Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 111
Filter
1.
J Clin Invest ; 129(10): 4180-4193, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31265436

ABSTRACT

Dengue virus (DENV) infection causes a characteristic pathology in humans involving dysregulation of the vascular system. In some patients with dengue hemorrhagic fever (DHF), vascular pathology can become severe, resulting in extensive microvascular permeability and plasma leakage into tissues and organs. Mast cells (MCs), which line blood vessels and regulate vascular function, are able to detect DENV in vivo and promote vascular leakage. Here, we identified that a MC-derived protease, tryptase, is consequential for promoting vascular permeability during DENV infection, through inducing breakdown of endothelial cell tight junctions. Injected tryptase alone was sufficient to induce plasma loss from the circulation and hypovolemic shock in animals. A potent tryptase inhibitor, nafamostat mesylate, blocked DENV-induced vascular leakage in vivo. Importantly, in two independent human dengue cohorts, tryptase levels correlated with the grade of DHF severity. This study defines an immune mechanism by which DENV can induce vascular pathology and shock.


Subject(s)
Capillary Permeability , Dengue Virus/metabolism , Dengue/enzymology , Endothelium, Vascular/enzymology , Mast Cells/enzymology , Shock/enzymology , Tight Junctions/metabolism , Tryptases/metabolism , Animals , Benzamidines , Cell Line , Dengue/drug therapy , Dengue/pathology , Dengue/virology , Endothelium, Vascular/pathology , Endothelium, Vascular/virology , Guanidines/pharmacology , Humans , Mast Cells/pathology , Mast Cells/virology , Mice , Shock/drug therapy , Shock/pathology , Shock/virology , Tight Junctions/pathology , Tryptases/antagonists & inhibitors , Tryptases/genetics
2.
Nat Cell Biol ; 19(10): 1237-1247, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28920952

ABSTRACT

TNF is a master proinflammatory cytokine whose pathogenic role in inflammatory disorders can, in certain conditions, be attributed to RIPK1 kinase-dependent cell death. Survival, however, is the default response of most cells to TNF stimulation, indicating that cell demise is normally actively repressed and that specific checkpoints must be turned off for cell death to proceed. We identified RIPK1 as a direct substrate of MK2 in the TNFR1 signalling pathway. Phosphorylation of RIPK1 by MK2 limits cytosolic activation of RIPK1 and the subsequent assembly of the death complex that drives RIPK1 kinase-dependent apoptosis and necroptosis. In line with these in vitro findings, MK2 inactivation greatly sensitizes mice to the cytotoxic effects of TNF in an acute model of sterile shock caused by RIPK1-dependent cell death. In conclusion, we identified MK2-mediated RIPK1 phosphorylation as an important molecular mechanism limiting the sensitivity of the cells to the cytotoxic effects of TNF.


Subject(s)
Apoptosis/drug effects , Fibroblasts/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Shock/chemically induced , Tumor Necrosis Factor-alpha/toxicity , Animals , Cell Line , Cytosol/enzymology , Disease Models, Animal , Enzyme Activation , Female , Fibroblasts/enzymology , Fibroblasts/pathology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , Necrosis , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptors, Tumor Necrosis Factor, Type I/agonists , Receptors, Tumor Necrosis Factor, Type I/metabolism , Serine , Shock/enzymology , Shock/pathology , Shock/prevention & control , Signal Transduction/drug effects , Time Factors
3.
Shock ; 45(5): 483-9, 2016 May.
Article in English | MEDLINE | ID: mdl-26717111

ABSTRACT

There is currently no effective treatment for multiorgan failure following shock other than supportive care. A better understanding of the pathogenesis of these sequelae to shock is required. The intestine plays a central role in multiorgan failure. It was previously suggested that bacteria and their toxins are responsible for the organ failure seen in circulatory shock, but clinical trials in septic patients have not confirmed this hypothesis. Instead, we review here evidence that the digestive enzymes, synthesized in the pancreas and discharged into the small intestine as requirement for normal digestion, may play a role in multiorgan failure. These powerful enzymes are nonspecific, highly concentrated, and fully activated in the lumen of the intestine. During normal digestion they are compartmentalized in the lumen of the intestine by the mucosal epithelial barrier. However, if this barrier becomes permeable, e.g. in an ischemic state, the digestive enzymes escape into the wall of the intestine. They digest tissues in the mucosa and generate small molecular weight cytotoxic fragments such as unbound free fatty acids. Digestive enzymes may also escape into the systemic circulation and activate other degrading proteases. These proteases have the ability to clip the ectodomain of surface receptors and compromise their function, for example cleaving the insulin receptor causing insulin resistance. The combination of digestive enzymes and cytotoxic fragments leaking into the central circulation causes cell and organ dysfunction, and ultimately may lead to complete organ failure and death. We summarize current evidence suggesting that enteral blockade of digestive enzymes inside the lumen of the intestine may serve to reduce acute cell and organ damage and improve survival in experimental shock.


