Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters











Publication year range
1.
Mol Microbiol ; 41(6): 1237-47, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11580831

ABSTRACT

The Rho-GTPases-activating toxin CNF1 (cytotoxic necrotizing factor 1) delivers its catalytic activity into the cytosol of eukaryotic cells by a low pH membrane translocation mechanism reminiscent of that used by diphtheria toxin (DT). As DT, CNF1 exhibits a translocation domain (T) containing two predicted hydrophobic helices (H1-2) (aa 350-412) separated by a short peptidic loop (CNF1-TL) (aa 373-386) with acidic residues. In the DT loop, the loss of charge of acidic amino acids, as a result of protonation at low pH, is a critical step in the transfer of the DT catalytic activity into the cytosol. To determine whether the CNF1 T domain operates similarly to the DT T domain, we mutated several ionizable amino acids of CNF1-TL to lysine. Single substitutions such as D373K or D379K strongly decreased the cytotoxic effect of CNF1 on HEp-2 cells, whereas the double substitution D373K/D379K induced a nearly complete loss of cytotoxic activity. These single or double substitutions did not modify the cell-binding, enzymatic or endocytic activities of the mutant toxins. Unlike the wild-type toxin, single- or double-substituted CNF1 molecules bound to the HEp-2 plasma membrane could not translocate their enzymatic activity directly into the cytosol following a low pH pulse.


Subject(s)
Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Cytotoxins/chemistry , Cytotoxins/genetics , Escherichia coli Proteins , Amino Acid Substitution , Bacterial Toxins/metabolism , Base Sequence , Biological Transport, Active , Cell Line , Cell Membrane/metabolism , Cytotoxins/metabolism , DNA Primers/genetics , Endocytosis , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli/pathogenicity , Humans , Hydrogen-Ion Concentration , Point Mutation , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
2.
Cardiovasc Drugs Ther ; 15(6): 479-86, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11916356

ABSTRACT

We have shown that chronic treatment with carteolol, a non-selective beta-adrenergic receptor antagonist, improved left ventricular (LV) function and survival in an avian model of dilated cardiomyopathy (DCM). The aim of the present study was to compare ex vivo heart function with and without beta-agonist and antagonist challenge. We investigated whether intracoronary infusion of a beta-blocker, carteolol or beta-agonist, isoproterenol decreased contractility. In the DCM group, isoproterenol resulted in a significantly greater increase in heart rate (71% vs. 28% compared to control hearts). To investigate the mechanism for the increase in heart rate, we exposed spontaneously beating neonatal cardiomyocytes to serum immunoglobulin (IgG) isolated from DCM animals. Serum IgG resulted in a significant increase in spontaneous beating rate in neonatal rat cardiomyocytes that was blocked by pre-treatment with a beta-blocker. Carteolol challenge did not significantly change heart rate but did significantly increase LV peak pressure in DCM hearts (62%) while coronary artery flow remained unchanged (2.7+/-0.1 vs 2.7+/-0.5 ml/min/g). These results show that 1) beta-receptor stimulation results in a greater tachycardic response in DCM animals, and 2) carteolol challenge improves myocardial contractility in hearts from DCM animals independent of heart rate or changes in coronary artery flow.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Heart Failure/drug therapy , Heart/drug effects , Adrenergic beta-Agonists/therapeutic use , Adrenergic beta-Antagonists/therapeutic use , Animals , Animals, Newborn , Blood Pressure/drug effects , Cardiomyopathy, Dilated/drug therapy , Cardiomyopathy, Dilated/physiopathology , Carteolol/pharmacology , Carteolol/therapeutic use , Cell Size/drug effects , Cells, Cultured , Disease Models, Animal , Heart/physiopathology , Heart Failure/physiopathology , Heart Rate/drug effects , Immunoglobulin G/metabolism , Immunoglobulin G/pharmacology , Isoproterenol/pharmacology , Isoproterenol/therapeutic use , Myocardium/cytology , Myocardium/metabolism , Rats , Turkeys , Ventricular Function, Left/drug effects
3.
EMBO J ; 19(23): 6361-70, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11101509

