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










Publication year range
1.
J Surg Res ; 168(2): e195-202, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21470634

ABSTRACT

BACKGROUND: Accelerated cardiovascular disease in patients with type I diabetes (TID) is a well-described condition and serious clinical obstacle. At present, the notion that early atherogenesis is largely dependent on sustained hyperglycemia remains in question. We hypothesize that an alteration in T lymphocyte homeostasis may result in early vascular inflammation, which might amplify subsequent blood vessel injury in euglycemia. METHODS: A murine model of carotid arterial ligation was employed to induce neointimal hyperplasia (NIH) in C57/Bl6 (non-autoimmune) and non-obese diabetic (NOD) mice. Additionally, adoptive transfer of NOD splenocytes into immunodeficient NOD mice (NOD.scid) was undertaken to evaluate the influence of restored autoimmunity on NIH development. RESULTS: Interestingly, compared with C57/Bl6 mice, the NOD demonstrate a significant increase in neointimal area. Conversely, the NOD.scid mice (immunodeficient control) reveal almost no evidence of vascular injury. While evidence of early vascular inflammation can be detected in the injured NOD vasculature, uninjured contralateral vessels and those of the NOD.scid have minimal T cell infiltration. Following reconstitution of autoimmune responses via NOD splenocyte adoptive transfer, accelerated vascular pathology is restored. CONCLUSIONS: These observations suggest that autoimmunity, in the setting of impending hyperglycemia, may contribute to accelerated vascular inflammation and subsequent pathology.


Subject(s)
Carotid Artery Diseases/immunology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 1/complications , Diabetic Angiopathies/immunology , T-Lymphocytes/physiology , Adoptive Transfer , Animals , CD4 Lymphocyte Count , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Neointima/immunology , Spleen/immunology
2.
Biochem J ; 432(1): 35-45, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20799926

ABSTRACT

Lyso-PCs (lysophosphatidylcholines) are a mixture of lipids that accumulate during storage of cellular blood components, have been implicated in TRALI (transfusion-related acute lung injury) and directly affect the physiology of neutrophils [PMNs (polymorphonuclear leucocytes)]. Because the G2A receptor, expressed on PMNs, has been reported to recognize lyso-PCs, we hypothesize that lyso-PC activation of G2A causes the increases in cytosolic Ca²(+) via release of G(α) and G(ßγ) subunits, kinase activation, and the recruitment of clathrin, ß-arrestin-1 and GRK6 (G-protein receptor kinase 6) to G2A for signal transduction. PMNs were isolated by standard techniques, primed with lyso-PCs for 5-180 s, and lysed for Western blot analysis, immunoprecipitation or subcellular fractionation, or fixed and smeared on to slides for digital microscopy. The results demonstrated that lyso-PCs cause rapid activation of the G2A receptor through S-phosphorylation and internalization resulting in G(αi)₋1 and G(αq/)11 release leading to increases in cytosolic Ca²(+), which was inhibited by an antibody to G2A or intracellular neutralization of these subunits. Lyso-PCs also caused the release of the G(ßγ) subunit which demonstrated a physical interaction (FRET+) with activated Hck (haemopoietic cell kinase; Tyr4¹¹). Moreover, G2A recruited clathrin, ß-arrestin-1 and GRK6: clathrin is important for signal transduction, GRK6 for receptor de-sensitization, and ß-arrestin-1 both propagates and terminates signals. We conclude that lyso-PC activation of G2A caused release of G(αi)₋1, G(αq/)11 and G(ßγ), resulting in cytosolic Ca²(+) flux, Hck activation, and recruitment of clathrin, ß-arrestin-1 and GRK6.


Subject(s)
Calcium/metabolism , Cell Cycle Proteins/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Lysophosphatidylcholines/pharmacology , Neutrophils/drug effects , Protein Kinases/metabolism , Receptors, G-Protein-Coupled/metabolism , Arrestins/metabolism , Blotting, Western , Cells, Cultured , Clathrin/metabolism , Cytosol/drug effects , Cytosol/metabolism , Enzyme Activation/drug effects , Fluorescence Resonance Energy Transfer , G-Protein-Coupled Receptor Kinases/metabolism , GTP-Binding Protein alpha Subunits/metabolism , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Humans , Ion Transport/drug effects , Microscopy, Fluorescence/methods , Neutrophils/cytology , Neutrophils/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-hck/metabolism , Signal Transduction/drug effects , Time Factors , beta-Arrestin 1 , beta-Arrestins
3.
Dis Esophagus ; 23(5): 430-6, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20095996

ABSTRACT

Exposure of esophageal epithelium to gastric and duodenal contents results in the histologic changes of hyperproliferation and mucosal thickening. We have previously shown that presence of secretory phospholipase A(2) (sPLA(2)) is necessary to produce these histologic changes in a murine model of gastroduodenal reflux. We sought to determine the influence of gastroduodenal reflux (GDR) on sPLA(2) protein and mRNA levels as well as enzyme activity in esophageal tissue. BALB/c (sPLA(2)(+/+)) mice (n= 28) underwent side-to-side surgical anastomosis of the first portion of the duodenum and GE junction (DGEA) resulting in continuous exposure of esophageal mucosa to mixed gastric and duodenal contents. Sham control mice (n= 14) underwent laparotomy, esophagotomy and closure. Real-time RT PCR was used to quantitate the influence of GDR on group IIa sPLA(2) expression. Immunofluorescent staining was quantitated by digital microscopy using a specific antibody to identify and locate sPLA(2) protein. A colorimetric assay was used to quantify total sPLA(2) activity after standardization of protein levels. Statistical analysis was conducted using Student's t-test. Group IIa sPLA(2) mRNA and protein levels were increased at 4 and 8 weeks compared with sham controls. This increase occurred in a time-dependent manner and correlated with esophageal mucosal thickness. Furthermore, sPLA(2) enzyme activity was increased significantly at 4 and 8 weeks compared with untreated controls. The expression of group IIa sPLA(2) as well as sPLA(2) activity is induced by GDR. This novel finding indicates that sPLA(2) may play a role in the development of the histologic changes produced by GDR in esophageal mucosa.


Subject(s)
Esophagitis, Peptic/enzymology , Esophagus/enzymology , Gastroesophageal Reflux/enzymology , Group II Phospholipases A2/metabolism , Animals , Disease Models, Animal , Esophagitis, Peptic/pathology , Esophagus/pathology , Fluorescent Antibody Technique , Gastroesophageal Reflux/pathology , Group II Phospholipases A2/genetics , Hyperplasia/pathology , Male , Mice , Mice, Inbred BALB C , Mucous Membrane/enzymology , Mucous Membrane/pathology , RNA, Messenger , Reverse Transcriptase Polymerase Chain Reaction
4.
Am J Physiol Cell Physiol ; 298(3): C714-24, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19907017

ABSTRACT

Neutrophils (PMNs) are a vital part of host defense and are the principal leukocyte in innate immunity. Interleukin (IL)-18 is a proinflammatory cytokine with roles in both innate and adaptive immunity. We hypothesize that PMNs contain preformed IL-18, which is released in response to specific inflammatory stimuli. Isolated PMNs were stimulated with a battery of chemoattractants (5 min to 24 h), and IL-18 release was measured. PMNs were also separated into subcellular fractions and immunoblotted with antibodies against IL-18 or were fixed and probed with antibodies to IL-18 as well as to the contents of granules, intracellular organelles, and filamentous actin (F-actin), incubated with fluorescent secondary antibodies, and examined by digital microscopy. Quiescent PMNs contained IL-18 in the cytoplasm, associated with F-actin, as determined by positive fluorescence resonance energy transfer (FRET+). In turn, TNF-alpha stimulation disrupted the association of IL-18 with F-actin, induced a FRET+ interaction of IL-18 with lipid rafts, and elicited IL-18 release. Manipulation of F-actin status confirmed the relationship between IL-18 and F-actin in resting PMNs. Consequently, incubation with monomeric IL-18 binding protein inhibited TNF-alpha-mediated priming of the PMN oxidase. We conclude that human PMNs contain IL-18 associated with F-actin in the cytoplasm and TNF-alpha stimulation causes dissociation of IL-18 from F-actin, association with lipid rafts, and extracellular release. Extracellular IL-18 participates in TNF-alpha priming of the PMN oxidase as demonstrated by inhibition with the IL-18 binding protein.


Subject(s)
Cytosol/immunology , Inflammation Mediators/metabolism , Interleukin-18/metabolism , Neutrophils/immunology , Tumor Necrosis Factor-alpha/metabolism , Actins/metabolism , Adaptive Immunity , Fluorescence Resonance Energy Transfer , Fluorescent Antibody Technique, Indirect , Humans , Immunity, Innate , Immunoblotting , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Microdomains/immunology , Microscopy, Fluorescence , Phosphoproteins/metabolism , Time Factors
5.
J Gastrointest Surg ; 13(12): 2212-8, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19672667

ABSTRACT

BACKGROUND: The initial response of esophageal mucosa to gastroduodenal reflux is inflammation and hyperplasia. Secretory phospholipase A(2) (sPLA(2)) is a known mediator of gut inflammation, and its levels are increased in Barrett's esophagus. We hypothesized that the sPLA(2) gene is required to produce esophageal mucosal hyperplasia in response to gastroduodenal reflux. METHODS: C57BL/6 (n = 5) sPLA(2) (-/-) mice and C57BL/6( Cg-Tg(PLA2G2A)703N16 ) mice (n = 4) sPLA(2) (-/+) underwent a side-to-side surgical anastomosis between the duodenum and gastroesophageal junction (DGEA). Control animals [sPLA(2) (-/-) (n = 5), sPLA(2) (-/+) (n = 4)] underwent laparotomy with incision and repair of the esophagus. Tissue was harvested after 4 weeks, and H&E staining was performed to quantify esophageal mucosal thickness. Ki67 and sPLA(2) immunostaining were performed to quantitate differences in cell division and sPLA(2) expression. RESULTS: Mice expressing human sPLA(2) had a 2.5-fold increase in thickness of the esophageal mucosa as compared to controls (p = 0.01). A 6.5-fold increase in proliferation (p = 0.02) and a twofold increase in sPLA(2) expression (p = 0.04) were demonstrated in animals exposed to gastroduodenal reflux. CONCLUSIONS: The presence of sPLA(2) is necessary for early mucosal hyperplasia produced by exposure of the esophagus to gastroduodenal contents. sPLA(2) expression is upregulated by gastroduodenal reflux, strengthening its role as a critical mediator of early mucosal hyperplasia.


Subject(s)
Esophagus/pathology , Gastroesophageal Reflux/pathology , Phospholipases A2, Secretory/genetics , Animals , Cell Division , Duodenogastric Reflux/metabolism , Duodenogastric Reflux/pathology , Fluorescent Antibody Technique , Gene Expression , Mice , Mice, Inbred C57BL , Mucous Membrane/pathology , Phospholipases A2, Secretory/analysis
6.
Am J Physiol Cell Physiol ; 297(4): C886-97, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19295175

ABSTRACT

Receptor signaling is integral for adhesion, emigration, phagocytosis, and reactive oxygen species production in polymorphonuclear neutrophils (PMNs). Priming is an important part of PMN emigration, but it can also lead to PMN-mediated organ injury in the host. Platelet-activating factor (PAF) primes PMNs through activation of a specific G protein-coupled receptor. We hypothesize that PAF priming of PMNs requires clathrin-mediated endocytosis (CME) of the PAF receptor (PAFr), and, therefore, amantadine, known to inhibit CME, significantly antagonizes PAF signaling. PMNs were isolated by standard techniques to >98% purity and tested for viability. Amantadine (1 mM) significantly inhibited the PAF-mediated changes in the cellular distribution of clathrin and the physical colocalization [fluorescence resonance energy transfer positive (FRET+)] of early endosome antigen-1 and Rab5a, known components of CME and similar to hypertonic saline, a known inhibitor of CME. Furthermore, amantadine had no effect on the PAF-induced cytosolic calcium flux; however, phosphorylation of p38 MAPK was significantly decreased. Amantadine inhibited PAF-mediated changes in PMN physiology, including priming of the NADPH oxidase and shape change with lesser inhibition of increases in CD11b surface expression and elastase release. Furthermore, rimantadine, an amantadine analog, was a more potent inhibitor of PAF priming of the N-formyl-methionyl-leucyl-phenylalanine-activated oxidase. PAF priming of PMNs requires clathrin-mediated endocytosis that is inhibited when PMNs are pretreated with either amantadine or rimantadine. Thus, amantadine and rimantadine have the potential to ameliorate PMN-mediated tissue damage in humans.


Subject(s)
Amantadine/pharmacology , Clathrin/metabolism , Endocytosis , Neutrophils/physiology , Platelet Activating Factor/physiology , Platelet Membrane Glycoproteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Antigens, CD1/metabolism , Enzyme Activation , Humans , In Vitro Techniques , NADPH Oxidases/metabolism , Neutrophils/drug effects , Platelet Activating Factor/pharmacology , Platelet Membrane Glycoproteins/antagonists & inhibitors , Receptors, G-Protein-Coupled/antagonists & inhibitors , Rimantadine/pharmacology , Signal Transduction
7.
Blood ; 113(9): 2079-87, 2009 Feb 26.
Article in English | MEDLINE | ID: mdl-19131548

ABSTRACT

Transfusion-related acute lung injury (TRALI) is the leading cause of transfusion death. We hypothesize that TRALI requires 2 events: (1) the clinical condition of the patient and (2) the infusion of antibodies against MHC class I antigens or the plasma from stored blood. A 2-event rat model was developed with saline (NS) or endotoxin (LPS) as the first event and the infusion of plasma from packed red blood cells (PRBCs) or antibodies (OX18 and OX27) against MHC class I antigens as the second event. ALI was determined by Evans blue dye leak from the plasma to the bronchoalveolar lavage fluid (BALF), protein and CINC-1 concentrations in the BALF, and the lung histology. NS-treated rats did not evidence ALI with any second events, and LPS did not cause ALI. LPS-treated animals demonstrated ALI in response to plasma from stored PRBCs, both prestorage leukoreduced and unmodified, and to OX18 and OX27, all in a concentration-dependent fashion. ALI was neutrophil (PMN) dependent, and OX18/OX27 localized to the PMN surface in vivo and primed the oxidase of rat PMNs. We conclude that TRALI is the result of 2 events with the second events consisting of the plasma from stored blood and antibodies that prime PMNs.


Subject(s)
Acute Lung Injury/etiology , Antibodies/adverse effects , Erythrocyte Transfusion/adverse effects , Erythrocytes/physiology , Histocompatibility Antigens Class I/immunology , Plasma/physiology , Acute Lung Injury/immunology , Acute Lung Injury/pathology , Animals , Blood Preservation/adverse effects , Bronchoalveolar Lavage Fluid/immunology , Disease Models, Animal , Humans , Male , Neutrophil Activation/immunology , Plasma/immunology , Rats , Rats, Sprague-Dawley
8.
Shock ; 31(5): 466-72, 2009 May.
Article in English | MEDLINE | ID: mdl-18948845

ABSTRACT

Resuscitation with hypertonic saline (HTS) attenuates acute lung injury (ALI) and modulates postinjury hyperinflammation. TNF-alpha-stimulated pulmonary epithelium is a major contributor to hemorrhage-induced ALI. We hypothesized that HTS would inhibit TNF-alpha-induced nuclear factor (NF)-kappaB proinflammatory signaling in pulmonary epithelial cells. Therefore, we pretreated human pulmonary epithelial cells (A549) with hypertonic medium (180 mM NaCl) for 30 min, followed by TNF-alpha stimulation (10 ng/mL). Key regulatory steps and protein concentrations in this pathway were assessed for significant alterations. Hypertonic saline significantly reduced TNF-alpha-induced intercellular adhesion molecule 1 levels and NF-kappaB nuclear localization. The mechanism is attenuated phosphorylation and delayed degradation of IkappaB alpha. Hypertonic saline did not alter TNF-alpha-induced p38 mitogen-activated protein kinase phosphorylation or constitutive vascular endothelial growth factor expression, suggesting that the observed inhibition is not a generalized suppression of protein phosphorylation or cellular function. These results show that HTS inhibits TNF-alpha-induced NF-kappaB activation in the pulmonary epithelium and, further, our understanding of its beneficial effects in hemorrhage-induced ALI.


Subject(s)
Epithelial Cells/drug effects , Epithelial Cells/metabolism , Lung/cytology , NF-kappa B/metabolism , Saline Solution, Hypertonic/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Blotting, Western , Cell Line , Cell Survival/drug effects , Flow Cytometry , Humans , Microscopy, Fluorescence , Phosphorylation/drug effects , Protein Transport/drug effects
9.
J Thorac Cardiovasc Surg ; 135(6): 1220-7, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18544357

ABSTRACT

OBJECTIVE: The earliest response of esophageal mucosa to gastric reflux is the development of oxidative damage and inflammation. These processes contribute to the development of metaplasia known as Barrett's esophagus, as well as the progression to malignancy. Secretory phospholipase A(2) is a mediator of inflammation with levels that are increased in Barrett's metaplasia and carcinoma when compared with levels in normal samples. Our goal is to determine the role of secretory phospholipase A(2) in the development of reflux-associated changes in the esophageal mucosa. METHODS: Secretory phospholipase A(2)-deficient mice (C57BL/6, n = 5) and mice known to express high levels of secretory phospholipase A(2) (BALB/c, n = 5) underwent side-to-side surgical anastomosis of the first portion of the duodenum and gastroesophageal junction, allowing exposure of esophageal mucosa to duodenal and gastric contents duodeno-gastroesophageal anastomosis. Control animals (n = 5) of each strain underwent laparotomy with esophagotomy and repair. Tissue was frozen in embedding medium. Hematoxylin and eosin staining and Ki67 and secretory phospholipase A(2) immunohistochemistry were used to evaluate esophageal tissue and its response to duodeno-gastroesophageal anastomosis. RESULTS: Immunofluorescent staining confirmed the absence of secretory phospholipase A(2) in C57BL/6 mice and its presence in BALB/c mice. Hematoxylin and eosin staining demonstrated significant thickening of the esophageal mucosa in response to gastroesophageal reflux in the presence of secretory phospholipase A(2). Mice known to express high levels of secretory phospholipase A(2) also demonstrated increased numbers of proliferating cells. Secretory phospholipase A(2)-deficient mice were immune to the early changes induced by mixed reflux. CONCLUSIONS: The presence of secretory phospholipase A(2) appears necessary for early histologic changes produced by exposure of the esophagus to gastroduodenal contents. This enzyme is identified as a promising target for evaluation of mechanisms of carcinogenesis and chemoprevention of esophageal carcinoma.


Subject(s)
Barrett Esophagus/pathology , Cell Transformation, Neoplastic/pathology , Disease Models, Animal , Esophageal Neoplasms/pathology , Phospholipases A2/metabolism , Analysis of Variance , Animals , Barrett Esophagus/enzymology , Biomarkers, Tumor/analysis , Biopsy, Needle , Esophageal Neoplasms/metabolism , Fluorescent Antibody Technique , Gastroesophageal Reflux/enzymology , Gastroesophageal Reflux/pathology , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Phospholipases A2/deficiency , Precancerous Conditions/pathology , Probability , Random Allocation , Sensitivity and Specificity , Tissue and Organ Harvesting
10.
J Immunol ; 180(12): 8192-203, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18523285

ABSTRACT

Neutrophils (polymorphonuclear leukocytes, PMNs) are vital to innate immunity and receive proinflammatory signals that activate G protein-coupled receptors (GPCRs). Because GPCRs transduce signals through clathrin-mediated endocytosis (CME), we hypothesized that platelet-activating factor (PAF), an effective chemoattractant that primes the PMN oxidase, would signal through CME, specifically via dynamin-2 activation and endosomal formation resulting in membrane translocation of cytosolic phagocyte oxidase (phox) proteins. PMNs were incubated with buffer or 2 muM PAF for 1-3 min, and in some cases activated with PMA, and O(2)(-) was measured, whole-cell lysates and subcellular fractions were prepared, or the PMNs were fixed onto slides for digital or electron microscopy. PAF caused activation of dynamin-2, resulting in endosomal formation that required PI3K and contained early endosomal Ag-1 (EEA-1) and Rab5a. The apoptosis signal-regulating kinase-1/MAPK kinase-3/p38 MAPK signalosome assembled on Rab5a and phosphorylated EEA-1 and Rab GDP dissociation inhibitor, with the latter causing Rab5a activation. Electron microscopy demonstrated that PAF caused two distinct sites for activation of p38 MAPK. EEA-1 provided a scaffold for recruitment of the p40(phox)-p67(phox) complex and PI3K-dependent Akt1 phosphorylation of these two phox proteins. PAF induced membrane translocation of p40(phox)-p67(phox) localizing to gp91(phox), which was PI3K-, but not p47(phox)-, dependent. In conclusion, PAF transduces signals through CME, and such GPCR signaling may allow for pharmacological manipulation of these cells to decrease PMN-mediated acute organ injury.


Subject(s)
Cell Membrane/metabolism , Endosomes/metabolism , Neutrophils/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Platelet Activating Factor/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , rab5 GTP-Binding Proteins/metabolism , Cell Membrane/enzymology , Dynamin II/metabolism , Endosomes/enzymology , Enzyme Activation/physiology , Fluorescence Resonance Energy Transfer , Humans , Ligands , MAP Kinase Signaling System/physiology , Neutrophils/enzymology , Platelet Activating Factor/metabolism , Platelet Membrane Glycoproteins/metabolism , Protein Transport/physiology , Receptors, G-Protein-Coupled/metabolism , Vesicular Transport Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/physiology
11.
J Surg Res ; 143(1): 136-40, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17950083

ABSTRACT

BACKGROUND: Posthemorrhagic shock mesenteric lymph (PSML) has been shown to activate pulmonary endothelial cells and cause lung injury. Although multiple mediators may be involved, most of these effects are mediated by nuclear factor-kappa B (NF-kappaB) activation. Degradation of the inhibitor of kappa B (IkappaB) is a key regulatory step in the activation of NF-kappaB. We therefore hypothesized that PSML would cause IkappaB degradation with subsequent NF-kappaB phosphorylation and nuclear translocation. METHODS: Mesenteric lymph was collected from male rats before shock and each hour after shock for up to 3 h (n = 5). Buffer (control), buffer + 10% (v/v) lymph, or buffer + tumor necrosis factor (10 ng/mL) were incubated with human pulmonary endothelial cells for 30 min and then lysed. Immunoblots of lysates were probed for IkappaB and phospho-p65. Immunohistochemistry was performed on cells grown on glass slides and then treated as above with the third PSML sample. Cells were fixed and then probed for p65. Statistical analysis was performed with Student's t-test and analysis of variance with significance was set at P < 0.05. RESULTS: Western blots of cell lysates for IkappaB demonstrated a steady decrease in total IkappaB with each lymph sample. Phosphorylation of NF-kappaB , p65 component, steadily increased with each PSML sample, with a maximum reached during the third PSML sample, which also significantly increased translocation of NF-kappaB to the nucleus. CONCLUSION: Postshock mesenteric lymph bioactivity is mediated by pathways which involved IkappaB degradation. These pathways offer novel off targets for clinical intervention to prevent the distal organ injury caused by postinjury hemorrhagic shock.


