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










Publication year range
1.
Acta Haematol ; 144(6): 633-640, 2021.
Article in English | MEDLINE | ID: mdl-34237720

ABSTRACT

INTRODUCTION: Viscoelastic coagulation tests are useful to assess coagulation status in the clinical setting and to aid in understanding underlying pathophysiological mechanisms that affect coagulation status. Such tests also are useful for coagulation research. Because mouse models are widely used to study molecular mechanisms in fine detail, a simple viscoelastic coagulation test requiring small blood volumes would be convenient for such studies in mice. METHODS: We tested viscoelastic coagulation properties of normal healthy adult mice using a novel veterinary clinical point-of-care device, Viscoelastic Coagulation Monitor (VCM Vet™; Entegrion Corp.). Fresh whole blood was collected from 63 healthy mature adult C57 black 6N mice, with ultimately 54 mice, equal numbers of male and females, used to determine reference intervals (RIs) for VCM test parameters. RESULTS: RIs were determined for equal numbers of male and female mice: clot time: 43.0-353.0 s; clot formation time: 49.4-137.6 s; alpha angle: 54.4-62.2°; A10: 25.0-49.6 VCM units; A20: 31.0-56.5 VCM units; maximum clot firmness: 37.6-62.8 VCM units; Lysis Index 30 (Li30): 99.8-100.0%; and Li45: 99.7-100.0%. Significant differences were found between male and female subgroups, where females had higher mean A10 and A20 and median MCF values, indicating greater clot firmness in female versus male mice. CONCLUSION: VCM Vet is a feasible viscoelastic coagulation test device for studies with mature adult mice, including studying inherent sex differences in coagulation parameters. Inherent differences in coagulability of male and female mice warrant further investigation to determine if such differences underlie greater coagulopathic, hemorrhagic, or thromboembolic risk during trauma or other pathophysiologic conditions.


Subject(s)
Blood Coagulation Tests/standards , Animals , Blood Coagulation , Blood Coagulation Tests/methods , Blood Coagulation Tests/veterinary , Female , Kinetics , Male , Mice , Mice, Inbred C57BL , Reference Values , Sex Characteristics
2.
J Trauma Acute Care Surg ; 86(4): 583-591, 2019 04.
Article in English | MEDLINE | ID: mdl-30562326

ABSTRACT

BACKGROUND: Thromboembolic events within the pulmonary arterial vasculature are a troublesome complication of severe blunt thoracic trauma. Mechanisms underlying these events are currently in question as pulmonary thromboembolic events in this particular trauma population tend to be diagnosed more rapidly, more frequently and without an associated systemic thrombosis. This study investigates the role of P-selectin in thrombus formation through the use of in vivo blocking antibodies. We hypothesize that P-selectin plays a pivotal role in de novo pulmonary arterial thrombosis following blunt thoracic trauma. METHODS: A murine weight-drop model of lateral blunt thoracic trauma was used. Wild-type mice in the experimental group were given blocking antibodies against P-selectin prior to the trauma. All mice were euthanized at 24 hours for evaluation with hematoxylin-eosin staining or immunofluorescent staining for fibrin and P-selectin. RESULTS: Injured mice that did not receive the P-selectin antibody showed a robust fourfold to fivefold increase in fibrin accumulation in both coup and contrecoup tissues (fluorescence per um of arterial wall) compared to uninjured sham mice. In contrast, mice pretreated with P-selectin blocking antibody showed no significant increase in fibrin accumulation on either side of the lungs after blunt thoracic trauma. No difference in mean fibrin deposition was found between sham controls that received the P-selectin-blocking antibody and those that received an isotype control antibody. CONCLUSION: P-selectin expression increases at the pulmonary arterial luminal surface following blunt thoracic trauma. In addition, P-selectin-blocking in vivo prevents pulmonary arterial fibrin accumulation after blunt thoracic trauma, confirming that P-selectin is necessary for de novo pulmonary arterial thrombosis after traumatic injury.


Subject(s)
P-Selectin/physiology , Pulmonary Embolism/physiopathology , Wounds, Nonpenetrating/physiopathology , Animals , Antibodies, Blocking , Disease Models, Animal , Mice , P-Selectin/immunology
3.
Shock ; 50(6): 696-705, 2018 12.
Article in English | MEDLINE | ID: mdl-29394242

ABSTRACT

Pulmonary thromboembolic events cause significant morbidity and mortality after severe trauma. Clinically, these lesions are believed to be emboli arising secondary to deep venous thrombosis (DVT) in the lower extremities. Recently, this notion has been challenged by clinical studies, showing that pulmonary clots arise after trauma in the absence of DVT. This suggests that pulmonary blood clots arise in situ via de novo thrombosis. In the present study, we characterize a murine weight-drop model of lateral blunt thoracic trauma. Our model demonstrates severe unilateral lung contusion injury with low (10%) mortality in the absence of extrapulmonary injury, after impact with a 50-g weight dropped from 45 cm height (657 J/m). At 24 h after injury, immunofluorescence and histological evidence revealed early pulmonary arterial thrombosis in the form of eccentric accumulation of fibrin and CD41 positive eosinophilic proteinaceous material, on both coup and contrecoup lung lobes of injured mice, indicating early thrombotic events both within and outside of the area of primary lung injury. Our model is ideal in that lateral impact enables greater impact energy to be applied to achieve significant lung contusion without significant mortality or extrapulmonary injury, and the model has additional translational value in creating thrombosis analogous to pulmonary embolism observed clinically after blunt thoracic trauma. To our knowledge, this is the first demonstration of de novo pulmonary thrombosis in a clinically translational model of blunt thoracic trauma, and supports challenges to current assumptions about the origin of pulmonary blood clots in the wake of severe traumatic injury.


Subject(s)
Thoracic Injuries/metabolism , Venous Thrombosis/metabolism , Animals , Bronchoalveolar Lavage , Disease Models, Animal , Fibrin/metabolism , Fluorescent Antibody Technique , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C57BL , Platelet Membrane Glycoprotein IIb/metabolism , Pulmonary Embolism/metabolism
4.
Sci Rep ; 7(1): 14058, 2017 10 25.
Article in English | MEDLINE | ID: mdl-29070899

ABSTRACT

Large-conductance Ca2+-activated potassium (BKCa) channels are key determinants of vascular smooth muscle excitability. Impaired BKCa channel function through remodeling of BKCa ß1 expression and function contributes to vascular complications in animal models of diabetes. Yet, whether similar alterations occur in native vascular smooth muscle from humans with type 2 diabetes is unclear. In this study, we evaluated BKCa function in vascular smooth muscle from small resistance adipose arteries of non-diabetic and clinically diagnosed type 2 diabetic patients. We found that BKCa channel activity opposes pressure-induced constriction in human small resistance adipose arteries, and this is compromised in arteries from diabetic patients. Consistent with impairment of BKCa channel function, the amplitude and frequency of spontaneous BKCa currents, but not Ca2+ sparks were lower in cells from diabetic patients. BKCa channels in diabetic cells exhibited reduced Ca2+ sensitivity, single-channel open probability and tamoxifen sensitivity. These effects were associated with decreased functional coupling between BKCa α and ß1 subunits, but no change in total protein abundance. Overall, results suggest impairment in BKCa channel function in vascular smooth muscle from diabetic patients through unique mechanisms, which may contribute to vascular complications in humans with type 2 diabetes.


Subject(s)
Arteries/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Action Potentials , Arteries/physiopathology , Case-Control Studies , Cells, Cultured , Humans , Membrane Potentials
5.
Heart Rhythm ; 14(5): 727-736, 2017 05.
Article in English | MEDLINE | ID: mdl-28111350

ABSTRACT

BACKGROUND: Interleukin 1ß (IL-1ß) is a key regulator of the inflammatory response after myocardial infarction (MI) by modulating immune cell recruitment, cytokine production, and extracellular matrix turnover. Elevated levels of IL-1ß are associated with adverse remodeling, and inhibition of IL-1 signaling after MI results in improved contractile function. OBJECTIVE: The goal of this study was to determine whether IL-1 signaling also contributes to post-MI arrhythmogenesis. METHODS: MI was created in 2 murine models of elevated inflammation: atherosclerotic on the Western diet or wild-type with a subseptic dose of lipopolysaccharide. The role of IL-1ß was assessed with the IL-1 receptor antagonist anakinra (10 mg/(kg·d), starting 24 hours post-MI). RESULTS: In vivo and ex vivo molecular imaging showed reduced myocardial inflammation after a 4-day course of anakinra treatment, despite no change in infarct size. At day 5 post-MI, high-speed optical mapping of transmembrane potential and intracellular Ca2+ in isolated hearts revealed that IL-1ß inhibition improved conduction velocity, reduced action potential duration dispersion, improved intracellular Ca2+ handling, decreased transmembrane potential and Ca2+ alternans magnitude, and reduced spontaneous and inducible ventricular arrhythmias. These functional improvements were linked to increased expression of connexin 43 and sarcoplasmic reticulum Ca2+-ATPase. CONCLUSION: This study revealed a novel mechanism for IL-1ß in contributing to defective excitation-contraction coupling and arrhythmogenesis in the post-MI heart. Our results suggest that inhibition of IL-1 signaling post-MI may represent a novel antiarrhythmic therapy.


Subject(s)
Anti-Arrhythmia Agents/therapeutic use , Arrhythmias, Cardiac/physiopathology , Excitation Contraction Coupling/drug effects , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Interleukin-1beta/antagonists & inhibitors , Myocardial Infarction/complications , Animals , Anti-Arrhythmia Agents/pharmacology , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/prevention & control , Disease Models, Animal , Excitation Contraction Coupling/physiology , Interleukin 1 Receptor Antagonist Protein/pharmacology , Mice , Myocardial Infarction/physiopathology
6.
J Proteome Res ; 14(5): 2298-311, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25815641

ABSTRACT

The cardiac voltage-gated sodium channel, Na(V)1.5, drives the upstroke of the cardiac action potential and is a critical determinant of myocyte excitability. Recently, calcium (Ca(2+))/calmodulin(CaM)-dependent protein kinase II (CaMKII) has emerged as a critical regulator of Na(V)1.5 function through phosphorylation of multiple residues including S516, T594, and S571, and these phosphorylation events may be important for the genesis of acquired arrhythmias, which occur in heart failure. However, phosphorylation of full-length human Na(V)1.5 has not been systematically analyzed and Na(V)1.5 phosphorylation in human heart failure is incompletely understood. In the present study, we used label-free mass spectrometry to assess phosphorylation of human Na(V)1.5 purified from HEK293 cells with full coverage of phosphorylatable sites and identified 23 sites that were phosphorylated by CaMKII in vitro. We confirmed phosphorylation of S516 and S571 by LC-MS/MS and found a decrease in S516 phosphorylation in human heart failure, using a novel phospho-specific antibody. This work furthers our understanding of the phosphorylation of Na(V)1.5 by CaMKII under normal and disease conditions, provides novel CaMKII target sites for functional validation, and provides the first phospho-proteomic map of full-length human Na(V)1.5.


Subject(s)
Arrhythmias, Cardiac/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Heart Failure/metabolism , Heart Ventricles/metabolism , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Action Potentials , Amino Acid Sequence , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/pathology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Gene Expression Regulation , HEK293 Cells , Heart Failure/genetics , Heart Failure/pathology , Heart Ventricles/pathology , Humans , Mass Spectrometry/methods , Molecular Sequence Data , Myocardium/metabolism , Myocardium/pathology , NAV1.5 Voltage-Gated Sodium Channel/genetics , Phosphorylation , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine/metabolism , Signal Transduction
7.
Circ Res ; 114(9): 1398-409, 2014 04 25.
Article in English | MEDLINE | ID: mdl-24643961

ABSTRACT

RATIONALE: Both ß-adrenergic receptor (ß-AR) and Gq-coupled receptor (GqR) agonist-driven signaling play key roles in the events, leading up to and during cardiac dysfunction. How these stimuli interact at the level of protein kinase D (PKD), a nodal point in cardiac hypertrophic signaling, remains unclear. OBJECTIVE: To assess the spatiotemporal dynamics of PKD activation in response to ß-AR signaling alone and on coactivation with GqR-agonists. This will test our hypothesis that compartmentalized PKD signaling reconciles disparate findings of PKA facilitation and inhibition of PKD activation. METHODS AND RESULTS: We report on the spatial and temporal profiles of PKD activation using green fluorescent protein-tagged PKD (wildtype or mutant S427E) and targeted fluorescence resonance energy transfer-based biosensors (D-kinase activity reporters) in adult cardiomyocytes. We find that ß-AR/PKA signaling drives local nuclear activation of PKD, without preceding sarcolemmal translocation. We also discover pronounced interference of ß-AR/cAMP/PKA signaling on GqR-induced translocation and activation of PKD throughout the cardiomyocyte. We attribute these effects to direct, PKA-dependent phosphorylation of PKD-S427. We also show that phosphomimetic substitution of S427 likewise impedes GqR-induced PKD translocation and activation. In neonatal myocytes, S427E inhibits GqR-evoked cell growth and expression of hypertrophic markers. Finally, we show altered S427 phosphorylation in transverse aortic constriction-induced hypertrophy. CONCLUSIONS: ß-AR signaling triggers local nuclear signaling and inhibits GqR-mediated PKD activation by preventing its intracellular translocation. PKA-dependent phosphorylation of PKD-S427 fine-tunes the PKD responsiveness to GqR-agonists, serving as a key integration point for ß-adrenergic and Gq-coupled stimuli.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Myocytes, Cardiac/enzymology , Protein Kinase C/metabolism , Receptors, Adrenergic, beta/metabolism , Signal Transduction , Adrenergic beta-Agonists/pharmacology , Animals , Cardiomegaly/enzymology , Cardiomegaly/pathology , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Enzyme Activation , Fluorescence Resonance Energy Transfer , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mutation , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Phosphorylation , Protein Kinase C/genetics , Protein Transport , Rabbits , Rats , Receptors, Adrenergic, beta/drug effects , Recombinant Fusion Proteins/metabolism , Signal Transduction/drug effects , Time Factors , Transfection
8.
Med Res Rev ; 33(5): 911-33, 2013 Sep.
Article in English | MEDLINE | ID: mdl-22886693

ABSTRACT

Microvascular barrier dysfunction is a serious problem that occurs in many inflammatory conditions, including sepsis, trauma, ischemia-reperfusion injury, cardiovascular disease, and diabetes. Barrier dysfunction permits extravasation of serum components into the surrounding tissue, leading to edema formation and organ failure. The basis for microvascular barrier dysfunction is hyperpermeability at endothelial cell-cell junctions. Endothelial hyperpermeability is increased by actomyosin contractile activity in response to phosphorylation of myosin light chain by myosin light chain kinase (MLCK). MLCK-dependent endothelial hyperpermeability occurs in response to inflammatory mediators (e.g., activated neutrophils, thrombin, histamine, tumor necrosis factor alpha, etc.), through multiple cell signaling pathways and signaling molecules (e.g., Ca(++) , protein kinase C, Src kinase, nitric oxide synthase, etc.). Other signaling molecules protect against MLCK-dependent hyperpermeability (e.g., sphingosine-1-phosphate or cAMP). In addition, individual MLCK isoforms play specific roles in endothelial barrier dysfunction, suggesting that isoform-specific inhibitors could be useful for treating inflammatory disorders and preventing multiple organ failure. Because endothelial barrier dysfunction depends upon signaling through MLCK in many instances, MLCK-dependent signaling comprises multiple potential therapeutic targets for preventing edema formation and multiple organ failure. The following review is a discussion of MLCK-dependent mechanisms and cell signaling events that mediate endothelial hyperpermeability.


Subject(s)
Endothelium/enzymology , Myosin-Light-Chain Kinase/metabolism , Signal Transduction , Animals , Endothelium/drug effects , Endothelium/physiopathology , Humans , Molecular Targeted Therapy , Myosin-Light-Chain Kinase/chemistry , Permeability/drug effects , Protective Agents/pharmacology , Signal Transduction/drug effects
9.
Am J Physiol Lung Cell Mol Physiol ; 304(3): L135-42, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23161886

ABSTRACT

ADAM15 is a disintegrin and metalloprotease recently implicated in cancer and chronic immune disorders. We have recently characterized ADAM15 as a mediator of endothelial barrier dysfunction. Whether this molecule contributes to acute inflammation has not been evaluated. The purpose of this study was to investigate the role of ADAM15 in mediating pulmonary microvascular leakage during acute inflammatory injury. Immunofluorescent staining and Western blotting revealed that the endothelium was the main source of ADAM15 in lung tissue. In a mouse model of acute lung injury induced by lipopolysaccharide (LPS), upregulation of ADAM15 was observed in association with pulmonary edema and neutrophil infiltration. The LPS-induced inflammatory injury, as demonstrated by bronchoalveolar lavage neutrophil count, lung wet-to-dry weight ratio, and myeloperoxidase activity, was significantly attenuated in Adam15(-/-) mice. Studies with primary cell culture demonstrated abundant ADAM15 expression in endothelial cells (ECs) of mouse lung but not in neutrophils. Deficiency of ADAM15 in ECs had no obvious effect on basal permeability but significantly attenuated hyperpermeability response to LPS as evidenced by albumin flux assay and measurements of transendothelial electrical resistance, respectively. ADAM15 deficiency also reduced neutrophil chemotactic transmigration across endothelial barriers in the presence or absence of formyl-methionyl-leucyl-phenylalanine (fMLP). Rescue expression of ADAM15 in Adam15(-/-) ECs restored neutrophil transendothelial migration. These data indicate that ADAM15 upregulation contributes to inflammatory lung injury by promoting endothelial hyperpermeability and neutrophil transmigration.


Subject(s)
ADAM Proteins/genetics , Acute Lung Injury/metabolism , Endothelial Cells/metabolism , Lung/metabolism , Membrane Proteins/genetics , Neutrophils/metabolism , Pulmonary Edema/metabolism , ADAM Proteins/deficiency , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Animals , Bronchoalveolar Lavage Fluid/cytology , Electric Impedance , Endothelial Cells/pathology , Lipopolysaccharides/pharmacology , Lung/pathology , Membrane Proteins/deficiency , Mice , Mice, Knockout , Neutrophil Infiltration , Neutrophils/pathology , Permeability , Peroxidase/genetics , Peroxidase/metabolism , Primary Cell Culture , Pulmonary Edema/chemically induced , Pulmonary Edema/genetics , Pulmonary Edema/pathology , Transendothelial and Transepithelial Migration , Up-Regulation
10.
Am J Physiol Cell Physiol ; 302(10): C1513-22, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22403784

ABSTRACT

Blood-brain barrier dysfunction is a serious consequence of inflammatory brain diseases, cerebral infections, and trauma. The proinflammatory cytokine interleukin (IL)-1ß is central to neuroinflammation and contributes to brain microvascular leakage and edema formation. Although it is well known that IL-1ß exposure directly induces hyperpermeability in brain microvascular endothelium, the molecular mechanisms mediating this response are not completely understood. In the present study, we found that exposure of the human brain microvascular endothelium to IL-1ß triggered activation of novel PKC isoforms δ, µ, and θ, followed by decreased transendothelial electrical resistance (TER). The IL-1ß-induced decrease in TER was prevented by small hairpin RNA silencing of PKC-θ or by treatment with the isoform-selective PKC inhibitor Gö6976 but not by PKC inhibitors that are selective for all PKC isoforms other than PKC-θ. Decreased TER coincided with increased phosphorylation of regulatory myosin light chain and with increased proapoptotic signaling indicated by decreased uptake of mitotracker red in response to IL-1ß treatment. However, neither of these observed effects were prevented by Gö6976 treatment, indicating lack of causality with respect to decreased TER. Instead, our data indicated that the mechanism of decreased TER involves PKC-θ-dependent phosphorylation of the tight junction protein zona occludens (ZO)-1. Because IL-1ß is a central inflammatory mediator, our interpretation is that inhibition of PKC-θ or inhibition of ZO-1 phosphorylation could be viable strategies for preventing blood-brain barrier dysfunction under a variety of neuroinflammatory conditions.


Subject(s)
Brain/blood supply , Brain/enzymology , Endothelium, Vascular/enzymology , Interleukin-1beta/physiology , Isoenzymes/antagonists & inhibitors , Microvessels/enzymology , Protein Kinase C/antagonists & inhibitors , Signal Transduction/physiology , Brain/pathology , Carbazoles/pharmacology , Cell Membrane Permeability/drug effects , Cell Membrane Permeability/genetics , Cells, Cultured , Endothelium, Vascular/pathology , Humans , Inflammation Mediators/physiology , Isoenzymes/genetics , Isoenzymes/physiology , Microvessels/pathology , Protein Kinase C/genetics , Protein Kinase C/physiology , Protein Kinase C-theta , RNA, Catalytic/antagonists & inhibitors , RNA, Catalytic/physiology , Signal Transduction/drug effects , Signal Transduction/genetics
11.
Microvasc Res ; 83(1): 82-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21864543

ABSTRACT

Neutrophil activation is an essential component of innate immune defense against infection and injury. In response to inflammatory stimulation, circulating neutrophils undergo a series of dynamic and metabolic changes characterized by ß2-intergrin mediated adhesion to microvascular endothelium and subsequent transendothelial migration. During this process, neutrophils release granular contents containing digestive enzymes and produce cytotoxic agents such as reactive oxygen species and cytokines. These products target endothelial barriers inducing phosphorylation-triggered junction dissociation, actin stress fiber formation, and actomyosin contraction, manifest as paracellular hyperpermeability. Endothelial cell-matrix focal adhesions play an integral role in this process by providing structural support for endothelial conformational changes that facilitate neutrophil transmigration, as well as by recruiting intracellular molecules that constitute the hyperpermeability signaling cascades. As a central connector of the complex signaling network, focal adhesion kinase (FAK) is activated following neutrophil adhesion, and further mediates the reorganization of endothelial integrin-matrix attachments in a pattern coordinating with cytoskeleton contraction and junction opening. In this review, we present recent experimental evidence supporting the importance of FAK in neutrophil-dependent regulation of endothelial permeability. The discussion focuses on the mechanisms by which neutrophils activate FAK and its downstream effects on endothelial barriers.


Subject(s)
Capillary Permeability , Endothelial Cells/enzymology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/enzymology , Neutrophil Activation , Neutrophils/enzymology , Transendothelial and Transepithelial Migration , Animals , Cell Adhesion , Endothelial Cells/immunology , Enzyme Activation , Focal Adhesions/immunology , Humans , Neutrophils/immunology , Signal Transduction
12.
J Biol Chem ; 286(1): 634-48, 2011 Jan 07.
Article in English | MEDLINE | ID: mdl-20974853

ABSTRACT

We previously presented evidence that transmembrane domain (TM) IV and TM X-XI are important for inhibitor binding and ion transport by the human Na(+)/H(+) exchanger, hNHE1 (Pedersen, S. F., King, S. A., Nygaard, E. B., Rigor, R. R., and Cala, P. M. (2007) J. Biol. Chem. 282, 19716-19727). Here, we present a structural model of the transmembrane part of hNHE1 that further supports this conclusion. The hNHE1 model was based on the crystal structure of the Escherichia coli Na(+)/H(+) antiporter, NhaA, and previous cysteine scanning accessibility studies of hNHE1 and was validated by EPR spectroscopy of spin labels in TM IV and TM XI, as well as by functional analysis of hNHE1 mutants. Removal of all endogenous cysteines in hNHE1, introduction of the mutations A173C (TM IV) and/or I461C (TM XI), and expression of the constructs in mammalian cells resulted in functional hNHE1 proteins. The distance between these spin labels was ∼15 A, confirming that TM IV and TM XI are in close proximity. This distance was decreased both at pH 5.1 and in the presence of the NHE1 inhibitor cariporide. A similar TM IV·TM XI distance and a similar change upon a pH shift were found for the cariporide-insensitive Pleuronectes americanus (pa) NHE1; however, in paNHE1, cariporide had no effect on TM IV·TM XI distance. The central role of the TM IV·TM XI arrangement was confirmed by the partial loss of function upon mutation of Arg(425), which the model predicts stabilizes this arrangement. The data are consistent with a role for TM IV and TM XI rearrangements coincident with ion translocation and inhibitor binding by hNHE1.


Subject(s)
Cation Transport Proteins/chemistry , Cation Transport Proteins/metabolism , Electron Spin Resonance Spectroscopy/methods , Models, Molecular , Sodium-Hydrogen Exchangers/chemistry , Sodium-Hydrogen Exchangers/metabolism , Amino Acid Sequence , Animals , Arginine , Cation Transport Proteins/antagonists & inhibitors , Cation Transport Proteins/genetics , Cell Line , Cell Membrane/metabolism , Cricetinae , Cricetulus , Flounder , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Protein Structure, Tertiary , Sodium-Hydrogen Exchanger 1 , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium-Hydrogen Exchangers/genetics
13.
PLoS One ; 6(12): e29210, 2011.
Article in English | MEDLINE | ID: mdl-22216214

ABSTRACT

The Na(+)/H(+)Exchanger isoform 1 (NHE1) is a highly versatile, broadly distributed and precisely controlled transport protein that mediates volume and pH regulation in most cell types. NHE1 phosphorylation contributes to Na(+)/H(+) exchange activity in response to phorbol esters, growth factors or protein phosphatase inhibitors, but has not been observed during activation by osmotic cell shrinkage (OCS). We examined the role of NHE1 phosphorylation during activation by OCS, using an ideal model system, the Amphiuma tridactylum red blood cell (atRBC). Na(+)/H(+) exchange in atRBCs is mediated by an NHE1 homolog (atNHE1) that is 79% identical to human NHE1 at the amino acid level. NHE1 activity in atRBCs is exceptionally robust in that transport activity can increase more than 2 orders of magnitude from rest to full activation. Michaelis-Menten transport kinetics indicates that either OCS or treatment with the phosphatase inhibitor calyculin-A (CLA) increase Na(+) transport capacity without affecting transport affinity (K(m)=44 mM) in atRBCs. CLA and OCS act non-additively to activate atNHE1, indicating convergent, phosphorylation-dependent signaling in atNHE1 activation. In situ(32)P labeling and immunoprecipitation demonstrates that the net phosphorylation of atNHE1 is increased 4-fold during OCS coinciding with a more than 2-order increase in Na(+) transport activity. This is the first reported evidence of increased NHE1 phosphorylation during OCS in any vertebrate cell type. Finally, liquid chromatography and mass spectrometry (LC-MS/MS) analysis of atNHE1 immunoprecipitated from atRBC membranes reveals 9 phosphorylated serine/threonine residues, suggesting that activation of atNHE1 involves multiple phosphorylation and/or dephosphorylation events.


Subject(s)
Cation Transport Proteins/metabolism , Erythrocytes/metabolism , Sodium-Hydrogen Exchangers/metabolism , Amino Acid Sequence , Cation Transport Proteins/chemistry , Cell Size , Chromatography, Liquid , Erythrocytes/cytology , Humans , Immunoprecipitation , Ion Transport , Kinetics , Molecular Sequence Data , Osmosis , Phosphorylation , Sequence Homology, Amino Acid , Sodium/metabolism , Sodium-Hydrogen Exchanger 1 , Sodium-Hydrogen Exchangers/chemistry , Tandem Mass Spectrometry
14.
J Cereb Blood Flow Metab ; 30(9): 1593-7, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20628400

ABSTRACT

Blood-brain barrier (BBB) P-glycoprotein activity is rapidly reduced by vascular endothelial growth factor (VEGF) acting via Src and by tumor necrosis factor-alpha acting via protein kinase C (PKC)beta1. To probe underlying mechanism(s), we developed an in vivo, immunoblot-based proteinase K (PK) protection assay to assess the changes in the P-glycoprotein content of the BBB's luminal membrane. Infusion of PK into the brain vasculature selectively cleaved luminal membrane P-glycoprotein, leaving intracellular proteins intact. Intracerebroventricular injection of VEGF partially protected P-glycoprotein from proteolytic cleavage, consistent with transporter internalization. Activation of PKCbeta1 did not protect P-glycoprotein. Thus, VEGF and PKCbeta1 reduce P-glycoprotein activity by distinct mechanisms.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Endopeptidase K/pharmacology , Neuroprotective Agents , Animals , Blotting, Western , Capillaries/drug effects , Cerebrovascular Circulation/drug effects , Endocytosis/drug effects , Enzyme Activation/drug effects , Injections, Intraventricular , Male , Membrane Proteins/metabolism , Phorbol Esters/pharmacology , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A/pharmacology
15.
Cardiovasc Res ; 87(2): 272-80, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20479130

ABSTRACT

Microvascular barrier dysfunction is implicated in the initiation and progression of inflammation, posttraumatic complications, sepsis, ischaemia-reperfusion injury, atherosclerosis, and diabetes. Under physiological conditions, a precise equilibrium between endothelial cell-cell adhesion and actin-myosin-based centripetal tension tightly controls the semi-permeability of microvascular barriers. Myosin light chain kinase (MLCK) plays an important role in maintaining the equilibrium by phosphorylating myosin light chain (MLC), thereby inducing actomyosin contractility and weakening endothelial cell-cell adhesion. MLCK is activated by numerous physiological factors and inflammatory or angiogenic mediators, causing vascular hyperpermeability. In this review, we discuss experimental evidence supporting the crucial role of MLCK in the hyperpermeability response to key cell signalling events during inflammation. At the cellular level, in vitro studies of cultured endothelial monolayers treated with MLCK inhibitors or transfected with specific inhibiting peptides have demonstrated that induction of endothelial MLCK activity is necessary for hyperpermeability. Ex vivo studies of live microvessels, enabled by development of the isolated, perfused venule method, support the importance of MLCK in endothelial permeability regulation in an environment that more closely resembles in vivo tissues. Finally, the role of MLCK in vascular hyperpermeability has been confirmed with in vivo studies of animal disease models and the use of transgenic MLCK210 knockout mice. These approaches provide a more complete view of the role of MLCK in vascular barrier dysfunction.


Subject(s)
Body Fluids/metabolism , Capillary Permeability , Endothelium, Vascular/enzymology , Microvessels/enzymology , Myosin-Light-Chain Kinase/metabolism , Actomyosin/metabolism , Animals , Capillary Permeability/drug effects , Capillary Permeability/genetics , Cells, Cultured , Endothelium, Vascular/drug effects , Endothelium, Vascular/immunology , Endothelium, Vascular/physiopathology , Humans , Inflammation/enzymology , Inflammation/physiopathology , Inflammation Mediators/metabolism , Mice , Mice, Transgenic , Microvessels/drug effects , Microvessels/immunology , Microvessels/physiopathology , Models, Animal , Myosin Light Chains/metabolism , Myosin-Light-Chain Kinase/antagonists & inhibitors , Myosin-Light-Chain Kinase/genetics , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Signal Transduction
16.
J Cereb Blood Flow Metab ; 30(7): 1373-83, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20197783

ABSTRACT

P-glycoprotein is an ATP (adenosine triphosphate)-driven drug efflux transporter that is highly expressed at the blood-brain barrier (BBB) and is a major obstacle to the pharmacotherapy of central nervous system diseases, including brain tumors, neuro-AIDS, and epilepsy. Previous studies have shown that P-glycoprotein transport activity in rat brain capillaries is rapidly reduced by the proinflammatory cytokine, tumor necrosis factor-alpha (TNF-alpha) acting through protein kinase C (PKC)-dependent signaling. In this study, we used isolated rat brain capillaries to show that the TNF-alpha-induced reduction of P-glycoprotein activity was prevented by a PKCbeta(I/II) inhibitor, LY333531, and mimicked by a PKCbeta(I/II) activator, 12-deoxyphorbol-13-phenylacetate-20-acetate (dPPA). Western blotting of brain capillary extracts with phospho-specific antibodies showed that dPPA activated PKCbeta(I), but not PKCbeta(II). Moreover, in intact rats, intracarotid infusion of dPPA potently increased brain accumulation of the P-glycoprotein substrate, [(3)H]-verapamil without compromising tight junction integrity. Thus, PKCbeta(I) activation selectively reduced P-glycoprotein activity both in vitro and in vivo. Targeting PKCbeta(I) at the BBB may prove to be an effective strategy for enhancing the delivery of small molecule therapeutics to the brain.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Blood-Brain Barrier/metabolism , Brain , Isoenzymes/metabolism , Pharmaceutical Preparations , Protein Kinase C/metabolism , Animals , Brain/blood supply , Brain/metabolism , Calcium Channel Blockers/metabolism , Capillaries/metabolism , Enzyme Activation , Humans , Indoles/metabolism , Isoenzymes/antagonists & inhibitors , Maleimides/metabolism , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/metabolism , Phorbol Esters/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C beta , Rats , Signal Transduction/physiology , Sucrose/metabolism , Tumor Necrosis Factor-alpha/metabolism , Verapamil/metabolism
17.
Am J Physiol Cell Physiol ; 298(3): C510-20, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19940069

ABSTRACT

The Na(+)/H(+) and K(+)/H(+) exchange pathways of Amphiuma tridactylum red blood cells (RBCs) are quiescent at normal resting cell volume yet are selectively activated in response to cell shrinkage and swelling, respectively. These alkali metal/H(+) exchangers are activated by net kinase activity and deactivated by net phosphatase activity. We employed relaxation kinetic analyses to gain insight into the basis for coordinated control of these volume regulatory ion flux pathways. This approach enabled us to develop a model explaining how phosphorylation/dephosphorylation-dependent events control and coordinate the activity of the Na(+)/H(+) and K(+)/H(+) exchangers around the cell volume set point. We found that the transition between initial and final steady state for both activation and deactivation of the volume-induced Na(+)/H(+) and K(+)/H(+) exchange pathways in Amphiuma RBCs proceed as a single exponential function of time. The rate of Na(+)/H(+) exchange activation increases with cell shrinkage, whereas the rate of Na(+)/H(+) exchange deactivation increases as preshrunken cells are progressively swollen. Similarly, the rate of K(+)/H(+) exchange activation increases with cell swelling, whereas the rate of K(+)/H(+) exchange deactivation increases as preswollen cells are progressively shrunken. We propose a model in which the activities of the controlling kinases and phosphatases are volume sensitive and reciprocally regulated. Briefly, the activity of each kinase-phosphatase pair is reciprocally related, as a function of volume, and the volume sensitivities of kinases and phosphatases controlling K(+)/H(+) exchange are reciprocally related to those controlling Na(+)/H(+) exchange.


Subject(s)
Amphibian Proteins/blood , Cell Size , Erythrocytes/metabolism , Potassium-Hydrogen Antiporters/blood , Potassium/metabolism , Sodium-Hydrogen Exchangers/blood , Sodium/metabolism , Urodela/blood , Animals , Enzyme Activation , Kinetics , Models, Biological , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Protein Kinases/metabolism , Signal Transduction , Water-Electrolyte Balance
18.
Am J Physiol Cell Physiol ; 295(5): C1316-25, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18799654

ABSTRACT

Alteration in cell volume of vertebrates results in activation of volume-sensitive ion flux pathways. Fine control of the activity of these pathways enables cells to regulate volume following osmotic perturbation. Protein phosphorylation and dephosphorylation have been reported to play a crucial role in the control of volume-sensitive ion flux pathways. Exposing Amphiuma tridactylu red blood cells (RBCs) to phorbol esters in isotonic medium results in a simultaneous, dose-dependent activation of both Na(+)/H(+) and K(+)/H(+) exchangers. We tested the hypothesis that in Amphiuma RBCs, both shrinkage-induced Na(+)/H(+) exchange and swelling-induced K(+)/H(+) exchange are activated by phosphorylation-dependent reactions. To this end, we assessed the effect of calyculin A, a phosphatase inhibitor, on the activity of the aforementioned exchangers. We found that exposure of Amphiuma RBCs to calyculin-A in isotonic media results in simultaneous, 1-2 orders of magnitude increase in the activity of both K(+)/H(+) and Na(+)/H(+) exchangers. We also demonstrate that, in isotonic media, calyculin A-dependent increases in net Na(+) uptake and K(+) loss are a direct result of phosphatase inhibition and are not dependent on changes in cell volume. Whereas calyculin A exposure in the absence of volume changes results in stimulation of both the Na(+)/H(+) and K(+)/H(+) exchangers, superimposing cell swelling or shrinkage and calyculin A treatment results in selective activation of K(+)/H(+) or Na(+)/H(+) exchange, respectively. We conclude that kinase-dependent reactions are responsible for Na(+)/H(+) and K(+)/H(+) exchange activity, whereas undefined volume-dependent reactions confer specificity and coordinated control.


Subject(s)
Cell Size/drug effects , Enzyme Inhibitors/pharmacology , Erythrocytes/drug effects , Oxazoles/pharmacology , Phosphoprotein Phosphatases/antagonists & inhibitors , Potassium-Hydrogen Antiporters/agonists , Sodium-Hydrogen Exchangers/agonists , Urodela/blood , Amiloride/analogs & derivatives , Amiloride/pharmacology , Animals , Dose-Response Relationship, Drug , Erythrocytes/enzymology , Marine Toxins , Osmotic Pressure , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Potassium/metabolism , Potassium-Hydrogen Antiporters/metabolism , Protein Kinases/metabolism , Ruthenium/metabolism , Sodium/metabolism , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium-Hydrogen Exchangers/metabolism , Time Factors
19.
J Biol Chem ; 282(27): 19716-27, 2007 Jul 06.
Article in English | MEDLINE | ID: mdl-17493937

ABSTRACT

The interaction of the ubiquitous Na(+)/H(+) exchanger, NHE1, with its commonly used inhibitors, amiloride- and benzoylguanidine (Hoechst type inhibitor (HOE))-type compounds, is incompletely understood. We previously cloned NHE1 from Amphiuma tridactylum (AtNHE1) and Pleuronectes americanus (PaNHE1). Although highly homologous to the amiloride- and HOE-sensitive human NHE1 (hNHE1), AtNHE1 is insensitive to HOE-type and PaNHE1 to both amiloride- and HOE-type compounds. Here we generated chimeras to "knock in" amiloride and HOE sensitivity to PaNHE1, and we thereby identified several NHE1 regions involved in inhibitor interaction. The markedly different inhibitor sensitivities of hNHE1, AtNHE1, and PaNHE1 could not be accounted for by differences in transmembrane (TM) region 9. Replacing TM10 through the C-terminal tail of PaNHE1 with the corresponding region of AtNHE1 partially restored sensitivity to amiloride and the related compound 5'-(N-ethyl-N-isopropyl)amiloride (EIPA) but not to HOE694. This effect was not due to the tail region, but it was dependent on TM10-11, because replacing only this region with that of AtNHE1 also partially restored amiloride and EIPA but not HOE sensitivity. The converse mutant (TM10-11 of AtNHE1 replaced with those of PaNHE1) exhibited even higher amiloride and EIPA sensitivity and was also HOE-sensitive. Replacing an LFFFY motif in TM region 4 of PaNHE1 with the corresponding residues of hNHE1 (VFFLF) or AtNHE1 (TFFLF) greatly increased sensitivity to both amiloride- and HOE-type compounds, despite the fact that AtNHE1 is HOE694-insensitive. Gain of amiloride sensitivity appeared to correlate with increased Na(+)/H(+) exchange rates. It is concluded that regions within TM4 and TM10-11 contribute to amiloride and HOE sensitivity, with both regions imparting partial inhibitor sensitivity to NHE1.


Subject(s)
Amiloride/chemistry , Cation Transport Proteins/antagonists & inhibitors , Guanidines/chemistry , Sodium Channel Blockers/chemistry , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sulfones/chemistry , Animals , Binding Sites , Cation Transport Proteins/chemistry , Cation Transport Proteins/genetics , Cell Line , Cricetinae , Cricetulus , Flounder , Humans , Mutation , Protein Structure, Tertiary/genetics , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Sodium-Hydrogen Exchanger 1 , Sodium-Hydrogen Exchangers/chemistry , Sodium-Hydrogen Exchangers/genetics , Species Specificity , Urodela
20.
Am J Physiol Cell Physiol ; 284(6): C1561-76, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12734109

ABSTRACT

In this report, we describe the cloning, cellular localization, and functional characteristics of Na(+)/H(+) exchanger 1 (NHE1) from red blood cells of the winter flounder Pseudopleuronectes americanus (paNHE1). The paNHE1 protein localizes primarily to the marginal band and exhibits a 74% similarity to the trout beta-NHE, and 65% to the human NHE1 (hNHE1). Functionally, paNHE1 shares characteristics of both beta-NHE and hNHE1 in that it is activated both by manipulations that increase cAMP and by cell shrinkage, respectively. In accordance, the paNHE1 protein exhibits both protein kinase A consensus sites as in beta-NHE and a region of high homology to that required for shrinkage-dependent activation of hNHE1. After shrinkage-dependent activation of paNHE1 and resulting activation of a Cl(-)/HCO(3)(-) exchanger, their parallel operation results in net uptake of NaCl and osmotically obliged water. Activation of paNHE1 by cAMP is at least additive to that elicited by osmotic shrinkage, suggesting that these stimuli regulate paNHE1 by distinct mechanisms. Finally, exposure to the serine/threonine phosphatase inhibitor calyculin A potently activates paNHE1, and this activation is also additive to that induced by shrinkage or cAMP.


Subject(s)
Cyclic AMP/metabolism , Enzyme Inhibitors/metabolism , Erythrocytes/metabolism , Flounder/metabolism , Oxazoles/metabolism , Sodium-Hydrogen Exchangers/metabolism , Adrenergic beta-Agonists/pharmacology , Amino Acid Sequence , Animals , Base Sequence , Cell Membrane/metabolism , Cell Size , Chlorides/metabolism , Cloning, Molecular , Dose-Response Relationship, Drug , Erythrocytes/cytology , Erythrocytes/drug effects , Flounder/genetics , Humans , Isoproterenol/pharmacology , Marine Toxins , Molecular Sequence Data , Osmolar Concentration , Phylogeny , Potassium/metabolism , Sodium/metabolism , Sodium-Hydrogen Exchangers/chemistry , Sodium-Hydrogen Exchangers/classification , Sodium-Hydrogen Exchangers/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...