Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
3.
Biol Reprod ; 87(4): 81, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22875907

ABSTRACT

Endometrial decidualization, a process essential for blastocyst implantation in species with hemochorial placentation, is accompanied by an enormous but transient influx of natural killer (NK) cells. Mouse uterine NK (uNK) cell subsets have been defined by diameter and cytoplasmic granule number, reflecting stage of maturity, and by histochemical reactivity with Periodic Acid Schiff (PAS) reagent with or without co-reactivity with Dolichos biflorus agglutinin (DBA) lectin. We asked whether DBA- and DBA+ mouse uNK cells were equivalent using quantitative RT-PCR analyses of flow-separated, midpregnancy (Gestation Day [gd] 10) cells and immunohistochemistry. CD3E (CD3)-IL2RB (CD122)+DBA cells were identified as the dominant Ifng transcript source. Skewed IFNG production by uNK cell subsets was confirmed by analysis of uNK cells from eYFP-tagged IFNG-reporter mice. In contrast, CD3E-IL2RB+DBA+ uNK cells expressed genes compatible with significantly greater potential for IL22 synthesis, angiogenesis, and participation in regulation mediated by the renin-angiotensin system (RAS). CD3E-IL2RB+DBA+ cells were further divided into VEGFA+ and VEGFA- subsets. CD3E-IL2RB+DBA+ uNK cells but not CD3E-IL2RB+DBA- uNK cells arose from circulating, bone marrow-derived progenitor cells by gd6. These findings indicate the heterogeneous nature of mouse uNK cells and suggest that studies using only DBA+ uNK cells will give biased data that does not fully represent the uNK cell population.


Subject(s)
Gene Expression , Killer Cells, Natural/classification , Killer Cells, Natural/metabolism , Plant Lectins/metabolism , Uterus/immunology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/physiology , Female , Gene Expression/immunology , Killer Cells, Natural/drug effects , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Plant Lectins/pharmacology , Protein Binding , Stem Cells/cytology , Stem Cells/metabolism , Stem Cells/physiology , Uterus/drug effects , Uterus/metabolism
4.
Proc Natl Acad Sci U S A ; 108(10): 4012-7, 2011 Mar 08.
Article in English | MEDLINE | ID: mdl-21300875

ABSTRACT

The mammalian fetus represents a semiallograft within the maternal uterus yet is not rejected. This situation is particularly pronounced in species with a hemochorial type of placentation, such as humans and rodents, where maternal tissues and blood are in direct contact with fetal trophoblast and thus potentially with paternal antigens. The main polymorphic antigens responsible for graft rejection are MHC antigens. In humans the trophoblast cells invading into the decidua have a unique pattern of MHC class I expression characterized by both classical (HLA-C) and nonclassical (HLA-G and HLA-E) molecules. Whether such an unusual MHC repertoire on the surface of trophoblast is a conserved feature between species with hemochorial placentation has not been resolved. Here we demonstrate, using a range of methods, that C57BL/6 mouse trophoblast predominantly expresses only one MHC class I antigen, H2-K, at the cell surface of giant cells but lacks expression of nonclassical MHC molecules. Antigenic disparity between parental MHCs affects trophoblast-induced transformation of the uterine vasculature and, consequently, placental and fetal gowth. Maternal uterine blood vessels were more dilated, allowing for increased blood supply, in certain combinations of maternal and paternal MHC haplotypes, and these allogeneic fetuses and placentas were heavier at term compared with syngeneic controls. Thus, maternal-fetal immune interactions are instrumental to optimize reproductive success. This cross-talk has important implications for human disorders of pregnancy, such as preeclampsia and fetal growth restriction.


Subject(s)
Fetal Development/genetics , Genomic Imprinting , Major Histocompatibility Complex/genetics , Trophoblasts/immunology , Uterus/blood supply , Animals , Cell Separation , Female , Flow Cytometry , Immunohistochemistry , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
5.
Methods Mol Biol ; 612: 465-503, 2010.
Article in English | MEDLINE | ID: mdl-20033660

ABSTRACT

The term uterine natural killer (uNK) cell is applied in mice to an abundant but transient NK cell population that undergoes unique, terminal differentiation within embryo implantation sites during endometrial decidualization and pregnancy. In mice, decidualization is induced by attachment and implantation of hatched, blastocyst-stage embryos. Within each implantation site, uNK cells proliferate and rapidly differentiate into highly restricted regions called decidua basalis and the mesometrial lymphoid aggregate of pregnancy (MLAp). uNK cells begin to die within healthy decidua basalis by day 8 of the 19-20 day pregnancy of mice. By gestation day 12, uNK cell numbers have peaked and most uNK cells show in situ nuclear fragmentation indicative of disintegration. Morphological studies (standard histology, ultrastructure, immunohistochemistry, in situ hybridization, and RNA analyses from laser capture microdissected uNK cells) have provided most of the current understanding regarding this cell lineage. These approaches identified the special angiogenic properties of uNK cells and their regulatory relationships with normal physiological changes to the uterine (endometrial) arterial tree that accompany successful pregnancy. This chapter highlights key information needed for successful dissection of the dynamically changing decidua basalis that is enriched in uNK cells and special morphological procedures used for uNK cell study. Preparation of viable mouse uNK cell suspensions is difficult but can be achieved. This chapter includes techniques for isolation of uterine leukocyte suspensions and their enrichment for uNK cells that permit immediate downstream applications such as culture, isolation of high quality RNA, or flow cytometry.


Subject(s)
Cytological Techniques/methods , Killer Cells, Natural/cytology , Uterus/cytology , Animals , Cell Culture Techniques , Cell Extracts , Female , Flow Cytometry , Freezing , Humans , Killer Cells, Natural/metabolism , Killer Cells, Natural/ultrastructure , Magnetics , Male , Mice , Microscopy, Electron , Nucleic Acid Amplification Techniques , Pregnancy , RNA/genetics , RNA/isolation & purification , Staining and Labeling , Tissue Fixation
6.
Proc Natl Acad Sci U S A ; 106(14): 5795-800, 2009 Apr 07.
Article in English | MEDLINE | ID: mdl-19297623

ABSTRACT

The mechanisms that regulate NK cell trafficking are unclear. Phosphoinositide-3 kinases (PI3K) control cell motility and the p110gamma and p110delta isoforms are mostly expressed in leukocytes, where they transduce signals downstream of G protein coupled receptors (GPCR) or tyrosine kinase receptors, respectively. Here, we set out to determine the relative contribution of p110gamma and p110delta to NK cell migration in mice. Using a combination of single-cell imaging analysis of transgenic cells reporting on PI3K activity in real time and small molecule inhibitors of p110gamma and p110delta, we show here that the tyrosine-kinase coupled p110delta is linked to GPCR signaling and, depending on the GPCR, may even be preferentially activated over p110gamma. Using gene-targeted mice, we showed that both isoforms were essential for NK cell chemotaxis to CXCL12 and to CCL3 and, in vivo, for normal NK cell migration during pregnancy and to the inflamed peritoneum. By contrast, only p110delta was indispensable for chemotaxis to S1P and CXCL10 and for NK cell distribution throughout lymphoid and nonlymphoid tissues and for extravasation to tumors. These results implicate p110delta downstream of GPCRs in NK cells and highlight its nonredundant role as a key regulator of NK cell trafficking in health and disease.


Subject(s)
Chemotaxis, Leukocyte/immunology , Killer Cells, Natural/immunology , Phosphatidylinositol 3-Kinases/immunology , Animals , Chemokine CCL3 , Chemokine CXCL10 , Chemokine CXCL12 , Class I Phosphatidylinositol 3-Kinases , Disease Models, Animal , Female , Inflammation , Liver Neoplasms , Mice , Mice, Knockout , Peritoneum/pathology , Pregnancy , Receptors, G-Protein-Coupled/metabolism
7.
J Immunol ; 181(9): 6140-7, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18941204

ABSTRACT

Uterine NK (uNK) cells are a prominent feature of the uterine mucosa and regulate placentation. NK cell activity is regulated by a balance of activating and inhibitory receptors, however the receptor repertoire of mouse uNK cells is unknown. We describe herein two distinct subsets of CD3(-)CD122(+) NK cells in the mouse uterus (comprising decidua and mesometrial lymphoid aggregate of pregnancy) at mid-gestation: a small subset indistinguishable from peripheral NK cells, and a larger subset that expresses NKp46 and Ly49 receptors, but not NK1.1 or DX5. This larger subset reacts with Dolichus biflores agglutinin, a marker of uNK cells in the mouse, and is adjacent to the invading trophoblast. By multiparametric analysis we show that the phenotype of uNK cells is unique and unprecedented in terms of adhesion, activation, and MHC binding potential. Thus, the Ly49 repertoire and the expression of other differentiation markers strikingly distinguish uNK cells from peripheral NK cells, suggesting that a selection process shapes the receptor repertoire of mouse uNK cells.


Subject(s)
Immunophenotyping , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Receptors, Immunologic/biosynthesis , Uterus/immunology , Uterus/metabolism , Animals , Antigens, Ly , CD3 Complex/metabolism , Decidua/cytology , Decidua/immunology , Decidua/metabolism , Female , Gene Expression Regulation/immunology , Integrin alpha2/metabolism , Interleukin-2 Receptor beta Subunit/biosynthesis , Interleukin-2 Receptor beta Subunit/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , NK Cell Lectin-Like Receptor Subfamily A/biosynthesis , NK Cell Lectin-Like Receptor Subfamily A/genetics , NK Cell Lectin-Like Receptor Subfamily B/deficiency , Pregnancy , Receptors, Immunologic/genetics , Uterus/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...