 and Bmal-1 protein expression. Conclusions: Our findings demonstrate that metformin corrects abnormal circadian rhythm and Kir4.1 channels in db/db mouse a model of type 2 diabetes. Metformin could represent a critical pharmacological agent for preventing Müller cell dysfunction observed in human DR.


Subject(s)
Circadian Rhythm/physiology , Diabetes Mellitus, Experimental/drug therapy , Diabetic Retinopathy/drug therapy , Gene Expression Regulation , Metformin/pharmacology , Potassium Channels, Inwardly Rectifying/genetics , Retinal Ganglion Cells/metabolism , Animals , Cells, Cultured , Circadian Rhythm/drug effects , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetic Retinopathy/genetics , Diabetic Retinopathy/metabolism , Disease Models, Animal , Hypoglycemic Agents/pharmacology , Male , Mice , Mice, Transgenic , Potassium Channels, Inwardly Rectifying/biosynthesis , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology
4.
Glia ; 68(10): 2119-2135, 2020 10.
Article in English | MEDLINE | ID: mdl-32220118

ABSTRACT

Spinal microglia change their phenotype and proliferate after nerve injury, contributing to neuropathic pain. For the first time, we have characterized the electrophysiological properties of microglia and the potential role of microglial potassium channels in the spared nerve injury (SNI) model of neuropathic pain. We observed a strong increase of inward currents restricted at 2 days after injury associated with hyperpolarization of the resting membrane potential (RMP) in microglial cells compared to later time-points and naive animals. We identified pharmacologically and genetically the current as being mediated by Kir2.1 ion channels whose expression at the cell membrane is increased 2 days after SNI. The inhibition of Kir2.1 with ML133 and siRNA reversed the RMP hyperpolarization and strongly reduced the currents of microglial cells 2 days after SNI. These electrophysiological changes occurred coincidentally to the peak of microglial proliferation following nerve injury. In vitro, ML133 drastically reduced the proliferation of BV2 microglial cell line after both 2 and 4 days in culture. In vivo, the intrathecal injection of ML133 significantly attenuated the proliferation of microglia and neuropathic pain behaviors after nerve injury. In summary, our data implicate Kir2.1-mediated microglial proliferation as an important therapeutic target in neuropathic pain.


Subject(s)
Cell Proliferation/physiology , Microglia/metabolism , Neuralgia/metabolism , Potassium Channel Blockers/administration & dosage , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Spinal Cord/metabolism , Animals , Cell Line, Transformed , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Imidazoles/administration & dosage , Injections, Spinal , Male , Mice , Mice, Transgenic , Microglia/drug effects , Neuralgia/prevention & control , Phenanthrolines/administration & dosage , Potassium Channels, Inwardly Rectifying/biosynthesis , Spinal Cord/cytology , Spinal Cord/drug effects
5.
Ann Surg Oncol ; 27(7): 2559-2568, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32052303

ABSTRACT

BACKGROUND: We aimed to clarify the role of potassium voltage-gated channel subfamily J member 15 (KCNJ15) in esophageal squamous cell carcinoma (ESCC) cells and its potential as a prognosticator in ESCC patients. METHODS: KCNJ15 transcription levels were evaluated in 13 ESCC cell lines and polymerase chain reaction (PCR) array analysis was conducted to detect coordinately expressed genes with KCNJ15. The biological functions of KCNJ15 in cell invasion, proliferation, migration, and adhesion were validated through small interfering RNA-mediated knockdown experiments. Cell proliferation was further evaluated through the forced expression experiment. KCNJ15 expression was detected in 200 ESCC tissues by quantitative real-time reverse transcription PCR (qRT-PCR) and analyzed in 64 representative tissues by immunohistochemistry. Correlations between KCNJ15 expression levels and clinicopathological features were also analyzed. RESULTS: The KCNJ15 expression levels varied widely in ESCC cell lines and correlated with COL3A1, JAG1, and F11R. Knockdown of KCNJ15 expression significantly repressed cell invasion, proliferation, and migration of ESCC cells in vitro. Furthermore, overexpression of KCNJ15 resulted in increased cell proliferation. Patients were stratified using the cut-off value of KCNJ15 messenger RNA (mRNA) levels in 200 ESCC tissues using receiver operating characteristic curve analysis; the high KCNJ15 expression group had significantly shorter overall and disease-free survival times. In multivariable analysis, high expression of KCNJ15 was identified as an independent poor prognostic factor. Staining intensity of in situ KCNJ15 protein expression tended to be associated with KCNJ15 mRNA expression levels. CONCLUSIONS: KCNJ15 is involved in aggressive tumor phenotypes of ESCC cells and its tissue expression levels may be useful as a prognosticator of patients with ESCC.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Potassium Channels, Inwardly Rectifying , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Squamous Cell Carcinoma/pathology , Humans , Potassium Channels, Inwardly Rectifying/biosynthesis , Potassium Channels, Inwardly Rectifying/genetics , Prognosis
6.
Invest Ophthalmol Vis Sci ; 60(6): 1928-1936, 2019 05 01.
Article in English | MEDLINE | ID: mdl-31042800

ABSTRACT

Purpose: Diabetes leads to the downregulation of the retinal Kir4.1 channels and Müller cell dysfunction. The insulin receptor substrate-1 (IRS-1) is a critical regulator of insulin signaling in Müller cells. Circadian rhythms play an integral role in normal physiology; however, diabetes leads to a circadian dysrhythmia. We hypothesize that diabetes will result in a circadian dysrhythmia of IRS-1 and Kir4.1 and disturbed clock gene function will have a critical role in regulating Kir4.1 channels. Methods: We assessed a diurnal rhythm of retinal IRS-1 and Kir4.1 in db/db mice. The Kir4.1 function was evaluated using a whole-cell recording of Müller cells. The rat Müller cells (rMC-1) were used to undertake in vitro studies using a siRNA. Results: The IRS-1 exhibited a diurnal rhythm in control mice; however, with diabetes, this natural rhythm was lost. The Kir4.1 levels peaked and troughed at times similar to the IRS-1 rhythm. The IRS-1 silencing in the rMC-1 led to a decrease in Kir4.1 and BMAL1. The insulin treatment of retinal explants upregulated Kir4.1 possibly via upregulation of BMAL1 and phosphorylation of IRS-1 and Akt-1. Conclusions: Our studies highlight that IRS-1, by regulating BMAL1, is an important regulator of Kir4.1 in Müller cells and the dysfunctional signaling mediated by IRS-1 may be detrimental to Kir4.1.


Subject(s)
ARNTL Transcription Factors/genetics , Circadian Rhythm/physiology , Diabetic Retinopathy/genetics , Ependymoglial Cells/metabolism , Gene Expression Regulation , Insulin Receptor Substrate Proteins/genetics , Potassium Channels, Inwardly Rectifying/genetics , ARNTL Transcription Factors/biosynthesis , Animals , Blotting, Western , Cells, Cultured , Diabetes Mellitus, Experimental , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Ependymoglial Cells/pathology , Humans , Insulin Receptor Substrate Proteins/biosynthesis , Mice , Polymerase Chain Reaction , Potassium Channels, Inwardly Rectifying/biosynthesis , RNA/genetics , Rats
8.
Neuron ; 98(2): 306-319.e7, 2018 04 18.
Article in English | MEDLINE | ID: mdl-29606582

