Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
1.
Mediators Inflamm ; 2023: 8215567, 2023.
Article in English | MEDLINE | ID: mdl-37035756

ABSTRACT

This study explored the role of T cell subsets and the expression of related microRNAs in patients with recurrent early pregnancy loss (EPL). Fifty patients with EPL loss between May 2018 and May 2021 were randomly selected as the EPL group, and 50 pregnant women with normal pregnancies or normal delivery outcomes were randomly selected as the control group. The expression levels of T cell subset-related markers and T cell subset-related miRNAs, in addition to the frequencies of T cell subsets, in peripheral blood of the two groups were analyzed. In terms of T cell-related markers, the results showed that the expression levels of the transcriptional regulator TBX-21 (T-bet) and interferon regulatory factor 4 (IRF4) were significantly upregulated in peripheral blood of the patients in the EPL group (P < 0.05), whereas the expression levels of GATA binding protein 3 (GATA3) and glucocorticoid-induced tumor necrosis factor receptor (GITR) were significantly downregulated (P < 0.05). In the EPL group, the expression of mir-106b, mir-93, and mir-25 was upregulated (1.51 ± 0.129, 1.43 ± 0.132, and 1.73 ± 0.156, respectively) in regulatory T (Treg) cell-related T cell subsets, whereas the expression of miR-146a and miR-155 was downregulated (P < 0.05). The frequencies of Treg and exhausted T cells in the EPL group were significantly lower than those in the control group (P < 0.05). The cell frequencies of T helper 17 (Th17) cells and exhausted Treg cells in the EPL group were significantly higher than those in the control group (P < 0.05). In conclusion, immune cells and associated miRNA profiles can be used as prognostic biomarkers for the treatment of human reproductive disorders, such as EPL.


Subject(s)
Abortion, Habitual , Embryo Loss , MicroRNAs , T-Lymphocyte Subsets , Female , Humans , Pregnancy , Abortion, Habitual/genetics , Abortion, Habitual/immunology , Embryo Loss/genetics , Embryo Loss/immunology , Gene Expression , MicroRNAs/genetics , MicroRNAs/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology
2.
FASEB J ; 35(10): e21938, 2021 10.
Article in English | MEDLINE | ID: mdl-34547143

ABSTRACT

Uterine glands are essential for the establishment of pregnancy and have critical roles in endometrial receptivity to blastocyst implantation, stromal cell decidualization, and placentation. Uterine gland dysfunction is considered a major contributing factor to pregnancy loss, however our understanding of how glands impact embryo survival and stromal cell decidualization is incomplete. Forkhead box A2 (FOXA2) is expressed only in the glandular epithelium and regulates its development and function. Mice with a conditional deletion of FOXA2 in the uterus are infertile due to defective embryo implantation arising from a lack of leukemia inhibitory factor (LIF), a critical factor of uterine gland origin. Here, a glandless FOXA2-deficient mouse model, coupled with LIF repletion to rescue the implantation defect, was used to investigate the roles of uterine glands in embryo survival and decidualization. Studies found that embryo survival and decidualization were compromised in glandless FOXA2-deficient mice on gestational day 6.5, resulting in abrupt pregnancy loss by day 7.5. These findings strongly support the hypothesis that uterine glands secrete factors other than LIF that impact embryo survival and stromal cell decidualization for pregnancy success.


Subject(s)
Decidua/metabolism , Embryo Loss , Embryo, Mammalian/embryology , Embryonic Development , Stromal Cells/metabolism , Uterus/metabolism , Animals , Decidua/immunology , Embryo Loss/immunology , Embryo, Mammalian/immunology , Embryonic Development/immunology , Female , Hepatocyte Nuclear Factor 3-beta/deficiency , Leukemia Inhibitory Factor , Mice , Pregnancy , Pregnancy Outcome , Stromal Cells/immunology , Transcriptome , Uterus/immunology
3.
FASEB J ; 35(8): e21754, 2021 08.
Article in English | MEDLINE | ID: mdl-34191338

ABSTRACT

To obtain a successful pregnancy, the establishment of maternal-fetal tolerance and successful placentation are required to be established. Disruption of this immune balance and/or inadequate placental perfusion is believed to be associated with a lot of pregnancy-related complications, such as recurrent spontaneous abortion, pre-eclampsia, and fetal intrauterine growth restriction. Extravillous trophoblasts (EVTs) have the unique ability to instruct decidual immune cells (DICs) to develop a regulatory phenotype for fetal tolerance. Utilizing immortalized human first trimester extravillous trophoblast cells and primary EVTs, we found that DICs promote EVT function and placental development. We have previously shown that checkpoints T-cell immunoglobulin mucin-3 (Tim-3) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) are important for DIC function. In the present study, we showed that blockade of Tim-3 and CTLA-4 pathways leaded to the abnormal DICs-EVTs interaction, poor placental development, and increased fetal loss. Treatment with IL-4 and IL-10 could rescue the adverse effects of targeting Tim-3 and CTLA-4 on the pregnancy outcome. Hence, the reproductive safety must be a criterion considered in the assessment of immuno-therapeutic agents. In addition, IL-4 and IL-10 may represent novel therapeutic strategies to prevent pregnancy loss induced by checkpoint inhibition.


Subject(s)
CTLA-4 Antigen/immunology , Decidua/immunology , Hepatitis A Virus Cellular Receptor 2/immunology , Interleukin-10/immunology , Interleukin-4/immunology , Trophoblasts/immunology , Animals , CTLA-4 Antigen/antagonists & inhibitors , Cell Communication/immunology , Cells, Cultured , Decidua/cytology , Embryo Loss/immunology , Female , Hepatitis A Virus Cellular Receptor 2/antagonists & inhibitors , Humans , Immune Tolerance , Interleukin-10/administration & dosage , Interleukin-4/administration & dosage , Male , Maternal-Fetal Exchange/immunology , Mice, Inbred BALB C , Mice, Inbred CBA , Mice, Inbred DBA , Models, Immunological , Placentation/immunology , Pregnancy , Pregnancy Outcome , Signal Transduction/immunology , Trophoblasts/cytology
4.
Am J Reprod Immunol ; 86(5): e13480, 2021 11.
Article in English | MEDLINE | ID: mdl-34191381

ABSTRACT

PROBLEM: Decidual macrophages (dMφ ) play an important role in the formation of maternal-fetal immune tolerance. However, factors that influence the immune status of dMφ and the related potential mechanisms have not been elucidated to date. METHOD OF STUDY: The gene transcription in dMφ , decidual stromal cells (DSCs), extravillous trophoblasts (EVTs), and peripheral monocytes (pMo) from human samples were measured using real-time polymerase chain reaction (PCR). Monocyte-DSC co-culture was established to explore whether DSCs influenced dMφ polarization via C-C motif ligand 2 (CCL2)-C-C chemokine receptor (CCR2) binding using flow cytometry. In vivo, changes in dMφ percentage and M1 and M2 marker expression after treatment with CCR2 or Janus kinase 2 (JAK2) inhibitor were detected with flow cytometry. Embryo resorption percentages in the above groups were also analyzed. RESULTS: We found that dMφ were an M1/M2 mixed status at the maternal-fetal interface during early pregnancy. CCL2 influenced the immune status of dMφ in an autocrine and paracrine manner. As a downstream regulator of CCR2 and triggers the Stat3 pathway, JAK2 was found to be essential for dMφ homeostasis in vivo. JAK2 inhibitor decreased the dMφ proportion and attenuated Ki67, CD36, CD86, CD206, TNF, and IL-10 expression in dMφ at E8.5 d. Moreover, CCR2-JAK2 pathway inhibition decreased the width of the placental labyrinth layer, further influencing the pregnancy outcome. CONCLUSION: The M1/M2 mixed immune status of dMφ was regulated by DSCs via CCR2, and the CCL2/CCR2/JAK2 pathway was essential for the immune status of dMφ and the outcome of early pregnancy.


Subject(s)
Chemokine CCL2/metabolism , Decidua/enzymology , Histocompatibility, Maternal-Fetal , Immune Tolerance , Janus Kinase 2/metabolism , Macrophages/enzymology , Receptors, CCR2/metabolism , Stromal Cells/enzymology , Adult , Animals , Cells, Cultured , Coculture Techniques , Decidua/drug effects , Decidua/immunology , Embryo Loss/enzymology , Embryo Loss/immunology , Female , Humans , Janus Kinase 2/antagonists & inhibitors , Janus Kinase Inhibitors/pharmacology , Macrophages/drug effects , Macrophages/immunology , Mice, Inbred C57BL , Phenotype , Pregnancy , Pregnancy Outcome , Receptors, CCR2/antagonists & inhibitors , Signal Transduction , Stromal Cells/drug effects , Stromal Cells/immunology , Young Adult
5.
Sci Rep ; 11(1): 12771, 2021 06 17.
Article in English | MEDLINE | ID: mdl-34140546

