Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(8)2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38674162

ABSTRACT

The biologically significant phenomenon that the fetus can survive immune attacks from the mother has been demonstrated in mammals. The survival mechanism depends on the fetus and placenta actively defending themselves against attacks by maternal T cells, achieved through the localized depletion of the amino acid L-tryptophan by an enzyme called indoleamine 2,3-dioxygenase. These findings were entirely unexpected and pose important questions regarding diseases related to human pregnancy and their prevention during human pregnancy. Specifically, the role of this mechanism, as discovered in mice, in humans remains unknown, as does the extent to which impaired activation of this process contributes to major clinical diseases in humans. We have, thus, elucidated several key aspects of this enzyme expressed in the human placenta both in normal and abnormal human pregnancy. The questions addressed in this brief review are as follows: (1) localization and characteristics of human placental indoleamine 2,3-dioxygenas; (2) overall tryptophan catabolism in human pregnancy and a comparison of indoleamine 2,3-dioxygenase expression levels between normal and pre-eclamptic pregnancy; (3) controlling trophoblast invasion by indoleamine 2,3-dioxygenase and its relation to the pathogenesis of placenta accrete spectrum.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase , Placenta , Tryptophan , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Pregnancy , Female , Placenta/metabolism , Placenta/enzymology , Tryptophan/metabolism , Pre-Eclampsia/metabolism , Pre-Eclampsia/enzymology , Trophoblasts/metabolism , Animals
2.
Biomolecules ; 13(11)2023 11 07.
Article in English | MEDLINE | ID: mdl-38002309

ABSTRACT

Cell fusion in the placenta is tightly regulated. Suppressyn is a human placental endogenous retroviral protein that inhibits the profusogenic activities of another well-described endogenous retroviral protein, syncytin-1. In this study, we aimed to elucidate the mechanisms underlying suppressyn's placenta-specific expression. We identified the promoter region and a novel enhancer region for the gene encoding suppressyn, ERVH48-1, and examined their regulation via DNA methylation and their responses to changes in the oxygen concentration. Like other endogenous retroviral genes, the ERVH48-1 promoter sequence is found within a characteristic retroviral 5' LTR sequence. The novel enhancer sequence we describe here is downstream of this LTR sequence (designated EIEs: ERV internal enhancer sequence) and governs placental expression. The placenta-specific expression of ERVH48-1 is tightly controlled by DNA methylation and further regulated by oxygen concentration-dependent, hypoxia-induced transcription factors (HIF1α and HIF2α). Our findings highlight the involvement of (1) tissue specificity through DNA methylation, (2) expression specificity through placenta-specific enhancer regions, and (3) the regulation of suppressyn expression in differing oxygen conditions by HIF1α and HIF2α. We suggest that these regulatory mechanisms are central to normal and abnormal placental development, including the development of disorders of pregnancy involving altered oxygenation, such as preeclampsia, pregnancy-induced hypertension, and fetal growth restriction.


Subject(s)
Endogenous Retroviruses , Trophoblasts , Female , Humans , Pregnancy , Cell Fusion , Endogenous Retroviruses/genetics , Endogenous Retroviruses/metabolism , Gene Products, env/genetics , Gene Products, env/metabolism , Oxygen/metabolism , Placenta/metabolism , Trophoblasts/metabolism
3.
Anticancer Res ; 43(6): 2509-2517, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37247931

ABSTRACT

BACKGROUND/AIM: Ovarian clear cell carcinoma (OCCC) is a histological type of ovarian cancer that is refractory to chemotherapy and has poor prognosis, which necessitates the development of novel treatment therapies. In this study, we focused on L-type amino acid transporter 1 (LAT1), which is involved in cancer growth, and investigated the effect of its selective inhibition on cell proliferation in OCCC. MATERIALS AND METHODS: The inhibitory effect of nanvuranlat (JPH203), a LAT1 selective inhibitor, on the cellular uptake of [3H] leucine was evaluated using the OCCC cell line JHOC9, which expresses the LAT1 protein. In addition, the kinetics of cell proliferation and changes in phosphorylation of the mTOR pathway were analyzed. The correlation between LAT1 expression and progression-free survival (PFS) was evaluated using clinical specimens of OCCC. RESULTS: Nanvuranlat inhibited [3H] leucine intracellular uptake and cell proliferation in a dose-dependent manner in JHOC9 cells. In addition, it suppressed the activity of the mTOR signaling pathway, which is thought to inhibit cancer cell proliferation. LAT1 expression was most frequent in OCCC among clinical specimens of epithelial ovarian cancer. A correlation between LAT1 expression and PFS was observed in OCCC. CONCLUSION: LAT1 selective inhibition suppresses cell proliferation via the mTOR pathway by inhibiting leucine uptake in OCCC. This study illustrates the potential of using LAT1 selective inhibition as a treatment strategy for OCCC.


