Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Appl Physiol (1985) ; 91(5): 2213-23, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11641364

ABSTRACT

Pharmacokinetics and immunogenicity of six different recombinant human soluble p55 tumor necrosis factor (TNF) receptor I (sTNFR-I) constructs were evaluated in juvenile baboons. The constructs included either an sTNFR-I IgG1 immunoadhesin (p55 sTNFR-I Fc) or five different sTNFR-I constructs covalently linked to polyethylene glycol. The constructs were administered intravenously three times, and pharmacokinetics and immunogenicity were examined over 63 days. All of the constructs were immunogenic, with the exception of a 2.6-domain monomeric sTNFR-I. To evaluate whether the nonimmunogenic 2.6-domain monomeric construct could protect baboons against TNF-alpha-induced mortality, baboons were pretreated with 1, 5, or 10 mg/kg body wt and were compared with baboons receiving either placebo or 1 mg/kg body wt of the dimeric 4.0-domain sTNFR-I construct (n = 3 each) before lethal Escherichia coli bacteremia. The monomeric construct protected baboons and neutralized TNF bioactivity, although greater quantities were required compared with the dimeric 4.0-domain sTNFR-I construct. We conclude that E. coli-recombinant-derived human sTNFR-I constructs can be generated with minimal immunogenicity on repeated administration and still protect against the consequences of exaggerated TNF-alpha production.


Subject(s)
Immunoglobulin G/metabolism , Papio/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Amino Acid Sequence , Animals , Blood Cell Count , Cloning, Molecular , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Escherichia coli/genetics , Etanercept , Half-Life , Hemodynamics/physiology , Humans , Immunoglobulin G/immunology , Kinetics , Molecular Sequence Data , Polyethylene Glycols , Receptors, Tumor Necrosis Factor/immunology , Tumor Necrosis Factor-alpha/pharmacokinetics , Tumor Necrosis Factor-alpha/pharmacology
2.
Proc Natl Acad Sci U S A ; 98(20): 11503-8, 2001 Sep 25.
Article in English | MEDLINE | ID: mdl-11553765

ABSTRACT

Sepsis remains a significant clinical conundrum, and recent clinical trials with anticytokine therapies have produced disappointing results. Animal studies have suggested that increased lymphocyte apoptosis may contribute to sepsis-induced mortality. We report here that inhibition of thymocyte apoptosis by targeted adenovirus-induced thymic expression of human IL-10 reduced blood bacteremia and prevented mortality in sepsis. In contrast, systemic administration of an adenovirus expressing IL-10 was without any protective effect. Improvements in survival were associated with increases in Bcl-2 expression and reductions in caspase-3 activity and thymocyte apoptosis. These studies demonstrate that thymic apoptosis plays a critical role in the pathogenesis of sepsis and identifies a gene therapy approach for its therapeutic intervention.


Subject(s)
Apoptosis/drug effects , Interleukin-10/genetics , Sepsis/therapy , Animals , Disease Models, Animal , Female , Humans , Interleukin-10/therapeutic use , Mice , Mice, Inbred C57BL , Sepsis/immunology
3.
Gene Ther ; 7(16): 1425-30, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10981671

ABSTRACT

Cationic liposome and plasmid-mediated gene transfer has emerged as a novel technique for the targeted delivery of protein-based therapies in acute inflammatory diseases. However, concerns have arisen that cationic liposomes and plasmid DNA have inherent proinflammatory properties which could exacerbate pre-existing inflammatory processes. In healthy mice, intraperitoneal administration of cationic liposomes (200 nmol) complexed to plasmid DNA (100 microg) induced a proinflammatory response characterized by the induction of tumor necrosis factor alpha and interleukin-1beta mRNA expression. The plasma concentrations of the hepatic acute phase proteins interleukin-6, amyloid A, amyloid P, and seromucoid were also increased (P<0.05), and this response was seen in endotoxin-resistant (C3H/HeJ) mice. The inflammatory response associated with gene transfer increased the mortality and severity of experimentally induced sterile inflammation (pancreatitis). We conclude that systemic administration of cationic liposomes and plasmid DNA is associated with induction of the innate immune response which may exacerbate pre-existing inflammatory processes.


Subject(s)
DNA/adverse effects , Gene Transfer Techniques/adverse effects , Liposomes/adverse effects , Pancreatitis/immunology , Amyloid/blood , Animals , Cations , DNA/administration & dosage , Injections, Intraperitoneal , Interleukin-1/genetics , Interleukin-6/blood , Liposomes/administration & dosage , Mice , Mice, Inbred C3H , Orosomucoid/analysis , RNA, Messenger/analysis , Tumor Necrosis Factor-alpha/genetics
4.
Am J Physiol Regul Integr Comp Physiol ; 278(4): R824-30, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10749768

ABSTRACT

A complete understanding of the role for endogenously produced interleukin-1 (IL-1), tumor necrosis factor-alpha (TNF-alpha), and IL-1 receptor antagonist (IL-1ra) in the acute phase response to inflammation remains unknown. In the present studies, knockout mice lacking either a functional IL-1 type I receptor (IL-1RI(-/-)), a TNF type I receptor (TNFR-I(-/-)), or both IL-1 type I and TNF type I receptors (IL-1RI(-/-)/TNFR-I(-/-)) received a turpentine abscess. Additional mice deficient in IL-1ra protein (IL-1ra(-/-)) or overexpressing IL-1ra protein (IL-1ra(tg)) were similarly treated. After a turpentine abscess, IL-1 receptor knockout mice exhibited an attenuated inflammatory response compared with wild-type or animals lacking a functional TNFR-I. Mice overexpressing IL-1ra also had an attenuated hepatic acute phase protein response, whereas IL-1ra knockout mice had a significantly greater hepatic acute phase response. We conclude that the inflammatory response to a turpentine abscess is the result of a balance between IL-1ra expression and IL-1 binding to its type I receptor. Endogenously produced IL-1ra plays a central role in mitigating the magnitude of the IL-1-mediated inflammatory response and, ultimately, the outcome to a turpentine abscess.


Subject(s)
Acute-Phase Reaction/genetics , Acute-Phase Reaction/immunology , Receptors, Interleukin-1/genetics , Sialoglycoproteins/genetics , Abscess/chemically induced , Abscess/immunology , Abscess/physiopathology , Animals , Anorexia/immunology , Anorexia/physiopathology , Appetite/immunology , Body Weight , Cachexia/immunology , Cachexia/physiopathology , Eating , Female , Gene Expression/immunology , Interleukin 1 Receptor Antagonist Protein , Interleukin-6/immunology , Irritants , Liver/immunology , Liver/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Receptors, Tumor Necrosis Factor/immunology , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/immunology , Turpentine
5.
Wound Repair Regen ; 8(6): 547-53, 2000.
Article in English | MEDLINE | ID: mdl-11208182

ABSTRACT

Exogenous administration of tumor necrosis factor-alpha has been shown to both enhance and attenuate cutaneous healing in a dose-dependent manner. We examined the effects of tumor necrosis factor inhibition in the healing wound by both systemic and local administration of tumor necrosis factor-binding protein. Male Balb/C mice underwent dorsal skin incision with subcutaneous implantation of 20 mg polyvinyl alcohol sponges (4 per animal). In Experiment I, one group (n = 20) received intraperitoneal injections of tumor necrosis factor-binding protein (3 mg/kg) at the time of wounding, while another group (n = 20) received saline. Four animals from each group were euthanized on days 1, 3, 5, 7, and 14 postwounding. In Experiment II, one group (n = 10) received an intraperitoneal injection of tumor necrosis factor-binding protein (3 mg/kg) at the time of wounding and every third day thereafter. Another group (n = 10) received an intraperitoneal injection of saline at the time of wounding and every third day thereafter. In Experiment III, one group received a single intraperitoneal injection of tumor necrosis factor-binding protein (3 mg/kg) at the time of wounding (n = 7), or on postwounding day 4 (n = 7), or day 7 (n = 7). Another group received saline injections at the time of wounding (n = 7), or on postwounding days 4 or 7 (n = 7, respectively). All animals in Experiments II and III were killed at postwounding day 14. Wound breaking strengths were assessed using a tensiometer. Wound fluid collected from the implanted sponges was assayed for tumor necrosis factor-alpha and tumor necrosis factor-binding protein levels using a biological assay and enzyme-linked immunosorbent assay, respectively. Collagen gene expression in sponge granulomata was assessed by Northern analysis. Collagen deposition in sponges was quantified by measuring hydroxyproline content. Wounds were significantly weaker in the animals that received repeated injections of tumor necrosis factor-binding protein with a mean wound breaking strength of 93.1 g vs. 186.6 g in controls (p < 0.05). Wound breaking strength in groups that received a single injection of tumor necrosis factor-binding protein on either day 0, 4, or 7 postwounding were no different than their respective controls. There was no difference in the mean hydroxyproline content of sponges between any of the tumor necrosis factor-binding protein groups and their respective controls. Northern analysis for collagen I and III expression also revealed no differences. These data indicate that continued systemic administration of tumor necrosis factor-binding protein resulted in significantly weaker wounds with no corresponding differences in wound collagen content, and collagen gene expression. This suggests that tumor necrosis factor-alpha inhibition throughout healing leads to a qualitatively impaired wound without a quantitative alteration in collagen deposition.


Subject(s)
Carrier Proteins/administration & dosage , Receptors, Tumor Necrosis Factor , Skin/injuries , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Wound Healing/drug effects , Wound Healing/physiology , Wounds and Injuries/drug therapy , Analysis of Variance , Animals , Carrier Proteins/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Enzyme-Linked Immunosorbent Assay , Injections, Intraperitoneal , Male , Mice , Mice, Inbred BALB C , Rats , Receptors, Tumor Necrosis Factor, Type I , Reference Values , Sensitivity and Specificity , Skin/drug effects , Time Factors , Tumor Necrosis Factor Decoy Receptors , Tumor Necrosis Factor-alpha/metabolism
6.
J Gastrointest Surg ; 2(1): 95-101, 1998.
Article in English | MEDLINE | ID: mdl-9841974

ABSTRACT

Production of inflammatory cytokines in the pancreas, lung, and liver is believed to play a major role in the development of severe pancreatitis. This tissue-specific production could lend itself to directed anti-cytokine gene therapy if an appropriate delivery system could be developed. This study was undertaken to examine a novel approach for the delivery of protein-based therapies to the tissues involved during acute pancreatitis. Healthy mice received an intraperitoneal injection of cationic liposomes and a DNA plasmid containing the chloramphenicol acetyltransferase (CAT) reporter gene. Animals were killed at 12 hours and 1, 2, 3, 7, and 14 days with serum, pancreas, lung, and liver harvested. Acute pancreatitis was induced (cerulein, 50 micrograms/kg/hr intraperitoneally x4) in additional mice before or after CAT transfection. The presence of pancreatitis was established in all animals by histologic scoring of pancreata and by serum amylase and lipase levels. CAT transfection efficiency was determined by quantitative CAT enzyme activity within tissue homogenates. Animals that received the liposome were successfully transfected with the CAT gene into the pancreas, lungs, and liver. Maximal transfection in each tissue occurred at 12 hours with decreasing CAT activity over the ensuing 14 days. No healthy animals receiving the CAT gene developed elevations in amylase, lipase, or any histologic parameter of pancreatitis. Transfection efficiency in the pancreas was markedly increased by preexisting or delayed induction of pancreatitis, whereas transfection of the lung and liver was increased to a lesser extent. Gene transfection into the pancreas, liver, and lungs is possible using a cationic liposome delivery system that does not induce pancreatitis or pancreatic inflammation. Pancreatic expression of the gene product is equal to or greater than that of the organs of the reticuloendothelial system and continues at very high efficiency rates during acute pancreatitis.


Subject(s)
Gene Transfer Techniques , Pancreatitis/therapy , Acute Disease , Amylases/blood , Animals , Cations , Chloramphenicol O-Acetyltransferase/analysis , Chloramphenicol O-Acetyltransferase/blood , Chloramphenicol O-Acetyltransferase/genetics , Cytomegalovirus/genetics , Disease Models, Animal , Follow-Up Studies , Gene Expression Regulation, Enzymologic , Genes, Reporter/genetics , Lipase/blood , Liposomes , Liver/enzymology , Lung/enzymology , Male , Mice , Pancreas/enzymology , Pancreas/pathology , Pancreatitis/blood , Pancreatitis/enzymology , Pancreatitis/pathology , Plasmids , Species Specificity , Time Factors , Transfection
7.
Am J Physiol ; 274(6): R1518-25, 1998 06.
Article in English | MEDLINE | ID: mdl-9608004

ABSTRACT

The ob gene product leptin is known to produce anorexia and loss of body fat when chronically administered to both lean and genetically obese mice. The current study was undertaken to examine whether administration of recombinant leptin in quantities sufficient to produce decreases in food intake and body weight and alterations in body composition would elicit either an hepatic acute phase protein response or preferential loss of carcass lean tissue. Mice were administered increasing quantities of recombinant human leptin or human tumor necrosis factor-alpha as a positive control. Although leptin (at 10 mg/kg body wt) produced significant anorexia and weight loss (both P < 0.05), human leptin administration did not appear to induce an hepatic acute phase protein response in either lean or genetically obese mice, as determined by protein synthetic rates in the liver or changes in the plasma concentration of the murine acute phase protein reactants, amyloid A, amyloid P, or seromucoid (alpha1-acid glycoprotein). In addition, human leptin administration did not induce a loss of fat-free dry mass (protein) in lean or obese animals. The findings suggest that at doses adequate to alter food intake and body weight leptin is not a significant inducer of the hepatic acute phase response nor does leptin promote the preferential loss of somatic protein characteristic of a chronic inflammatory process.


