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










Publication year range
1.
Article in English | MEDLINE | ID: mdl-23360294

ABSTRACT

We herein investigated the in vitro effect of hypoxanthine on the activities of antioxidant enzymes such as catalase (CAT), glutathione peroxidase (GSH-Px), and superoxide dismutase (SOD) in erythrocytes, as well as on thiobarbituric acid-reactive substances (TBA-RS), in the plasma of rats. Results showed that hypoxanthine, when added to the incubation medium, enhanced CAT (10.0 µM), GSH-Px and SOD (8.5 µM and 10.0 µM) activities in erythrocytes of 15-day-old rats, reduced CAT activity (10.0 µM) and enhanced GSH-Px activity (10.0 µM) in erythrocytes of 30-day-old rats, reduced CAT activity (10.0 µM) and enhanced GSH-Px activity (8.5 µM and 10.0 µM) in erythrocytes of 60-day-old rats, as compared to controls. In addition, hypoxanthine (10.0 µM) enhanced TBA-RS levels in the plasma of 30- and 60-day old rats. Furthermore, we also tested the influence of allopurinol, trolox, and ascorbic acid on the effects elicited by hypoxanthine on the antioxidant enzymes and TBA-RS. Allopurinol and/or administration of antioxidants prevented most alterations caused by hypoxanthine in the oxidative stress parameters evaluated. Findings suggest that hypoxanthine alters antioxidant defenses and induces lipid peroxidation in the blood of rats; however, in the presence of allopurinol and antioxidants, some of these alterations in oxidative stress caused are prevented. Data indicate that, in humans, antioxidant administration might serve as a potential adjuvant therapy for ameliorating the damage caused by hypoxanthine.


Subject(s)
Allopurinol/pharmacology , Ascorbic Acid/pharmacology , Erythrocytes/enzymology , Hypoxanthines/physiology , Oxidative Stress , Vitamin E/pharmacology , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Catalase/metabolism , Chromans/pharmacology , Erythrocytes/drug effects , Erythrocytes/metabolism , Glutathione Peroxidase/metabolism , Hypoxanthines/pharmacology , Malondialdehyde/metabolism , Rats , Rats, Wistar , Superoxide Dismutase/metabolism , Thiobarbituric Acid Reactive Substances/metabolism
2.
J Cell Physiol ; 162(1): 147-53, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7814447

ABSTRACT

Changes in intracellular Ca2+ homeostasis are thought to contribute to cell dysfunction in oxidative stress. The hypoxanthine-xanthine oxidase system (X-XO) mobilizes Ca2+ from intracellular stores and induces a marked rise in cytosolic calcium in different cell types. To identify the reactive O2 species involved in the disruption of calcium homeostasis by X-XO, we studied the effect of X-XO on [Ca2+]i by spectrofluorimetry with fura-2 in human umbilical vein endothelial cells (HUVEC). The [Ca2+]i response to X-XO was essentially diminished by superoxide dismutase (SOD) (200 U/ml) and catalase (CAT) (200 U/ml), which scavenge the superoxide anion, O2-, or H2O2, respectively. The [Ca2+]i increase stimulated by 10 nmol H2O2/ml/min, generated from the glucose-glucose oxidase system, or 10 microM H2O2, given as bolus, was about a third of that induced by X-XO (10 nmol O2-/ml/min) but was comparable to that induced by X-XO in the presence of SOD. The X-XO-stimulated [Ca2+]i increase was significantly reduced by 100 microM o-phenanthroline, which inhibits the iron-catalysed formation of the hydroxyl radical. On the other hand, the [Ca2+]i response to low dose X-XO (1 nmol O2-/ml/min) was markedly enhanced in the presence of 1 microM H2O2, which itself had no effect on [Ca2+]i. More than 50% of this synergistic effect was prevented by o-phenanthroline. These results indicate that the effect of X-XO on calcium homeostasis appears to result from an interaction of O2- and H2O2, which could be explained by the formation of the hydroxyl radical.


Subject(s)
Calcium/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Hydrogen Peroxide/pharmacology , Hydroxyl Radical/pharmacology , Superoxides/pharmacology , Calcium/analysis , Cells, Cultured , Endothelium, Vascular/chemistry , Homeostasis , Humans , Hypoxanthine , Hypoxanthines/metabolism , Hypoxanthines/pharmacology , Hypoxanthines/physiology , Oxidative Stress , Phenanthrolines/pharmacology , Superoxide Dismutase/pharmacology , Xanthine Oxidase/metabolism , Xanthine Oxidase/pharmacology , Xanthine Oxidase/physiology
3.
Ukr Biokhim Zh (1978) ; 66(2): 85-9, 1994.
Article in Ukrainian | MEDLINE | ID: mdl-7998346

ABSTRACT

The incorporation of [8-14C]hypoxanthine radioactive label into the nucleic acids of the mature thymocytes (that do not bind peanut agglutinin) of CBA mice and Wistar rats and the action of non-peptide thymic mitogenic factor (factor) on this process have been studied. It was shown, that the factor, shifting AMP catabolism towards hypoxanthine accumulation, accelerated by 50% the incorporation of hypoxanthine radioactive label into nucleic acids during 3-hour incubation. It is concluded that the factor being an activator of the hypoxanthine accumulation, abrogates the purine limits in thymocytes and activates the "salvage pathway" of purine synthesis which is the primary pathway in thymocytes. The factor is assumed to act as the mitogen on one of the stages of nucleic acids or nucleotides synthesis, but not to act on the guanine-hypoxanthine phosphoribosyl transferase. It was shown that the intrathymocyte ratio of hypoxanthine and factor concentrations (hypoxanthine/factor) is higher in rats than in mice. The dependence of proliferative activity on relative levels of both substances was revealed. The stimulation of cell proliferation by the system: factor--hypoxanthine is one of the mechanisms of thymus regeneration at the expense of mature thymocyte population.


Subject(s)
Hypoxanthines/physiology , Mitogens/physiology , Thymus Gland/cytology , Thymus Hormones/physiology , Animals , Cell Division/physiology , Hypoxanthine , Hypoxanthines/chemistry , Mice , Mice, Inbred CBA , Mitogens/chemistry , Rats , Rats, Wistar , Species Specificity , Thymus Hormones/chemistry
4.
Crit Care Med ; 21(9): 1376-86, 1993 Sep.
Article in English | MEDLINE | ID: mdl-8370303

ABSTRACT

OBJECTIVE: Review the histologic and pathophysiologic alterations seen after intestinal ischemia and reperfusion. DATA SOURCE: Current literature review. STUDY SELECTION: The most pertinent, current, and representative articles describing results from both animal and human investigations are utilized and discussed. DATA SYNTHESIS: Postischemic intestinal tissue damage appears to be due to the formation of oxygen radicals and the activation of phospholipase A2. The initial source of oxygen radicals seems to be the hypoxanthine-xanthine oxidase system. Oxygen radicals react directly with poly-unsaturated fatty acids, leading to lipid peroxidation within the cell membranes. Indirectly, the radicals trigger the accumulation of neutrophils within the affected tissue initiating inflammatory processes that lead to severe mucosal lesions. Similarly, phospholipase A2 also initiates postischemic mucosal lesions. Phospholipase A2 is a hydrolytic enzyme capable of increasing formation of cytotoxic lysophospholipids within the tissue. Enhanced activity of phospholipase A2 also stimulates the production of prostaglandins and leukotrienes. Various substances (superoxide dismutase, catalase, dimethyl sulfoxide, allopurinol, and deferoxamine, etc.) are able to detoxify oxygen radicals or inhibit the mechanisms leading to their enhanced generation, thus attenuating the postischemic lesions of the mucosa. CONCLUSIONS: Oxygen radicals and the activation of phospholipase A2 during reperfusion seem to be instrumental for the development of hemorrhagic mucosal lesions after intestinal ischemia. Radical scavengers and phospholipase A2 inhibitors may prevent reperfusion damage of the intestine, even when the treatment starts during ischemia but before reperfusion.


