Publications by Year: 1999

1999
 The biology of leukocyte-mediated proteolysis
Ginsburg I.  The biology of leukocyte-mediated proteolysis. Journal of Leukocyte Biology. 1999;66 (6) :1057.Abstract
Comment on The cell biology of leukocyte-mediated proteolysis. [J Leukoc Biol. 1999]
Signal-averaged P-wave duration does not predict atrial fibrillation after thoracic surgery
Amar D, Roistacher N, Zhang H, Baum MS, Ginsburg I, Steinberg JS. Signal-averaged P-wave duration does not predict atrial fibrillation after thoracic surgery. Anesthesiology. 1999;91 (1) :16-23.Abstract
BACKGROUND: Atrial fibrillation (AF) is the most common dysrhythmia seen early after major thoracic surgery but occurs infrequently after minor thoracic or other operations. A prolonged signal-averaged P-wave duration (SAPWD) has been shown to be an independent predictor of AF after cardiac surgery. The authors sought to determine whether a prolonged SAPWD alone or in combination with clinical or echocardiographic correlates predicts AF after elective noncardiac thoracic surgery. METHODS: Of the 250 patients enrolled, 228 were included in the final analysis. Preoperative SAPWD was obtained in 155 patients who had major thoracic surgery and in 73 patients undergoing minor thoracic or other operations who served as comparison control subjects. The SAPWD was recorded from three orthogonal leads using a sinus P-wave template. The filtered vector composite was used to measure total P-wave duration. Clinical, surgical, and echocardiographic parameters were collected and patients followed for 30 days after surgery for the development of symptomatic AF. RESULTS: Symptomatic AF developed in 18 of 155 (12%) patients undergoing major thoracic surgery and in 1 of 73 (1%) patients having minor thoracic or abdominal surgery, most commonly 2 or 3 days after surgery. In comparison with similar patients undergoing major thoracic surgery without AF, those who developed AF were older (66+/-8 vs. 62+/-10 yr; P = 0.04) but did not differ in SAPWD (145+/-17 vs. 147+/-16, ms) in standard electrocardiographic P-wave duration (105+/-7 vs. 107+/-10 mns), incidence of left-ventricular hypertrophy on 12-lead electrocardiography, male sex, history of hypertension, diabetes, or coronary heart disease. Thoracic-surgery patients at risk for postoperative AF did not differ from all other patients at low risk for AF in clinical or SAPWD parameters. CONCLUSIONS: Under the conditions of this study, SAPWD did not differentiate patients who did or did not develop AF after noncardiac thoracic surgery, and therefore its measurement cannot be recommended for the routine evaluation of these patients. Older age continues to be a risk factor for AF after thoracic surgery.
Hemolysis of human red blood cells induced by the combination of diethyldithiocarbamate (DDC) and divalent metals: Modulation by anaerobiosis, certain antioxidants and oxidants
Ginsburg I, Sadovnic M, Varani J, Tirosh O, Kohen R. Hemolysis of human red blood cells induced by the combination of diethyldithiocarbamate (DDC) and divalent metals: Modulation by anaerobiosis, certain antioxidants and oxidants. Free Radical Research. 1999;31 (2) :79-91.Abstract
The objective of the present communication is to describe the role played by combinations between diethydithiocarbamate (DDC) and divalent metals in hemolysis of human RBC. RBC which had been treated with DDC (10-50 microM) were moderately hemolyzed (about 50%) upon the addition of subtoxic amounts of Cu2+ (50 microM). However, a much stronger and a faster hemolysis occurred either if mixtures of RBC-DDC were immediately treated either by Co2+ (50 microM) or by a premixture of Cu2+ and Co2+ (Cu:Co) (50 microM). While Fe2+ and Ni2+, at 50 microM, initiated 30-50% hemolysis when combined with DDC (50 microM), on a molar basis, Cd2+ was at least 50 fold more efficient than any of the other metals in the initiation of hemolysis by DDC. On the other hand, neither Mn2+ nor Zn2+, had any hemolysis-initiating effects. Co2+ was the only metal which totally blocked hemolysis if added to DDC prior to the addition of the other metals. Hemolysis by mixtures of DDC + (Cu:Co) was strongly inhibited by anaerobiosis (flushing with nitrogen gas), by the reducing agents glutathione, N-acetyl cysteine, mercaptosuccinate, ascorbate, TEMPO, and alpha-tocopherol, by the PLA2 inhibitorbromophenacylbromide (BrPACBr), by tetracycline as well as by phosphatidyl choline, cholesterol and by trypan blue. However, TEMPO, BrPACBr and PC were the only agents which inhibited hemolysis induced by DDC: Cd2+ complexes. On the other hand, none of the classical scavengers of reactive oxygen species (ROS) employed e.g dimethylthiourea, catalase, histidine, mannitol, sodium benzoate, nor the metal chelators desferal and phenanthroline, had any appreciable inhibitory effects on hemolysis induced by DDC + (Cu:Co). DDC oxidized by H2O2 lost its capacity to act in concert either with Cu2+ or with Cd2+ to hemolyze RBC. While either heating RBC to temperatures greater than 37 degrees C or exposure of the cells to glucose-oxidase-generated peroxide diminished their susceptibility to hemolysis, exposure to the peroxyl radical from AAPH, enhanced hemolysis by DDC + (Cu:Co). The cyclovoltammetry patterns of DDC were drastically changed either by Cu2+, Co2+ or by Cd2+ suggesting a strong interaction of the metals with DDC. Also, while the absorbance spectrum of DDC at 280 nm was decreased by 50% either by Co2+, Cd2+ or by H2O2, a 90% reduction in absorbance occurred if DDC + H2O2 mixtures were treated either by Cu2+ or by Co2+, but not by Cd2+. Taken together, it is suggested that DDC-metal chelates can induce hemolysis by affecting the stability and the integrity of the RBC membrane, and possibly also of the cytoskeleton and the role played by reducing agents as inhibitors might be related to their ability to deplete oxygen which is also supported by the inhibitory effects of anaeobiosis.
Novel anthraquinone derivatives with redox-active functional groups capable of producing free radicals by metabolism: are free radicals essential for cytotoxicity?
Barasch D, Zipori O, Ringel I, Ginsburg I, Samuni A, Katzhendler J. Novel anthraquinone derivatives with redox-active functional groups capable of producing free radicals by metabolism: are free radicals essential for cytotoxicity?. European Journal of Medicinal Chemistry. 1999;34 (7-8) :597-615.Abstract
The mode of action of antitumour anthraquinone derivatives (i.e. mitoxantrone) is not clearly established yet. It includes, among others, intercalation and binding to DNA, bioreduction and aerobic redox cycling. A series of anthraquinone derivatives, with potentially bioreducible groups sited in the side chain, have been synthesized and biologically evaluated. Their redox and cytotoxic activities were screened. Derivatives which bear a 2-(dimethylamino)ethylamino substituent, known to confer high DNA affinity, demonstrated cytotoxicity but not redox activity (beside the anthraquinone reduction). Conversely, derivatives which showed redox activity were not cytotoxic toward the P388 cell line. The results suggest that bioreduction is not the main mode of action in the cytotoxicity of anthraquinones.
Antibacterial synergistic effect of chlorhexidine and hydrogen peroxide against Streptococcus sobrinus, Streptococcus faecalis and Staphylococcus aureus
Steinberg D, Heling I, Daniel I, Ginsburg I. Antibacterial synergistic effect of chlorhexidine and hydrogen peroxide against Streptococcus sobrinus, Streptococcus faecalis and Staphylococcus aureus. Journal of Oral Rehabilitation. 1999;26 (2) :151-156.Abstract
Chlorhexidine (CHX) and Hydrogen peroxide (HP) are potent antibacterial agents that are used in controlling dental plaque. However, both agents bear undesired side-effects. We have tested the hypothesis that an antibacterial synergistic effect can occur between the two agents against Streptococcus sobrinus, Streptococcus faecalis and Staphylococcus aureus. We have found that at several combinations of HP and CHX an antibacterial synergistic effect does occur, while at other combinations a on-significant synergism was noticed. No antagonism between the two agents was found in our experimental system. It can be postulated that the mechanism of this synergistic effect is via alteration of the bacterial cell-surface by CHX thereby allowing for an increased amount of HP to penetrate and to react with the intercellular organelles of the bacteria. These results suggest that CHX and HP can be of use in controlling the dental plaque in the oral cavity.
PADMA-28, a traditional tibetan herbal preparation inhibits the respiratory burst in human neutrophils, the killing of epithelial cells by mixtures of oxidants and pro-inflammatory agonists and peroxidation of lipids
Ginsburg I, Sadovnik M, Sallon S, Milo-Goldzweig I, Mechoulam R, Breuer A, Gibbs D, Varani J, Roberts S, Cleator E, et al. PADMA-28, a traditional tibetan herbal preparation inhibits the respiratory burst in human neutrophils, the killing of epithelial cells by mixtures of oxidants and pro-inflammatory agonists and peroxidation of lipids. Inflammopharmacology. 1999;7 (1) :47-62.Abstract
Both aqueous and methanolic fractions derived from the Tibetan preparation PADMA-28 (a mixture of 22 plants) used as an anti-atherosclerotic agent, and which is non-cytolytic to a variety of mammalian cells, were found to strongly inhibit (1) the killing of epithelial cells in culture induced by 'cocktails' comprising oxidants, membrane perforating agents and proteinases; (2) the generation of luminol-dependent chemiluminescence in human neutrophils stimulated by opsonized bacteria; (3) the peroxidation of intralipid (a preparation rich in phopholipids) induced in the presence of copper; and (4) the activity of neutrophil elastase. It is proposed that PADMA-28 might prove beneficial for the prevention of cell damage induced by synergism among pro-inflammatory agonists which is central in the initiation of tissue destruction in inflammatory and infectious conditions.
Multi-drug strategies are necessary to inhibit the synergistic mechanism causing tissue damage and organ failure in post infectious sequelae
Ginsburg I. Multi-drug strategies are necessary to inhibit the synergistic mechanism causing tissue damage and organ failure in post infectious sequelae. Inflammopharmacology. 1999;7 (3) :207-217.Abstract
The paper discusses the principal evidence that supports the concept that cell and tissue injury in infectious and post-infectious and inflammatory sequelae might involve a deleterious synergistic interaction among microbial- and host-derived pro-inflammatory agonists. Experimental models had proposed that a rapid cell and tissue injury might be induced by combinations among subtoxic amounts of three major groups of agonists generated both by microorganisms and by the host's own defense systems. These include: (1) oxidants: Superoxide, H(2)O(2), OH', oxidants generated by xanthine-xanthine-oxidase, ROO; HOC1, NO, OONO'-, (2) the membrane-injuring and perforating agents, microbial hemolysins, phospholipases A(2) and C, lysophosphatides, bactericidal cationic proteins, fatty acids, bile salts and the attack complex of complement a, certain xenobics and (3) the highly cationic proteinases, elastase and cathepsin G, as well as collagenase, plasmin, trypsin and a variety of microbial proteinases. Cell killing by combinations among the various agonists also results in the release of membrane-associated arachidonate and metabolites. Cell damage might be further enhanced by certain cytokines either acting directly on targets or through their capacity to prime phagocytes to generate excessive amounts of oxidants. The microbial cell wall components, lipoteichoic acid (LTA), lipopolysaccharides (LPS) and peptidoglycan (PPG), released following bacteriolysis, induced either by cationic proteins from neutrophils and eosinophils or by beta lactam antibiotics, are potent activators of macrophages which can release oxidants, cytolytic cytokines and NO. The microbial cell wall components can also activate the cascades of coagulation, complement and fibrinolysis. All these cascades might further synergize with microbial toxins and metabolites and with phagocyte-derived agonsits to amplify tissue damage and to induce septic shock, multiple organ failure, 'flesh-eating' syndromes, etc. The long persistence of non-biodegradable bacterial cell wall components within activated macrophages in granulomatous inflammation might be the result of the inactivation by oxidants and proteinases of bacterial autolytic wall enzymes (muramidases). The unsuccessful attempts in recent clinical trials to prevent septic shock by the administration of single antagonists is disconcerting. It does suggest however that, since tissue damage in post-infectious syndromes is most probably the end result of synergistic interactions among a multiplicity of agents, only agents which might depress bacteriolysis in vivo and 'cocktails' of appropriate antagonists, but not single antagonists, if administered at the early phases of infection especially to patients at high risk, might help to control the development of post-infectious syndromes. However, the use of adequate predictive markers for sepsis and other post-infectious complications is highly desirable. Although it is conceivable that anti-inflammatory strategies might also be counter-productive as they might act as 'double-edge swords', intensive investigations to devise combination therapies are warranted. The present review also lists the major anti-inflammatory agents and strategies and combinations among them which have been proposed in the last few years for clinical treatments of sepsis and other post-infectious complications.
Is streptolysin S of group A streptococci a virulence factor?
Ginsburg I. Is streptolysin S of group A streptococci a virulence factor?. APMIS. 1999;107 (12) :1051-1059.Abstract
The possible role played by streptolysin S (SLS) of group A streptococci in the pathophysiology of streptococcal infections and in post-streptococcal sequelae is discussed. The following properties of SLS justify its definition as a distinct virulence factor: 1) its presence on the streptococcus surface in a cell-bound form, 2) its continuous and prolonged synthesis by resting streptococci, 3) its non-immunogenicity, 4) its extractability by serum proteins (albumin, alpha lipoprotein), 5) its ability to become transferred directly to target cells while being protected from inhibitory agents in the milieu of inflammation, 6) its ability to bore holes in the membrane phospholipids in a large variety of mammalian cells, 7) its ability to synergize with oxidants, proteolytic enzymes, and with additional host-derived proinflammatory agonists, and 8) its absence in streptococcal mutants associated with a lower pathogenicity for animals. Because tissue damage in streptococcal and post-streptococcal sequelae might be the end result of a distinct synergism between streptococcal and host-derived proinflammatory agonists it is proposed that only cocktails of anti-inflammatory agents including distinct inhibitors of SLS (phospholipids), gamma globulin, inhibitors of reactive oxygen species, proteinases, cationic proteins cytokines etc., will be effective in inhibiting the multiple synergistic interactions which lead to fasciitis, myositis and the flesh-eating syndromes, and often develop into sepsis, septic shock and multiple organ failure. The creation of mutants deficient in SLS and in proteases will help shed light on the specific role played by SLS in the virulence of group A hemolytic streptococci.
"Cross-talk" among a multiplicity of pro-inflammatory agents: main cause of tissue damage in pulmonary inflammation?
Ginsburg I. "Cross-talk" among a multiplicity of pro-inflammatory agents: main cause of tissue damage in pulmonary inflammation?. European Respiratory Journal. 1999;14 (2) :481-482.Abstract
Comment on Proteolytic enzymes and airway diseases. [Eur Respir J. 1998] Neutrophil serine proteinases and defensins in chronic obstructive pulmonary disease: effects on pulmonary epithelium. [Eur Respir J. 1998] To the Editor: I have recently read with much interest two excellent reports in the European Respiratory Journal which discussed the role of neutrophil proteinases and defensins in chronic obstructive pulmonary disease [1] and in airway diseases [2]. Reading through these articles, it was surprising not to find any considerations of a major aspect related to the elucidation of the possible mechanisms of tissue damage in the lungs during inflammation. I refer to extensive studies from several laboratories which had proposed that tissue damage in inflammatory and infectious processes may primarily be the result of a synergistic "cross talk" among a multiplicity of pro-inflammatory agents (a multi-component system) [3, 4]. A series of publications [5±14] have shown that a severe and rapid membrane injury (necrosis) could be initiated in mammalian cells by a synergism among subtoxic concen- trations of three major groups of agonists. These included a) oxidants (H2O2, peroxyl radical, oxidants generated by xanthine-xanthine-oxidase, NO, HOCl, OONO-), b) mem- brane -perforating agents (microbial haemolysins/phospho- lipases A2 and C, lysophosphatides, free fatty acids, cationic proteins, histone [9] and defensins [5], and c) highly cationic proteolytic enzymes, (elastase, cathepsin G) [3, 4, 12]. These synergistic cytotoxic effects can be further amplified by certain cytokines. Furthermore, combinations of oxidants and elastase have also been shown to synergize to cause severe lung damage in animal models [6±10]. It has also been proposed that a deleterious synergism among microbial and host-derived pro-inflammatory agonists may frequently contribute to tissue injury in many infectious and post- infection complications [3, 4]. A notable example is, sepsis and the "flesh-eating" syndrome caused by highly toxigenic and invasive bacteria. Other studies had also shown that subtoxic amounts of the membrane-active xenobiotics, ethanol, methanol, n-butanol and the pesticide linden [13], could also synergize with subtoxic concentrations of peroxide, proteinases and cationic agents to amplify the damage to endothelial cells in culture. The results with the xenobiotics are of especial interest and concern to pulmonologists as these volatile agents may be inhaled and might then synergize with oxidants, proteinases and cationic proteins released either by accumulating neu- trophils or by activated lung macrophages to cause damage to both epithelial and endothelial cells. It has also been documented that ˜-lactam antibiotics and a large variety of cationic agents including, elastase, cath- epsin G, defensins, lysozyme, myeloperoxidase, spermine, spermidine, histones, polymyxin B and chlorhexidine are all capable of activating the autolytic wall enzymes (murami- dases) in bacteria leading to bacteriolysis [14]. Bacteriolysis at least in Gram-positive bacteria induced either by ˜- lactams or by cationic agents can, however, be strongly inhibited by sulphated polyanions presumably by inactivat- ing the autolytic wall enzymes responsible for breaking down the rigid cell wall. It is accepted that the massive release widely of bacterial wall components (lipopolysac- charide, lipoteichoic acid (LTA), peptidoglycan), in vivo, can activate macrophages to release cytotoxic cytokines, NO and also to activate the complement and coagulation cascades leading to sepsis, systemic inflammatory response syndrome (SIRS), multiple organ disfunction syndrome (MODS) and multiple organ failure (MOF) [15]. Today there are controversial opinions and hot debates regarding the approaches to treat sepsis, adult respiratory distress syndrome (ARDS) and additional post-infectious and inflammatory sequelae [15]. Unfortunately, the exclu- sive use of single antagonists to treat these syndromes has yielded poor results. Such failures may principally be due to, a) the lack of adequate and rapid tests to predict the onset of such complications so that treatment of patients usually starts too late, and b) a lack of sufficient awareness that fighting the deleterious effects caused by synergistic cytotoxic mechan- isms necessitates the use not of single antagonists but of cocktails comprised of a multiplicity of anti-inflammatory agents. Hopefully, a wider recognition of synergism concept of cellular injury [3, 4, 11±13] might offer a new and more realistic approach to this complex and still unsolved clinical problem. I. Ginsburg Dept of Oral Biology, Hebrew University - Hadassah Faculty of Dental Medicine, Jerusalem, Israel. Fax: 972 26758583.
Can we learn from the pathogenetic strategies of group A hemolytic streptococci how tissues are injured and organs fail in post-infectious and inflammatory sequelae?
Ginsburg I, Ward PA, Varani J. Can we learn from the pathogenetic strategies of group A hemolytic streptococci how tissues are injured and organs fail in post-infectious and inflammatory sequelae?. FEMS Immunology & Medical Microbiology. 1999;25 (4) :325-338.Abstract
The purpose of this review-hypothesis is to discuss the literature which had proposed the concept that the mechanisms by which infectious and inflammatory processes induce cell and tissue injury, in vivo, might paradoxically involve a deleterious synergistic ‘cross-talk’, among microbial- and host-derived pro-inflammatory agonists. This argument is based on studies of the mechanisms of tissue damage caused by catalase-negative group A hemolytic streptococci and also on a large body of evidence describing synergistic interactions among a multiplicity of agonists leading to cell and tissue damage in inflammatory and infectious processes. A very rapid cell damage (necrosis), accompanied by the release of large amounts of arachidonic acid and metabolites, could be induced when subtoxic amounts of oxidants (superoxide, oxidants generated by xanthine-xanthine oxidase, HOCl, NO), synergized with subtoxic amounts of a large series of membrane-perforating agents (streptococcal and other bacterial-derived hemolysins, phospholipases A2 and C, lysophosphatides, cationic proteins, fatty acids, xenobiotics, the attack complex of complement and certain cytokines). Subtoxic amounts of proteinases (elastase, cathepsin G, plasmin, trypsin) very dramatically further enhanced cell damage induced by combinations between oxidants and the membrane perforators. Thus, irrespective of the source of agonists, whether derived from microorganisms or from the hosts, a triad comprised of an oxidant, a membrane perforator, and a proteinase constitutes a potent cytolytic cocktail the activity of which may be further enhanced by certain cytokines. The role played by non-biodegradable microbial cell wall components (lipopolysaccharide, lipoteichoic acid, peptidoglycan) released following polycation- and antibiotic-induced bacteriolysis in the activation of macrophages to release oxidants, cytolytic cytokines and NO is also discussed in relation to the pathophysiology of granulomatous inflammation and sepsis. The recent failures to prevent septic shock by the administration of only single antagonists is disconcerting. It suggests, however, that since tissue damage in post-infectious syndromes is caused by synergistic interactions among a multiplicity of agents, only cocktails of appropriate antagonists, if administered at the early phase of infection and to patients at high risk, might prevent the development of post-infectious syndromes.