Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Drug Metab Dispos ; 34(6): 895-900, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16510538

ABSTRACT

Diallyl sulfone (DASO2) is a garlic derivative formed during cooking or after ingestion. Bioactivation of DASO2 in murine lung and liver results in formation of an epoxide that inactivates CYP2E1 and significantly decreases cytochrome P450 and heme levels. In this study, we tested the hypothesis that DASO2 metabolism leads to production of the heme adduct, N-alkylprotoporphyrin IX (N-alkylPP). Formation of N-alkylPP in vivo and in vitro was determined by spectrophotometric and fluorometric methods, respectively. In in vivo studies, N-alkylPP was generated in the livers of male and female mice treated with DASO2, but was not detectable in the lungs of DASO2-treated mice. In in vitro studies, rates of formation of N-alkylPP in liver and lung microsomes incubated with DASO2 and NADPH were dependent on time and protein concentrations, but were negligible in control incubations performed in the absence of NADPH or DASO2 or with boiled microsomes. The rates of N-alkylPP formation generated in murine liver were higher than those in either murine lung or human liver. Kinetic analysis revealed that murine liver microsomes metabolized DASO2 to N-alkylPP with higher affinity and catalytic efficiency than did murine lung or human liver microsomes. Recombinant rat CYP2E1 also metabolized DASO2 to N-alkylPP; however, rates of formation of the heme adduct was minimal in incubations of recombinant human CYP2E1 with DASO2. These findings demonstrated that the N-alkylPP adduct was produced via metabolism of DASO2 in murine liver and lung microsomes, in human liver microsomes, in recombinant CYP2E1, and in vivo in murine liver.


Subject(s)
Allyl Compounds/metabolism , Liver/metabolism , Lung/metabolism , Protoporphyrins/metabolism , Sulfones/metabolism , Alkylation , Allyl Compounds/administration & dosage , Animals , Cytochrome P-450 CYP2E1/genetics , Cytochrome P-450 CYP2E1/metabolism , Female , Humans , Kinetics , Male , Mice , Microsomes, Liver/metabolism , Protoporphyrins/biosynthesis , Protoporphyrins/chemistry , Recombinant Proteins/metabolism , Sex Factors , Sulfones/administration & dosage
2.
J Immunol ; 176(1): 148-56, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16365405

ABSTRACT

Prenatal mortality reaching 30% occurs during the first weeks of gestation in commercial swine. Mechanisms for this are unknown although poor uterine blood supply has been postulated. In other species, vascular endothelial growth factor, hypoxia-inducible factor 1-alpha, and IFN-gamma regulate gestational endometrial angiogenesis. Vascular endothelial growth factor and hypoxia-inducible factor 1-alpha are also important for placental angiogenesis while trophoblastic expression of Fas ligand is thought to protect conceptuses against immune-mediated pregnancy loss. In this study, we document dynamic, peri-implantation differences in transcription of genes for angiogenesis, cytokine production, and apoptosis regulation in the endometrium, and laser capture microdissected endometrial lymphocytes and trophoblasts associated with healthy or viable but arresting porcine fetuses. In healthy implantation sites, endometrial gene expression levels differed between anatomic subregions and endometrial lymphocytes showed much greater transcription of angiogenic genes than trophoblasts. In arresting fetal sites, uterine lymphocytes had no angiogenic gene transcription and showed rapid elevation in transcription of proinflammatory cytokines Fas and Fas ligand while trophoblasts showed elevated transcription of IFN-gamma and Fas. This model of experimentally accessible spontaneous fetal loss, involving blocked maternal angiogenesis, should prove valuable for further investigations of peri-implantation failure of normally conceived and surgically transferred embryos in many species, including the human.


Subject(s)
Abortion, Spontaneous/genetics , Embryo Implantation/genetics , Endometrium/physiology , Lymphocytes/metabolism , Trophoblasts/metabolism , Abortion, Spontaneous/immunology , Animals , Apoptosis/immunology , Cytokines/immunology , Cytokines/metabolism , DNA Primers , Embryo Implantation/immunology , Endometrium/blood supply , Female , Gene Expression , Lasers , Lymphocytes/immunology , Microdissection , Molecular Sequence Data , Neovascularization, Physiologic/physiology , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction , Swine , Time Factors , Transcription, Genetic , Trophoblasts/immunology
3.
J Leukoc Biol ; 78(6): 1347-55, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16204645

ABSTRACT

During human and rodent uterine decidualization, transient but abundant numbers of uterine natural killer (uNK) cells appear, proliferate, and differentiate. uNK cells share features with peripheral NK cells but are specialized to promote interferon-gamma (IFN-gamma)-mediated, pregnancy-associated, structural changes in maternal placental arteries. In CD8+ T cells and NK cells, the transcription factors T-bet and eomesodermin (Eomes) regulate maturation and effector functions, including IFN-gamma production. No studies are reported for uNK cells. Implantation sites in T-bet null mice, which have a defect in NK cell maturation, had uNK cells normal in morphology and number and normally modified spiral arteries. As Eomes null mice are not viable, real-time polymerase chain reaction comparisons between C57Bl/6J (B6) and alymphoid (Rag2(0/0)gammac0/0) mice were used to assess uNK cell expression of T-bet, Eomes, and the target genes IFN-gamma, granzyme A, and perforin. Gestation dated (gd) uterine tissues (mixed cell composition) and 200 morphologically homogeneous, laser-capture, microdissected uNK cells of different maturation stages were used. In uterus, Eomes transcripts greatly outnumbered those of T-bet, whether donors were nonpregnant or pregnant, and increased to gd10. In uNK cells, transcripts for T-bet, Eomes, and IFN-gamma were most abundant in mature stage cells, and transcripts for granzyme A and perforin were lower at this stage than in immature or senescent cells. Thus, Eomes dominance to T-bet discriminates regulation of the uNK cell subset from that observed for peripheral NK cells.


Subject(s)
Cell Differentiation/immunology , Killer Cells, Natural/immunology , T-Box Domain Proteins/genetics , Transcription Factors/genetics , Uterus/immunology , Animals , Estrous Cycle/genetics , Estrous Cycle/immunology , Estrous Cycle/metabolism , Female , Granzymes , Interferon-gamma/genetics , Killer Cells, Natural/metabolism , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Perforin , Pore Forming Cytotoxic Proteins , Pregnancy , Regulatory Elements, Transcriptional/genetics , Regulatory Elements, Transcriptional/physiology , Serine Endopeptidases/genetics , T-Box Domain Proteins/immunology , Transcription Factors/immunology , Transcriptional Activation/genetics , Uterus/cytology
4.
J Immunol ; 171(6): 2937-44, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-12960317

ABSTRACT

In mouse and human, precursors of NK cell lineage home to decidualizing uteri. To assess the requirement for IL-15, an essential cytokine for NK differentiation in lymphoid tissue, on uterine NK (uNK) cell differentiation, implantation sites from IL-15(-/-) mice were analyzed histologically. IL-15(-/-) implantation sites had no uNK cells, no spiral-artery modification, and lacked the decidual integrity found in normal mice. IL-15(-/-) recipients of C57BL/6 marrow displayed similar pathology. However, implantation sites from recombination-activating gene-2(-/-)gamma(c)(-/-) (alymphoid) recipients of IL-15(-/-) marrow showed normal uNK cells, modified spiral arteries, and well-developed decidua basalis. Deletion of the IFN-regulatory factor (IRF)-1, but not IRF-2 (factors important in peripheral NK cell differentiation) limited but did not prevent uNK cell development. In situ hybridization localized IRF-1 largely to placental trophoblast cells. IRF-1(-/-) marrow transplanted into recombination-activating gene-2(-/-)gamma(c)(-/-) displayed competence for full uNK cell differentiation. IL-15 mRNA expression at implantation sites of IRF-1(-/-) and C57BL/6 was similar, suggesting that, unlike in bone marrow and spleen, IRF-1 does not regulate IL-15 in the pregnant uterus. Terminal differentiation of uNK cells was not promoted in pregnant IRF-1(-/-) mice by 5-day infusion of murine rIL-15, suggesting that IRF-1 deficiency rather than IL-15 deficiency limits uNK cell differentiation in these mice. Further, IRF-1 regulates placental growth, birth weight, and postnatal growth of offspring. These studies indicate that uNK cell development and maturation share some aspects with NK cell development in other tissues, but also display distinctive tissue-specific regulation.


Subject(s)
DNA-Binding Proteins/physiology , Interferon-gamma/physiology , Interleukin-15/physiology , Killer Cells, Natural/immunology , Phosphoproteins/physiology , Pregnancy Proteins/physiology , Repressor Proteins , Transcription Factors , Uterus/cytology , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Differentiation/immunology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Embryo Implantation/drug effects , Embryo Implantation/genetics , Embryo Implantation/immunology , Female , Interferon Regulatory Factor-1 , Interferon Regulatory Factor-2 , Interleukin-15/biosynthesis , Interleukin-15/deficiency , Interleukin-15/genetics , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphoproteins/deficiency , Phosphoproteins/genetics , Pregnancy , Pregnancy Proteins/biosynthesis , Pregnancy Proteins/deficiency , Pregnancy Proteins/genetics , Recombinant Proteins/administration & dosage , Stem Cells/cytology , Stem Cells/immunology , Stem Cells/metabolism , Uterus/immunology , Uterus/metabolism
5.
J Reprod Immunol ; 59(2): 175-91, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12896821

ABSTRACT

Virgin adult C57Bl/6J mouse uterus contains a population of small, non-granulated Natural Killer (NK) cells with balanced expression of NK cell activating and inhibiting LY49 receptors. Coincident with blastocyst implantation and decidualization, uterine (u)NK cells become activated. The surface glycoslyation of uNK changes, the cells proliferate and they induce production of interferon (IFN)gamma, perforin, serine esterases and other molecules, including angiogenic factors. Mouse strains genetically ablated in uNK cells fail to undergo modification of spiral artery segments that branch from the uterine artery and feed into the placenta and these mice do not sustain a robust decidualization response. IFN-gamma is thought, from bone marrow transplantation and therapeutic studies, to be the key uNK-cell derived mediator regulating gene expression in vascular and decidual tissues. Here, we review recent studies showing that IL-15 is the critical cytokine controlling uNK cell differentiation and that uNK cells are activated by either IL-12 or IL-18 and by other factors when both IL-12 and IL-18 are genetically absent from implantation sites. We address possible roles of the IFN-gamma regulated gene alpha2-macroglobulin (alpha2-M) in regulation of the position of fetal trophoblast within the walls of the spiral arteries, and we discuss approaches that have been successful in evaluating mechanisms involved in homing of mouse uNK cell precursors to the uterus. These approaches maybe applicable to studies in women. Our studies show that complex immuno-physiological events contribute to spiral artery modification by mid-gestation in mice.


Subject(s)
Decidua/blood supply , Killer Cells, Natural/immunology , Lymphocyte Activation , Uterus/immunology , Adult , Animals , Antigens, Ly/genetics , Antigens, Ly/immunology , Arteries/physiology , Female , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukins/immunology , Lectins, C-Type , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy , Receptors, NK Cell Lectin-Like , Trophoblasts/physiology , Uterus/cytology , alpha-Macroglobulins/genetics , alpha-Macroglobulins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...