Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Language
Publication year range
1.
Crit Care Med ; 40(2): 379-87, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22020238

ABSTRACT

OBJECTIVES: Matrix metalloproteinase-8 messenger RNA expression was previously found to be increased in whole blood of children with septic shock. The impact of this finding on the severity and inflammatory response to sepsis is unknown. Here, we investigate the relationship between matrix metalloproteinase-8 and disease severity in children with septic shock. We further corroborate the role of matrix metalloproteinase-8 in sepsis in a murine model. DESIGN: Retrospective observational clinical study and randomized controlled laboratory experiments. SETTING: Pediatric intensive care units and an animal research facility at an academic children's hospital. PATIENTS AND SUBJECTS: Patients age ≤10 yrs admitted to the intensive care unit with a diagnosis of septic shock. For laboratory studies, we utilized male mice deficient for matrix metalloproteinase-8 and male wild-type C57BL/6J mice. INTERVENTIONS: Blood from children with septic shock was analyzed for matrix metalloproteinase-8 messenger RNA expression and matrix metalloproteinase-8 activity, and correlated with disease severity based on mortality and degree of organ failure. A murine model of sepsis was used to explore the effect of genetic and pharmacologic inhibition of matrix metalloproteinase-8 on the inflammatory response to sepsis. Finally, activation of nuclear factor-κB was assessed both in vitro and in vivo. MEASUREMENTS AND MAIN RESULTS: Increased matrix metalloproteinase-8 mRNA expression and activity in septic shock correlates with decreased survival and increased organ failure in pediatric patients. Genetic and pharmacologic inhibition of matrix metalloproteinase-8 leads to improved survival and a blunted inflammatory profile in a murine model of sepsis. We also identify matrix metalloproteinase-8 as a direct in vitro activator of the proinflammatory transcription factor, nuclear factor-κB. CONCLUSIONS: Matrix metalloproteinase-8 is a novel modulator of inflammation during sepsis and a potential therapeutic target.


Subject(s)
Inflammation Mediators/blood , Matrix Metalloproteinase 8/blood , Multiple Organ Failure/blood , Shock, Septic/blood , Shock, Septic/mortality , Animals , Cells, Cultured , Child , Child, Preschool , Cohort Studies , Disease Models, Animal , Disease Progression , Female , Humans , Inflammation Mediators/metabolism , Intensive Care Units, Pediatric , Leukocytes, Mononuclear/metabolism , Macrophages, Peritoneal/metabolism , Male , Matrix Metalloproteinase 8/metabolism , Mice , Mice, Inbred C57BL , Multiple Organ Failure/mortality , Prognosis , Retrospective Studies , Sepsis/blood , Sepsis/drug therapy , Sepsis/physiopathology , Severity of Illness Index , Shock, Septic/physiopathology , Survival Analysis , Treatment Outcome
2.
Immunol Lett ; 135(1-2): 173-9, 2011 Mar 30.
Article in English | MEDLINE | ID: mdl-21094186

ABSTRACT

Heat shock protein 72 (Hsp72), a canonical intracellular molecular chaperone, may also function as an extracellular danger signal for the innate immune system. To further delineate the biological role of Hsp72 in the innate immune system, we generated two truncated versions of the full length human Hsp72 (N-terminus Hsp72, amino acids 1-430; and C-terminus Hsp72 amino acids 420-641) and directly compared their ability to activate cells from the macrophage/monocyte lineage. In RAW 264.7 macrophages transfected with a NF-κB-dependent luciferase reporter plasmid, C-terminus Hsp72 was a more potent inducer of NF-κB activity than N-terminus Hsp72, and this effect did not seem to be secondary to endotoxin contamination. C-terminus Hsp72-mediated activation of the NF-κB pathway was corroborated by increased activation of IκB kinase, degradation of IκBα, and increased NF-κB-DNA binding. C-terminus Hsp72 was a more potent inducer of tumor necrosis factor-α (TNFα) expression in RAW 264.7 macrophages and in primary murine peritoneal macrophages from wild-type mice. C-terminus Hsp72 did not induce TNFα expression in primary murine peritoneal macrophages from Toll-like receptor (TLR4) mutant mice, indicating a role for TLR4. In human THP-1 mononuclear cells, C-terminus Hsp72 induced tolerance to subsequent LPS stimulation, whereas N-terminus Hsp72 did not induce tolerance. Finally, control experiments using equimolar amounts of N-terminus or C-terminus Hsp72 demonstrated a higher biological potency for C-terminus Hsp72. These data demonstrate that the ability of human Hsp72 to serve as an activator for cells of the macrophage/monocyte lineage primarily lies in the C-terminus region spanning amino acids 420-641.