ABSTRACT

The pathogenic bacterium Helicobacter pylori produces the cytotoxin VacA, which is implicated in the genesis of gastric epithelial lesions. By transfect ing HEp-2 cells with DNAs encoding either the N-terminal (p34) or the C-terminal (p58) fragment of VacA, p34 was found localized specifically to mitochondria, whereas p58 was cytosolic. Incubated in vitro with purified mitochondria, VacA and p34 but not p58 translocated into the mitochondria. Microinjection of DNAs encoding VacA-GFP and p34-GFP, but not GFP-VacA or GFP-p34, induced cell death by apoptosis. Transient transfection of HeLa cells with p34-GFP or VacA-GFP induced the release of cytochrome c from mitochondria and activated the executioner caspase 3, as determined by the cleavage of poly(ADP-ribose) polymerase (PARP). PARP cleavage was antagonized specifically by co-transfection of DNA encoding Bcl-2, known to block mitochondria-dependent apoptotic signals. The relevance of these observations to the in vivo mechanism of VacA action was supported by the fact that purified activated VacA applied externally to cells induced cytochrome c release into the cytosol.


Subject(s)
Bacterial Proteins/metabolism , Cytochrome c Group/metabolism , Mitochondria/metabolism , Animals , Apoptosis , Caspase 3 , Caspases/metabolism , Cell Line , Cell Nucleus/metabolism , Cytosol/metabolism , Digitonin/pharmacology , Electrophoresis, Polyacrylamide Gel , Fluorescent Antibody Technique , Green Fluorescent Proteins , HeLa Cells , Humans , Immunohistochemistry , Luminescent Proteins/metabolism , Microscopy, Electron , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rabbits , Reticulocytes/metabolism , Stomach Diseases/metabolism , Transfection
4.
Mol Biol Cell ; 11(11): 3897-909, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11071915

ABSTRACT

Helicobacter pylori vacuolating toxin (VacA) causes vacuolation in a variety of cultured cell lines, sensitivity to VacA differing greatly, however, among the different cell types. We found that the high sensitivity of HEp-2 cells to VacA was impaired by treating the cells with phosphatidylinositol-specific phospholipase C (PI-PLC) which removes glycosylphosphatidylinositol (GPI)-anchored proteins from the cell surface. Incubation of cells with a cholesterol-sequestering agent, that impairs both structure and function of sphingolipid-cholesterol-rich membrane microdomains ("lipid rafts"), also impaired VacA-induced cell vacuolation. Overexpression into HEp-2 cells of proteins inhibiting clathrin-dependent endocytosis (i.e., a dominant-negative mutant of Eps15, the five tandem Src-homology-3 domains of intersectin, and the K44A dominant-negative mutant of dynamin II) did not affect vacuolation induced by VacA. Nevertheless, F-actin depolymerization, known to block the different types of endocytic mechanisms, strongly impaired VacA vacuolating activity. Taken together, our data suggest that the high cell sensitivity to VacA depends on the presence of one or several GPI-anchored protein(s), intact membrane lipid rafts, and an uptake mechanism via a clathrin-independent endocytic pathway.


Subject(s)
Bacterial Proteins/pharmacology , Bacterial Toxins/pharmacology , Clathrin/metabolism , Endocytosis/drug effects , Phosphatidylinositols/metabolism , Actin Cytoskeleton/drug effects , Actins/drug effects , Animals , Bacterial Proteins/metabolism , CHO Cells/drug effects , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Cell Line/drug effects , Cricetinae , Cytochalasin D/pharmacology , Dogs , Dose-Response Relationship, Drug , Endocytosis/physiology , Humans , Iodine Radioisotopes , Nystatin/pharmacology , Proteins/drug effects , Proteins/metabolism , Type C Phospholipases/pharmacology , Vacuoles/drug effects
5.
J Comp Physiol B ; 170(4): 295-306, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10935520

ABSTRACT

Global contractile heart failure was induced in turkey poults by furazolidone feeding (700 ppm). Abnormal calcium regulation appears to be a key factor in the pathophysiology of heart failure, but the cellular mechanisms contributing to changes in calcium fluxes have not been clearly defined. Isolated ventricular myocytes from non-failing and failing hearts were therefore used to determine whether the whole heart and ventricular muscle contractile dysfunctions were realized at the single cell level. Whole cell current- and voltage-clamp techniques were used to evaluate action potential configurations and L-type calcium currents, respectively. Intracellular calcium transients were evaluated in isolated myocytes with fura-2 and in isolated left ventricular muscles using aequorin. Action potential durations were prolonged in failing myocytes, which correspond to slowed cytosolic calcium clearing. Calcium current-voltage relationships were normal in failing myocytes; preliminary evidence suggests that depressed transient outward potassium currents contribute to prolonged action potential durations. The number of calcium channels (as measured by radioligand binding) were also similar in non-failing and failing hearts. Isolated ventricular muscles from failing hearts had enhanced inotropic responses, in a dose-dependent fashion, to a calcium channel agonist (Bay K 8644). These data suggest that changes in intracellular calcium mobilization kinetics and longer calcium-myofilament interaction may be able to compensate for contractile failure. We conclude that the relationship between calcium current density and sarcoplasmic reticulum calcium release is a dynamic process that may be altered in the setting of heart failure at higher contraction rates.


Subject(s)
Calcium/metabolism , Disease Models, Animal , Heart Failure/physiopathology , Myocardial Contraction/physiology , Turkeys , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Action Potentials/drug effects , Action Potentials/physiology , Aequorin , Animals , Calcium Channel Agonists/pharmacology , Dose-Response Relationship, Drug , Electrophysiology , Heart Failure/metabolism , Muscle Fibers, Skeletal/physiology , Myocardium/cytology , Organ Size
6.
Mol Biol Cell ; 11(5): 1775-87, 2000 May.
Article in English | MEDLINE | ID: mdl-10793151

ABSTRACT

Cytotoxic necrotizing factor 1 (CNF1), a protein produced by pathogenic strains of Escherichia coli, activates the p21 Rho-GTP-binding protein, inducing a profound reorganization of the actin cytoskeleton. CNF1 binds to its cell surface receptor on HEp-2 cells with high affinity (K(d) = 20 pM). In HEp-2 cells the action of CNF1 is not blocked in the presence of filipin, a drug described to reduce cholera toxin internalization by the caveolae-like mechanism. Moreover, HEp-2 cells, which express a dominant negative form of proteins that impair the formation of clathrin coated-vesicles and internalization of transferrin (Eps15, dynamin or intersectin-Src homology 3), are still sensitive to CNF1. In this respect, the endocytosis of CNF1 is similar to the plant toxin ricin. However, unlike ricin toxin, CNF1 does not cross the Golgi apparatus and requires an acidic cell compartment to transfer its enzymatic activity into the cytosol in a manner similar to that required by diphtheria toxin. As shown for diphtheria toxin, the pH-dependent membrane translocation step of CNF1 could be mimicked at the level of the plasma membrane by a brief exposure to a pH of

Subject(s)
Bacterial Toxins/metabolism , Caveolins , Clathrin/metabolism , Cytosol/metabolism , Cytotoxins/metabolism , Endocytosis/physiology , Escherichia coli Proteins , Animals , Bacterial Toxins/genetics , Bacterial Toxins/pharmacokinetics , Binding Sites , Biological Transport , Catalytic Domain , Caveolin 1 , Cell Compartmentation , Cell Membrane/metabolism , Coated Vesicles/metabolism , Cytotoxins/genetics , Cytotoxins/pharmacokinetics , Dogs , Endocytosis/drug effects , Golgi Apparatus/metabolism , Humans , Membrane Proteins/metabolism , Microtubules/metabolism , Receptors, Cell Surface/metabolism , Tumor Cells, Cultured
7.
Circulation ; 101(14): 1679-85, 2000 Apr 11.
Article in English | MEDLINE | ID: mdl-10758050

