Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
J Infect Dis ; 222(6): 1037-1045, 2020 08 17.
Article in English | MEDLINE | ID: mdl-32285112

ABSTRACT

BACKGROUND: Sepsis is a life-threatening systemic disease with severe microvascular dysfunction. Pericytes preserve vascular homeostasis. To our knowledge, the potential roles of microRNAs in sepsis-induced pericyte dysfunction have not been explored. METHODS: We determined lung pericyte expression of miR-145a in cecal ligation and puncture (CLP)-induced sepsis. Mouse lung pericytes were isolated and transfected with a miR-145a mimic, followed by stimulation with lipopolysaccharide (LPS). We measured inflammatory cytokine levels. To assess the functions of miR-145a in vivo, we generated a pericyte-specific miR-145a-knockout mouse and determined sepsis-induced organ injury, lung and renal vascular leakage, and mouse survival rates. We used RNA sequencing and Western blotting to analyze the signaling pathways regulated by miR-145a. RESULTS: CLP led to decreased miR-145a expression in lung pericytes. The miR-145a mimic inhibited LPS-induced increases in cytokines. In CLP-induced sepsis, pericytes lacking miR-145a exhibited increased lung and kidney vascular leakage and reduced survival rates. We found that miR-145a could suppress LPS-induced NF-κB activation. In addition, we confirmed that the transcription factor Friend leukemia virus integration 1 (Fli-1) is a target of miR-145a and that Fli-1 activates NF-κB signaling. CONCLUSION: Our results demonstrated that pericyte miR-145a mediates sepsis-associated microvascular dysfunction, potentially by means of Fli-1-mediated modulation of NF-κB signaling.


Subject(s)
Host-Pathogen Interactions/genetics , MicroRNAs/genetics , Pericytes/metabolism , Sepsis/etiology , Animals , Cytokines/blood , Cytokines/metabolism , Disease Models, Animal , Gene Expression Regulation , Inflammation Mediators/blood , Inflammation Mediators/metabolism , Mice , Mice, Knockout , Models, Biological , NF-kappa B/metabolism , Prognosis , RNA Interference , Sepsis/mortality , Signal Transduction
2.
Am J Physiol Lung Cell Mol Physiol ; 318(6): L1261-L1269, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32321279

ABSTRACT

Circulating microRNAs (miRNAs) can be taken up by recipient cells and have been recently associated with the acute respiratory distress syndrome (ARDS). Their role in host predisposition to the syndrome is unknown. The objective of the study was to identify circulating miRNAs associated with the development of sepsis-related ARDS and examine their impact on endothelial cell gene expression and function. We determined miRNA levels in plasma collected from subjects during the first 24 h of admission to a tertiary intensive care unit for sepsis. A miRNA that was differentially expressed between subjects who did and did not develop ARDS was identified and was transfected into human pulmonary microvascular endothelial cells (HPMECs). RNA sequencing, in silico analysis, cytokine expression, and leukocyte migration assays were used to determine the impact of this miRNA on gene expression and cell function. In two cohorts, circulating miR-887-3p levels were elevated in septic patients who developed ARDS compared with those who did not. Transfection of miR-887-3p into HPMECs altered gene expression, including the upregulation of several genes previously associated with ARDS (e.g., CXCL10, CCL5, CX3CL1, VCAM1, CASP1, IL1B, IFNB, and TLR2), and activation of cellular pathways relevant to the response to infection. Functionally, miR-887-3p increased the endothelial release of chemokines and facilitated trans-endothelial leukocyte migration. Circulating miR-887-3p is associated with ARDS in critically ill patients with sepsis. In vitro, miR-887-3p regulates the expression of genes relevant to ARDS and neutrophil tracking. This miRNA may contribute to ARDS pathogenesis and could represent a novel therapeutic target.


Subject(s)
Circulating MicroRNA/blood , Circulating MicroRNA/genetics , Endothelial Cells/metabolism , Gene Expression Regulation , MicroRNAs/blood , MicroRNAs/genetics , Respiratory Distress Syndrome/blood , Respiratory Distress Syndrome/genetics , Cell Movement , Chemokines/metabolism , Circulating MicroRNA/metabolism , Cohort Studies , Female , Humans , Lung/blood supply , Male , MicroRNAs/metabolism , Microvessels/pathology , Middle Aged , Neutrophils/metabolism
3.
Mol Immunol ; 108: 1-7, 2019 04.
Article in English | MEDLINE | ID: mdl-30739075

ABSTRACT

Our previous data demonstrated that Friend leukemia virus integration 1 (Fli-1), an ETS transcription factor, governs pericyte loss and vascular dysfunction in cecal ligation and puncture-induced murine sepsis by regulating essential pyroptosis markers including caspase-1. However, whether Fli-1 regulates caspase-1 expression levels in vitro and how Fli-1 regulates caspase-1 remain unknown. Our present work further demonstrated that overexpressed Fli-1 significantly increased caspase-1 and IL-18 expression levels in cultured mouse lung pericytes. Bacterial outer membrane vesicles (OMVs) have been found to induce cell pyroptosis through transferring LPS intracellularly. Using OMVs to induce an in vitro model of pyroptosis, we observed that OMVs significantly increased protein levels of Fli-1 in mouse lung pericytes. Furthermore, knockdown of Fli-1 by siRNA blocked OMVs-induced caspase-1, caspase-11 and IL-18 expression levels. As caspase-1 was predicted as a potential target of Fli-1, we cloned murine caspase-1 promoter into a luciferase construct. Our data demonstrate for the first time that Fli-1 regulates caspase-1 expression by directly binding to its promoter regions measured by chromatin immunoprecipitation (ChIP) assay and luciferase reporter system. In summary, our findings demonstrated a novel role and mechanism of Fli-1 in regulating caspase-1 expression in lung pericytes.


Subject(s)
Caspase 1/immunology , Gene Expression Regulation, Enzymologic/immunology , Proto-Oncogene Protein c-fli-1/immunology , Animals , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/pharmacology , Caspase 1/genetics , Escherichia coli K12/chemistry , Gene Expression Regulation, Enzymologic/drug effects , Gene Knockdown Techniques , Interleukin-18/genetics , Interleukin-18/immunology , Lung , Mice , Pericytes , Proto-Oncogene Protein c-fli-1/genetics
4.
Crit Care ; 23(1): 44, 2019 02 13.
Article in English | MEDLINE | ID: mdl-30760290

ABSTRACT

BACKGROUND: The acute respiratory distress syndrome (ARDS) is characterized by disruption of the alveolar-capillary barrier resulting in accumulation of proteinaceous edema and increased inflammatory cells in the alveolar space. We previously found that endothelial progenitor cell (EPC) exosomes prevent endothelial dysfunction and lung injury in sepsis in part due to their encapsulation of miRNA-126. However, the effects of EPC exosomes in acute lung injury (ALI) remain unknown. METHODS: To determine if EPC exosomes would have beneficial effects in ALI, intratracheal administration of lipopolysaccharide (LPS) was used to induce ALI in mice. Lung permeability, inflammation, and the role of miRNA-126 in the alveolar-epithelial barrier function were examined. RESULTS: The intratracheal administration of EPC exosomes reduced lung injury following LPS-induced ALI at 24 and 48 h. Compared to placebo, intratracheal administration of EPC exosomes significantly reduced the cell number, protein concentration, and cytokines/chemokines in the bronchoalveolar lavage fluid (BALF), indicating a reduction in permeability and inflammation. Further, EPC exosomes reduced myeloperoxidase (MPO) activity, lung injury score, and pulmonary edema, demonstrating protection against lung injury. Murine fibroblast (NIH3T3) exosomes, which do not contain abundant miRNA-126, did not provide these beneficial effects. In human small airway epithelial cells (SAECs), we found that overexpression of miRNA-126-3p can target phosphoinositide-3-kinase regulatory subunit 2 (PIK3R2), while overexpression of miRNA-126-5p inhibits the inflammatory alarmin HMGB1 and permeability factor VEGFα. Interestingly, both miR-126-3p and 5p increase the expression of tight junction proteins suggesting a potential mechanism by which miRNA-126 may mitigate LPS-induced lung injury. CONCLUSIONS: Our data demonstrated that human EPC exosomes are beneficial in LPS-induced ALI mice, in part through the delivery of miRNA-126 into the injured alveolus.


Subject(s)
Acute Lung Injury/drug therapy , Endothelial Progenitor Cells/enzymology , Inflammation/physiopathology , Acute Lung Injury/physiopathology , Animals , Blotting, Western/methods , Exosomes/metabolism , HMGB1 Protein/metabolism , Inflammation/metabolism , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/adverse effects , Mice , MicroRNAs/physiology , Peroxidase/metabolism , Peroxidase/physiology , Phosphatidylinositol 3-Kinases/metabolism , Real-Time Polymerase Chain Reaction/methods , Severity of Illness Index , Trachea/drug effects , Vascular Endothelial Growth Factor A/metabolism
5.
Inflammation ; 42(1): 170-184, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30244405

ABSTRACT

Sepsis is an acute inflammatory syndrome in response to infection. In some cases, excessive inflammation from sepsis results in endothelial dysfunction and subsequent increased vascular permeability leading to organ failure. We previously showed that treatment with endothelial progenitor cells, which highly express microRNA-126 (miR-126), improved survival in mice subjected to cecal ligation and puncture (CLP) sepsis. miRNAs are important regulators of gene expression and cell function, play a major role in endothelial homeostasis, and may represent an emerging therapeutic modality. However, delivery of miRNAs to cells in vitro and in vivo is challenging due to rapid degradation by ubiquitous RNases. Herein, we developed a nanoparticle delivery system separately combining deacetylated poly-N-acetyl glucosamine (DEAC-pGlcNAc) polymers with miRNA-126-3p and miRNA-126-5p and testing these combinations in vitro and in vivo. Our results demonstrate that DEAC-pGlcNAc polymers have an appropriate size and zeta potential for cellular uptake and when complexed, DEAC-pGlcNAc protects miRNA from RNase A degradation. Further, DEAC-pGlcNAc efficiently encapsulates miRNAs as evidenced by preventing their migration in an agarose gel. The DEAC-pGlcNAc-miRNA complexes were taken up by multiple cell types and the delivered miRNAs had biological effects on their targets in vitro including pERK and DLK-1. In addition, we found that delivery of DEAC-pGlcNAc alone or DEAC-pGlcNAc:miRNA-126-5p nanoparticles to septic animals significantly improved survival, preserved vascular integrity, and modulated cytokine production. These composite studies support the concept that DEAC-pGlcNAc nanoparticles are an effective platform for delivering miRNAs and that they may provide therapeutic benefit in sepsis.