Subject(s)
Multiple Organ Failure/enzymology , Multiple Organ Failure/metabolism , Shock/enzymology , Shock/metabolism , Animals , Enzymes/metabolism , Humans , Insulin Resistance/physiology , Multiple Organ Failure/etiology , Pancreas/enzymology , Peptide Hydrolases/metabolism , Shock/complications
4.
Sci Transl Med ; 5(169): 169ra11, 2013 Jan 23.
Article in English | MEDLINE | ID: mdl-23345609

ABSTRACT

Shock, sepsis, and multiorgan failure are associated with inflammation, morbidity, and high mortality. The underlying pathophysiological mechanism is unknown, but evidence suggests that pancreatic enzymes in the intestinal lumen autodigest the intestine and generate systemic inflammation. Blocking these enzymes in the intestine reduces inflammation and multiorgan dysfunction. We investigated whether enzymatic blockade also reduces mortality after shock. Three rat shock models were used here: hemorrhagic shock, peritonitis shock induced by placement of cecal material into the peritoneum, and endotoxin shock. One hour after initiation of hemorrhagic, peritonitis, or endotoxin shock, animals were administered one of three different pancreatic enzyme inhibitors--6-amidino-2-naphtyl p-guanidinobenzoate dimethanesulfate, tranexamic acid, or aprotinin--into the lumen of the small intestine. In all forms of shock, blockade of digestive proteases with protease inhibitor attenuated entry of digestive enzymes into the wall of the intestine and subsequent autodigestion and morphological damage to the intestine, lung, and heart. Animals treated with protease inhibitors also survived in larger numbers than untreated controls over a period of 12 weeks. Surviving animals recovered completely and returned to normal weight within 14 days after shock. The results suggest that the active and concentrated digestive enzymes in the lumen of the intestine play a central role in shock and multiorgan failure, which can be treated with protease inhibitors that are currently available for use in the clinic.


Subject(s)
Digestion , Intestines/enzymology , Pancreas/enzymology , Protease Inhibitors/therapeutic use , Serine Proteases/metabolism , Shock/drug therapy , Shock/enzymology , Animals , Digestion/drug effects , Disease Models, Animal , Hemorrhage/chemically induced , Hemorrhage/drug therapy , Hemorrhage/enzymology , Hemorrhage/pathology , Intestines/drug effects , Male , Organ Specificity/drug effects , Pancreas/drug effects , Peritonitis/chemically induced , Peritonitis/drug therapy , Peritonitis/enzymology , Peritonitis/pathology , Protease Inhibitors/pharmacology , Rats , Rats, Wistar , Shock/blood , Shock/chemically induced , Survival Analysis , Troponin/blood
5.
Shock ; 38(3): 262-7, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22576000

ABSTRACT

In bowel ischemia, impaired mucosal integrity may allow intestinal pancreatic enzyme products to become systemic and precipitate irreversible shock and death. This can be attenuated by pancreatic enzyme inhibition in the small-bowel lumen. It is unresolved, however, whether ischemically mediated mucosal disruption is the key event allowing pancreatic enzyme products systemic access and whether intestinal digestive enzyme activity in concert with increased mucosal permeability leads to shock in the absence of ischemia. To test this possibility, the small intestinal lumen of nonischemic rats was perfused for 2 h with either digestive enzymes, a mucin disruption strategy (i.e., mucolytics) designed to increase mucosal permeability, or both, and animals were observed for shock. Digestive enzymes perfused included trypsin, chymotrypsin, elastase, amylase, and lipase. Control (n = 6) and experimental animals perfused with pancreatic enzymes only (n = 6) or single enzymes (n = 3 for each of the five enzyme groups) maintained stable hemodynamics. After mucin disruption using a combination of enteral N-acetylcysteine, atropine, and increased flow rates, rats (n = 6) developed mild hypotension (P < 0.001 compared with groups perfused with pancreatic enzymes only after 90 min) and increased intestinal permeability to intralumenally perfused fluorescein isothiocyanate-dextran 20 kd (P < 0.05) compared with control and enzyme-only groups, but there were no deaths. All animals perfused with both digestive enzymes and subjected to mucin disruption (n = 6) developed hypotension and increased intestinal permeability (P < 0.001 after 90 min). Pancreatic enzymes were measured in the intestinal wall of both groups subjected to mucin disruption, but not in the enzyme-only or control groups. Depletion of plasma protease inhibitors was found only in animals perfused with pancreatic enzymes plus mucin disruption, implicating increased permeability and intralumenal pancreatic enzyme egress in this group. These experiments demonstrate that increased bowel permeability via mucin disruption in the presence of pancreatic enzymes can induce shock and increase systemic protease activation in the absence of ischemia, implicating bowel mucin disruption as a key event in early ischemia. Digestive enzymes and their products, if allowed to penetrate the gut wall, may trigger multiorgan failure and death.