ABSTRACT

BACKGROUND: We investigated whether decreased myofilament calcium contractile activation may, in part, contribute to heart failure. METHODS AND RESULTS: Calcium concentration required for 50% activation and Hill coefficient for fibers from nonfailing and failing human hearts at pH 7.1 were not different. Maximum calcium-activated force (F(max)) was also not different. However, at pH 6.8 and 6.9, differences were seen in myofilament calcium activation between nonfailing and failing hearts. At lower pH, failing myocardium was shifted left on the calcium axis compared with nonfailing myocardium, which suggested an increase in myofilament calcium responsiveness. Increased inorganic phosphate concentration decreased maximal force development by 56% in nonfailing and 36% in failing myocardium and shifted the calcium-force relationship by 2.01+/-0.22 versus 0.86+/-0.13 micromol/L, respectively (P<0.05). Addition of cAMP resulted in a 0. 56 micromol/L shift toward higher intracellular calcium concentrations in nonfailing myocardium and a 1.04 micromol/L shift in failing myocardium. Protein kinase A in the presence of cAMP resulted in a further rightward shift in nonfailing human myocardium but did not further shift the calcium-force relationship in fibers from failing hearts. cGMP also resulted in a greater decrease in myofilament calcium sensitivity in fibers from failing hearts. CONCLUSIONS: We propose that changes at the level of the thin myofilaments result in differential responses to changes in the intracellular milieu in nonfailing versus failing myocardium.


Subject(s)
Actin Cytoskeleton/metabolism , Calcium/metabolism , Myocardium/metabolism , Cadaver , Cardiac Output, Low/metabolism , Cardiac Output, Low/physiopathology , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic GMP/metabolism , Heart Ventricles , Humans , Hydrogen-Ion Concentration , Myocardial Contraction , Osmolar Concentration , Phosphates/metabolism
8.
Cardiovasc Drugs Ther ; 13(5): 455-63, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10547227

ABSTRACT

We have previously demonstrated that turkey poults fed furazolidone (Fz) in high concentrations (700 ppm) develop dilated cardiomyopathy (DCM) which approximates the human condition [1-3]. We wanted to study the effects of a calcium channel blocker in an animal model with a documented decrease in beta-receptor density, increased levels of circulating catecholamines, and abnormal calcium metabolism. The effects of a third generation calcium channel blocker has not been studied in our model. We hypothesized that the model would be predictive of the human condition and provide additional insights into the potential use of Ca2+ channel blockers in the setting of DCM. In the present study, we examined the effect of pranidipine, a new dihydropyridine calcium antagonist, in the setting of DCM on the gross and microscopic morphology of the heart and the overall contractile performance of the myocardium. A state of symptomatic to mild cardiomyopathy was induced in Broad-Breasted White turkey poults by administration of Fz for three weeks. Blood pressure, heart rate, fractional shortening, and body weight were monitored and compared in DCM animals treated with pranidipine and those given a placebo. After four weeks of treatment or no treatment with pranidipine, animals were euthanized and heart weight, cardiac dimensions, and microscopic morphology were compared. Progressive left ventricular (LV) dilatation and wall thinning was prevented with pranidipine treatment. In addition, microscopic examination demonstrated myocyte hypertrophy regression in DCM animals treated with pranidipine. In DCM animals, treatment with pranidipine resulted in significantly smaller left ventricular dimensions. We conclude that the calcium channel blocker pranidipine was not detrimental to global cardiac function in animals with dilated cardiomyopathy.


