Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Shock ; 59(4): 612-620, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36640152

ABSTRACT

ABSTRACT: Increased epithelial permeability in sepsis is mediated via disruptions in tight junctions, which are closely associated with the perijunctional actin-myosin ring. Genetic deletion of myosin light chain kinase (MLCK) reverses sepsis-induced intestinal hyperpermeability and improves survival in a murine model of intra-abdominal sepsis. In an attempt to determine the generalizability of these findings, this study measured the impact of MLCK deletion on survival and potential associated mechanisms following pneumonia-induced sepsis. MLCK -/- and wild-type mice underwent intratracheal injection of Pseudomonas aeruginosa . Unexpectedly, survival was significantly worse in MLCK -/- mice than wild-type mice. This was associated with increased permeability to Evans blue dye in bronchoalveolar lavage fluid but not in tissue homogenate, suggesting increased alveolar epithelial leak. In addition, bacterial burden was increased in bronchoalveolar lavage fluid. Cytokine array using whole-lung homogenate demonstrated increases in multiple proinflammatory and anti-inflammatory cytokines in knockout mice. These local pulmonary changes were associated with systemic inflammation with increased serum levels of IL-6 and IL-10 and a marked increase in bacteremia in MLCK -/- mice. Increased numbers of both bulk and memory CD4 + T cells were identified in the spleens of knockout mice, with increased early and late activation. These results demonstrate that genetic deletion of MLCK unexpectedly increases mortality in pulmonary sepsis, associated with worsened alveolar epithelial leak and both local and systemic inflammation. This suggests that caution is required in targeting MLCK for therapeutic gain in sepsis.


Subject(s)
Lung , Myosin-Light-Chain Kinase , Pneumonia , Sepsis , Animals , Mice , Cytokines , Inflammation , Intestinal Mucosa , Lung/metabolism , Lung/pathology , Mice, Knockout , Myosin-Light-Chain Kinase/genetics , Permeability , Pneumonia/complications , Sepsis/pathology , Tight Junctions/physiology
2.
Methods Mol Biol ; 2367: 137-148, 2021.
Article in English | MEDLINE | ID: mdl-33460025

ABSTRACT

Lung fluid balance is maintained in part by the barriers formed by the pulmonary microvasculature and alveolar epithelium. Failure of either of these barriers leads to pulmonary edema, which limits lung function and exacerbates the severity of acute lung injury. Here we describe a method using Evans Blue dye to simultaneously measure the function of vascular and epithelial barriers of murine lungs in vivo.


Subject(s)
Lung , Acute Lung Injury , Animals , Capillary Permeability , Evans Blue , Mice , Permeability , Pulmonary Alveoli , Pulmonary Edema
3.
Sci Rep ; 10(1): 11017, 2020 07 03.
Article in English | MEDLINE | ID: mdl-32620847

ABSTRACT

The previous reports on an addiction vulnerability marker in the human SLC4A7 gene encoding the Na/HCO3 transporter NBCn1 suggest that this pH-regulating protein may affect alcohol-related behavior and response. Here, we examined alcohol consumption and sensitivity to the sedative effects of alcohol in male NBCn1 knockout mice. These mice displayed lower pH in neurons than wildtype controls, determined by intracellular pH in hippocampal neuronal cultures. Neurons from knockout mice had a higher action potential threshold and a more depolarized membrane potential, thus reducing membrane excitability. In a two-bottle free choice procedure, knockout mice consumed more alcohol than controls and consistently increased alcohol consumption after repeated alcohol deprivation periods. Quinine and sucrose preference was similar between genotypes. Knockout mice showed increased propensity for alcohol-induced conditioned place preference. In loss of righting reflex assessment, knockout mice revealed increased sensitivity to alcohol-induced sedation and developed tolerance to the sedation after repeated alcohol administrations. Furthermore, chronic alcohol consumption caused NBCn1 downregulation in the hippocampus and striatum of mice and humans. These results demonstrate an important role of NBCn1 in regulation of alcohol consumption and sensitivity to alcohol-induced sedation.


