Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Nat Commun ; 9(1): 816, 2018 02 26.
Article in English | MEDLINE | ID: mdl-29483579

ABSTRACT

Myocardial ischemia-reperfusion injury (IRI) leads to the stabilization of the transcription factors hypoxia-inducible factor 1-alpha (HIF1-alpha) and hypoxia-inducible factor 2-alpha (HIF2-alpha). While previous studies implicate HIF1-alpha in cardioprotection, the role of HIF2-alpha remains elusive. Here we show that HIF2-alpha induces the epithelial growth factor amphiregulin (AREG) to elicit cardioprotection in myocardial IRI. Comparing mice with inducible deletion of Hif1a or Hif2a in cardiac myocytes, we show that loss of Hif2-alpha increases infarct sizes. Microarray studies in genetic models or cultured human cardiac myocytes implicate HIF2-alpha in the myocardial induction of AREG. Likewise, AREG increases in myocardial tissues from patients with ischemic heart disease. Areg deficiency increases myocardial IRI, as does pharmacologic inhibition of Areg signaling. In contrast, treatment with recombinant Areg provides cardioprotection and reconstitutes mice with Hif2a deletion. These studies indicate that HIF2-alpha induces myocardial AREG expression in cardiac myocytes, which increases myocardial ischemia tolerance.


Subject(s)
Amphiregulin/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Myocardial Reperfusion Injury/genetics , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Amphiregulin/metabolism , Amphiregulin/pharmacology , Animals , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Integrases/genetics , Integrases/metabolism , Male , Mice , Mice, Transgenic , Microarray Analysis , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/prevention & control , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Myosins/genetics , Myosins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction
2.
Sci Transl Med ; 9(408)2017 Sep 20.
Article in English | MEDLINE | ID: mdl-28931657

ABSTRACT

Intercellular transfer of microRNAs can mediate communication between critical effector cells. We hypothesized that transfer of neutrophil-derived microRNAs to pulmonary epithelial cells could alter mucosal gene expression during acute lung injury. Pulmonary-epithelial microRNA profiling during coculture of alveolar epithelial cells with polymorphonuclear neutrophils (PMNs) revealed a selective increase in lung epithelial cell expression of microRNA-223 (miR-223). Analysis of PMN-derived supernatants showed activation-dependent release of miR-223 and subsequent transfer to alveolar epithelial cells during coculture in vitro or after ventilator-induced acute lung injury in mice. Genetic studies indicated that miR-223 deficiency was associated with severe lung inflammation, whereas pulmonary overexpression of miR-223 in mice resulted in protection during acute lung injury induced by mechanical ventilation or by infection with Staphylococcus aureus Studies of putative miR-223 gene targets implicated repression of poly(adenosine diphosphate-ribose) polymerase-1 (PARP-1) in the miR-223-dependent attenuation of lung inflammation. Together, these findings suggest that intercellular transfer of miR-223 from neutrophils to pulmonary epithelial cells may dampen acute lung injury through repression of PARP-1.


Subject(s)
Acute Lung Injury/genetics , Acute Lung Injury/pathology , Epithelial Cells/metabolism , Lung/pathology , MicroRNAs/metabolism , Neutrophils/metabolism , Animals , Cell Communication , Gene Knockdown Techniques , Humans , Mice, Inbred C57BL , MicroRNAs/genetics , Nanoparticles/chemistry , Pneumonia/genetics , Pneumonia/pathology , Poly(ADP-ribose) Polymerases/metabolism , RNA Transport
4.
J Exp Med ; 214(6): 1737-1752, 2017 06 05.
Article in English | MEDLINE | ID: mdl-28487310

ABSTRACT

MicroRNA (miRNA)-mediated RNA interference regulates many immune processes, but how miRNA circuits orchestrate aberrant intestinal inflammation during inflammatory bowel disease (IBD) is poorly defined. Here, we report that miR-223 limits intestinal inflammation by constraining the nlrp3 inflammasome. miR-223 was increased in intestinal biopsies from patients with active IBD and in preclinical models of intestinal inflammation. miR-223-/y mice presented with exacerbated myeloid-driven experimental colitis with heightened clinical, histopathological, and cytokine readouts. Mechanistically, enhanced NLRP3 inflammasome expression with elevated IL-1ß was a predominant feature during the initiation of colitis with miR-223 deficiency. Depletion of CCR2+ inflammatory monocytes and pharmacologic blockade of IL-1ß or NLRP3 abrogated this phenotype. Generation of a novel mouse line, with deletion of the miR-223 binding site in the NLRP3 3' untranslated region, phenocopied the characteristics of miR-223-/y mice. Finally, nanoparticle-mediated overexpression of miR-223 attenuated experimental colitis, NLRP3 levels, and IL-1ß release. Collectively, our data reveal a previously unappreciated role for miR-223 in regulating the innate immune response during intestinal inflammation.


Subject(s)
Inflammasomes/metabolism , Inflammation/genetics , Intestines/pathology , MicroRNAs/metabolism , Myeloid Cells/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Adult , Animals , Antibodies/metabolism , Base Sequence , Colitis/chemically induced , Colitis/genetics , Colitis/pathology , Dextran Sulfate , Disease Susceptibility , Hematopoiesis , Humans , Inflammation/pathology , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Interleukin-1beta/metabolism , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Middle Aged , Monocytes/metabolism , Nanoparticles/chemistry , Neutrophils/metabolism , Receptors, CCR2/metabolism
5.
Am J Physiol Renal Physiol ; 313(2): F553-F559, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28515178

ABSTRACT

Sepsis outcomes are heavily dependent on the development of septic organ injury, but no interventions exist to interrupt or reverse this process. microRNA-223 (miR-223) is known to be involved in both inflammatory gene regulation and host-pathogen interactions key to the pathogenesis of sepsis. The goal of this study was to determine the role of miR-223 as a mediator of septic kidney injury. Using miR-223 knockout mice and multiple models of experimental sepsis, we found that miR-223 differentially influences acute kidney injury (AKI) based on the model used. In the absence of miR-223, mice demonstrated exaggerated AKI in sterile models of sepsis (LPS injection) and attenuated AKI in a live-infection model of sepsis (cecal ligation and puncture). We demonstrated that miR-223 expression is induced in kidney homogenate after cecal ligation and puncture, but not after LPS or fecal slurry injection. We investigated additional potential mechanistic explanations including differences in peritoneal bacterial clearance and host stool virulence. Our findings highlight the complex role of miR-223 in the pathogenesis of septic kidney injury, as well as the importance of differences in experimental sepsis models and their consequent translational applicability.


Subject(s)
Acute Kidney Injury/etiology , Disease Models, Animal , MicroRNAs/metabolism , Sepsis/complications , Acute Kidney Injury/metabolism , Animals , Lipopolysaccharides , Male , Methicillin-Resistant Staphylococcus aureus , Mice, Inbred C57BL , Mice, Knockout , Sepsis/metabolism
6.
J Leukoc Biol ; 101(2): 471-480, 2017 02.
Article in English | MEDLINE | ID: mdl-27601626

ABSTRACT

A critical step in the pathogenesis of acute lung injury (ALI) is excessive recruitment of polymorphonuclear neutrophils (PMNs) into the lungs, causing significant collateral tissue damage. Defining the molecular and cellular steps that control neutrophil infiltration and activation during ALI is therefore of important therapeutic relevance. Based on previous findings implicating the transcription factor Tbet in mucosal Th1-inflammation, we hypothesized a detrimental role for Tbet during ALI. In line with our hypothesis, initial studies of endotoxin-induced lung injury revealed a marked protection of Tbet-/- mice, including attenuated neutrophilia compared to WT counterparts. Surprisingly, subsequent studies identified natural killer (NK) cells as the major source of pulmonary Tbet during ALI. In addition, a chemokine screen suggested that mature Tbet+ NK-cells are critical for the production of pulmonary CXCL1 and -2, thereby contributing to pulmonary PMN recruitment. Indeed, both NK-cell Ab depletion and adoptive transfer studies provide evidence for NK cells in the orchestration of neutrophil recruitment during endotoxin-induced ALI. Taken together, these findings identify a novel role for Tbet+ NK-cells in initiating the early events of noninfectious pulmonary inflammation.


Subject(s)
Acute Lung Injury/immunology , Acute Lung Injury/pathology , Killer Cells, Natural/immunology , Neutrophil Infiltration/immunology , Receptors, Interleukin-8B/metabolism , Acute Lung Injury/complications , Adoptive Transfer , Animals , Antibodies/metabolism , Cytokines/metabolism , Inflammation Mediators/metabolism , Lipopolysaccharides , Mice, Inbred C57BL , Pneumonia/complications , Pneumonia/immunology , Pneumonia/pathology , T-Box Domain Proteins/metabolism
7.
Anesthesiology ; 124(2): 489-506, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26632665

ABSTRACT

MicroRNAs (miRNAs) are small, non-protein-coding, single-stranded RNAs. They function as posttranscriptional regulators of gene expression by interacting with target mRNAs. This process prevents translation of target mRNAs into a functional protein. miRNAs are considered to be functionally involved in virtually all physiologic processes, including differentiation and proliferation, metabolism, hemostasis, apoptosis, and inflammation. Many of these functions have important implications for anesthesiology and critical care medicine. Studies indicate that miRNA expression levels can be used to predict the risk for eminent organ injury or sepsis. Pharmacologic approaches targeting miRNAs for the treatment of human diseases are currently being tested in clinical trials. The present review highlights the important biological functions of miRNAs and their usefulness as perioperative biomarkers and discusses the pharmacologic approaches that modulate miRNA functions for disease treatment. In addition, the authors discuss the pharmacologic interactions of miRNAs with currently used anesthetics and their potential to impact anesthetic toxicity and side effects.


Subject(s)
Anesthesiology/methods , MicroRNAs/blood , Perioperative Care/methods , Biomarkers/blood , Humans , MicroRNAs/drug effects
8.
J Immunol ; 195(10): 4973-85, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26453755

ABSTRACT

NK cells are innate lymphoid cells important for immune surveillance, identifying and responding to stress, infection, and/or transformation. Whereas conventional NK (cNK) cells circulate systemically, many NK cells reside in tissues where they appear to be poised to locally regulate tissue function. In the present study, we tested the contribution of tissue-resident NK (trNK) cells to tissue homeostasis by studying ischemic injury in the mouse kidney. Parabiosis experiments demonstrate that the kidney contains a significant fraction of trNK cells under homeostatic conditions. Kidney trNK cells developed independent of NFIL3 and T-bet, and they expressed a distinct cell surface phenotype as compared with cNK cells. Among these, trNK cells had reduced asialo-GM1 (AsGM1) expression relative to cNK cells, a phenotype observed in trNK cells across multiple organs and mouse strains. Strikingly, anti-AsGM1 Ab treatment, commonly used as an NK cell-depleting regimen, resulted in a robust and selective depletion of cNKs, leaving trNKs largely intact. Using this differential depletion, we tested the relative contribution of cNK and trNK cells in ischemic kidney injury. Whereas anti-NK1.1 Ab effectively depleted both trNK and cNK cells and protected against ischemic/reperfusion injury, anti-AsGM1 Ab preferentially depleted cNK cells and failed to protect against injury. These data demonstrate unanticipated specificity of anti-AsGM1 Ab depletion on NK cell subsets and reveal a new approach to study the contributions of cNK and trNK cells in vivo. In total, these data demonstrate that trNK cells play a key role in modulating local responses to ischemic tissue injury in the kidney and potentially other organs.


Subject(s)
Antibodies/pharmacology , G(M1) Ganglioside/immunology , Ischemia/immunology , Kidney Diseases/immunology , Kidney/blood supply , Kidney/immunology , Killer Cells, Natural/immunology , Animals , G(M1) Ganglioside/antagonists & inhibitors , Ischemia/pathology , Kidney/pathology , Kidney Diseases/pathology , Killer Cells, Natural/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Knockout
9.
J Immunol ; 195(4): 1815-24, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26188061

ABSTRACT

Acute lung injury (ALI) is an acute inflammatory lung disease that causes morbidity and mortality in critically ill patients. However, there are many instances where ALI resolves spontaneously through endogenous pathways that help to control excessive lung inflammation. Previous studies have implicated the extracellular signaling molecule adenosine and signaling events through the A2B adenosine receptor in lung protection. In this context, we hypothesized that tissue-specific expression of the A2B adenosine receptor is responsible for the previously described attenuation of ALI. To address this hypothesis, we exposed mice with tissue-specific deletion of Adora2b to ALI, utilizing a two-hit model where intratracheal LPS treatment is followed by injurious mechanical ventilation. Interestingly, a head-to-head comparison of mice with deletion of Adora2b in the myeloid lineage (Adora2b(loxP/loxP) LysM Cre(+)), endothelial cells (Adora2b(loxP/loxP) VE-cadherin Cre(+)), or alveolar epithelial cells (Adora2b(loxP/loxP) SPC Cre(+)) revealed a selective increase in disease susceptibility in Adora2b(loxP/loxP) SPC Cre(+) mice. More detailed analysis of Adora2b(loxP/loxP) SPC Cre(+) mice confirmed elevated lung inflammation and attenuated alveolar fluid clearance. To directly deliver an A2B adenosine receptor-specific agonist to alveolar epithelial cells, we subsequently performed studies with inhaled BAY 60-6583. Indeed, aerosolized BAY 60-6583 treatment was associated with attenuated pulmonary edema, improved histologic lung injury, and dampened lung inflammation. Collectively, these findings suggest that alveolar epithelial A2B adenosine receptor signaling contributes to lung protection, and they implicate inhaled A2B adenosine receptor agonists in ALI treatment.


Subject(s)
Acute Lung Injury/metabolism , Alveolar Epithelial Cells/metabolism , Receptor, Adenosine A2B/metabolism , Acute Lung Injury/drug therapy , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Adenosine A2 Receptor Agonists/administration & dosage , Administration, Inhalation , Animals , Disease Models, Animal , Gene Deletion , Genetic Predisposition to Disease , Humans , Macrophages, Alveolar/metabolism , Mice , Mice, Knockout , Neutrophils/metabolism , Organ Specificity/genetics , Receptor, Adenosine A2B/genetics , Severity of Illness Index
10.
Mol Endocrinol ; 29(3): 460-72, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25650755

ABSTRACT

The genetic and molecular mechanisms that initiate and maintain pituitary tumorigenesis are poorly understood. Nonfunctioning tumors of the gonadotrope lineage represent 35% of all tumors; are usually macroadenomas, often resulting in hypopituitarism; and have no medical treatments. Using expression microarrays combined with whole-genome copy number screens on individual human tumors, we identified the mammalian sterile-20-like kinase (MST4) transcript, which was amplified within chromosome Xq26.2 in one tumor and up-regulated in all gonadotrope tumor samples. MST4 mRNA and protein were consistently overexpressed in human tumors compared with normal pituitaries. To mimic the pituitary tumor microenvironment, a hypoxia model using LßT2 murine gonadotrope cells was created to examine the functional role of the kinase. During long-term hypoxia, MST4 expression increased colony formation in a soft agar assay and rates of cell proliferation by activating p38 MAPK and AKT. Under short-term severe hypoxic stress, MST4 decreased the rates of apoptosis via p38 MAPK, AKT, hypoxia-inducible factor-1, and its cell-specific downstream targets. Analysis of MST4 mutants confirmed the importance of the kinase sequence but not the regulatory C terminus for its functional effects. Together these data identify the MST4 kinase as a novel candidate to mediate human pituitary tumorigenesis in a hypoxic environment and position it as a potential therapeutic target.


Subject(s)
Pituitary Gland/enzymology , Pituitary Gland/pathology , Protein Serine-Threonine Kinases/metabolism , Apoptosis , Cell Hypoxia , Cell Proliferation , Cell Survival , Cytoprotection , DNA Copy Number Variations/genetics , Gene Amplification , Gene Expression Regulation, Neoplastic , Gonadotrophs/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Pituitary Neoplasms/genetics , Pituitary Neoplasms/pathology , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Stress, Physiological , Tumor Stem Cell Assay , Up-Regulation/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
11.
J Immunol ; 192(3): 1249-56, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24391213

ABSTRACT

Although acute lung injury (ALI) contributes significantly to critical illness, resolution often occurs spontaneously through endogenous pathways. We recently found that mechanical ventilation increases levels of pulmonary adenosine, a signaling molecule known to attenuate lung inflammation. In this study, we hypothesized a contribution of transcriptionally controlled pathways to pulmonary adenosine receptor (ADOR) signaling during ALI. We gained initial insight from microarray analysis of pulmonary epithelia exposed to conditions of cyclic mechanical stretch, a mimic for ventilation-induced lung disease. Surprisingly, these studies revealed a selective induction of the ADORA2B. Using real-time RT-PCR and Western blotting, we confirmed an up to 9-fold induction of the ADORA2B following cyclic mechanical stretch (A549, Calu-3, or human primary alveolar epithelial cells). Studies using ADORA2B promoter constructs identified a prominent region within the ADORA2B promoter conveying stretch responsiveness. This region of the promoter contained a binding site for the transcription factor hypoxia-inducible factor (HIF)-1. Additional studies using site-directed mutagenesis or transcription factor binding assays demonstrated a functional role for HIF-1 in stretch-induced increases of ADORA2B expression. Moreover, studies of ventilator-induced lung injury revealed induction of the ADORA2B during ALI in vivo that was abolished following HIF inhibition or genetic deletion of Hif1a. Together, these studies implicate HIF in the transcriptional control of pulmonary adenosine signaling during ALI.


Subject(s)
Acute Lung Injury/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Receptor, Adenosine A2B/genetics , Stress, Mechanical , Ventilator-Induced Lung Injury/physiopathology , Acute Lung Injury/metabolism , Adenosine/physiology , Animals , Binding Sites , Cells, Cultured , Epithelial Cells/physiology , Female , Genes, Reporter , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Lung/metabolism , Lung/physiopathology , Male , Mice , Mice, Knockout , Mice, Transgenic , Promoter Regions, Genetic/genetics , Receptor, Adenosine A2B/biosynthesis , Receptor, Adenosine A2B/physiology , Transcription, Genetic
12.
Proc Natl Acad Sci U S A ; 110(29): 12012-7, 2013 Jul 16.
Article in English | MEDLINE | ID: mdl-23812746

ABSTRACT

Ischemia and reperfusion significantly contributes to the morbidity and mortality of liver surgery and transplantation. Based on studies showing a critical role for adenosine signaling in mediating tissue adaptation during hypoxia, we hypothesized that signaling events through adenosine receptors (ADORA1, ADORA2A, ADORA2B, or ADORA3) attenuates hepatic ischemia and reperfusion injury. Initial screening studies of human liver biopsies obtained during hepatic transplantation demonstrated a selective and robust induction of ADORA2B transcript and protein following ischemia and reperfusion. Subsequent exposure of gene-targeted mice for each individual adenosine receptor to liver ischemia and reperfusion revealed a selective role for the Adora2b in liver protection. Moreover, treatment of wild-type mice with an Adora2b-selective antagonist resulted in enhanced liver injury, whereas Adora2b-agonist treatment was associated with attenuated hepatic injury in wild-type, but not in Adora2b(-/-) mice. Subsequent studies in mice with Adora2b deletion in different tissues--including vascular endothelia, myeloid cells, and hepatocytes--revealed a surprising role for hepatocellular-specific Adora2b signaling in attenuating nuclear factor NF-κB activation and thereby mediating liver protection from ischemia and reperfusion injury. These studies provide a unique role for hepatocellular-specific Adora2b signaling in liver protection during ischemia and reperfusion injury.


Subject(s)
Liver/physiopathology , Receptor, Adenosine A2B/metabolism , Reperfusion Injury/physiopathology , Signal Transduction/physiology , Animals , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Gene Expression Profiling , Humans , Immunoblotting , Liver/metabolism , Mice , Mice, Knockout , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Receptor, Adenosine A2B/genetics , Reperfusion Injury/metabolism
13.
FASEB J ; 27(8): 3078-89, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23603835

ABSTRACT

The signaling molecule adenosine has been implicated in attenuating acute lung injury (ALI). Adenosine signaling is terminated by its uptake through equilibrative nucleoside transporters (ENTs). We hypothesized that ENT-dependent adenosine uptake could be targeted to enhance adenosine-mediated lung protection. To address this hypothesis, we exposed mice to high-pressure mechanical ventilation to induce ALI. Initial studies demonstrated time-dependent repression of ENT1 and ENT2 transcript and protein levels during ALI. To examine the contention that ENT repression represents an endogenous adaptive response, we performed functional studies with the ENT inhibitor dipyridamole. Dipyridamole treatment (1 mg/kg; EC50=10 µM) was associated with significant increases in ALI survival time (277 vs. 395 min; P<0.05). Subsequent studies in gene-targeted mice for Ent1 or Ent2 revealed a selective phenotype in Ent2(-/-) mice, including attenuated pulmonary edema and improved gas exchange during ALI in conjunction with elevated adenosine levels in the bronchoalveolar fluid. Furthermore, studies in genetic models for adenosine receptors implicated the A2B adenosine receptor (Adora2b) in mediating ENT-dependent lung protection. Notably, dipyridamole-dependent attenuation of lung inflammation was abolished in mice with alveolar epithelial Adora2b gene deletion. Our newly identified crosstalk pathway between ENT2 and alveolar epithelial Adora2b in lung protection during ALI opens possibilities for combined therapies targeted to this protein set.


Subject(s)
Acute Lung Injury/metabolism , Equilibrative-Nucleoside Transporter 2/metabolism , Receptor, Adenosine A2B/metabolism , Signal Transduction , Acute Lung Injury/genetics , Acute Lung Injury/prevention & control , Adenosine/metabolism , Animals , Bronchoalveolar Lavage Fluid/chemistry , Cell Line , Dipyridamole/pharmacology , Equilibrative Nucleoside Transporter 1/genetics , Equilibrative Nucleoside Transporter 1/metabolism , Equilibrative-Nucleoside Transporter 2/antagonists & inhibitors , Equilibrative-Nucleoside Transporter 2/genetics , Gene Expression , Humans , Immunoblotting , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Alveoli/metabolism , Pulmonary Gas Exchange/drug effects , Pulmonary Gas Exchange/genetics , Receptor, Adenosine A2B/genetics , Reverse Transcriptase Polymerase Chain Reaction , Vasodilator Agents/pharmacology
14.
FASEB J ; 27(6): 2207-19, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23413361

ABSTRACT

Acute lung injury (ALI) is characterized by alveolar injury and uncontrolled inflammation. Since most cases of ALI resolve spontaneously, understanding the endogenous mechanisms that promote ALI resolution is important to developing effective therapies. Previous studies have implicated extracellular adenosine signaling in tissue adaptation and wound healing. Therefore, we hypothesized a functional contribution for the endogenous production of adenosine during ALI resolution. As a model, we administered intratracheal LPS and observed peak lung injury at 3 d, with resolution by d 14. Treatment with pegylated adenosine-deaminase to enhance extracellular adenosine breakdown revealed impaired ALI resolution. Similarly, genetic deletion of cd73, the pacemaker for extracellular adenosine generation, was associated with increased mortality (0% wild-type and 40% in cd73(-/-) mice; P<0.05) and failure to resolve ALI adequately. Studies of inflammatory cell trafficking into the lungs during ALI resolution revealed that regulatory T cells (Tregs) express the highest levels of CD73. While Treg numbers in cd73(-/-) mice were similar to controls, cd73-deficient Tregs had attenuated immunosuppressive functions. Moreover, adoptive transfer of cd73-deficient Tregs into Rag(-/-) mice emulated the observed phenotype in cd73(-/-) mice, while transfer of wild-type Tregs was associated with normal ALI resolution. Together, these studies implicate CD73-dependent adenosine generation in Tregs in promoting ALI resolution.


Subject(s)
5'-Nucleotidase/physiology , Acute Lung Injury/immunology , Acute Lung Injury/metabolism , Adenosine/physiology , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/immunology , 5'-Nucleotidase/deficiency , Acute Lung Injury/pathology , Adenosine/deficiency , Adenosine Deaminase/administration & dosage , Adoptive Transfer , Animals , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Lipopolysaccharides/toxicity , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , T-Lymphocytes, Regulatory/pathology
15.
J Clin Invest ; 122(2): 693-710, 2012 02.
Article in English | MEDLINE | ID: mdl-22269324

ABSTRACT

A complex biologic network regulates kidney perfusion under physiologic conditions. This system is profoundly perturbed following renal ischemia, a leading cause of acute kidney injury (AKI) - a life-threatening condition that frequently complicates the care of hospitalized patients. Therapeutic approaches to prevent and treat AKI are extremely limited. Better understanding of the molecular pathways promoting postischemic reflow could provide new candidate targets for AKI therapeutics. Due to its role in adapting tissues to hypoxia, we hypothesized that extracellular adenosine has a regulatory function in the postischemic control of renal perfusion. Consistent with the notion that equilibrative nucleoside transporters (ENTs) terminate adenosine signaling, we observed that pharmacologic ENT inhibition in mice elevated renal adenosine levels and dampened AKI. Deletion of the ENTs resulted in selective protection in Ent1-/- mice. Comprehensive examination of adenosine receptor-knockout mice exposed to AKI demonstrated that renal protection by ENT inhibitors involves the A2B adenosine receptor. Indeed, crosstalk between renal Ent1 and Adora2b expressed on vascular endothelia effectively prevented a postischemic no-reflow phenomenon. These studies identify ENT1 and adenosine receptors as key to the process of reestablishing renal perfusion following ischemic AKI. If translatable from mice to humans, these data have important therapeutic implications.


Subject(s)
Acute Kidney Injury/metabolism , Equilibrative Nucleoside Transporter 1/metabolism , Ischemia/metabolism , Regional Blood Flow/physiology , Acute Kidney Injury/drug therapy , Acute Kidney Injury/pathology , Adenosine/metabolism , Animals , Cell Line , Chimerism , Dipyridamole/therapeutic use , Equilibrative Nucleoside Transporter 1/antagonists & inhibitors , Equilibrative Nucleoside Transporter 1/genetics , Humans , Kidney/metabolism , Kidney/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , No-Reflow Phenomenon , Nucleoside Transport Proteins/antagonists & inhibitors , Nucleoside Transport Proteins/metabolism , Phosphodiesterase Inhibitors/therapeutic use , Receptors, Purinergic P1/genetics , Receptors, Purinergic P1/metabolism
16.
PLoS One ; 6(5): e14812, 2011.
Article in English | MEDLINE | ID: mdl-21625583

ABSTRACT

The netrin family of secreted proteins provides migrational cues in the developing central nervous system. Recently, netrins have also been shown to regulate diverse processes beyond their functions in the brain, incluing the ochrestration of inflammatory events. Particularly netrin-1 has been implicated in dampening hypoxia-induced inflammation. Here, we hypothesized an anti-inflammatory role of endogenous netrin-1 in acute kidney injury (AKI). As homozygous deletion of netrin-1 is lethal, we studied mice with partial netrin-1 deletion (Ntn-1(+/-) mice) as a genetic model. In fact, Ntn-1(+/-) mice showed attenuated Ntn-1 levels at baseline and following ischemic AKI. Functional studies of AKI induced by 30 min of renal ischemia and reperfusion revealed enhanced kidney dysfunction in Ntn-1(+/-) mice as assessed by measurements of glomerular filtration, urine flow rate, urine electrolytes, serum creatinine and creatinine clearance. Consistent with these findings, histological studies indicated a more severe degree kidney injury. Similarly, elevations of renal and systemic inflammatory markers were enhanced in mice with partial netrin-1 deficiency. Finally, treatment of Ntn-1(+/-) mice with exogenous netrin-1 restored a normal phenotype during AKI. Taking together, these studies implicate endogenous netrin-1 in attenuating renal inflammation during AKI.


Subject(s)
Acute Kidney Injury/etiology , Acute Kidney Injury/pathology , Nerve Growth Factors/metabolism , Nerve Growth Factors/physiology , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/physiology , Acute Kidney Injury/metabolism , Animals , Blotting, Western , Cells, Cultured , Creatinine/blood , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Glomerular Filtration Rate , Humans , Immunoenzyme Techniques , Kidney/cytology , Kidney/metabolism , Kidney Function Tests , Mice , Mice, Knockout , Nerve Growth Factors/genetics , Netrin-1 , Phenotype , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Proteins/genetics
17.
Exp Biol Med (Maywood) ; 234(10): 1155-65, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19596832

ABSTRACT

Autosomal dominant polycystic kidney (ADPKD) is highly prevalent genetic disease. Liver cyst disease is the most common extrarenal manifestation in ADPKD and accounts for up to 10% of ADPKD morbidity and mortality. The clinical features of ADPKD liver disease arise from dramatic increases in liver cyst volumes. To identify mechanisms that promote liver cyst growth, the present study characterized the degree of vascularization of liver cyst walls and determined that cyst-specific cytokines and growth factors can drive endothelial cell proliferation and development. Microscopic techniques demonstrated liver cyst walls are well vascularized. A comparative analysis found the vascular density in free liver cyst walls was greater in mice than in humans. Treatment of human micro-vascular endothelial cells (HMEC-1) with human liver cyst fluid (huLCF) induced a rapid increase in vascular endothelium growth factor receptor 2 (VEGFR2) phosphorylation that persisted for 45-60 min and was blocked by 20 microM SU5416, a VEGFR tyrosine kinase inhibitor. Similarly, huLCF treatment of HMEC-1 cells induced an increase in the cell proliferation rate (131 +/- 6% of control levels; P > 0.05) and the degree of vascular development ('tube' diameter assay: 92 +/- 14 microm for huLCF vs. 12 +/- 7 microm for vehicle); P > 0.05). Both cell proliferation and vascular development were sensitive to SU5416. These studies indicate that factors secreted by liver cyst epithelia can activate VEGF signaling pathways and induce endothelial cell proliferation and differentiation. The present studies suggest that targeting VEGFR2-dependent angiogenesis may be an effective therapeutic strategy in blocking ADPKD liver cyst vascularization and growth.


Subject(s)
Cell Proliferation/drug effects , Cysts/metabolism , Cytokines/pharmacology , Endothelial Cells/physiology , Liver Diseases/metabolism , Animals , Cells, Cultured , Cyst Fluid/metabolism , Cysts/blood supply , Cytokines/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Enzyme Inhibitors/pharmacology , Humans , Indoles/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , NIMA-Interacting Peptidylprolyl Isomerase , Neovascularization, Pathologic/metabolism , Peptidylprolyl Isomerase/pharmacology , Phosphorylation , Polycystic Kidney, Autosomal Dominant/metabolism , Pyrroles/pharmacology , TRPP Cation Channels/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
18.
Am J Physiol Cell Physiol ; 294(3): C786-96, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18199703

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) is a highly prevalent genetic disease that results in cyst formation in kidney and liver. Cytokines and growth factors secreted by the cyst-lining epithelia are positioned to initiate autocrine/paracrine signaling and promote cyst growth. Comparative analyses of human kidney and liver cyst fluids revealed disparate cytokine/growth factor profiles. CXCR2 agonists, including IL-8, epithelial neutrophil-activating peptide (ENA-78), growth-related oncogene-alpha (GRO-alpha), are potent proliferative agents that were found at high levels in liver but not kidney cyst fluids. Liver cysts are lined by epithelial cells derived from the intrahepatic bile duct (i.e., cholangiocytes). In polarized pkd2(WS25/-) mouse liver cyst epithelial monolayers, CXCR2 agonists were released both apically and basally, indicating that they may act both on the endothelial and epithelial cells within or lining the cyst wall. IL-8 and human liver cyst fluid induced cell proliferation of HMEC-1 cells, a human microvascular endothelial cell line, and Mz-ChA1 cells, a human cholangiocyte cell model. IL-8 expression can be regulated by specific stresses. Hypoxia and mechanical stretch, two likely stressors acting on the liver cyst epithelia, significantly increased IL-8 secretion and promoter activity. AP-1, c/EBP, and NF-kappaB were required but not sufficient to drive the stress-induced increase in IL-8 transcription. An upstream element between -272 and -1,481 bp allowed for the stress-induced increase in IL-8 transcription. These studies support the hypothesis that CXCR2 signaling promotes ADPKD liver cyst growth.


Subject(s)
Cell Proliferation , Cyst Fluid/metabolism , Cysts/metabolism , Interleukin-8/metabolism , Liver Diseases/metabolism , Liver/metabolism , Polycystic Kidney, Autosomal Dominant/complications , Receptors, Interleukin-8B/metabolism , Animals , Cell Hypoxia , Cell Line , Cell Line, Tumor , Cell Polarity , Cell Proliferation/drug effects , Cell Shape , Cells, Cultured , Chemokine CXCL1/metabolism , Chemokine CXCL5/metabolism , Cysts/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Interleukin-8/genetics , Interleukin-8/pharmacology , Kidney/metabolism , Kidney/pathology , Liver/drug effects , Liver/pathology , Liver Diseases/etiology , Liver Diseases/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Dominant/pathology , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Receptors, Interleukin-8B/agonists , Signal Transduction , Stress, Mechanical , TRPP Cation Channels/metabolism , Transcription, Genetic , Transfection
19.
Am J Physiol Cell Physiol ; 293(1): C419-28, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17475663

ABSTRACT

Proliferation of cyst-lining epithelial cells is an integral part of autosomal dominant polycystic kidney disease (ADPKD) cyst growth. Cytokines and growth factors within cyst fluids are positioned to induce cyst growth. Vascular endothelial growth factor (VEGF) is a pleiotropic growth factor present in ADPKD liver cyst fluids (human 1,128 +/- 78, mouse 2,787 +/- 136 pg/ml) and, to a lesser extent, in ADPKD renal cyst fluids (human 294 +/- 41, mouse 191 +/- 90 pg/ml). Western blotting showed that receptors for VEGF (VEGFR1 and VEGFR2) were present in both normal mouse bile ducts and pkd2(WS25/-) liver cyst epithelial cells. Treatment of pkd2(WS25/-) liver cyst epithelial cells with VEGF (50-50,000 pg/ml) or liver cyst fluid induced a proliferative response. The effect on proliferation of liver cyst fluid was inhibited by SU-5416, a potent VEGF receptor inhibitor. Treatment of pkd2(WS25/-) mice between 4 and 8 mo of age with SU-5416 markedly reduced the cyst volume density of the liver (vehicle 9.9 +/- 4.3%, SU-5416 1.8 +/- 0.7% of liver). SU-5416 treatment between 4 and 12 mo of age markedly protected against increases in liver weight [pkd2(+/+) 4.8 +/- 0.2%, pkd2(WS25/-)-vehicle 10.8 +/- 1.9%, pkd2(WS25/-)-SU-5416 4.8 +/- 0.4% body wt]. The capacity of VEGF signaling to induce in vitro proliferation of pkd2(WS25/-) liver cyst epithelial cells and inhibition of in vivo VEGF signaling to retard liver cyst growth in pkd2(WS25/-) mice indicates that the VEGF signaling pathway is a potentially important therapeutic target in the treatment of ADPKD liver cyst disease.


Subject(s)
Cysts/prevention & control , Indoles/pharmacology , Liver Diseases/prevention & control , Liver/drug effects , Polycystic Kidney, Autosomal Dominant/complications , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , TRPP Cation Channels/metabolism , Animals , Cell Proliferation/drug effects , Cells, Cultured , Cyst Fluid/metabolism , Cysts/genetics , Cysts/metabolism , Cysts/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Indoles/therapeutic use , Liver/metabolism , Liver/pathology , Liver Diseases/genetics , Liver Diseases/metabolism , Liver Diseases/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Polycystic Kidney, Autosomal Dominant/drug therapy , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Dominant/pathology , Protein Kinase Inhibitors/therapeutic use , Pyrroles/therapeutic use , Receptors, Vascular Endothelial Growth Factor/metabolism , Signal Transduction/drug effects , Time Factors , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism
20.
Biochim Biophys Acta ; 1772(3): 345-54, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17208416

ABSTRACT

Derived from bile duct epithelia (BDE), secretion by liver cyst-lining epithelia is positioned to drive cyst expansion but the responsible ion flux pathways have not been characterized. Cyst-lining epithelia were isolated and cultured into high resistance monolayers to assess the ion secretory pathways. Electrophysiologic studies showed a marked rate of constitutive transepithelial ion transport, including Cl(-) secretion and Na(+) absorption. Na(+) absorption was amiloride-sensitive, suggesting the activation of epithelial sodium channels (ENaC). Further, both cAMP(i) and extracellular ATP induced robust secretory responses. Western blotting and immunohistologic analysis of liver cyst epithelia demonstrated expression of P2X4, a potent purinergic receptor in normal BDE. Luminometry and bioassaying measured physiologically relevant levels of ATP in a subset of liver cyst fluid samples. Liver cyst epithelia also displayed a significant capacity to degrade extracellular ATP. In conclusion, regulated ion transport pathways are present in liver cyst epithelia and are positioned to direct fluid secretion into the lumen of liver cysts and promote increases in liver cyst expansion and growth.


Subject(s)
Bile Ducts/metabolism , Cysts/metabolism , Epithelial Cells/metabolism , Epithelial Sodium Channels/metabolism , Liver Neoplasms/metabolism , Adenosine Triphosphate/analysis , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Amiloride/pharmacology , Animals , Bile Ducts/pathology , Chlorine/metabolism , Cyclic AMP/pharmacology , Cysts/chemistry , Epithelial Sodium Channels/drug effects , Ion Transport , Liver Neoplasms/chemistry , Mice , Mice, Inbred Strains , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2X4 , Sodium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...