Subject(s)
I-kappa B Kinase/metabolism , Lymph/metabolism , Mesentery/metabolism , NF-kappa B/metabolism , Shock, Hemorrhagic/metabolism , Animals , Cell Nucleus/metabolism , Cells, Cultured , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Male , Mesentery/pathology , Phosphorylation , Rats , Rats, Sprague-Dawley , Shock, Hemorrhagic/pathology
12.
Surgery ; 140(2): 268-72, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16904979

ABSTRACT

BACKGROUND: Tumor necrosis factor receptor I recruits tumor necrosis factor receptor-associated death domain (TRADD) and multiple kinases that ultimately phosphorylate inhibitor kappa B (IKB alpha). Degradation of phospho-IKB alpha (p-IKB alpha) frees nuclear factor kappa B (NFKB) to be active and phosphorylated. Many receptors require clathrin-mediated endocytosis to provide the scaffolds necessary for signaling. Therefore, we investigated the role of clathrin heavy chain (CHC) in tumor necrosis factor alpha (TNF-alpha)-induced IKB alpha phosphorylation and NFKB activation. We hypothesized that CHC was required for TNF-alpha-induced inflammatory signaling. METHODS: We treated human pulmonary epithelial cells with small interfering RNA to knock down intracellular CHC (CHCsil). TRADD and scrambled (noncoding) small interfering RNA sequences were used as positive and negative controls, respectively. Treatment groups were exposed to 10 ng/mL of TNF-alpha. Total I kappaB alpha, p-I kappaB alpha, and phosphorylated P65 (a subunit of NFKB) were determined by immunoblot staining. Densitometry was normalized to controls for the analysis of the stains. TNF-alpha-induced release of monocyte chemoattractant protein 1 (MCP-1) was determined by enzyme-linked immunosorbent assay. Statistical analyses were determined by analysis of variance or paired t test as appropriate. RESULTS: TNF-alpha-induced I kappaB alpha phosphorylation and degradation at 5 and 30 minutes, respectively, and induced P65 phosphorylation. CHCsil diminished p-I kappaB alpha by 91% (P < .03); however, I kappaB alpha degradation was not affected. CHC knockdown caused a 66% decrease in P65 phosphorylation after 3 minutes of TNF-alpha. CHCsil decreased TNF-alpha-induced MCP-1 by 46% (P < .05), compared with control. CONCLUSIONS: CHCsil significantly impairs phosphorylation of both I kappaB alpha and P65. CHCsil also significantly decreased MCP-1 production. These data suggest that CHC is required for certain TNF-alpha-induced, inflammatory signaling pathways.


Subject(s)
Clathrin Heavy Chains/physiology , Epithelial Cells/metabolism , I-kappa B Proteins/metabolism , Signal Transduction/physiology , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/physiology , Cell Culture Techniques , Chemokine CCL2/metabolism , Humans , NF-KappaB Inhibitor alpha , Phosphorylation , RNA, Small Interfering , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism
13.
J Immunol ; 176(11): 7039-50, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16709866

ABSTRACT

Clathrin-mediated endocytosis (CME) is a common pathway used by G protein-linked receptors to transduce extracellular signals. We hypothesize that platelet-activating factor (PAF) receptor (PAFR) ligation requires CME and causes engagement of beta-arrestin-1 and recruitment of a p38 MAPK signalosome that elicits distinct actin rearrangement at the receptor before endosomal scission. Polymorphonuclear neutrophils were stimulated with buffer or 2 microM PAF (1 min), and whole cell lysates or subcellular fractions were immunoprecipitated or slides prepared for colocalization and fluorescent resonance energy transfer analysis. In select experiments, beta-arrestin-1 or dynamin-2 were neutralized by intracellular introduction of specific Abs. PAFR ligation caused 1) coprecipitation of the PAFR and clathrin with beta-arrestin-1, 2) fluorescent resonance energy transfer-positive interactions among the PAFR, beta-arrestin-1, and clathrin, 3) recruitment and activation of the apoptosis signal-regulating kinase-1/MAPK kinase-3/p38 MAPK (ASK1/MKK3/p38 MAPK) signalosome, 4) cell polarization, and 5) distinct actin bundle formation at the PAFR. Neutralization of beta-arrestin-1 inhibited all of these cellular events, including PAFR internalization; conversely, dynamin-2 inhibition only affected receptor internalization. Selective p38 MAPK inhibition globally abrogated actin rearrangement; however, inhibition of MAPK-activated protein kinase-2 and its downstream kinase leukocyte-specific protein-1 inhibited only actin bundle formation and PAFR internalization. In addition, ASK1/MKK3/p38 MAPK signalosome assembly appears to occur in a novel manner such that the ASK1/p38 MAPK heterodimer is recruited to a beta-arrestin-1 bound MKK3. In polymorphonuclear neutrophils, leukocyte-specific protein-1 may play a role similar to fascin for actin bundle formation. We conclude that PAF signaling requires CME, beta-arrestin-1 recruitment of a p38 MAPK signalosome, and specific actin bundle formation at the PAFR for transduction before endosomal scission.