Subject(s)
Alcohol Drinking/genetics , Down-Regulation , Hippocampus/cytology , Sodium-Bicarbonate Symporters/genetics , Animals , Cells, Cultured , Gene Knockout Techniques , Hippocampus/chemistry , Humans , Hydrogen-Ion Concentration , Male , Mice , Neurons/chemistry , Neurons/cytology , Quinine/pharmacology , Sucrose/pharmacology
4.
Front Physiol ; 11: 209, 2020.
Article in English | MEDLINE | ID: mdl-32231591

ABSTRACT

Endothelin-1 (ET-1) is a peptide hormone that functions as a vasoconstrictor in the vasculature, whereas in the collecting duct of the kidney it exerts blood pressure-lowering effects via natriuretic actions. Aberrant ET-1 signaling is associated with several pathological states including hypertension and chronic kidney disease. ET-1 expression is regulated largely through transcriptional control of the gene that encodes ET-1, EDN1. Here we report a long, non-coding RNA (lncRNA) that appears to be antisense to the EDN1 gene, called EDN1-AS. Because EDN1-AS represents a potential novel mechanism to regulate ET-1 expression, we examined the regulation of EDN1-AS expression and action. A putative glucocorticoid receptor response (GR) element upstream of the predicted EDN1-AS transcription start site was identified using the ENCODE database and the UCSC genome browser. Two homozygous deletion clones of the element were generated using CRISPR/Cas9. This deletion resulted in a significant increase in the expression of EDN1-AS, which was associated with increased secretion of ET-1 peptide from HK-2 cells (two-fold increase in KO cells vs. CNTL, n = 7, P < 0.05). Phenotypic characterization of these CRISPR clones revealed a difference in cell growth rates. Using a standard growth assay, we determined that the KO1 clone exhibited a three-fold increase in growth over 8 days compared to control cells (n = 4, P < 0.01) and the KO2 clone exhibited a two-fold increase (n = 4, P < 0.01). These results support a role for EDN1-AS as a novel regulatory mechanism of ET-1 expression and cellular proliferation.

5.
Alcohol ; 80: 81-89, 2019 11.
Article in English | MEDLINE | ID: mdl-31278041

ABSTRACT

In the lung, chronic alcohol consumption is a risk factor for acute respiratory distress syndrome (ARDS), a disorder that can be fatal due to airspace flooding. The severity of pulmonary edema is controlled by multiple barriers, and in particular the alveolar epithelial barrier and pulmonary microvasculature. However, to date, the effects of chronic alcohol ingestion on both of these barriers in the lung has not been directly and simultaneously measured. In addition the effects of alcohol on systemic, indirect lung injury versus direct injury have not been compared. In this study, we used tissue morphometry and Evans Blue permeability assays to assess the effects of alcohol and endotoxemia injury on pulmonary barrier function comparing intraperitoneal (IP) administration of lipopolysaccharide (LPS) to intratracheal (IT) administration. Consistent with previous reports, we found that in alcohol-fed mice, the alveolar barrier was impaired, allowing Evans Blue to permeate into the airspaces. Moreover, IT administered LPS caused a significant breach of both the alveolar epithelial and vascular barriers in alcohol-fed mice, whereas the endothelial barrier was less affected in response to IP administered LPS. The alveolar barrier of control mice remained intact for both IP and IT administered LPS. However, both injuries caused significant interstitial edema, independently of whether the mice were fed alcohol or not. These data suggest that in order to properly target pulmonary edema due to alcoholic lung syndrome, both the alveolar and endothelial barriers need to be considered as well as the nature of the "second hit" that initiates ARDS.


Subject(s)
Alcoholism/complications , Lung Diseases/chemically induced , Animals , Disease Models, Animal , Endotoxins/pharmacology , Ethanol/adverse effects , Lipopolysaccharides/pharmacology , Lung/drug effects , Lung/pathology , Lung Diseases/pathology , Lung Injury/chemically induced , Lung Injury/pathology , Male , Mice , Mice, Inbred C57BL , Peroxidase/metabolism , Syndrome
6.
Life Sci ; 118(2): 195-9, 2014 Nov 24.
Article in English | MEDLINE | ID: mdl-24632479

ABSTRACT

AIMS: Recently, microRNAs (miRNAs) have been implicated in control of Edn1 mRNA in several tissues. Here we examined the role of miRNA action on Edn1 mRNA expression in renal distal collecting duct cells. MAIN METHODS: A microarray study was conducted to provide a comprehensive assessment of miRNAs present in a murine inner medullary collecting duct (mIMCD-3) cell line. The experiment was designed as a comparison between mIMCD-3 cells grown in the presence and absence of aldosterone. Argonaute (Ago) immunoprecipitation experiments were used to investigate binding of the RNA induced silencing complex (RISC) to Edn1 mRNA. KEY FINDINGS: Thirty-four miRNAs were detected in very high abundance in mIMCD-3 cells, and a large number of others were present at lower levels. The microarray experiments were validated by quantitative PCR analysis of selected miRNAs. The microarray data, in combination with in silico examination of the Edn1 3' UTR provided a panel of candidate miRNAs that could act upon the Edn1 expression. Edn1 mRNA was co-immunoprecipitated with an Argonaute protein antibody, and this interaction was blocked by anti-miR-709 oligonucleotides. SIGNIFICANCE: These results define the miRNA landscape of the mIMCD-3 cell line. Moreover, Edn1 was shown to interact with Argonaute protein suggesting that it is a target of the RNA induced silencing complex (RISC).


Subject(s)
Endothelin-1/genetics , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/metabolism , MicroRNAs/metabolism , 3' Untranslated Regions/genetics , Aldosterone/pharmacology , Animals , Argonaute Proteins/metabolism , Binding Sites , Cell Line , Endothelin-1/metabolism , Gene Expression Regulation/drug effects , Immunoprecipitation , Kidney Tubules, Collecting/drug effects , Mice , MicroRNAs/genetics , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
7.
Am J Physiol Renal Physiol ; 305(12): F1697-704, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24154698

ABSTRACT

The circadian clock plays an important role in the regulation of physiological processes, including renal function and blood pressure. We have previously shown that the circadian protein period (Per)1 regulates the expression of multiple Na(+) transport genes in the collecting duct, including the α-subunit of the renal epithelial Na(+) channel. Consistent with this finding, Per1 knockout mice exhibit dramatically lower blood pressure than wild-type mice. We have also recently demonstrated the potential opposing actions of cryptochrome (Cry)2 on Per1 target genes. Recent work by others has demonstrated that Cry1/2 regulates aldosterone production through increased expression of the adrenal gland-specific rate-limiting enzyme 3ß-dehydrogenase isomerase (3ß-HSD). Therefore, we tested the hypothesis that Per1 plays a role in the regulation of aldosterone levels and renal Na(+) retention. Using RNA silencing and pharmacological blockade of Per1 nuclear entry in the NCI-H295R human adrenal cell line, we showed that Per1 regulates 3ß-HSD expression in vitro. These results were confirmed in vivo: mice with reduced levels of Per1 had decreased levels of plasma aldosterone and decreased mRNA expression of 3ß-HSD. We postulated that mice with reduced Per1 would have a renal Na(+)-retaining defect. Indeed, metabolic cage experiments demonstrated that Per1 heterozygotes excreted more urinary Na(+) compared with wild-type mice. Taken together, these data support the hypothesis that Per1 regulates aldosterone levels and that Per1 plays an integral role in the regulation of Na(+) retention.


Subject(s)
Aldosterone/metabolism , Kidney/metabolism , Period Circadian Proteins/metabolism , Sodium/metabolism , 3-Hydroxysteroid Dehydrogenases/metabolism , Adrenal Glands/cytology , Adrenal Glands/metabolism , Animals , Cell Line , Cells, Cultured , Cryptochromes/metabolism , Humans , In Vitro Techniques , Male , Mice , Mice, Knockout , Models, Animal , Period Circadian Proteins/deficiency , Period Circadian Proteins/drug effects , Period Circadian Proteins/genetics , RNA, Small Interfering/pharmacology
8.
Front Physiol ; 4: 253, 2013.
Article in English | MEDLINE | ID: mdl-24062694

ABSTRACT

Renal function and blood pressure (BP) exhibit a circadian pattern of variation, but the molecular mechanism underlying this circadian regulation is not fully understood. We have previously shown that the circadian clock protein Per1 positively regulates the basal and aldosterone-mediated expression of the alpha subunit of the renal epithelial sodium channel (αENaC). The mechanism of this regulation has not been determined however. To further elucidate the mechanism of mineralocorticoid receptor (MR) and Per1 action, site-directed mutagenesis, DNA pull-down assays and chromatin immunoprecipitation (ChIP) methods were used to investigate the coordinate regulation of αENaC by Per1 and MR. Mutation of two circadian response E-boxes in the human αENaC promoter abolished both basal and aldosterone-mediated promoter activity. DNA pull down assays demonstrated the interaction of both MR and Per1 with the E-boxes from the αENaC promoter. These observations were corroborated by ChIP experiments showing increased occupancy of MR and Per1 on an E-box of the αENaC promoter in the presence of aldosterone. This is the first report of an aldosterone-mediated increase in Per1 on a target gene promoter. Taken together, these results demonstrate the novel finding that Per1 and MR mediate the aldosterone response of αENaC through DNA/protein interaction in renal collecting duct cells.

9.
Am J Physiol Regul Integr Comp Physiol ; 305(7): R735-47, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23824961

ABSTRACT

Mounting evidence suggests that the circadian clock plays an integral role in the regulation of many physiological processes including blood pressure, renal function, and metabolism. The canonical molecular clock functions via activation of circadian target genes by Clock/Bmal1 and repression of Clock/Bmal1 activity by Per1-3 and Cry1/2. However, we have previously shown that Per1 activates genes important for renal sodium reabsorption, which contradicts the canonical role of Per1 as a repressor. Moreover, Per1 knockout (KO) mice exhibit a lowered blood pressure and heavier body weight phenotype similar to Clock KO mice, and opposite that of Cry1/2 KO mice. Recent work has highlighted the potential role of Per1 in repression of Cry2. Therefore, we postulated that Per1 potentially activates target genes through a Cry2-Clock/Bmal1-dependent mechanism, in which Per1 antagonizes Cry2, preventing its repression of Clock/Bmal1. This hypothesis was tested in vitro and in vivo. The Per1 target genes αENaC and Fxyd5 were identified as Clock targets in mpkCCDc14 cells, a model of the renal cortical collecting duct. We identified PPARα and DEC1 as novel Per1 targets in the mouse hepatocyte cell line, AML12, and in the liver in vivo. Per1 knockdown resulted in upregulation of Cry2 in vitro, and this result was confirmed in vivo in mice with reduced expression of Per1. Importantly, siRNA-mediated knockdown of Cry2 and Per1 demonstrated opposing actions for Cry2 and Per1 on Per1 target genes, supporting the potential Cry2-Clock/Bmal1-dependent mechanism underlying Per1 action in the liver and kidney.


Subject(s)
Cryptochromes/metabolism , Kidney/metabolism , Liver/metabolism , Period Circadian Proteins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line , Cryptochromes/deficiency , Cryptochromes/genetics , Epithelial Sodium Channels/genetics , Epithelial Sodium Channels/metabolism , Gene Expression Regulation , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Ion Channels , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, 129 Strain , Mice, Knockout , Microfilament Proteins , PPAR alpha/genetics , PPAR alpha/metabolism , Period Circadian Proteins/deficiency , Period Circadian Proteins/genetics , RNA Interference , RNA, Messenger/metabolism , Transfection
10.
Am J Physiol Renal Physiol ; 303(7): F918-27, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22832921

ABSTRACT

Increasing evidence suggests that the circadian clock plays an important role in the control of renal function and blood pressure. We previously showed that the circadian clock protein Period (Per)1, positively regulates the expression of the rate limiting subunit of the renal epithelial sodium channel (αENaC), which contributes to blood pressure regulation. Casein kinases 1δ and 1ε (CK1δ/ε) are critical regulators of clock proteins. CK1δ/ε must phosphorylate the circadian clock protein Per1 in order for the latter to enter the nucleus. We used a commercially available CK1δ/ε inhibitor, PF670462, to test the effect of CK1δ/ε blockade and inhibited Per1 nuclear entry on αENaC in a model of the renal cortical collecting duct (mpkCCD(c14) cells). CK1δ/ε blockade prevented Per1 and Clock from interacting with an E-box from the αENaC promoter. CK1δ/ε inhibition reduced αENaC mRNA levels by <60%. A similar decrease in αENaC mRNA was observed following siRNA-mediated CK1δ/ε knock-down. Inhibition of CK1δ/ε effectively prevented the transcriptional response of αENaC to aldosterone, suggesting an interaction between the circadian clock and aldosterone-mediated regulation of αENaC. CK1δ/ε inhibition significantly reduced αENaC but increased Caveolin-1 membrane protein levels; transepithelial current, a measure of ENaC activity, was decreased. Importantly, single channel analysis in amphibian renal cells demonstrated a dramatic decrease in the number of patches with observable ENaC current following CK1δ/ε inhibition. The present study shows for the first time that CK1δ/ε inhibition and impaired Per1 nuclear entry results in decreased αENaC expression and ENaC activity, providing further support for direct control of ENaC by the circadian clock.


Subject(s)
Casein Kinase 1 epsilon/antagonists & inhibitors , Casein Kinase Idelta/antagonists & inhibitors , Epithelial Sodium Channels/metabolism , Kidney Tubules, Collecting/metabolism , Animals , CLOCK Proteins/metabolism , Casein Kinase 1 epsilon/metabolism , Casein Kinase Idelta/metabolism , Cell Line , Cells, Cultured , Epithelial Sodium Channels/genetics , Kidney Tubules, Collecting/drug effects , Mice , Period Circadian Proteins/metabolism , Phosphorylation , Pyrimidines/pharmacology
11.
Hypertension ; 59(6): 1151-6, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22526258

ABSTRACT

The circadian clock protein period 1 (Per1) contributes to the regulation of expression of the α subunit of the renal epithelial sodium channel at the basal level and in response to the mineralocorticoid hormone aldosterone. The goals of the present study were to define the role of Per1 in the regulation of additional renal sodium handling genes in cortical collecting duct cells and to evaluate blood pressure (BP) in mice lacking functional Per1. To determine whether Per1 regulates additional genes important in renal sodium handling, a candidate gene approach was used. Immortalized collecting duct cells were transfected with a nontarget small interfering RNA or a Per1-specific small interfering RNA. Expression of the genes for α-epithelial sodium channel and Fxyd5, a positive regulator of Na, K-ATPase activity, decreased in response to Per1 knockdown. Conversely, mRNA expression of caveolin 1, Ube2e3, and ET-1, all negative effectors of epithelial sodium channel, was induced after Per1 knockdown. These results led us to evaluate BP in Per1 KO mice. Mice lacking Per1 exhibit significantly reduced BP and elevated renal ET-1 levels compared with wild-type animals. Given the established role of renal ET-1 in epithelial sodium channel inhibition and BP control, elevated renal ET-1 is one possible explanation for the lower BP observed in Per1 KO mice. These data support a role for the circadian clock protein Per1 in the coordinate regulation of genes involved in renal sodium reabsorption. Importantly, the lower BP observed in Per1 KO mice compared with wild-type mice suggests a role for Per1 in BP control as well.


Subject(s)
Blood Pressure/physiology , Kidney Tubules, Collecting/metabolism , Period Circadian Proteins/metabolism , Sodium/metabolism , Animals , Blood Pressure/genetics , Blotting, Western , Caveolin 1/genetics , Caveolin 1/metabolism , Cell Line , Cell Line, Transformed , Endothelin-1/genetics , Endothelin-1/metabolism , Epithelial Sodium Channels/genetics , Epithelial Sodium Channels/metabolism , Gene Expression Regulation , Ion Transport/genetics , Male , Mice , Mice, 129 Strain , Mice, Knockout , Models, Genetic , Period Circadian Proteins/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...