Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
Mol Pharmacol ; 87(2): 251-62, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25468882

ABSTRACT

Chemokines and chemokine receptors are key modulators in inflammatory diseases and malignancies. Here, we describe the identification and pharmacologic characterization of nanobodies selectively blocking CXCR2, the most promiscuous of all chemokine receptors. Two classes of selective monovalent nanobodies were identified, and detailed epitope mapping showed that these bind to distinct, nonoverlapping epitopes on the CXCR2 receptor. The N-terminal-binding or class 1 monovalent nanobodies possessed potencies in the single-digit nanomolar range but lacked complete efficacy at high agonist concentrations. In contrast, the extracellular loop-binding or class 2 monovalent nanobodies were of lower potency but were more efficacious and competitively inhibited the CXCR2-mediated functional response in both recombinant and neutrophil in vitro assays. In addition to blocking CXCR2 signaling mediated by CXCL1 (growth-related oncogene α) and CXCL8 (interleukin-8), both classes of nanobodies displayed inverse agonist behavior. Bivalent and biparatopic nanobodies were generated, respectively combining nanobodies from the same or different classes via glycine/serine linkers. Interestingly, receptor mutation and competition studies demonstrated that the biparatopic nanobodies were able to avidly bind epitopes within one or across two CXCR2 receptor molecules. Most importantly, the biparatopic nanobodies were superior over their monovalent and bivalent counterparts in terms of potency and efficacy.


Subject(s)
Receptors, Interleukin-8B/antagonists & inhibitors , Receptors, Interleukin-8B/metabolism , Signal Transduction/physiology , Single-Domain Antibodies/metabolism , Single-Domain Antibodies/pharmacology , Amino Acid Sequence , Animals , Binding Sites/physiology , CHO Cells , Camelids, New World , Cricetinae , Cricetulus , Humans , Molecular Sequence Data , Receptors, Interleukin-8B/genetics , Signal Transduction/drug effects , Single-Domain Antibodies/genetics
2.
Br J Pharmacol ; 159(7): 1429-39, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20233217

ABSTRACT

BACKGROUND AND PURPOSE: We have previously shown that SB265610 (1-(2-bromo-phenyl)-3-(7-cyano-3H-benzotriazol-4-yl)-urea) behaves as an allosteric, inverse agonist at the C-X-C chemokine (CXCR)2 receptor. The aim of this study was to determine whether SB265610, in addition to two other known antagonists, bind to either of the two putative, topographically distinct, allosteric binding sites previously reported in the Literature. EXPERIMENTAL APPROACH: Ten single point mutations were introduced into the CXCR2 receptor using site-directed mutagenesis. Three CXCR2 antagonists were investigated, SB265610, Pteridone-1 (2-(2,3 difluoro-benzylsulphanyl)-4-((R)-2-hydroxy-1-methyl-ethylamino)-8H-pteridin-7-one) and Sch527123 (2-hydroxy-N,N-dimethyl-3-{2-[[(R)-1-(5-methyl-furan-2-yl)-propyl]amino]-3,4-dioxo-cyclobut-1enylamino}-benzamide), and the effect of these mutations on their binding affinity and ability to inhibit interleukin-8-stimulated binding of [(35)S]GTPgammaS was examined. KEY RESULTS: Seven of the nine mutations introduced into the C-terminal domain and intracellular loops of the receptor produced a significant reduction in affinity at least one of the antagonists tested. Of those seven mutations, three produced a significant reduction in the affinity of all three antagonists, namely K320A, Y314A and D84N. In all but one mutation, the changes observed on antagonist affinity were matched with effects on inhibition of interleukin-8-stimulated [(35)S]GTPgammaS binding. CONCLUSIONS AND IMPLICATIONS: These antagonists bind to a common intracellular, allosteric, binding site of the CXCR2 receptor, which has been further delineated. As many of these mutations are close to the site of G protein coupling or to a region of the receptor that is responsible for the transduction of the activation signal, our results suggest a molecular mechanism for the inhibition of receptor activation.


Subject(s)
Benzamides/pharmacology , Cyclobutanes/pharmacology , Phenylurea Compounds/pharmacology , Receptors, Interleukin-8B/antagonists & inhibitors , Triazoles/pharmacology , Allosteric Site , Animals , Benzamides/chemistry , CHO Cells , Cricetinae , Cricetulus , Cyclobutanes/chemistry , Flow Cytometry , Humans , Models, Molecular , Phenylurea Compounds/chemistry , Point Mutation , Radioligand Assay , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/metabolism , Triazoles/chemistry
3.
Br J Pharmacol ; 158(1): 328-38, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19422399