Subject(s)
Actins/metabolism , Arrestins/metabolism , Cell Membrane/enzymology , Clathrin/physiology , Endocytosis/physiology , MAP Kinase Signaling System/physiology , Platelet Activating Factor/physiology , p38 Mitogen-Activated Protein Kinases/physiology , Arrestins/physiology , Calcium/metabolism , Cell Membrane/metabolism , Clathrin/metabolism , Dynamin II/physiology , Endosomes/enzymology , Endosomes/metabolism , Enzyme Activation/physiology , Humans , MAP Kinase Kinase Kinase 5/isolation & purification , MAP Kinase Kinase Kinase 5/metabolism , Microfilament Proteins/metabolism , Microfilament Proteins/physiology , Neutrophils/cytology , Neutrophils/enzymology , Neutrophils/metabolism , Platelet Activating Factor/metabolism , Platelet Membrane Glycoproteins/metabolism , Platelet Membrane Glycoproteins/physiology , Protein Transport/physiology , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/physiology , Subcellular Fractions/enzymology , beta-Arrestin 1 , beta-Arrestins , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Am J Physiol Cell Physiol ; 290(3): C917-24, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16267105

ABSTRACT

High mobility group box 1 (HMGB1), originally described as a DNA-binding protein, can also be released extracellularly and functions as a late mediator of inflammatory responses. Although recent reports have indicated that the receptor for advanced glycation end products (RAGE) as well as Toll-like receptor (TLR)2 and TLR4 are involved in cellular activation by HMGB1, there has been little evidence of direct association between HMGB1 and these receptors. To examine this issue, we used fluorescence resonance energy transfer (FRET) and immunoprecipitation to directly investigate cell surface interactions of HMGB1 with TLR2, TLR4, and RAGE. FRET images in RAW264.7 macrophages demonstrated association of HMGB1 with TLR2 and TLR4 but not RAGE. Transient transfections into human embryonic kidney-293 cells showed that HMGB1 induced cellular activation and NF-kappaB-dependent transcription through TLR2 or TLR4 but not RAGE. Coimmunoprecipitation also found interaction between HMGB1 and TLR2 as well as TLR4, but not with RAGE. These studies provide the first direct evidence that HMGB1 can interact with both TLR2 and TLR4 and also supply an explanation for the ability of HMGB1 to induce cellular activation and generate inflammatory responses that are similar to those initiated by LPS.


Subject(s)
HMGB1 Protein/metabolism , Receptors, Immunologic/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Animals , Cell Line , Fluorescence Resonance Energy Transfer , Gene Expression Regulation , HMGB1 Protein/genetics , Humans , Macrophages/cytology , Macrophages/metabolism , Protein Binding , Receptor for Advanced Glycation End Products , Receptors, Immunologic/blood , Signal Transduction , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics
15.
J Am Coll Surg ; 201(3): 418-25, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16125076

ABSTRACT

BACKGROUND: Activated macrophages defend against tumors by secreting cytokines to recruit secondary immune cells, presenting antigen to T cells, and by direct tumor cytotoxicity. Peritoneal macrophages harvested from melanoma-bearing mice are less cytotoxic to melanoma cells, and produce less superoxide, nitric oxide, and tumor necrosis factor-alpha (TNF-alpha) than those from nontumor-bearing mice. Similar impairment of macrophage activation occurs in vitro using media harvested from cultured melanoma cells. Stimulation of Toll-like receptor 4 (TLR-4) activates macrophages and results in the release of TNF-alpha. We hypothesized that melanoma inhibits macrophage activation by suppressing TLR-4 signaling. STUDY DESIGN: Melanoma conditioned media (MCM) was generated from B16 melanoma cells. Peritoneal macrophages from TLR-4 competent or TLR-4 incompetent mice were exposed to control or MCM for 24 hours; then stimulated with lipopolysaccharide. TNF-alpha secretion, TNF-alpha mRNA production, nuclear factor-kappaB (NF-kappaB) activation, and TLR-4 surface expression were measured. RESULTS: Peritoneal macrophages exposed to MCM produced considerably less TNF-alpha in response to stimulus than controls (691 pg/mL versus 2,066 pg/mL, p < 0.001). TNF-alpha production by TLR-4 incompetent macrophages was not affected by MCM (454 pg/mL versus 480 pg/mL). Stimulated TNF-alpha mRNA and activated NF-kappaB were decreased in MCM treated C57BL/6 macrophages (by 38% and 33%, respectively). TLR-4 surface expression, however, was not decreased by exposure to MCM. CONCLUSIONS: Melanoma inhibits macrophage activation by suppressing TLR-4 signaling downstream of the TLR-4 receptor.


Subject(s)
Macrophage Activation , Melanoma, Experimental/immunology , Membrane Glycoproteins/immunology , Receptors, Cell Surface/immunology , Signal Transduction , Animals , Culture Media , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , NF-kappa B/pharmacology , RNA, Messenger/metabolism , Signal Transduction/physiology , Toll-Like Receptor 4 , Toll-Like Receptors , Tumor Necrosis Factor-alpha/biosynthesis
16.
Nutrition ; 21(2): 214-23, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15723751

ABSTRACT

OBJECTIVE: Septic shock leads to derangement of cellular metabolism. Enhanced heat shock protein 70 (HSP-70) can preserve cellular metabolism after other forms of cellular stress. Glutamine (GLN) can enhance lung HSP-70 expression after lethal endotoxemia. However, it is unknown whether GLN can enhance HSP-70 expression and attenuate lung metabolic dysfunction after sublethal endotoxemia. Our aim was to determine whether GLN could upregulate HSP-70 and attenuate metabolic dysfunction in lung tissue after sublethal endotoxemia. METHODS: Sprague-Dawley rats were assigned to one of five groups. The first two groups were treated with Escherichia coli lipopolysaccharide (LPS; 1 mg/kg intravenously). GLN (0.75 g/kg intravenously) or balanced salt solution as a control was administered 5 min after LPS administration. The next two groups of rats were treated with quercetin (HSP-70 inhibitor; 400 mg/kg intraperitoneally) 6 h before LPS administration. The final group received no treatment. Lung tissue was harvested 24-h after LPS and analyzed with immunofluorescence and western blot for HSP-70. Tissue metabolites were quantified by 1H and 31P nuclear magnetic resonance spectroscopy. RESULTS: GLN compared with balanced salt solution (BSS) administration in LPS-treated animals led to significant increases in lung HSP-70. Increased HSP-70 expression was observed in lung epithelial cells and macrophages. GLN significantly improved the ratio of adenosine triphosphate to adenosine diphosphate in the lung after LPS. Quercetin inhibited a GLN-mediated increase in lung HSP-70 and blocked a beneficial effect of GLN on the ratio of adenosine triphosphate to adenosine diphosphate after LPS. CONCLUSIONS: A single dose of GLN can enhance HSP-70 in pulmonary epithelial cells and macrophages after sublethal endotoxemia. Further, GLN can attenuate endotoxin-induced lung metabolic dysfunction. GLN's beneficial effect on lung tissue after metabolic dysfunction caused by sublethal endotoxemia may be mediated in part by enhanced HSP-70.


Subject(s)
Glutamine/pharmacology , HSP70 Heat-Shock Proteins/metabolism , Lung/metabolism , Animals , Blotting, Western , Fluorescent Antibody Technique , Gene Expression Regulation/drug effects , HSP70 Heat-Shock Proteins/antagonists & inhibitors , HSP70 Heat-Shock Proteins/drug effects , Lipopolysaccharides/toxicity , Lung/cytology , Lung/drug effects , Lung/immunology , Magnetic Resonance Spectroscopy , Male , Quercetin/pharmacology , Random Allocation , Rats , Rats, Sprague-Dawley
17.
Biochem J ; 381(Pt 2): 503-10, 2004 Jul 15.
Article in English | MEDLINE | ID: mdl-15046617

ABSTRACT

IL-1F7b, a novel homologue of the IL-1 (interleukin 1) family, was discovered by computational cloning. We demonstrated that IL-1F7b shares critical amino acid residues with IL-18 and binds to the IL-18-binding protein enhancing its ability to inhibit IL-18-induced interferon-gamma. We also showed that low levels of IL-1F7b are constitutively present intracellularly in human blood monocytes. In this study, we demonstrate that similar to IL-18, both mRNA and intracellular protein expression of IL-1F7b are up-regulated by LPS (lipopolysaccharide) in human monocytes. In stable transfectants of murine RAW264.7 macrophage cells, there was no IL-1F7b protein expression despite a highly active CMV promoter. We found that IL-1F7b-specific mRNA was rapidly degraded in transfected cells, via a 3'-UTR (untranslated region)-independent control of IL-1F7b transcript stability. After LPS stimulation, there was a rapid transient increase in IL-1F7b-specific mRNA and concomitant protein levels. Using sequence alignment, we found a conserved ten-nucleotide homology box within the open reading frame of IL-F7b, which is flanking the coding region instability elements of some selective genes. In-frame deletion of downstream exon 5 from the full-length IL-1F7b cDNA markedly increased the levels of IL-1F7b mRNA. A similar coding region element is located in IL-18. When transfected into RAW264.7 macrophages, IL-18 mRNA was also unstable unless treated with LPS. These results indicate that both IL-1F7b and IL-18 mRNA contain functional instability determinants within their coding region, which influence mRNA decay as a novel mechanism to regulate the expression of IL-1 family members.


Subject(s)
Interleukin-18/genetics , Interleukin-1/genetics , Lipopolysaccharides/immunology , RNA Stability/physiology , RNA, Messenger/physiology , Animals , Base Sequence/genetics , CHO Cells/chemistry , CHO Cells/metabolism , COS Cells/chemistry , COS Cells/metabolism , Cell Line , Chlorocebus aethiops , Conserved Sequence/genetics , Cricetinae , Exons/genetics , Exons/physiology , Gene Expression Regulation/physiology , Humans , Interleukin-1/immunology , Interleukin-1/metabolism , Interleukin-1/physiology , Interleukin-18/immunology , Interleukin-18/metabolism , Macrophages/chemistry , Macrophages/metabolism , Mice , Molecular Sequence Data , Monocytes/chemistry , Monocytes/metabolism , Monocytes/physiology , Sequence Deletion/genetics , Sequence Deletion/physiology , Sequence Homology, Nucleic Acid , Transfection/methods , Up-Regulation/physiology
18.
Proc Natl Acad Sci U S A ; 99(21): 13723-8, 2002 Oct 15.
Article in English | MEDLINE | ID: mdl-12381835