Subject(s)
Acute-Phase Reaction/chemically induced , Anorexia/chemically induced , Body Weight/drug effects , Cachexia/chemically induced , Proteins/pharmacology , Animals , Female , Humans , Leptin , Mice , Mice, Inbred C57BL , Time Factors
8.
Biochem Biophys Res Commun ; 246(3): 654-9, 1998 May 29.
Article in English | MEDLINE | ID: mdl-9618268

ABSTRACT

Inducible nitric oxide synthase (iNOS) is expressed during cutaneous wound repair. Mounting evidence suggests that wound nitric oxide (NO) augments collagen accumulation. We hypothesized that in vivo transfection of wound cells with the iNOS gene would increase physiological wound NO levels and thus augment collagen accumulation. Polyvinyl alcohol sponges were instilled with a mammalian expression plasmid (pMP6) containing either the chloramphenicol acetyl transferase (CAT) reporter or murine iNOS gene driven by a CMV immediate-early promoter. Plasmid DNA was injected alone or in complex with cationic liposomes, and the sponges were placed subcutaneously in male Sprague-Dawley rats which had received a longitudinal dorsal midline incision. Animals were sacrificed at different time points post-wounding and the sponges assayed for CAT activity, transfected iNOS mRNA, total nitrate and nitrite concentration (NOx) (as an index of wound NO synthesis), and hydroxyproline content (as an index of sponge collagen accumulation). The results demonstrate that wound cells were more efficiently transfected by naked DNA than by liposome mediated transfection and that maximal expression of both iNOS and CAT occurred at 48 hrs with a rapid decline after this time point. After 7 days, iNOS transfected sponges had accumulated significantly more collagen than those transfected with CAT. We conclude that cutaneous wounds can be successfully transfected by direct injection of naked DNA and that increased iNOS expression precedes an increase in collagen synthesis.


Subject(s)
Collagen/biosynthesis , Nitric Oxide Synthase/biosynthesis , Skin/injuries , Wounds, Penetrating/metabolism , Animals , DNA , Drug Carriers , Genetic Therapy/methods , Liposomes , Male , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Rats , Rats, Sprague-Dawley , Transfection , Wounds, Penetrating/therapy
9.
Cytokine ; 10(6): 452-6, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9632532

ABSTRACT

The acute phase response to inflammation is mediated in part by the endogenous production of pro-inflammatory cytokines. Interleukin 6 (IL-6) and members of its superfamily, including ciliary neurotrophic factor (CNTF) and leukaemia inhibitory factor (LIF) have been implicated as primary mediators of the hepatic acute phase response. In the present report, mice suffering a turpentine-induced myositis were passively immunized with antibodies against either IL-6, CNTF or LIF. Passive immunization against IL-6 attenuated the anorexia and completely prevented the hypoalbuminaemia, and increases in the serum concentration of the acute phase reactants, amyloid P, amyloid A and seromucoid. In contrast, passive immunization against either CNTF or LIF failed to modulate the anorexia, weight loss or hepatic acute phase protein responses. The findings suggest that IL-6, but not other members of its superfamily, is primarily responsible for the hepatic acute phase response, and contributes to the anorexia, associated with turpentine-induced myositis.


Subject(s)
Acute-Phase Reaction/physiopathology , Growth Inhibitors/physiology , Interleukin-6/physiology , Lymphokines/physiology , Myositis/immunology , Nerve Tissue Proteins/physiology , Animals , Antibodies, Monoclonal/pharmacology , Body Weight/drug effects , Ciliary Neurotrophic Factor , Eating/drug effects , Female , Growth Inhibitors/immunology , Immunization, Passive , Interleukin-6/immunology , Leukemia Inhibitory Factor , Lymphokines/immunology , Mice , Mice, Inbred C57BL , Myositis/chemically induced , Nerve Tissue Proteins/immunology , Orosomucoid/analysis , Serum Albumin/analysis , Serum Amyloid A Protein/analysis , Serum Amyloid P-Component/analysis , Turpentine
10.
J Appl Physiol (1985) ; 84(4): 1119-30, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9516174

ABSTRACT

Immunogenicity, pharmacokinetics, and therapeutic efficacy of three novel dimeric soluble tumor necrosis factor (TNF)-receptor I constructs [TNF-binding protein (bp)] were evaluated in 28 baboons, 12 of which were healthy and 16 were challenged with a lethal Escherichia coli bacteremia. The three constructs differed only in the number of extracellular domains of the TNF receptor I and were dimerized with polyethylene glycol. Although all three constructs had generally similar pharmacokinetics when administered to a naive animal, they differed quantitatively in their immunogenicity. Antibodies were detected more frequently, and titers were significantly higher (P < 0.05) in both healthy and septic baboons that received the 4.0-domain TNF-bp construct, compared with animals receiving the 2.6-domain construct. When the TNF-bp constructs were administered a second time (21 days later), the half-lives of the three constructs were significantly shorter in animals that had an antibody response after the first injection. In contrast, all three TNF-bp constructs were equally effective at improving outcome, blocking a systemic TNF-alpha response, and attenuating the cytokine responses when administered at a dose of 1.0 mg/kg body wt 1 h before a lethal E. coli infusion. The findings suggest that immunogenicity of TNF-bp constructs can be altered by changing the number of functional domains, without affecting their capacity to neutralize TNF-alpha and to abrogate TNF-mediated pathology.


Subject(s)
Bacteremia/immunology , Escherichia coli Infections/immunology , Receptors, Tumor Necrosis Factor/immunology , Receptors, Tumor Necrosis Factor/metabolism , Animals , Antibody Formation/physiology , Bacteremia/pathology , Crystallography, X-Ray , Escherichia coli Infections/pathology , Female , Half-Life , Immunoglobulin G/biosynthesis , Kidney/pathology , Kinetics , Leukocyte Count , Male , Molecular Conformation , Papio , Protein Binding
11.
J Immunol ; 158(1): 414-9, 1997 Jan 01.
Article in English | MEDLINE | ID: mdl-8977217

ABSTRACT

TNF-alpha is a pleiotropic cytokine that exists both as a 26-kDa cell-associated and a 17-kDa soluble form. Recently, a class of matrix metalloproteinase inhibitors has been identified that can prevent the processing by TNF convertase of 26-kDa TNF-alpha to its 17-kDa form and can reduce mortality from normally lethal doses of D-galactosamine plus LPS (D-GalN/LPS). Here we report that a matrix metalloproteinase inhibitor, GM-6001, improves survival but does not protect against liver injury from D-GalN/LPS-induced shock in the mouse. In Con A-induced hepatitis, GM-6001 actually exacerbates hepatocellular necrosis and apoptosis despite greater than 90% reduction in plasma TNF-alpha concentrations. Treatment with GM-6001 also has minimal effect on the concentration of membrane-associated TNF-alpha in the livers of animals with Con A induced hepatitis. In contrast, a TNF binding protein (TNF-bp), which neutralizes both membrane-associated and soluble TNF-alpha, prevents D-GalN/LPS- and Con A-induced hepatitis. Our studies suggest that cell-associated TNF-alpha plays a role in the hepatocellular necrosis and apoptosis that accompany D-GalN/LPS- or Con A-induced hepatitis, and that matrix metalloproteinase inhibitors are ineffective in preventing this hepatic injury.


Subject(s)
Chemical and Drug Induced Liver Injury/pathology , Dipeptides/toxicity , Metalloendopeptidases/antagonists & inhibitors , Protease Inhibitors/toxicity , Tumor Necrosis Factor-alpha/chemistry , Tumor Necrosis Factor-alpha/toxicity , Animals , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/mortality , Concanavalin A/toxicity , Drug Synergism , Galactosamine/chemistry , Galactosamine/toxicity , Lipopolysaccharides/toxicity , Liver/drug effects , Liver/injuries , Mice , Mice, Inbred C57BL , Receptors, Tumor Necrosis Factor/blood , Tumor Necrosis Factor-alpha/analysis
12.
Am J Physiol ; 273(6): R1998-2004, 1997 12.
Article in English | MEDLINE | ID: mdl-9435654

ABSTRACT

Exposing rats to a single session of inescapable tail shock (IS) reduces corticosteroid binding globulin (CBG) 24 h later (Fleshner et al., Endocrinology 136: 5336-5342, 1995). The present experiments examined whether reductions in CBG are differentially affected by controllable vs. identical uncontrollable tail shock, are mediated by IS-induced glucocorticoid elevation, or reflect IS-induced activation of the acute phase response and whether IS produces fever. The results demonstrate that 1) equivalent reductions in CBG are observed in response to escapable tail shock or yoked IS, 2) IS-induced CBG reduction is not blocked by adrenalectomy in rats that receive basal corticosteroid replacement or by pretreatment with RU-38486, and 3) IS appears to activate the acute phase response, since IS reduces serum levels of an acute-phase negative reactant (CBG), increases serum levels of acute-phase positive reactants (haptoglobin and alpha 1-acid glycoprotein), and increases core body temperature 20-24 h later.


Subject(s)
Corticosterone/pharmacology , Stress, Psychological/physiopathology , Transcortin/biosynthesis , Adrenalectomy , Animals , Electroshock , Escape Reaction , Male , Rats , Rats, Sprague-Dawley , Restraint, Physical , Stress, Psychological/blood , Time Factors
13.
Shock ; 6(3): 171-6, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8885081

ABSTRACT

Acute visceral ischemia and subsequent reperfusion injury, which accompanies the surgical repair of a thoracoabdominal aorta aneurysm, is associated with high rates of morbidity and mortality. The purpose of the present study was to determine whether endogenous tumor necrosis factor-alpha (TNF-alpha) and interleukin-1 (IL-1) production contributes to organ dysfunction in animals subjected to visceral ischemia secondary to 30 min of supraceliac aortic occlusion. C57BL6/j mice were treated with either a TNF binding protein (TNF-bp-10 mg/kg) or an anti-IL-1 receptor type 1 antibody (150 micrograms) 2 h prior to 30 min of supraceliac aortic occlusion. An additional group of mice received 30 min of infrarenal aortic occlusion to determine the contribution of lower torso ischemia-reperfusion injury to the changes seen following supraceliac aortic occlusion. Visceral organ ischemia for 30 min produced by supraceliac aortic occlusion followed by 2 h of reperfusion produced measurable TNF-alpha in 38% of untreated mice, but TNF-alpha was undetectable in both sham-operated mice and following infrarenal aortic occlusion. After 2 h of reperfusion, lung myeloperoxidase levels were significantly elevated in the mice experiencing visceral ischemia-reperfusion compared with either a sham operation or infrarenal ischemia-reperfusion (11.6 +/- 1.3 U/g vs. 3.4 +/- .2 U/g and 3.7 +/- 1.0 U/g, respectively, p < .05). Pretreatment with TNF-bp and anti-IL-1 antibody decreased lung neutrophil recruitment (7.2 +/- 1.2 U/g and 4.6 +/- 1.1 U/g) and capillary membrane permeability changes in mice following visceral ischemia-reperfusion. The present study demonstrates that brief (30 min) clinically relevant visceral ischemia produces TNF-alpha and IL-1 dependent lung injury.


Subject(s)
Interleukin-1/metabolism , Lung Injury , Reperfusion Injury/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Aorta, Abdominal/surgery , Aspartate Aminotransferases/blood , Aspartate Aminotransferases/metabolism , Capillary Permeability , Female , Interleukin-6/blood , Interleukin-6/metabolism , Liver/injuries , Liver/metabolism , Lung/blood supply , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Peroxidase/metabolism , Time Factors , Vascular Surgical Procedures/methods
14.
Ann Surg ; 223(4): 428-33, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8633922

ABSTRACT

OBJECTIVE: The present study was designed to evaluate whether pre-existing hepatic dysfunction (cirrhosis) leads to increased morbidity and mortality, in part through an inappropriate in vivo tumor necrosis factor-alpha response. SUMMARY BACKGROUND DATA: Vibrio vulnificus is the most commonly isolated member of the noncholera Vibrio sp., responsible for fulminant and frequently fatal septicemia. A strong clinical association exists between hepatic dysfunction and increased morbidity and mortality from Vibrio sp. infection. However, the underlying mechanism behind this association has not been fully delineated. METHODS: Cirrhosis was induced in C57BL/6 (15 to 20 g) mice using thrice-weekly injections of carbon tetrachloride (CCl4) for 7 weeks. Either a 7.0 to 9.5 X 10(7) (low dose) or a 0.8 to 1.2 X 10(9) colony-forming unit (high dose) of V. vulnificus was administered through a mini-laparotomy incision via transgastric puncture into both cirrhotic and control animals. RESULTS: Mortality in cirrhotic mice to low- and high-dose Vibrio infection was 88% (7/8) and 100% (8/8), respectively, whereas mortality in control animals was 0% (0/8) and 12% (1/8), respectively (p<0.01). Tumor necrosis factor-alpha mRNA could be detected by reverse transcriptase polymerase chain reaction in livers and lungs from infected animals 2 and 4 hours after Vibrio administration in both control and cirrhotic animals. Lung and liver tumor necrosis factor-alpha bioactivity, however, was significantly lower in cirrhotic animals infected with Vibrio when compared with controls. Serum tumor necrosis factor-alpha was only sporadically detected in both groups of Vibrio-infected animals. When cirrhotic mice challenged with a low dose of Vibrio sp. were pretreated with 1.0 mg/kg body weight of a novel tumor necrosis factor-alpha receptor immunoadhesin, the increased mortality was completely prevented. CONCLUSIONS: Cirrhotic mice show increased mortality to Vibrio infection, and this increased mortality is dependent on an in vivo tumor necrosis factor-alpha response.


Subject(s)
Liver Cirrhosis, Experimental/physiopathology , Tumor Necrosis Factor-alpha/physiology , Vibrio Infections/physiopathology , Animals , Female , Liver/chemistry , Liver Cirrhosis, Experimental/metabolism , Liver Cirrhosis, Experimental/virology , Lung/chemistry , Mice , Mice, Inbred C57BL , RNA, Messenger/analysis , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/genetics , Vibrio Infections/metabolism
15.
Surgery ; 116(2): 276-83; discussion 283-4, 1994 Aug.
Article in English | MEDLINE | ID: mdl-7519362

ABSTRACT

BACKGROUND: Glutamine metabolism in fibroblasts is essential for energy production, nucleotide biosynthesis, and growth during wound healing. Because cytokines can impair fibroblast proliferation, we tested the hypothesis that cytokines impair glutamine metabolism. We studied the influence of several cytokines on the expression of glutaminase, the major enzyme of intracellular glutamine metabolism in fibroblasts. METHODS: Human foreskin fibroblasts were incubated for 6 and 12 hours with varying doses (10, 100, or 1000 units/ml) of interleukin (IL)-1, IL-6, tumor necrosis factor-alpha, or gamma-interferon. Cell lysates were assayed for glutaminase-specific activity, and glutaminase protein content was measured by Western blotting with a polyclonal antibody. Total cellular RNA was extracted, and relative glutaminase messenger RNA levels were determined by Northern blotting with a 32P-labeled glutaminase complement DNA-derived probe. These mRNA levels were normalized by blotting with a beta-actin cDNA-derived probe as control. Cell nuclei were isolated, and nuclear run-ons were used to determine relative glutaminase mRNA transcription rates. RESULTS: IL-1, IL-6, tumor necrosis factor-alpha, and gamma-interferon decreased glutaminase activity and protein concentration after a 12-hour incubation in a dose-independent fashion. No difference was noted at 6 hours. Western blot analysis showed a 30% to 60% reduction in glutaminase protein in treated cells. These cytokines also decreased glutaminase mRNA levels, consistent with transcriptional regulation. This was confirmed by nuclear run-on assays that showed a decrease in the number of glutaminase transcripts. CONCLUSIONS: A variety of different pro-inflammatory cytokines decrease glutaminase expression in cultured human fibroblasts. This cytokine-mediated inhibition of glutamine metabolism may limit the availability of key glutamine-derived intermediates and impair fibroblast proliferation in certain patients.


Subject(s)
Cytokines/pharmacology , Glutaminase/analysis , Cells, Cultured , Fibroblasts/enzymology , Glutaminase/genetics , Glutamine/metabolism , Humans , Interferons/pharmacology , Interleukin-1/pharmacology , Interleukin-6/pharmacology , RNA, Messenger/analysis , Tumor Necrosis Factor-alpha/pharmacology
16.
Arch Surg ; 129(1): 59-65, 1994 Jan.
Article in English | MEDLINE | ID: mdl-7904150

ABSTRACT

PURPOSE: Although glutamine is required to maintain gut mucosal metabolism and function, intestinal glutamine uptake from the gut lumen and from the bloodstream is decreased during sepsis. We hypothesized that endogenous mucosal glutamine biosynthesis is increased during endotoxemia, and we attempted to define the "stress" mediators that regulate the activity of small intestinal glutamine synthetase (GS), the principal enzyme of de novo glutamine biosynthesis in the gut. METHODS: Adult rats received Escherichia coli lipopolysaccharide (LPS) (7.5 mg/kg intraperitoneally), RU 38486 (a glucocorticoid antagonist; 10 mg/kg by gavage) 2 hours prior to LPS administration, antibody to tumor necrosis factor (TNF) (4 mg/kg intraperitoneally) prior to LPS administration, or ketorolac tromethamine (a prostaglandin synthesis inhibitor; 1 mg/kg intraperitoneally) followed by LPS administration. Mucosal GS activity was assayed 12 hours after LPS administration. In a separate set of studies, cultured intestinal mucosal cells (Caco-2) were exposed to LPS, interleukin 1 (IL-1), IL-6, TNF-alpha, interferon-gamma, prostaglandin E2, or dexamethasone. Twelve hours later, GS activity was assayed and messenger RNA was extracted. The GS transcripts were labeled with a GS complementary DNA probe radiolabeled with phosphorus 32, were quantitated by phosphoimaging, and were normalized to beta-actin. RESULTS: In vivo LPS treatment increased mucosal GS activity by 250%. Pretreatment with antibody to TNF or ketorolac did not inhibit the LPS-induced increase in mucosal GS, whereas pretreatment with RU 38486 attenuated the increase in gut GS activity by 60%. Lipopolysaccharide, IL-1, IL-6, TNF-alpha, gamma-interferon, and prostaglandin E2 did not increase GS activity in Caco-2 cells, whereas dexamethasone increased GS activity and messenger RNA 2.5-fold and threefold, respectively. These data indicate that cytokines and prostaglandins (prostaglandin E2) do not regulate mucosal GS expression during endotoxemia. Glucocorticoids, however, stimulate GS gene expression directly. CONCLUSIONS: This hormonally mediated response may support de novo mucosal GS during septic states when uptake of glutamine from the lumen and blood is decreased.


Subject(s)
Endotoxins/blood , Glutamate-Ammonia Ligase/metabolism , Toxemia/enzymology , Animals , Cytokines/physiology , Dexamethasone/pharmacology , Gene Expression , Glutamate-Ammonia Ligase/genetics , Humans , Male , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Tumor Cells, Cultured
17.
Surgery ; 112(2): 278-83, 1992 Aug.
Article in English | MEDLINE | ID: mdl-1641766

ABSTRACT

BACKGROUND: The metabolism of glutamine by the small intestinal mucosal cells is highly dependent on the glutaminase enzyme. Because mucosal glutamine utilization is increased after operation, we hypothesized that the elevated glucocorticoid hormones that occur after surgical stress regulate expression of mucosal glutaminase at the molecular level. METHODS: Adult rats received saline solution or dexamethasone (0.5 mg/kg, one dose) and were sacrificed at various times after treatment. Jejunal mucosal total RNA was extracted for Northern hybridization with an alpha-32P-labeled rat glutaminase cDNA. The mRNA of the constitutively expressed gene beta-actin was the control for RNA loading. Quantitation of glutaminase transcripts was determined by densitometric scanning and normalized to actin. Glutaminase activity (micromoles per milligram of protein per hour) and its kinetic parameters, maximal transport velocity (micromoles per milligram of protein per hour) and Michaelis-Menten constant (micromoles per liter), were also determined. RESULTS: Dexamethasone increased glutaminase mRNA (twofold at 4 hours, sixfold at 12 hours; p less than 0.01) and glutaminase-specific activity. The increase in message preceded the increase in activity by 4 hours, consistent with de novo RNA synthesis followed by protein synthesis. The increase in glutaminase activity was the result of a 21% increase in the maximal enzyme capacity (maximal transport velocity = 8.6 +/- 0.5 mumol/mg protein/hr in control rats vs 10.4 +/- 0.3 mumol/mg protein/hr in rats treated with dexamethasone; p less than 0.01) rather than a change in enzyme affinity (Michaelis-Menten constant). CONCLUSION: Glucocorticoids may accelerate intestinal glutamine utilization by increasing glutaminase expression, an adaptive response that could provide more energy for mucosal cells in stress states.


Subject(s)
Dexamethasone/pharmacology , Glutaminase/metabolism , Jejunum/enzymology , Animals , Autoradiography , Glutaminase/genetics , Male , RNA, Messenger/metabolism , Rats , Rats, Inbred Strains , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...