ABSTRACT

BACKGROUND AND PURPOSE: In several previous studies, the C-X-C chemokine receptor (CXCR)2 antagonist 1-(2-bromo-phenyl)-3-(7-cyano-3H-benzotriazol-4-yl)-urea (SB265610) has been described as binding competitively with the endogenous agonist. This is in contrast to many other chemokine receptor antagonists, where the mechanism of antagonism has been described as allosteric. EXPERIMENTAL APPROACH: To determine whether it displays a unique mechanism among the chemokine receptor antagonists, the mode of action of SB265610 was investigated at the CXCR2 receptor using radioligand and [(35)S]-GTPgammaS binding approaches in addition to chemotaxis of human neutrophils. KEY RESULTS: In equilibrium saturation binding studies, SB265610 depressed the maximal binding of [(125)I]-interleukin-8 ([(125)I]-IL-8) without affecting the K(d). In contrast, IL-8 was unable to prevent binding of [(3)H]-SB265610. Kinetic binding experiments demonstrated that this was not an artefact of irreversible or slowly reversible binding. In functional experiments, SB265610 caused a rightward shift of the concentration-response curves to IL-8 and growth-related oncogene alpha, but also a reduction in maximal response elicited by each agonist. Fitting these data to an operational allosteric ternary complex model suggested that, once bound, SB265610 completely blocks receptor activation. SB265610 also inhibited basal [(35)S]-GTPgammaS binding in this preparation. CONCLUSIONS AND IMPLICATIONS: Taken together, these data suggest that SB265610 behaves as an allosteric inverse agonist at the CXCR2 receptor, binding at a region distinct from the agonist binding site to prevent receptor activation, possibly by interfering with G protein coupling.


Subject(s)
Drug Inverse Agonism , Phenylurea Compounds/pharmacology , Receptors, Interleukin-8B/agonists , Receptors, Interleukin-8B/antagonists & inhibitors , Triazoles/pharmacology , Allosteric Regulation/physiology , Animals , CHO Cells , Chemokines/chemistry , Chemokines/metabolism , Cricetinae , Cricetulus , Humans , Protein Binding/physiology , Receptors, Interleukin-8B/metabolism
4.
Eur J Clin Microbiol Infect Dis ; 26(4): 271-6, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17333081

ABSTRACT

The study presented here was performed in order to create a rule that identifies subjects at high risk for invasive candidiasis in the intensive care setting. Retrospective review and statistical modelling were carried out on 2,890 patients who stayed at least 4 days in nine hospitals in the USA and Brazil; the overall incidence of invasive candidiasis in this group was 3% (88 cases). The best performing rule was as follows: Any systemic antibiotic (days 1-3) OR presence of a central venous catheter (days 1-3) AND at least TWO of the following-total parenteral nutrition (days 1-3), any dialysis (days 1-3), any major surgery (days -7-0), pancreatitis (days -7-0), any use of steroids (days -7-3), or use of other immunosuppressive agents (days -7-0). The rate of invasive candidiasis among patients meeting the rule was 9.9%, capturing 34% of cases in the units, with the following performance: relative risk 4.36, sensitivity 0.34, specificity 0.90, positive predictive value 0.01, and negative predictive value 0.97. The rule may identify patients at high risk of invasive candidiasis.


Subject(s)
Candidiasis/epidemiology , Cross Infection/epidemiology , Adult , Aged , Aged, 80 and over , Brazil/epidemiology , Candidiasis/diagnosis , Candidiasis/microbiology , Cross Infection/diagnosis , Cross Infection/microbiology , Female , Humans , Intensive Care Units , Male , Middle Aged , Models, Statistical , Predictive Value of Tests , Reproducibility of Results , Retrospective Studies , Risk Factors , Sensitivity and Specificity , United States/epidemiology
5.
J Pharmacol Exp Ther ; 299(2): 748-52, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11602690

ABSTRACT

We examined the effects of purines and the pyrimidine UTP on cellular proliferation in the human astrocytoma cell line 1321N1. Treatment of cultured cells with 100 microM ATP or 2-chloroadenosine (2-CA) resulted in significant reductions in cell numbers after 2 days, whereas adenosine (ADO) exhibited a slower time course of inhibition of cell growth. Treatment with 100 microM UTP had no effect on cell numbers. 2-Chloroadenosine but neither ATP nor ADO resulted in an increase in cell death rates. A significant portion of the inhibitory response to ATP, ADO, or 2-CA was sensitive to the purine nucleoside transport inhibitor S-(p-nitrobenzyl)-6-thioguanosine, suggesting that uptake into cells was required for the inhibitory response. At least the majority of the observed responses to purines was not mediated by P1 (adenosine) receptors, because effects of ATP, ADO, or 2-CA were not affected by treatment of cells with the P1 receptor antagonist 8-(p-sulfophenyl)-theophylline. The absence of any known P2 (nucleotide) receptors in 1321N1 cells, coupled with the failure of the relatively stable ATP analog adenosine 5'-O-(3-thiotriphosphate) to alter cell growth rates, suggests that ATP acts indirectly to inhibit proliferation via one or more metabolic products. Although intracellular effects of purine nucleosides should be taken into account in future studies using 1321N1 cells, our findings also suggest 1321N1 cells as an excellent model for intracellular actions of nucleosides.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Astrocytoma/pathology , Brain Neoplasms/pathology , Purine Nucleosides/pharmacology , Theophylline/analogs & derivatives , 2-Chloroadenosine/pharmacology , Adenosine/pharmacology , Adenosine Triphosphate/pharmacology , Cell Death/drug effects , Cell Division/drug effects , Humans , Purinergic P1 Receptor Antagonists , Theophylline/pharmacology , Tumor Cells, Cultured , Uridine Triphosphate/pharmacology
6.
Cell ; 106(2): 171-82, 2001 Jul 27.
Article in English | MEDLINE | ID: mdl-11511345

ABSTRACT

Prions are self-propagating protein conformations. Recent research brought insight into prion propagation, but how they first appear is unknown. We previously established that the yeast non-Mendelian trait [PIN(+)] is required for the de novo appearance of the [PSI(+)] prion. Here, we show that the presence of prions formed by Rnq1 or Ure2 is sufficient to make cells [PIN(+)]. Thus, [PIN(+)] can be caused by more than one prion. Furthermore, an unbiased functional screen for [PIN(+)] prions uncovered the known prion gene, URE2, the proposed prion gene, NEW1, and nine novel candidate prion genes all carrying prion domains. Importantly, the de novo appearance of Rnq1::GFP prion aggregates also requires the presence of other prions, suggesting the existence of a general mechanism by which the appearance of prions is enhanced by heterologous prion aggregates.


Subject(s)
Prions/genetics , Prions/metabolism , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Fungal Proteins/chemistry , Fungal Proteins/genetics , Fungal Proteins/metabolism , Genes, Fungal/genetics , Glutathione Peroxidase , Models, Biological , Phenotype , Plasmids/genetics , Prions/chemistry , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/chemistry
7.
Peptides ; 22(3): 439-44, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11287099

ABSTRACT

ATP increases cAMP formation in bovine chromaffin cells, EC(50) = 7.1 x 10(-6) M. NPY, EC(50) = 4.1 x 10(-8) M, increases the efficacy of ATP (1.5-2 fold). Inclusion of the selective Y1 receptor antagonist 1229U91 produced a decrease in NPY potency (EC(50) = 2.7 x 10(-7) M). PTX pretreatment did not abolish either the effect of ATP nor the enhancement by NPY. NPY could also enhance the ability of angiotensin and bradykinin to increase cAMP formation. The selective phospholipase C inhibitor, U73122, and the selective protein kinase C inhibitors, bisindolylmaleimide I and RO-31-8425, were effective inhibitors of the enhancing effect of NPY.


Subject(s)
Adenosine Triphosphate/metabolism , Chromaffin Cells/metabolism , Cyclic AMP/metabolism , Neuropeptide Y/metabolism , Animals , Cattle , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Estrenes/pharmacology , Indoles/pharmacology , Maleimides/pharmacology , Neuropeptide Y/antagonists & inhibitors , Peptides, Cyclic/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Protein Kinase C/antagonists & inhibitors , Pyrrolidinones/pharmacology , Type C Phospholipases/antagonists & inhibitors , Virulence Factors, Bordetella/pharmacology
8.
J Pharmacol Exp Ther ; 297(2): 718-26, 2001 May.
Article in English | MEDLINE | ID: mdl-11303063

ABSTRACT

The molecular and pharmacological characteristics of muscarinic receptor subtypes in the rat parotid acinar cell line, PAR-C5, were determined and compared with native rat parotid glands to evaluate the PAR-C5 cell line as a model to study receptor-mediated secretion. Reverse transcription-polymerase chain reaction (RT-PCR) identified mRNAs for M(3), M(4), and M(5) receptor subtypes in both PAR-C5 cells and parotid glands. Specific [N-methyl-(3)H]scopolamine binding in PAR-C5 and parotid membranes was to a single class of sites with mean K(D) values of 0.38 and 0.64 nM, respectively. Binding affinities (K(I) values) of muscarinic receptor subtype-selective drugs were obtained in side-by-side experiments comparing PAR-C5 cells with parotid glands. Nonlinear regression analysis indicated that competition binding curves for drugs in PAR-C5 cells and parotid glands fit best to a one-site binding model. K(I) values (nM) in PAR-C5 cells and parotid glands, respectively, for atropine (1.0, 2.1), darifenacin (1.2, 2.0), 4-diphenylacetoxy-N-methylpiperidine methiodide (4-DAMP) (2.9, 2.4), tripitramine (220, 180), pirenzepine (320, 720), and methoctramine (1400, 1700) were consistent with their known affinities at the M(3) receptor subtype. Affinities (K(B) values) of muscarinic receptor subtype-selective drugs for blocking methacholine-stimulated Ca(2+) mobilization were determined to show which subtype mediates Ca(2+)-dependent secretion in Fura-2-loaded PAR-C5 cells. K(B) values (nM) for atropine (0.44), 4-DAMP (0.38), pirenzepine (140), and methoctramine (320) for blocking Ca(2+) responses correlated well with their known affinities at the M(3) receptor (r(2) = 0.99). These results show that at the level of mRNA, receptor protein and function, PAR-C5 cells and parotid glands are similar, establishing PAR-C5 cells as an important model for muscarinic receptor-mediated secretion.


Subject(s)
Parotid Gland/metabolism , Receptors, Muscarinic/drug effects , Animals , Calcium/metabolism , Cell Line , Cell Membrane/metabolism , Cytosol/metabolism , In Vitro Techniques , Muscarinic Antagonists/metabolism , Parotid Gland/drug effects , RNA, Messenger/biosynthesis , RNA, Messenger/isolation & purification , Radioligand Assay , Rats , Receptors, Muscarinic/biosynthesis , Receptors, Muscarinic/genetics , Reverse Transcriptase Polymerase Chain Reaction
9.
EMBO J ; 19(9): 1942-52, 2000 May 02.
Article in English | MEDLINE | ID: mdl-10790361

ABSTRACT

The [PSI(+)] prion can be induced by overproduction of the complete Sup35 protein, but only in strains carrying the non-Mendelian [PIN(+)] determinant. Here we demonstrate that just as [psi (-)] strains can exist as [PIN(+)] and [pin(-)] variants, [PSI(+)] can also exist in the presence or absence of [PIN(+)]. [PSI(+)] and [PIN(+)] tend to be cured together, but can be lost separately. [PSI(+)]-related phenotypes are not affected by [PIN(+)]. Thus, [PIN(+)] is required for the de novo formation of [PSI(+)], not for [PSI(+)] propagation. Although [PSI(+)] induction is shown to require [PIN(+)] even when the only overexpressed region of Sup35p is the prion domain, two altered prion domain fragments circumventing the [PIN(+)] requirement are characterized. Finally, in strains cured of [PIN(+)], prolonged incubation facilitates the reappearance of [PIN(+)]. Newly appearing [PIN(+)] elements are often unstable but become stable in some mitotic progeny. Such reversibility of curing, together with our previous demonstration that the inheritance of [PIN(+)] is non-Mendelian, supports the hypothesis that [PIN(+)] is a prion. Models for [PIN(+)] action, which explain these findings, are discussed.


Subject(s)
Biological Factors/physiology , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Prions/chemistry , Prions/metabolism , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Amino Acid Sequence , Biological Factors/genetics , Cold Temperature , Crosses, Genetic , Fungal Proteins/genetics , Gene Expression , Guanidine/pharmacology , Mitosis , Models, Biological , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptide Termination Factors , Phenotype , Plasmids/genetics , Prions/genetics , Protein Structure, Tertiary/drug effects , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Sequence Deletion/genetics , Time Factors
10.
Am J Physiol ; 277(1): G127-36, 1999 07.
Article in English | MEDLINE | ID: mdl-10409159

ABSTRACT

Voltage-activated K(+) (K(V)) channels play an important role in regulating the membrane potential in excitable cells. In gastrointestinal (GI) smooth muscles, these channels are particularly important in modulating spontaneous electrical activities. The purpose of this study was to identify the molecular components that may be responsible for the K(V) currents found in the canine GI tract. In this report, we have examined the qualitative expression of eighteen different K(V) channel genes in canine GI smooth muscle cells at the transcriptional level using RT-PCR analysis. Our results demonstrate the expression of K(V)1.4, K(V)1.5, K(V)1.6, K(V)2.2, and K(V)4.3 transcripts in all regions of the GI tract examined. Transcripts encoding K(V)1.2, K(V)beta1.1, and K(V)beta1.2 subunits were differentially expressed. K(V)1.1, K(V)1.3, K(V)2.1, K(V)3.1, K(V)3.2, K(V)3.4, K(V)4.1, K(V)4.2, and K(V)beta2.1 transcripts were not detected in any GI smooth muscle cells. We have also determined the protein expression for a subset of these K(V) channel subunits using specific antibodies by immunoblotting and immunohistochemistry. Immunoblotting and immunohistochemistry demonstrated that K(V)1.2, K(V)1.4, K(V)1.5, and K(V)2.2 are expressed at the protein level in GI tissues and smooth muscle cells. K(V)2.1 was not detected in any regions of the GI tract examined. These results suggest that the wide array of electrical activity found in different regions of the canine GI tract may be due in part to the differential expression of K(V) channel subunits.


Subject(s)
Digestive System/metabolism , Muscle, Smooth/metabolism , Potassium Channels/chemistry , Potassium Channels/metabolism , Animals , DNA, Complementary/metabolism , Dogs , Immunoblotting , Immunohistochemistry , Potassium Channels/genetics , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism
11.
Curr Genet ; 35(2): 59-67, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10079323

ABSTRACT

We have previously described different variants of the yeast prion [PSI+] that can be obtained and maintained in the same genetic background. These [PSI+] variants, which differ in the efficiency of nonsense suppression, mitotic stability and the efficiency of curing by GuHCl, may correspond to different [PSI+] prion conformations of Sup35p or to different types of prion aggregates. Here we investigate the effects of overexpressing a mutant allele of SUP35 and find different effects on weak and strong [PSI+] variants: the suppressor phenotype of weak [PSI+] factors is increased, whereas the suppressor effect of strong [PSI+] factors is reduced. The SUP35 mutation used was originally described as a "Psi no more" mutation (PNM2) because it caused loss of [PSI+]. However, none of the [PSI+] variants in the strains used in our study were cured by PNM2. Indeed, when overexpressed, PNM2 induced the de novo appearance of both weak and strong [PSI+] variants with approximately the same efficiency as the overexpressed wild-type SUP35 allele. Our data suggest that the change in the region of oligopeptide repeats in the Sup35p N-terminus due to the PNM2 mutation modifies, but does not impair, the function of the prion domain of Sup35p.


Subject(s)
Fungal Proteins/genetics , Prions/genetics , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/genetics , Alleles , Binding Sites , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Gene Expression Regulation , Gene Expression Regulation, Fungal , Genetic Variation , Green Fluorescent Proteins , Luminescent Proteins/genetics , Mutation , Peptide Termination Factors , Prions/metabolism , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/metabolism
12.
Am J Physiol ; 276(3): H1091-7, 1999 03.
Article in English | MEDLINE | ID: mdl-10070096

ABSTRACT

Endothelial cells express receptors for ATP and UTP, and both UTP and ATP elicit endothelial release of vasoactive compounds such as prostacyclin and nitric oxide; however, the distinction between purine and pyrimidine nucleotide signaling is not known. We hypothesized that UTP plays a more important role in endothelial mitogenesis and chemotaxis than does ATP and that UTP is angiogenic. In cultured endothelial cells from guinea pig cardiac vasculature (CEC), both UTP and vascular endothelial growth factor (VEGF) were significant mitogenic and chemotactic factors; in contrast, ATP demonstrated no significant chemotaxis in CEC. In chick chorioallantoic membranes (CAM), UTP and VEGF treatments produced statistically significant increases in CAM vascularity compared with controls. These findings are the first evidence of chemotactic or angiogenic effects of pyrimidines; they suggest a role for pyrimidine nucleotides that is distinct from those assumed by purine nucleotides and provide for the possibility that UTP serves as an extracellular signal for processes such as endothelial repair and angiogenesis.


Subject(s)
Chemotactic Factors/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Mitogens/pharmacology , Neovascularization, Physiologic/physiology , Uridine Triphosphate/pharmacology , Adenosine Triphosphate/pharmacology , Animals , Chick Embryo , Coronary Vessels/cytology , Coronary Vessels/drug effects , Coronary Vessels/physiology , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/cytology , Extraembryonic Membranes/blood supply , Guinea Pigs , Humans , Lymphokines/pharmacology , Placenta Growth Factor , Pregnancy Proteins/pharmacology , Recombinant Proteins , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
13.
Am J Physiol ; 275(6): C1668-73, 1998 12.
Article in English | MEDLINE | ID: mdl-9843729

ABSTRACT

The role of intracellular guanosine 3',5'-cyclic monophosphate concentration ([cGMP]i) in nitric oxide (NO)-mediated relaxations in the uterus has become controversial. We found the NO donor S-nitroso-L-cysteine (CysNO) to potently (IC50 = 30 nM) inhibit spontaneous contractions in the nonpregnant human myometrium. CysNO treatment increased [cGMP]i significantly (P < 0.001), and this increase was blocked by the guanylyl cyclase inhibitors methylene blue (10 microM) or LY-83583 (1 microM); however, pretreatment with these guanylyl cyclase inhibitors failed to block CysNO-mediated relaxations. Intracellular cAMP concentrations were not altered by treatment of tissues with 10 microM CysNO. Incubation with the cGMP analogs 8-bromo-cGMP or beta-phenyl-1,N2-etheno-cGMP did not significantly affect spontaneous contractility. Pretreatment of tissues with charybdotoxin [a calcium-dependent potassium channel (BK) blocker] completely reversed CysNO-induced relaxations. We conclude that NO is a potent inhibitor of spontaneous contractile activity in the nonpregnant human uterus and that, although guanylyl cyclase and BK activities are increased by NO, increases in [cGMP]i are not required for NO-induced relaxations in this tissue.


Subject(s)
Cyclic GMP/physiology , Myometrium/physiology , Nitric Oxide/physiology , S-Nitrosothiols , Uterine Contraction/physiology , Aminoquinolines/pharmacology , Charybdotoxin/pharmacology , Cyclic GMP/metabolism , Cysteine/analogs & derivatives , Cysteine/pharmacology , Enzyme Inhibitors/pharmacology , Female , Guanylate Cyclase/antagonists & inhibitors , Humans , Methylene Blue/pharmacology , Myometrium/drug effects , Nitric Oxide Donors/pharmacology , Nitroso Compounds/pharmacology , Uterine Contraction/drug effects
14.
Br J Pharmacol ; 124(1): 63-8, 1998 May.
Article in English | MEDLINE | ID: mdl-9630344

ABSTRACT

1. We evaluated the effect of the nitric oxide (NO) donor CysNO (S-nitroso-L-cysteine) and endogenous NO upon spontaneous contractility in non-pregnant cynomolgus monkeys. We also assessed the role of intracellular guanosine 3',5'-cyclic monophosphate ([cyclic GMP]i) as a second messenger for NO in monkey uterine smooth muscle. 2. CysNO reduced spontaneous contractility by 84% (P < 0.05) at maximal concentrations, and significantly elevated [cyclic GMP]i (P < 0.05). However, increases in [cyclic GMP]i were not required for CysNO-induced relaxations; CysNO inhibited contractile activity despite the complete inhibition of guanylyl cyclase by methylene blue or LY83,583. 3. Analogues of cyclic GMP had no significant effect upon spontaneous contractile activity. L-arginine produced a 62% reduction in spontaneous activity (P < 0.05) while D-arginine had no effect. The competitive nitric oxide synthase (NOS) inhibitor N(omega)-nitro-L-arginine (L-NOARG) not only blocked L-arginine-induced relaxations, but also significantly increased spontaneous contractile activity when added alone (P < 0.05); the inactive D-enantiomer of NOARG had no such effect. 4. While both endogenous NO and the NO donor CysNO relax monkey myometrium, this effect is not causally related to CysNO-induced elevations in [cyclic GMP]i. The failure of cyclic GMP analogues to alter monkey uterine smooth muscle tension also argues against a role for [cyclic GMP]i in the regulation of uterine contractility. Not only do these findings argue for the existence of a functionally-relevant NOS in the monkey uterus, but increases in contractile activity seen in the presence of NOS inhibitors suggest a role for NO in the moment-to-moment regulation of contractile activity in this organ.


Subject(s)
Nitric Oxide/pharmacology , S-Nitrosothiols , Uterine Contraction/drug effects , Animals , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , Cyclic GMP/pharmacology , Cysteine/analogs & derivatives , Cysteine/pharmacology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Female , Guanylate Cyclase/antagonists & inhibitors , In Vitro Techniques , Macaca fascicularis , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Nitroso Compounds/pharmacology , Substrate Specificity , Uterus/drug effects , Uterus/enzymology , Uterus/metabolism
15.
Nucleic Acids Res ; 26(13): 3215-20, 1998 Jul 01.
Article in English | MEDLINE | ID: mdl-9628921

ABSTRACT

Potent viral promoters/enhancers are often used to achieve high level expression of ectopic genes in transient transfection analysis. By using a GAL4-responsive transcription assay system, we show that the use of potent eukaryotic expression vectors can lead to biased transcriptional effects. Three functionally diverse transcription factors, YY1, SRF and Msx-1, were examined and each was found to exhibit a strong transrepression function in the context of the DNA binding domain of GAL4 when expressed from the cytomegalovirus (pCMV) or simian virus 40 (pSV) promoters/enhancers. An internal 15 amino acid domain of YY1 mediating transrepression in the viral promoter setting was identified. This GAL4-mediated transcriptional repression could, however, be completely relieved by using the yeast alcohol dehydrogenase promoter (pADH) to drive gene expression, which is approximately 100-fold weaker than canonical pCMV and pSV in cultured mammalian cells. In addition, low level expression achieved with the pADH vector unveiled the intrinsic transactivation functions of YY1 and SRF previously not observed with the GAL4 assay system. Our results highlight a potential pitfall in conventional pCMV- and pSV-based transfection assays and suggest that the use of a low level expression system may be preferable in most transcriptional analysis.


Subject(s)
DNA-Binding Proteins/metabolism , Homeodomain Proteins/metabolism , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Saccharomyces cerevisiae Proteins , Transcription Factors/metabolism , Transcription, Genetic , Alcohol Dehydrogenase/genetics , Animals , Base Sequence , Cell Line , DNA Primers , Erythroid-Specific DNA-Binding Factors , Genetic Vectors , MSX1 Transcription Factor , Mice , Serum Response Factor , Transfection , YY1 Transcription Factor
16.
Ann Epidemiol ; 8(4): 212-6, 1998 May.
Article in English | MEDLINE | ID: mdl-9590599

ABSTRACT

PURPOSE: Although cryptococcosis is a significant opportunistic infection among patients with human immunodeficiency virus (HIV), there is conflicting information on rates of cryptococcosis among HIV-positive and HIV-negative patients. Precise state-wide epidemiologic data for cryptococcosis are not available in Alabama. METHODS: We conducted an active laboratory and hospital medical record-based surveillance for cryptococcosis in Alabama from October 1, 1992 to September 30, 1994. A case of cryptococcosis was defined as a patient's initial episode of cryptococcal disease and based on either a positive culture for C. neoformans from any normally sterile site, a positive latex agglutination serologic test for cryptococcal antigen in CSF or serum, or histopathologic findings consistent with C. neoformans. RESULTS: Over the two year period, 153 cases were identified. The diagnosis was based on positive culture (37%), positive antigen (24%), positive autopsy culture (2%), and histopathologic findings (4%). Further, 33% of the total cases were diagnosed from combined positive culture, antigen, or histopathology. Of the total 153 cases, 55% were in HIV-positive patients and 44% were in HIV-negative individuals and one case (1%) had an unknown HIV status. The overall annual incidence rate of cryptococcosis was 1.89 cases per 100,000 population. The incidence was 1638.7 per 100,000 in the HIV-positive population and 0.84 per 100,000 in the HIV-negative population. CONCLUSION: The first Alabama statewide active surveillance system for cryptococcosis confirms previous observations that rates of cryptococcosis are consistently higher in HIV-infected individuals than in their HIV-negative counterparts. In Alabama, cryptococcosis occurs more commonly in urban residents and in men. Cryptococcosis in HIV-positive persons is more likely to occur in the 20 to 44 year age group, whereas cryptococcosis in HIV-negative persons is more likely to occur in those greater than 45 years old.


Subject(s)
Cryptococcosis/epidemiology , AIDS-Related Opportunistic Infections/epidemiology , Adult , Alabama/epidemiology , Cryptococcus neoformans/isolation & purification , Female , Humans , Male , Middle Aged , Population Surveillance
17.
Proc Natl Acad Sci U S A ; 95(5): 2400-5, 1998 Mar 03.
Article in English | MEDLINE | ID: mdl-9482897

ABSTRACT

[PSI+], a non-Mendelian element found in some strains of Saccharomyces cerevisiae, is presumed to be the manifestation of a self-propagating prion conformation of eRF3 (Sup35p). Translation termination factor eRF3 enhances the activity of release factor eRF1 (Sup45p). As predicted by the prion model, overproduction of Sup35p induces the de novo appearance of [PSI+]. However, another non-Mendelian determinant, [PIN+], is required for this induction. We now show that SUP45 overexpression inhibits the induction of [PSI+] by Sup35p overproduction in [PIN+] strains, but has no effect on the propagation of [PSI+] or on the [PIN] status of the cells. We also show that SUP45 overexpression counteracts the growth inhibition usually associated with overexpression of SUP35 in [PSI+] strains. We argue that excess Sup45p inhibits [PSI+] seed formation. Because Sup45p complexes with Sup35p, we hypothesize that excess Sup45p may sequester Sup35p, thereby reducing the opportunity for Sup35p conformational flips and/or self-interactions leading to prion formation. This in vivo yeast result is reminiscent of the in vitro finding by investigators of Alzheimer disease that apolipoprotein E inhibits amyloid nucleation, but does not reduce seeded growth of amyloid.


Subject(s)
Fungal Proteins/biosynthesis , Peptide Termination Factors , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/genetics , Fungal Proteins/genetics , Plasmids , Prions/genetics , Protein Biosynthesis , Saccharomyces cerevisiae/physiology , Species Specificity , Suppression, Genetic , Terminator Regions, Genetic
18.
J Biol Chem ; 273(12): 6656-61, 1998 Mar 20.
Article in English | MEDLINE | ID: mdl-9506962

ABSTRACT

Regulated proteolysis has been postulated to be critical for proper control of cell functions. Muscle development, in particular, involves a great deal of structural adaptation and remodeling mediated by proteases. The transcription factor YY1 represses muscle-restricted expression of the sarcomeric alpha-actin genes. Consistent with this repressor function of YY1, the nuclear regulator is down-regulated at the protein level during skeletal as well as cardiac muscle cell differentiation. However, the YY1 message remains relatively unaltered throughout the myoblast-myotube transition, implicating a post-translational regulatory mechanism. We show that YY1 can be a substrate for cleavage by the calcium-activated neutral protease calpain II (m-calpain) and the 26 S proteasome. The calcium ionophore A23187 destabilized YY1 in cultured myoblasts, and the decrease in YY1 protein levels could be prevented by calpain inhibitor II and calpeptin. Treatment with the proteasome inhibitors MG132 and lactacystin resulted in the stabilization of YY1 protein, which is consistent with the finding that YY1 is readily polyubiquitinated in reticulocyte lysates. We further show that proteolytic targeting by calpain II and the proteasome involves different structural elements of YY1. This study thus illustrates two proteolytic pathways through which the transcriptional regulator can be differentially targeted under different cell growth conditions.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Muscle, Skeletal/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Zinc Fingers , Animals , Calpain/antagonists & inhibitors , Calpain/metabolism , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Down-Regulation , Erythroid-Specific DNA-Binding Factors , Hydrolysis , Multienzyme Complexes/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/embryology , Proteasome Endopeptidase Complex , Rats , Rats, Sprague-Dawley , Substrate Specificity , YY1 Transcription Factor
19.
Mol Cell Biochem ; 178(1-2): 141-9, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9546593

ABSTRACT

Myocardial infarctions and stroke arise primarily as a result of hypoxia/ischemia-induced cell injury. However, the molecular mechanism of cardiac cell death due to hypoxia has not been elucidated. We showed here that chemical hypoxia induced by 1 mM azide triggered apoptosis of isolated neonatal rat ventricular cardiac myocytes but had no effect on cardiac fibroblasts. The azide-induced cardiomyocyte apoptosis could be characterized by a reversible initiation phase (0-46 h after azide exposure) during which cytosolic ATP levels remained little affected. This was followed by an irreversible execution phase (12-18 h) exhibiting prominent internucleosomal DNA fragmentation, cell membrane leakage, mitochondrial dysfunction, and increased calpain messenger RNA. Blocking extracellular calcium influx or intracellular calcium release was each effective in suppressing myocyte apoptosis. Cell death was also found to be mediated by calcium sensitive signal transduction events based on the use of specific antagonists. Consistent with the induction of calpain expression during apoptosis, blocking de novo protein synthesis and calpain activity inhibited cell death. These regulatory features coupled with the ease of the cell system suggest that the myocyte apoptosis model described here should be useful in the study of events leading to the demise of the myocardium.


Subject(s)
Apoptosis , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Heart/physiology , Myocardium/metabolism , Protein Kinase C/metabolism , Animals , Animals, Newborn , Azides/pharmacology , Calpain/genetics , Calpain/metabolism , Cell Hypoxia , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA Fragmentation , Fibroblasts/metabolism , Heart/drug effects , L-Lactate Dehydrogenase/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
20.
Genetics ; 147(2): 507-19, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9335589

ABSTRACT

It has previously been shown that yeast prion [PSI+] is cured by GuHCl, although reports on reversibility of curing were contradictory. Here we show that GuHCl treatment of both [PSI+] and [psi-] yeast strains results in two classes of [psi-] derivatives: Pin+, in which [PSI+] can be reinduced by Sup35p overproduction, and Pin-, in which overexpression of the complete SUP35 gene does not lead to the [PSI+] appearance. However, in both Pin+ and Pin- derivatives [PSI+] is reinduced by overproduction of a short Sup35p N-terminal fragment, thus, in principle, [PSI+] curing remains reversible in both cases. Neither suppression nor growth inhibition caused by SUP35 overexpression in Pin+ [psi-] derivatives are observed in Pin- [psi-] derivatives. Genetic analyses show that the Pin+ phenotype is determined by a non-Mendelian factor, which, unlike the [PSI+] prion, is independent of the Sup35p N-terminal domain. A Pin- [psi-] derivative was also generated by transient inactivation of the heat shock protein, Hsp104, while [PSI+] curing by Hsp104 overproduction resulted exclusively in Pin+ [psi-] derivatives. We hypothesize that in addition to the [PSI+] prion-determining domain in the Sup35p N-terminus, there is another self-propagating conformational determinant in the C-proximal part of Sup35p and that this second prion is responsible for the Pin+ phenotype.


Subject(s)
Prions/metabolism , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/genetics , Fungal Proteins/metabolism , Guanidine , Heat-Shock Proteins/metabolism , Peptide Termination Factors , Phenotype , Saccharomyces cerevisiae/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL