Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 22(18)2021 Sep 14.
Article in English | MEDLINE | ID: mdl-34576081

ABSTRACT

Pulmonary hypertension (PH) is a progressive cardiovascular disorder in which local vascular inflammation leads to increased pulmonary vascular remodeling and ultimately to right heart failure. The HDAC inhibitor butyrate, a product of microbial fermentation, is protective in inflammatory intestinal diseases, but little is known regarding its effect on extraintestinal diseases, such as PH. In this study, we tested the hypothesis that butyrate is protective in a Sprague-Dawley (SD) rat model of hypoxic PH. Treatment with butyrate (220 mg/kg intake) prevented hypoxia-induced right ventricular hypertrophy (RVH), hypoxia-induced increases in right ventricular systolic pressure (RVSP), pulmonary vascular remodeling, and permeability. A reversal effect of butyrate (2200 mg/kg intake) was observed on elevated RVH. Butyrate treatment also increased the acetylation of histone H3, 25-34 kDa, and 34-50 kDa proteins in the total lung lysates of butyrate-treated animals. In addition, butyrate decreased hypoxia-induced accumulation of alveolar (mostly CD68+) and interstitial (CD68+ and CD163+) lung macrophages. Analysis of cytokine profiles in lung tissue lysates showed a hypoxia-induced upregulation of TIMP-1, CINC-1, and Fractalkine and downregulation of soluble ICAM (sICAM). The expression of Fractalkine and VEGFα, but not CINC-1, TIMP-1, and sICAM was downregulated by butyrate. In rat microvascular endothelial cells (RMVEC), butyrate (1 mM, 2 and 24 h) exhibited a protective effect against TNFα- and LPS-induced barrier disruption. Butyrate (1 mM, 24 h) also upregulated tight junctional proteins (occludin, cingulin, claudin-1) and increased the acetylation of histone H3 but not α-tubulin. These findings provide evidence of the protective effect of butyrate on hypoxic PH and suggest its potential use as a complementary treatment for PH and other cardiovascular diseases.


Subject(s)
Butyrates/pharmacology , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/physiopathology , Hypoxia/complications , Lung/physiopathology , Pneumonia/physiopathology , Vascular Remodeling/drug effects , Acetylation/drug effects , Animals , Blood Pressure/drug effects , Cytokines/metabolism , Endothelial Cells/drug effects , Heart Ventricles/drug effects , Heart Ventricles/physiopathology , Hypertrophy, Right Ventricular/complications , Hypertrophy, Right Ventricular/physiopathology , Lung/blood supply , Lung/drug effects , Macrophages/drug effects , Macrophages/pathology , Microvessels/pathology , Pneumonia/complications , Rats, Sprague-Dawley , Systole/drug effects , Tight Junctions/drug effects , Tight Junctions/metabolism , Up-Regulation/drug effects
2.
Int J Mol Sci ; 21(18)2020 Sep 18.
Article in English | MEDLINE | ID: mdl-32962005

ABSTRACT

Purinergic G-protein-coupled receptors are ancient and the most abundant group of G-protein-coupled receptors (GPCRs). The wide distribution of purinergic receptors in the cardiovascular system, together with the expression of multiple receptor subtypes in endothelial cells (ECs) and other vascular cells demonstrates the physiological importance of the purinergic signaling system in the regulation of the cardiovascular system. This review discusses the contribution of purinergic P2Y receptors to endothelial dysfunction (ED) in numerous cardiovascular diseases (CVDs). Endothelial dysfunction can be defined as a shift from a "calm" or non-activated state, characterized by low permeability, anti-thrombotic, and anti-inflammatory properties, to a "activated" state, characterized by vasoconstriction and increased permeability, pro-thrombotic, and pro-inflammatory properties. This state of ED is observed in many diseases, including atherosclerosis, diabetes, hypertension, metabolic syndrome, sepsis, and pulmonary hypertension. Herein, we review the recent advances in P2Y receptor physiology and emphasize some of their unique signaling features in pulmonary endothelial cells.


Subject(s)
Cardiovascular Diseases/metabolism , Endothelial Cells/metabolism , Endothelium/metabolism , Neovascularization, Pathologic/metabolism , Receptors, Purinergic P2Y/metabolism , Signal Transduction/physiology , Animals , Endothelium/pathology , Humans , Inflammation/metabolism , Inflammation/physiopathology , Oxidative Stress/physiology , Receptors, Purinergic P2Y/physiology
3.
Front Mol Neurosci ; 11: 45, 2018.
Article in English | MEDLINE | ID: mdl-29515368

ABSTRACT

Alcohol exposure can affect brain development, leading to long-lasting behavioral problems, including cognitive impairment, which together is defined as fetal alcohol spectrum disorder (FASD). However, the fundamental mechanisms through which this occurs are largely unknown. In this study, we report that the exposure of postnatal day 7 (P7) mice to ethanol activates caspase-3 via cannabinoid receptor type-1 (CB1R) in neonatal mice and causes a reduction in methylated DNA binding protein (MeCP2) levels. The developmental expression of MeCP2 in mice is closely correlated with synaptogenesis and neuronal maturation. It was shown that ethanol treatment of P7 mice enhanced Mecp2 mRNA levels but reduced protein levels. The genetic deletion of CB1R prevented, and administration of a CB1R antagonist before ethanol treatment of P7 mice inhibited caspase-3 activation. Additionally, it reversed the loss of MeCP2 protein, cAMP response element binding protein (CREB) activation, and activity-regulated cytoskeleton-associated protein (Arc) expression. The inhibition of caspase-3 activity prior to ethanol administration prevented ethanol-induced loss of MeCP2, CREB activation, epigenetic regulation of Arc expression, long-term potentiation (LTP), spatial memory deficits and activity-dependent impairment of several signaling molecules, including MeCP2, in adult mice. Collectively, these results reveal that the ethanol-induced CB1R-mediated activation of caspase-3 degrades the MeCP2 protein in the P7 mouse brain and causes long-lasting neurobehavioral deficits in adult mice. This CB1R-mediated instability of MeCP2 during active synaptic maturation may disrupt synaptic circuit maturation and lead to neurobehavioral abnormalities, as observed in this animal model of FASD.

5.
Front Physiol ; 5: 259, 2014.
Article in English | MEDLINE | ID: mdl-25076911

ABSTRACT

RATIONALE: Antibiotic treatment of patients infected with G(-) or G(+) bacteria promotes release of the toxins lipopolysaccharide (LPS) and pneumolysin (PLY) in their lungs. Growth Hormone-releasing Hormone (GHRH) agonist JI-34 protects human lung microvascular endothelial cells (HL-MVEC), expressing splice variant 1 (SV-1) of the receptor, from PLY-induced barrier dysfunction. We investigated whether JI-34 also blunts LPS-induced hyperpermeability. Since GHRH receptor (GHRH-R) signaling can potentially stimulate both cAMP-dependent barrier-protective pathways as well as barrier-disruptive protein kinase C pathways, we studied their interaction in GHRH agonist-treated HL-MVEC, in the presence of PLY, by means of siRNA-mediated protein kinase A (PKA) depletion. METHODS: Barrier function measurements were done in HL-MVEC monolayers using Electrical Cell substrate Impedance Sensing (ECIS) and VE-cadherin expression by Western blotting. Capillary leak was assessed by Evans Blue dye (EBD) incorporation. Cytokine generation in broncho-alveolar lavage fluid (BALF) was measured by multiplex analysis. PKA and PKC-α activity were assessed by Western blotting. RESULTS: GHRH agonist JI-34 significantly blunts LPS-induced barrier dysfunction, at least in part by preserving VE-cadherin expression, while not affecting inflammation. In addition to activating PKA, GHRH agonist also increases PKC-α activity in PLY-treated HL-MVEC. Treatment with PLY significantly decreases resistance in control siRNA-treated HL-MVEC, but does so even more in PKA-depleted monolayers. Pretreatment with GHRH agonist blunts PLY-induced permeability in control siRNA-treated HL-MVEC, but fails to improve barrier function in PKA-depleted PLY-treated monolayers. CONCLUSIONS: GHRH signaling in HL-MVEC protects from both LPS and PLY-mediated endothelial barrier dysfunction and concurrently induces a barrier-protective PKA-mediated and a barrier-disruptive PKC-α-induced pathway in the presence of PLY, the former of which dominates the latter.

6.
Int J Neuropsychopharmacol ; 18(5)2014 Oct 31.
Article in English | MEDLINE | ID: mdl-25609594

ABSTRACT

BACKGROUND: Ethanol exposure to rodents during postnatal day 7 (P7), which is comparable to the third trimester of human pregnancy, induces long-term potentiation and memory deficits. However, the molecular mechanisms underlying these deficits are still poorly understood. METHODS: In the present study, we explored the potential role of epigenetic changes at cannabinoid type 1 (CB1R) exon1 and additional CB1R functions, which could promote memory deficits in animal models of fetal alcohol spectrum disorder. RESULTS: We found that ethanol treatment of P7 mice enhances acetylation of H4 on lysine 8 (H4K8ace) at CB1R exon1, CB1R binding as well as the CB1R agonist-stimulated GTPγS binding in the hippocampus and neocortex, two brain regions that are vulnerable to ethanol at P7 and are important for memory formation and storage, respectively. We also found that ethanol inhibits cyclic adenosine monophosphate response element-binding protein (CREB) phosphorylation and activity-regulated cytoskeleton-associated protein (Arc) expression in neonatal and adult mice. The blockade or genetic deletion of CB1Rs prior to ethanol treatment at P7 rescued CREB phosphorylation and Arc expression. CB1R knockout mice exhibited neither ethanol-induced neurodegeneration nor inhibition of CREB phosphorylation or Arc expression. However, both neonatal and adult mice did exhibit enhanced CREB phosphorylation and Arc protein expression. P7 ethanol-treated adult mice exhibited impaired spatial and social recognition memory, which were prevented by the pharmacological blockade or deletion of CB1Rs at P7. CONCLUSIONS: Together, these findings suggest that P7 ethanol treatment induces CB1R expression through epigenetic modification of the CB1R gene, and that the enhanced CB1R function induces pCREB, Arc, spatial, and social memory deficits in adult mice.


