Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Phys Rev Lett ; 127(6): 060505, 2021 Aug 06.
Article in English | MEDLINE | ID: mdl-34420337

ABSTRACT

Cross-resonance (CR) gates have emerged as a promising scheme for fault-tolerant quantum computation with fixed-frequency qubits. We experimentally implement an entangling CR gate by using a microwave-only control in a tunable coupling superconducting circuit, where the tunable coupler provides extra degrees of freedom to verify optimal conditions for constructing a CR gate. By developing a three-qubit Hamiltonian tomography protocol, we systematically investigate the dependency of gate fidelities on spurious qubit interactions and present the first experimental approach to the evaluation of the perturbation impact arising from spectator qubits. Our results reveal that the spectator qubits lead to reductions in CR gate fidelity dependent on ZZ interactions and particular frequency detunings between spectator and gate qubits. The target spectator demonstrates a more serious impact than the control spectator under a standard echo pulse scheme, whereas the degradation of gate fidelity is observed up to 22.5% as both the spectators are present with a modest ZZ coupling to the computational qubits. Our experiments uncover an optimal CR operation regime, and the method we develop here can readily be applied to improving other kinds of two-qubit gates in large-scale quantum circuits.

2.
J Biol Chem ; 276(52): 48702-8, 2001 Dec 28.
Article in English | MEDLINE | ID: mdl-11641412

ABSTRACT

ATP-binding cassette transporter A1 (ABCA1) mediates an active efflux of cholesterol and phospholipids and is mutated in patients with Tangier disease. Expression of ABCA1 may be increased by certain oxysterols such as 22(R)-hydroxycholesterol via activation of the nuclear hormone receptor liver X receptor (LXR). In searching for potential modulators of ABCA1 expression, we have studied the effects of various mevalonate metabolites on the expression of ABCA1 in two human cell lines, THP-1 and Caco-2 cells. Most of the tested metabolites, including mevalonate, geranyl pyrophosphate, farnesyl pyrophosphate, and ubiquinone, failed to significantly change the expression levels of ABCA1. However, treatment with geranylgeranyl pyrophosphate resulted in a dose- and time-dependent reduction of ABCA1 expression. Geranylgeranyl pyrophosphate appears to reduce ABCA1 expression via two different mechanisms. One of these mechanisms is by acting directly as an antagonist of LXR since it reduces the interaction between LXR alpha or -beta with nuclear coactivator SRC-1. Another mechanism appears to involve activation of the Rho GTP-binding proteins since treatment of Caco-2 cells with inhibitors of geranylgeranyl transferase or the Rho proteins significantly increased the expression and promoter activity of ABCA1. Further studies showed that mutations in the DR4 element of the ABCA1 promoter completely eliminate the inducible activities of these inhibitors. These data indicate that activation of the Rho proteins may change the activation status of LXR.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Gene Expression Regulation/drug effects , Organic Chemicals , Polyisoprenyl Phosphates/pharmacology , Receptors, Retinoic Acid/metabolism , Receptors, Thyroid Hormone/metabolism , Transcription Factors/metabolism , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/genetics , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/physiology , Histone Acetyltransferases , Humans , Hydroxycholesterols/pharmacology , Interleukin-8/genetics , Interleukin-8/metabolism , Liver X Receptors , Mevalonic Acid/metabolism , Mutation , Nuclear Receptor Coactivator 1 , Orphan Nuclear Receptors , Promoter Regions, Genetic , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Retinoic Acid/genetics , Receptors, Thyroid Hormone/genetics , Recombinant Fusion Proteins/metabolism , Tangier Disease/genetics , Tangier Disease/metabolism
3.
J Leukoc Biol ; 69(6): 959-62, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11404382

ABSTRACT

Macrophages secrete matrix metalloproteinase 9 (MMP-9), an enzyme that weakens the fibrous cap of atherosclerotic plaques, predisposing them to plaque rupture and subsequent ischemic events. Recent work indicates that statins strongly reduce the possibility of heart attack. Furthermore, these compounds appear to exert beneficial effects not only by lowering plasma low-density-lipoprotein cholesterol but also by directly affecting the artery wall. To evaluate whether statins influence the proinflammatory responses of monocytic cells, we studied their effects on the chemotactic migration and MMP-9 secretion of human monocytic cell line THP-1. Simvastatin dose dependently inhibited THP-1 cell migration mediated by monocyte chemoattractant protein 1, with a 50% inhibitory concentration of about 50 nM. It also inhibited bacterial lipopolysaccharide-stimulated secretion of MMP-9. The effects of simvastatin were completely reversed by mevalonate and its derivatives, farnesylpyrophosphate and geranylgeranyl pyrophosphate, but not by ubiquinone. Additional studies revealed similar but more profound inhibitory effects with L-839,867, a specific inhibitor of geranylgeranyl transferase. However, alpha-hydroxyfarnesyl phosphonic acid, an inhibitor of farnesyl transferase, had no effect. C3 exoenzyme, a specific inhibitor of the prenylated small signaling Rho proteins, mimicked the inhibitory effects of simvastatin and L-839,867. These data supported the role of geranylgeranylation in the migration and MMP-9 secretion of monocytes.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Botulinum Toxins , Chemotaxis/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Matrix Metalloproteinase 9/metabolism , Monocytes/drug effects , Organic Chemicals , Protein Prenylation/drug effects , Protein Processing, Post-Translational/drug effects , Simvastatin/pharmacology , ADP Ribose Transferases/pharmacology , Alkyl and Aryl Transferases/antagonists & inhibitors , Cell Movement/drug effects , Chemokine CCL2/pharmacology , Depression, Chemical , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Leukemia, Monocytic, Acute/pathology , Lipopolysaccharides/pharmacology , Mevalonic Acid/pharmacology , Monocytes/enzymology , Monocytes/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Polyisoprenyl Phosphates/pharmacology , Sesquiterpenes , Simvastatin/antagonists & inhibitors , Tumor Cells, Cultured/drug effects
4.
J Immunol ; 167(1): 30-5, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11418628

ABSTRACT

11beta-hydroxysteroid dehydrogenases (11beta-HSD) perform prereceptor metabolism of glucocorticoids through interconversion of the active glucocorticoid, cortisol, with inactive cortisone. Although the immunosuppressive and anti-inflammatory activities of glucocorticoids are well documented, the expression of 11beta-HSD enzymes in immune cells is not well understood. Here we demonstrate that 11beta-HSD1, which converts cortisone to cortisol, is expressed only upon differentiation of human monocytes to macrophages. 11beta-HSD1 expression is concomitant with the emergence of peroxisome proliferator activating receptor gamma, which was used as a surrogate marker of monocyte differentiation. The type 2 enzyme, 11beta-HSD2, which converts cortisol to cortisone, was not detectable in either monocytes or cultured macrophages. Incubation of monocytes with IL-4 or IL-13 induced 11beta-HSD1 activity by up to 10-fold. IFN-gamma, a known functional antagonist of IL-4 and IL-13, suppressed the induction of 11beta-HSD1 by these cytokines. THP-1 cells, a human macrophage-like cell line, expressed 11beta-HSD1 and low levels of 11beta-HSD2. The expression of 11beta-HSD1 in these cells is up-regulated 4-fold by LPS. In summary, we have shown strong expression of 11beta-HSD1 in cultured human macrophages and THP-1 cells. The presence of the enzyme in these cells suggests that it may play a role in regulating the immune function of these cells.