Subject(s)
Gastrointestinal Hemorrhage/metabolism , Gastrointestinal Hemorrhage/pathology , Intestines/blood supply , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Cats , Disease Models, Animal , Free Radical Scavengers , Free Radicals , Gangrene , Gastrointestinal Hemorrhage/drug therapy , Gastrointestinal Hemorrhage/etiology , Humans , Hypoxanthines/physiology , Inflammation , Leukotrienes/physiology , Lipid Peroxidation , Neutrophils/physiology , Phospholipases A/antagonists & inhibitors , Phospholipases A/physiology , Phospholipases A2 , Prostaglandins/physiology , Rats , Reactive Oxygen Species , Reperfusion Injury/drug therapy , Reperfusion Injury/etiology , Xanthine Oxidase/physiology
5.
Early Hum Dev ; 30(2): 171-7, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1493769

ABSTRACT

Hyperoxia has been suggested as a risk factor for kernicterus. The toxicity of hyperoxia may be mediated by free radicals. We investigated the effects of free radicals, formed by the hypoxanthine/xanthine oxidase system, with and without additional hyperoxia, on the accumulation of bilirubin and albumin in rat brain. Hypoxanthine was infused for 60 min into retrograde carotid catheters in awake, young, male SPRD rats. After 30 min the infusion was briefly interrupted to inject xanthine oxidase 1 U/kg through the same catheter. Group I (controls) received 0.9% NaCl in lieu of hypoxanthine/xanthine oxidase. Groups I and II breathed room air at all times, while group III breathed 90% O2. After 60 min all groups received a bolus dose of 125I-albumin through a peripheral venous catheter, followed by bilirubin 25 mg/kg for 5 min, then bilirubin 35 mg/kg for 55 min. There were no significant differences between the groups as regards serum bilirubin, serum albumin, brain bilirubin, or brain albumin. Neither during normoxic nor hyperoxic conditions did the hypoxanthine/xanthine oxidase system increase the accumulation of bilirubin or albumin in rat brain.


Subject(s)
Albumins/metabolism , Bilirubin/metabolism , Brain/metabolism , Hypoxanthines/physiology , Oxygen/poisoning , Xanthine Oxidase/physiology , Animals , Free Radicals , Hypoxanthine , Male , Oxygen/metabolism , Rats , Rats, Sprague-Dawley
6.
J Pharmacol Exp Ther ; 250(2): 470-5, 1989 Aug.
Article in English | MEDLINE | ID: mdl-2547932

ABSTRACT

The purpose of this study was to evaluate the possible involvement of xanthine and xanthine oxidase in reperfusion injury in a low-flow, reflow model of liver perfusion. Livers were perfused at flow rates around 25% of normal for 90 min and then at normal flow rates (4 ml/g/min) for 30 min. When flow was restored to normal, malondialdehyde and lactic dehydrogenase (LDH) were released into the effluent perfusate. Malondialdehyde production rapidly reached values of 300 nmol/g/hr whereas LDH increased from basal levels of 100 to 600 U/l upon reperfusion. Trypan blue was taken up exclusively in cells in pericentral regions of the liver lobule under these conditions. Xanthine and hypoxanthine in the effluent perfusate increased steadily during the low-flow period reaching values around 5 and 10 microM, respectively, and decreased rapidly after the flow was restored to normal. Perfusion with nitrogen-saturated buffer for 3 min before restoration of normal flow rates or infusion of the radical scavenger catechin (400 microM) reduced cell damage by about 50%. Infusion of allopurinol (2-6 mM), an inhibitor of xanthine oxidase, prevented reperfusion injury in a dose-dependent manner. Taken together, these data indicate that a reperfusion injury occurs in liver upon reintroduction of oxygen which is initiated by oxidation of xanthine and hypoxanthine via xanthine oxidase and ultimately leads to production of lipid peroxides. Surprisingly, infusion of xanthine (4 mM), substrate for xanthine oxidase, reduced hepatocellular injury on reperfusion. LDH release was decreased from values around 700 to less than 200 U/l and trypan blue uptake in pericentral region was prevented totally by xanthine.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Liver Circulation , Reperfusion Injury/etiology , Xanthine Oxidase/physiology , Xanthines/physiology , Animals , Female , Hypoxanthine , Hypoxanthines/physiology , L-Lactate Dehydrogenase/metabolism , Malondialdehyde/metabolism , Rats , Rats, Inbred Strains , Superoxides/metabolism , Uric Acid/metabolism , Uric Acid/pharmacology , Xanthine
7.
Gamete Res ; 23(3): 323-34, 1989 Jul.
Article in English | MEDLINE | ID: mdl-2476369

ABSTRACT

Hypoxanthine and adenosine are present in preparations of mouse ovarian follicular fluid, and these purines maintain mouse oocytes in meiotic arrest in vitro (Eppig et al.: Biology of Reproduction 33:1041-1049. 1985). The first hypothesis tested in this study is that purines which maintain meiotic arrest act by maintaining meiosis-arresting levels of cyclic adenosine monophosphate (cAMP) in the oocyte. Oocyte-cumulus cell complexes were incubated in control medium (no added purines), or medium containing 0.75 mM adenosine, 4 mM hypoxanthine, or both for 3 hr and the percentage of the oocytes that underwent germinal vesicle breakdown (GVB) and the cAMP content of the intact complexes and the oocytes were determined. Adenosine alone had little inhibitory effect on GVB at this time point but sustained higher levels of cAMP in the oocytes. Hypoxanthine maintained 80% of cumulus cell-enclosed oocytes in meiotic arrest and also sustained higher cAMP levels in the oocytes. The addition of adenosine to hypoxanthine-containing medium increased the percentage of oocytes maintained in meiotic arrest, and increased the amount of cAMP in the oocytes above that maintained by either hypoxanthine or adenosine alone. Neither hypoxanthine, adenosine, nor hypoxanthine plus adenosine altered the cAMP content of intact complexes when assayed after 3 hr culture. Microinjection of an inhibitor of the catalytic subunit of cAMP-dependent protein kinase induced GVB in denuded oocytes cultured in medium containing hypoxanthine. This purine, therefore, maintained meiotic arrest by sustaining elevated cAMP levels within the oocytes. The second hypothesis tested in this study is that purines maintain meiosis-arresting levels of cAMP, at least in part, by inhibiting cAMP phosphodiesterase activity. In descending order of potency, 3-isobutyl-1-methylxanthine (IBMX), guanosine, hypoxanthine, adenosine, and xanthosine inhibited cAMP phosphodiesterase in oocyte lysates. Moreover, like the potent phosphodiesterase inhibitor IBMX, hypoxanthine augmented the meiotic arrest and cAMP accumulation mediated by follicle-stimulating hormone (FSH) in intact complexes. Therefore, inhibition of oocyte phosphodiesterase appears to be one mechanism by which the purines could maintain meiosis-arresting levels of cAMP.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Cyclic AMP/metabolism , Oocytes/cytology , Purines/physiology , 1-Methyl-3-isobutylxanthine/pharmacology , 3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Adenosine/pharmacology , Adenosine/physiology , Animals , Cells, Cultured , Female , Follicle Stimulating Hormone/pharmacology , Hypoxanthine , Hypoxanthines/pharmacology , Hypoxanthines/physiology , Meiosis/drug effects , Mice , Mice, Inbred C57BL , Microinjections , Oocytes/drug effects , Oocytes/metabolism , Purines/pharmacology
8.
Circulation ; 78(5 Pt 2): III224-35, 1988 Nov.
Article in English | MEDLINE | ID: mdl-3180402

ABSTRACT

The aim of this study was to differentiate myocardial reperfusion injury from that of ischemia. We assessed the role of the myocardial adenosine 5'-triphosphate (ATP) catabolites, hypoxanthine and xanthine, generated during ischemia and the early phase of reperfusion, in reperfusion injury by modulating adenosine transport and metabolism with specific metabolic inhibitors. This was followed by intracoronary infusion of exogenous hypoxanthine and xanthine. Twenty-four dogs instrumented with minor-axis piezoelectric crystals and intraventricular pressure transducers were subjected to 30 minutes of normothermic global myocardial ischemia and 60 minutes of reperfusion. In Group 1 (n = 7), normal saline was infused into the cardiopulmonary bypass reservior before ischemia and before reperfusion. Saline solution containing 25 microM p-nitrobenzylthioinosine (NBMPR) and 100 microM erythro-9-(2-hydroxy-3-nonyl)adenine (EHNA) was infused in Group 2 (n = 10) dogs. Group 3 (n = 7) dogs were treated exactly like those in Group 2 except, at the end of the ischemic period and immediately before releasing the cross-clamp, a solution of EHNA-NBMPR containing 100 microM hypoxanthine and 100 microM xanthine was infused into the aortic root. Left ventricular performance and myocardial adenine nucleotide pool intermediates were determined before and after ischemia. ATP was depleted by about 50% (p less than 0.05 vs. preischemia) in all groups after 30 minutes of ischemia. Inosine was the major ATP catabolite (9.29 +/- 1.2 nmol/mg protein) in Group 1, while adenosine (9.91 +/- 0.7 nmol/mg protein) was the major metabolite in EHNA-NBMPR-treated dogs (Groups 2 and 3). Hypoxanthine levels were fivefold more in Group 1 compared with Groups 2 and 3 (p less than 0.05). Left ventricular performance in Group 1 decreased from 76.8 +/- 7.6 to 42.9 +/- 9.8 and 52.3 +/- 8.4 dynes/cm2 x 10(3) (p less than 0.05), while myocardial ATP decreased from 30.9 +/- 2.2 to 17.2 +/- 1.0 and 16.5 +/- 1.0 nmol/mg protein during 30 and 60 minutes of reperfusion, respectively (p less than 0.05 vs. preischemia). Ventricular function in Group 2 dogs completely recovered within 30 minutes of reperfusion, and myocardial ATP recovered to the preischemic level at 60 minutes of reperfusion. In Group 3, left ventricular performance was depressed by 39% and 30% during 30 and 60 minutes of reperfusion (p less than 0.05), respectively, and myocardial ATP did not recover during reperfusion despite a significant intramyocardial adenosine accumulation.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Hypoxanthines/physiology , Myocardial Reperfusion Injury/etiology , Myocardium/metabolism , Xanthines/physiology , Adenine/analogs & derivatives , Adenine/pharmacology , Adenine Nucleotides/metabolism , Animals , Coronary Disease/physiopathology , Coronary Disease/therapy , Dogs , Female , Free Radicals , Heart/physiopathology , Heart Ventricles , Hypoxanthine , Male , Myocardial Reperfusion , Thioinosine/analogs & derivatives , Thioinosine/pharmacology , Xanthine
9.
Endocr Res ; 13(4): 399-405, 1987.
Article in English | MEDLINE | ID: mdl-2834191

ABSTRACT

Different concentrations of cyclic adenosine 3',5'-pyrophosphate (cAPP), adenosine, hypoxanthine, 17 beta-estradiol and testosterone were tested to determine their effect on spontaneous germinal vesicle breakdown (GVBD) in mouse oocytes. Individually, these compounds at concentrations of 200 microM or less, had no or minimal inhibitory effect on the spontaneous GVBD of mouse oocytes. Dibutyryl 3',5' cyclic adenosine monophosphate (dbcAMP) at a concentration of 50 microM or higher blocked GVBD but had no effect at concentrations of 20 microM or less. With dbcAMP at a concentration of 20 microM, cAPP blocked GVBD at a concentration of 10 microM. To affect inhibition of GVBD with adenosine, 17 beta-estradiol and testosterone and with hypoxanthine, concentrations of 100-200 microM and 500 microM, respectively, were required. In conclusion cAPP is the most potent compound among those tested in blocking spontaneous GVBD of mouse oocytes when combined with dbcAMP.


Subject(s)
Cyclic AMP/physiology , Oocytes/physiology , Adenosine/physiology , Animals , Cells, Cultured , Estradiol/physiology , Female , Hypoxanthine , Hypoxanthines/physiology , Mice , Mice, Inbred ICR , Testosterone/physiology
10.
Ups J Med Sci ; 92(3): 265-75, 1987.
Article in English | MEDLINE | ID: mdl-3448801

ABSTRACT

The effects of intra-arterial and intravenous injections of adenosine and hypoxanthine were investigated with special reference to respiratory variables in anesthetized young cats. Studies were made of the effects on inspiratory activity (phrenic nerve activity), heart rate, blood pressure and central venous pressure. To assess the risk of accumulation of adenosine degeneration products after several injections measurements were also made of hypoxanthine, xanthine and urate in plasma at intervals after the injections. It was found that intra-arterial and intravenous injections of adenosine increased central inspiratory activity during the first few breaths after the injection. The blood pressure and heart rate decreased slightly and central venous pressure increased slightly after the injection. Degradation of adenosine and its metabolites takes place rapidly and it is therefore unlikely that metabolites influence the results. It is concluded that adenosine causes brief stimulation of inspiratory activity.


Subject(s)
Adenosine/pharmacology , Hypoxanthines/pharmacology , Respiration/drug effects , Adenosine/administration & dosage , Adenosine/blood , Adenosine/physiology , Animals , Cats , Female , Hydrogen-Ion Concentration , Hypoxanthine , Hypoxanthines/administration & dosage , Hypoxanthines/blood , Hypoxanthines/physiology , Injections, Intra-Arterial , Injections, Intravenous , Male , Phrenic Nerve/drug effects , Respiratory Function Tests
12.
Radiobiologiia ; 25(4): 521-4, 1985.
Article in Russian | MEDLINE | ID: mdl-3875877

ABSTRACT

Hypoxanthine in vitro causes death of thymocytes and concurrent intranucleosome degradation of chromatin. This process is more manifest in a more radiosensitive thymocyte fraction and prevented by protein synthesis inhibitors. The increase in the yield of hypoxanthine after the effect of lympholytic agents of different nature is not the result of cell death. It is assumed that hypoxanthine, formed in the exposed cells, may be an additional cytotoxic factor on reaching a subliminal concentration.


Subject(s)
Hypoxanthines/physiology , T-Lymphocytes/radiation effects , Animals , Cell Survival/drug effects , Cell Survival/radiation effects , Cesium Radioisotopes , Cycloheximide/pharmacology , Gamma Rays , Hypoxanthine , In Vitro Techniques , Mice , Puromycin/pharmacology , T-Lymphocytes/drug effects
14.
J Clin Invest ; 72(3): 965-70, 1983 Sep.
Article in English | MEDLINE | ID: mdl-6193145

ABSTRACT

In previous studies from this laboratory, human bone marrow hypoxanthine concentrations were found to average 7.1 microM, three times higher than plasma hypoxanthine concentrations measured simultaneously. To assess the significance of this finding, the relationship between hypoxanthine concentration and the rate of purine nucleotide synthesis by the de novo pathway was studied in normal human bone marrow mononuclear cells and in the human promyelocytic cell line, HL-60, in vitro. Utilizing a [14C]formate incorporation technique, rates of total cellular de novo purine synthesis as well as rates of de novo adenine, de novo guanine, and thymine synthesis and incorporation into RNA and DNA were measured as a function of hypoxanthine concentration. In normal human marrow cells, the rate of total de novo purine synthesis declined by 81%, while the rate of de novo adenine and de novo guanine synthesis and incorporation into macromolecules declined by 89 and 75%, respectively, when media hypoxanthine was increased from 0 to 10 microM. Similar results were seen in the HL-60 cell line. In contrast, rates of thymine synthesis and incorporation into DNA as well as overall rates of RNA and DNA synthesis did not change with varying media hypoxanthine concentrations. In addition, hypoxanthine salvage and incorporation into RNA and DNA was shown to progressively increase with increasing media hypoxanthine concentrations. These results indicate that physiologic concentrations of hypoxanthine are sufficient to regulate the rate of de novo purine synthesis in human bone marrow in vivo.


Subject(s)
Bone Marrow/metabolism , Hypoxanthines/physiology , Leukocytes/metabolism , Purines/biosynthesis , Adenine/metabolism , Bone Marrow Cells , DNA/biosynthesis , Guanine/metabolism , Humans , Hypoxanthine , Hypoxanthines/metabolism , Leukemia, Myeloid, Acute/metabolism , Purines/metabolism , RNA/biosynthesis , Thymine/metabolism
15.
J Bacteriol ; 153(2): 837-45, 1983 Feb.
Article in English | MEDLINE | ID: mdl-6401706

ABSTRACT

Data are presented which indicate that the repression of pur gene expression seen after the addition of preformed purines to cultures of Salmonella typhimurium is the consequence of the presence or the formation of the purine bases, hypoxanthine and guanine. This conclusion is based on the following observations. First, it was impossible to find a correlation between the size of any individual purine nucleotide pool and the level of the first four enzymes in the de novo biosynthetic pathway. Second, adenine plus guanosine served as a perfect source of purine nucleotides, but their presence caused no repression of pur gene expression if the cells lacked purine nucleoside phosphorylase activity. This enzyme is needed to convert adenine and guanosine to hypoxanthine and guanine, but not for their conversion to nucleotides. Third, addition of guanine to a strain lacking guanine phosphoribosyltransferase (gpt) resulted in a repression of the level of the purine de novo biosynthetic enzymes, a reduction of the growth rate, and a fall in the pools of ATP and GTP. Addition of hypoxanthine to a strain lacking hypoxanthine phosphoribosyltransferase (hpt) had a similar, although weaker, effect. If the cells lacked both hypoxanthine and guanine phosphoribosyltransferases (hpt gpt), their basal level of the purine de novo biosynthetic enzymes was repressed in minimal medium. Such cells grow slower than wild-type cells and excrete purines, probably due to the inability to salvage endogenously formed hypoxanthine and guanine.


Subject(s)
Gene Expression Regulation , Guanine/physiology , Hypoxanthines/physiology , Purines/biosynthesis , Salmonella typhimurium/genetics , Adenosine Triphosphate/metabolism , Amidophosphoribosyltransferase/metabolism , Guanosine Triphosphate/metabolism , Hypoxanthine Phosphoribosyltransferase/metabolism , Mutation , Pentosyltransferases/metabolism , Salmonella typhimurium/enzymology , Salmonella typhimurium/metabolism
16.
Transfusion ; 21(2): 138-49, 1981.
Article in English | MEDLINE | ID: mdl-7222198

ABSTRACT

Red blood cells were stored at 4 C in the primary bag with an integrally attached empty transfer pack so that the red blood cells could be rejuvenated or not, as desired before glycerolization and freezing. The rejuvenation and glycerol solutions were added through ports in the system. After glycerolization, the red blood cells were concentrated by centrifugation to remove the supernatant glycerol before freezing with 40% w/v glycerol in the primary polyvinylchloride (PVC) plastic container at -80 C. After thawing, the red blood cells were washed using either the Haemonetics Blood Processor 115 or the IBM Blood Processor 2991-1 or 2991-2. In each system, 50 ml of 12% sodium chloride and 1.5 to 1.6 liters of 0.9% sodium chloride-0.2% glucose-25 meq/l disodium phosphate were used. Recovery of red blood cells in vitro was 91 per cent. After three days of postwash storage at 4 C, nonrejuvenated red blood cells had a mean 24-hour posttransfusion survival of 88 per cent, and outdated-rejuvenated red blood cells a value of 81 per cent. This new system is simpler and safer than methods previously used in this laboratory, and red blood cell recovery and 24-hour posttransfusion survivals were comparable or better.


Subject(s)
Blood Specimen Collection/methods , Erythrocytes/physiology , Polyvinyl Chloride , Polyvinyls , Blood Bactericidal Activity , Blood Preservation , Freezing , Glycerol/pharmacology , Humans , Hypoxanthines/physiology , Leukocytes , Rejuvenation , Serum Albumin/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...