Subject(s)
Adenocarcinoma, Clear Cell , Ovarian Neoplasms , Female , Humans , Leucine/pharmacology , Large Neutral Amino Acid-Transporter 1 , Cell Line, Tumor , TOR Serine-Threonine Kinases/metabolism , Cell Proliferation , Ovarian Neoplasms/pathology , Adenocarcinoma, Clear Cell/drug therapy
4.
Biomolecules ; 12(8)2022 07 26.
Article in English | MEDLINE | ID: mdl-35892338

ABSTRACT

Preterm birth is one of the most significant obstetric complications. Inflammation reportedly promotes uterine contraction and weakening of the fetal membrane, which induces preterm birth. Previous studies using animal models of lipopolysaccharide-induced acute inflammation have shown that progesterone (P4) promotes uterine quiescence. However, this effect is not fully understood in chronic inflammation. This study aimed to investigate the effects of P4 on uterine contractility and inflammation of the fetal membrane in mice infected with Porphyromonas gingivalis (P.g.), a major periodontal pathogen as a model of preterm birth caused by chronic inflammation. Mice were injected with 1 mg of P4 from day 15.5 to 17.5. P4 prolonged the mean gestation period of P.g mice from 18.3 to 20.4 days, and no reduction in the gestation period was observed. P4 treatment suppressed spontaneous uterine contractility and decreased oxytocin sensitivity. In addition, the expression of inflammatory cytokines in the fetal membrane was significantly reduced. Thus, P4 prevented preterm birth by suppressing enhanced uterine contractility induced by chronic inflammation in this model. This result describes the effects of P4 in a chronic inflammation model, which may lead to a better understanding of the efficacy of P4 in preventing preterm birth in humans.


Subject(s)
Premature Birth , Uterine Contraction , Animals , Female , Humans , Infant, Newborn , Inflammation/metabolism , Mice , Porphyromonas gingivalis , Pregnancy , Premature Birth/prevention & control , Progesterone/pharmacology
5.
Sci Rep ; 12(1): 10552, 2022 06 22.
Article in English | MEDLINE | ID: mdl-35732788

ABSTRACT

Suppressyn (SUPYN) is the first host-cell encoded mammalian protein shown to inhibit cell-cell fusion. Its expression is restricted to the placenta, where it negatively regulates syncytia formation in villi. Since its chromosomal localization overlaps with the Down syndrome critical region and the TS21 placenta is characterized by delayed maturation of cytotrophoblast cells and reduced syncytialization, we hypothesized a potential link between changes in SUPYN expression and morphologic abnormalities in the TS21 placenta. Here we demonstrate that an increase in chromosomal copy number in the TS21 placenta is associated with: (1) reduced fusion of cytotrophoblast cells into syncytiotrophoblast in vivo, (2) increased SUPYN transcription, translation and secretion in vivo, (3) increased SUPYN/syncytin-1 receptor degradation in vivo, (4) increased SUPYN transcription and secretion ex vivo, (5) decreased cytotrophoblast cell fusion ex vivo, and (6) reciprocal response of changes in SUPYN and CGB in TS21 placental cells ex vivo. These data suggest direct links between immature placentation in Down syndrome and increased SUPYN. Finally, we report a significant increase in secreted SUPYN concentration in maternal serum in women with pregnancies affected by Down syndrome, suggesting that SUPYN may be useful as an alternate or additional diagnostic marker for this disease.


Subject(s)
Down Syndrome , Endogenous Retroviruses , Animals , Cell Fusion , Down Syndrome/genetics , Down Syndrome/metabolism , Female , Humans , Mammals , Placenta/metabolism , Pregnancy , Trisomy , Trophoblasts/metabolism
6.
Microscopy (Oxf) ; 71(1): 66-76, 2022 Jan 29.
Article in English | MEDLINE | ID: mdl-34536007

ABSTRACT

Amnion membrane studies related to miscarriage have been conducted in the field of obstetrics and gynecology. However, the distribution of stem cells within the amnion and the differences in the properties of each type of stem cells are still not well understood. We address this gap in knowledge in the present study where we morphologically classified the amnion membrane, and we clarified the distribution of stem cells here to identify functionally different amniotic membrane-derived stem cells. The amnion can be divided into a site that is continuous with the umbilical cord (region A), a site that adheres to the placenta (region B), and a site that is located opposite the placenta (region C). We found that human amnion epithelial stem cells (HAECs) that strongly express stem cell markers were abundant in area A. HAEC not only expressesed stem cell-specific surface markers TRA-1-60, Tra-1-81, SSEA4, SSEA3, but was also OCT-3/4 positive and had alkaline phosphatase activity. Human amniotic mesenchymal stem cells expressed KLF-A, OCTA, Oct3/4, c-MYC and Sox2 which is transcription factor. Especially, in regions A and B they have expressed CD73, and the higher expression of BCRP which is drug excretion transporter protein than the other parts. These data suggest that different types of stem cells may have existed in different area. The understanding the relation with characteristics of the stem cells in each area and function would allow for the efficient harvest of suitable HAE and HAM stem cells as using tool for regenerative medicine.


Subject(s)
Amnion , Epithelial Cells , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Amnion/metabolism , Cell Differentiation/physiology , Female , Humans , Neoplasm Proteins/metabolism , Pregnancy , Stem Cells/metabolism
7.
Int J Mol Sci ; 22(19)2021 Sep 23.
Article in English | MEDLINE | ID: mdl-34638599

ABSTRACT

Proper placental development relies on tightly regulated trophoblast differentiation and interaction with maternal cells. Human endogenous retroviruses (HERVs) play an integral role in modulating cell fusion events in the trophoblast cells of the developing placenta. Syncytin-1 (ERVW-1) and its receptor, solute-linked carrier family A member 5 (SLC1A5/ASCT2), promote fusion of cytotrophoblast (CTB) cells to generate the multi-nucleated syncytiotrophoblast (STB) layer which is in direct contact with maternal blood. Another HERV-derived protein known as Suppressyn (ERVH48-1/SUPYN) is implicated in anti-fusogenic events as it shares the common receptor with ERVW-1. Here, we explore primary tissue and publicly available datasets to determine the distribution of ERVW-1, ERVH48-1 and SLC1A5 expression at the maternal-fetal interface. While SLC1A5 is broadly expressed in placental and decidual cell types, ERVW-1 and ERVH48-1 are confined to trophoblast cell types. ERVH48-1 displays higher expression levels in CTB and extravillous trophoblast, than in STB, while ERVW-1 is generally highest in STB. We have demonstrated through gene targeting studies that suppressyn has the ability to prevent ERVW-1-induced fusion events in co-culture models of trophoblast cell/maternal endometrial cell interactions. These findings suggest that differential HERV expression is vital to control fusion and anti-fusogenic events in the placenta and consequently, any imbalance or dysregulation in HERV expression may contribute to adverse pregnancy outcomes.


Subject(s)
Endogenous Retroviruses/metabolism , Gene Products, env/metabolism , Pregnancy Proteins/metabolism , Cell Communication/physiology , Cell Differentiation/physiology , Cell Fusion/methods , Cell Line, Tumor , Decidua/metabolism , Female , Humans , Minor Histocompatibility Antigens/metabolism , Placenta/metabolism , Pregnancy , Trophoblasts/metabolism
8.
Int J Mol Sci ; 22(4)2021 Feb 10.
Article in English | MEDLINE | ID: mdl-33578919

ABSTRACT

Multinucleate syncytialized trophoblast is found in three forms in the human placenta. In the earliest stages of pregnancy, it is seen at the invasive leading edge of the implanting embryo and has been called primitive trophoblast. In later pregnancy, it is represented by the immense, multinucleated layer covering the surface of placental villi and by the trophoblast giant cells found deep within the uterine decidua and myometrium. These syncytia interact with local and/or systemic maternal immune effector cells in a fine balance that allows for invasion and persistence of allogeneic cells in a mother who must retain immunocompetence for 40 weeks of pregnancy. Maternal immune interactions with syncytialized trophoblast require tightly regulated mechanisms that may differ depending on the location of fetal cells and their invasiveness, the nature of the surrounding immune effector cells and the gestational age of the pregnancy. Some specifically reflect the unique mechanisms involved in trophoblast cell-cell fusion (aka syncytialization). Here we will review and summarize several of the mechanisms that support healthy maternal-fetal immune interactions specifically at syncytiotrophoblast interfaces.


Subject(s)
Trophoblasts/immunology , Animals , Chorionic Villi/immunology , Extracellular Vesicles/immunology , Female , Humans , Immunity , Placenta/immunology , Placentation , Pregnancy , Toll-Like Receptors/immunology
9.
Placenta ; 113: 8-14, 2021 09 15.
Article in English | MEDLINE | ID: mdl-33504453

ABSTRACT

Three versions of syncytiotrophoblast exist in the human placenta: an invasive type associated with the implanting conceptus, non-invasive villous type of definitive placenta, and placental bed giant cells. Syncytins are encoded by modified env genes of endogenous retroviruses (ERV), but how they contribute functionally to placental syncytial structures is unclear. A minimum of eight genes (ERVW1, ERVFRD-1, ERVV-1, ERVV-2, ERVH48-1, ERVMER34-1, ERV3-1, & ERVK13-1) encoding syncytin family members are expressed in human trophoblast, the majority from implantation to term. ERVW1 (Syncytin 1) and ERVFRD-1 (Syncytin 2) are considered the major fusogens, but, when the expression of their genes is analyzed by single cell RNAseq in first trimester placenta, their transcripts are distinctly patterned and also differ from those of their proposed binding partners, SLC1A5 and MFSD2A, respectively. ERVRH48-1 (suppressyn or SUPYN) and ERVMER34-1 are probable negative regulators of fusion and co-expressed, primarily in cytotrophoblast. The remaining genes and their products have been little studied. Syncytin expression is a feature of placental development in almost all eutherian mammals studied, in at least one marsupial, and in viviparous lizards, which lack the trophoblast lineage. Their expression has been inferred to be essential for pregnancy success in the mouse. All the main human ERV genes arose following independent retroviral insertion events, none of which trace back to the divergence of eutherians and metatherians (marsupials). While syncytins may be crucial for placental development, it seems unlikely that they helped orchestrate the divergence of eutherians and marsupials.


Subject(s)
Biological Evolution , Endogenous Retroviruses/genetics , Gene Products, env/metabolism , Placentation , Pregnancy Proteins/metabolism , Trophoblasts/metabolism , Cell Fusion , Female , Gene Products, env/genetics , Humans , Pregnancy , Pregnancy Proteins/genetics
10.
Placenta ; 99: 21-26, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32738645

ABSTRACT

INTRODUCTION: Inflammation and infection, including dental infectious diseases, are factors that can induce preterm birth. We previously reported that mice with dental Porphyromonas gingivalis infection could be used as a model of preterm birth. In this model, cyclooxygenase (COX)-2 and interleukin (IL)-1ß levels are increased, and P. gingivalis colonies are observed in the fetal membrane. However, the mechanism underlying fetal membrane inflammation remains unknown. Therefore, we investigated the immune responses of human amnion to P. gingivalis in vitro. METHODS: Epithelial and mesenchymal cells were isolated from human amnion using trypsin and collagenase, and primary cell cultures were obtained. Confluent cells were stimulated with P. gingivalis lipopolysaccharide (P.g-LPS) or P. gingivalis. mRNA expressions of IL-1ß, IL-8, IL-6 and COX-2, protein expressions of nuclear factor (NF)-κB pathway components and culture medium levels of prostaglandin E2 were evaluated. RESULTS: Following stimulation with 1 µg/mL P.g-LPS, the mRNA expression levels of IL-1ß, IL-8, IL-6 and COX-2 in mesenchymal cells were increased 5.9-, 3.3-, 4.2- and 3.1-fold, respectively. Similarly, the expression levels of IL-1ß, IL-8, IL-6 and COX-2 in mesenchymal cells were increased by 7.6-, 8.2-, 13.4- and 9.3-fold, respectively, after coculture with P. gingivalis. Additionally, stimulation with P.g-LPS or P. gingivalis resulted in the activation of NF-κB signaling and increased production of IL-1ß and prostaglandin E2. In contrast, no significant changes were observed in epithelial cells. DISCUSSION: Our findings suggest that mesenchymal cells might mediate the inflammatory responses to P. gingivalis and P.g-LPS, thereby producing inflammation that contributes to the induction of preterm birth.


Subject(s)
Amnion/drug effects , Epithelial Cells/drug effects , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Porphyromonas gingivalis , Amnion/metabolism , Animals , Cyclooxygenase 2/metabolism , Epithelial Cells/metabolism , Humans , Interleukin-1beta , Interleukin-6/metabolism , Interleukin-8/metabolism , Premature Birth/metabolism
11.
J Reprod Immunol ; 138: 103099, 2020 04.
Article in English | MEDLINE | ID: mdl-32050141

ABSTRACT

Immunohistochemical localisation of indoleamine 2,3-dioxygenase was studied in order to better understand the pathophysiology of placenta accreta spectrum. In the decidua staining for indoleamine 2,3-dioxygenase was found in the glandular epithelium with some additional positive cells. Extravillous cytotrophoblast invasion was present in the myometrium which was not covered by the decidual tissue whereas myometrial invasion of cytotrophoblasts was absent where this tissue lay deep to decidua. These results suggest that indoleamine 2,3-dioxygenase expression in the decidua may normally control trophoblast invasion and absence of its expression where decidua is absent may be involved in the pathogenesis of the over-invaded placenta.


Subject(s)
Cesarean Section/adverse effects , Cicatrix/pathology , Decidua/pathology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Placenta Accreta/etiology , Cicatrix/etiology , Decidua/surgery , Female , Humans , Hysterectomy , Indoleamine-Pyrrole 2,3,-Dioxygenase/analysis , Placenta Accreta/pathology , Placenta Accreta/surgery , Pregnancy , Pregnancy Trimester, First , Trophoblasts/pathology
12.
Int J Tryptophan Res ; 13: 1178646920984163, 2020.
Article in English | MEDLINE | ID: mdl-33447047

ABSTRACT

Immunohistochemical localization of indoleamine 2,3-dioxygenase-1 and indoleamine 2,3-dioxygenase-2, the first and rate-limiting enzyme in tryptophan metabolism along the kynurenine pathway, has been studied in order to better understand the physiological significance of these enzymes at the maternal-fetal interface of human pregnancy with a gestational age of 7 weeks (n = 1) and term placentas (37-40 weeks of gestation, n = 5). Indoleamine 2,3-dioxygenase-1 protein immunoreactivity was found in glandular epithelium of the decidua and the endothelium of the fetal blood vessels in the villous stroma with some additional positive cells in the villous core and in the decidua. The syncytiotrophoblast stained strongly for indoleamine 2,3-dioxygenase-2. Immunoreactivity of kynurenine, the immediate downstream product of indoleamine 2,3-dioxygenase-mediated tryptophan metabolism, showed the same localization as that of indoleamine 2,3-dioxygenase-1 and indoleamine 2,3-dioxygenase-2, suggesting these are functional enzymes. Interferon-γ added to placental villous explant culture markedly stimulated expression level of both mRNA and immunoreactivity of indoleamine 2,3-dioxygenase-1. The different cellular expression and interferon-γ sensitivity of these enzymes at the maternal-fetal interface suggests distinct physiological roles for each enzyme in normal human viviparity.

13.
Sci Rep ; 9(1): 19502, 2019 12 20.
Article in English | MEDLINE | ID: mdl-31862915

ABSTRACT

We previously identified suppressyn (SUPYN), a placental protein that negatively regulates the cell fusion essential for trophoblast syncytialization via binding to the trophoblast receptor for syncytin-1, ASCT2, and hypothesized that SUPYN may thereby regulate cell-cell fusion in the placenta. Here, we redefine in vivo SUPYN localization using specific monoclonal antibodies in a rare early placental sample, showing SUPYN localization in villous and extravillous trophoblast subtypes, the decidua and even in placental debris in the maternal vasculature. In human trophoblast cell lines, we show SUPYN alters ASCT2 glycosylation within the secretory pathway and that this binding is associated with inhibition of cell fusion. Using newly-optimized trophoblast isolation protocols that allow tracking of ex vivo cell fusion, we present transcription and translation dynamics of fusion-related proteins over 96 hours in culture and the effects of changes in ambient oxygen levels on these processes. We report converse syncytin-1 and SUPYN transcriptional and translational responses to surrounding oxygen concentrations that suggest both are important in the effects of hypoxia and hyperoxia on placental syncytialization. Our results suggest that SUPYN's anti-fusogenic properties may be exerted at several sites in the maternal body and its dysregulation may be associated with diseases of abnormal placentation.