Subject(s)
Enzymes/pharmacology , Expectorants/pharmacology , Intestine, Small/drug effects , Pancreas/enzymology , Shock/enzymology , Animals , Blotting, Western , Hemodynamics/drug effects , Immunohistochemistry , Intestine, Small/metabolism , Male , Pancreatic Elastase/metabolism , Peptide Hydrolases/metabolism , Permeability/drug effects , Rats , Rats, Wistar , Shock/etiology , Trypsin/metabolism
6.
Article in English | MEDLINE | ID: mdl-23366584

ABSTRACT

Physiological shock and subsequent multi-organ failure is one of the most important medical problems from a mortality point of view. No agreement exists for mechanisms that lead to the relative rapid cell and organ failure during this process and no effective treatment. We postulate that the digestive enzymes synthesized in the pancreas and transported in the lumen of the small intestine as requirement of normal food digestion play a central role in multi-organ failure. These powerful enzymes are usually compartmentalized in the lumen of the intestine by the mucosal barrier, but may escape into the wall of the intestine if the permeability of the mucosal lining increases. Entry of the digestive enzymes into the wall of the intestine precipitates an autodigestion process as well as an escape of pancreatic enzymes and breakdown products generated by them into the system circulation. The consequence of autodigestion is multiorgan failure. We discuss the possibility to block the digestive enzymes in acute forms of shock as a potential therapeutic intervention.


Subject(s)
Multiple Organ Failure/enzymology , Shock/enzymology , Humans , Intestines/enzymology , Pancreas/enzymology
8.
J Biol Chem ; 283(22): 15258-70, 2008 May 30.
Article in English | MEDLINE | ID: mdl-18378686

ABSTRACT

Tumor necrosis factor-alpha (TNF-alpha) produced by macrophages in response to CpG DNA induces severe liver injury and subsequent death of D-galactosamine (D-GalN)-sensitized mice. In the present study we demonstrate that mice pre-exposed to CpG DNA are resistant to liver injury and death induced by CpG DNA/D-GalN. CpG DNA/D-GalN failed to induce TNF-alpha production and hepatocyte apoptosis in the mice pre-exposed to CpG DNA. In addition, macrophages isolated from the CpG DNA-pretreated mice showed suppressed activation of MAPKs and NF-kappaB and production of TNF-alpha in response to CpG DNA, indicating that the CpG DNA-mediated protection of CpG DNA/D-GalN-challenged mice is due to the hyporesponsiveness of macrophages to CpG DNA. CpG DNA pretreatment in vivo inhibited expression of interleukin-1 receptor-associated kinase (IRAK)-1 while inducing IRAK-M expression in macrophages. Suppressed expression of IRAK-1 was responsible for the macrophage hyporesponsiveness to CpG DNA. However, increased expression of IRAK-M was not sufficient to render macrophages hyporesponsive to CpG DNA but was required for induction of the optimal level of macrophage hyporesponsiveness. Taken together, reduced expression of IRAK-1 and increased expression of IRAK-M after CpG DNA pretreatment resulted in the hyporesponsiveness of macrophages that leads to the protection of mice from hepatic injury and death caused by CpG DNA/D-GalN.


Subject(s)
Galactosamine/toxicity , Gene Expression Regulation, Enzymologic/drug effects , Interleukin-1 Receptor-Associated Kinases/biosynthesis , Liver/enzymology , Macrophages/enzymology , Oligodeoxyribonucleotides/toxicity , Shock/enzymology , Animals , Apoptosis/drug effects , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Galactosamine/agonists , Liver/injuries , Mice , Mice, Inbred BALB C , Oligodeoxyribonucleotides/agonists , Shock/chemically induced , Tumor Necrosis Factor-alpha/biosynthesis
9.
Int Immunopharmacol ; 7(14): 1845-51, 2007 Dec 20.
Article in English | MEDLINE | ID: mdl-18039521

ABSTRACT

My association with Tony Hugli, long-term editor of Immunopharmacology and International Immunopharmacology, came about by a specific and long-standing problem in inflammation research. What is the trigger mechanism of inflammation in physiological shock? This is an important clinical problem due to the high mortality associated with physiological shock. We joined forces in the search of the answer to this question for more than a decade. Our journey eventually led to development of the hypothesis that shock may be associated with pancreatic enzymes, a set of powerful digestive enzymes that are an integral part of human digestion. The digestive enzymes need to be compartmentalized in the lumen of the intestine where they break down a broad spectrum of biological molecules into their building blocks, suitable for molecular transport across the mucosal epithelium into the circulation. The mucosal epithelial barrier is the key element for compartmentalization of the digestive enzymes. But under conditions when the mucosal barrier is compromised, the fully activated digestive enzymes in the lumen of the intestine are transported into the wall of the intestine, starting an auto-digestion process. In the process several classes of mediators are generated that by themselves have inflammatory activity and upon entry into the central circulation generate the hallmarks of inflammation and eventually cause multi-organ failure. Thus, our journey led to a new hypothesis, which is potentially of fundamental importance for death by multi-organ failure. The auto-digestion hypothesis is in line with the century old observation that the intestine plays a special role on shock - indeed it is the organ for digestion. Auto-digestion may be the prize to pay for life-long nutrition.


Subject(s)
Digestion/physiology , Inflammation Mediators/physiology , Inflammation/enzymology , Pancreas/enzymology , Animals , Digestion/drug effects , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Inflammation/physiopathology , Inflammation Mediators/pharmacology , Intestinal Mucosa/metabolism , Intestines/enzymology , Multiple Organ Failure/physiopathology , Rats , Shock/enzymology , Shock/physiopathology , Systemic Inflammatory Response Syndrome/physiopathology
10.
Shock ; 27(2): 113-23, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17224784

ABSTRACT

After the discovery that glycogen synthase kinase (GSK) 3beta plays a fundamental role in the regulation of the activity of nuclear factor kappaB, a number of studies have investigated the effects of this protein kinase in the regulation of the inflammatory process. The GSK-3beta inhibition, using genetically modified cells and chemically different pharmacological inhibitors, affects the regulation of various inflammatory mediators in vitro and in vivo. Insulin, an endogenous inhibitor of GSK-3 in the pathway leading to the regulation of glycogen synthase activity, has recently been clinically used in the therapy for septic shock. The beneficial anti-inflammatory effects of insulin in preclinical and clinical studies could possibly be due, at least in part, to the inhibition of GSK-3 and not directly correlated to the regulation of blood glucose. We describe the latest studies describing the effects of GSK-3 inhibition as potential target of the therapy for diseases associated with inflammation, ischemia/reperfusion, and shock.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase/metabolism , NF-kappa B/metabolism , Shock/enzymology , Animals , Anti-Inflammatory Agents , Blood Glucose/metabolism , Enzyme Inhibitors/therapeutic use , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Humans , Hypoglycemic Agents/therapeutic use , Inflammation/drug therapy , Inflammation/enzymology , Insulin/therapeutic use , Reperfusion Injury/drug therapy , Reperfusion Injury/enzymology , Shock/drug therapy
11.
Curr Drug Targets ; 7(9): 1195-204, 2006 Sep.
Article in English | MEDLINE | ID: mdl-17017895