Subject(s)
Calcium Channel Blockers/therapeutic use , Cardiomyopathy, Dilated/drug therapy , Dihydropyridines/therapeutic use , Animals , Anti-Infective Agents, Local , Calcium/metabolism , Cardiomyopathy, Dilated/chemically induced , Disease Models, Animal , Furazolidone , Hemodynamics/drug effects , Myocardial Contraction/drug effects , Organ Size/drug effects , Turkeys
9.
Alcohol Clin Exp Res ; 23(10): 1668-72, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10550000

ABSTRACT

BACKGROUND: Excessive alcohol consumption is recognized as a common cause of left ventricular (LV) dysfunction. It is currently thought that 36% of all cases of dilated cardiomyopathy are due to excessive alcohol intake. Suitable animal models are needed to study the pathogenic mechanisms of ethanol-induced LV dysfunction. We have therefore created a new model of ethanol-induced LV dysfunction in the chicken. METHODS: For 12 weeks, adult chickens were given, twice a day, by gavage, 73% of their total calculated daily water intake containing a 20% ethanol concentration. Twenty percent ethanol also was placed in the water and provided ad libitum. Control chickens received the same volume of water by gavage twice a day without ethanol. Water without ethanol was given ad libitum to control birds. RESULTS: Our study shows that after a relatively short duration of ethanol ingestion, chickens developed LV dilatation and LV dysfunction. The serum concentrations of ethanol attained in this new model were similar to those reported in humans. Furthermore, unlike other currently available animal models of ethanol-induced cardiac disease, this model demonstrates myocyte hypertrophy, interstitial fibrosis, and myocytolysis, similar to observations in human ethanol-induced cardiac dysfunction. CONCLUSIONS: We conclude that this new avian model should provide a useful tool for investigating the mechanism(s) and pathophysiology of ethanol-induced dilated cardiomyopathy and heart failure.


Subject(s)
Cardiomyopathy, Alcoholic/etiology , Central Nervous System Depressants/adverse effects , Disease Models, Animal , Ethanol/adverse effects , Animals , Cardiomyopathy, Alcoholic/diagnostic imaging , Central Nervous System Depressants/blood , Chickens , Echocardiography , Ethanol/blood , Myocardium/pathology , Pilot Projects , Radiography , Random Allocation
10.
Am J Physiol ; 277(2): H474-80, 1999 08.
Article in English | MEDLINE | ID: mdl-10444471

ABSTRACT

Failing human myocardium has been associated with decreased sarcoplasmic reticulum (SR) Ca(2+)-ATPase activity. There remains controversy as to whether the regulation of SR Ca(2+)-ATPase activity is altered in heart failure or whether decreased SR Ca(2+)-ATPase activity is due to changes in SR Ca(2+)-ATPase or phospholamban expression. We therefore investigated whether alterations in cAMP-dependent phosphorylation of phospholamban may be responsible for the reduced SR Ca(2+)-ATPase activity in human heart failure. Protein levels of phospholamban and SR Ca(2+)-ATPase, detected by Western blot, were unchanged in failing compared with nonfailing human myocardium. There was decreased responsiveness to the direct activation of the SR Ca(2+)-ATPase activity by either cAMP (0.01-100 micromol/l) or protein kinase A (1-30 microgram) in failing myocardium. Using the backphosphorylation technique, we observed a decrease of the cAMP-dependent phosphorylation level of phospholamban by 20 +/- 2%. It is concluded that the impaired SR function in human end-stage heart failure may be due, in part, to a reduced cAMP-dependent phosphorylation of phospholamban.


Subject(s)
Calcium-Binding Proteins/metabolism , Calcium-Transporting ATPases/metabolism , Cardiac Output, Low/metabolism , Cyclic AMP/pharmacology , Sarcoplasmic Reticulum/enzymology , Adult , Cardiac Output, Low/enzymology , Cyclic AMP-Dependent Protein Kinases/pharmacology , Female , Humans , Male , Middle Aged , Myocardium/enzymology , Myocardium/metabolism , Phosphorylation/drug effects
11.
J Cell Biol ; 146(4): 819-30, 1999 Aug 23.
Article in English | MEDLINE | ID: mdl-10459016

ABSTRACT

Villin is an actin-binding protein localized in intestinal and kidney brush borders. In vitro, villin has been demonstrated to bundle and sever F-actin in a Ca(2+)-dependent manner. We generated knockout mice to study the role of villin in vivo. In villin-null mice, no noticeable changes were observed in the ultrastructure of the microvilli or in the localization and expression of the actin-binding and membrane proteins of the intestine. Interestingly, the response to elevated intracellular Ca(2+) differed significantly between mutant and normal mice. In wild-type animals, isolated brush borders were disrupted by the addition of Ca(2+), whereas Ca(2+) had no effect in villin-null isolates. Moreover, increase in intracellular Ca(2+) by serosal carbachol or mucosal Ca(2+) ionophore A23187 application abolished the F-actin labeling only in the brush border of wild-type animals. This F-actin disruption was also observed in physiological fasting/refeeding experiments. Oral administration of dextran sulfate sodium, an agent that causes colonic epithelial injury, induced large mucosal lesions resulting in a higher death probability in mice lacking villin, 36 +/- 9.6%, compared with wild-type mice, 70 +/- 8.8%, at day 13. These results suggest that in vivo, villin is not necessary for the bundling of F-actin microfilaments, whereas it is necessary for the reorganization elicited by various signals. We postulate that this property might be involved in cellular plasticity related to cell injury.


Subject(s)
Actins/metabolism , Calcium/metabolism , Carrier Proteins/metabolism , Intestinal Mucosa/metabolism , Microfilament Proteins/metabolism , Microvilli/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Animals , Carbachol/pharmacology , Carrier Proteins/genetics , Colitis/chemically induced , Colitis/pathology , Culture Techniques , Dextran Sulfate/pharmacology , Fasting , Female , Gene Deletion , Intestinal Mucosa/drug effects , Intestinal Mucosa/ultrastructure , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/genetics , Microvilli/drug effects , Microvilli/pathology , Microvilli/ultrastructure , Polymers
12.
J Mol Cell Cardiol ; 30(10): 1929-37, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9799647

ABSTRACT

Two of the most significant characteristics of failing human myocardium are an increased diastolic [Ca2+]i and a prolonged diastolic relaxation. These abnormalities are more pronounced at higher frequencies of stimulation and may be caused by an altered Ca2+ resequestration into the sarcoplasmic reticulum (SR). The force-frequency relationship was determined in multicellular preparations obtained from non-failing (n=6) and failing human myocardium (n=11). The active force in non-failing tissue increased as a function of the frequency of stimulation. In failing myocardium, an increase in frequency of stimulation (>1 Hz) was accompanied by a decrease in active force. Changes in the frequency of stimulation and active force were also associated with changes in intracellular calcium concentrations. The diastolic force in failing myocardium was augmented following an increase in frequency of stimulation, whereas in non-failing tissue, no increase in diastolic force was observed. Associated with the increase in diastolic force was an increase in intracellular diastolic calcium concentrations. The SR Ca2+ ATPase activity was reduced in failing compared to non-failing myocardium. SR Ca2+ ATPase was positively correlated with diastolic force in non-failing myocardium. The relationship between Ca2+ ATPase activity at 1 micromol/l [Ca2+] and active force between 0.5 and 2.0 Hz was different between failing and non-failing myocardium. The diastolic force demonstrate an inverse relationship with the SR Ca2+ ATPase activity in failing myocardium. These data suggest that a reduction in SR Ca2+ ATPase activity contributes to the impairment in both systolic and diastolic function of failing human hearts.


Subject(s)
Adenosine Triphosphatases/metabolism , Blood Pressure , Cardiomyopathy, Dilated/physiopathology , Heart Failure/physiopathology , Myocardial Contraction , Sarcoplasmic Reticulum/enzymology , Cardiomyopathies/enzymology , Cardiomyopathies/physiopathology , Cardiomyopathy, Dilated/enzymology , Diastole , Female , Heart Failure/enzymology , Humans , Male , Myocardial Ischemia/enzymology , Myocardial Ischemia/physiopathology , Reference Values , Systole
13.
Neuron ; 17(5): 823-35, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8938116

ABSTRACT

Serotonin (5-HT) has been shown to affect the development and patterning of the mouse barrelfield. We show that the dense transient 5-HT innervation of the somatosensory, visual, and auditory cortices originates in the thalamus rather than in the raphe: 5-HT is detected in thalamocortical fibers and most 5-HT cortical labeling disappears after thalamic lesions. Thalamic neurons do not synthesize 5-HT but take up exogenous 5-HT through 5-HT high affinity uptake sites located on thalamocortical axons and terminals. 3H-5-HT injected into the cortex is retrogradely transported to thalamic neurons. In situ hybridization shows a transient expression of the genes encoding the serotonin transporter and the vesicular monoamine transporter in thalamic sensory neurons. In these glutamatergic neurons, internalized 5-HT might thus be stored and used as a "borrowed transmitter" for extraneuronal signaling or could exert an intraneuronal control on thalamic maturation.


Subject(s)
Membrane Transport Proteins , Nerve Tissue Proteins , Neurons, Afferent/metabolism , Neuropeptides , Serotonin/pharmacokinetics , Thalamus/cytology , Age Factors , Animals , Antibody Specificity , Biological Transport/physiology , Carrier Proteins/analysis , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Gene Expression/physiology , Immunohistochemistry , In Situ Hybridization , Membrane Glycoproteins/analysis , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Neurons, Afferent/chemistry , Neurotransmitter Agents/analysis , Neurotransmitter Agents/genetics , Neurotransmitter Agents/metabolism , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Serotonin/biosynthesis , Serotonin/immunology , Serotonin Plasma Membrane Transport Proteins , Synaptic Vesicles/metabolism , Thalamus/growth & development , Thalamus/metabolism , Time Factors , Tritium , Vesicular Biogenic Amine Transport Proteins , Vesicular Monoamine Transport Proteins
14.
Pharm Res ; 5(12): 749-52, 1988 Dec.
Article in English | MEDLINE | ID: mdl-3247283

ABSTRACT

The effects of the administration of methemoglobin (MetHb) prepared in vitro were evaluated in Sprague-Dawley rats given increasing doses of potassium cyanide (KCN). Median lethal dose (LD50) studies were conducted by giving intraperitoneal injections of KCN (in 0.3- to 0.5-ml volumes), then 2 min later administering intravenous (iv) doses of 1000, 1500, or 2500 mg/kg of MetHb through the tail vein. Control rats received an equivalent volume of saline. The resulting LD50 values for KCN were 7.4 +/- 1.1, 11.7 +/- 1.1, 13.9 +/- 1.0, and 14.2 +/- 1.0 mg/kg (mean +/- SD) for the control (no MetHb) and 1000-, 1500-, and 2500-mg/kg dose groups, respectively. Additional groups of rats were given 1000, 1500, or 2500 mg/kg MetHb and submitted for necropsy. The gross finding of darkened kidneys was present in both dose groups, but became consistent and more prominent in the 2500-mg/kg dose group. Evidence of pathologic changes was not present in other organs. Single-dose pharmacokinetic studies were conducted using iv doses of 1600 and 2500 mg/kg MetHb. The elimination half-life was similar in both doses (62.6 min), but the volume of distribution (95.3 +/- 7.2 and 126.3 +/- 5.2 ml/kg, mean +/- SE) and clearance (1.1 +/- 0.1 and 1.5 +/- 0.1 ml/min/kg) were significantly different (P less than 0.05) for the 1600- and 2500-mg/kg dose groups, respectively. From these data we conclude that although MetHb is cleared from the vascular system rapidly, it may be an effective and nontoxic antidote for doses of cyanide up to twice that of the control LD50.


Subject(s)
Antidotes , Cyanides/poisoning , Methemoglobin/pharmacology , Potassium Cyanide/poisoning , Animals , Kidney/drug effects , Kidney/pathology , Lethal Dose 50 , Male , Methemoglobin/pharmacokinetics , Potassium Cyanide/antagonists & inhibitors , Rats , Rats, Inbred Strains
15.
Arch Androl ; 11(3): 233-7, 1983 Dec.
Article in English | MEDLINE | ID: mdl-6660973

ABSTRACT

An analysis of sperm abnormalities can be used as a rapid method to determine the mutagenic potential of chemical agents as well as the toxic potential of chemicals on the whole animal. O-Xylene was investigated for its potential mutagenicity by the sperm abnormality assay. Sprague Dawley rats 10-16 weeks old were injected intraperitoneally (ip) with 0.5 and 1.5 ml/kg body weight of o-xylene in corn oil. Five weeks after treatment, animals were sacrificed and sperm examined for morphological abnormalities. Several morphologically abnormal sperm types were observed; among these were those with amorphous heads, and banana-like heads. Animals housed at room temperatures of 20-24 degrees C showed no significant increase in abnormal sperm over the control. Rats housed at temperatures between 24 and 30 degrees C after ip injection with 0.5 ml/kg o-xylene showed a significant increase in abnormal sperm.


Subject(s)
Mutagens , Spermatozoa/abnormalities , Xylenes/toxicity , Animals , Kinetics , Male , Rats , Rats, Inbred Strains , Spermatozoa/drug effects , Temperature
16.
J Submicrosc Cytol ; 14(4): 621-6, 1982 Oct.
Article in English | MEDLINE | ID: mdl-7143516

ABSTRACT

Pulmonary alveolar macrophages (PAM) from rats exposed to a single dose of ZnO or NiO showed distinct changes in size and ultrastructure. ZnO exposure brought about considerable reduction in the size of the cells, which can be distinguished into two types of macrophages. The macrophages contained a prominent nucleolus, several primary and secondary lysosomes, membrane formations, electron dense structures, and small dense mitochondria. In addition, electron dense and membrane bound structures were seen in the interstitial macrophages and in the intercellular spaces. NiO exposure induced hypertrophy and hyperplasia of the alveolar walls. The macrophages contained invaginated bizarre-shaped nuclei, pigment aggregations, and membrane bound structures. The results seem to suggest that after a one week recovery period following injections with ZnO and NiO, ZnO-related accumulations were transferred from alveolar to interstitial macrophages, however NiO-associated structures were contained within the alveolar macrophages.


Subject(s)
Macrophages/drug effects , Nickel/toxicity , Pulmonary Alveoli/drug effects , Zinc Oxide/toxicity , Zinc/toxicity , Animals , Intracellular Membranes/ultrastructure , Lysosomes/ultrastructure , Macrophages/ultrastructure , Male , Microscopy, Electron , Pulmonary Alveoli/ultrastructure , Rats , Rats, Inbred Strains
17.
J Submicrosc Cytol ; 14(2): 347-53, 1982 Apr.
Article in English | MEDLINE | ID: mdl-7077715

ABSTRACT

The morphological effects of single intratracheal injections of copper oxide (CuO) and cadmium oxide (CdO) (5 mg) on rat alveolar macrophages were studied. Rats sacrificed after a one week recovery period showed distinct morphological changes induced by CuO and CdO. Copper oxide induced hypertrophy of both the alveolar macrophages as well as the epithelial lining. The macrophages contained polymorphic nuclei with margination of the chromatin, crystalloid-like inclusions, concentric and parallel lamellar structures, lattice formationa and degenerative membranous structures. After CdO treatment, a slight increase in the number of alveolar macrophages and an attenuated thickening of the epithelial wall were noted. Two types of macrophages were identified: one type was large in size, with numerous vacuoles, electron dense aggregations, phagocytized mitochondria and fragmented chromatin. The other type was smaller in size, with fewer vacuoles, a distinct rough endoplasmic reticulum, mitochondria, and a highly convoluted plasma membrane. The results were suggestive of degenerative morphological alterations due to CuO and CdO treatments which were incompatible with normal macrophage function.


Subject(s)
Cadmium Compounds , Cadmium/pharmacology , Copper/pharmacology , Macrophages/ultrastructure , Oxides , Animals , Macrophages/drug effects , Male , Microscopy, Electron , Rats , Rats, Inbred Strains , Vacuoles/drug effects , Vacuoles/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL