Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
1.
Eur J Immunol ; 30(6): 1551-61, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10898490

ABSTRACT

Previous studies from our laboratory have shown that prolonged exposure of mouse macrophages to IFN-beta interferes with their subsequent ability to become activated for tumor cell killing. Data reported here show that such inhibition is due to reduced production of NO, resulting from decreased transcription of the gene that encodes inducible NO synthase (iNOS; EC 1.14.13.39). The molecular basis for such suppression was shown to be, at least in part, decreased nuclear accumulation of tyrosine-phosphorylated Stat1alpha (pStat1alpha), and a consequent change in the nuclear ratio of pStat1alpha to non-transactivating pStat1beta. Reduced phosphorylation was observed despite the fact that time-course studies revealed greater than normal quantities of both Stat1alpha and Stat1beta proteins in macrophages that had been pre-exposed to IFN-beta. The decrease in nuclear pStat1alpha was demonstrated to involve an increase in the rate of turnover of phosphorylated protein. The homodimeric form of pStat1alpha is essential for the expression of both the iNOS and IFN-regulatory factor-1 genes (the product of the latter is necessary for full expression of the iNOS gene). These results have broad implications, because they suggest that limiting the availability of homodimeric pStat1alpha is a means by which down-regulation of genes containing promoter-linked IFN-gamma-activated sites might be achieved.


Subject(s)
DNA-Binding Proteins/metabolism , Interferon-beta/metabolism , Macrophages/metabolism , Nitric Oxide Synthase/genetics , Trans-Activators/metabolism , Transcription, Genetic , Animals , Cell Nucleus/metabolism , Cells, Cultured , Culture Media , Cytotoxicity, Immunologic/immunology , Enhancer Elements, Genetic , Interferon Regulatory Factor-1 , Interferon-beta/pharmacology , Interferon-gamma/pharmacology , Kinetics , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Male , Mice , Mice, Inbred C3H , NF-kappa B/metabolism , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase Type II , Phosphoproteins/metabolism , Phosphorylation , Promoter Regions, Genetic , Recombinant Proteins , STAT1 Transcription Factor , Time Factors , Tumor Cells, Cultured
2.
Biochem J ; 349(Pt 1): 99-104, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10861216

ABSTRACT

It has previously been reported by us that a brief prior exposure of mouse bone marrow culture-derived macrophages to bacterial lipopolysaccharide (LPS) resulted in a dramatic reduction in their ability to produce NO in response to a subsequent stimulus with either interferon-gamma (IFN-gamma) or IFN-gamma plus LPS. We show here that this brief exposure to LPS results in an impaired response to subsequently added IFN-gamma. A 2--4 h pretreatment with LPS leads to a dramatic reduction in the IFN-gamma-induced DNA-binding of the transcription factor, signal transducer and activator of transcription 1 alpha (STAT1 alpha). This loss in ability to activate STAT1 alpha temporally correlates with the LPS-induced accumulation of mRNA encoding the suppressor of cytokine signalling-1 (SOCS-1). However, LPS does not directly induce the synthesis of SOCS-1. Rather, LPS induces the synthesis of autocrine/paracrine factors that are the true mediators of SOCS-1 induction. IFN-alpha/beta is one of these mediators, but plays only a partial role in the induction of SOCS-1 because neutralization of LPS-induced IFN-alpha/beta production incompletely inhibits the induction of SOCS-1. We show that mouse IFN-beta directly induces the synthesis of SOCS-1, without the need for prior protein synthesis, and does so with faster kinetics than does LPS. Our results are consistent with the non-specific nature of LPS-induced tolerance and provide a mechanistic insight into nonspecificity; LPS indirectly induces the synthesis of a protein mediator, SOCS-1, which inhibits the signalling that is induced by IFN-gamma.


Subject(s)
Carrier Proteins/metabolism , Interferon-alpha/physiology , Interferon-beta/physiology , Lipopolysaccharides/metabolism , Macrophages/metabolism , Repressor Proteins , Animals , Blotting, Northern , Bone Marrow Cells/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Kinetics , Male , Mice , Mice, Inbred C3H , RNA, Messenger/metabolism , Signal Transduction , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins , Time Factors
3.
J Leukoc Biol ; 65(5): 605-13, 1999 May.
Article in English | MEDLINE | ID: mdl-10331488

ABSTRACT

Bone marrow-culture-derived macrophages activated with interferon-gamma and lipopolysaccharide produced less nitric oxide (NO) when cultured with vesicular stomatitis virus (VSV)-infected BALB/c3T3 (3T3-VSV) than macrophages activated in an identical manner and cultured alone, with uninfected BALB/c3T3 (3T3), or with P815. However, all four groups of macrophages produced nearly the same amount of interleukin-6 (IL-6). Addition of VSV to activated macrophages did not change the amount of NO produced. The amount of NO generated by two non-macrophage sources of NO was not affected by the presence of either P815 or 3T3-VSV. Reverse transcriptase-polymerase chain reaction showed a decrease in the amount of inducible nitric oxide synthase (iNOS) but not IL-6 mRNA from macrophages cocultured with 3T3-VSV compared with macrophages cocultured with P815. The reduction in iNOS mRNA was confirmed by ribonuclease protection assay. When RAW 264.7 transfected with an iNOS regulatory construct were activated and incubated with 3T3-VSV there was a decrease in the expression of the reporter luciferase gene and NO production but not IL-6 production compared with cells incubated with either medium alone or with P815.


Subject(s)
3T3 Cells/virology , Bone Marrow Cells/virology , Cell Communication/immunology , Macrophage Activation/immunology , Macrophages/virology , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/biosynthesis , Vesicular stomatitis Indiana virus/immunology , 3T3 Cells/metabolism , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Coculture Techniques , Interferon-gamma/pharmacology , Interleukin-6/antagonists & inhibitors , Interleukin-6/biosynthesis , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Male , Mast-Cell Sarcoma/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred DBA , Nitric Oxide/physiology , Vesicular stomatitis Indiana virus/metabolism
4.
J Immunol ; 161(9): 4803-10, 1998 Nov 01.
Article in English | MEDLINE | ID: mdl-9794412

ABSTRACT

We have examined the role of Stat1alpha in the induction by LPS of the mouse inducible nitric oxide synthase (EC 1.14.13.39) gene. LPS induced both the tyrosine phosphorylation of Stat1alpha and the production of nitric oxide in a time- and dose-dependent manner. The phosphorylation of Stat1alpha elicited by LPS differed from that observed using IFN-gamma or IFN-beta, in that LPS induced less phosphorylated protein and the time course of induction was much delayed (2-4 h compared with 30 min). Cycloheximide inhibited LPS-mediated Stat1alpha phosphorylation. In addition, cell culture supernatants derived from macrophages treated with LPS for 4 h could be transferred to naive macrophage cultures resulting in rapid (30 min), rather than delayed (4 h), phosphorylation of Stat1alpha. Together, these results implicated an autocrine/paracrine effector protein(s) in the phosphorylation process. LPS stimulated phosphorylation of Stat1alpha in peritoneal macrophages derived from IFN-gamma-knockout mice, negating any possibility that IFN-gamma was the mediator. By contrast, neutralizing Ig raised against mouse IFN-alphabeta inhibited both the delayed LPS-mediated phosphorylation of Stat1alpha and the rapid induction of phosphorylation induced by supernatants from LPS-stimulated cultures. Collectively, these results show that LPS-induced IFN-alphabeta production, Stat1alpha activation, and nitrite accumulation closely parallel one another, suggesting that indirect activation of transcription factor Stat1alpha by IFN-alphabeta is a critical determinant of LPS-mediated inducible nitric oxide synthase gene expression.


Subject(s)
Interferon-alpha/physiology , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Macrophages, Peritoneal/metabolism , Nitric Oxide Synthase/biosynthesis , Nitric Oxide/biosynthesis , Transcription Factors/physiology , Animals , Culture Media, Conditioned/pharmacology , Cycloheximide/pharmacology , Enzyme Induction , Female , Interferon-Stimulated Gene Factor 3 , Interferon-gamma/deficiency , Interferon-gamma/genetics , Interferon-gamma/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Protein Synthesis Inhibitors/pharmacology
5.
Biochim Biophys Acta ; 1443(3): 315-22, 1998 Dec 22.
Article in English | MEDLINE | ID: mdl-9878805

ABSTRACT

The promoter of the gene encoding the inducible nitric oxide synthase (iNOS) contains an octamer motif which is of importance for its activation by specific stimuli. We show that in contrast to the promoter of the neuronal nitric oxide synthase gene (nNOS) which is strongly activated by the Oct-2 octamer-binding POU family transcription factor, the iNOS gene is only weakly activated by Oct-2 via its octamer motif. Unlike the nNOS promoter, however, the iNOS promoter is strongly activated by the POU family transcription factors Brn-3a and Brn-3b. This activation is dependent upon the octamer motif in the iNOS promoter and requires the activation domain located within the POU domain of Brn-3a or Brn-3b but not the N-terminal activation domain of Brn-3a. Thus different but related POU proteins play important roles in the regulation of the genes encoding different forms of nitric oxide synthase.