ABSTRACT

IL-1F7 was discovered in expressed sequence tag databases as a member of the increasing family of proteins sharing sequence homology to IL-1alpha/beta, IL-1Ra, and IL-18. In the present study using immunohistochemical staining, IL-1F7 was localized in human peripheral monocytic cells, suggesting its role in immune regulation. Recombinant human IL-1F7b was shown to bind to the IL-18Ralpha but without IL-18 agonistic or antagonistic function. Using chemical cross-linking, we observed that, unlike IL-18, IL-1F7b fails to recruit the IL-18Rbeta chain to form a functionally active, ternary complex with the IL-18Ralpha chain. IL-1F7b shares two conserved amino acids with IL-18 (Glu-35 and Lys-124), which participate in the interaction of IL-18 with the IL-18Ralpha chain as well as the IL-18-binding protein (IL-18BP), a secreted protein that neutralizes IL-18 activity. In testing whether IL-1F7b interacts with IL-18BP, we unexpectedly observed that IL-1F7b enhanced the ability of IL-18BP to inhibit IL-18-induced IFNgamma by 25-30% in a human natural killer cell line. This effect was observed primarily at limiting concentrations of IL-18BP (3.12-12.5 ng/ml) and at a 50- to 100-fold molar excess of IL-1F7b. Similar results were obtained by using isolated human peripheral blood mononuclear cells. To study the molecular basis of this effect we performed binding studies of IL-1F7b and IL-18BP. After cross-linking, a high molecular weight complex consisting of IL-1F7b and IL-18BP was observed on SDS/PAGE. We propose that after binding to IL-18BP, IL-1F7b forms a complex with IL-18Rbeta, depriving the beta-chain of forming a functional receptor complex with IL-18Ralpha and thus inhibiting IL-18 activity.


Subject(s)
Glycoproteins/chemistry , Glycoproteins/metabolism , Interleukin-18/antagonists & inhibitors , Interleukin-1/chemistry , Interleukin-1/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Gene Expression , Humans , In Vitro Techniques , Intercellular Signaling Peptides and Proteins , Interferon-gamma/biosynthesis , Interleukin-1/genetics , Interleukin-18/genetics , Interleukin-18 Receptor alpha Subunit , Killer Cells, Natural/immunology , Macromolecular Substances , Mice , Molecular Sequence Data , Monocytes/immunology , Protein Structure, Tertiary , Receptors, Interleukin/chemistry , Receptors, Interleukin/metabolism , Receptors, Interleukin-18 , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Transfection
19.
Am J Physiol Heart Circ Physiol ; 283(4): H1455-61, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12234797

ABSTRACT

Although monocyte chemotactic protein-1 (MCP-1) is best known for its ability to recruit mononuclear cells, few studies have examined the effects of this chemokine on other events in the vascular response to injury. The purpose of the present study was to determine the influence of MCP-1 on human vascular smooth muscle (VSMC) proliferation. MCP-1 induced concentration-dependent VSMC proliferation as measured by bromodeoxyuridine (BrdU) uptake. Direct cell counting demonstrated a twofold increase in VSMC after stimulation with MCP-1. This mitogenic effect was similar to that observed with the prototypical atherogenic cytokine platelet-derived growth factor. Immunohistochemistry and Western blot analysis revealed that MCP-1 increased both proliferating nuclear cell antigen and cyclin A expression. Whereas MCP-1 did not promote nuclear factor-kappaB activation, MCP-1-induced VSMC proliferation appeared to be dependent on phosphotidylinositol 3-kinase activation. In conclusion, MCP-1 directly induces VSMC growth, which is associated with activation of cell cycle proteins and intracellular proliferative signals. Within the inflammatory paradigm of vascular remodeling, these data suggest that MCP-1 is more than simply a chemokine but also a potent mitogen for VSMC proliferation.


Subject(s)
Chemokine CCL2/pharmacology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Aorta, Thoracic/cytology , Cell Division/drug effects , Cells, Cultured , Chemokine CCL2/metabolism , Cyclin A/metabolism , Humans , Mitogens/pharmacology , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Platelet-Derived Growth Factor/pharmacology , Proliferating Cell Nuclear Antigen/metabolism , Signal Transduction/drug effects
20.
J Leukoc Biol ; 72(2): 401-9, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12149432

ABSTRACT

Patients with sepsis and acute lung injury have increased interleukin (IL)-18 levels systemically. We hypothesize that IL-18 stimulates neutrophils (PMNs) at physiologic concentrations. IL-18 primed the oxidase at 15 min (10-100 ng/ml), 30 min (0.1-100 ng/ml), and 60 min (100 ng/ml; P<0.05) and caused translocation of p47(phox) to the membrane similar to lipopolysaccharides. CD11b surface expression was increased by IL-18 in a time- and concentration-dependent manner. IL-18 caused up-regulation of the formyl-Met-Leu-Phe receptor, changes in PMN size, and elastase release. Investigation of signaling demonstrated IL-18-mediated activation of p38 mitogen-activated protein (MAP) kinase in a concentration (0.1-100 ng/ml)-, time (5-15 min)-, and Ca2+-dependent manner. IL-18 directly increased cytosolic Ca2+ concentration. IL-18 activation of PMNs was blocked by inhibition of p38 MAP kinase activity (SB203580) or by inhibition of p38 MAP kinase activation by chelation of cytosolic Ca2+. We conclude that IL-18, at physiologic concentrations, is an effective PMN priming agent that requires p38 MAP kinase activity.


Subject(s)
Egtazic Acid/analogs & derivatives , Interleukin-18/physiology , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/physiology , Neutrophils/drug effects , Adult , Calcium , Calcium Signaling/drug effects , Cell Size , Chelating Agents/pharmacology , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Imidazoles/pharmacology , Interleukin-18/pharmacology , Lipopolysaccharides/pharmacology , MAP Kinase Signaling System/drug effects , Macrophage-1 Antigen/biosynthesis , Macrophage-1 Antigen/genetics , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , NADPH Oxidases/metabolism , Neutrophils/enzymology , Neutrophils/ultrastructure , Phosphoproteins/metabolism , Protein Transport/drug effects , Pyridines/pharmacology , Receptors, Formyl Peptide , Receptors, Immunologic/biosynthesis , Receptors, Immunologic/genetics , Receptors, Peptide/biosynthesis , Receptors, Peptide/genetics , Respiratory Burst/drug effects , Up-Regulation/drug effects , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...