Subject(s)
Hydroxysteroid Dehydrogenases/biosynthesis , Macrophages/cytology , Macrophages/enzymology , Monocytes/cytology , Monocytes/enzymology , 11-beta-Hydroxysteroid Dehydrogenases , Animals , Calcitriol/pharmacology , Cell Differentiation/immunology , Cell Line , Enzyme Induction/drug effects , Enzyme Induction/immunology , Humans , Interleukin-13/pharmacology , Interleukin-4/pharmacology , Lipopolysaccharides/pharmacology , Mice , Tetradecanoylphorbol Acetate/pharmacology , Tumor Cells, Cultured , U937 Cells
5.
J Steroid Biochem Mol Biol ; 77(2-3): 117-22, 2001 May.
Article in English | MEDLINE | ID: mdl-11377976

ABSTRACT

The 11beta-hydroxysteroid dehydrogenase (11beta-HSD) enzymes catalyze the interconversion of active glucocorticoids (GC) with their inert metabolites, thereby regulating the functional activity of GC. While 11beta-HSD type 1 (11beta-HSD1) activates GC from their 11-keto metabolites, 11beta-HSD type 2 (11beta-HSD2) inactivates GC. Here we report that both of these enzymes are expressed in human aortic smooth muscle cells (SMC), and that 11beta-HSD1 is more abundant and is differentially regulated relative to 11beta-HSD2. Stimulation of SMC with IL-1beta or TNFalpha led to a time- and dose-dependent increase of mRNA levels for 11beta-HSD1, while 11beta-HSD2 mRNA levels decreased. Parallel enzyme activity studies showed increased conversion of 3H-cortisone to 3H-cortisol but not 3H-cortisol to 3H-cortisone, demonstrating 11beta-HSD1 in SMC acts primarily as a reductase. A similar increase of 11beta-HSD1 mRNA expression was also found in human bronchial SMC upon stimulation, indicating the regulatory effect is not limited to vascular smooth muscle. Additional parallel studies revealed a similar pattern of induction for 11beta-HSD1 and monocyte chemoattractant protein-1, a well-defined proinflammatory molecule. These data suggest 11beta-HSD1 may play an important role in regulating inflammatory responses in the artery wall and lung.


Subject(s)
Aorta/enzymology , Hydroxysteroid Dehydrogenases/biosynthesis , Inflammation Mediators/metabolism , Muscle, Smooth, Vascular/enzymology , 11-beta-Hydroxysteroid Dehydrogenase Type 2 , Aorta/cytology , Base Sequence , Cells, Cultured , DNA Primers , Enzyme Induction , Humans , Hydroxysteroid Dehydrogenases/genetics , Muscle, Smooth, Vascular/cytology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
6.
J Interferon Cytokine Res ; 21(2): 93-8, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11244573

ABSTRACT

Matrix metalloproteinase-9 (MMP-9) may play an important role in the development of inflammatory bowel disease (IBD). However, the cellular source of MMP-9 in the inflamed mucosa of IBD remains unclear. Here we report that MMP-9 mRNA is expressed in CaCO-2 cells, an intestinal epithelial cell line, and that its expression is upregulated by inflammatory stimuli. Stimulation of CaCO-2 cells with interleukin-1beta (IL-1beta) or tumor necrosis factor-alpha (TNF-alpha) led to a dose-dependent increase in expression and secretion of MMP-9. In contrast, bacterial lipopolysaccharide (LPS) failed to induce expression or secretion of MMP-9, suggesting that an inflammatory reaction leading to cytokine release is a necessary step for the induction of MMP-9 release in intestinal epithelial cells. Additional studies show that induction of MMP-9 mRNA peaked at 16 h of IL-1beta stimulation, whereas expression of monocyte chemoattractant protein-1 (MCP-1) and IL-8 both peaked at 3 h of stimulation. Treatment of CaCO-2 cells with rosiglitazone, a peroxisome proliferator-activated receptor-gamma (PPAR-gamma) agonist, significantly reduced secretion of MMP-9, indicating that agents that activate PPAR-gamma may have therapeutic use in patients with IBD.


Subject(s)
Inflammatory Bowel Diseases/enzymology , Inflammatory Bowel Diseases/etiology , Matrix Metalloproteinase 9/biosynthesis , Thiazolidinediones , Caco-2 Cells , Humans , Inflammation Mediators/pharmacology , Inflammatory Bowel Diseases/genetics , Interleukin-1/pharmacology , Lipopolysaccharides/pharmacology , Matrix Metalloproteinase 9/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Rosiglitazone , Thiazoles/pharmacology , Transcription Factors/agonists , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects
7.
Biochem Biophys Res Commun ; 267(1): 345-9, 2000 Jan 07.
Article in English | MEDLINE | ID: mdl-10623622

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors that directly control numerous genes of lipid metabolism by binding to response elements in the promoter. It has recently been proposed that PPARgamma may also regulate genes for proinflammatory proteins, not through PPRE binding but by interaction with transcription factors AP-1, STAT, and NF-kappaB. Recent studies with cultured human monocytes, however, have failed to observe an inhibitory effect of PPARgamma agonists on induced expression of TNFalpha and IL-6, genes known to be controlled by AP-1, STAT, and NF-kappaB. In a similar fashion, we show here that PPARalpha (fenofibrate) or PPARgamma (rosiglitazone) agonists failed to modulate LPS-induced secretion of IL-8 in THP-1 cells. When we made parallel observations on another gene, matrix metalloproteinase 9 (MMP-9), we were surprised to find profound downregulation of LPS-induced secretion by both PPARalpha or PPARgamma agonists. These findings suggest that PPAR may regulate only a subset of the proinflammatory genes controlled by AP-1, STAT, and NF-kappaB. Effects of PPARs on MMP-9 may account for the beneficial effect of PPAR agonists in animal models of atherosclerosis.


Subject(s)
Fenofibrate/pharmacology , Gene Expression Regulation , Interleukin-8/genetics , Matrix Metalloproteinase 9/genetics , Receptors, Cytoplasmic and Nuclear/physiology , Thiazoles/pharmacology , Thiazolidinediones , Transcription Factors/physiology , Cell Nucleus/metabolism , Cytosol/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation/drug effects , Humans , Kinetics , Lipopolysaccharides/pharmacology , Monocytes , Promoter Regions, Genetic , Receptors, Cytoplasmic and Nuclear/drug effects , Rosiglitazone , Transcription Factors/drug effects , Tumor Cells, Cultured
8.
J Leukoc Biol ; 65(6): 750-6, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10380895

ABSTRACT

Secretory nonpancreatic group IIA phospholipase A2 (sPLA2), a lipolytic enzyme found in plasma, is thought to play an important role in inflammation. In patients with sepsis, a strong positive correlation is observed between the plasma level of sPLA2 and poor clinical outcome in sepsis. We have thus asked whether sPLA2 could play a role in enabling responses of cells to bacterial lipopolysaccharide (LPS), a key contributor to sepsis. In the presence of sPLA2, cellular responses to LPS were significantly increased. This was demonstrated in assays of LPS-stimulated interleukin-6 (IL-6) production in whole blood and binding of freshly isolated human polymorphonuclear neutrophils (PMN) to fibrinogen-coated surfaces. We further found that sPLA2 enhanced binding of labeled LPS to PMN, and that the sPLA2-mediated cell responses to LPS were all blocked by monoclonal antibodies directed against membrane CD14. Two properties ofsPLA2 may contribute to its activity to mediate responses to LPS. sPLA2 appears to bind LPS because pre-exposure of sPLA2 to LPS led to a dose-dependent increase in its ability to hydrolyze phospholid substrate, and incubation of sPLA2 with BODIPY-LPS micelles resulted in enhanced fluorescence, presumably from the disaggregation of the LPS aggregates. Additional studies demonstrated that the esterolytic function of sPLA2 is also needed both for the disaggregation of LPS and CD14-dependent cell stimulation. The precise mechanisms by which LPS-binding and esterolytic activity contribute to sPLA2 activity are not clear but our data strongly suggest that these activities result in interaction of LPS with CD14 and subsequent cell activation.


Subject(s)
Leukocytes/physiology , Lipopolysaccharides/pharmacology , Phospholipases A/pharmacology , Boron Compounds/blood , Fluorescent Dyes/metabolism , Humans , Interleukin-6/blood , Leukocytes/drug effects , Neutrophils/drug effects , Phospholipases A2 , Shock, Septic/enzymology
9.
Nat Med ; 3(4): 414-20, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9095175

ABSTRACT

We have studied the interactions of phosphodiester and phosphorothioate oligodeoxynucleotides with Mac-1 (CD11b/CD18; alpha M beta 2), a heparin-binding integrin found predominantly on the surface of polymorphonuclear leukocytes (PMNs), macrophages and natural killer cells. Binding of a homopolymer of thymidine occurred on both the alpha M and beta 2 subunits. Soluble fibrinogen, a natural ligand for Mac-1, was an excellent competitor of the binding of a phosphorothioate oligodeoxynucleotide to both TNF-alpha-activated and nonactivated PMNs. Upregulation of cell-surface Mac-1 expression increased cell-surface binding of oligodeoxynucleotides. Binding was inhibited by anti-Mac-1 monoclonal antibodies, and the increase in cell-surface binding was correlated with a three- to fourfold increase in internalization by PMNs. An oligodeoxynucleotide inhibited beta 2-dependent migration through Matrigel, but the production of reactive oxygen species in PMNs adherent to fibrinogen dramatically increased. Thus, our data demonstrate that Mac-1 is a cell-surface receptor for oligodeoxynucleotides that can mediate their internalization and that this binding may have important functional consequences.


Subject(s)
CD18 Antigens/metabolism , DNA-Binding Proteins/metabolism , Macrophage-1 Antigen/metabolism , Membrane Proteins , Neutrophils/metabolism , Oligodeoxyribonucleotides/metabolism , Receptors, Lipoprotein , Antibodies, Monoclonal/pharmacology , Binding, Competitive , CD18 Antigens/genetics , CD18 Antigens/immunology , Chemotaxis, Leukocyte/drug effects , Humans , Ligands , Macrophage-1 Antigen/genetics , Macrophage-1 Antigen/immunology , Protein Binding/drug effects , Reactive Oxygen Species/metabolism , Receptors, Immunologic/metabolism , Receptors, Scavenger , Scavenger Receptors, Class B , Signal Transduction , Thionucleotides/metabolism , Up-Regulation
10.
J Exp Med ; 184(4): 1213-23, 1996 Oct 01.
Article in English | MEDLINE | ID: mdl-8879192

ABSTRACT

Integrin CR3 (CD11b/CD18, Mac-1, alpha M beta 2) mediates the transient adhesion of polymorphonuclear leukocytes (PMN) to surfaces coated with fibrinogen, C3bi, ICAM-1, and other ligands. Recent studies (Cai, T.-Q., and S.D. Wright 1995. J. Biol. Chem. 270:14358) suggest that adhesion may be favored by stimulus-dependent changes in the kinetics of ligand binding by CR3. Cell detachment, on the other hand, must occur by a different mechanism because binding kinetics cannot affect cell adhesion after binding of ligand has occurred. We have sought a mechanism that would reverse binding of ligand to CR3 and report here that lysates of PMN contain an endogenous ligand that binds CR3 and competes the binding of C3bi. Purification and sequence analysis identified the structurally homologous azurophilic granule proteins, elastase, protease 3, and azurocidin as candidates. Studies with purified elastase and azurocidin showed that each bound specifically to purified, immobilized CR3. Elastase may play a role in modulating integrin-mediated cell adhesion because it is expressed at the cell surface, and the expression level is inversely proportional to cell adhesivity. Furthermore, a monoclonal antibody against elastase prevented detachment of PMN from fibrinogen-coated surfaces and blocked chemotaxis, confirming a role for this protein in regulating integrin-mediated adhesion. These studies suggest a model for release of integrin-mediated cell adhesion in which endogenous ligands such as elastase may release adhesion by "'eluting" substrate-bound ligand. A role for the proteolytic activity of elastase appears likely but is not demonstrated in this study.


Subject(s)
CD18 Antigens/metabolism , Carrier Proteins , Cell Adhesion/physiology , Leukocyte Elastase/metabolism , Macrophage-1 Antigen/metabolism , Neutrophils/physiology , Amino Acid Sequence , Antimicrobial Cationic Peptides , Binding, Competitive , Blood Proteins/metabolism , Chemotaxis, Leukocyte/drug effects , Complement C3b/metabolism , Fibrinogen/metabolism , Humans , Kinetics , Leukocyte Elastase/isolation & purification , Leukocyte Elastase/pharmacology , Ligands , Molecular Sequence Data , Neutrophils/enzymology , Protease Inhibitors/pharmacology , Protein Binding/drug effects
11.
Cell Adhes Commun ; 3(5): 399-406, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8640377

ABSTRACT

Integrins exhibit reversible changes in their ability to bind ligands and these changes enable transient cell adhesion. We recently showed that leukocyte integrin CR3 (complement receptor type three, CD11b/CD18, alpha m beta 2) may be purified in a form that is either capable or incapable of binding soluble, monomeric ligand and that "inactive" CR3 may be rendered capable of binding ligand by addition of an anti-CR3 mAb known as KIM-127 (Cai and Wright, JBC. 270: 14358, 1995). Here, we demonstrate that active CR3 may be rendered inactive by treatment of immobilized receptor with EDTA. EDTA-treated CR3 failed to bind ligand even in the presence of mM Ca2+ and Mg2+, suggesting that EDTA-treatment caused a change in the receptor that is not readily reversed. EDTA-treated receptor did, however, bind ligand upon addition of KIM-127 plus Mg2+ with an affinity (17.8 +/- 4.5 nM) similar to untreated, active receptor (12.5 +/- 4.7 nM). EDTA-treated CR3 thus exhibits the properties of inactive CR3, in which the ligand binding site is cryptic but subject to exposure by KIM-127. A candidate for the cryptic ligand binding site is the I-domain, a Mg2+-binding region in the alpha chain of CR3. We found that monomeric C3bi binds directly to recombinant I-domain in a Mg(2+)-dependent fashion with an affinity of 300 +/- 113 nM. These results thus suggest that CR3 may be inactivated by removing tightly bound divalent cation from a cryptic site in CR3.


Subject(s)
Cations, Divalent/metabolism , Macrophage-1 Antigen/metabolism , Neutrophils/metabolism , Antibodies, Monoclonal , Base Sequence , Binding Sites/physiology , Calcium/metabolism , Dose-Response Relationship, Drug , Edetic Acid , Hydrogen-Ion Concentration , Macrophage-1 Antigen/chemistry , Macrophage-1 Antigen/immunology , Magnesium/metabolism , Molecular Sequence Data , Neutrophils/chemistry , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Protein Structure, Tertiary , Temperature
12.
J Biol Chem ; 270(24): 14358-65, 1995 Jun 16.
Article in English | MEDLINE | ID: mdl-7782296

ABSTRACT

Cell adhesion mediated by leukocyte integrin CR3 (CD11b/CD18, alpha m beta 2) may be rapidly modulated without changes in receptor number, and transient changes in adhesivity are thought to be driven by reversible alteration of the affinity of CR3 for ligand. Here we measure the binding affinity of CR3 using purified active and inactive receptor and the ligand, C3bi, coupled to alkaline phosphatase. Immobilized, active CR3 bound saturably and with high affinity (12.5 +/- 4.7 nM). In contrast, inactive CR3 exhibited no measurable binding. High affinity binding could be restored by the addition of the activating anti-CR3 monoclonal antibody KIM-127 to inactive CR3. Since the affinity of KIM-127 for active and inactive receptor was identical, it cannot contribute the energy to convert a low affinity receptor into a high affinity receptor. Rather, KIM-127 appears to facilitate binding of C3bi by lowering the activation energy for the shift from an inactive to an active state. These results suggest that CR3-mediated binding and detachment of cells is not driven by a reversible change in affinity but by two mechanistically distinct processes, an energetically neutral activation step for binding and an energy-dependent step that reverses binding of ligand.


Subject(s)
Macrophage-1 Antigen/metabolism , Antibodies, Monoclonal/immunology , Binding Sites , Cations, Divalent , Complement C3b/metabolism , Energy Metabolism , Humans , Kinetics , Macrophage-1 Antigen/immunology , Temperature , Time Factors
13.
Am J Vet Res ; 55(7): 934-43, 1994 Jul.
Article in English | MEDLINE | ID: mdl-7526753

ABSTRACT

Neutrophil functions were examined in healthy periparturient dairy cows (n = 46) and in cows with retained placenta and metritis complex (n = 20); metritis (n = 18); or mastitis (n = 13). Blood samples (50 ml) were collected from each cow via jugular vein twice weekly from 1.5 weeks before to 4 weeks after parturition. Neutrophil function was evaluated, using 6 tests: random migration, chemotaxis, ingestion, myeloperoxidase activity (iodination), superoxide production (cytochrome C reduction), and antibody-dependent cell-mediated cytotoxicity. Ability to ingest bacteria and random migration activity of neutrophils from clinically normal cows were high around parturition and increased immediately after parturition, whereas myeloperoxidase activity and antibody-dependent cell-mediated cytotoxicity ability of neutrophils from these cows decreased after parturition. Measurement of neutrophil function in 4 ovariectomized cows revealed significant (P < 0.0005) seasonal changes in results of all 6 functional assays. We observed various defects of neutrophil function in all cows with abnormal conditions after parturition. Before parturition, superoxide production activity by neutrophils from cows with metritis and chemotaxis by neutrophils from cows with mastitis were significantly (P < 0.001 and P < 0.05, respectively) lower, indicating that a defect of neutrophil function may be a predisposing factor in the development of these disorders. In conclusion, the host defense role of neutrophils in periparturient cows was impaired, principally because of a defect in killing capacity, which may increase susceptibility to infections. We also investigated the in vitro effects of arachidonic acid metabolites and recombinant human colony-stimulating factors (rhCSF) on functions of neutrophils from clinically normal and postparturient cows with abnormalities, including retained placenta, metritis, or mastitis (n = 5/group). Each abnormal cow was matched for postpartum period with a clinically normal cow. Neutrophils from individual cows were preincubated with arachidonic acid metabolites (prostaglandin F2 alpha, 10(-7) M; prostaglandin E2, 10(-6) M; leukotriene B4, 10(-8) M; and lipoxin B, 10(-8) M) and rhCSF (rh-granulocyte-CSF, 1,000 or 6,000 U/ml; rh-granulocyte-macrophage-CSF, 5 or 15 ng/ml) in a 37 C water bath for 30 minutes before submitting them to function assays. There was no response by neutrophils from either clinically normal or abnormal postparturient cows to treatment with either arachidonic acid metabolites or rhCSF in any of the 6 functional assays. However, preincubation of neutrophils alone in a 37 C water bath for 30 minutes resulted in some alteration of neutrophil function.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Cattle Diseases , Mastitis, Bovine/blood , Neutrophils/physiology , Placenta, Retained/veterinary , Pregnancy Complications/veterinary , Uterine Diseases/veterinary , Animals , Arachidonic Acids/blood , Cattle , Female , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Leukocyte Count , Neutrophils/drug effects , Placenta Diseases , Placenta, Retained/blood , Pregnancy , Pregnancy Complications/blood , Recombinant Proteins/pharmacology , Reference Values , Uterine Diseases/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...