Subject(s)
DNA-Binding Proteins/physiology , Homeodomain Proteins/physiology , Nitric Oxide Synthase/genetics , Promoter Regions, Genetic/genetics , Transcription Factors/physiology , Animals , Binding Sites/genetics , Cell Line , DNA-Binding Proteins/genetics , Gene Expression Regulation , Genetic Vectors/genetics , Homeodomain Proteins/genetics , Mutation , Nitric Oxide Synthase Type II , Recombinant Fusion Proteins/genetics , Transcription Factor Brn-3 , Transcription Factor Brn-3B , Transcription Factors/genetics
6.
Biochim Biophys Acta ; 1351(3): 287-95, 1997 Apr 10.
Article in English | MEDLINE | ID: mdl-9130592

ABSTRACT

Upon cytokine induction, inducible nitric oxide synthase (iNOS) mRNA and enzyme activity in DLD-1 cells reached maximal levels at 6 and 8 h, respectively. A 3.7 kb 5'-flanking region of the human iNOS gene was used to prepare luciferase reporter constructs. Upon transfection of these constructs into DLD-1 cells and primary monkey hepatocytes, significant promoter activity was detected in the absence of cytokines, and this activity decreased with successive truncations of the human iNOS promoter. No increase in luciferase activity was observed after cytokine treatment, in spite of the fact that nuclear run-on analysis indicated that iNOS induction in DLD-1 cells was due, in part, to an increase in transcription rate. These results suggest that 3.7 kb of 5'-flanking DNA do not contain all of the elements required for transcriptional induction of the human iNOS gene. This differs from the mouse iNOS gene for which 1.7 kb of 5'-flanking DNA contain most or all of the elements that control iNOS expression in mouse macrophages. Thus, important cell- and species-specific mechanisms may exist for the control of iNOS expression.


Subject(s)
Adenocarcinoma/enzymology , Colorectal Neoplasms/enzymology , Liver/enzymology , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Cells, Cultured , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Dactinomycin/pharmacology , Half-Life , Haplorhini , Humans , Interferon-gamma/pharmacology , Interleukin-1/pharmacology , Interleukin-6/pharmacology , Lipopolysaccharides/pharmacology , Liver/cytology , Liver/drug effects , Mice , Nitric Oxide Synthase/drug effects , Promoter Regions, Genetic , Protein Biosynthesis , Protein Synthesis Inhibitors/pharmacology , Proteins/drug effects , RNA/biosynthesis , RNA/drug effects , RNA, Messenger/biosynthesis , RNA, Messenger/drug effects , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Time Factors , Transcription, Genetic , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology
7.
J Biol Chem ; 272(2): 1226-30, 1997 Jan 10.
Article in English | MEDLINE | ID: mdl-8995425

ABSTRACT

Mouse macrophages can be stimulated by interferon (IFN)-gamma and bacterial lipopolysaccharide (LPS) to produce nitric oxide (NO) as the result of expression of the inducible NO synthase (iNOS; EC 1.14.13.39) gene. The iNOS gene promoter contains a candidate gamma-interferon-activated site (GAS). In transfection studies reported here, it was demonstrated that a luciferase reporter-gene construct, containing four synthetic copies of the iNOS GAS, was inducible when transfected macrophages were stimulated with either IFN-gamma, LPS, or a combination of the two. Consistent with this finding were other transfection analyses, which showed that responsiveness of the intact iNOS promoter to these same agents was significantly reduced when two conserved nucleotide positions within the GAS were mutated. Oligonucleotide probes, which mimicked the iNOS GAS, formed a complex with proteins that appeared in the nuclei of IFN-gamma or IFN-gamma + LPS-treated macrophages within 30 min of stimulation, as shown by electrophoretic mobility shift assay. LPS alone also caused the the appearance of a nuclear protein capable of binding the iNOS GAS-containing oligonucleotide; however, in contrast to binding induced by IFN-gamma, approximately 2 h of stimulation with LPS were required. The protein bound to the iNOS GAS-containing oligonucleotide reacted specifically with an antibody raised against Stat1a, regardless of the stimulus used. These data collectively support the conclusion that binding of Stat1 alpha to the iNOS promoter's GAS is required for optimal induction of the iNOS gene by IFN-gamma and LPS.


Subject(s)
Gene Expression Regulation, Enzymologic , Interferon-gamma/physiology , Lipopolysaccharides/pharmacology , Nitric Oxide Synthase/genetics , Animals , DNA Mutational Analysis , Enzyme Induction , Mice , Nuclear Proteins/metabolism , Promoter Regions, Genetic
8.
Infect Immun ; 64(10): 4288-98, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8926101

ABSTRACT

We investigated the capacity of mouse macrophages obtained from senescent animals to respond in vitro to microbial stimuli. Significant hypersecretion of nitric oxide (NO) was observed in thioglycolate-elicited macrophages from senescent mice compared with those obtained from young mice in response to lipopolysaccharide (LPS). In contrast, both cell populations manifested equivalent responses to LPS with respect to tumor necrosis factor alpha secretion. Further, macrophages from senescent animals also showed potentiated responses to both zymosan and heat-killed Staphylococcus aureus, as assessed by NO production. Both cell populations were equivalently inhibited by a competitive inhibitor of NO synthase NG-monomethyl-L-arginine. Since endogenous beta interferon (IFN-beta) is recognized as an essential cofactor for LPS-induced NO production by macrophages, we investigated the role of IFN-beta in enhancing the capacity of both macrophage populations for LPS-induced NO production. Macrophages from young mice were minimally activated by LPS alone to express inducible NO synthase (iNOS), and the response was significantly potentiated by the addition of IFN-beta. These findings were confirmed by immunocytochemical staining of iNOS in which the frequency of iNOS-positive cells in response to LPS was enhanced in the presence of IFN-beta. Reverse transcription-PCR analyses revealed that macrophages from senescent animals produced larger amounts of iNOS mRNA in response to LPS. Further, exogenous IFN-beta potentiated iNOS mRNA expression in macrophages from young mice. In contrast, the frequency of LPS-activated macrophages for iNOS expression was markedly increased during senescence and addition of IFN-beta did not significantly change this frequency. These results correlated with reverse transcription PCR data showing high levels of iNOS mRNA in LPS-stimulated macrophages from senescent mice. LPS-induced NO production in macrophages from both young and senescent mice was inhibited by neutralizing antibody to either IFN-beta or IFN-gamma. Mixed cultures of macrophages from young and senescent mice stimulated with LPS manifested significantly enhanced NO production relative to that which would be predicted from an additive response of the two macrophage populations stimulated separately. The differential responsiveness of NO production observed with thioglycolate-elicited macrophages from young and senescent mice was also observed in resident macrophages but, interestingly, not in bone marrow culture-derived macrophages. These results suggest that environmental factors may be responsible for the potentiated NO responses of macrophages from senescent mice. Collectively, these data suggest that macrophages from senescent animals manifest an altered mechanism for regulation of macrophage function in NO production and iNOS expression by constitutive and/or induced expression of autoregulatory cytokines.


Subject(s)
Aging/metabolism , Gene Expression Regulation, Enzymologic , Macrophages/metabolism , Nitric Oxide Synthase/genetics , Animals , Immunohistochemistry , Interferons/physiology , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred CBA , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/analysis , Tumor Necrosis Factor-alpha/biosynthesis , omega-N-Methylarginine/pharmacology
9.
Dermatol Surg ; 22(7): 613-6, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8680783

ABSTRACT

BACKGROUND: The continuing development of new types and applications of lasers has appeared to surpass the development of specific eye protection for these lasers. There are a variety of eye shields on the market, but few are specifically designed for laser protection. OBJECTIVE: Our purpose was to test a variety of eye shields by two parameters, light transmission and temperature rise, and to determine from these measurements the most protective shield for patients. METHODS: We tested four plastic shields, one metal shield, and two sets of tanning goggles for temperature rise and light transmission when irradiated with a beam from a flashlamp-pumped, pulsed-dye laser. RESULTS: The temperature rise at the surface of the shield opposite the laser impacts was no more than 0.2 degree C in any case. White light was transmitted at significant levels through several of the shields, but yellow light transmittance was noted only through the green eye shield. CONCLUSION: Our measurements indicate that all except the green shield appeared safe from transmission of the 585-nm radiant energy. However, the optimal laser eye shield, in our opinion, would be a composite of several different shields' characteristics.


Subject(s)
Cornea , Eye Protective Devices , Laser Therapy , Color , Equipment Design , Hot Temperature , Humans , Laser Therapy/adverse effects , Light , Metals , Plastics , Thermometers
10.
J Leukoc Biol ; 59(4): 575-85, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8613707

ABSTRACT

The work reported here resolves, at the level of gene regulation, the controversy as to whether or not human monocytes/macrophages can produce nitric oxide (NO) when stimulated with lipopolysaccharide (LPS), with or without co-stimulation by interferon-gamma (IFN-gamma). Studies included structural comparison of the promoters for human and mouse inducible NO synthase (iNOS) genes, transfection and assay of human and mouse iNOS promoter regions in response to LPS +/- IFN-gamma, and electrophoretic mobility shift assays of kappa B response elements. Two explanations for hyporesponsiveness of the human iNOS promoter to LPS +/- IFN-gamma were found: (1) multiple inactivating nucleotide substitutions in the human counterpart of the enhancer element that has been shown to regulate LPS/IFN-gamma induced expression of the mouse iNOS gene; and (2) and absence of one or more nuclear factors in human macrophages (e.g., an LPS-inducible nuclear factor-kappa B/Rel complex), that is (are) required for maximal expression of the gene. The importance of resolution of this controversy is that future research in this area should be directed toward the understanding of alternative mechanisms that can result in the successful production of NO.


Subject(s)
Gene Expression Regulation, Enzymologic/drug effects , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Nitric Oxide Synthase/genetics , Animals , Base Sequence , Humans , Leukocytes, Mononuclear/enzymology , Leukocytes, Mononuclear/physiology , Mice , Molecular Sequence Data , NF-kappa B/physiology , Nitric Oxide Synthase/biosynthesis , Phagocytes/metabolism , Promoter Regions, Genetic/physiology , Recombinant Proteins , Sequence Homology, Nucleic Acid , Transcription, Genetic
11.
J Leukoc Biol ; 59(3): 442-50, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8604025

ABSTRACT

The purpose of this study was to determine whether the female hormones estradiol-l7 beta (E2) and progesterone (P4) influence inducible nitric oxide synthase (iNOS) and the production of nitric oxide (NO) by interferon-gamma(IFN-gamma)-and lipopolysaccharide (LPS)-activated mouse macrophages. Treatment with P4 alone caused a time- and dose-dependent inhibition of NO production by macrophage cell lines (RAW 264.7, J774) and mouse bone marrow culture-derived macrophages as assessed by nitrite accumulation. RAW 264.7 cells transiently transfected with an iNOS gene promoter/luciferase reporter-gene construct that were stimulated with IFN-gamma/LPS in the presence of P4 displayed reduced luciferase activity and NO production. Analysis of RAW 264.7 cells by Northern blot hybridization revealed concurrent P4-mediated reduction in iNOS mRNA. These observations suggest that P4-mediated inhibition of NO may be an important gender-based difference within females and males that relates to macrophage-mediated host defense.


Subject(s)
Macrophage Activation/drug effects , Macrophages/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide/biosynthesis , Progesterone/pharmacology , Animals , Cells, Cultured , Dexamethasone/pharmacology , Estradiol/pharmacology , Gene Expression/drug effects , Glucocorticoids/pharmacology , Hormone Antagonists/pharmacology , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Mice , Mifepristone/pharmacology , Promoter Regions, Genetic , RNA, Messenger/genetics , Receptors, Estrogen/genetics , Receptors, Progesterone/genetics , Transcription, Genetic/drug effects
12.
J Biomed Mater Res ; 30(2): 165-74, 1996 Feb.
Article in English | MEDLINE | ID: mdl-9019480

ABSTRACT

Ion beam technology may be applied in a straightforward fashion to the analysis and modification of biomaterials. For analytical purposes, characterization using megaelectron-volt He2+ ions provides a standardless, nondestructive means for accurately quantifying the composition of material surfaces and the thickness of thin films. In this study, three complementary ion backscattering techniques were utilized to characterize hydroxyapatite (HA) films: Rutherford backscattering spectrometry (RBS) can determine composition and amounts of elements heavier than He; forward recoil elastic spectrometry (FRES) can determine hydrogen content; resonance-enhanced RBS can quantify small amounts of light elements, e.g. carbon, by choosing a particular incident beam energy resulting in excitation of the light element nucleus. At this resonance energy, the scattering cross section greatly increases, improving elemental sensitivity. Sol-gel chemistry was used to synthesize HA films by spin coating and annealing in a rapid thermal processor. Using these techniques, the chemical composition of unfired films was Ca1.63O5.4H1.8C0.24P with a thickness of 3.01 x 10(18) atoms/cm2 and after firing at 800 degrees C as Ca1.66O4.0H0.26C0.09P with a thickness of 2.11 x 10(18) atoms/cm2. This compares favorably to stoichiometric HA, which has a composition of Ca1.67O4.33H0.33P.


Subject(s)
Biocompatible Materials/chemistry , Calcium Phosphates/chemistry , Scattering, Radiation , Crystallography, X-Ray , Helium , Hydrogen/analysis , Hydroxyapatites/chemistry , Ions , Spectrometry, Mass, Secondary Ion , Temperature
13.
Gene ; 158(2): 247-51, 1995 Jun 09.
Article in English | MEDLINE | ID: mdl-7541763

ABSTRACT

The ability of macrophages to kill some kinds of tumor cells is dependent upon the production of the free radical nitric oxide (NO) by the inducible enzyme NO synthase (iNOS; EC 1.14.13.39). Expression of the iNOS gene is induced by lipopolysaccharide (LPS) and augmented by interferon-gamma (IFN-gamma). Two regions of the iNOS promoter are known to regulate induction, a promoter proximal region I (RI) and a more distal region II (RII). Reconfiguration of RI within the iNOS regulatory region revealed its dependence upon native position and orientation for maximal activity, suggesting that it is a core promoter module, and further implicated the putative octamer element as a contributor to promoter activity. RII, however, functioned in a relatively orientation- and position-independent manner. Therefore, it had the characteristics of a classical enhancer element.


Subject(s)
Amino Acid Oxidoreductases/genetics , Enhancer Elements, Genetic/genetics , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Animals , Enzyme Induction , Macrophages/drug effects , Mice , Nitric Oxide Synthase , Promoter Regions, Genetic/genetics
14.
J Bone Miner Res ; 10(3): 359-67, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7785456

ABSTRACT

In the United States, the higher prevalence of osteoporosis and the higher incidence of fractures in whites than in blacks may be attributed to the finding of lower bone density (BD) in both white children and adults. In South Africa, osteoporosis and fractures also occur more frequently in whites than in blacks. Appendicular BD has been found to be similar in black and white children in South Africa, but there is little information available on BD of adults in South Africa. This cross-sectional study aimed to assess changes in BD with age in adult females in South Africa and to assess possible differences in peak BD and in the rate of postmenopausal bone loss between blacks and whites. Data for 180 black and 184 white female nurses aged 20-64 years were analyzed. The distal radius bone density (RBD) was measured by single photon absorptiometry. The lumbar spine bone density (SBD) and the femur bone density (FBD) were measured by dual-energy X-ray absorptiometry. Blacks were shorter than whites (p = 0.0001), and blacks' weight, body mass index, and skinfold thickness increased with age. Peak SBD and RBD were similar in blacks and whites, but peak FBD was higher in blacks (p = 0.0001). This ethnic difference in peak FBD became apparent in the fourth decade. Peak FBD was similar in black and white subjects with normal body mass indices (p = 0.09), but in overweight subjects peak FBD was higher in blacks than in whites (p = 0.0001).(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Black People , Bone Density/physiology , Osteoporosis, Postmenopausal/ethnology , White People , Absorptiometry, Photon , Adult , Age Factors , Body Mass Index , Cohort Studies , Cross-Sectional Studies , Female , Femoral Fractures/epidemiology , Femur/physiology , Humans , Lumbar Vertebrae/physiology , Middle Aged , Nurses , Osteoporosis, Postmenopausal/physiopathology , Radius/physiology , Risk Assessment , South Africa/epidemiology , Spinal Fractures/epidemiology
15.
Gene ; 154(2): 219-23, 1995 Mar 10.
Article in English | MEDLINE | ID: mdl-7890167

ABSTRACT

The purpose of this investigation was to characterize the gene that encodes the receptor for mouse interferon-gamma (IFN-gamma R), including determination of its size, intronic boundaries and its transcription start points (tsp). The mouse IFN-gamma R gene is 22-kb long, with six introns that range in size from approx. 1 to 7 kb. The first six exons encode the extracellular and transmembrane (TM) domains of the protein, while the last exon of about 1 kb encodes most of the intracellular domain. No canonical TATA box can be found in the 5' flanking sequence of the gene, and primer extension analysis indicates multiple tsp. In addition, the gene's 5' promoter region was sequenced to identify candidate responsive elements that might regulate expression of the gene. Among the putative regulatory motifs identified by computer-assisted analysis are multiple SP1 and AP-2 sites, an NF1 and CCAAT box, as well as a potential cyclic AMP-responsive element (CRE).


Subject(s)
Receptors, Interferon/biosynthesis , Receptors, Interferon/genetics , Animals , Base Sequence , Binding Sites , Chromosome Mapping , Cyclic AMP Receptor Protein/metabolism , DNA-Binding Proteins/metabolism , Exons , Genes , Introns , Mice , Molecular Sequence Data , NFI Transcription Factors , Operator Regions, Genetic , Promoter Regions, Genetic , Sp1 Transcription Factor/metabolism , Transcription Factor AP-2 , Transcription Factors/metabolism , Transcription, Genetic , Interferon gamma Receptor
16.
J Leukoc Biol ; 57(1): 27-35, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7530281

ABSTRACT

Nitric oxide (NO), a potent and versatile free radical, is synthesized in macrophages and mast cells as well as in other types of cells by the inducible form of nitric oxide synthase (iNOS). In this study, cells containing iNOS were identified in the uteri of cycling mice by using a rabbit antibody generated to an iNOS-specific peptide. Macrophages were identified in semiserial sections of the same tissues with the monoclonal antibody, F4/80, and mast cells were identified by toluidine blue staining. In tissue sections of uteri obtained from mice in the four stages of the estrous cycle (8 to 11 mice per stage), iNOS immunoreactivity was strongest in diestrus-I uteri and weakest in diestrus-II uteri. Myometrial mast cells and endometrial epithelial cells were prominent locations of iNOS, and specific protein was also present in myometrial smooth muscle and macrophage-like cells in the endometrial stroma. Because cyclic variations suggested regulation of iNOS expression by ovarian steroid hormones, studies were done using ovariectomized mice. Seven days after ovariectomy, immunoreactive iNOS was low but detectable in mast cells and luminal epithelial cells. In the uteri of ovariectomized, estradiol-17 beta (E2)-treated mice, mast cells were iNOS+ after 24 h whereas epithelial cells were negative; the reverse was observed in progesterone (P4)-treated mice. Both mast cells and epithelial cells were iNOS+ in the uteri of mice that had received a combination of E2 + P4. These results indicate that several types of uterine cells produce iNOS and that expression of this enzyme in specific cell lineages is governed by ovarian steroid hormones. The data are consistent with the postulate that NO derived from uterine leukocytes and other types of cells plays a role in uterine cyclicity and preparation for pregnancy.


Subject(s)
Amino Acid Oxidoreductases/analysis , Amino Acid Oxidoreductases/biosynthesis , Estradiol/pharmacology , Progesterone/pharmacology , Uterus/enzymology , Animals , Blotting, Western , Endometrium/cytology , Endometrium/enzymology , Enzyme Induction/drug effects , Estrus/physiology , Female , Histocytochemistry , Macrophages/cytology , Macrophages/enzymology , Mast Cells/cytology , Mast Cells/enzymology , Mice , Myometrium/cytology , Myometrium/enzymology , NADPH Dehydrogenase/analysis , Nitric Oxide Synthase , Uterus/cytology , Uterus/physiology
17.
J Leukoc Biol ; 55(5): 617-25, 1994 May.
Article in English | MEDLINE | ID: mdl-8182339

ABSTRACT

Macrophages and natural killer (NK)-like cells are the major hematopoietic cell populations in the cycling and pregnant mouse uterus and are also found in the embryo. In order to evaluate potential receptivity of these cells to interferon-gamma (IFN-gamma), tissues taken from cycling and pregnant mice were tested for IFN-gamma receptor (IFN-gamma R) mRNA and protein. Macrophages were identified immunohistochemically by using the specific monoclonal antibody F4/80. NK cells were identified by their large size, distinctive intracellular granules, and binding of a monoclonal antibody to the common leukocyte antigen. In cycling uteri, the abundance of IFN-gamma R mRNA relative to an invariant message (glyceraldehyde-3-phosphate dehydrogenase) increased during progression of the hormonally regulated estrous cycle. IFN-gamma R mRNA in situ hybridization signals were slightly higher in macrophage-like than in other types of endometrial stromal cells. In pregnant uteri, the highest proportions of IFN-gamma R mRNA were observed at gestation day (g.d.) 16. Specific message and protein were present in uterine macrophages by g.d. 7 and in NK cells by g.d. 9. IFN-gamma R expression in both lineages remained stable through the balance of pregnancy. In embryos, IFN-gamma R mRNA increased between g.d. 14 and 16. Specific transcripts were present in many cells at g.d. 14, but none were detected in embryonic liver macrophages until g.d. 16. The results of this study suggest relationships between IFN-gamma R expression and ovarian hormones as well as cell maturation and support the postulate that IFN-gamma receptor-ligand interactions may improve the ability of uterine and embryonic hematopoietic cells to perform specific tasks during gestation.


Subject(s)
Embryo, Mammalian/cytology , Gene Expression Regulation/genetics , Hematopoietic Stem Cells/physiology , Pregnancy, Animal/genetics , Receptors, Interferon/genetics , Uterus/cytology , Animals , Antibodies, Monoclonal , Blotting, Northern , Cell Line , Embryo, Mammalian/chemistry , Embryo, Mammalian/ultrastructure , Female , Hematopoietic Stem Cells/chemistry , Hematopoietic Stem Cells/ultrastructure , Immunohistochemistry , In Situ Hybridization , Killer Cells, Natural/chemistry , Killer Cells, Natural/physiology , Killer Cells, Natural/ultrastructure , Macrophages/chemistry , Macrophages/physiology , Macrophages/ultrastructure , Mice , Pregnancy , RNA, Messenger/analysis , RNA, Messenger/genetics , Receptors, Interferon/analysis , Transcription, Genetic , Uterus/chemistry , Uterus/ultrastructure , Interferon gamma Receptor
18.
J Biol Chem ; 269(17): 12773-8, 1994 Apr 29.
Article in English | MEDLINE | ID: mdl-7513694

ABSTRACT

The role of endogenous tumor necrosis factor alpha (TNF-alpha) and interferon-beta (IFN-beta) in lipopolysaccharide (LPS)-induced activation of the inducible nitric-oxide synthase (i-NOS) gene was investigated. By Northern analysis or reverse-transcription polymerase chain reaction, the mouse macrophage cell line (J774) was found to respond to LPS treatment by increased expression of mRNAs specific for TNF-alpha, IFN-beta, and i-NOS with the kinetics unique for each gene. Bioassay of the culture supernatants showed that TNF-alpha and IFN-beta secreted by J774 cells increased from an undetectable level to about 300 and 340 units/ml, respectively, 3-6 h after LPS stimulation. Nitrite concentration was found to increase from 0 to 7.8 and 28.5 microM by 12 and 24 h, respectively, in the culture supernatant of LPS-treated J774 cells. The presence of a neutralizing dose of antibodies against IFN-beta, but not against TNF-alpha, during treatment with either 10 ng or 1 microgram of LPS/ml significantly, but not completely decreased the level of i-NOS-specific mRNA expression and NO production. The incubation of J774 cells with mouse natural IFN-beta itself (up to the level of 1,200 units/ml) did not induce i-NOS-specific mRNA and therefore did not stimulate J774 cells to produce NO. However, natural IFN-beta synergistically augmented the expression of i-NOS mRNA and the production of NO by J774 cells triggered by suboptimal concentrations of LPS (1 to 5 ng/ml). These data thus suggest that endogenous IFN-beta, but not TNF-alpha, produced by LPS-stimulated J774 cells specifically contributes, probably in an auto/paracrine fashion, to the activation of the i-NOS gene expression by LPS.


Subject(s)
Amino Acid Oxidoreductases/genetics , Gene Expression Regulation, Enzymologic/physiology , Interferon-beta/physiology , Lipopolysaccharides/pharmacology , Macrophages/enzymology , Animals , Base Sequence , Cell Line , DNA Primers , Enzyme Induction/genetics , Female , Gene Expression Regulation, Enzymologic/drug effects , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Nitric Oxide/biosynthesis , Nitric Oxide Synthase , Polymerase Chain Reaction , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/genetics
19.
Placenta ; 15(2): 109-21, 1994.
Article in English | MEDLINE | ID: mdl-8008727

ABSTRACT

In order to evaluate the potential of placental cells to bind the multifunctional cytokine, interferon-gamma (IFN-gamma), tissues collected from pregnant Swiss mice were analysed for IFN-gamma receptor (IFN-gamma R) mRNA and protein. Northern blot hybridization studies indicated that the relative abundance of IFN-gamma R mRNA increased as gestation progressed to term. Analysis by in situ hybridization revealed that trophoblast cells first contained high steady state levels of IFN-gamma R mRNA at g.d. 12. At g.d. 12 and 14, transcription was restricted to cells in the spongiotrophoblast region and nests of similar cells in the labyrinthine region. These cells also contained immunoreactive IFN-gamma R protein. By g.d. 18, IFN-gamma R mRNA was clearly detectable in large spongiotrophoblast cells and labyrinthine trophoblast. IFN-gamma R mRNA was low to absent in giant trophoblast cells at all stages of gestation. Specific mRNA was present in parietal and visceral yolk sac cells by g.d. 14. Thus, expression of the IFN-gamma R gene in mouse placental cells is influenced by stage of gestation, cell lineage and state of differentiation. Whether or not these cells respond vigorously to IFN-gamma with induction of antiviral proteins, increased MHC class I antigens and growth modulation may therefore be determined by their expression of specific receptors for this pluripotent cytokine.


Subject(s)
Gene Expression , Interferon-gamma , Placenta/metabolism , Receptors, Interferon/genetics , Trophoblasts/metabolism , Allantois/metabolism , Animals , Blotting, Northern , Chorion/metabolism , Female , Immunoenzyme Techniques , In Situ Hybridization , Mice , Pregnancy , RNA, Messenger/metabolism , Receptors, Interferon/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...