ABSTRACT

Diversified neurons are essential for sensorimotor function, but whether astrocytes become specialized to optimize circuit performance remains unclear. Large fast α-motor neurons (FαMNs) of spinal cord innervate fast-twitch muscles that generate peak strength. We report that ventral horn astrocytes express the inward-rectifying K+ channel Kir4.1 (a.k.a. Kcnj10) around MNs in a VGLUT1-dependent manner. Loss of astrocyte-encoded Kir4.1 selectively altered FαMN size and function and led to reduced peak strength. Overexpression of Kir4.1 in astrocytes was sufficient to increase MN size through activation of the PI3K/mTOR/pS6 pathway. Kir4.1 was downregulated cell autonomously in astrocytes derived from amyotrophic lateral sclerosis (ALS) patients with SOD1 mutation. However, astrocyte Kir4.1 was dispensable for FαMN survival even in the mutant SOD1 background. These findings show that astrocyte Kir4.1 is essential for maintenance of peak strength and suggest that Kir4.1 downregulation might uncouple symptoms of muscle weakness from MN cell death in diseases like ALS.


Subject(s)
Astrocytes/metabolism , Motor Neurons/metabolism , Potassium Channels, Inwardly Rectifying/biosynthesis , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Animals, Newborn , Astrocytes/chemistry , Astrocytes/pathology , Cells, Cultured , Female , Humans , Induced Pluripotent Stem Cells/chemistry , Induced Pluripotent Stem Cells/metabolism , Male , Mice , Mice, Transgenic , Motor Neurons/chemistry , Motor Neurons/pathology , Organ Culture Techniques , Potassium Channels, Inwardly Rectifying/analysis
9.
PLoS One ; 13(2): e0193280, 2018.
Article in English | MEDLINE | ID: mdl-29474462

ABSTRACT

Diabetic retinopathy (DR) is a major cause of adult blindness. Retinal Müller cells maintain water homeostasis and potassium concentration via inwardly rectifying Kir4.1 channels. Accumulation of advanced glycation end products (AGEs) is a major pathologic event in DR. While diabetes leads to a decrease in the Kir4.1 channels, it remains unknown whether AGEs-linked to the basement membrane (BM) affect normal Kir4.1 channels. For this study, we hypothesized that AGE-modification of laminin is detrimental to Kir4.1 channels, therefore, disrupting Müller cell function. The AGE-modified laminin-coated substrates were prepared by incubating Petri-dishes with laminin and methylglyoxal for seven days. The rat Müller cells (rMC-1) were propagated on AGE-modified laminin, and Kir4.1 expression and function were evaluated. Quantification of AGEs using ELISA revealed a dose-dependent increase in methylglyoxal-hydro-imidazolone adducts. The rMC-1 propagated on AGE-modified laminin demonstrated a decrease in Kir4.1 levels in immunofluorescence and western blot studies and a decrease in the Kir4.1 channel function. Kir4.1 decrease on AGE-modified laminin resulted in a disorganization of an actin cytoskeleton and disruption of α-dystroglycan-syntrophin-dystrophin complexes. Our studies suggest that AGE-modification of laminin is detrimental to Kir4.1 channels. By studying the role of AGEs in Kir4.1 channels we have identified a novel mechanism of Müller cell dysfunction and its subsequent involvement in DR.


Subject(s)
Diabetic Retinopathy/metabolism , Ependymoglial Cells/metabolism , Glycation End Products, Advanced/metabolism , Laminin/metabolism , Potassium Channels, Inwardly Rectifying/biosynthesis , Retina/metabolism , Animals , Cell Line , Diabetic Retinopathy/pathology , Ependymoglial Cells/pathology , Glycation End Products, Advanced/chemistry , Glycation End Products, Advanced/pharmacology , Laminin/chemistry , Laminin/pharmacology , Rats , Retina/pathology
10.
Int J Cardiol ; 255: 61-68, 2018 Mar 15.
Article in English | MEDLINE | ID: mdl-29290419

ABSTRACT

BACKGROUND: Basal and acetylcholine-gated inward-rectifier K+-currents (IK1 and IK,ACh, respectively) are altered in atrial fibrillation (AF). Gi-protein-coupled muscarinic (M) receptors type-2 are considered the predominant receptors activating IK,ACh. Although a role for Gq-coupled non-M2-receptor subtypes has been suggested, the precise regulation of IK,ACh by multiple M-receptor subtypes in the human atrium is unknown. Here, we investigated M1-receptor-mediated IK,ACh regulation and its remodeling in chronic AF (cAF). METHODS AND RESULTS: M1-receptor mRNA and protein abundance were increased in atrial cardiomyocyte fractions and atrial homogenates from cAF patients, whereas M2-receptor levels were unchanged. The regulation of IK,ACh by M1-receptors was investigated in right-atrial cardiomyocytes using two applications of the M-receptor agonist carbachol (CCh, 2µM), with pharmacological interventions during the second application. CCh application produced a rapid current increase (Peak-IK,ACh), which declined to a quasi-steady-state level (Qss-IK,ACh). In sinus rhythm (Ctl) the selective M1-receptor antagonists pirenzepine (10nM) and muscarinic toxin-7 (MT-7, 10nM) significantly inhibited CCh-activated Peak-IK,ACh, whereas in cAF they significantly reduced both Peak- and Qss-IK,ACh, with no effects on basal inward-rectifier currents in either group. Conversely, the selective M1-receptor agonist McN-A-343 (100µM) induced a current similar to the CCh-activated current in Ctl atrial cardiomyocytes pretreated with pertussis toxin to inhibit M2-receptor-mediated Gi-protein signaling, which was abolished by MT-7. Computational modeling indicated that M1- and M2-receptors redundantly activate IK,ACh to abbreviate APD, albeit with predominant effects of M2-receptors. CONCLUSION: Our data suggest that Gq-coupled M1-receptors also regulate human atrial IK,ACh and that their relative contribution to IK,ACh activation is increased in cAF patients. We provide novel insights about the role of non-M2-receptors in human atrial cardiomyocytes, which may have important implications for understanding AF pathophysiology.


Subject(s)
Acetylcholine/pharmacology , Atrial Fibrillation/metabolism , Myocytes, Cardiac/physiology , Potassium Channels, Inwardly Rectifying/biosynthesis , Receptor, Muscarinic M1/biosynthesis , Up-Regulation/physiology , Atrial Fibrillation/pathology , Cells, Cultured , Chronic Disease , Dose-Response Relationship, Drug , Heart Atria/metabolism , Heart Atria/pathology , Humans , Muscarinic Antagonists/pharmacology , Myocytes, Cardiac/drug effects , Receptor, Muscarinic M1/antagonists & inhibitors , Up-Regulation/drug effects
11.
Mech Ageing Dev ; 166: 48-54, 2017 09.
Article in English | MEDLINE | ID: mdl-28705548

ABSTRACT

INTRODUCTION: The left ventricular hypertrophy (LVH)-ventricular arrhythmias relationship associated with arterial hypertension and aging remains controversial. We aimed to assess the age-dependency of ventricular arrhythmias in spontaneously hypertensive rats (SHRs) and the corresponding ventricular structural and molecular remodeling. MATERIALS AND METHODS: Ventricular arrhythmias were quantified using 24-h radiotelemetry ECG monitoring in eight SHRs and four Wistar-Kyoto (WKY) rats at 14 (young), 24 (adult), and 48 (aging) weeks of age. Left ventricular histology and mRNA expressions of 89 proarrhythmogenic genes were assessed in six additional groups (n=4 each) of young, adult, and aging SHRs and WKYs. RESULTS: Regardless of their age, SHRs presented more premature ventricular contractions (PVCs) than age-matched WKYs (p<0.01). The arrhythmogenicity peak occurred in adult SHRs; ventricular tachycardias only occurred in adult SHRs. Among the SHRs, LV thickness, interstitial fibrosis, and the number of deregulated genes increased with age. Kcnj11 expression was deregulated in adult, but not in young or aging SHRs. DISCUSSION: This study confirms the presence of higher ventricular ectopy in SHRs than in age-matched WKYs. LVH appeared to be an adaptive, antiarrhythmic process. Myocardial energetic changes with advancing age, as reflected by Kcnj11 expression changes, could underlie this age-dependency of ventricular arrhythmias.


Subject(s)
Aging/metabolism , Arrhythmias, Cardiac/metabolism , Gene Expression Regulation , Hypertension/metabolism , Hypertrophy, Left Ventricular/metabolism , Potassium Channels, Inwardly Rectifying/biosynthesis , Ventricular Remodeling , Aging/pathology , Animals , Arrhythmias, Cardiac/pathology , Arrhythmias, Cardiac/physiopathology , Hypertension/pathology , Hypertension/physiopathology , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Male , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Risk Factors
12.
Invest Ophthalmol Vis Sci ; 58(5): 2473-2482, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28460049

ABSTRACT

Purpose: Diabetic patients often are affected by vision problems. We previously identified diabetic retinopathy (DR) as a disease of clock gene dysregulation. TNF-α, a proinflammatory cytokine, is known to be elevated in DR. Müller cells maintain retinal water homeostasis and K+ concentration via Kir4.1 channels. Notably, Kir4.1 expression is reduced in diabetes; however, the interplay of TNF-α, Kir4.1, and clock genes in Müller cells remains unknown. We hypothesize that the Kir4.1 in Müller cells is under clock regulation, and increase in TNF-α is detrimental to Kir4.1. Methods: Long-Evans rats were made diabetic using streptozotocin (STZ). Retinal Kir4.1 expression was determined at different time intervals. Rat Müller (rMC-1) cells were transfected with siRNA for Per2 or Bmal1 and in parallel treated with TNF-α (5-5000 pM) to determine Kir4.1 expression. Results: Kir4.1 expression exhibited a diurnal rhythm in the retina; however, with STZ-induced diabetes, Kir4.1 was reduced overall. Kir4.1 rhythm was maintained in vitro in clock synchronized rMC-1 cells. Clock gene siRNA-treated rMC-1 exhibited a decrease in Kir4.1 expression. TNF-α treatment of rMCs lead to a profound decrease in Kir4.1 due to reduced colocalization of Kir4.1 channels with synapse-associated protein (SAP97) and disorganization of the actin cytoskeleton. Conclusions: Our findings demonstrate that Kir4.1 channels possess a diurnal rhythm, and this rhythm is dampened with diabetes, thereby suggesting that the increase in TNF-α is detrimental to normal Kir4.1 rhythm and expression.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Retinopathy/genetics , Ependymoglial Cells/metabolism , Gene Expression Regulation , Potassium Channels, Inwardly Rectifying/genetics , RNA, Messenger/genetics , Tumor Necrosis Factor-alpha/therapeutic use , Animals , Blotting, Western , Cells, Cultured , Circadian Rhythm , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/metabolism , Ependymoglial Cells/drug effects , Ependymoglial Cells/pathology , Immunohistochemistry , Potassium Channels, Inwardly Rectifying/biosynthesis , Rats , Rats, Long-Evans , Real-Time Polymerase Chain Reaction , Retina/drug effects , Retina/metabolism , Retina/pathology
13.
Invest Ophthalmol Vis Sci ; 58(3): 1656-1664, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28297725

ABSTRACT

Purpose: To test the effects of rearing light intensity on retinal function and morphology in the retinoschisis knockout (Rs1-KO) mouse model of X-linked retinoschisis, and whether it affects functional outcome of RS1 gene replacement. Methods: Seventy-six Rs1-KO mice were reared in either cyclic low light (LL, 20 lux) or moderate light (ML, 300 lux) and analyzed at 1 and 4 months. Retinal function was assessed by electroretinogram and cavity size by optical coherence tomography. Expression of inward-rectifier K+ channel (Kir4.1), water channel aquaporin-4 (AQP4), and glial fibrillary acidic protein (GFAP) were analyzed by Western blotting. In a separate study, Rs1-KO mice reared in LL (n = 29) or ML (n = 27) received a unilateral intravitreal injection of scAAV8-hRs-IRBP at 21 days, and functional outcome was evaluated at 4 months by electroretinogram. Results: At 1 month, no functional or structural differences were found between LL- or ML-reared Rs1-KO mice. At 4 months, ML-reared Rs1-KO mice showed significant reduction of b-wave amplitude and b-/a-wave ratio with no changes in a-wave, and a significant increase in cavity size, compared to LL-reared animals. Moderate light rearing increased Kir4.1 expression in Rs1-KO mice by 4 months, but not AQP4 and GFAP levels. Administration of scAAV8-hRS1-IRBP to Rs1-KO mice showed similar improvement of inner retinal ERG function independent of LL or ML rearing. Conclusions: Rearing light conditions affect the development of retinal cavities and post-photoreceptor function in Rs1-KO mice. However, the effect of rearing light intensity does not interact with the efficacy of RS1 gene replacement in Rs1-KO mice.