Subject(s)
Drug Carriers/chemistry , MicroRNAs/administration & dosage , Nanoparticles/chemistry , Sepsis/drug therapy , Acetylglucosamine/therapeutic use , Animals , Cecum/microbiology , Cytokines/metabolism , Endothelium, Vascular/metabolism , Ligation , Mice , Punctures/adverse effects , Sepsis/etiology , Sepsis/metabolism , Sepsis/mortality , Survival Rate
6.
J Infect Dis ; 218(12): 1995-2005, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30053030

ABSTRACT

Background: Pericytes are vascular mural cells and are embedded in the basement membrane of the microvasculature. Recent studies suggest a role for pericytes in lipopolysaccharide (LPS)-induced microvascular dysfunction and mortality, but the mechanisms of pericyte loss in sepsis are largely unknown. Methods: By using a cecal ligation and puncture (CLP)-induced murine model of sepsis, we observed that CLP led to lung and renal pericyte loss and reduced lung pericyte density and pericyte/endothelial cell (EC) coverage. Results: Up-regulated Friend leukemia virus integration 1 (Fli-1) messenger ribonucleic acid (RNA) and protein levels were found in lung pericytes from CLP mice in vivo and in LPS-stimulated lung pericytes in vitro. Knockout of Fli-1 in Foxd1-derived pericytes prevented CLP-induced pericyte loss, vascular leak, and improved survival. Disrupted Fli-1 expression by small interfering RNA inhibited LPS-induced inflammatory cytokines and chemokines in cultured lung pericytes. Furthermore, CLP-induced pericyte pyroptosis was mitigated in pericyte Fli-1 knockout mice. Conclusions: Our findings suggest that Fli-1 is a potential therapeutic target in sepsis.


Subject(s)
Pericytes/physiology , Proto-Oncogene Protein c-fli-1/metabolism , Sepsis/metabolism , Animals , Cecum , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation , Inflammation/metabolism , Ligation , Lipopolysaccharides , Lung/cytology , Mice , Mice, Knockout , Pyroptosis , Sepsis/immunology , Up-Regulation
7.
Mol Ther ; 26(5): 1375-1384, 2018 05 02.
Article in English | MEDLINE | ID: mdl-29599080

ABSTRACT

Microvascular dysfunction leads to multi-organ failure and mortality in sepsis. Our previous studies demonstrated that administration of exogenous endothelial progenitor cells (EPCs) confers protection in sepsis as evidenced by reduced vascular leakage, improved organ function, and increased survival. We hypothesize that EPCs protect the microvasculature through the exosomes-mediated transfer of microRNAs (miRNAs). Mice were rendered septic by cecal ligation and puncture (CLP), and EPC exosomes were administered intravenously at 4 hr after CLP. EPC exosomes treatment improved survival, suppressing lung and renal vascular leakage, and reducing liver and kidney dysfunction in septic mice. EPC exosomes attenuated sepsis-induced increases in plasma levels of cytokines and chemokine. Moreover, we determined miRNA contents of EPC exosomes with next-generation sequencing and found abundant miR-126-3p and 5p. We demonstrated that exosomal miR-126-5p and 3p suppressed LPS-induced high mobility group box 1 (HMGB1) and vascular cell adhesion molecule 1 (VCAM1) levels, respectively, in human microvascular endothelial cells (HMVECs). Inhibition of microRNA-126-5p and 3p through transfection with microRNA-126-5p and 3p inhibitors abrogated the beneficial effect of EPC exosomes. The inhibition of exosomal microRNA-126 failed to block LPS-induced increase in HMGB1 and VCAM1 protein levels in HMVECs and negated the protective effect of exosomes on sepsis survival. Thus, EPC exosomes prevent microvascular dysfunction and improve sepsis outcomes potentially through the delivery of miR-126.


Subject(s)
Endothelial Progenitor Cells/metabolism , Exosomes/metabolism , Sepsis/metabolism , Animals , Biomarkers , Capillary Permeability , Cell Line , Cytokines/metabolism , Disease Models, Animal , Endothelial Progenitor Cells/cytology , Humans , Mice , MicroRNAs/genetics , Organ Specificity , Prognosis , Sepsis/etiology , Sepsis/mortality
8.
Atherosclerosis ; 254: 271-281, 2016 11.
Article in English | MEDLINE | ID: mdl-27693002

ABSTRACT

Coronary artery disease and atherosclerosis are complex pathologies that develop over time due to genetic and environmental factors. Differential expression of miRNAs has been identified in patients with coronary artery disease and atherosclerosis, however, their association with cardiovascular disease risk factors, including hyperlipidemia, hypertension, obesity, diabetes, lack of physical activity and smoking, remains unclear. This review examines the role of miRNAs as either biomarkers or potential contributors to the pathophysiology of these aforementioned risk factors. It is intended to provide an overview of the published literature which describes alterations in miRNA levels in both human and animal studies of cardiovascular risk factors and when known, the possible mechanism by which these miRNAs may exert either beneficial or deleterious effects. The intent of this review is engage clinical, translational, and basic scientists to design future collaborative studies to further elucidate the potential role of miRNAs in cardiovascular diseases.


Subject(s)
Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/genetics , MicroRNAs/genetics , Animals , Biomarkers/blood , Cardiovascular Diseases/blood , Cell Communication , Cholesterol/blood , Coronary Artery Disease/blood , Coronary Artery Disease/genetics , Homeostasis , Humans , Hyperlipidemias/complications , Hypertension/complications , Lipoproteins, HDL/metabolism , Lipoproteins, LDL/metabolism , Mice , Obesity/complications , Risk Factors , Smoking/adverse effects , Nicotiana , Treatment Outcome
9.
Mol Cancer Ther ; 15(10): 2344-2356, 2016 10.
Article in English | MEDLINE | ID: mdl-27439478

ABSTRACT

The PI3K/AKT/mTOR pathway is among the most frequently altered pathways in cancer cell growth and survival. LY3023414 is a complex fused imidazoquinolinone with high solubility across a wide pH range designed to inhibit class I PI3K isoforms and mTOR kinase. Here, we describe the in vitro and in vivo activity of LY3023414. LY3023414 was highly soluble at pH 2-7. In biochemical testing against approximately 266 kinases, LY3023414 potently and selectively inhibited class I PI3K isoforms, mTORC1/2, and DNA-PK at low nanomolar concentrations. In vitro, inhibition of PI3K/AKT/mTOR signaling by LY3023414 caused G1 cell-cycle arrest and resulted in broad antiproliferative activity in cancer cell panel screens. In vivo, LY3023414 demonstrated high bioavailability and dose-dependent dephosphorylation of PI3K/AKT/mTOR pathway downstream substrates such as AKT, S6K, S6RP, and 4E-BP1 for 4 to 6 hours, reflecting the drug's half-life of 2 hours. Of note, equivalent total daily doses of LY3023414 given either once daily or twice daily inhibited tumor growth to similar extents in multiple xenograft models, indicating that intermittent target inhibition is sufficient for antitumor activity. In combination with standard-of-care drugs, LY3023414 demonstrated additive antitumor activity. The novel, orally bioavailable PI3K/mTOR inhibitor LY3023414 is highly soluble and exhibits potent in vivo efficacy via intermittent target inhibition. It is currently being evaluated in phase I and II trials for the treatment of human malignancies. Mol Cancer Ther; 15(10); 2344-56. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Biological Availability , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Drug Synergism , Enzyme Activation/drug effects , Humans , Hydrogen-Ion Concentration , Mice , Models, Molecular , Molecular Conformation , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Binding , Protein Kinase Inhibitors/chemistry , Signal Transduction/drug effects , Solubility , TOR Serine-Threonine Kinases/chemistry , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays
10.
Crit Care ; 19: 440, 2015 Dec 18.
Article in English | MEDLINE | ID: mdl-26683209

ABSTRACT

BACKGROUND: Endothelial dysfunction plays a critical role in the development of sepsis-related organ failure; however, the mechanisms that govern its development are not fully understood. Endothelial progenitor cells (EPCs) reduce vascular leak and organ failure in experimental sepsis while modulating plasma expression of microRNA (miRNA). MicroRNAs are small, noncoding segments of RNA that regulate gene expression and are known to modulate endothelial cell function and inflammatory signaling pathways. We hypothesized that miRNA may play an etiologic role in the endothelial dysfunction of sepsis and that their extracellular expression levels would be altered in those with shock. METHODS: Thirteen miRNAs were identified by literature search and analysis of the contents of human EPC-derived exosomes using real-time PCR. Plasma samples were obtained from patients within 24 hours of their admission to ICUs with severe sepsis (n = 62) and from healthy controls (n = 32) and real-time PCR was used to measure the expression of the candidate miRNAs. The Wilcoxon rank sum test was used to compare expression levels of the 13 candidate miRNAs in septic patients with (n = 29) and without (n = 33) shock while logistic regression was used to determine the area under the curve for associations between miRNA expression and shock. Bioinformatic analyses using miRNA databases were performed to identify pathways and gene targets of differentially expressed miRNA with potential relevance to sepsis-related shock. RESULTS: MiRNA-34a expression was significantly increased in the group who developed shock (p = 0.03) while miR-15a and miR-27a expressions were significantly decreased in this group (p = 0.006 and 0.03, respectively). The combined expression of these three miRNAs predicted shock with an area under the curve of 0.78 (95 % CI 0.66-0.90). In silico analyses predict that these three miRNAs regulate genes involved in endothelial cell cycle, apoptosis, VEGF signaling, LPS-stimulated MAPK signaling, and nuclear factor kappa B signaling. CONCLUSIONS: The plasma levels of miRNA are altered in patients with severe sepsis complicated by shock and may offer prognostic value as well as insights into the mechanisms of endothelial dysfunction in sepsis.


Subject(s)
MicroRNAs/analysis , Shock, Septic/pathology , Adult , Endothelial Cells/metabolism , Female , Humans , Male , MicroRNAs/blood , Middle Aged , Plasma/metabolism , Prognosis , Shock, Septic/complications , Shock, Septic/etiology , Shock, Septic/metabolism
11.
Innate Immun ; 21(6): 609-18, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25956304

ABSTRACT

The severity of sepsis is significantly affected by advanced age; however, age-dependent molecular mechanisms of this susceptibility are unknown. Nuclear liver X receptor-α (LXRα) is a regulator of lipid metabolism with associated anti-inflammatory properties. Here, we investigated the role of LXRα in age-dependent lung injury and outcome of sepsis. Male C57BL/6, LXRα-deficient (LXRα(-/-)) and wild type (WT) (LXRα(+/+)) mice of different ages were subjected to sepsis by cecal ligation and puncture (CLP). In pharmacological studies, treatment with the LXRα ligand T0901317 reduced lung neutrophil infiltration in C57BL/6 mice aged from 1 to 8 mo when compared with vehicle-treated animals subjected to CLP. The LXRα ligand improved survival in young mice (2-3 mo old) but did not affect survival or neutrophil infiltration in mature adult mice (11-13 mo old). Immunoblotting revealed an age-dependent decrease of lung LXRα levels. Young LXRα(-/-) mice (2-3 mo old) exhibited earlier mortality than age-matched WT mice after CLP. Lung damage and neutrophil infiltration, lung activation of the pro-inflammatory NF-κB and plasma IL-6 levels were higher in LXRα(-/-) mice 18 h after CLP compared with LXRα(+/+) mice. This study suggests that the anti-inflammatory properties of LXRα in sepsis are age-dependent and severely compromised in mature adult animals.


Subject(s)
Age Factors , Neutrophils/physiology , Orphan Nuclear Receptors/metabolism , Sepsis/immunology , Animals , Cecum/surgery , Cell Movement/drug effects , Cell Movement/genetics , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Hydrocarbons, Fluorinated/administration & dosage , Interleukin-6/blood , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Neutrophils/drug effects , Orphan Nuclear Receptors/agonists , Orphan Nuclear Receptors/genetics , Sulfonamides/administration & dosage
12.
Immunology ; 2014 Sep 08.
Article in English | MEDLINE | ID: mdl-25201453

ABSTRACT

Previous studies demonstrated that the CXCL12 peptide analogue CTCE-0214 (CTCE) has beneficial effects in experimental sepsis induced by cecal ligation and puncture (CLP). We examined the hypothesis that CTCE recruits neutrophils (PMN) to the site of infection, enhances PMN function and improves survival of mice in CLP-induced sepsis with antibiotic treatment. Septic mice (n=15) were administered imipenem (25mg/kg) and CTCE (10 mg/kg) subcutaneously vs. vehicle control at designated intervals post-CLP. CTCE treatment increased PMN recruitment in CLP-induced sepsis as evidenced by increased PMN in blood by 2.4±0.6 fold at 18h, 2.9±0.6 fold at 24h, respectively and in peritoneal fluid by 2.0±0.2 fold at 24h vs. vehicle control. CTCE treatment reduced bacterial invasion in blood (CFU decreased 77±11%), peritoneal fluid (CFU decreased 78±9%) and lung (CFU decreased 79±8% vs. CLP vehicle). The improved PMN recruitment and bacterial clearance correlated with reduced mortality with CTCE treatment (20% vs. 67% vehicle controls). In vitro studies support the notion that CTCE augments PMN function by enhancing phagocytic activity (1.25±0.02 fold), increasing intracellular production of ROS (32±4%) and improving bacterial killing (CFU decreased 27±3%). These composite findings support the hypothesis that specific CXCL12 analogues with ancillary antibiotic treatment are beneficial in experimental sepsis, in part, by augmenting PMN recruitment and function. This article is protected by copyright. All rights reserved.

13.
Am J Respir Crit Care Med ; 189(12): 1509-19, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24707934

ABSTRACT

RATIONALE: Endothelial progenitor cells (EPCs) have been associated with human sepsis but their role is incompletely understood. Stromal cell-derived factor (SDF)-1α facilitates EPC recruitment and is elevated in murine sepsis models. Previous studies have demonstrated that the SDF-1α analog CTCE-0214 (CTCE) is beneficial in polymicrobial sepsis induced by cecal ligation and puncture (CLP) in mice. OBJECTIVES: We hypothesized that exogenously administered EPCs are also beneficial in CLP sepsis and that CTCE provides synergistic benefit. METHODS: Mice were subjected to CLP and administered EPCs at varying doses, CTCE, or a combination of the two. Mouse survival, plasma miRNA expression, IL-10 production, and lung vascular leakage were determined. The in vitro effect of CTCE on miRNA expression and EPC function were determined. MEASUREMENTS AND MAIN RESULTS: Survival was improved with EPC therapy at a threshold of 10(6) cells. In coculture studies, EPCs augmented LPS-induced macrophage IL-10 production. In vivo EPC administration in sepsis increased plasma IL-10, suppressed lung vascular leakage, attenuated liver and kidney injury, and augmented miR-126 and -125b expression, which regulate endothelial cell function and/or inflammation. When subthreshold numbers of EPCs were coadministered with CTCE in CLP mice they synergistically improved survival. We demonstrated that CTCE recruits endogenous EPCs in septic mice. In in vitro analysis, CTCE enhanced EPC proliferation, angiogenesis, and prosurvival signaling while inhibiting EPC senescence. These cellular effects were, in part, explained by the effect of CTCE on miR-126, -125b, -34a, and -155 expression in EPCs. CONCLUSIONS: EPCs and CTCE represent important potential therapeutic strategies in sepsis.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Chemokine CXCL12/therapeutic use , Cord Blood Stem Cell Transplantation , Endothelial Cells/transplantation , Sepsis/therapy , Animals , Anti-Inflammatory Agents/pharmacology , Biomarkers/metabolism , Chemokine CXCL12/pharmacology , Combined Modality Therapy , Endothelial Cells/drug effects , Humans , Interleukin-10/metabolism , Male , Mice , MicroRNAs/metabolism , Sepsis/immunology , Sepsis/metabolism , Sepsis/mortality , Stem Cells/drug effects , Treatment Outcome
14.
Inflammation ; 35(5): 1611-7, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22581266

ABSTRACT

Previous studies have implicated a role of Gα(i) proteins as co-regulators of Toll-like receptor (TLR) activation. These studies largely derived from examining the effect of Gα(i) protein inhibitors or genetic deletion of Gα(i) proteins. However, the effect of increased Gα(i) protein function or Gα(i) protein expression on TLR activation has not been investigated. We hypothesized that gain of function or increased expression of Gα(i) proteins suppresses TLR2- and TLR4-induced inflammatory cytokines. Novel transgenic mice with genomic "knock-in" of a regulator of G protein signaling (RGS)-insensitive Gnai2 allele (Gα(i2)(G184S/G184S) ; GS/GS) were employed. These mice express essentially normal levels of Gα(i2) protein; however, the Gα(i2) is insensitive to its negative regulator RGS thus rendering more sustained Gα(i2) protein activation following ligand/receptor binding. In subsequent studies, we generated Raw 264.7 cells that stably overexpress Gα(i2) protein (Raw Gα(i2)). Peritoneal macrophages, splenocytes, and mouse embryonic fibroblasts (MEF) were isolated from WT and GS/GS mice and were stimulated with LPS, Pam3CSK4, or Poly (I:C). We also subjected WT and GS/GS mice to endotoxic shock (LPS, 25 mg/kg i.p.) and plasma tumor necrosis factor alpha (TNF-α) and interleukin (IL)-6 production were determined. We found that in vitro LPS and Pam3CSK4-induced TNF-α, and IL-6 production are decreased in macrophages from GS/GS mice compared with WT mice (p < 0.05). In vitro, LPS and Pam3CSK4 induced IL-6 production in splenocytes, and in vivo, LPS-induced IL-6 were suppressed in GS/GS mice. Poly (I:C)-induced TNF-α, and IL-6 in vitro demonstrated no difference between GS/GS mice and WT mice. LPS-induced IL-6 production was inhibited in MEFs from GS/GS mice similarly to macrophage and splenocytes. In parallel studies, Raw Gα(i2) cells also exhibit decreased TNF-α and IL-6 production in response to LPS and Pam3CSK4. These studies support our hypothesis that Gα(i2) proteins are novel negative regulators of TLR activation.


Subject(s)
GTP-Binding Protein alpha Subunit, Gi2/metabolism , Interleukin-6/biosynthesis , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Cell Line , Endotoxemia/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , GTP-Binding Protein alpha Subunit, Gi2/genetics , Lipopeptides/immunology , Lipopeptides/pharmacology , Lipopolysaccharides/immunology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Poly I-C/immunology
15.
Inflammation ; 35(1): 130-7, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21274742

ABSTRACT

The chemokine CXC receptor 4 (CXCR4) is activated by stromal cell-derived factor (SDF-1α). CXCR4 may be part of a lipopolysaccharide (LPS) sensing co-clustering complex that modulates TLR4 activation and evidence suggest that SDF-1α can activate anti-inflammatory signaling pathways and suppress inflammation. In the present study we examined the hypothesis that the SDF-1α peptide analog and CXCR4 agonist CTCE-0214 is anti-inflammatory in three distinct models of murine systemic inflammation. Our findings demonstrate that CTCE-0214 in vivo significantly suppressed plasma tumor necrosis factor alpha (TNF-α) increases in acute endotoxemia and following zymosan-induced multiple organ dysfunction syndrome (MODS). In both models, CTCE-0214 did not suppress plasma increases in the anti-inflammatory cytokine interleukin (IL)-10. CTCE-0214 improved survival without antibiotics in a model of severe sepsis induced by cecal ligation and puncture (CLP). CTCE-0214 also decreased plasma increases in IL-6 but not TNF-α and IL-10 in response to CLP-induced inflammation. We demonstrated in a moderately severe model of CLP (one puncture) that IL-6 levels at 24 h were similar to sham controls. However in severe CLP (two punctures) plasma IL-6 levels were markedly elevated. Plasma SDF-1α levels varied inversely with the plasma IL-6. In addition to the beneficial effect of CTCE-0214 in these models of systemic inflammation in vivo, we also demonstrated that the analog dose dependently suppressed LPS-induced IL-6 production in bone marrow-derived macrophages. CTCE-0214 therefore may be beneficial in controlling inflammation sepsis and systemic inflammatory syndromes.


Subject(s)
Chemokine CXCL12/metabolism , Multiple Organ Failure/drug therapy , Receptors, CXCR4/agonists , Receptors, CXCR4/immunology , Systemic Inflammatory Response Syndrome/drug therapy , Animals , Chemokine CXCL12/blood , Chemokine CXCL12/pharmacology , Disease Models, Animal , Endotoxemia/pathology , Interleukin-10/biosynthesis , Interleukin-10/blood , Interleukin-6/blood , Lipopolysaccharides/immunology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Multiple Organ Failure/chemically induced , Sepsis/drug therapy , Tumor Necrosis Factor-alpha/blood , Zymosan
16.
Mol Immunol ; 49(1-2): 64-74, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21855149

ABSTRACT

Pro-inflammatory cytokines and chemokines play critical roles in autoimmune diseases including rheumatoid arthritis (RA). Recently, it has been reported that ß-arrestin 1 and 2 are involved in the regulation of inflammation. We hypothesized that ß-arrestin 1 and 2 play critical roles in murine models of RA. Using a collagen-induced arthritis (CIA) and a human TNFα transgenic (TNFtg) mouse model, we demonstrated that ß-arrestin 1 and 2 expression are significantly increased in joint tissue of CIA mice and TNFtg mice. In fibroblast-like synoviocytes (FLS) isolated from hind knee joint of CIA mice, we observed an increase of ß-arrestin 1 and 2 protein and mRNA levels in the early stage of arthritis. In FLS, low molecular weight hyaluronan (HA)-induced TNFα and IL-6 production was increased by overexpression of ß-arrestin 1 but decreased by overexpression of ß-arrestin 2 demonstrating isoform specific regulation. TNFα and HA induced an increase of ß-arrestin 1 and 2 expression in FLS, while high mobility group box (HMGB)-1 only stimulated ß-arrestin 1 expression. TNFα- or HA-induced ß-arrestin 2 expression was blocked by a p38 inhibitor. To examine the in vivo role of ß-arrestin 2 in the pathogenesis of arthritis, WT and ß-arrestin 2 KO mice were subjected to collagen antibody-induced arthritis (CAIA). ß-Arrestin 2 KO mice exhibited more severe arthritis in CAIA. Thus ß-arrestin 2 is anti-inflammatory in CAIA. These composite observations suggest that ß-arrestin 1 and 2 differentially regulate FLS inflammation and increased ß-arrestin 2 may reduce experimental arthritis severity.


Subject(s)
Arrestins/biosynthesis , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/metabolism , Animals , Arrestins/immunology , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Blotting, Western , Disease Models, Animal , Fibroblasts/immunology , Fibroblasts/metabolism , Humans , Inflammation/immunology , Inflammation/metabolism , Mice , Mice, Inbred DBA , Mice, Knockout , Mice, Transgenic , Protein Isoforms/immunology , Protein Isoforms/metabolism , Real-Time Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , beta-Arrestin 1 , beta-Arrestin 2 , beta-Arrestins
17.
Biochim Biophys Acta ; 1813(3): 466-72, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21255617

ABSTRACT

Previous studies have implicated a role of heterotrimeric Gα(i) proteins in lipopolysaccharide (LPS)-induced inflammatory responses. We hypothesized that Toll-like receptor (TLR) signaling regulates Gα(i) proteins, which are anti-inflammatory in endotoxemia and polymicrobial sepsis. RAW 264.7 cells were stimulated with LPS and the Gα(i)-GTP protein complex was immunoprecipitated with a Gα(i) protein activation assay. In subsequent in vivo studies, the Gα(i) protein inhibitor pertussis toxin (PTx) or G(i) protein agonist mastoparan (MP-7) were administrated prior to endotoxemia. LPS-induced pro-inflammatory cytokines and mortality were determined. To examine the role of Gα(i2) in sepsis, Gα(i2) (-/-) and wildtype (WT) mice were subjected to cecal ligation and puncture (CLP) and monitored every 24 h for 120 h. Other mice were sacrificed 24 h after CLP. Peritoneal fluid, blood, and tissue samples were collected. Plasma pro-inflammatory cytokine production, bacterial load in peritoneal fluid, blood and lung tissue, myeloperoxidase (MPO) activity in lung and liver and different immune cell populations in spleen were studied. We found that Gα(i) proteins are rapidly activated by LPS followed by rapid inactivation. These studies provide the first direct evidence that Gα(i) proteins are modulated by TLR signaling. In following studies, PTx augmented LPS-induced plasma TNFα, IL-6, whereas MP-7 suppressed LPS-induced TNFα and decreased LPS-induced mortality. In sepsis studies, the survival rate post-CLP was significantly decreased in the Gα(i2) (-/-) mice compared to WT mice. CLP-induced plasma TNFα, IL-6, bacterial load in peritoneal fluid, blood and lung tissue and lung and liver MPO activity were significantly increased in Gα(i2) (-/-) compared to WT mice. Gα(i2) (-/-) mice also exhibited increased Th1 and Th2 responses compared to WT mice. Taken together, Gα(i) proteins are activated by LPS and negatively regulate endotoxemia and sepsis. Understanding the role of Gα(i2) protein in regulation of the inflammatory response in sepsis may provide novel targets for treatment of sepsis.


Subject(s)
Endotoxemia/immunology , GTP-Binding Protein alpha Subunits, Gi-Go/immunology , Lipopolysaccharides/immunology , Sepsis/immunology , Animals , Bacterial Load , Cecum/injuries , Cecum/surgery , Cell Line , Cytokines/blood , Cytokines/immunology , GTP-Binding Protein alpha Subunit, Gi2/genetics , GTP-Binding Protein alpha Subunit, Gi2/immunology , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Gene Deletion , Intercellular Signaling Peptides and Proteins , Mice , Mice, Knockout , Peptides/immunology , Peroxidase/immunology , Pertussis Toxin/immunology , Spleen/cytology , T-Lymphocytes, Helper-Inducer/cytology , Wasp Venoms/immunology
18.
Am J Pathol ; 177(4): 1834-47, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20709805

ABSTRACT

The nuclear peroxisome proliferator-activated receptor δ (PPARδ) is an important regulator of lipid metabolism. In contrast to its known effects on energy homeostasis, its biological role on inflammation is not well understood. We investigated the role of PPARδ in the modulation of the nuclear factor-κB (NF-κB)-driven inflammatory response to polymicrobial sepsis in vivo and in macrophages in vitro. We demonstrated that administration of GW0742, a specific PPARδ ligand, provided beneficial effects to rats subjected to cecal ligation and puncture, as shown by reduced systemic release of pro-inflammatory cytokines and neutrophil infiltration in lung, liver, and cecum, when compared with vehicle treatment. Molecular analysis revealed that treatment with GW0742 reduced NF-κB binding to DNA in lung and liver. In parallel experiments, heterozygous PPARδ-deficient mice suffered exaggerated lethality when subjected to cecal ligation and puncture and exhibited severe lung injury and higher levels of circulating tumor necrosis factor-α (TNFα) and keratinocyte-derived chemokine than wild-type mice. Furthermore, in lipopolysaccharide-stimulated J774.A1 macrophages, GW0742 reduced TNFα production by inhibiting NF-κB activation. RNA silencing of PPARδ abrogated the inhibitory effects of GW0742 on TNFα production. Chromatin immunoprecipitation assays revealed that PPARδ displaced the NF-κB p65 subunit from the κB elements of the TNFα promoter, while recruiting the co-repressor BCL6. These data suggest that PPARδ is a crucial anti-inflammatory regulator, providing a basis for novel sepsis therapies.


Subject(s)
Bacteremia/prevention & control , Inflammation/prevention & control , NF-kappa B/metabolism , PPAR delta/physiology , Sepsis/metabolism , Sepsis/microbiology , Animals , Bacteremia/etiology , Bacteremia/metabolism , Blotting, Western , Cecum/immunology , Cecum/metabolism , Cecum/microbiology , Cell Nucleus/metabolism , Cells, Cultured , Chromatin Immunoprecipitation , Disease Models, Animal , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Hypotension , Immunoenzyme Techniques , Inflammation/etiology , Inflammation/metabolism , Luciferases/metabolism , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Knockout , NF-kappa B/genetics , PPAR delta/agonists , PPAR delta/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Sepsis/immunology , Signal Transduction , Survival Rate , Thiazoles/pharmacology
19.
Immunology ; 130(3): 344-51, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20465566

ABSTRACT

SUMMARY: Beta-arrestins 1 and 2 are ubiquitously expressed proteins that alter signalling by G-protein-coupled receptors. beta-arrestin 2 plays an important role as a signalling adaptor and scaffold in regulating cellular inflammatory responses. We hypothesized that beta-arrestin 2 is a critical modulator of inflammatory response in experimental sepsis. beta-arrestin 2(-/-) and wild-type (WT) mice were subjected to caecal ligation and puncture (CLP). The survival rate was significantly decreased (P < 0.05) in beta-arrestin 2(-/-) mice (13% survival) compared with WT mice (53% survival). A second group of mice were killed 18 hr after CLP for blood, peritoneal lavage and tissue sample collection. CLP-induced plasma interleukin (IL)-6 was significantly increased 25 +/- 12 fold and caecal myeloperoxidase (MPO) activity was increased 2.4 +/- 0.3 fold in beta-arrestin 2(-/-) compared with WT mice. beta-arrestin 2(-/-) mice exhibited more severe lung damage and higher bacterial loads compared with WT mice post CLP challenge as measured by histopathology and colony-forming unit count. In subsequent experiments, splenocytes, peritoneal macrophages and bone marrow-derived macrophages (BMDMs) were isolated and cultured from beta-arrestin 2(-/-) and WT mice and stimulated in vitro with lipopolysaccharide (LPS). Tumour necrosis factor (TNF)-alpha, IL-6 and IL-10 production induced by LPS was significantly augmented (2.2 +/- 0.2 fold, 1.8 +/- 0.1 fold, and 2.2 +/- 0.4 fold, respectively; P < 0.05) in splenocytes from beta-arrestin 2(-/-) mice compared with WT mice. The splenocyte response was different from that of peritoneal macrophages or BMDMs, which exhibited no difference in TNF-alpha and IL-6 production upon LPS stimulation between WT and beta-arrestin 2(-/-) mice. Our data demonstrate that beta-arrestin 2 functions to negatively regulate the inflammatory response in polymicrobial sepsis.


Subject(s)
Arrestins/metabolism , Inflammation/etiology , Inflammation/metabolism , Sepsis/complications , Sepsis/metabolism , Animals , Arrestins/genetics , Blood/microbiology , Cecum/enzymology , Inflammation/genetics , Inflammation/immunology , Interleukin-10/metabolism , Interleukin-6/blood , Interleukin-6/metabolism , Liver/enzymology , Liver/pathology , Lung/microbiology , Lung/pathology , Macrophages/metabolism , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Peritoneal Cavity/microbiology , Peroxidase/metabolism , Sepsis/genetics , Sepsis/pathology , Spleen/cytology , Spleen/metabolism , Survival Analysis , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha/metabolism , beta-Arrestin 2 , beta-Arrestins
20.
Int J Clin Exp Med ; 1(1): 32-41, 2008.
Article in English | MEDLINE | ID: mdl-19079685

ABSTRACT

Heterotrimeric Gi proteins have been previously implicated in signaling leading to inflammatory mediator production induced by bacterial lipopolysaccharide (LPS). beta-arrestins are ubiquitously expressed proteins that alter G-protein-coupled receptors signaling. beta-arrestin 2 plays a multifaceted role as a scaffold protein in regulating cellular inflammatory responses. Polymorphonuclear leukocytes (PMNs) activated by LPS induce inflammatory responses resulting in organ injury during sepsis. We hypothesized that beta-arrestin 2 is a critical modulator of inflammatory responses in PMNs. To examine the potential role of beta-arrestin 2 in LPS-induced cellular activation, we studied homozygous beta-arrestin 2 (-/-), heterozygous (+/-), and wildtype (+/+) mice. PMNs were stimulated with LPS for 16h. There was increased basal TNFalpha and IL-6 production in the beta-arrestin 2 (-/-) compared to both beta-arrestin 2 (+/-) and (+/+) cells. LPS failed to stimulate TNFalpha production in the beta-arrestin 2 (-/-) PMNs. However, LPS stimulated IL-6 production was increased in the beta-arrestin 2 (-/-) cells compared to (+/+) cells. In subsequent studies, peritoneal PMN recruitment was increased 81% in the beta-arrestin 2 (-/-) mice compared to (+/+) mice (p<0.05). beta-arrestin 2 deficiency resulted in an augmented expression of CD18 and CD62L (p<0.05). In subsequent studies, beta-arrestin 2 (-/-) and (+/+) mice were subjected to cecal ligation and puncture (CLP) and lung was collected and analyzed for myeloperoxidase activity (MPO) as index of PMNs infiltrate. CLP-induced MPO activity was significantly increased (p<0.05) in the beta-arrestin 2 (-/-) compared to (+/+) mice. These studies demonstrate that beta-arrestin 2 is a negative regulator of PMN activation and pulmomary leukosequestration in response to polymicrobial sepsis.

SELECTION OF CITATIONS
SEARCH DETAIL
...