ABSTRACT

There were strong evidences that NO has capital importance in the progressive vasodilatation that associates to the varied circulatory shock forms. The decreased systemic vascular resistance observed in irreversible hemorrhagic (hypovolemic) and septic shock may be due to the excess production of nitric oxide. Other forms of shock associated to anaphylaxis (anaphylactic shock, SIRS) and ischemia reperfusion injury (cardiogenic shock, organ transplants), may involve nitric oxide overproduction. In these situations, the nitric oxide-induced loss of vascular sensitivity to catecholamines and myocardial depression contributes to lethal hypotension. As NO vasodilatation is cyclic GMP-mediated, there were two therapeutical options: a) The unspecific NO synthesis inhibition by L-arginine analogs, iNOS-specific inhibition by corticoids and/or aminoguanidine and; b) Guanylyl cyclase inhibition by MB. As the NO synthesis inhibition is associated to tissue necrosis and adverse hemodynamic effects and its clinical use was associated with high mortality, the second option using MB is safer and more rational. The elaboration of this text was motivated to suggest the guanylyl cyclase inhibition by MB as vasoplegic circulatory shock therapeutical target.


Subject(s)
Drug Delivery Systems/methods , Enzyme Inhibitors/therapeutic use , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/metabolism , Methylene Blue/therapeutic use , Shock/enzymology , Animals , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Humans , Methylene Blue/chemistry , Methylene Blue/metabolism , Shock/blood , Shock/drug therapy
12.
Acta Paediatr ; 95(6): 747-50, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16754559

ABSTRACT

UNLABELLED: We studied the association between genetic polymorphisms of the renin-angiotensin system and the risk for circulatory failure (CF) during the first three postnatal days in 104 very-low-birthweight preterm infants. CONCLUSION: Infants with angiotensin-converting enzyme DD genotype were protected against CF (adjusted OR 0.41, 95% CI 0.19-0.89).


Subject(s)
Infant, Very Low Birth Weight , Peptidyl-Dipeptidase A/genetics , Polymorphism, Genetic , Shock/genetics , Female , Genotype , Humans , Infant, Newborn , Male , Shock/enzymology
13.
Clin Exp Pharmacol Physiol ; 33(3): 264-8, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16487272

ABSTRACT

In a previous study, we demonstrated that heat shock augments the contractility of vascular smooth muscle through the stress response. 2. In the present study, we investigated whether Rho-kinases play a role in heat shock-induced augmentation of vascular contractility in rat isolated aorta. 3. Rat aortic strips were mounted in organ baths, exposed to 42 C for 45 min and subjected to contractile or relaxant agents 5 h later. 4. The level of expression of Rho-kinases in heat shock-exposed tissues was no different to that of control tissues, whereas heat shock induced heat shock protein (Hsp) 72 at 3 and 5 h. Heat shock resulted in an increase in vascular contractility in response to phenylephrine 5 h later. 5. The Rho-kinase inhibitors Y27632 (30 nmol/L-10 mmol/L) or HA 1077 (10 nmol/L-10 mmol/L) relaxed 1.0 mmol/L phenylephrine-precontracted vascular strips in a concentration-dependent manner; these effects were attenuated in heat shock-exposed strips. Pretreatment with Y27632 resulted in greater inhibition of the maximum contraction in control strips compared with those in heat shock-exposed strips. 6. The results of the present study suggest that Rho-kinases are unlikely to be involved in heat shock-induced augmentation of vascular contractility.


Subject(s)
Hot Temperature , Muscle, Smooth, Vascular/physiology , Protein Serine-Threonine Kinases/physiology , Shock/enzymology , Shock/physiopathology , Actins/biosynthesis , Amides/pharmacology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Blotting, Western , HSP72 Heat-Shock Proteins/metabolism , In Vitro Techniques , Intracellular Signaling Peptides and Proteins , Male , Muscle Contraction/physiology , Muscle Relaxation/physiology , Myosin Light Chains/metabolism , Phenylephrine/antagonists & inhibitors , Phenylephrine/pharmacology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Vasoconstrictor Agents/pharmacology , rho-Associated Kinases
14.
J Intern Med ; 258(4): 385-7, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16164579

ABSTRACT

In adverse reactions with shock, tripled tryptase values can support a diagnosis of anaphylaxis. A 51-year old physically fit woman experienced angio-oedema and hypotensive shock after irbesartan ingestion requiring noradrenaline infusion. Serum tryptase rose to three times the normal value. Total immunoglobulin E and skin prick tests were normal, however. As nonallergic increases in tryptase have been observed, e.g. during angio-oedema from angiotensin-converting enzyme inhibitors, and bradykinin itself can degranulate mast cells acutely, we interpret the reaction as a class effect. To our knowledge, our report is one of the first on shock and angio-oedema from irbesartan.