Subject(s)
HSP72 Heat-Shock Proteins/immunology , Immunity, Innate/immunology , Macrophage Activation/immunology , Macrophages, Peritoneal/immunology , Monocytes/immunology , Animals , Cell Line , HSP72 Heat-Shock Proteins/genetics , HSP72 Heat-Shock Proteins/pharmacology , Humans , Immunity, Innate/drug effects , Macrophage Activation/drug effects , Macrophages, Peritoneal/metabolism , Mice , Mice, Mutant Strains , Monocytes/metabolism , Protein Structure, Tertiary , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism
3.
Shock ; 30(3): 285-92, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18323737

ABSTRACT

Intracellular heat shock protein 72 (Hsp72) is known to serve a broad cytoprotective role. Recent data indicate that stressed cells can release Hsp72 into the extracellular compartment, although the biological function of extracellular Hsp72 remains to be fully elucidated. Because extracellular Hsp72 has been demonstrated to interact with Toll-like receptor 4, we hypothesized that endogenously produced and released Hsp72 would reprogram the mononuclear cell responses to LPS. THP-1 cells treated with LPS were used as a model for nuclear factor (NF)-kappaB activation. Heat shock conditions consisted of incubation at 43 degrees C for 1 h. Control cells were incubated at 37 degrees C. Twenty four hours after incubation, heat shock conditioned media (HSCM) and control media (CM) were centrifuged, and the respective cells were discarded. A separate group of naive THP-1 cells were then incubated with either HSCM or CM for 18 h and then stimulated with LPS (1 mug/mL). Heat shock significantly increased Hsp72 in HSCM compared with CM. In THP-1 cells transfected with an NF-kappaB luciferase reporter plasmid, the addition of HSCM attenuated subsequent LPS-mediated luciferase activity compared with cells incubated in CM. The addition of HSCM also attenuated LPS-mediated NF-kappaB-DNA binding and IkappaBalpha degradation. Heat shock protein 72-mediated inhibition of NF-kappaB activation was further corroborated by a significant decrease in TNF-alpha production. When HSCM and CM were subjected to Hsp72 depletion via adenosine triphosphate-agarose binding, LPS-mediated activation of NF-kappaB was partially restored, suggesting that Hsp72 is partially responsible for cellular reprogramming in response to HSCM. These data demonstrate that endogenously produced and released extracellular Hsp72 has the ability to reprogram the in vitro response to endotoxin in cultured human mononuclear cells.


Subject(s)
HSP72 Heat-Shock Proteins/metabolism , Leukocytes, Mononuclear/metabolism , Cell Line, Tumor , Endotoxins/metabolism , Humans , Hypoxia/metabolism , Immune System , Inflammation , Ischemic Preconditioning , Lipopolysaccharides/metabolism , Models, Biological , Monocytes/metabolism , NF-kappa B/metabolism , Sepharose/chemistry , Signal Transduction
4.
J Neurochem ; 94(2): 360-6, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15998287

ABSTRACT

Induction of heat shock protein 70 (Hsp70) via sublethal stress protects neurons from subsequent lethal injuries. Here we show that specific and efficient intracellular transduction of Hsp70 can be achieved utilizing an 11 amino acid leading sequence from human immunodeficiency virus (TAT-Hsp70) in primary neuronal cultures. Western blot and immunohistochemistry demonstrated intracellular accumulation of Hsp70 in insoluble protein fractions and mitochondrial compartments. We then examined the effects of Hsp70 overexpression using TAT-Hsp70 in models of nitrosative and excitotoxic neuronal death in vitro. Neurons were pre-incubated with 300 nM TAT-Hsp 70 overnight, then exposed to either peroxynitrite (ONOO-) or glutamate. TAT-Hsp70 maintained cellular respiration, inhibited extracellular lactate dehydrogenase release, and/or reduced cell death assessed by flow cytometry vs. vehicle, wild-type Hsp70, and TAT-beta-galactosidase controls. Hsp70 transduction using a TAT fusion protein is an effective method to selectively increase Hsp70 in neurons and is sufficient to provide neuroprotection from nitrosative stress and excitotoxicity. Further study is needed to confirm whether TAT-Hsp70 is protective in in vivo models of brain injury.


Subject(s)
Gene Products, tat/physiology , Glutamic Acid/toxicity , HSP70 Heat-Shock Proteins/physiology , Neurons/drug effects , Peroxynitrous Acid/toxicity , Transduction, Genetic/methods , Animals , Animals, Newborn , Blotting, Western/methods , Cell Death/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cloning, Molecular/methods , Dose-Response Relationship, Drug , Flow Cytometry/methods , HSP70 Heat-Shock Proteins/metabolism , Humans , Immunohistochemistry/methods , Lactate Dehydrogenases/metabolism , Neurons/physiology , Rats , Rats, Sprague-Dawley , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Tetrazolium Salts , Thiazoles
5.
Am J Physiol Cell Physiol ; 289(5): C1152-8, 2005 Nov.
Article in English | MEDLINE | ID: mdl-15958526

ABSTRACT

Heat shock modulates cellular proinflammatory responses, and we have been interested in elucidating the mechanisms that govern this modulation. The dual specific phosphatase, MAP kinase phosphatase-1 (MKP-1), is an important modulator of cellular inflammatory responses, and we recently reported that heat shock increases expression of MKP-1. Herein we sought to elucidate the mechanisms by which heat shock modulates MKP-1 gene expression. Subjecting RAW264.7 macrophages to heat shock increased MKP-1 gene expression in a time-dependent manner. Transfection with a wild-type murine MKP-1 promoter luciferase reporter plasmid demonstrated that heat shock activates the MKP-1 promoter. When the reporter plasmid was transfected into heat shock factor-1 (HSF-1)-null fibroblasts, the MKP-1 promoter was activated in response to heat shock in a manner similar to that of wild-type fibroblasts with intact HSF-1. Site-directed mutagenesis of two potential heat shock elements in the MKP-1 promoter demonstrated that both sites are required for basal promoter activity. mRNA stability assays demonstrated that heat shock increased MKP-1 mRNA stability compared with cells maintained at 37 degrees C. Inhibition of p38 MAP kinase activity inhibited heat shock-mediated expression of MKP-1. These data demonstrate that heat shock regulates MKP-1 gene expression at both the transcriptional and posttranscriptional levels. Transcriptional mechanisms are HSF-1 independent but are dependent on putative heat shock elements in the MKP-1 promoter. Posttranscriptional mechanisms involve increased stability of MKP-1 mRNA that is partially dependent on p38 MAP kinase activity. These data demonstrate another potential mechanism by which heat shock can modulate inflammation-related signal transduction.


Subject(s)
Cell Cycle Proteins/biosynthesis , Immediate-Early Proteins/biosynthesis , Macrophages/physiology , Phosphoprotein Phosphatases/biosynthesis , Protein Tyrosine Phosphatases/biosynthesis , Animals , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Dactinomycin/pharmacology , Dual Specificity Phosphatase 1 , Heat Shock Transcription Factors , Heat-Shock Proteins/metabolism , Hot Temperature , Macrophages/drug effects , Mice , Mice, Knockout , Promoter Regions, Genetic/physiology , Protein Phosphatase 1 , RNA, Messenger/biosynthesis , Time Factors , Transcription Factors/genetics , Transcription Factors/physiology , Up-Regulation , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Am J Respir Cell Mol Biol ; 32(3): 225-31, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15576670

ABSTRACT

German cockroach extract synergistically regulates tumor necrosis factor-alpha (TNF-alpha)-induced interleukin (IL)-8 expression in human airway epithelial cells. The IL-8 promoter contains nuclear factor (NF)-kappaB, activating protein (AP)-1, and NF for IL-6 (NF-IL6) transcription factor binding regions. Because cockroach extract activates extracellular regulated kinase (ERK), a known activator of AP-1 and NF-IL6, we focused on the regulation of these transcription factors. Although TNF-alpha and cockroach extract both increased AP-1 translocation, mutation of the AP-1 site in the context of the wild-type promoter had no effect on cockroach extract-induced synergy. Mutation of the NF-IL6 site in the context of the wild-type IL-8 promoter, or overexpression of a dominant-negative NF-IL6 mutant, each abolished cockroach extract-induced synergy. Cockroach extract induced NF-IL6 translocation and DNA binding, an effect that was further increased in the presence of TNF-alpha. Cockroach extract-induced regulation of NF-IL6 was due to active serine proteases in the extract as well as activation of protease activated receptor (PAR)-2, but not PAR-1. Chemical inhibition of ERK also attenuated cockroach extract-induced NF-IL6-DNA binding. We conclude that proteases in German cockroach extract regulate PAR-2 and ERK to increase NF-IL6 activity and synergistically regulate TNF-alpha-induced IL-8 promoter activity in human airway epithelium.


Subject(s)
Blattellidae/enzymology , CCAAT-Enhancer-Binding Protein-beta/metabolism , Gene Expression Regulation/physiology , Interleukin-8/genetics , Peptide Hydrolases/metabolism , Animals , Blattellidae/genetics , Blattellidae/metabolism , Bronchi/metabolism , Epithelium/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Interleukin-8/metabolism , NF-kappa B/metabolism , Promoter Regions, Genetic , Protein Transport/physiology , Transcription Factor AP-1/metabolism , Tumor Necrosis Factor-alpha/metabolism
7.
Biochem Biophys Res Commun ; 301(1): 54-9, 2003 Jan 31.
Article in English | MEDLINE | ID: mdl-12535640

ABSTRACT

Heat shock protein 70 (HSP70) is an intracellular stress protein that confers cytoprotection to a variety of cellular stressors. Several lines of evidence have suggested that augmentation of the heat shock response by increasing the expression of HSP70 represents a potential therapeutic strategy for the treatment of critically ill patients. The Tat protein of human immunodeficiency virus 1 (HIV-1) has been used previously to deliver functional cargo proteins intracellularly when added exogenously to cultured cells. We generated a Tat-HSP70 fusion protein using recombinant methods and treated HSF -/- cells with either Tat-HSP70 or recombinant HSP70 prior to exposure to hyperoxia or lethal heat shock. We showed that biologically active, exogenous HSP70 can be delivered into cells using the HIV-1 Tat protein, and that the Tat-mediated delivery of HSP70 confers cytoprotection against thermal stress and hyperoxia and may represent a novel approach to augmenting intracellular HSP70 levels.


Subject(s)
Gene Products, tat/metabolism , HSP70 Heat-Shock Proteins/metabolism , Protein Transport/physiology , Transduction, Genetic , Animals , Cell Line , Cell Survival , DNA-Binding Proteins/genetics , Gene Products, tat/chemistry , Gene Products, tat/genetics , HSP70 Heat-Shock Proteins/genetics , Heat Shock Transcription Factors , Hot Temperature , Humans , Mice , Mice, Knockout , Oxygen/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transcription Factors
8.
Exp Lung Res ; 28(8): 609-22, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12490036

ABSTRACT

High concentrations of oxygen (hyperoxia) are known to cause cellular injury and death. The heat shock response is a highly conserved cellular defense mechanism that protects cells against various environmental stressors, including hyperoxia. Herein we determined the role of heat shock factor-1 (HSF-1), a major component of the heat shock response, in protecting cells against hyperoxia. Embryonic fibroblasts from HSF-1-null mutant mice (HSF-1 -/- cells) were compared to wild-type embryonic fibroblasts (HSF-1 +/+ cells) following 24 hours' exposure to room air or hyperoxia (95% O(2)). Acute survival in hyperoxia was decreased in HSF-1 -/- cells as compared to HSF-1 +/+ cells. Intracellular ATP levels were significantly lower in the HSF-1 -/- cells as compared to the HSF-1 +/+ cells exposed to hyperoxia. Isoprostane levels, a marker of membrane lipid peroxidation, were significantly higher in the HSF-1 -/- cells as compared to the HSF-1 +/+ cells exposed to hyperoxia. Restoration of HSF-1 in the HSF-1 -/- cells by stable transfection with a HSF-1 expression plasmid improved survival in hyperoxia when compared to HSF-1 -/- cells stably transfected with the empty expression vector. Hyperoxia increased activation of HSF-1 in HSF-1 +/+ cells and in HSF-1 -/- cells stably transfected with the HSF-1 expression plasmid. These data demonstrate that HSF-1 plays an important role in conferring resistance to hyperoxia in vitro.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Hyperoxia/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Death/physiology , Cell Survival/physiology , Cells, Cultured , Fibroblasts/cytology , HSP70 Heat-Shock Proteins/metabolism , Heat Shock Transcription Factors , Hyperoxia/pathology , Isoprostanes/biosynthesis , Mice , Mice, Mutant Strains , Oxidative Stress/physiology , Oxygen/pharmacology , Transcription Factors , Transfection
9.
Inflammation ; 26(3): 129-37, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12083419

ABSTRACT

Previous studies demonstrated important interactions between the heat shock response and the IkappaBalpha/NF-kappaB pathway when these two pathways are induced sequentially. One such interaction involves the ability of heat shock to inhibit subsequent degradation of IkappaBalpha in response to a proinflammatory signal. Herein we investigated the temporal relationship between recovery from heat shock and inhibition of IkappaBalpha degradation, and the proximal mechanisms by which heat shock inhibits degradation of IkappaBalpha in macrophages. In RAW 264.7 murine macrophages, prior heat shock inhibited LPS-mediated IkappaBalpha degradation up to 4 h after recovery from heat shock, and this effect correlated with inhibition of LPS-mediated activation of NF-kappaB. Beyond these recovery periods, heat shock did not inhibit IkappaBalpha degradation. IkappaB kinase (IKK) assays demonstrated that heat shock inhibited LPS-mediated activation of IKK up to 1 h after recovery from heat shock. Heat shock also increased intracellular phosphatase activity, and inhibition of intracellular phosphatase activity partially reversed the ability of heat shock to inhibit both LPS-mediated degradation of IkappaBalpha and LPS-mediated activation of IKK. These data demonstrate that the ability of heat shock to inhibit degradation of IkappaBalpha is dependent on the recovery period between the heat shock stimulus and the proinflammatory stimulus. The mechanism by which heat shock inhibits degradation of IkappaBalpha involves dual modulation of IKK and intracellular phosphatase activity.


Subject(s)
Heat-Shock Response , I-kappa B Proteins/metabolism , Lipopolysaccharides/pharmacology , Macrophages/metabolism , NF-kappa B/metabolism , Animals , Cell Line , Immunologic Factors/pharmacology , Mice , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL