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(5): e0266890, 2022.
Article in English | MEDLINE | ID: mdl-35503765

ABSTRACT

Sodium-hydrogen exchangers (NHEs) tightly regulate intracellular pH (pHi), proliferation, migration and cell volume. Heterogeneity exists between pulmonary endothelial cells derived from different vascular segments, yet the activity and isoform expression of NHEs between these vascular segments has not been fully examined. Utilizing the ammonium-prepulse and recovery from acidification technique in a buffer lacking bicarbonate, pulmonary microvascular and pulmonary artery endothelial cells exhibited unique recovery rates from the acid load dependent upon the concentration of the sodium transport inhibitor, amiloride; further, pulmonary artery endothelial cells required a higher dose of amiloride to inhibit sodium-dependent acid recovery compared to pulmonary microvascular endothelial cells, suggesting a unique complement of NHEs between the different endothelial cell types. While NHE1 has been described in pulmonary endothelial cells, all NHE isoforms have not been accounted for. To address NHE expression in endothelial cells, qPCR was performed. Using a two-gene normalization approach, Sdha and Ywhag were identified for qPCR normalization and analysis of NHE isoforms between pulmonary microvascular and pulmonary artery endothelial cells. NHE1 and NHE8 mRNA were equally expressed between the two cell types, but NHE5 expression was significantly higher in pulmonary microvascular versus pulmonary artery endothelial cells, which was confirmed at the protein level. Thus, pulmonary microvascular and pulmonary artery endothelial cells exhibit unique NHE isoform expression and have a unique response to acid load revealed through recovery from cellular acidification.


Subject(s)
Amiloride , Endothelial Cells , Acids/metabolism , Amiloride/pharmacology , Endothelial Cells/metabolism , Hydrogen-Ion Concentration , Protein Isoforms/genetics , Protein Isoforms/metabolism , Sodium-Hydrogen Exchanger 1/genetics , Sodium-Hydrogen Exchangers/metabolism
2.
Am J Physiol Lung Cell Mol Physiol ; 316(4): L691-L700, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30758991

ABSTRACT

The second messenger, cAMP, is highly compartmentalized to facilitate signaling specificity. Extracellular vesicles (EVs) are submicron, intact vesicles released from many cell types that can act as biomarkers or be involved in cell-to-cell communication. Although it is well recognized that EVs encapsulate functional proteins and RNAs/miRNAs, currently it is unclear whether cyclic nucleotides are encapsulated within EVs to provide an additional second messenger compartment. Using ultracentrifugation, EVs were isolated from the culture medium of unstimulated systemic and pulmonary endothelial cells. EVs were also isolated from pulmonary microvascular endothelial cells (PMVECs) following stimulation of transmembrane adenylyl cyclase (AC) in the presence or absence of the phosphodiesterase 4 inhibitor rolipram over time. Whereas cAMP was detected in EVs isolated from endothelial cells derived from different vascular beds, it was highest in EVs isolated from PMVECs. Treatment of PMVECs with agents that increase near-membrane cAMP led to an increase in cAMP within corresponding EVs, yet there was no increase in EV number. Elevated cell cAMP, measured by whole cell measurements, peaked 15 min after treatment, yet in EVs the peak increase in cAMP was delayed until 60 min after cell stimulation. Cyclic AMP was also increased in EVs collected from the perfusate of isolated rat lungs stimulated with isoproterenol and rolipram, thus corroborating cell culture findings. When added to unperturbed confluent PMVECs, EVs containing elevated cAMP were not barrier disruptive like cytosolic cAMP but maintained monolayer resistance. In conclusion, PMVECs release EVs containing cAMP, providing an additional compartment to cAMP signaling.


Subject(s)
Cell Communication , Cyclic AMP/metabolism , Endothelial Cells/metabolism , Extracellular Vesicles/metabolism , Lung/metabolism , Second Messenger Systems , Adenylyl Cyclases/metabolism , Animals , Endothelial Cells/cytology , Lung/cytology , Male , Rats , Rats, Sprague-Dawley
3.
BMC Immunol ; 19(1): 9, 2018 02 17.
Article in English | MEDLINE | ID: mdl-29452585

ABSTRACT

BACKGROUND: The potency of T regulatory (TREG) cells to inhibit T helper (Th)-driven immune cell responses has been linked to increased intracellular cyclic-AMP (cAMP) levels of TREG cells. In an ovalbumin (OVA)-driven allergic asthma mouse model, moderate aerobic exercise increases TREG cell function in a contact-dependent manner that leads to a significant reduction in chronic inflammation and restoration of lung function. However, the mechanism, whereby exercise increases TREG function, remains unknown and was the focus of these investigations. Exercise can communicate with TREG cells by their expression of ß2-adrenergic receptors (ß2-AR). Activation of these receptors results in an increase in intracellular levels of cyclic-AMP, potentially creating a potent inhibitor of Th cell responses. RESULTS: For the allergic asthma model, female wildtype BALB/c mice were challenged with OVA, and exercised (13.5 m/min for 45 min) 3×/week for 4 weeks. TREG cells were isolated from all mouse asthma/exercise groups, including ß2-AR-/- mice, to test suppressive function and intracellular cAMP levels. In these studies, cAMP levels were increased in TREG cells isolated from exercised mice. When ß2-AR expression was absent on TREG cells, cAMP levels were significantly decreased. Correlatively, their suppressive function was compromised. Next, TREG cells from all mouse groups were tested for suppressive function after treatment with either a pharmaceutical ß2-adrenergic agonist or an effector-specific cAMP analogue. These experiments showed TREG cell function was increased when treated with either a ß2-adrenergic agonist or effector-specific cAMP analogue. Finally, female wildtype BALB/c mice were antibody-depleted of CD25+CD4+ TREG cells (anti-CD25). Twenty-four hours after TREG depletion, either ß2-AR-/- or wildtype TREG cells were adoptively transferred. Recipient mice underwent the asthma/exercise protocols. ß2-AR-/- TREG cells isolated from these mice showed no increase in TREG function in response to moderate aerobic exercise. CONCLUSION: These studies offer a novel role for ß2-AR in regulating cAMP intracellular levels that can modify suppressive function in TREG cells.


Subject(s)
Asthma/immunology , Physical Conditioning, Animal/methods , Receptors, Adrenergic, beta-2/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Asthma/metabolism , Cyclic AMP/immunology , Cyclic AMP/metabolism , Disease Models, Animal , Female , Intracellular Space/immunology , Intracellular Space/metabolism , Mice, Inbred BALB C , Mice, Transgenic , Ovalbumin/immunology , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/metabolism , T-Lymphocytes, Regulatory/metabolism
4.
Am J Physiol Lung Cell Mol Physiol ; 309(12): L1430-7, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26475732

ABSTRACT

Bacteria-induced sepsis is a common cause of pulmonary endothelial barrier dysfunction and can progress toward acute respiratory distress syndrome. Elevations in intracellular cAMP tightly regulate pulmonary endothelial barrier integrity; however, cAMP signals are highly compartmentalized: whether cAMP is barrier-protective or -disruptive depends on the compartment (plasma membrane or cytosol, respectively) in which the signal is generated. The mammalian soluble adenylyl cyclase isoform 10 (AC10) is uniquely stimulated by bicarbonate and is expressed in pulmonary microvascular endothelial cells (PMVECs). Elevated extracellular bicarbonate increases cAMP in PMVECs to disrupt the endothelial barrier and increase the filtration coefficient (Kf) in the isolated lung. We tested the hypothesis that sepsis-induced endothelial barrier disruption and increased permeability are dependent on extracellular bicarbonate and activation of AC10. Our findings reveal that LPS-induced endothelial barrier disruption is dependent on extracellular bicarbonate: LPS-induced barrier failure and increased permeability are exacerbated in elevated bicarbonate compared with low extracellular bicarbonate. The AC10 inhibitor KH7 attenuated the bicarbonate-dependent LPS-induced barrier disruption. In the isolated lung, LPS failed to increase Kf in the presence of minimal perfusate bicarbonate. An increase in perfusate bicarbonate to the physiological range (24 mM) revealed the LPS-induced increase in Kf, which was attenuated by KH7. Furthermore, in PMVECs treated with LPS for 6 h, there was a dose-dependent increase in AC10 expression. Thus these findings reveal that LPS-induced pulmonary endothelial barrier failure requires bicarbonate activation of AC10.


Subject(s)
Adenylyl Cyclases/metabolism , Bicarbonates/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Lipopolysaccharides/pharmacology , Protein Isoforms/metabolism , Pulmonary Edema/chemically induced , Animals , Capillary Permeability/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Cyclic AMP/metabolism , Cytosol/drug effects , Cytosol/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Lung/drug effects , Lung/metabolism , Male , Pulmonary Edema/metabolism , Rats, Sprague-Dawley , Respiratory Distress Syndrome/metabolism , Sepsis/metabolism
5.
Am J Physiol Lung Cell Mol Physiol ; 305(2): L185-92, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23686854

ABSTRACT

It is becoming increasingly apparent that cAMP signals within the pulmonary endothelium are highly compartmentalized, and this compartmentalization is critical to maintaining endothelial barrier integrity. Studies demonstrate that the exogenous soluble bacterial toxin, ExoY, and heterologous expression of the forskolin-stimulated soluble mammalian adenylyl cyclase (AC) chimera, sACI/II, elevate cytosolic cAMP and disrupt the pulmonary microvascular endothelial barrier. The barrier-disruptive effects of cytosolic cAMP generated by exogenous soluble ACs are in contrast to the barrier-protective effects of subplasma membrane cAMP generated by transmembrane AC, which strengthens endothelial barrier integrity. Endogenous soluble AC isoform 10 (AC10 or commonly known as sAC) lacks transmembrane domains and localizes within the cytosolic compartment. AC10 is uniquely activated by bicarbonate to generate cytosolic cAMP, yet its role in regulation of endothelial barrier integrity has not been addressed. Here we demonstrate that, within the pulmonary circulation, AC10 is expressed in pulmonary microvascular endothelial cells (PMVECs) and pulmonary artery endothelial cells (PAECs), yet expression in PAECs is lower. Furthermore, pulmonary endothelial cells selectively express bicarbonate cotransporters. While extracellular bicarbonate generates a phosphodiesterase 4-sensitive cAMP pool in PMVECs, no such cAMP response is detected in PAECs. Finally, addition of extracellular bicarbonate decreases resistance across the PMVEC monolayer and increases the filtration coefficient in the isolated perfused lung above osmolality controls. Collectively, these findings suggest that PMVECs have a bicarbonate-sensitive cytosolic cAMP pool that disrupts endothelial barrier integrity. These studies could provide an alternative mechanism for the controversial effects of bicarbonate correction of acidosis of acute respiratory distress syndrome patients.


Subject(s)
Adenylyl Cyclases/biosynthesis , Bicarbonates/metabolism , Blood-Air Barrier/enzymology , Endothelium/enzymology , Gene Expression Regulation, Enzymologic , Acidosis/enzymology , Animals , Blood-Air Barrier/pathology , Cells, Cultured , Cyclic AMP/metabolism , Endothelium/pathology , Humans , Rats , Respiratory Distress Syndrome/enzymology
6.
Am J Physiol Cell Physiol ; 302(6): C839-52, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22116306

ABSTRACT

Cyclic AMP signals encode information required to differentially regulate a wide variety of cellular responses; yet it is not well understood how information is encrypted within these signals. An emerging concept is that compartmentalization underlies specificity within the cAMP signaling pathway. This concept is based on a series of observations indicating that cAMP levels are distinct in different regions of the cell. One such observation is that cAMP production at the plasma membrane increases pulmonary microvascular endothelial barrier integrity, whereas cAMP production in the cytosol disrupts barrier integrity. To better understand how cAMP signals might be compartmentalized, we have developed mathematical models in which cellular geometry as well as total adenylyl cyclase and phosphodiesterase activities were constrained to approximate values measured in pulmonary microvascular endothelial cells. These simulations suggest that the subcellular localizations of adenylyl cyclase and phosphodiesterase activities are by themselves insufficient to generate physiologically relevant cAMP gradients. Thus, the assembly of adenylyl cyclase, phosphodiesterase, and protein kinase A onto protein scaffolds is by itself unlikely to ensure signal specificity. Rather, our simulations suggest that reductions in the effective cAMP diffusion coefficient may facilitate the formation of substantial cAMP gradients. We conclude that reductions in the effective rate of cAMP diffusion due to buffers, structural impediments, and local changes in viscosity greatly facilitate the ability of signaling complexes to impart specificity within the cAMP signaling pathway.


Subject(s)
Cell Compartmentation/physiology , Cyclic AMP/metabolism , Endothelial Cells/metabolism , Models, Biological , Signal Transduction/physiology , Adenylyl Cyclases/metabolism , Animals , Cell Culture Techniques , Cell Membrane/metabolism , Computer Simulation , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytosol/metabolism , Endothelial Cells/cytology , Lung/blood supply , Lung/cytology , Phosphoric Diester Hydrolases/metabolism , Rats , Receptors, G-Protein-Coupled/physiology
7.
Am J Physiol Lung Cell Mol Physiol ; 301(1): L117-24, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21478251

ABSTRACT

Transmembrane adenylyl cyclase (AC) generates a cAMP pool within the subplasma membrane compartment that strengthens the endothelial cell barrier. This cAMP signal is steered toward effectors that promote junctional integrity and is inactivated before it accesses microtubules, where the cAMP signal causes phosphorylation of tau, leading to microtubule disassembly and barrier disruption. During infection, Pseudomonas aeruginosa uses a type III secretion system to inject a soluble AC, ExoY, into the cytosol of pulmonary microvascular endothelial cells. ExoY generates a cAMP signal that disrupts the endothelial cell barrier. We tested the hypothesis that this ExoY-dependent cAMP signal causes phosphorylation of tau, without inducing phosphorylation of membrane effectors that strengthen endothelial barrier function. To approach this hypothesis, we first discerned the membrane compartment in which endogenous transmembrane AC6 resides. AC6 was resolved in caveolin-rich lipid raft fractions with calcium channel proteins and the cell adhesion molecules N-cadherin, E-cadherin, and activated leukocyte adhesion molecule. VE-cadherin was excluded from the caveolin-rich fractions and was detected in the bulk plasma membrane fractions. The actin binding protein, filamin A, was detected in all membrane fractions. Isoproterenol activation of ACs promoted filamin phosphorylation, whereas thrombin inhibition of AC6 reduced filamin phosphorylation within the membrane fraction. In contrast, ExoY produced a cAMP signal that did not cause filamin phosphorylation yet induced tau phosphorylation. Hence, our data indicate that cAMP signals are strictly compartmentalized; whereas cAMP emanating from transmembrane ACs activates barrier-enhancing targets, such as filamin, cAMP emanating from soluble ACs activates barrier-disrupting targets, such as tau.


Subject(s)
Adenylyl Cyclases/metabolism , Cell Membrane/enzymology , Contractile Proteins/metabolism , Cytosol/enzymology , Microfilament Proteins/metabolism , Animals , Bacterial Proteins/pharmacology , Calcium Channels/metabolism , Caveolin 1/metabolism , Cell Adhesion Molecules , Cell Compartmentation/drug effects , Cell Membrane/drug effects , Cyclic AMP/metabolism , Cytosol/drug effects , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Enzyme Activation/drug effects , Filamins , Glucosyltransferases/pharmacology , Isoproterenol/pharmacology , Lung/blood supply , Membrane Microdomains/drug effects , Membrane Microdomains/enzymology , Microvessels/cytology , Models, Biological , ORAI1 Protein , Phosphorylation/drug effects , Rats , Subcellular Fractions/drug effects , Subcellular Fractions/enzymology , TRPC Cation Channels/metabolism , Thrombin/pharmacology , tau Proteins/metabolism
8.
Am J Physiol Lung Cell Mol Physiol ; 300(5): L667-78, 2011 May.
Article in English | MEDLINE | ID: mdl-21335524

ABSTRACT

The presence of excess fluid in the interstitium and air spaces of the lung presents severe restrictions to gas exchange. The pulmonary endothelial barrier regulates the flux of fluid and plasma proteins from the vascular space into the underlying tissue. The integrity of this endothelial barrier is dynamically regulated by transitions in cAMP (3',5'-cyclic adenosine monophosphate), which are synthesized in discrete subcellular compartments. Cyclic AMP generated in the subplasma membrane compartment acts through PKA and Epac (exchange protein directly activated by cAMP) to tighten cell adhesions, strengthen cortical actin, reduce actomyosin contraction, and decrease permeability. Confining cAMP within the subplasma membrane space is critical to its barrier-protective properties. When cAMP escapes the near membrane compartment and gains access to the cytosolic compartment, or when soluble adenylyl cyclases generate cAMP within the cytosolic compartment, this second messenger activates established cytosolic cAMP signaling cascades to perturb the endothelial barrier through PKA-mediated disruption of microtubules. Thus the concept of cAMP compartmentalization in endothelial barrier regulation is gaining momentum and new possibilities are being unveiled for cytosolic cAMP signaling with the emergence of the bicarbonate-regulated mammalian soluble adenylyl cyclase (sAC or AC10).


Subject(s)
Cyclic AMP/metabolism , Guanine Nucleotide Exchange Factors/physiology , Lung/metabolism , Adenylyl Cyclases/metabolism , Bicarbonates/pharmacology , Capillary Permeability/drug effects , Capillary Permeability/physiology , Cell Adhesion Molecules/physiology , Cell Compartmentation/physiology , Cyclic AMP-Dependent Protein Kinases/physiology , Cytosol/metabolism , Endothelium/drug effects , Endothelium/physiology , Microfilament Proteins/physiology , Phosphoproteins/physiology , Pulmonary Edema/etiology , Second Messenger Systems/physiology
9.
Circ Res ; 98(5): 675-81, 2006 Mar 17.
Article in English | MEDLINE | ID: mdl-16469954

ABSTRACT

Subtle elevations in cAMP localized to the plasma membrane intensely strengthen endothelial barrier function. Paradoxically, pathogenic bacteria insert adenylyl cyclases (ACs) into eukaryotic cells generating a time-dependent cytosolic cAMP-increase that disrupts rather than strengthens the endothelial barrier. These findings bring into question whether membrane versus cytosolic AC activity dominates in control of cell adhesion. To address this problem, a mammalian forskolin-sensitive soluble AC (sACI/II) was expressed in pulmonary microvascular endothelial cells. Forskolin stimulated this sACI/II construct generating a small cytosolic cAMP-pool that was not regulated by phosphodiesterases or Galphas. Whereas forskolin simultaneously activated the sACI/II construct and endogenous transmembrane ACs, the modest sACI/II activity overwhelmed the barrier protective effects of plasma membrane activity to induce endothelial gap formation. Retargeting sACI/II to the plasma membrane retained AC activity but protected the endothelial cell barrier. These findings demonstrate for the first time that the intracellular location of cAMP synthesis critically determines its physiological outcome.


Subject(s)
Adenylyl Cyclases/physiology , Capillary Permeability , Cyclic AMP/physiology , Endothelial Cells/metabolism , Adenylyl Cyclases/analysis , Animals , Bacterial Proteins/physiology , Cell Membrane/enzymology , Cells, Cultured , Colforsin/pharmacology , Cytosol/enzymology , GTP-Binding Protein alpha Subunits/physiology , Glucosyltransferases/physiology , Lung/blood supply , Rats , Signal Transduction
10.
Circ Res ; 96(8): 856-63, 2005 Apr 29.
Article in English | MEDLINE | ID: mdl-15790951

ABSTRACT

Store-operated calcium (SOC) entry is sufficient to disrupt the extra-alveolar, but not the alveolar, endothelial cell barrier. Mechanism(s) underlying such insensitivity to transitions in cytosolic calcium ([Ca2+]i) in microvascular endothelial cells are unknown. Depletion of stored Ca2+ activates a larger SOC entry response in extra-alveolar (pulmonary artery; PAECs) than alveolar (pulmonary microvascular; PMVECs) endothelial cells. In vivo permeation studies revealed that Ca2+ store depletion activates similar nonselective cationic conductances in PAECs and PMVECs, while only PAECs possess the calcium-selective, store-operated Ca2+ entry current, I(SOC). Pretreatment with the type 4 phosphodiesterase inhibitor, rolipram, abolished thapsigargin-activated I(SOC) in PAECs, and revealed I(SOC) in PMVECs. Rolipram pretreatment shifted the thapsigargin-induced fluid leak site from extra-alveolar to alveolar vessels in the intact pulmonary circulation. Thus, our results indicate I(SOC) provides a [Ca2+]i source that is needed to disrupt the endothelial cell barrier, and demonstrate that intracellular events controlling I(SOC) activation coordinate the site-specific vascular response to inflammation.


Subject(s)
Calcium Channels/physiology , Calcium/metabolism , Capillary Permeability , Endothelial Cells/metabolism , Adenylyl Cyclases/physiology , Animals , Cyclic AMP/metabolism , Ion Channels/physiology , Lanthanum/pharmacology , Models, Molecular , Rats , Rolipram/pharmacology , TRPC Cation Channels , Thapsigargin/pharmacology
11.
Circ Res ; 95(2): 196-203, 2004 Jul 23.
Article in English | MEDLINE | ID: mdl-15192021

ABSTRACT

Mammalian transmembrane adenylyl cyclases synthesize a restricted plasmalemmal cAMP pool that is intensely endothelial barrier protective. Bacteria have devised mechanisms of transferring eukaryotic factor-dependent adenylyl cyclases into mammalian cells. Pseudomonas aeruginosa ExoY is one such enzyme that catalyzes cytosolic cAMP synthesis, with unknown function. Pseudomonas aeruginosa genetically modified to introduce only the ExoY toxin elevated cAMP 800-fold in pulmonary microvascular endothelial cells over 4 hours, whereas a catalytically deficient (ExoY(K81M)) strain did not increase cAMP. ExoY-derived cAMP was localized to a cytosolic microdomain not regulated by phosphodiesterase activity. In contrast to the barrier-enhancing actions of plasmalemmal cAMP, the ExoY cytosolic cAMP pool induced endothelial gap formation and increased the filtration coefficient in the isolated perfused lung. These findings collectively illustrate a previously unrecognized mechanism of hyperpermeability induced by rises in cytosolic cAMP.


Subject(s)
Bacterial Proteins/physiology , Cyclic AMP/physiology , Endothelial Cells/microbiology , Endothelium, Vascular/cytology , Glucosyltransferases/physiology , Lung/blood supply , Pseudomonas aeruginosa/enzymology , Second Messenger Systems/physiology , 3',5'-Cyclic-AMP Phosphodiesterases/physiology , Adenylyl Cyclase Inhibitors , Adenylyl Cyclases/analysis , Adenylyl Cyclases/metabolism , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Capillaries/cytology , Catalysis , Cell Compartmentation , Cell Membrane Permeability , Cells, Cultured/metabolism , Cells, Cultured/microbiology , Colforsin/pharmacology , Cyclic AMP/biosynthesis , Cyclic Nucleotide Phosphodiesterases, Type 4 , Cytosol/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/microbiology , Glucosyltransferases/chemistry , Glucosyltransferases/genetics , Intercellular Junctions/drug effects , Male , Phosphodiesterase Inhibitors/pharmacology , Pseudomonas aeruginosa/physiology , Rats , Rats, Sprague-Dawley , Rolipram/pharmacology , Second Messenger Systems/drug effects , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...