Subject(s)
Angioedema/chemically induced , Angiotensin II Type 1 Receptor Blockers/adverse effects , Biphenyl Compounds/adverse effects , Hypotension/chemically induced , Serine Endopeptidases/blood , Shock/chemically induced , Tetrazoles/adverse effects , Angioedema/drug therapy , Angioedema/enzymology , Biomarkers/blood , Female , Humans , Hypotension/drug therapy , Hypotension/enzymology , Irbesartan , Mast Cells/drug effects , Middle Aged , Norepinephrine/therapeutic use , Shock/drug therapy , Shock/enzymology , Skin Tests , Tryptases
16.
J Trauma ; 56(4): 749-58; discussion 758-9, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15187737

ABSTRACT

BACKGROUND: Hypertonic saline (HTS) has been shown to modulate the inflammatory response after shock. We have previously demonstrated that heme oygenase-1 (HO-1) induction is protective against gut dysfunction in models of shock-induced gut ischemia/reperfusion (I/R). We therefore hypothesized that HTS prevents gut inflammation, injury, and impaired transit by inducing HO-1 in a model of gut I/R. METHODS: Rats underwent 60 minutes of superior mesenteric artery occlusion (SMAO) and then were resuscitated with 4 mL/kg of HTS, an equal volume of lactated Ringer's (LR) solution (4 mL/kg, low volume), or equal salt LR solution (32 mL/kg, high volume) and compared with SMAO alone or shams. A separate group was pretreated with the HO-1 blocker Sn protoporphyrin IX (SNP IX) before SMAO plus HTS. At 6 hours of reperfusion, transit was determined and ileum harvested for HO-1 (anti-inflammatory) and inducible nitric oxide synthase (proinflammatory) immunoreactivity, myeloperoxidase (MPO) activity, and histologic injury. Data are expressed as mean +/- SEM (analysis of variance). RESULTS: Intestinal transit was severely impaired after SMAO (2.5 +/- 0.1), improved with low- and high-volume LR solution (3.2 +/- 0.2 and 3.1 +/- 0.1, not significant), but returned to sham (4.6 +/- 0.2) with HTS (4.8 +/- 0.2). Pretreatment with SNP abrogated this protective effect. Myeloperoxidase activity was significantly increased by SMAO (SMAO, 2.3 +/- 0.3; sham, 0.4 +/- 0.05), lessened by low- and high-volume LR solution (1.5 +/- 0.3 and 1.7 +/- 0.4), but returned to sham levels with HTS (1.0 +/- 0.01). Activity with SNP IX pretreatment was significantly increased (4.04 +/- 0.8). Mucosal injury followed a similar pattern. Inducible nitric oxide synthase was increased by SMAO and low- and high-volume LR solution (0.8 +/- 0.2, 0.8 +/- 0.03, and 0.8 +/- 0.02, respectively; sham, 0.5 +/- 0.02), but significantly reduced by HTS (0.7 +/- 0.02). HO-1 was induced by SMAO and low- and high-volume LR solution (0.33 +/- 0.02, 0.32 +/- 0.03, and 0.37 +/- 0.4, respectively; sham, 0.0 +/- 0.0), but was further increased with HTS (0.49 +/- 0.04). CONCLUSION: HTS resuscitation protects against inflammation, injury, and impaired intestinal transit after gut I/R in part by inducing HO-1. This is a novel mechanism of HO-1 protection.


Subject(s)
Enzyme Induction/drug effects , Heme Oxygenase (Decyclizing)/biosynthesis , Inflammation/prevention & control , Reperfusion Injury/prevention & control , Saline Solution, Hypertonic/therapeutic use , Animals , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1 , Intestinal Mucosa/drug effects , Intestinal Mucosa/injuries , Male , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Rats , Rats, Sprague-Dawley , Resuscitation/methods , Shock/enzymology , Shock/metabolism , Shock/therapy
17.
JOP ; 3(5): 139-43, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12221328

ABSTRACT

BACKGROUND: There is a lack of data concerning pancreatic involvement during shock. AIM: To evaluate possible pancreatic alterations in the early phase of shock. SETTING: Twelve consecutive patients with shock were studied within 2 hours from the onset of illness. Seven patients died during the hospital stay: 3 within 4 hours from admission, 3 within 4-8 hours and 1 within 12 hours. MAIN OUTCOME MEASURES: Amylase, lipase, C-reactive protein, amyloid A, interleukin 6, procalcitonin and vascular cell adhesion molecule-1 serum concentrations were determined on admission and 4, 8, and 12 hours afterward. All patients underwent imaging studies of the pancreas. RESULTS: None of the patients developed clinical signs or morphological alterations compatible with acute pancreatitis. Serum amylase levels were above the upper reference limit in 7 patients (58.3%) and serum lipase levels in 2 patients (16.7%; P=0.062). There were no significant differences found between survivors and non-survivors in the serum concentrations of all the proteins studied. CONCLUSIONS: In patients with shock, amylase seems to be more frequently elevated than lipase. None of the patients showed pancreatic alterations at imaging techniques.


Subject(s)
Pancreas/physiopathology , Shock/physiopathology , Aged , Aged, 80 and over , Amylases/blood , Female , Humans , Lipase/blood , Male , Pancreas/enzymology , Shock/blood , Shock/enzymology , Shock/mortality
18.
Diabetes ; 49(3): 346-55, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10868955

ABSTRACT

In this study, the anti-inflammatory actions of the peroxisome proliferator-activated receptor (PPAR)-gamma agonists 15-deoxy-delta 12,14-prostaglandin J2 (15-d-delta 12,14-PGJ2) and troglitazone have been examined. Treatment of RAW 264.7 cells and CD-1 mouse peritoneal macrophages with lipopolysaccharide (LPS) + interferon-gamma (IFN-gamma) results in inducible nitric oxide synthase (iNOS), inducible cyclooxygenase (COX-2) and interleukin-1 (IL-1) expression, increased production of nitric oxide, and the release of IL-1. In a concentration-dependent manner, 15-d-delta 12,14-PGJ2 inhibits each of these proinflammatory actions of LPS + IFN-gamma, with half-maximal inhibition at approximately 0.5 microg/ml and complete inhibition at 1-5 microg/ml. The inhibitory actions of 15-d-delta 12,14-PGJ2 on LPS + IFN-gamma-induced inflammatory events are not associated with the inhibition of iNOS enzymatic activity or macrophage cell death, but appear to result from an inhibition of iNOS and IL-1 transcription. In addition, the anti-inflammatory actions of 15-d-delta 12,14-PGJ2 are not limited to peritoneal macrophages, as 15-d-delta 12,14-PGJ2 prevents TNF-alpha + LPS-induced resident islet macrophage expression of IL-1beta and beta-cell expression of iNOS stimulated by the local release of IL-1 in rat islets. 15-d-delta 12,14-PGJ2 appears to be approximately 10-fold more effective at inhibiting resident islet macrophage activation (in response to TNF + LPS) than IL-1-induced nitrite production by beta-cells. Two mechanisms appear to be associated with the antiinflammatory actions of both 15-d-delta 12,14-PGJ2 and troglitazone: 1) the direct inhibition of cytokine- and endotoxin-stimulated iNOS and IL-1 transcription; and 2) the inhibition of IL-1 signaling, an event associated with PPAR-gamma agonist-induced activation of the heat shock response (as assayed by heat shock protein 70 expression). These findings indicate that the PPAR-gamma agonists, troglitazone and the J series of prostaglandins, are potent anti-inflammatory agents that prevent cytokine- and endotoxin-stimulated activation of peripheral and resident tissue macrophages and cytokine-induced iNOS expression by beta-cells by the inhibition of transcriptional activation and induction of the heat shock response.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Chromans/pharmacology , Prostaglandin D2/analogs & derivatives , Thiazoles/pharmacology , Thiazolidinediones , Animals , Cell Line , Cytokines/pharmacology , Hot Temperature , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/pharmacology , Interleukin-1/metabolism , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred Strains , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type II , Nitrites/metabolism , Prostaglandin D2/pharmacology , Rats , Rats, Sprague-Dawley , Shock/enzymology , Troglitazone
19.
Crit Care Med ; 27(2): 356-64, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10075061