Subject(s)
Ethanol/toxicity , Gene Expression Regulation/drug effects , Memory Disorders/chemically induced , Neurodegenerative Diseases/chemically induced , Receptor, Cannabinoid, CB1/drug effects , Receptor, Cannabinoid, CB1/metabolism , AIDS-Related Complex/metabolism , Acetylation/drug effects , Age Factors , Animals , Animals, Newborn/metabolism , Animals, Newborn/psychology , CREB-Binding Protein/metabolism , Central Nervous System Depressants/toxicity , Epigenesis, Genetic/drug effects , Exons/drug effects , Female , Hippocampus/drug effects , Hippocampus/metabolism , Histones/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neocortex/drug effects , Neocortex/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/psychology , Phosphorylation/drug effects , Receptor, Cannabinoid, CB1/deficiency , Receptor, Cannabinoid, CB1/genetics , Social Behavior , Up-Regulation/drug effects
7.
Neurobiol Dis ; 54: 475-85, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23396011

ABSTRACT

Rodent exposure to binge-like ethanol during postnatal day 7 (P7), which is comparable to the third trimester of human pregnancy, induces neuronal cell loss. However, the molecular mechanisms underlying these neuronal losses are still poorly understood. Here, we tested the possibility of histone methylation mediated by G9a (lysine dimethyltransferase) in regulating neuronal apoptosis in P7 mice exposed to ethanol. G9a protein expression, which is higher during embryogenesis and synaptogenic period compared to adult brain, is entirely confined to the cell nuclei in the developing brain. We found that ethanol treatment at P7, which induces apoptotic neurodegeneration in neonatal mice, enhanced G9a activity followed by increased histone H3 lysine 9 (H3K9me2) and 27 (H3K27me2) dimethylation. In addition, it appears that increased dimethylation of H3K9 makes it susceptible to proteolytic degradation by caspase-3 in conditions in which ethanol induces neurodegeneration. Further, pharmacological inhibition of G9a activity prior to ethanol treatment at P7 normalized H3K9me2, H3K27me2 and total H3 proteins to basal levels and prevented neurodegeneration in neonatal mice. Together, these data demonstrate that G9a mediated histone H3K9 and K27 dimethylation critically regulates ethanol-induced neurodegeneration in the developing brain. Furthermore, these findings reveal a novel link between G9a and neurodegeneration in the developing brain exposed to postnatal ethanol and may have a role in fetal alcohol spectrum disorders.


Subject(s)
Central Nervous System Depressants/toxicity , Ethanol/toxicity , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Nerve Degeneration/chemically induced , Nerve Degeneration/enzymology , Animals , Animals, Newborn , Blotting, Western , Brain/drug effects , Brain/enzymology , Brain/growth & development , Enzyme Activation/drug effects , Immunohistochemistry , Methylation , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
8.
Am J Respir Cell Mol Biol ; 47(4): 445-53, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22582175

ABSTRACT

Antibiotics-induced release of the pore-forming virulence factor pneumolysin (PLY) in patients with pneumococcal pneumonia results in its presence days after lungs are sterile and is a major factor responsible for the induction of permeability edema. Here we sought to identify major mechanisms mediating PLY-induced endothelial dysfunction. We evaluated PLY-induced endothelial hyperpermeability in human lung microvascular endothelial cells (HL-MVECs) and human lung pulmonary artery endothelial cells in vitro and in mice instilled intratracheally with PLY. PLY increases permeability in endothelial monolayers by reducing stable and dynamic microtubule content and modulating VE-cadherin expression. These events, dependent upon an increased calcium influx, are preceded by protein kinase C (PKC)-α activation, perturbation of the RhoA/Rac1 balance, and an increase in myosin light chain phosphorylation. At later time points, PLY treatment increases the expression and activity of arginase in HL-MVECs. Arginase inhibition abrogates and suppresses PLY-induced endothelial barrier dysfunction by restoring NO generation. Consequently, a specific PKC-α inhibitor and the TNF-derived tonoplast intrinsic protein peptide, which blunts PLY-induced PKC-α activation, are able to prevent activation of arginase in HL-MVECs and to reduce PLY-induced endothelial hyperpermeability in mice. Arginase I (AI)(+/-)/arginase II (AII)(-/-) C57BL/6 mice, displaying a significantly reduced arginase I expression in the lungs, are significantly less sensitive to PLY-induced capillary leak than their wild-type or AI(+/+)/AII(-/-) counterparts, indicating an important role for arginase I in PLY-induced endothelial hyperpermeability. These results identify PKC-α and arginase I as potential upstream and downstream therapeutic targets in PLY-induced pulmonary endothelial dysfunction.


Subject(s)
Arginase/metabolism , Capillary Permeability , Endothelial Cells/metabolism , Lung/pathology , Protein Kinase C-alpha/metabolism , Streptolysins/pharmacology , Animals , Antigens, CD/metabolism , Arginase/antagonists & inhibitors , Bacterial Proteins/pharmacology , Cadherins/metabolism , Calcium Signaling , Cells, Cultured , Endothelial Cells/enzymology , Enzyme Inhibitors/pharmacology , Humans , Lung/blood supply , Lung/immunology , Male , Mice , Mice, Inbred C57BL , Microtubules/metabolism , Microvessels/pathology , Pneumonia/enzymology , Pneumonia/immunology , Pneumonia/pathology , Protein Kinase C-alpha/antagonists & inhibitors , rhoA GTP-Binding Protein/metabolism
9.
Proc Natl Acad Sci U S A ; 109(6): 2084-9, 2012 Feb 07.
Article in English | MEDLINE | ID: mdl-22308467

ABSTRACT

Aggressive treatment with antibiotics in patients infected with Streptococcus pneumoniae induces release of the bacterial virulence factor pneumolysin (PLY). Days after lungs are sterile, this pore-forming toxin can still induce pulmonary permeability edema in patients, characterized by alveolar/capillary barrier dysfunction and impaired alveolar liquid clearance (ALC). ALC is mainly regulated through Na(+) transport by the apically expressed epithelial sodium channel (ENaC) and the basolaterally expressed Na(+)/K(+)-ATPase in type II alveolar epithelial cells. Because no standard treatment is currently available to treat permeability edema, the search for novel therapeutic candidates is of high priority. We detected mRNA expression for the active receptor splice variant SV1 of the hypothalamic polypeptide growth hormone-releasing hormone (GHRH), as well as for GHRH itself, in human lung microvascular endothelial cells (HL-MVEC). Therefore, we have evaluated the effect of the GHRH agonist JI-34 on PLY-induced barrier and ALC dysfunction. JI-34 blunts PLY-mediated endothelial hyperpermeability in monolayers of HL-MVEC, in a cAMP-dependent manner, by means of reducing the phosphorylation of myosin light chain and vascular endothelial (VE)-cadherin. In human airway epithelial H441 cells, PLY significantly impairs Na(+) uptake, but JI-34 restores it to basal levels by means of increasing cAMP levels. Intratracheal instillation of PLY into C57BL6 mice causes pulmonary alveolar epithelial and endothelial hyperpermeability as well as edema formation, all of which are blunted by JI-34. These findings point toward a protective role of the GHRH signaling pathway in PLY-induced permeability edema.


Subject(s)
Growth Hormone-Releasing Hormone/agonists , Pulmonary Edema/pathology , Streptolysins/toxicity , Animals , Antigens, CD/metabolism , Bacterial Proteins/toxicity , Cadherins/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology , Gene Expression Regulation , Growth Hormone-Releasing Hormone/genetics , Growth Hormone-Releasing Hormone/metabolism , Humans , Ion Channel Gating , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred C57BL , Microvessels/pathology , Myosin Light Chains/metabolism , Permeability , Phosphorylation , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/pathology , Pulmonary Edema/genetics , Pulmonary Edema/physiopathology , RNA Splicing/genetics , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/genetics , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Sodium Channels/metabolism
10.
J Cell Physiol ; 227(4): 1701-8, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21678426

ABSTRACT

The phosphorylation status of myosin light chain (MLC) is regulated by both MLC kinases and type 1 Ser/Thr phosphatase (PPase 1), MLC phosphatase (MLCP) activities. The activity of the catalytic subunit of MLCP (CS1ß) towards myosin depends on its associated regulatory subunit, namely myosin PPase targeting subunit 1 (MYPT1). Our previously published data strongly suggested the involvement of MLCP in endothelial cell (EC) barrier regulation. In this study, our new data demonstrate that inhibition of MLCP by either CS1ß or MYPT1 siRNA-based depletion results in significant attenuation of purine nucleotide (ATP and adenosine)-induced EC barrier enhancement. Consistent with the data, thrombin-induced EC F-actin stress fiber formation and permeability increase were attenuated by the ectopic expression of constitutively active (C/A) MYPT1. The data demonstrated for the first time direct involvement of MLCP in EC barrier enhancement/protection. Cloning of MYPT1 in human pulmonary artery EC (HPAEC) revealed the presence of two MYPT1 isoforms, long and variant 2 (V2) lacking 56 amino acids from 553 to 609 of human MYPT1 long, which were previously identified in HeLa and HEK 293 cells. Our data demonstrated that in Cos-7 cells ectopically expressed EC MYPT1 isoforms co-immunoprecipitated with intact CS1ß suggesting the importance of PPase 1 activity for the formation of functional complex of MYPT1/CS1ß. Interestingly, MYPT1 V2 shows decreased binding affinity compared to MYPT1 long for radixin (novel MLCP substrate and a member of ERM family proteins). These results suggest functional difference between EC MYPT1 isoforms in the regulation of MLCP activity and cytoskeleton.


Subject(s)
Endothelial Cells/enzymology , Myosin-Light-Chain Phosphatase/metabolism , Animals , Base Sequence , COS Cells , Capillary Permeability , Cells, Cultured , Chlorocebus aethiops , Cloning, Molecular , Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/metabolism , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Membrane Proteins/metabolism , Myosin-Light-Chain Phosphatase/chemistry , Myosin-Light-Chain Phosphatase/genetics , Protein Interaction Domains and Motifs , Protein Subunits , RNA, Small Interfering/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity , Transcription Factors/metabolism
11.
J Cardiovasc Dis Res ; 2(1): 14-22, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21716747

ABSTRACT

Endothelial cells (ECs), forming a semi-permeable barrier between the interior space of blood vessels and underlying tissues, control such diverse processes as vascular tone, homeostasis, adhesion of platelets, and leukocytes to the vascular wall and permeability of vascular wall for cells and fluids. Mechanisms which govern the highly clinically relevant process of increased EC permeability are under intense investigation. It is well known that loss of this barrier (permeability increase) results in tissue inflammation, the hall mark of inflammatory diseases such as acute lung injury and its severe form, acute respiratory distress syndrome. Little is known about processes which determine the endothelial barrier enhancement or protection against permeability increase. It is now well accepted that extracellular purines and pyrimidines are promising and physiologically relevant barrier-protective agents and their effects are mediated by interaction with cell surface P2Y receptors which belong to the superfamily of G-protein-coupled receptors. The therapeutic potential of P2Y receptors is rapidly expanding field in pharmacology and some selective agonists became recently available. Here, we present an overview of recently identified P2Y receptor agonists that enhance the pulmonary endothelial barrier and inhibit and/or reverse endothelial barrier disruption.

12.
Am J Physiol Cell Physiol ; 300(5): C1181-92, 2011 May.
Article in English | MEDLINE | ID: mdl-21289285

ABSTRACT

Enhanced vascular arginase activity impairs endothelium-dependent vasorelaxation by decreasing l-arginine availability to endothelial nitric oxide (NO) synthase, thereby reducing NO production. Elevated angiotensin II (ANG II) is a key component of endothelial dysfunction in many cardiovascular diseases and has been linked to elevated arginase activity. We determined signaling mechanisms by which ANG II increases endothelial arginase function. Results show that ANG II (0.1 µM, 24 h) elevates arginase activity and arginase I expression in bovine aortic endothelial cells (BAECs) and decreases NO production. These effects are prevented by the arginase inhibitor BEC (100 µM). Blockade of ANG II AT(1) receptors or transfection with small interfering RNA (siRNA) for Gα12 and Gα13 also prevents ANG II-induced elevation of arginase activity, but siRNA for Gαq does not. ANG II also elevates active RhoA levels and induces phosphorylation of p38 MAPK. Inhibitors of RhoA activation (simvastatin, 0.1 µM) or Rho kinase (ROCK) (Y-27632, 10 µM; H1152, 0.5 µM) block both ANG II-induced elevation of arginase activity and phosphorylation of p38 MAPK. Furthermore, pretreatment of BAECs with p38 inhibitor SB-202190 (2 µM) or transfection with p38 MAPK siRNA prevents ANG II-induced increased arginase activity/expression and maintains NO production. Additionally, inhibitors of p38 MAPK (SB-203580, 5 µg·kg(-1)·day(-1)) or arginase (ABH, 8 mg·kg(-1)·day(-1)) or arginase gene knockout in mice prevents ANG II-induced vascular endothelial dysfunction and associated enhancement of arginase. These results indicate that ANG II increases endothelial arginase activity/expression through Gα12/13 G proteins coupled to AT(1) receptors and subsequent activation of RhoA/ROCK/p38 MAPK pathways leading to endothelial dysfunction.


Subject(s)
Angiotensin II/physiology , Arginase/physiology , Endothelium, Vascular/physiopathology , p38 Mitogen-Activated Protein Kinases/physiology , rho-Associated Kinases/physiology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Amides/pharmacology , Angiotensin II/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Arginase/antagonists & inhibitors , Boronic Acids/pharmacology , Cattle , Cell Line , Endothelial Cells , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Enzyme Inhibitors/pharmacology , GTP-Binding Protein alpha Subunits, G12-G13/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, Gq-G11/antagonists & inhibitors , Imidazoles/pharmacology , Mice , Phosphorylation , Pyridines/pharmacology , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Simvastatin/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , rho-Associated Kinases/antagonists & inhibitors
13.
Biochim Biophys Acta ; 1798(6): 1164-71, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20211600

ABSTRACT

Pyroglutamate, also known as 5-oxoproline, is a structural analog of proline. This amino acid derivative is a byproduct of glutathione metabolism, and is reabsorbed efficiently in kidney by Na(+)-coupled transport mechanisms. Previous studies have focused on potential participation of amino acid transport systems in renal reabsorption of this compound. Here we show that it is not the amino acid transport systems but instead the Na(+)-coupled monocarboxylate transporter SLC5A8 that plays a predominant role in this reabsorptive process. Expression of cloned human and mouse SLC5A8 in mammalian cells induces Na(+)-dependent transport of pyroglutamate that is inhibitable by various SLC5A8 substrates. SLC5A8-mediated transport of pyroglutamate is saturable with a Michaelis constant of 0.36+/-0.04mM. Na(+)-activation of the transport process exhibits sigmoidal kinetics with a Hill coefficient of 1.8+/-0.4, indicating involvement of more than one Na(+) in the activation process. Expression of SLC5A8 in Xenopuslaevis oocytes induces Na(+)-dependent inward currents in the presence of pyroglutamate under voltage-clamp conditions. The concentration of pyroglutamate necessary for induction of half-maximal current is 0.19+/-0.01mM. The Na(+)-activation kinetics is sigmoidal with a Hill coefficient of 2.3+/-0.2. Ibuprofen, a blocker of SLC5A8, suppressed pyroglutamate-induced currents in SLC5A8-expressing oocytes; the concentration of the blocker necessary for causing half-maximal inhibition is 14+/-1microM. The involvement of SLC5A8 can be demonstrated in rabbit renal brush border membrane vesicles by showing that the Na(+)-dependent uptake of pyroglutamate in these vesicles is inhibitable by known substrates of SLC5A8. The Na(+) gradient-driven pyroglutamate uptake was stimulated by an inside-negative K(+) diffusion potential induced by valinomycin, showing that the uptake process is electrogenic.


Subject(s)
Cation Transport Proteins/metabolism , Cell Membrane/metabolism , Pyrrolidonecarboxylic Acid/metabolism , Sodium/metabolism , Animals , Biological Transport/drug effects , Biological Transport/physiology , Cation Transport Proteins/genetics , Cell Line , Cell Membrane/genetics , Gene Expression , Humans , Ionophores/pharmacology , Kidney/metabolism , Kinetics , Mice , Microvilli/genetics , Microvilli/metabolism , Monocarboxylic Acid Transporters , Oocytes , Patch-Clamp Techniques , Potassium/metabolism , Rabbits , Rats , Valinomycin/pharmacology , Xenopus laevis
14.
Vascul Pharmacol ; 52(5-6): 199-206, 2010.
Article in English | MEDLINE | ID: mdl-20045081

ABSTRACT

Extracellular adenosine is a physiologically relevant agonist released by various sources, including endothelial cells (EC) and activated platelets, with complex effects mediated via activation of P1 purinergic receptors. Adenosine-induced EC production of glutathione peroxidase1 and nitric oxide is recognized, and an anti-inflammatory mechanism has been described. Effects of extracellular adenosine on the pulmonary EC barrier function and vascular permeability, however, remain poorly characterized. In this study, we demonstrated the adenosine-induced rapid dose-dependent barrier enhancement in human pulmonary artery EC (HPAEC) as measured by an increase in transendothelial electrical resistance (TER). We have shown that HPAEC express only A2A and A2B adenosine receptors. Pharmacological and siRNA depletion studies indicate that A2A, but not A2B receptor activation is required for the adenosine-induced TER increase. Depletion of Galphas with a specific siRNA significantly attenuated the adenosine-induced TER response in HPAEC. In contrast, depletion of either Galphaq or Galphai2 did not affect the adenosine-induced TER increase. This suggests that the adenosine-induced TER increase is cAMP-dependent. The adenosine-induced barrier enhancement effects were associated with a rearrangement of the EC F-actin component of the cytoskeleton, enhanced cell-surface presentation of cell-cell junctional protein VE-cadherin and an involvement of Myosin-light-chain phosphatase (MLCP). Our results suggest, for the first time, that adenosine regulates the EC barrier function via A2A receptors followed by Galphas engagement and is associated with cytoskeletal activation.


Subject(s)
Adenosine/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Receptor, Adenosine A2A/metabolism , Adenosine/administration & dosage , Cells, Cultured , Cyclic AMP/metabolism , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Dose-Response Relationship, Drug , Electric Impedance , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , GTP-Binding Protein alpha Subunits/metabolism , Humans , Permeability , Pulmonary Artery/cytology , Pulmonary Artery/metabolism , RNA, Small Interfering/administration & dosage
15.
J Cell Physiol ; 223(1): 215-23, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20054824

ABSTRACT

Extracellular beta-NAD is known to elevate intracellular levels of calcium ions, inositol 1,4,5-trisphate and cAMP. Recently, beta-NAD was identified as an agonist for P2Y1 and P2Y11 purinergic receptors. Since beta-NAD can be released extracellularly from endothelial cells (EC), we have proposed its involvement in the regulation of EC permeability. Here we show, for the first time, that endothelial integrity can be enhanced in EC endogenously expressing beta-NAD-activated purinergic receptors upon beta-NAD stimulation. Our data demonstrate that extracellular beta-NAD increases the transendothelial electrical resistance (TER) of human pulmonary artery EC (HPAEC) monolayers in a concentration-dependent manner indicating endothelial barrier enhancement. Importantly, beta-NAD significantly attenuated thrombin-induced EC permeability as well as the barrier-compromising effects of Gram-negative and Gram-positive bacterial toxins representing the barrier-protective function of beta-NAD. Immunofluorescence microscopy reveals more pronounced staining of cell-cell junctional protein VE-cadherin at the cellular periphery signifying increased tightness of the cell-cell contacts after beta-NAD stimulation. Interestingly, inhibitory analysis (pharmacological antagonists and receptor sequence specific siRNAs) indicates the participation of both P2Y1 and P2Y11 receptors in beta-NAD-induced TER increase. beta-NAD-treatment attenuates the lipopolysaccharide (LPS)-induced phosphorylation of myosin light chain (MLC) indicating its involvement in barrier protection. Our studies also show the involvement of cAMP-dependent protein kinase A and EPAC1 pathways as well as small GTPase Rac1 in beta-NAD-induced EC barrier enhancement. With these results, we conclude that beta-NAD regulates the pulmonary EC barrier integrity via small GTPase Rac1- and MLCP- dependent signaling pathways.


Subject(s)
Actins/metabolism , Capillary Permeability , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytoskeleton/metabolism , Endothelial Cells/enzymology , Guanine Nucleotide Exchange Factors/metabolism , NAD/metabolism , Pulmonary Artery/enzymology , rac1 GTP-Binding Protein/metabolism , Antigens, CD/metabolism , Bacterial Proteins/pharmacology , Cadherins/metabolism , Capillary Permeability/drug effects , Cells, Cultured , Cytoskeleton/drug effects , Electric Impedance , Endothelial Cells/drug effects , Guanine Nucleotide Exchange Factors/genetics , Humans , Intercellular Junctions/metabolism , Lipopolysaccharides/pharmacology , Myosin Light Chains/metabolism , Myosin-Light-Chain Phosphatase/metabolism , Phosphorylation , Pulmonary Artery/cytology , Pulmonary Artery/drug effects , RNA Interference , RNA, Messenger/metabolism , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2Y1 , Signal Transduction , Streptolysins/pharmacology , Thrombin/metabolism , Time Factors , rac1 GTP-Binding Protein/genetics
16.
Vascul Pharmacol ; 52(5-6): 207-13, 2010.
Article in English | MEDLINE | ID: mdl-20074664

ABSTRACT

Listeriosis can lead to potentially lethal pulmonary complications in newborns and immune compromised patients, characterized by extensive permeability edema. Listeriolysin (LLO), the main virulence factor of Listeria monocytogenes, induces a dose-dependent hyperpermeability in monolayers of human lung microvascular endothelial cells in vitro. The permeability increasing activity of LLO, which is accompanied by an increased reactive oxygen species (ROS) generation, RhoA activation and myosin light chain (MLC) phosphorylation, can be completely inhibited by the protein kinase C (PKC) alpha/beta inhibitor GO6976, indicating a crucial role for PKC in the induction of barrier dysfunction. The TNF-derived TIP peptide, which mimics the lectin-like domain of the cytokine, blunts LLO-induced hyperpermeability in vitro, upon inhibiting LLO-induced protein kinase C-alpha activation, ROS generation and MLC phosphorylation and upon restoring the RhoA/Rac 1 balance. These results indicate that the lectin-like domain of TNF has a potential therapeutic value in protecting from LLO-induced pulmonary endothelial hyperpermeability.


Subject(s)
Bacterial Toxins/toxicity , Endothelium, Vascular/metabolism , Heat-Shock Proteins/toxicity , Hemolysin Proteins/toxicity , Listeria monocytogenes/pathogenicity , Tumor Necrosis Factor-alpha/metabolism , Animals , Cattle , Cells, Cultured , Endothelium, Vascular/microbiology , Humans , Lung/cytology , Lung/metabolism , Lung/microbiology , Myosin Light Chains/metabolism , Peptides/pharmacology , Permeability , Phosphorylation , Protein Kinase C-alpha/antagonists & inhibitors , Pulmonary Artery/metabolism , Pulmonary Artery/microbiology , Reactive Oxygen Species/metabolism , Sheep , Tumor Necrosis Factor-alpha/chemistry , rhoA GTP-Binding Protein/metabolism
17.
J Biol Chem ; 284(21): 14469-75, 2009 May 22.
Article in English | MEDLINE | ID: mdl-19304661

ABSTRACT

Atypical protein kinase Cs (PKCs) (aPKCzeta and lambda/iota) have emerged as important binding partners for ceramide, a membrane-resident cell signaling lipid that is involved in the regulation of apoptosis as well as cell polarity. Using ceramide overlay assays with proteolytic fragments of PKCzeta and vesicle binding assays with ectopically expressed protein, we show that a protein fragment comprising the carboxyl-terminal 20-kDa sequence of PKCzeta (C20zeta, amino acids 405-592) bound to C16:0 ceramide. This sequence is not identical to the C1 domain (amino acids 131-180), which has been suggested to serve as a potential ceramide binding domain. Using immunocytochemistry, we found that a C20zeta protein fragment ectopically expressed in two epithelial cell types (neural progenitors and Madin-Darby canine kidney cells) co-distributed with ceramide. Stable expression of C20zeta-EGFP in Madin-Darby canine kidney cells disrupted the formation of adherens and tight junctions and impaired the epithelium integrity by reducing transepithelial electrical resistance. Disruption of cell adhesion and loss of transepithelial electrical resistance was prevented by incubation with C16:0 ceramide. Our results show, for the first time, that there is a novel ceramide binding domain (C20zeta) in the carboxyl terminus of aPKC. Our results also show that the interaction of ceramide with this binding domain is essential for cell-to-cell contacts in epithelia. Therefore, ceramide interaction with the C20zeta binding domain is a potential mechanism by which ceramide and aPKC regulate the formation of junctional complexes in epithelial cells.


Subject(s)
Ceramides/metabolism , Epithelial Cells/cytology , Epithelial Cells/enzymology , Intercellular Junctions/enzymology , Protein Kinase C/chemistry , Protein Kinase C/metabolism , Animals , Cadherins/metabolism , Cell Line , Dogs , Down-Regulation , Genes, Dominant , Green Fluorescent Proteins/metabolism , Humans , Mutant Proteins/metabolism , Neurons/cytology , Neurons/enzymology , Peptide Fragments/metabolism , Protein Binding , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Stem Cells/enzymology
18.
DNA Cell Biol ; 28(3): 119-29, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19105596

ABSTRACT

We have previously shown that endothelial nitric oxide synthase (eNOS) promoter activity is decreased in endothelial cells in response to the addition of hydrogen peroxide (H(2)O(2)), and this involves, at least in part, the inhibition of AP-1 activity. Thus, the objective of this study was to determine if other cis-element(s) and transcription factor(s) are involved in the oxidant-mediated downregulation of eNOS. Our initial experiments indicated that although H(2)O(2) treatment increased eNOS mRNA levels in ovine pulmonary arterial endothelial cells (OPAECs), there was a significant decrease in the promoter activity of an eNOS promoter construct containing 840 bp of upstream sequence. However, a truncated promoter construct that lacked the AP-1 element (650 bp) was also inhibited by H(2)O(2). A similar effect was observed when the 650 bp human eNOS promoter construct was transfected into human PAECs. We also found that although exposure of the cells to PEG-catalase prevented the inhibitory effect on eNOS promoter activity, the hydroxyl radical scavenger, deferoxamine myslate, did not. Nor could we identify an increase in hydroxyl radical levels in cells exposed to H(2)O(2). Exposure of PAECs caused a significant increase in labile zinc levels in response to H(2)O(2). As the eNOS promoter has a cis-element for Sp1 binding, we evaluated the role of Sp1 in response to H(2)O(2). As previously reported, mutation of the Sp1 consensus lead to the complete loss of eNOS promoter activity, confirming the key role of Sp1 in regulating basal eNOS promoter activity. In addition, we found, using electrophoretic mobility and supershift assays, that H(2)O(2) decreased Sp1 binding. Finally, using chromatin immunoprecipitation analysis, we found a significant decrease in Sp1 binding to the eNOS promoter in vivo in response to treatment with H(2)O(2). Together, these data suggest that the inhibition of Sp1 activity, possibly through loss of zinc in the protein, plays a role in the H(2)O(2)-induced inhibition of eNOS promoter activity.


Subject(s)
Hydrogen Peroxide/pharmacology , Nitric Oxide Synthase Type III/genetics , Sp1 Transcription Factor/genetics , Transcription Factor AP-1/genetics , Zinc/metabolism , Animals , Cells, Cultured , Down-Regulation/drug effects , Down-Regulation/genetics , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Hydroxyl Radical/metabolism , Nitric Oxide Synthase Type III/drug effects , Nitric Oxide Synthase Type III/metabolism , Promoter Regions, Genetic , Pulmonary Artery/cytology , Pulmonary Artery/metabolism , Regulatory Sequences, Nucleic Acid , Sheep , Sp1 Transcription Factor/metabolism , Transcription Factor AP-1/metabolism
19.
Invest Ophthalmol Vis Sci ; 49(11): 5151-60, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18689705

ABSTRACT

PURPOSE: Glutamine transport is essential for the glutamate-glutamine cycle, which occurs between neurons and glia. System N, consisting of SN1 (SNAT3) and SN2 (SNAT5), is the principal mediator of glutamine transport in retinal Müller cells. Mediators of glutamine transport in retinal ganglion cells were investigated. METHODS: The relative contributions of various transport systems for glutamine uptake (systems N, A, L, y+L, ASCT, and ATB(0,+)) were examined in RGC-5 cells based on differential features of the individual transport systems. mRNA for the genes encoding members of these transport systems were analyzed by RT-PCR. Based on these data, SN1 and SN2 were analyzed in mouse retina, RGC-5 cells, and primary mouse ganglion cells (GCs) by in situ hybridization (ISH), immunofluorescence (IF), and Western blotting. RESULTS: Three transport systems--N, A, and L--participated in glutamine uptake in RGC-5 cells. System N was the principal contributor; systems A and L contributed considerably less. ISH and IF revealed SN1 and SN2 expression in the ganglion, inner nuclear, and photoreceptor cell layers. SN1 and SN2 colocalized with the ganglion cell marker Thy 1.2 and with the Müller cell marker vimentin, confirming their presence in both retinal cell types. SN1 and SN2 proteins were detected in primary mouse GCs. CONCLUSIONS: These findings suggest that in addition to its role in glutamine uptake in retinal glial cells, system N contributes significantly to glutamine uptake in ganglion cells and, hence, contributes to the retinal glutamate-glutamine cycle.


Subject(s)
Amino Acid Transport Systems, Neutral/genetics , Gene Expression , RNA, Messenger/genetics , Retinal Ganglion Cells/metabolism , Amino Acid Transport Systems, Neutral/biosynthesis , Animals , Blotting, Western , Cells, Cultured , In Situ Hybridization , Mice , Neurotransmitter Agents , Retinal Ganglion Cells/cytology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
20.
Biochem J ; 414(3): 343-55, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18522536

ABSTRACT

ATB(0,+) [SLC6A14 (solute carrier family 6 member 14)] is an Na(+)/Cl(-)-coupled amino acid transporter whose expression is upregulated in cancer. 1-Methyltryptophan is an inducer of immune surveillance against tumour cells through its ability to inhibit indoleamine dioxygenase. In the present study, we investigated the role of ATB(0,+) in the uptake of 1-methyltryptophan as a potential mechanism for entry of this putative anticancer drug into tumour cells. These studies show that 1-methyltryptophan is a transportable substrate for ATB(0,+). The transport process is Na(+)/Cl(-)-dependent with an Na(+)/Cl(-)/1-methyltryptophan stoichiometry of 2:1:1. Evaluation of other derivatives of tryptophan has led to identification of alpha-methyltryptophan as a blocker, not a transportable substrate, for ATB(0,+). ATB(0,+) can transport 18 of the 20 proteinogenic amino acids. alpha-Methyltryptophan blocks the transport function of ATB(0,+) with an IC(50) value of approximately 250 muM under conditions simulating normal plasma concentrations of all these 18 amino acids. These results suggest that alpha-methyltryptophan may induce amino acid deprivation in cells which depend on the transporter for their amino acid nutrition. Screening of several mammary epithelial cell lines shows that ATB(0,+) is expressed robustly in some cancer cell lines, but not in all; in contrast, non-malignant cell lines do not express the transporter. Treatment of ATB(0,+)-positive tumour cells with alpha-methyltryptophan leads to suppression of their colony-forming ability, whereas ATB(0,+)-negative cell lines are not affected. The blockade of ATB(0,+) in these cells with alpha-methyltryptophan is associated with cell cycle arrest. These studies reveal the potential of ATB(0,+) as a drug target for cancer chemotherapy.


Subject(s)
Amino Acid Transport Systems, Neutral/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Tryptophan/analogs & derivatives , Amino Acid Transport Systems/metabolism , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism , Animals , Antineoplastic Agents/therapeutic use , Biological Transport, Active/drug effects , Cell Line , Cell Line, Tumor , Humans , Large Neutral Amino Acid-Transporter 1/genetics , Large Neutral Amino Acid-Transporter 1/metabolism , Mice , Oocytes/metabolism , Plasma Membrane Neurotransmitter Transport Proteins/metabolism , Tryptophan/metabolism , Tryptophan/pharmacology , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...