Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters










Publication year range
1.
Placenta ; 36 Suppl 1: S5-10, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25703592

ABSTRACT

Workshops are an important part of the IFPA annual meeting as they allow for discussion of specialized topics. At IFPA meeting 2014 there were six themed workshops, five of which are summarized in this report. These workshops related to various aspects of placental biology but collectively covered areas of animal models, xenobiotics, pathological biomarkers, genetics and epigenetics, and stillbirth and fetal growth restriction.


Subject(s)
Biomarkers/analysis , Disease Models, Animal , Placenta/drug effects , Placenta/metabolism , Pregnancy Complications/pathology , Xenobiotics/toxicity , Animals , Epigenesis, Genetic/physiology , Female , Fetal Growth Retardation/genetics , Fetal Growth Retardation/pathology , Humans , Maternal Exposure/adverse effects , Placenta Diseases/chemically induced , Placenta Diseases/genetics , Placenta Diseases/metabolism , Pregnancy , Pregnancy Complications/diagnosis , Stillbirth
2.
J Endocrinol ; 216(3): R33-45, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23291503

ABSTRACT

The development of the placenta is imperative for successful pregnancy establishment, yet the earliest differentiation events of the blastocyst-derived trophectoderm that forms the placenta remain difficult to study in humans. Human embryonic stem cells (hESC) display a unique ability to form trophoblast cells when induced to differentiate either by the addition of exogenous BMP4 or by the formation of cellular aggregates called embryoid bodies. While mouse trophoblast stem cells (TSC) have been isolated from blastocyst outgrowths, mouse ESC do not spontaneously differentiate into trophoblast cells. In this review, we focus on addressing the similarities and differences between mouse TSC differentiation and hESC-derived trophoblast differentiation. We discuss the functional and mechanistic diversity that is found in different species models. Of central importance are the unique signaling events that trigger downstream gene expression that create specific cellular fate decisions. We support the idea that we must understand the nuances that hESC differentiation models display so that investigators can choose the appropriate model system to fit experimental needs.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Signal Transduction/physiology , Trophoblasts/cytology , Animals , Embryonic Stem Cells/physiology , Female , Humans , Mice , Placenta/cytology , Placenta/physiology , Pregnancy , Trophoblasts/physiology
3.
Placenta ; 34 Suppl: S56-61, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23261342

ABSTRACT

The human embryo is not a feasible experimental system for the detailed study of implantation and early placentation, so surrogate systems have been sought for investigating the determination of the trophectoderm lineage, its differentiation into trophoblasts of the early implantation site, and subsequently the morphogenesis of the definitive placenta. An alternative to the use of embryos for studying early placental development was revealed by work with human embryonic stem cells (hESC), demonstrating BMP2/4-stimulated trophoblast differentiation, and spontaneous formation from embryoid bodies (EBs). These cells display a trophoblastic transcriptome, as well as a placental protein and steroid hormone secretory profile, and invasive and chemotactic behavior resembling human placental trophoblasts. With EB-derived trophoblasts, two-dimensional and three-dimensional paradigms and other modifications of the culture environment, including extracellular matrix and aggregation with placental fibroblasts, impact on trophoblast differentiation. Recent studies have questioned the identity of the trophoblasts directed by BMP treatment of hESC, and careful attention to culture conditions is needed to interpret different results among research groups. Although the precise placental counterpart of the hESC-derived trophoblast remains unclear, hESC-derived trophoblasts remain an intriguing platform for modeling early implantation.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Trophoblasts/cytology , Animals , Embryoid Bodies/physiology , Female , Humans , Pregnancy , Primates
4.
Placenta ; 34 Suppl: S3-5, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23206905

ABSTRACT

Workshops are an important part of the IFPA annual meeting as they allow for discussion of specialized topics. At IFPA meeting 2012 there were twelve themed workshops, three of which are summarized in this report. These workshops related to various aspects of placental biology but collectively covered areas of models and technical issues involved in placenta research: 1) comparative placentation and animal models; 2) advanced techniques in placental histopathology; 3) human pluripotent stem cells as a model for trophoblast differentiation.


Subject(s)
Cell Differentiation/physiology , Models, Animal , Placenta/pathology , Placentation/physiology , Pluripotent Stem Cells/physiology , Trophoblasts/physiology , Animals , Female , Humans , Placenta/cytology , Pregnancy
6.
Placenta ; 34 Suppl: S6-10, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23253784

ABSTRACT

Workshops are an important part of the IFPA annual meeting as they allow for discussion of specialized topics. At IFPA meeting 2012 there were twelve themed workshops, four of which are summarized in this report. These workshops related to various aspects of placental biology: 1) epigenetics and imprinting in the placenta; 2) growth factors and villous trophoblast differentiation; 3) role of the placenta in regulating fetal exposure to xenobiotics during pregnancy; 4) infection and the placenta.


Subject(s)
Epigenesis, Genetic/physiology , Genomic Imprinting/physiology , Intercellular Signaling Peptides and Proteins/physiology , Placenta/physiology , Pregnancy Complications, Infectious/physiopathology , Prenatal Exposure Delayed Effects/chemically induced , Trophoblasts/physiology , Xenobiotics/adverse effects , Cell Differentiation/physiology , Female , Humans , Placenta/drug effects , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Prenatal Exposure Delayed Effects/physiopathology
7.
Placenta ; 29(1): 58-70, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17996936

ABSTRACT

The aim of this study was to characterize the expression of the rhesus HLA-E ortholog Mamu-E, particularly at the maternal-fetal interface. Mamu-E expression was confirmed by locus-specific RT-PCR in the placenta as well as in peripheral blood mononuclear cells (PBMC) and other organs. We evaluated the utility of antibodies recognizing HLA-E (MEM-E/06 against native HLA-E, MEM-E/02 against denatured HLA-E) to detect Mamu-E by flow cytometry/immunofluorescence, Western blot, and immunohistochemistry (IHC). Western blot analysis of cells and selected transfectants confirmed the recognition of Mamu-E but not Mamu-AG by antibodies MEM-E/06 and HC10 but not MEM-E/02. Immunohistochemical staining of frozen sections of rhesus placenta with the MEM-E/06 antibody demonstrated expression in most populations of rhesus monkey trophoblast cells, including villous cytotrophoblasts (strong positive staining), apical membrane of syncytiotrophoblasts (light to moderate staining) and extravillous cytotrophoblasts (moderate to strong staining, especially endovascular trophoblasts in early pregnancy). Expression was not trophoblast cell-specific, especially at term, when endothelial cells in both the chorionic plate and placental villi showed strong staining for Mamu-E. Staining of rhesus extravillous trophoblast cells suggested the co-expression of Mamu-E and Mamu-AG (the rhesus HLA-G homolog) on these cells. MEM-E/06 was shown also to react with differentiating rhesus placental syncytiotrophoblasts in primary culture, detecting intracellular and weak surface expression of Mamu-E. We conclude that the gestation-dependent co-expression of Mamu-E with Mamu-AG in villous and extravillous trophoblast cells suggests important and perhaps complementary but distinct roles of these two non-classical MHC class I loci in pregnancy at the maternal-fetal interface. In addition, the MEM-E/06 antibody will be useful for the detection of Mamu-E at the maternal-fetal interface in the rhesus monkey.


Subject(s)
HLA Antigens/metabolism , Histocompatibility Antigens Class I/metabolism , Placenta/immunology , Animals , Female , HLA Antigens/analysis , Histocompatibility Antigens Class I/analysis , Macaca mulatta/immunology , Models, Animal , Pregnancy , Recombinant Proteins/analysis , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , HLA-E Antigens
8.
Placenta ; 28(5-6): 383-9, 2007.
Article in English | MEDLINE | ID: mdl-16797695

ABSTRACT

Trophoblast cell lines are important research tools used as a surrogate for primary trophoblast cells in the study of placental function. Because the cellular origins of transformed trophoblasts are likely to be diverse, it would be of value to understand the unique and shared phenotypes of the cells on a global scale. We have compared two widely used cell lines, BeWo and JEG3, by microarray analysis in order to identify differentially expressed genes. Results indicated that approximately 2700 genes were differentially expressed between the cell lines, with principal differences observed in the biological processes of response to stress, cell adhesion, signal transduction, and protein and nucleobase metabolisms. These data suggest that BeWo and JEG3 cell lines, and perhaps other trophoblast cell lines, are sufficiently dissimilar from each other such that they will be differentially suited for specific experimental paradigms.


Subject(s)
Gene Expression Regulation, Developmental , Oligonucleotide Array Sequence Analysis , Trophoblasts/cytology , Trophoblasts/physiology , Cell Line , DNA Primers , Female , Humans , Integrins/genetics , Placenta/cytology , Placenta/physiology , Polymerase Chain Reaction , Pregnancy , Proteins/genetics , Transcription, Genetic
9.
Tissue Antigens ; 67(4): 331-7, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16634871

ABSTRACT

Leukocyte Ig-like receptors (LILRs) are a family of receptors that have inhibitory and activating functions and widely expressed by lymphoid and myeloid cells. Here we report the identification of the rhesus monkey LILRs by screening of rhesus spleen and decidua cDNA libraries and RT-PCR cloning. We obtained eight different full-length clones with structural and functional diversity similar to human LILRs, including LILRs with immunoreceptor tyrosine-based inhibitory motifs, LILRs with truncated cytoplasmic tails containing positively charged arginine residues in the transmembrane domain, and putative soluble receptors lacking transmembrane or cytoplasmic domains. Characterization of rhesus LILRs will facilitate use of this non-human primate model for the study of the functional role(s) of LILRs, including immune regulation through interaction with non-classical MHC class I molecules.


Subject(s)
Macaca mulatta/genetics , Receptors, Immunologic/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Decidua/metabolism , Female , Gene Library , Leukocytes/metabolism , Molecular Sequence Data , Multigene Family , Phylogeny , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Spleen/metabolism
10.
Placenta ; 27(1): 11-21, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16310033

ABSTRACT

We performed immunohistochemical analysis of DC-SIGN expression at the maternal-fetal interface at different stages of pregnancy in the rhesus monkey. Natural killer cells, monocytes and macrophages were observed in the nonpregnant endometrium, particularly in the luteal phase, and were increased in pregnant endometrium. No DC-SIGN+ cells were observed in the nonpregnant uterus. We observed decidual DC-SIGN+ cells within 1 week of implantation, and they increased in number during the first 5 weeks of gestation. DC-SIGN+ cells showed a clear differential distribution in the decidua in the first 2 weeks of pregnancy, being found only adjacent to the implantation site, in marked contrast to the widespread distribution of CD68+ macrophages and CD56+ NK cells throughout the endometrium. DC-SIGN+ cells also showed a more dendritic morphology than the general CD68+ cell population, and analysis of serial sections indicated an overlapping but not identical localization of these markers. Mature dendritic cells could not be detected as judged by total absence of immunostaining for CD83, CD86, DEC-205, or CD1a. DC-SIGN+ cells were defined as MHC class II+ and CD14+ by flow cytometry. We conclude that DC-SIGN expression is an early response by the primate maternal immune system to the implanting embryo. The selectively distributed population of DC-SIGN+ decidual leukocytes may represent a morphologically and phenotypically distinct subpopulation of decidual macrophages of early pregnancy that could contribute to the establishment of maternal-fetal immune tolerance.


Subject(s)
Cell Adhesion Molecules/metabolism , Gene Expression Regulation , Lectins, C-Type/metabolism , Macaca mulatta/physiology , Maternal-Fetal Exchange/physiology , Pregnancy/physiology , Receptors, Cell Surface/metabolism , Animals , Antigens, CD/metabolism , Biomarkers/analysis , Female , Fetus/physiology , Immunohistochemistry
11.
J Clin Endocrinol Metab ; 89(11): 5837-46, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15531550

ABSTRACT

A novel cDNA was cloned from human endometrium, matching a human gene with the interim name KIAA1463. An mRNA identified by 5'-rapid amplification of cDNA ends was found to be 3349 nt in length. PCR analysis also identified another transcript of 6626 nt, with an open reading frame encoding a 900 amino acid protein. A fold recognition program identified similarity to firefly luciferase containing an AMP-binding motif; hence, we refer to the predicted protein as the AMP binding/luciferase-like protein (ALLP). ALLP mRNA and protein were expressed throughout the female reproductive tract with the highest levels found in the ovary and uterus. In situ hybridization and immunohistochemistry showed predominant localization of the ALLP mRNA/protein in endometrial glandular epithelium and within the theca and granulosa cells in the ovary. In the endometrium expression of ALLP, mRNA and protein were higher during d 16-21 of the secretory phase of the cycle. Western blot analysis showed decreased expression of ALLP in the postmenopausal endometrium, and hormone replacement therapy increased the expression of ALLP. Endometrial adenocarcinoma cell lines expressed more ALLP, compared with cultured primary endometrial cells or normal endometrial tissue. The ubiquitous expression of ALLP in reproductive and nonreproductive tissues suggests that this protein, which is probably regulated by ovarian steroids, plays an important metabolic role and may be involved in such processes as implantation and tumorigenesis.


Subject(s)
Adenosine Monophosphate/metabolism , Carrier Proteins/analysis , Genitalia, Female/chemistry , Luciferases/analysis , Amino Acid Sequence , Base Sequence , Carrier Proteins/chemistry , Endometrium/chemistry , Female , Humans , Luciferases/chemistry , Menstrual Cycle , Molecular Sequence Data , RNA, Messenger/analysis
12.
Placenta ; 25(4): 297-307, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15028422

ABSTRACT

The distribution of uterine leukocytes during the periimplantation period cannot be readily evaluated in human pregnancy. Using immunohistochemistry we examined the distribution of macrophages, natural killer (NK) cells, and T cells in the non-pregnant endometrium and in the decidua at early stages of implantation and pregnancy in the rhesus monkey. CD68+ macrophages, CD56+ lymphocytes and CD3+ T cells were present in the proliferative and secretory endometrium. The number of macrophages and CD56+ lymphocytes dramatically increased at implantation (day 14-15 of pregnancy) and continued to be high in early pregnancy decidua. Macrophages were conspicuously more numerous in proximity to implantation site (decidua basalis) as compared to sites peripheral to the developing placenta (decidua parietalis), and were found in close association with cytotrophoblasts adjacent to the decidua, as well as around arteries invaded by extravillous cytotrophoblasts. In contrast to macrophages, CD56+ lymphocytes were more evenly distributed throughout the decidua. Few CD3+ T cells were seen in pregnancy, being scattered in the endometrial stroma with occasional aggregate formation. The distribution of uterine leukocytes vis-à-vis trophoblasts at the rhesus monkey implantation site and in early pregnancy suggests different roles for macrophages and uterine NK cells in the response to trophoblast invasion.


Subject(s)
Endometrium/cytology , Killer Cells, Natural/cytology , Macaca mulatta/physiology , Macrophages/cytology , Animals , Antigens, CD/metabolism , Endometrium/metabolism , Female , Immunoenzyme Techniques , Killer Cells, Natural/metabolism , Macrophages/metabolism , Pregnancy , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Trophoblasts/cytology , Trophoblasts/metabolism
13.
J Soc Gynecol Investig ; 11(1): 36-41, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14706681

ABSTRACT

OBJECTIVE: Type I diabetes mellitus during pregnancy is associated with dysregulation of the oxygen and glucose metabolic pathways, both of which affect placental villous growth and function. Alteration of placental development in women with diabetes may contribute to the increased risk of preeclampsia, macrosomia, or fetal growth restriction. METHODS: To evaluate placental growth in the setting of maternal diabetes, immunohistochemical techniques were used to examine fibroblast growth factor-2 (FGF-2) expression, cell proliferation (Ki67), and apoptosis (Apo-Tag) in placentas from diabetic and nondiabetic patients. RESULTS: Immunostaining for FGF-2 in placentas from diabetic women demonstrated an increase in intensity within the villous stroma and syncytiotrophoblast (P<.05). Associated with these changes in FGF-2 expression, placentas from diabetic women showed no change in villous mitotic activity but did show decreased stromal compartment apoptosis. When expressed as a ratio of Ki67-positive:Apo-Tag-positive nuclei as an index of relative cell turnover, the stromal compartment showed a significant trend towards decreased nuclei turnover (P<.05), suggesting relative tissue growth in diabetic patients. CONCLUSION: Increased FGF-2 expression and decreased stromal cell compartment turnover in the diabetic placenta might be a compensatory mechanism in response to the altered physiologic milieu of maternal diabetes on placental function.


Subject(s)
Apoptosis , Cell Division , Diabetes Mellitus, Type 1/metabolism , Fibroblast Growth Factor 2/analysis , Placenta/chemistry , Pregnancy in Diabetics , Diabetes Mellitus, Type 1/pathology , Female , Gestational Age , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Ki-67 Antigen/analysis , Placenta/cytology , Pregnancy
14.
J Med Primatol ; 32(1): 57-66, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12733603

ABSTRACT

To reduce the number of animals required for controlled studies of marmoset oocytes and early embryos, a superovulation protocol was developed for the common marmoset. Females were given up to 50 i.u./day recombinant human follicle stimulating hormone (FSH)--(r-hFSH) for 6 days. Ovaries were visualized by a modified laparoscopic technique and follicular aspiration was performed using a needle and suction apparatus inserted directly through an otoscope speculum. The number of follicles + ovulation points (+/- S.E.) was 2.9 (+/- 0.2) in controls and 14.1 (+/- 1.6; P < or = 0.001) in the 50 i.u. r-hFSH per day animals. Oocytes, typically at the germinal vesicle stage at collection, extruded a first polar body within 26 hours. In vitro fertilization was performed and embryos developed to the hatched blastocyst stage (34%). With many high quality oocytes and the ability to synchronize cycles, the marmoset is a valuable primate model for examining nuclear reprograming and early embryonic events.


Subject(s)
Callithrix/physiology , Follicle Stimulating Hormone/pharmacology , Ovary/drug effects , Animals , Blastocyst/drug effects , Embryo, Mammalian/drug effects , Female , Fertilization in Vitro , Humans , Male , Oocytes/drug effects , Recombinant Proteins/pharmacology , Superovulation/drug effects
15.
Proc Natl Acad Sci U S A ; 98(19): 10728-32, 2001 Sep 11.
Article in English | MEDLINE | ID: mdl-11553810

ABSTRACT

Transgenic mice have provided invaluable information about gene function and regulation. However, because of marked differences between rodents and primates, some areas of human biology such as early embryonic development, aging, and maternal-fetal interactions would be best studied in a nonhuman primate model. Here, we report that gene transfer into rhesus monkey (Macaca mulatta) preimplantation embryos gives rise to transgenic placentas that express a reporter transgene (eGFP). Blastocysts resulting from culture of in vitro fertilized ova were transduced with a self-inactivating lentiviral vector and transferred into recipient females. One twin and one singleton pregnancy were produced from a single stimulation cycle, and one live rhesus monkey was born from each pregnancy. Placentas from all conceptuses showed expression of the transgene as detected by reverse transcription-PCR, ribonuclease protection assay, direct epifluorescence, immunohistochemistry, and Western blot analysis. Integration in somatic tissues of the offspring was not detected. A maternal immune response to the xenogeneic placental antigen was shown by the presence of anti-GFP antibodies in peripheral blood of the recipient females by day 99 of gestation (term = 165 days). These results demonstrate that transgene expression during gestation is compatible with successful pregnancy in nonhuman primates and provides an approach that could be broadly applicable to the development of novel models for primate biomedical research.


Subject(s)
Embryonic Development/physiology , Gene Transfer Techniques , Genetic Vectors , Lentivirus , Placenta/metabolism , Animals , Cell Line, Transformed , Female , Gene Expression , Genes, Reporter , Green Fluorescent Proteins , Humans , Luminescent Proteins/genetics , Macaca mulatta , Pregnancy , Transgenes , Tumor Cells, Cultured
16.
J Med Primatol ; 30(3): 148-55, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11515670

ABSTRACT

Embryo transfer in the rhesus monkey has been historically limited to transfer of cleavage stage embryos. In order to allow genetic manipulation of rhesus embryos in vitro, without using invasive surgical techniques, it is important to explore the transfer of morula and blastocyst stage embryos. Embryos were produced by in vitro fertilization from gonadotropin-stimulated monkeys, or were obtained by nonsurgical uterine flushing of naturally mated or artificially inseminated females. Nonsurgical transfer was accomplished by inserting a metal guide through the cervix into the uterus, after which a hollow cell sampler was inserted over the guide. The guide was removed and a catheter was inserted containing one to five embryos. Several pregnancies resulted from in vitro- and in vivo-derived blastocysts, and two pregnancies were carried to term resulting in one live birth. Blood samples were collected regularly to monitor plasma levels of chorionic gonadotropin, luteinizing hormone, and progesterone. The recipients received progesterone as a subcutaneous implant or daily injections from the day of transfer. The approach described in this study provides the opportunity to explore transgenic and chimeric models in the monkey by the development of noninvasive methods to transfer late-stage embryos that have been manipulated in vitro.


Subject(s)
Blastocyst/physiology , Embryo Transfer/veterinary , Genetic Engineering/veterinary , Macaca mulatta , Animals , Animals, Genetically Modified , Catheterization/veterinary , Chimera , Chorionic Gonadotropin/administration & dosage , Chorionic Gonadotropin/pharmacology , Embryo Implantation , Female , Insemination, Artificial/veterinary , Pregnancy , Specimen Handling
18.
Am J Primatol ; 53(3): 123-30, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11253847

ABSTRACT

In humans, placental corticotropin-releasing hormone (CRH) production has been linked to the determination of gestational length, and a late gestational fall in CRH-binding protein (CRH-BP) has been linked to the onset of parturition. Expression of placental CRH mRNA is limited to primates, and only in man has a circulating CRH-BP been described. As the fall in CRH-BP in late gestation has been associated with parturition in humans, we sought to determine whether a CRH-BP circulated in the plasma of other primates. It is unclear whether maternal plasma CRH concentrations are elevated in New World monkeys and prosimians. We have therefore performed CRH plasma measurements in the blood of pregnant marmosets, in several species of lemur, and in pregnant and fetal rhesus monkeys as a positive control. Using gel chromatography, CRH-BP was detected in the human, gorilla, chimpanzee, orangutan, gibbon, macaque, squirrel monkey, and marmoset, but was absent in the mandrill, spider monkey, and lemur. CRH was detected in the plasma of pregnant marmosets and rhesus monkeys. CRH was also detected in the fetal rhesus monkey, but at lower concentrations than in maternal plasma. CRH immunoreactivity was not detectable in the plasma of pregnant lemurs or in extracts of lemur placenta. In conclusion, a circulating binding protein for CRH exists in all species of apes but occurs variably among New World and Old World monkeys and is absent in lemurs. The variable occurrence of the CRH-BP does not support a role for this protein in the mechanism of parturition in primates. Maternal CRH is elevated in the pregnant marmoset and rhesus, and may play a role in the pregnancy of New and Old World monkeys.


Subject(s)
Carrier Proteins/analysis , Pregnancy/physiology , Primates/physiology , Animals , Carrier Proteins/pharmacology , Chromatography, Gel , Corticotropin-Releasing Hormone/analysis , Corticotropin-Releasing Hormone/pharmacology , Female , Humans
19.
J Reprod Immunol ; 50(1): 57-79, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11254941

ABSTRACT

In this study, we carried out a phenotypic and functional characterization of lymphocytes isolated from the uterine endometrium of the pregnant rhesus monkey. A majority (80%) of these cells were CD56(bright+), CD3- had typical large granular lymphocyte/uterine natural killer (NK) cell morphology and contained numerous cytoplasmic granules. Flow cytometric evaluation showed that rhesus decidual CD56(bright+) cells shared other phenotypic features of human uterine NK cells, including low levels of CD45RA and CD62L expression. A majority of the rhesus uterine CD56(bright+) cells expressed low levels of CD 16 but were CD2-. In contrast, most rhesus CD16+ peripheral blood cells were CD56-. In addition to the primary population of CD56(bright+) cells, a minor subset of smaller and less granular CD56(intermediate+) decidual lymphocytes was identified, the majority of which were CD16-, CD2(+). Decidual CD56+ cells did not express monocyte/macrophage markers, including CD14, CD64 and CD68. Decidual lymphocytes effectively lysed K562, Raji and particularly 721.221 targets in cytotoxicity assays. Together, these results suggest that as in human pregnancy, rhesus decidual CD56(bright+) cells represent a distinct lymphocyte subset that belongs to the NK cell lineage.


Subject(s)
CD56 Antigen/biosynthesis , Cytotoxicity, Immunologic , Decidua/immunology , Decidua/metabolism , Killer Cells, Natural/immunology , Lymphocyte Subsets/immunology , Animals , Decidua/cytology , Female , Humans , Immunophenotyping , K562 Cells , Killer Cells, Natural/metabolism , Lymphocyte Subsets/metabolism , Macaca mulatta , Macrophages/immunology , Macrophages/metabolism , Pregnancy , Receptors, IgG/biosynthesis , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tumor Cells, Cultured
20.
Tissue Antigens ; 58(5): 329-34, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11844144

ABSTRACT

To define the killer-cell Ig-like receptors (KIR) molecules expressed within the decidua in the rhesus monkey, an early pregnancy rhesus decidual library was screened with an oligonucleotide located within the KIR D2 domain. We obtained 26 full-length clones which represented 11 mRNAs. The cDNAs encode proteins containing 2 or 3 Ig domains, and short or long cytoplasmic domains. The long domains contained immunotyrosine inhibitory motifs (ITIM) motifs indicating inhibitory function. Three clones contained single or dinucleotide deletions within the D1 domain, which would result in proteins truncated within this domain. Phylogenetic analysis of the Ig domains supports a D0 - D2 organization of rhesus KIR2DL4.1, similar to human KIR2DL4 and KIR2DL5. The rhesus KIR2DL4.1 transcript contains a long cytoplasmic tail homologous with human KIR2DL4.


Subject(s)
Decidua/metabolism , Macaca mulatta/genetics , Macaca mulatta/metabolism , Receptors, Immunologic/genetics , Amino Acid Motifs , Amino Acid Sequence , Animals , Cloning, Molecular , Female , Phylogeny , Receptors, Immunologic/metabolism , Receptors, KIR , Receptors, KIR2DL4 , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...