ABSTRACT

OBJECTIVE: To examine the role of constitutive and inducible nitric oxide synthases (cNOS and iNOS) in platelet-activating factor (PAF)-induced shock and intestinal injury. DESIGN: Prospective, randomized, controlled experimental study. SETTING: Hospital research laboratory. SUBJECTS: Young adult male Sprague-Dawley rats were anesthetized and studied. INTERVENTIONS: Rats were injected with PAF, either alone or after the following pretreatments: a) selective iNOS inhibitors aminoguanidine or S-methylisothiourea; b) 3-morpholinosydnonimine, a NO donor; c) S-methylisothiourea + 3-morpholinosydnonimine; and d) antineutrophil antibody (to deplete neutrophils). MEASUREMENTS AND MAIN RESULTS: Blood pressure, hematocrit, white blood cell counts, intestinal injury, and intestinal cNOS and iNOS activities were assessed. We found that: a) cNOS is the predominant NOS in the intestine and its activity is inversely correlated to the level of tissue injury; b) there is a time-dependent increase in cNOS activity in sham-operated animals, which was abolished by PAF; c) Western blotting and immunohistochemistry showed iNOS present in the normal intestine, localizing mainly in crypt cells; d) iNOS inhibitors attenuated PAF-induced injury in animals with high cNOS activity, but had no protective effect in animals with low cNOS activity; e) 3-morpholinosydnonimine, alone or together with S-methylisothiourea, alleviated PAF-induced injury; and f) neutrophil depletion blocked the suppressive effect of PAF on cNOS and prevented injury. CONCLUSIONS: We conclude that cNOS and iNOS play different roles in PAF-induced intestinal injury. Caution should be exerted concerning potential therapeutic uses of iNOS inhibitors.


Subject(s)
Intestine, Small/drug effects , Intestine, Small/enzymology , Nitric Oxide Synthase/metabolism , Platelet Activating Factor/pharmacology , Animals , Drug Evaluation, Preclinical , Enzyme Inhibitors/therapeutic use , Intestine, Small/pathology , Male , Necrosis , Neutrophils/drug effects , Nitric Oxide Donors/therapeutic use , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/drug effects , Nitric Oxide Synthase Type I , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Prospective Studies , Random Allocation , Rats , Rats, Sprague-Dawley , Shock/chemically induced , Shock/drug therapy , Shock/enzymology , Shock/pathology
20.
Endocrinology ; 139(12): 5050-7, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9832444

ABSTRACT

In this study the effects of heat shock on interleukin-1beta (IL-1)-induced inhibition of islet metabolic function were examined. Treatment of rat islets for 18 h with IL-1 results in a potent inhibition of glucose-stimulated insulin secretion. The inhibitory effects of IL-1 on insulin secretion are completely prevented if islets are pretreated for 60 min at 42 C before cytokine stimulation. Heat shock also prevents IL-1-induced inhibition of insulinoma RINm5F cell mitochondrial aconitase activity. The protective effects of heat shock on islet metabolic function are associated with the inhibition of IL-1-stimulated inducible nitric oxide synthase (iNOS or NOS II) expression. Islets heat shocked for 60 min at 42 C fail to express iNOS (messenger RNA or protein) or produce nitrite in response to IL-1. IL-1-induced iNOS expression by rat islets requires activation of the transcriptional regulator nuclear factor kappaB (NF-kappaB). Heat shock prevents IL-1-induced NF-kappaB nuclear localization by inhibiting inhibitory protein kappaB (IkappaB) degradation in rat islets. Similar to rat islets, heat shock (stimulated by 90 min incubation at 42 C) prevents IL-1 + interferon gamma-induced iNOS expression and NF-kappaB nuclear localization in human islets. IL-1 also stimulates heat-shock protein 70 (hsp 70) expression by rat islets, and hsp 70 expression is dependent on islet production of nitric oxide. Last, evidence is presented that implicates nitric oxide as a stimulus for the expression of proteins that participate in islet recovery from nitric oxide-mediated damage. These studies indicate that heat shock prevents cytokine-induced islet damage by inhibiting iNOS expression, and suggest that nitric oxide is one effector molecule that stimulates the expression of factors involved in beta-cell recovery from nitric oxide-mediated damage.


Subject(s)
Cytokines/pharmacology , Hot Temperature , Islets of Langerhans/enzymology , Nitric Oxide Synthase/antagonists & inhibitors , Shock/enzymology , Aconitate Hydratase/metabolism , Animals , DNA-Binding Proteins/metabolism , HSP70 Heat-Shock Proteins/metabolism , Humans , Hydrazines/pharmacology , I-kappa B Proteins , Insulin/metabolism , Insulin Secretion , Interleukin-1/pharmacology , Islets of Langerhans/metabolism , Male , Mitochondria/enzymology , NF-kappa B/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Nitrogen Oxides , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...