Subject(s)
Gene Products, env/metabolism , Placentation/physiology , Pregnancy Proteins/metabolism , Trophoblasts/metabolism , Adult , Amino Acid Transport System ASC/metabolism , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Female , Glycosylation , Humans , Minor Histocompatibility Antigens/metabolism , Pregnancy , Young Adult
14.
Cancer Genomics Proteomics ; 16(6): 481-490, 2019.
Article in English | MEDLINE | ID: mdl-31659102

ABSTRACT

BACKGROUND/AIM: The biological importance of the caudal-related homeobox transcription factor CDX2 in acquiring resistance to anticancer drugs has been studied in ovarian mucinous carcinoma. CDX2 promotes the expression of multidrug resistance 1 (MDR1) and confers resistance to paclitaxel. The regenerating islet-derived family member 4 (REG4) gene is a potential target gene of CDX2. In this study, we investigated the relationship between the expression of CDX2 and Reg IV and the regulation of Reg IV expression and examined novel chemotherapeutic regimens. MATERIALS AND METHODS: The regulation of Reg IV expression by CDX2 and sensitivity of 5-fluorouracil (5-FU) were evaluated using ovarian mucinous cancer cell lines. RESULTS: The correlation of CDX2 with Reg IV expression was demonstrated in ovarian mucinous carcinoma. Reg IV expression was enhanced by transfection of CDX2 and was suppressed by inhibition of CDX2 expression. OMC-3 cells with ectopically overexpressed CDX2 showed enhanced apoptosis and sensitivity to 5-FU. CONCLUSION: CDX2 promotes resistance to paclitaxel and sensitivity to 5-FU. Novel 5-FU-based chemotherapy based on CDX2 may be used in ovarian mucinous carcinoma.


Subject(s)
Adenocarcinoma, Mucinous , CDX2 Transcription Factor/metabolism , Drug Resistance, Neoplasm , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Neoplasm Proteins/metabolism , Ovarian Neoplasms , Pancreatitis-Associated Proteins/biosynthesis , Adenocarcinoma, Mucinous/drug therapy , Adenocarcinoma, Mucinous/metabolism , Adenocarcinoma, Mucinous/pathology , Cell Line, Tumor , Female , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Predictive Value of Tests
15.
Cell Transplant ; 24(10): 2065-76, 2015.
Article in English | MEDLINE | ID: mdl-25333453

ABSTRACT

Human amnion-derived cells are considered to be a promising alternative cell source for their potential clinical use in tissue engineering and regenerative medicine because of their proliferation and differentiation ability. The cells can easily be obtained from human amnion, offering a potential source without medical intervention. It has been proven that human amnion-derived cells express immunosuppressive factors CD59 and HLA-G, implying that they may have an immunosuppressive function. To assess the immunosuppressive activity, we investigated the effect of human amnion-derived cells on NK cell and monocyte function. Amnion-derived cells inhibited the cytotoxicity of NK cells to K562 cells. The inhibition depended on the NK/amnion-derived cell ratio. The inhibition of NK cytotoxicity was recovered by continuous culturing without amnion-derived cells. The inhibition of NK cytotoxicity was related to the downregulation of the expression of the activated NK receptors and the production of IFN-γ, as well as the upregulation of the expression of IL-10 and PGE2 in human amnion-derived cells. The addition of antibody to IL-10 or PGE2 inhibitor tended to increase NK cytotoxicity. IL-10 and PGE2 might be involved in the immunosuppressive activity of amniotic cells toward NK cells. Amniotic cells also suppressed the activity of cytokine production in monocytes analyzed with TNF-α and IL-6. These data suggested that amniotic cells have immunosuppressive activity.


Subject(s)
Amnion/cytology , Amnion/immunology , Cell Differentiation/physiology , Killer Cells, Natural/cytology , Monocytes/cytology , Stem Cells/cytology , Antigens, CD/metabolism , Cells, Cultured , Cytotoxicity, Immunologic/immunology , Humans , Interleukin-6/metabolism , Killer Cells, Natural/immunology , Monocytes/immunology , Stem Cells/immunology , Tumor Necrosis Factor-alpha/metabolism
16.
Am J Reprod Immunol ; 70(3): 213-20, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23590581

ABSTRACT

PROBLEM: Magnesium sulfate (MgSO4 ) exposure reduces the risk of cerebral palsy. As neonatal inflammatory cytokine levels strongly correlate with neurologic outcome, we hypothesize that MgSO4 decreases inflammatory cytokine production. METHOD OF STUDY: We assessed the effect of MgSO4 on cellular magnesium levels, cytokine production, and release within THP-1 and cord blood mononuclear cells. RESULTS: MgSO4 exposure increased intracellular magnesium levels, reducing the frequency of THP-1 cells producing IL-1ß, IL-8, and TNF-α following LPS stimulation. Significant reductions in the frequency of neonatal monocytes producing TNF-α (48%) and IL-6 (37%) were seen following LPS stimulation, and MgSO4 also significantly decreased the frequency of monocytes producing TNF-α (35%) under basal conditions. Decreased cytokine production was confirmed via ELISA, demonstrating a sustained effect and dose response. Magnesium also reduced cytokine production following stimulation with TLR ligands representing obstetrical infections (group B Streptococcus and Mycoplasma) and in preterm neonatal monocytes. CONCLUSION: MgSO4 increases intracellular magnesium, reducing inflammatory cytokine production and release, potentially elucidating the mechanism by which MgSO4 prevents cerebral palsy, eclampsia, and preterm birth.


Subject(s)
Cytokines/drug effects , Inflammation/immunology , Leukocytes, Mononuclear/drug effects , Magnesium Sulfate/pharmacology , Magnesium/metabolism , Monocytes/drug effects , Cell Line , Cells, Cultured , Cytokines/metabolism , Fetal Blood/cytology , Fetal Blood/immunology , Humans , Infant, Newborn , Interleukin-6/immunology , Interleukin-6/pharmacology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Magnesium/pharmacology , Monocytes/immunology , Monocytes/metabolism , Premature Birth/immunology , Tumor Necrosis Factor-alpha
17.
Sci Rep ; 3: 1462, 2013.
Article in English | MEDLINE | ID: mdl-23492904

ABSTRACT

While common in viral infections and neoplasia, spontaneous cell-cell fusion, or syncytialization, is quite restricted in healthy tissues. Such fusion is essential to human placental development, where interactions between trophoblast-specific human endogenous retroviral (HERV) envelope proteins, called syncytins, and their widely-distributed cell surface receptors are centrally involved. We have identified the first host cell-encoded protein that inhibits cell fusion in mammals. Like the syncytins, this protein, called suppressyn, is HERV-derived, placenta-specific and well-conserved over simian evolution. In vitro, suppressyn binds to the syn1 receptor and inhibits syn1-, but not syn2-mediated trophoblast syncytialization. Suppressyn knock-down promotes cell-cell fusion in trophoblast cells and cell-associated and secreted suppressyn binds to the syn1 receptor, ASCT2. Identification of the first host cell-encoded inhibitor of mammalian cell fusion may encourage improved understanding of cell fusion mechanisms, of placental morphogenesis and of diseases resulting from abnormal cell fusion.


Subject(s)
Endogenous Retroviruses/metabolism , Gene Products, env/metabolism , Placenta/metabolism , Pregnancy Proteins/metabolism , Trophoblasts/physiology , Viral Envelope Proteins/metabolism , Amino Acid Sequence , Amino Acid Transport System ASC/metabolism , Animals , Base Sequence , Blotting, Western , Cell Fusion , Cell Line , Cell Line, Tumor , Endogenous Retroviruses/genetics , Female , Gene Products, env/genetics , Gene Products, env/pharmacology , Humans , Minor Histocompatibility Antigens , Molecular Sequence Data , Pregnancy , Pregnancy Proteins/genetics , Pregnancy Proteins/pharmacology , Protein Binding , RNA Interference , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Trophoblasts/cytology , Trophoblasts/drug effects , Viral Envelope Proteins/genetics , Viral Envelope Proteins/pharmacology
18.
J Immunol ; 188(12): 6338-46, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22611240

ABSTRACT

MgSO(4) exposure before preterm birth is neuroprotective, reducing the risk of cerebral palsy and major motor dysfunction. Neonatal inflammatory cytokine levels correlate with neurologic outcome, leading us to assess the effect of MgSO(4) on cytokine production in humans. We found reduced maternal TNF-α and IL-6 production following in vivo MgSO(4) treatment. Short-term exposure to a clinically effective MgSO(4) concentration in vitro substantially reduced the frequency of neonatal monocytes producing TNF-α and IL-6 under constitutive and TLR-stimulated conditions, decreasing cytokine gene and protein expression, without influencing cell viability or phagocytic function. In summary, MgSO(4) reduced cytokine production in intrapartum women, term and preterm neonates, demonstrating effectiveness in those at risk for inflammation-associated adverse perinatal outcomes. By probing the mechanism of decreased cytokine production, we found that the immunomodulatory effect was mediated by magnesium and not the sulfate moiety, and it was reversible. Cellular magnesium content increased rapidly upon MgSO(4) exposure, and reduced cytokine production occurred following stimulation with different TLR ligands as well as when magnesium was added after TLR stimulation, strongly suggesting that magnesium acts intracellularly. Magnesium increased basal IĸBα levels, and upon TLR stimulation was associated with reduced NF-κB activation and nuclear localization. These findings establish a new paradigm for innate immunoregulation, whereby magnesium plays a critical regulatory role in NF-κB activation, cytokine production, and disease pathogenesis.


Subject(s)
Immunologic Factors/pharmacology , Immunomodulation/immunology , Inflammation/immunology , Magnesium Sulfate/pharmacology , Monocytes/drug effects , Blotting, Western , Cells, Cultured , Cytokines/biosynthesis , Female , Fetal Blood/drug effects , Fetal Blood/immunology , Humans , Immunity, Innate/drug effects , Immunity, Innate/immunology , Infant, Newborn , Infant, Premature/immunology , Monocytes/immunology , Phagocytosis/drug effects , Phagocytosis/immunology , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction
19.
Curr HIV Res ; 10(3): 218-27, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22384841

ABSTRACT

Among the now pandemic sexually transmitted infections (STIs), Chlamydia trachomatis (C. trachomatis) is the predominant bacterial pathogen and human immunodeficiency virus type 1 (HIV-1) is the most lethal of the viral pathogens. The female genital tract is the primary site for heterosexual transmission of both C. trachomatis and HIV-1. Infection with C. trachomatis, and with a variety of other STIs, increases the risk for transmission of HIV-1, although the mechanisms for this finding remain unclear. We have used in vitro modeling to assess the mechanisms by which infection with genital C. trachomatis serovars might increase the transmission of HIV-1 across the female genital tract. C. trachomatis infection of an immortalized endocervical epithelial cell line (A2EN) increases the cell surface expression of the HIV-1 alternative primary receptor, galactosyl ceramide (GalCer), and of the HIV-1 co-receptors, CXCR4 and CCR5. C. trachomatis infection also increases the binding of HIV-1 to A2EN cells, and, subsequently, increases levels of virus in co-cultures of HIV-exposed A2EN and susceptible MT4-R5 T cells. Finally, in vivo endocervical cell sampling reveals a dramatic increase in the number of CD4+, CXCR4 and/or CCR5 positive T cell targets in the endocervix of C. trachomatis positive women when compared to those who are C. trachomatis negative. This combination of in vitro and in vivo results suggests several mechanisms for increased transmission of HIV-1 across the endocervices of C. trachomatis-infected women.


Subject(s)
Cervix Uteri/immunology , Chlamydia Infections/immunology , Chlamydia trachomatis/pathogenicity , HIV Seropositivity/transmission , HIV-1 , Receptors, CCR5/immunology , Receptors, CXCR4/immunology , Base Sequence , Cell Line , Cervix Uteri/microbiology , Chlamydia Infections/microbiology , Chlamydia Infections/transmission , Disease Susceptibility , Female , HIV Seropositivity/immunology , HIV Seropositivity/microbiology , Humans , Virus Replication
20.
Infect Dis Obstet Gynecol ; 2011: 420905, 2011.
Article in English | MEDLINE | ID: mdl-21747639

ABSTRACT

Genital C. trachomatis infections typically last for many months in women. This has been attributed to several strategies by which C. trachomatis evades immune detection, including well-described methods by which C. trachomatis decreases the cell surface expression of the antigen presenting molecules major histocompatibility complex (MHC) class I, MHC class II, and CD1d in infected genital epithelial cells. We have harnessed new methods that allow for separate evaluation of infected and uninfected cells within a mixed population of chlamydia-infected endocervical epithelial cells to demonstrate that MHC class I downregulation in the presence of C. trachomatis is mediated by direct and indirect (soluble) factors. Such indirect mechanisms may aid in priming surrounding cells for more rapid immune evasion upon pathogen entry and help promote unfettered spread of C. trachomatis genital infections.


Subject(s)
Chlamydia Infections/microbiology , Chlamydia trachomatis/immunology , Histocompatibility Antigens Class I/biosynthesis , Cell Line, Tumor , Cervix Uteri/cytology , Cervix Uteri/microbiology , Chlamydia Infections/immunology , Chlamydia Infections/metabolism , Down-Regulation , Epithelial Cells/cytology , Epithelial Cells/microbiology , Female , Flow Cytometry , Histocompatibility Antigens Class I/immunology , Host-Pathogen Interactions , Humans , Microscopy, Fluorescence , Models, Biological
SELECTION OF CITATIONS
SEARCH DETAIL
...