Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Am J Physiol Lung Cell Mol Physiol ; 312(1): L143-L153, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27913425

ABSTRACT

Elevated levels of reactive oxygen species and intracellular Ca2+ play a key role in endothelial barrier dysfunction in acute lung injury. We previously showed that H2O2-induced increases in intracellular calcium concentrations ([Ca2+]i) in lung microvascular endothelial cells (LMVECs) involve the membrane Ca2+ channel, transient receptor potential vanilloid-4 (TRPV4) and that inhibiting this channel attenuated H2O2-induced barrier disruption in vitro. We also showed that phosphorylation of TRPV4 by the Src family kinase, Fyn, contributes to H2O2-induced Ca2+ influx in LMVEC. In endothelial cells, Fyn is tethered to the cell membrane by CD36, a fatty acid transporter. In this study, we assessed the effect of genetic loss or pharmacological inhibition of CD36 on Ca2+ responses to H2O2 H2O2-induced Ca2+ influx was attenuated in LMVEC isolated from mice lacking CD36 (CD36-/-). TRPV4 expression and function was unchanged in LMVEC isolated from wild-type (WT) and CD36-/- mice, as well as mice with deficiency for Fyn (Fyn-/-). TRPV4 immunoprecipitated with Fyn, but this interaction was decreased in CD36-/- LMVEC. The amount of phosphorylated TRPV4 was decreased in LMVEC from CD36-/- mice compared with WT controls. Loss of CD36 altered subcellular localization of Fyn, while inhibition of CD36 fatty acid transport with succinimidyl oleate did not attenuate H2O2-induced Ca2+ influx. Lastly, we found that CD36-/- mice were protected from ischemia-reperfusion injury in vivo. In conclusion, our data suggest that CD36 plays an important role in H2O2-mediated lung injury and that the mechanism may involve CD36-dependent scaffolding of Fyn to the cell membrane to facilitate TRPV4 phosphorylation.


Subject(s)
CD36 Antigens/metabolism , Calcium/metabolism , Endothelial Cells/metabolism , Hydrogen Peroxide/pharmacology , Lung/blood supply , Microvessels/cytology , Animals , Endothelial Cells/drug effects , Fatty Acids/metabolism , Gene Deletion , Lipoproteins, LDL/pharmacology , Mice, Inbred C57BL , Oleic Acid/pharmacology , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Transport/drug effects , Proto-Oncogene Proteins c-fyn/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , TRPV Cation Channels/metabolism
2.
Am J Physiol Cell Physiol ; 306(6): C559-69, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24401847

ABSTRACT

Oxidant injury contributes to acute lung injury (ALI). We previously reported that activation of protein kinase GI (PKGI) posttranscriptionally increased the key antioxidant enzymes catalase and glutathione peroxidase 1 (Gpx-1) and attenuated oxidant-induced cytotoxicity in mouse lung microvascular endothelial cells (MLMVEC). The present studies tested the hypothesis that the antioxidant effect of PKGI is mediated via inhibition of the c-Abl tyrosine kinase. We found that activation of PKGI with the cGMP analog 8pCPT-cGMP inhibited c-Abl activity and decreased c-Abl expression in wild-type but not PKGI(-/-) MLMVEC. Treatment of wild-type MLMVEC with atrial natriuretic peptide also inhibited c-Abl activation. Moreover, treatment of MLMVEC with the c-Abl inhibitor imatinib increased catalase and GPx-1 protein in a posttranscriptional fashion. In imatinib-treated MLMVEC, there was no additional effect of 8pCPT-cGMP on catalase or GPx-1. The imatinib-induced increase in antioxidant proteins was associated with an increase in extracellular H2O2 scavenging by MLMVEC, attenuation of oxidant-induced endothelial barrier dysfunction, and prevention of oxidant-induced endothelial cell death. Finally, in the isolated perfused lung, imatinib prevented oxidant-induced endothelial toxicity. We conclude that cGMP, through activation of PKGI, inhibits c-Abl, leading to increased key antioxidant enzymes and resistance to lung endothelial oxidant injury. Inhibition of c-Abl by active PKGI may be the downstream mechanism underlying PKGI-mediated antioxidant signaling. Tyrosine kinase inhibitors may represent a novel therapeutic approach in oxidant-induced ALI.


Subject(s)
Acute Lung Injury/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic GMP/metabolism , Lung/metabolism , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Active Transport, Cell Nucleus/physiology , Animals , Apoptosis/drug effects , Atrial Natriuretic Factor/metabolism , Benzamides/pharmacology , Catalase/metabolism , Cells, Cultured , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinases/drug effects , Cyclic GMP-Dependent Protein Kinases/genetics , Endothelial Cells/metabolism , Enzyme Activation , Glutathione Peroxidase/metabolism , Hydrogen Peroxide/metabolism , Imatinib Mesylate , Lung/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction/drug effects , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-abl/metabolism , Pyrimidines/pharmacology , RNA, Messenger/biosynthesis , Signal Transduction/drug effects , Glutathione Peroxidase GPX1
3.
Am J Physiol Lung Cell Mol Physiol ; 301(2): L161-70, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21571906

ABSTRACT

Phosphodiesterase 2A (PDE2A) is stimulated by cGMP to hydrolyze cAMP, a potent endothelial barrier-protective molecule. We previously found that lung PDE2A contributed to a mouse model of ventilator-induced lung injury (VILI). The purpose of the present study was to determine the contribution of PDE2A in a two-hit mouse model of 1-day intratracheal (IT) LPS followed by 4 h of 20 ml/kg tidal volume ventilation. Compared with IT water controls, LPS alone (3.75 µg/g body wt) increased lung PDE2A mRNA and protein expression by 6 h with a persistent increase in protein through day 4 before decreasing to control levels on days 6 and 10. Similar to the PDE2A time course, the peak in bronchoalveolar lavage (BAL) neutrophils, lactate dehydrogenase (LDH), and protein concentration also occurred on day 4 post-LPS. IT LPS (1 day) and VILI caused a threefold increase in lung PDE2A and inducible nitric oxide synthase (iNOS) and a 24-fold increase in BAL neutrophilia. Compared with a control adenovirus, PDE2A knockdown with an adenovirus expressing a short hairpin RNA administered IT 3 days before LPS/VILI effectively decreased lung PDE2A expression and significantly attenuated BAL neutrophilia, LDH, protein, and chemokine levels. PDE2A knockdown also reduced lung iNOS expression by 53%, increased lung cAMP by nearly twofold, and improved survival from 47 to 100%. We conclude that in a mouse model of LPS/VILI, a synergistic increase in lung PDE2A expression increased lung iNOS and alveolar inflammation and contributed significantly to the ensuing acute lung injury.


Subject(s)
Acute Lung Injury/etiology , Cyclic Nucleotide Phosphodiesterases, Type 2/deficiency , Lung/metabolism , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Adenoviridae/enzymology , Adenoviridae/genetics , Animals , Bronchoalveolar Lavage Fluid/cytology , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 2/genetics , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Inflammation/metabolism , Inflammation/pathology , Lipopolysaccharides/administration & dosage , Lung/virology , Male , Mice , Mice, Inbred C57BL , Neutrophils/pathology , Nitric Oxide Synthase Type II/metabolism , Proteins/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Tidal Volume , Time Factors , Trachea , Ventilator-Induced Lung Injury/metabolism , Ventilator-Induced Lung Injury/pathology
4.
Am J Physiol Lung Cell Mol Physiol ; 299(3): L323-33, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20453163

ABSTRACT

Increasing evidence suggests that endothelial cytotoxicity from reactive oxygen species (ROS) contributes to the pathogenesis of acute lung injury. Treatments designed to increase intracellular cGMP attenuate ROS-mediated apoptosis and necrosis in several cell types, but the mechanisms are not understood, and the effect of cGMP on pulmonary endothelial cell death remains controversial. In the current study, increasing intracellular cGMP by either 8pCPT-cGMP (50 microM) or atrial natriuretic peptide (10 nM) significantly attenuated cell death in H(2)O(2)-challenged mouse lung microvascular (MLMVEC) monolayers. 8pCPT-cGMP also decreased perfusate LDH release in isolated mouse lungs exposed to H(2)O(2) or ischemia-reperfusion. The protective effect of increasing cGMP in MLMVECs was accompanied by enhanced endothelial H(2)O(2) scavenging (measured by H(2)O(2) electrode) and decreased intracellular ROS concentration (measured by 2',7'-dichlorofluorescin fluorescence) as well as decreased phosphorylation of p38 MAPK and Akt. The cGMP-mediated cytoprotection and increased H(2)O(2) scavenging required >2 h of 8pCPT-cGMP incubation in wild-type MLMVEC and were absent in MLMVEC from protein kinase G (PKG(I))-/- mice suggesting a PKG(I)-mediated effect on gene regulation. Catalase and glutathione peroxidase 1 (Gpx-1) protein were increased by cGMP in wild-type but not PKG(I)-/- MLMVEC monolayers. Both the cGMP-mediated increases in antioxidant proteins and H(2)O(2) scavenging were prevented by inhibition of translation with cycloheximide. 8pCPT-cGMP had minimal effects on catalase and Gpx-1 mRNA. We conclude that cGMP, through PKG(I), attenuated H(2)O(2)-induced cytotoxicity in MLMVEC by increasing catalase and Gpx-1 expression through an unknown posttranscriptional effect.


Subject(s)
Antioxidants/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic GMP/metabolism , Endothelial Cells/metabolism , Hydrogen Peroxide/pharmacology , Lung/blood supply , Microvessels/physiopathology , Oxidants/pharmacology , Animals , Atrial Natriuretic Factor/pharmacology , Catalase/metabolism , Cell Death , Cells, Cultured , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinase Type I , Cyclic GMP-Dependent Protein Kinases/deficiency , Endothelial Cells/drug effects , Glutathione Peroxidase/metabolism , Intracellular Membranes/metabolism , Isoenzymes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidoreductases/metabolism , Protein Processing, Post-Translational , Reactive Oxygen Species/metabolism , Signal Transduction , Thionucleotides/pharmacology
5.
Toxicology ; 402-403: 37-49, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29689364

ABSTRACT

Chemically modified hemoglobin (Hb)-based oxygen carriers are promising oxygen replacement therapeutics however their potential renal effects are not fully understood. Using a guinea pig exchange transfusion model, we examined the effects of glutaraldehyde-polymerized bovine hemoglobin (HbG) on the permeability and integrity of the glomerular filtration barrier (GFB), which is comprised of podocytes, fenestrated endothelium, and the glomerular basement membrane. HbG induced marked proteinuria characterized in part by the loss of high molecular weight proteins, including albumin, immunoglobulin, and transferrin, at 4 and 12 h post-infusion that resolved by 72 h. This correlated with HbG-induced GFB alterations based on the reduced expression of specific markers of podocytes (podocin, nephrin, podocalyxin, and Wilms Tumor 1 protein) and endothelial cells (ETS-related gene and claudin-5). Lectin binding studies also demonstrated marked but reversible alterations to the GFB glycocalyx accompanied by increased intraglomerular HbG deposition and 4-HNE protein adduct expression indicative of oxidative damage. Together, these findings indicate that HbG induces reversible glomerular barrier dysfunction in conjunction with transient GFB changes providing new insight into the renal response to chemically modified Hb therapeutics.


Subject(s)
Glutaral/toxicity , Hemoglobins/toxicity , Kidney Glomerulus/drug effects , Kidney Glomerulus/pathology , Polymerization , Polymers/toxicity , Animals , Guinea Pigs , Kidney Glomerulus/physiopathology , Male , Proteinuria/chemically induced , Proteinuria/pathology , Proteinuria/physiopathology
6.
Biochem J ; 389(Pt 3): 705-16, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-15804237

ABSTRACT

The matrilin-1 gene has the unique feature that it is expressed in chondrocytes in a developmental stage-specific manner. Previously, we found that the chicken matrilin-1 long promoter with or without the intronic enhancer and the short promoter with the intronic enhancer restricted the transgene expression to the columnar proliferative chondroblasts and prehypertrophic chondrocytes of growth-plate cartilage in transgenic mice. To study whether the short promoter shared by these transgenes harbours cartilage-specific control elements, we generated transgenic mice expressing the LacZ reporter gene under the control of the matrilin-1 promoter between -338 and +67. Histological analysis of the founder embryos demonstrated relatively weak transgene activity in the developing chondrocranium, axial and appendicular skeleton with highest level of expression in the columnar proliferating chondroblasts and prehypertrophic chondrocytes. Computer analysis of the matrilin-1 genes of amniotes revealed a highly conserved Pe1 (proximal promoter element 1) and two less-conserved sequence blocks in the distal promoter region. The inverted Sox motifs of the Pe1 element interacted with chondrogenic transcription factors Sox9, L-Sox5 and Sox6 in vitro and another factor bound to the spacer region. Point mutations in the Sox motifs or in the spacer region interfered with or altered the formation of nucleoprotein complexes in vitro and significantly decreased the reporter gene activity in transient expression assays in chondrocytes. In vivo occupancy of the Sox motifs in genomic footprinting in the expressing cell type, but not in fibroblasts, also supported the involvement of Pe1 in the tissue-specific regulation of the gene. Our results indicate that interaction of Pe1 with distal DNA elements is required for the high level, cartilage- and developmental stage-specific transgene expression.


Subject(s)
Extracellular Matrix Proteins/genetics , Gene Expression Regulation, Developmental/physiology , Glycoproteins/genetics , High Mobility Group Proteins/physiology , Promoter Regions, Genetic/genetics , Transcription Factors/physiology , Animals , Base Sequence , Binding Sites , Cells, Cultured , Chick Embryo , Chondrocytes/metabolism , Conserved Sequence , DNA-Binding Proteins/chemistry , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/physiology , Fibroblasts/metabolism , Glycoproteins/chemistry , Glycoproteins/physiology , High Mobility Group Proteins/chemistry , High Mobility Group Proteins/genetics , High Mobility Group Proteins/metabolism , Matrilin Proteins , Mice , Mice, Transgenic , Molecular Sequence Data , Nuclear Proteins/chemistry , Point Mutation , Protein Binding , Repressor Proteins/chemistry , SOX9 Transcription Factor , SOXD Transcription Factors , Sequence Homology, Nucleic Acid , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism
7.
Toxics ; 4(1)2016.
Article in English | MEDLINE | ID: mdl-27471729

ABSTRACT

Hemoglobin-based oxygen carriers (HBOCs) are being developed as oxygen and plasma volume-expanding therapeutics though their potential to promote oxidative tissue injury has raised safety concerns. Using a guinea pig exchange transfusion model, we examined the effects of polymerized bovine hemoglobin (HbG) on the transcriptional regulation, activity, and expression of the renal antioxidant enzymes; superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx). HbG infusion downregulated the mRNA levels for genes encoding SOD isoforms 1-3, GPx1, GPx3, GPx4, and CAT. This transcriptional suppression correlated with decreased enzymatic activities for SOD, CAT, and GPx. Immunostaining revealed decreased protein expression of SOD1, CAT, and GPx1 primarily in renal cortical tubules. DNA methylation analyses identified CpG hypermethylation in the gene promoters for SOD1-3, GPx1, GPx3, and GPx4, suggesting an epigenetic-based mechanism underlying the observed gene repression. HbG also induced oxidative stress as evidenced by increased renal lipid peroxidation end-products and 4-HNE immunostaining, which could be the result of the depleted antioxidant defenses and/or serve as a trigger for increased DNA methylation. Together, these findings provide evidence that the renal exposure to HbG suppresses the function of major antioxidant defense systems which may have relevant implications for understanding the safety of hemoglobin-based products.

8.
Matrix Biol ; 22(8): 605-18, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15062854

ABSTRACT

Matrilin-1 is a non-collagenous protein, which functions in the organization of the extracellular matrix by forming collagen-dependent and -independent filamentous networks. It is secreted primarily by chondrocytes in a characteristic spatial, temporal and developmental stage-specific pattern during skeletogenesis. As a first step to define the tissue- and site-specific regulatory regions of the chicken matrilin-1 gene in vivo, we generated transgenic mice harboring various promoter and intronic fragments fused to the LacZ reporter gene. Histological analysis of the transgene expression pattern during ontogenic development revealed specific X-gal staining in most primordial elements of endochondral bones of transgenic mouse lines carrying either the long promoter between -2011 and +67 or the intronic fragment with a short promoter between -338 and +1819. The cartilage-specific activity of the latter transgene, however, was accompanied with variable ectopic expression pattern in neural and other tissues depending on the site of integration. The presence of both promoter upstream and intronic elements was necessary for the high level transgene activity in all chondrogenic tissues and for the extraskeletal transgene expression pattern resembling the most to that of the chicken matrilin-1 gene, e.g. expression in the eye, and lack of expression in the diminishing notochord and nucleus pulposus. The activity of the transgenes was restricted to the columnar proliferating and pre-hypertrophic chondrocytes visualized by BrdU incorporation and distribution of phosphorylated Sox9, respectively. DNA elements between -2011 and -338 also mediated ectopic LacZ expression in cells of neural crest origin. These results suggest that an interplay of modularly arranged cartilage- and neural crest-specific DNA elements control the expression of the matrilin-1 gene. The dispersal of cartilage-specific elements in the promoter upstream and intronic regions shows similarity to the transcriptional regulation of the Col11a2 gene.


Subject(s)
Extracellular Matrix Proteins/genetics , Gene Expression Regulation, Developmental , Glycoproteins/genetics , Introns , Promoter Regions, Genetic , Animals , Bone and Bones/cytology , Bone and Bones/embryology , Bone and Bones/physiology , Cartilage/cytology , Cartilage/embryology , Cartilage/physiology , Chickens , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Embryo, Nonmammalian , Extracellular Matrix Proteins/metabolism , Female , Genes, Reporter , Glycoproteins/metabolism , Matrilin Proteins , Mice , Mice, Transgenic , Pregnancy , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transgenes
9.
J Leukoc Biol ; 96(5): 907-15, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25063878

ABSTRACT

PDE2A is a dual-function PDE that is stimulated by cGMP to hydrolyze cAMP preferentially. In a two-hit model of ALI, we found previously that PDE2A decreased lung cAMP, up-regulated lung iNOS, and exacerbated ALI. Recent data suggest that macrophage iNOS expression contributes to ALI but later, promotes lung-injury resolution. However, macrophage iNOS is increased by cAMP, suggesting that PDE2A could negatively regulate macrophage iNOS expression. To test this, we examined the effects of manipulating PDE2A expression and function on LPS-induced iNOS expression in a mouse AM cell line (MH-S) and primary mouse AMs. In MH-S cells, LPS (100 ng/ml) increased PDE2A expression by 15% at 15 min and 50% at 6 h before decreasing at 24 h and 48 h. iNOS expression appeared at 6 h and remained increased 48 h post-LPS. Compared with control Ad, Ad.PDE2A-shRNA enhanced LPS-induced iNOS expression further by fourfold, an effect mimicked by the PDE2A inhibitor BAY 60-7550. Adenoviral PDE2A overexpression or treatment with ANP decreased LPS-induced iNOS expression. ANP-induced inhibition of iNOS was lost by knocking down PDE2A and was not mimicked by 8-pCPT-cGMP, a cGMP analog that does not stimulate PDE2A activity. Finally, we found that in primary AMs from LPS-treated mice, PDE2A knockdown also increased iNOS expression, consistent with the MH-S cell data. We conclude that increased AM PDE2A is an important negative regulator of macrophage iNOS expression.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Gene Expression Regulation , Macrophages, Alveolar/metabolism , Nitric Oxide Synthase Type II/genetics , Animals , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 2/antagonists & inhibitors , Cyclic Nucleotide Phosphodiesterases, Type 2/genetics , Down-Regulation , Gene Expression Regulation/drug effects , Lipopolysaccharides/pharmacology , Macrophages, Alveolar/immunology , Male , Mice , Nitric Oxide Synthase Type II/metabolism , Thionucleotides/metabolism
10.
PLoS One ; 9(9): e106092, 2014.
Article in English | MEDLINE | ID: mdl-25181540

ABSTRACT

The process of leukocyte recruitment to the airways in real time has not been extensively studied, yet airway inflammation persists as a major contributor to lung pathology. We showed previously in vivo, that neutrophils are recruited acutely to the large airways after periods of airway distension imposed by the application of positive end-expiratory pressure (PEEP). Given extensive literature implicating products of nitric oxide synthase (NOS) in lung injury after ventilatory over-distension, we questioned whether similar mechanisms exist in airway post-capillary venules. Yet, endothelial nitric oxide has been shown to be largely anti-inflammatory in other systemic venules. Using intravital microscopy to visualize post-capillary tracheal venules in anesthetized, ventilated mice, the number of adherent leukocytes was significantly decreased in eNOS-/- mice under baseline conditions (2±1 cell/60 min observation) vs wild type (WT) C57BL/6 mice (7±2 cells). After exposure to PEEP (8 cmH2O for 1 min; 5 times), adherent cells increased significantly (29±5 cells) in WT mice while eNOS-/- mice demonstrated a significantly decreased number of adherent cells (11±4 cells) after PEEP. A similar response was seen when thrombin was used as the pro-inflammatory stimulus. In addition, mouse tracheal venular endothelial cells studied in vitro after exposure to cyclic stretch (18% elongation) or thrombin both demonstrated increased p-selectin expression that was significantly attenuated by NG-nitro-L-arginine methyl ester, N-acetylcysteine amide (NACA) and excess BH4. In vivo treatment with the ROS inhibitor NACA or co-factor BH4 abolished completely the PEEP-induced leukocyte adherence. These results suggest that pro-inflammatory stimuli cause leukocyte recruitment to tracheal endothelium in part due to eNOS uncoupling.


Subject(s)
Leukocytes/enzymology , Leukocytes/pathology , Nitric Oxide Synthase Type III/metabolism , Stress, Mechanical , Trachea/pathology , Venules/pathology , Animals , Cell Adhesion/drug effects , Leukocytes/drug effects , Male , Mice, Inbred C57BL , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/deficiency , P-Selectin/metabolism , Phosphorylation/drug effects , Positive-Pressure Respiration , Reactive Oxygen Species/metabolism , Thrombin/pharmacology
11.
PLoS One ; 8(8): e71010, 2013.
Article in English | MEDLINE | ID: mdl-23967147

ABSTRACT

Severe malaria can trigger acute lung injury characterized by pulmonary edema resulting from increased endothelial permeability. However, the mechanism through which lung fluid conductance is altered during malaria remains unclear. To define the role that the scavenger receptor CD36 may play in mediating this response, C57BL/6J (WT) and CD36-/- mice were infected with P. berghei ANKA and monitored for changes in pulmonary endothelial barrier function employing an isolated perfused lung system. WT lungs demonstrated a >10-fold increase in two measures of paracellular fluid conductance and a decrease in the albumin reflection coefficient (σalb) compared to control lungs indicating a loss of barrier function. In contrast, malaria-infected CD36-/- mice had near normal fluid conductance but a similar reduction in σalb. In WT mice, lung sequestered iRBCs demonstrated production of reactive oxygen species (ROS). To determine whether knockout of CD36 could protect against ROS-induced endothelial barrier dysfunction, mouse lung microvascular endothelial monolayers (MLMVEC) from WT and CD36-/- mice were exposed to H2O2. Unlike WT monolayers, which showed dose-dependent decreases in transendothelial electrical resistance (TER) from H2O2 indicating loss of barrier function, CD36-/- MLMVEC demonstrated dose-dependent increases in TER. The differences between responses in WT and CD36-/- endothelial cells correlated with important differences in the intracellular compartmentalization of the CD36-associated Fyn kinase. Malaria infection increased total lung Fyn levels in CD36-/- lungs compared to WT, but this increase was due to elevated production of the inactive form of Fyn further suggesting a dysregulation of Fyn-mediated signaling. The importance of Fyn in CD36-dependent endothelial signaling was confirmed using in vitro Fyn knockdown as well as Fyn-/- mice, which were also protected from H2O2- and malaria-induced lung endothelial leak, respectively. Our results demonstrate that CD36 and Fyn kinase are critical mediators of the increased lung endothelial fluid conductance caused by malaria infection.


Subject(s)
Alveolar Epithelial Cells/metabolism , Blood-Air Barrier/metabolism , CD36 Antigens/metabolism , Malaria/metabolism , Plasmodium berghei , Proto-Oncogene Proteins c-fyn/metabolism , Alveolar Epithelial Cells/pathology , Animals , Blood-Air Barrier/physiopathology , CD36 Antigens/genetics , Erythrocytes/metabolism , Erythrocytes/parasitology , Gene Knockdown Techniques , Lung/metabolism , Lung/pathology , Malaria/pathology , Mice , Mice, Knockout , Permeability , Reactive Oxygen Species/metabolism , Signal Transduction
12.
Mol Cell Biol ; 31(4): 686-99, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21173167

ABSTRACT

To help uncover the mechanisms underlying the staggered expression of cartilage-specific genes in the growth plate, we dissected the transcriptional mechanisms driving expression of the matrilin-1 gene (Matn1). We show that a unique assembly of evolutionarily conserved cis-acting elements in the Matn1 proximal promoter restricts expression to the proliferative and prehypertrophic zones of the growth plate. These elements functionally interact with distal elements and likewise are capable of restricting the domain of activity of a pancartilaginous Col2a1 enhancer. The proximal elements include a Pe1 element binding the chondrogenic L-Sox5, Sox6, and Sox9 proteins, a SI element binding Nfi proteins, and an initiator Ine element binding the Sox trio and other factors. Sox9 binding to Pe1 is indispensable for functional interaction with the distal promoter. Binding of L-Sox5/Sox6 to Ine and Nfib to SI modulates Sox9 transactivation in a protein dose-dependent manner, possibly to enhance Sox9 activity in early stages of chondrogenesis and repress it at later stages. Hence, our data suggest a novel model whereby Sox and Nfi proteins bind to conserved Matn1 proximal elements and functionally interact with each other to finely tune gene expression in specific zones of the cartilage growth plate.


Subject(s)
Extracellular Matrix Proteins/genetics , Glycoproteins/genetics , Growth Plate/embryology , Growth Plate/metabolism , NFI Transcription Factors/metabolism , Promoter Regions, Genetic , SOX9 Transcription Factor/metabolism , SOXD Transcription Factors/metabolism , Animals , Animals, Genetically Modified , Base Sequence , Binding Sites/genetics , COS Cells , Cells, Cultured , Chick Embryo , Chlorocebus aethiops , Chondrocytes/metabolism , Chondrogenesis/genetics , Chondrogenesis/physiology , Conserved Sequence , DNA/genetics , DNA/metabolism , Enhancer Elements, Genetic , Evolution, Molecular , Gene Expression Regulation, Developmental , Matrilin Proteins , Molecular Sequence Data , Mutation , NFI Transcription Factors/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , SOX9 Transcription Factor/genetics , SOXD Transcription Factors/genetics , Sequence Homology, Nucleic Acid , TATA Box
13.
Am J Physiol Lung Cell Mol Physiol ; 294(4): L686-97, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18281604

ABSTRACT

Increased pulmonary endothelial cGMP was shown to prevent endothelial barrier dysfunction through activation of protein kinase G (PKG(I)). Vasodilator-stimulated phosphoprotein (VASP) has been hypothesized to mediate PKG(I) barrier protection because VASP is a cytoskeletal phosphorylation target of PKG(I) expressed in cell-cell junctions. Unphosphorylated VASP was proposed to increase paracellular permeability through actin polymerization and stress fiber bundling, a process inhibited by PKG(I)-mediated phosphorylation of Ser(157) and Ser(239). To test this hypothesis, we examined the role of VASP in the transient barrier dysfunction caused by H(2)O(2) in human pulmonary artery endothelial cell (HPAEC) monolayers studied without and with PKG(I) expression introduced by adenoviral infection (Ad.PKG). In the absence of PKG(I) expression, H(2)O(2) (100-250 microM) caused a transient increased permeability and pSer(157)-VASP formation that were both attenuated by protein kinase C inhibition. Potentiation of VASP Ser(157) phosphorylation by either phosphatase 2B inhibition with cyclosporin or protein kinase A activation with forskolin prolonged, rather than inhibited, the increased permeability caused by H(2)O(2). With Ad.PKG infection, inhibition of VASP expression with small interfering RNA exacerbated H(2)O(2)-induced barrier dysfunction but had no effect on cGMP-mediated barrier protection. In addition, expression of a Ser-double phosphomimetic mutant VASP failed to reproduce the protective effects of activated PKG(I). Finally, expression of a Ser-double phosphorylation-resistant VASP failed to interfere with the ability of cGMP/PKG(I) to attenuate H(2)O(2)-induced disruption of VE-cadherin homotypic binding. Our results suggest that VASP phosphorylation does not explain the protective effect of cGMP/PKG(I) on H(2)O(2)-induced endothelial barrier dysfunction in HPAEC.


Subject(s)
Cyclic GMP/pharmacology , Endothelium, Vascular/physiology , Phosphoproteins/metabolism , Pulmonary Artery/physiology , Vasodilator Agents/pharmacology , Colforsin/pharmacology , Cyclosporine/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Humans , Hydrogen Peroxide/pharmacology , Kinetics , Phosphoserine/metabolism , Plasmids , Pulmonary Artery/cytology , Pulmonary Artery/drug effects , RNA, Small Interfering/genetics , Transfection
14.
Am J Physiol Lung Cell Mol Physiol ; 295(6): L1056-65, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18849438

ABSTRACT

High tidal volume (HV(T)) ventilation causes pulmonary endothelial barrier dysfunction. HV(T) ventilation also increases lung nitric oxide (NO) and cGMP. NO contributes to HV(T) lung injury, but the role of cGMP is unknown. In the current study, ventilation of isolated C57BL/6 mouse lungs increased perfusate cGMP as a function of V(T). Ventilation with 20 ml/kg V(T) for 80 min increased the filtration coefficient (K(f)), an index of vascular permeability. The increased cGMP and K(f) caused by HV(T) were attenuated by nitric oxide synthase (NOS) inhibition and in lungs from endothelial NOS knockout mice. Inhibition of soluble guanylyl cyclase (sGC) in wild-type lungs (10 muM ODQ) also blocked cGMP generation and inhibited the increase in K(f), suggesting an injurious role for sGC-derived cGMP. sGC inhibition also attenuated lung Evans blue dye albumin extravasation and wet-to-dry weight ratio in an anesthetized mouse model of HV(T) injury. Additional activation of sGC (1.5 muM BAY 41-2272) in isolated lungs at 40 min increased cGMP production and K(f) in lungs ventilated with 15 ml/kg V(T). HV(T) endothelial barrier dysfunction was attenuated with a nonspecific phosphodiesterase (PDE) inhibitor (100 muM IBMX) as well as an inhibitor (10 muM BAY 60-7550) specific for the cGMP-stimulated PDE2A. Concordantly, we found a V(T)-dependent increase in lung cAMP hydrolytic activity and PDE2A protein expression with a decrease in lung cAMP concentration that was blocked by BAY 60-7550. We conclude that HV(T)-induced endothelial barrier dysfunction resulted from a simultaneous increase in NO/sGC-derived cGMP and PDE2A expression causing decreased cAMP.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 2/biosynthesis , Guanylate Cyclase/metabolism , Ventilator-Induced Lung Injury/enzymology , 1-Methyl-3-isobutylxanthine/pharmacology , Animals , Blood-Air Barrier , Cyclic AMP/genetics , Cyclic AMP/metabolism , Cyclic GMP/genetics , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 2/genetics , Disease Models, Animal , Enzyme Activation/drug effects , Enzyme Activation/genetics , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Guanylate Cyclase/genetics , Mice , Mice, Knockout , Nitric Oxide/genetics , Nitric Oxide/metabolism , Phosphodiesterase Inhibitors/pharmacology , Ventilator-Induced Lung Injury/genetics
SELECTION OF CITATIONS
SEARCH DETAIL