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










Publication year range
1.
PLoS One ; 17(11): e0276462, 2022.
Article in English | MEDLINE | ID: mdl-36413536

ABSTRACT

The long-sought-after "magic bullet" in systemic therapy remains unrealized for disease targets existing inside most tissues, theoretically because vascular endothelium impedes passive tissue entry and full target engagement. We engineered the first "dual precision" bispecific antibody with one arm pair to precisely bind to lung endothelium and drive active delivery and the other to precisely block TGF-ß effector function inside lung tissue. Targeting caveolae for transendothelial pumping proved essential for delivering most of the injected intravenous dose precisely into lungs within one hour and for enhancing therapeutic potency by >1000-fold in a rat pneumonitis model. Ultra-low doses (µg/kg) inhibited inflammatory cell infiltration, edema, lung tissue damage, disease biomarker expression and TGF-ß signaling. The prodigious benefit of active vs passive transvascular delivery of a precision therapeutic unveils a new promising drug design, delivery and therapy paradigm ripe for expansion and clinical testing.


Subject(s)
Antibodies, Bispecific , Caveolae , Rats , Animals , Caveolae/metabolism , Transforming Growth Factor beta/metabolism , Antibodies, Bispecific/metabolism , Lung/metabolism , Endothelium, Vascular/metabolism
2.
Genomics ; 112(6): 4769-4776, 2020 11.
Article in English | MEDLINE | ID: mdl-32882326

ABSTRACT

The several virulence and drug-resistance mechanisms of Pseudomonas aeruginosa responsible for poor clinical outcomes in keratitis patients remain largely unknown. Here, we investigated the distribution of virulence factors and drug resistance by genes, mutations, efflux-pump systems of P. aeruginosa strains from keratitis patients with different clinical outcomes, included of whole-genome sequenced and annotated our five P. aeruginosa strains. Of the large number of virulence genes detected in all the genomes, MDR/XDR strains carry exoU and non-MDR strains carry exoS exotoxin of the type III secretion system, considered as main contributors of keratitis pathogenesis. However, several strain-specific virulence and resistance genes were detected in keratitis strains with poor outcome. Mainly, the flagellar genes fliC and fliD detected in the exoS carrying strains, reported to alter the host immune response, might impact the clinical outcome. This study may provide new insights into the genome of ocular strains and requires further functional studies.


Subject(s)
Keratitis/microbiology , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/genetics , Drug Resistance, Bacterial/genetics , Genome, Bacterial , Genomics , Humans , Microbial Sensitivity Tests , Virulence Factors/genetics
3.
JCI Insight ; 2(11)2017 Jun 02.
Article in English | MEDLINE | ID: mdl-28570277

ABSTRACT

Pulmonary function is dependent upon the precise regulation of alveolar surfactant. Alterations in pulmonary surfactant concentrations or function impair ventilation and cause tissue injury. Identification of the molecular pathways that sense and regulate endogenous alveolar surfactant concentrations, coupled with the ability to pharmacologically modulate them both positively and negatively, would be a major therapeutic advance for patients with acute and chronic lung diseases caused by disruption of surfactant homeostasis. The orphan adhesion GPCR GPR116 (also known as Adgrf5) is a critical regulator of alveolar surfactant concentrations. Here, we show that human and mouse GPR116 control surfactant secretion and reuptake in alveolar type II (AT2) cells by regulating guanine nucleotide-binding domain α q and 11 (Gq/11) signaling. Synthetic peptides derived from the ectodomain of GPR116 activated Gq/11-dependent inositol phosphate conversion, calcium mobilization, and cortical F-actin stabilization to inhibit surfactant secretion. AT2 cell-specific deletion of Gnaq and Gna11 phenocopied the accumulation of surfactant observed in Gpr116-/- mice. These data provide proof of concept that GPR116 is a plausible therapeutic target to modulate endogenous alveolar surfactant pools to treat pulmonary diseases associated with surfactant dysfunction.

4.
Am J Pathol ; 187(4): 921-935, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28188112

ABSTRACT

Activation of plasma membrane receptors initiates compartmentalized second messenger signaling. Whether this compartmentalization facilitates the preferential intercellular diffusion of specific second messengers is unclear. Toward this, the receptor-mediated agonist, thrombin, was instilled into microvessels in a restricted region of isolated blood-perfused mouse lungs. Subsequently, the thrombin-induced increase in endothelial F-actin was determined using confocal fluorescence microscopy. Increased F-actin was evident in microvessels directly treated with thrombin and in those located in adjoining thrombin-free regions. This increase was abrogated by inhibiting inositol trisphosphate-mediated calcium release with Xestospongin C (XeC). XeC also inhibited the thrombin-induced increase in the amplitude of endothelial cytosolic Ca2+ oscillations. Instillation of thrombin and XeC into adjacent restricted regions increased F-actin in microvessels in the thrombin-treated and adjacent regions but not in those in the XeC-treated region. Thus, inositol trisphosphate, and not calcium, diffused interendothelially to the spatially remote thrombin-free microvessels. Thus, activation of plasma membrane receptors increased the ambit of inflammatory responses via a second messenger different from that used by stimuli that induce cell-wide increases in second messengers. Thrombin however failed to induce the spatially extensive response in microvessels of mice lacking endothelial connexin43, suggesting a role for connexin43 gap junctions. Compartmental second messenger signaling and interendothelial communication define the specific second messenger involved in exacerbating proinflammatory responses to receptor-mediated agonists.


Subject(s)
Inositol 1,4,5-Trisphosphate/metabolism , Lung/blood supply , Microvessels/metabolism , Thrombin/pharmacology , Actins/metabolism , Animals , Calcium/metabolism , Cell Communication/drug effects , Connexin 43/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Mice, Inbred C57BL , Microvessels/drug effects , Permeability/drug effects , Stress Fibers/drug effects , Stress Fibers/metabolism
5.
Am J Physiol Lung Cell Mol Physiol ; 309(6): L584-92, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26163513

ABSTRACT

Endothelial barrier restoration reverses microvessel hyperpermeability and facilitates recovery from lung injury. Because inhibiting connexin 43 (Cx43)-dependent interendothelial communication blunts hyperpermeability in single microvessels, we determined whether endothelial Cx43 levels correlate with changes in microvessel permeability during recovery from lung injury. Toward this, bacterial endotoxin was instilled intratracheally into rat lungs, and at different durations postinstillation the lungs were isolated and blood perfused. Microvessel Cx43 expression was quantified by in situ immunofluorescence and microvessel permeability via a fluorescence method. To supplement the immunofluorescence data, protein levels were determined by immunoblots of lung tissue from endotoxin-instilled rats. Immunofluorescence and immunoblot together revealed that both Cx43 expression and microvessel permeability increased above baseline within a few hours after endotoxin instillation but declined progressively over the next few days. On day 5 postendotoxin, microvessel Cx43 declined to negligible levels, resulting in complete absence of intermicrovessel communication determined by photolytic uncaging of Ca(2+). However, by day 14, both Cx43 expression and microvessel permeability returned to baseline levels. In contrast to Cx43, expression of microvessel vascular endothelial (VE)-cadherin, a critical determinant of vascular barrier integrity, exhibited an inverse trend by initially declining below baseline and then returning to baseline at a longer duration. Knockdown of vascular Cx43 by tail vein injection of Cx43 shRNA increased VE-cadherin expression, suggesting that reduction in Cx43 levels may modulate VE-cadherin levels in lung microvessels. Together, the data suggest that endotoxin challenge initiates interrelated changes in microvessel Cx43, VE-cadherin, and microvessel permeability, with changes in Cx43 temporally leading the other responses.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Connexin 43/metabolism , Lung/immunology , Microvessels/metabolism , Animals , Capillary Permeability , Cell Movement , Cell Proliferation , Cells, Cultured , Chemokine CXCL2/antagonists & inhibitors , Chemokine CXCL2/metabolism , Connexin 43/genetics , Lipopolysaccharides/pharmacology , Lung/blood supply , Lung/growth & development , Lung/metabolism , Mice, Inbred C57BL , NF-kappa B/metabolism , Neovascularization, Physiologic/immunology , STAT1 Transcription Factor/metabolism , Signal Transduction
6.
Cardiovasc Pathol ; 24(1): 49-55, 2015.
Article in English | MEDLINE | ID: mdl-25294342

ABSTRACT

Chronic administration of Nω-nitro-L-arginine methyl ester (L-NAME) in rats is a chemical method to study the induction and progression of nitric oxide (NO) deficiency-induced endothelial dysfunction. Male Wistar rats received L-NAME (50 mg/kg/day in drinking water) or no drug for 6 weeks. Mean arterial pressure (MAP) was measured on Day 43 by carotid artery cannulation. Plasma interleukin 1ß (IL-1ß) level was measured by enzyme-linked immunosorbent assay. Aorta and carotid artery were isolated for determination of basal nitrite, cGMP production, soluble guanylylcyclase (sGC) activity, phosphodiesterase-5 (PDE5) activity, and dimethylarginine dimethylaminohydrolase (DDAH) activity. mRNA expression studies were done by real time-polymerase chain reaction. L-NAME induced an increase in MAP and plasma IL-1ß. The treatment had varied effect on endothelial nitric oxide synthase (eNOS), sGC, and PDE5 but showed an increase in inducible NOS (iNOS) mRNA expression and plasma asymmetric dimethyl arginine levels. Basal nitrite, cGMP levels, sGC activity, and DDAH activity were significantly decreased in the tissues. Brief incubation of tissues in vitro with 1400 W, a specific iNOS blocker, partially reversed sGC activity, and cGMP levels. The results of this study showed that L-NAME-mediated inhibition of eNOS is only partially responsible for the vascular pathology observed in this model. Secondary effects that include an increase in iNOS and a decrease in DDAH activity are likely to be the causative factors for the progression of vascular dysfunction.


Subject(s)
Amidohydrolases/metabolism , Aorta/metabolism , Carotid Arteries/metabolism , Hypertension/enzymology , Nitric Oxide Synthase Type II/metabolism , Animals , Disease Models, Animal , Enzyme Inhibitors/toxicity , Enzyme-Linked Immunosorbent Assay , Hypertension/chemically induced , Male , NG-Nitroarginine Methyl Ester/toxicity , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction
7.
J Vis Exp ; (88): e51552, 2014 Jun 30.
Article in English | MEDLINE | ID: mdl-25045895

ABSTRACT

The isolated blood-perfused lung preparation is widely used to visualize and define signaling in single microvessels. By coupling this preparation with real time imaging, it becomes feasible to determine permeability changes in individual pulmonary microvessels. Herein we describe steps to isolate rat lungs and perfuse them with autologous blood. Then, we outline steps to infuse fluorophores or agents via a microcatheter into a small lung region. Using these procedures described, we determined permeability increases in rat lung microvessels in response to infusions of bacterial lipopolysaccharide. The data revealed that lipopolysaccharide increased fluid leak across both venular and capillary microvessel segments. Thus, this method makes it possible to compare permeability responses among vascular segments and thus, define any heterogeneity in the response. While commonly used methods to define lung permeability require postprocessing of lung tissue samples, the use of real time imaging obviates this requirement as evident from the present method. Thus, the isolated lung preparation combined with real time imaging offers several advantages over traditional methods to determine lung microvascular permeability, yet is a straightforward method to develop and implement.


Subject(s)
Lung/blood supply , Microscopy, Fluorescence/methods , Models, Animal , Animals , Capillary Permeability , Dextrans/chemistry , Dextrans/pharmacokinetics , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/chemistry , Fluorescein-5-isothiocyanate/pharmacokinetics , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacokinetics , Lung/surgery , Male , Microscopy, Fluorescence/instrumentation , Perfusion/methods , Rats , Rats, Sprague-Dawley
8.
PLoS One ; 8(5): e63465, 2013.
Article in English | MEDLINE | ID: mdl-23675486

ABSTRACT

The pulmonary microvasculature plays a critical role in endotoxin-induced acute lung injury. However, the relevant signaling remain unclear. Specifically the role of endothelial Ca(2+) in the induction of endotoxin-mediated responses in lung microvessels remains undefined. Toward elucidating this, we used the isolated blood-perfused rat lung preparation. We loaded microvessels with the Ca(2+) indicator, Fura 2 AM and then determined Ca(2+) responses to infusions of lipopolysaccharide (LPS) into the microvessels. LPS induced a more than two-fold increase in the amplitude of cytosolic Ca(2+) oscillations. Inhibiting inositol 1,4,5 trisphosphate receptors on endoplasmic reticulum (ER) Ca(2+) stores with Xestospongin C (XeC), blocked the LPS-induced increase in the Ca(2+) oscillation amplitude. However, XeC did not affect entry of external Ca(2+) via plasma membrane Ca(2+) channels in lung microvascular endothelial cells. This suggested that LPS augmented the oscillations via release of Ca(2+) from ER stores. In addition, XeC also blocked LPS-mediated activation and nuclear translocation of nuclear factor-kappa B in lung microvessels. Further, inhibiting ER Ca(2+) release blunted increases in intercellular adhesion molecule-1 expression and retention of naïve leukocytes in LPS-treated microvessels. Taken together, the data suggest that LPS-mediated Ca(2+) release from ER stores underlies nuclear factor-kappa B activation and downstream inflammatory signaling in lung microvessels. Thus, we show for the first time a role for inositol 1,4,5 trisphosphate-mediated ER Ca(2+) release in the induction of LPS responses in pulmonary microvascular endothelium. Mechanisms that blunt this signaling may mitigate endotoxin-induced morbidity.


Subject(s)
Calcium/metabolism , Cytosol/metabolism , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Lung/metabolism , Microvessels/drug effects , Microvessels/metabolism , Animals , Calcium Signaling/drug effects , Endoplasmic Reticulum/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Enzyme Activation/drug effects , Inflammation/genetics , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Leukocytes/drug effects , Leukocytes/metabolism , Lung/drug effects , Lung/pathology , Male , NF-kappa B/metabolism , Rats
9.
Microvasc Res ; 83(3): 323-31, 2012 May.
Article in English | MEDLINE | ID: mdl-22342350

ABSTRACT

Endotoxemia, a major feature of sepsis, is a common cause of acute lung injury and initiates rapid accumulation of leukocytes in the lung vasculature. Endothelial mechanisms that underlie this accumulation remain unclear, as current experimental models of endotoxemia are less suitable for targeted activation of the endothelium. Toward elucidating this, we used the isolated blood-perfused rat lung preparation. With a microcatheter inserted through a left atrial cannula, we cleared blood cells from a small lung region and then infused lipopolysaccharide (LPS) into microvessels. After a Ringer's wash to remove residual LPS, we infused fluorescently-labeled autologous leukocytes and imaged their transit through the treated microvessels. Image analysis revealed that leukocytes infused 90 min after LPS treatment were retained more in treated venules and capillaries than untreated vessels. Further, pretreatment with either the intercellular adhesion molecule-1 (ICAM-1) mAb or polymyxin-B blunted LPS-induced leukocyte retention in both microvessel segments. In addition, retention of leukocytes treated ex vivo with LPS in LPS-treated microvessels was higher compared to retention of untreated leukocytes. In situ immunofluorescence experiments revealed that LPS significantly increased microvessel ICAM-1 expression at 90 min post treatment. Polymyxin pretreatment inhibited this increase. Taken together, the data suggest that LPS increased leukocyte retention in both venules and capillaries and this response was mediated by the increased expression of endothelial ICAM-1. Thus, endothelial mechanisms may themselves play a major role in LPS-induced leukocyte retention in lung microvessels. Blunting the endothelial responses may mitigate endotoxin-induced morbidity.


Subject(s)
Endothelium, Vascular/pathology , Leukocytes/cytology , Lipopolysaccharides/metabolism , Lung/blood supply , Microcirculation , Animals , Capillaries/pathology , Diagnostic Imaging/methods , Fluorescent Dyes/pharmacology , Humans , Intercellular Adhesion Molecule-1/biosynthesis , L-Selectin/biosynthesis , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence/methods , Perfusion , Polymyxin B/biosynthesis , Rats , Rats, Sprague-Dawley , Signal Transduction , Time Factors
10.
Am J Respir Cell Mol Biol ; 46(4): 461-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22052879

ABSTRACT

Both hyperoxia and mechanical ventilation can independently cause lung injury. In combination, these insults produce accelerated and severe lung injury. We recently reported that pre-exposure to hyperoxia for 12 hours, followed by ventilation with large tidal volumes, induced significant lung injury and epithelial cell apoptosis compared with either stimulus alone. We also reported that such injury and apoptosis are inhibited by antioxidant treatment. In this study, we hypothesized that apoptosis signal-regulating kinase-1 (ASK-1), a redox-sensitive, mitogen-activated protein kinase kinase kinase, plays a role in lung injury and apoptosis in this model. To determine the role of ASK-1 in lung injury, the release of inflammatory mediators and apoptosis, attributable to 12 hours of hyperoxia, were followed by large tidal volume mechanical ventilation with hyperoxia. Wild-type and ASK-1 knockout mice were subjected to hyperoxia (Fi(O(2)) = 0.9) for 12 hours before 4 hours of large tidal mechanical ventilation (tidal volume = 25 µl/g) with hyperoxia, and were compared with nonventilated control mice. Lung injury, apoptosis, and cytokine release were measured. The deletion of ASK-1 significantly inhibited lung injury and apoptosis, but did not affect the release of inflammatory mediators, compared with the wild-type mice. ASK-1 is an important regulator of lung injury and apoptosis in this model. Further study is needed to determine the mechanism of lung injury and apoptosis by ASK-1 and its downstream mediators in the lung.


Subject(s)
MAP Kinase Kinase Kinase 5/genetics , MAP Kinase Kinase Kinase 5/metabolism , Ventilator-Induced Lung Injury/enzymology , Ventilator-Induced Lung Injury/prevention & control , Animals , Apoptosis/genetics , Cytokines/metabolism , Disease Models, Animal , Enzyme Activation , Epithelial Cells/pathology , Female , Hyperoxia/enzymology , Inflammation Mediators/metabolism , Male , Mice , Mice, Knockout , Pulmonary Alveoli/pathology , Ventilator-Induced Lung Injury/pathology
11.
Shock ; 36(1): 76-82, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21412183

ABSTRACT

Hyporeactivity to vasoconstrictors is one of the clinical manifestations of sepsis in man and experimental animals. The objective of the investigation was to examine whether atorvastatin can prevent hyporeactivity to norepinephrine (NE) in mouse aorta in sepsis, and if so, what are the mechanisms involved. Sepsis in mice was induced by cecal ligation and puncture. The aorta was harvested for tension experiment, nitric oxide (NO) and cyclic guanosine monophosphate measurements, and inducible NO synthase (iNOS) and α(1D)-adrenoceptor mRNA expression studies. In comparison with sham-operated controls, sepsis significantly decreased the contractile response to NE in the mouse aorta. Pretreatment with atorvastatin of septic animals completely restored NE-induced contractions to levels similar to those of sham-operated controls and significantly increased survival time and mean arterial pressure. Atorvastatin also attenuated iNOS-induced overproduction of NO, as well as iNOS mRNA expression. Accordingly, hyporeactivity to NE was not evident in tissues pretreated with selective iNOS inhibitor 1400W in sepsis. Although basal cyclic guanosine monophosphate accumulation in the aorta was reduced in sepsis, pretreatment of the tissues with soluble guanylyl cyclase inhibitor 1H-(1,2,4)oxadiazolo(4,3-a)quinoxalin-1-one (ODQ) partially restored the reactivity to NE. Interestingly, hyporeactivity to NE in sepsis was associated with a decreased α(1D)-adrenoceptor mRNA expression in the mouse aorta. Atorvastatin pretreatment, however, prevented the decrease in α(1D)-adrenoceptor mRNA expression in septic animals. In conclusion, atorvastatin seems to prevent hyporeactivity to vasoconstrictor NE in the aorta from septic mice through attenuation of overproduction of NO as well as improved α(1D)-adrenoceptor mRNA expression. The findings of the present study may explain the beneficial effects of atorvastatin on improved hemodynamic functions in sepsis.


Subject(s)
Heptanoic Acids/therapeutic use , Nitric Oxide/metabolism , Norepinephrine/adverse effects , Pyrroles/therapeutic use , Receptors, Adrenergic, alpha-1/genetics , Sepsis/drug therapy , Vasoconstrictor Agents/adverse effects , Animals , Atorvastatin , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Mice , Norepinephrine/therapeutic use , Oxadiazoles/pharmacology , Polymerase Chain Reaction , Quinoxalines/pharmacology , RNA, Messenger , Vasoconstrictor Agents/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...