Subject(s)
Genetic Therapy/methods , Light , Retinal Photoreceptor Cell Inner Segment/pathology , Retinoschisis/therapy , Animals , Blotting, Western , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Disease Models, Animal , Electroretinography , Eye Proteins/genetics , Eye Proteins/metabolism , Follow-Up Studies , Gene Expression Regulation , Gene Transfer Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Potassium Channels, Inwardly Rectifying/biosynthesis , Potassium Channels, Inwardly Rectifying/genetics , RNA/genetics , Retinal Photoreceptor Cell Inner Segment/radiation effects , Retinoschisis/diagnosis , Retinoschisis/genetics , Retinoschisis/physiopathology , Time Factors , Tomography, Optical Coherence
14.
Int J Cardiol ; 232: 192-198, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28082085

ABSTRACT

BACKGROUND: The E23K variant of the Kir6.2 subunit of the ATP-sensitive potassium (KATP) channel has been implicated in cardiac remodeling. However, the effects of E23K variant on ventricular electrophysiology and arrhythmogenesis remain unclear. METHODS: Transgenic rats were generated to express human E23K-variant genomic DNA in the heart under the α-myosin heavy chain promoter. Electrophysiological parameters including electrocardiograph, ventricular action potential duration (APD), effective refractory period (ERP), electrical alternans and ventricle arrhythmia threshold were examined in wild type (WT) and transgenic rats. The KATP current in cardiomyocytes was recorded using whole-cell patch clamp techniques. RESULTS: No differences in the electrophysiological parameters between the two groups were found at baseline. However, after acute ischemic stress, shortened QT intervals were further aggravated in the E23K-variant rats. Additionally, the E23K variant exacerbated the decrease of APD70, APD90 and ERP. The ventricular arrhythmia and alternans thresholds were significantly attenuated, and the duration of ventricular arrhythmia induced by electrical stimulation was significantly prolonged in the E23K-variant rats. More importantly, the KATP current in cardiomyocytes was significantly increased in the E23K-variant rats after ischemia. CONCLUSION: The E23K variant of the KATP channel increased the susceptibility to ventricular arrhythmia under acute ischemia stress.


Subject(s)
DNA/genetics , Gene Expression Regulation , Myocardial Reperfusion Injury/complications , Potassium Channels, Inwardly Rectifying/genetics , Tachycardia, Ventricular/genetics , Ventricular Function, Left/physiology , Ventricular Remodeling , Animals , Blotting, Western , Disease Models, Animal , Electrocardiography , Genotype , Heart Ventricles/physiopathology , KATP Channels/biosynthesis , KATP Channels/genetics , Myocardial Reperfusion Injury/metabolism , Polymerase Chain Reaction , Potassium Channels, Inwardly Rectifying/biosynthesis , Rats , Rats, Transgenic , Tachycardia, Ventricular/etiology , Tachycardia, Ventricular/metabolism
15.
Oncol Rep ; 36(5): 3005-3013, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27599582

ABSTRACT

To elucidate the complex molecular mechanisms of anaplastic thyroid carcinoma (ATC), the mRNA and miRNA expression profiles of ATC were systematically explored. A total of 55 common differentially expressed genes (DEGs) were obtained from two mRNA expression datasets including 23 ATC samples and 24 paired normal samples. Gene expression levels of three randomly selected DEGs, VCAN, COL5A1 and KCNJ16, were examined using RT-PCR in 10 ATC samples. Notably, the ATC and normal samples were clearly classified into two groups based on their common DEGs. Moreover 23 common DEGs, such as TG, NKX2-1, KCNJ16 and CTHRC1, were predicted to be the potential targets of 17 identified miRNAs in ATC. Meanwhile, several miRNA target genes were associated with biological processes related to tumor progression such as angiogenesis, cell migration or growth and potassium channel regulation. In summary, the poor prognosis of ATC is possibly caused via complex biological processes. Firstly, angiogenesis was activated by the high expression of CTHRC1, VCAN and POSTN, providing necessary nutrition for tumor cells. Then tumor distant metastasis was induced via stimulation of cell migration and cell growth or regulation of cell-cell interaction. Moreover, intracellular potassium concentration changes promoted ATC progression indirectly. Hence, identification of these critical DEGs was valuable in understanding the molecular mechanisms of ATC.


Subject(s)
Cell Adhesion Molecules/biosynthesis , Collagen Type V/biosynthesis , Extracellular Matrix Proteins/biosynthesis , Potassium Channels, Inwardly Rectifying/biosynthesis , Thyroid Carcinoma, Anaplastic/genetics , Versicans/biosynthesis , Cell Adhesion Molecules/genetics , Cell Communication , Cell Movement/genetics , Cell Proliferation/genetics , Collagen Type V/genetics , Extracellular Matrix Proteins/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Male , MicroRNAs/biosynthesis , MicroRNAs/genetics , Neovascularization, Pathologic/genetics , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Potassium Channels, Inwardly Rectifying/genetics , Prognosis , Thyroid Carcinoma, Anaplastic/pathology , Thyroid Nuclear Factor 1 , Transcription Factors/biosynthesis , Transcription Factors/genetics , Versicans/genetics
16.
Am J Physiol Renal Physiol ; 311(1): F12-5, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27122539

ABSTRACT

Kir4.1 is an inwardly rectifying potassium (K(+)) channel and is expressed in the brain, inner ear, and kidney. In the kidney, Kir4.1 is expressed in the basolateral membrane of the late thick ascending limb (TAL), the distal convoluted tubule (DCT), and the connecting tubule (CNT)/cortical collecting duct (CCD). It plays a role in K(+) recycling across the basolateral membrane in corresponding nephron segments and in generating negative membrane potential. The renal phenotypes of the loss-function mutations of Kir4.1 include mild salt wasting, hypomagnesemia, hypokalemia, and metabolic alkalosis, suggesting that the disruption of Kir4.1 mainly impairs the transport in the DCT. Patch-clamp experiments and immunostaining demonstrate that Kir4.1 plays a predominant role in determining the basolateral K(+) conductance in the DCT. However, the function of Kir4.1 in the TAL and CNT/CCD is not essential, because K(+) channels other than Kir4.1 are also expressed. The downregulation of Kir4.1 in the DCT reduced basolateral chloride (Cl(-)) conductance, suppressed the expression of ste20 proline-alanine-rich kinase (SPAK), and decreased Na-Cl cotransporter (NCC) expression and activity. This suggests that Kir4.1 regulates NCC expression by the modulation of the Cl(-)-sensitive with-no-lysine kinase-SPAK pathway.


Subject(s)
Kidney/metabolism , Potassium Channels, Inwardly Rectifying/genetics , Animals , Humans , Kidney Tubules, Collecting/metabolism , Mammals , Potassium Channels, Inwardly Rectifying/biosynthesis
17.
J Vis Exp ; (103)2015 Sep 26.
Article in English | MEDLINE | ID: mdl-26436772

ABSTRACT

DNA methylation serves to regulate gene expression through the covalent attachment of a methyl group onto the C5 position of a cytosine in a cytosine-guanine dinucleotide. While DNA methylation provides long-lasting and stable changes in gene expression, patterns and levels of DNA methylation are also subject to change based on a variety of signals and stimuli. As such, DNA methylation functions as a powerful and dynamic regulator of gene expression. The study of neuroepigenetics has revealed a variety of physiological and pathological states that are associated with both global and gene-specific changes in DNA methylation. Specifically, striking correlations between changes in gene expression and DNA methylation exist in neuropsychiatric and neurodegenerative disorders, during synaptic plasticity, and following CNS injury. However, as the field of neuroepigenetics continues to expand its understanding of the role of DNA methylation in CNS physiology, delineating causal relationships in regards to changes in gene expression and DNA methylation are essential. Moreover, in regards to the larger field of neuroscience, the presence of vast region and cell-specific differences requires techniques that address these variances when studying the transcriptome, proteome, and epigenome. Here we describe FACS sorting of cortical astrocytes that allows for subsequent examination of a both RNA transcription and DNA methylation. Furthermore, we detail a technique to examine DNA methylation, methylation sensitive high resolution melt analysis (MS-HRMA) as well as a luciferase promoter assay. Through the use of these combined techniques one is able to not only explore correlative changes between DNA methylation and gene expression, but also directly assess if changes in the DNA methylation status of a given gene region are sufficient to affect transcriptional activity.


Subject(s)
Astrocytes/physiology , DNA Methylation , Potassium Channels, Inwardly Rectifying/genetics , Animals , Astrocytes/metabolism , Cytosine/metabolism , DNA/genetics , DNA/metabolism , Dinucleoside Phosphates/genetics , Dinucleoside Phosphates/metabolism , Gene Expression , Potassium Channels, Inwardly Rectifying/biosynthesis , Promoter Regions, Genetic , Rats, Transgenic , Transcriptional Activation , Transcriptome
20.
Cardiovasc Drugs Ther ; 29(3): 209-18, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26095682

ABSTRACT

PURPOSE: Myocardial infarction (MI) results in an increased susceptibility to ventricular arrhythmias, due in part to decreased inward-rectifier K+ current (IK1), which is mediated primarily by the Kir2.1 protein. The use of renin-angiotensin-aldosterone system antagonists is associated with a reduced incidence of ventricular arrhythmias. Casein kinase 2 (CK2) binds and phosphorylates SP1, a transcription factor of KCNJ2 that encodes Kir2.1. Whether valsartan represses CK2 activation to ameliorate IK1 remodeling following MI remains unclear. METHODS: Wistar rats suffering from MI received either valsartan or saline for 7 days. The protein levels of CK2 and Kir2.1 were each detected via a Western blot analysis. The mRNA levels of CK2 and Kir2.1 were each examined via quantitative real-time PCR. RESULTS: CK2 expression was higher at the infarct border; and was accompanied by a depressed IK1/Kir2.1 protein level. Additionally, CK2 overexpression suppressed KCNJ2/Kir2.1 expression. By contrast, CK2 inhibition enhanced KCNJ2/Kir2.1 expression, establishing that CK2 regulates KCNJ2 expression. Among the rats suffering from MI, valsartan reduced CK2 expression and increased Kir2.1 expression compared with the rats that received saline treatment. In vitro, hypoxia increased CK2 expression and valsartan inhibited CK2 expression. The over-expression of CK2 in cells treated with valsartan abrogated its beneficial effect on KCNJ2/Kir2.1. CONCLUSIONS: AT1 receptor antagonist valsartan reduces CK2 activation, increases Kir2.1 expression and thereby ameliorates IK1 remodeling after MI in the rat model.


Subject(s)
Casein Kinase II/metabolism , Myocardial Infarction/metabolism , Potassium Channels, Inwardly Rectifying/biosynthesis , Up-Regulation/drug effects , Valsartan/pharmacology , Animals , Male , Membrane Potentials/drug effects , Myocardial Infarction/enzymology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Primary Cell Culture , Rats
SELECTION OF CITATIONS
SEARCH DETAIL