ABSTRACT

Objective of experiment was to determine whether oxidative stress (OS) and inflammation altered embryonic loss in dairy cows. Blood samples were collected at days 0, 16, 32 and 60 after timed (AI) from 200 Holstein cows to determine embryonic loss based on interferon-stimulated gene-15 (ISG15) mRNA expression (day 16) and ultrasound at day 32 and day 60. Leucocyte expressions of mRNA TLR2, TLR4, TNF-α, IL1B, IL10, STAT3 (inflammation), PTGS2, PTGES (prostaglandin synthesis), and PLA2G4A and ALOX5AP (eicosanoid metabolism) at days 0 and 16 were determined. Plasma redox status for antioxidant enzymatic activities of glutathione peroxidase (GPX), superoxide dismutase (SOD), total antioxidant capacity (TAC), and concentrations of malondialdehyde (MDA) were determined at days 0, 16, 32 and 60. All antioxidant-redox responses were beneficially significant in pregnant cows diagnosed pregnant at day16 and sustained pregnancy to day 60 compared to non-pregnant cows at day16 or pregnant at day16 and lost embryos by days 32 or 60. The leucocyte mRNA expressions of TLR2, TLR4, STAT 3, IL1B, PTGS2, PLA2G4A and ALOX5AP were greater and PTGES was lower at day16 in pregnant cows that lost embryos early (P < 0.05). In conclusion peripheral leucocyte molecular indicators of inflammation and plasma indicators of OS were altered in pregnant cows undergoing embryonic losses compared to cows with a sustained pregnancy.


Subject(s)
Cattle/immunology , Embryo Loss/immunology , Embryo Loss/veterinary , Inflammation/immunology , Inflammation/veterinary , Leukocytes/metabolism , Oxidative Stress , Animals , Antioxidants/metabolism , Cattle/blood , Eicosanoids/metabolism , Embryo Loss/blood , Female , Gene Expression Regulation , Glutathione Peroxidase/metabolism , Inflammation/blood , Interferons/metabolism , Oxidation-Reduction , Pregnancy , Prostaglandins/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Superoxide Dismutase/metabolism , Ultrasonography
6.
Science ; 371(6534)2021 03 12.
Article in English | MEDLINE | ID: mdl-33707237

ABSTRACT

Antiphospholipid antibodies (aPLs) cause severe autoimmune disease characterized by vascular pathologies and pregnancy complications. Here, we identify endosomal lysobisphosphatidic acid (LBPA) presented by the CD1d-like endothelial protein C receptor (EPCR) as a pathogenic cell surface antigen recognized by aPLs for induction of thrombosis and endosomal inflammatory signaling. The engagement of aPLs with EPCR-LBPA expressed on innate immune cells sustains interferon- and toll-like receptor 7-dependent B1a cell expansion and autoantibody production. Specific pharmacological interruption of EPCR-LBPA signaling attenuates major aPL-elicited pathologies and the development of autoimmunity in a mouse model of systemic lupus erythematosus. Thus, aPLs recognize a single cell surface lipid-protein receptor complex to perpetuate a self-amplifying autoimmune signaling loop dependent on the cooperation with the innate immune complement and coagulation pathways.


Subject(s)
Antigen Presentation , Autoimmunity , Blood Coagulation/immunology , Endothelial Protein C Receptor/immunology , Lupus Erythematosus, Systemic/immunology , Lysophospholipids/immunology , Monoglycerides/immunology , Animals , Antibodies, Antiphospholipid/biosynthesis , Autoantibodies/biosynthesis , Disease Models, Animal , Embryo Loss/immunology , Endosomes/immunology , Endothelial Protein C Receptor/genetics , Humans , Immunity, Innate , Lupus Erythematosus, Systemic/blood , Mice , Mice, Mutant Strains , Sphingomyelin Phosphodiesterase/metabolism , Thrombosis/immunology , Toll-Like Receptor 7/immunology
7.
Drug Chem Toxicol ; 44(5): 544-549, 2021 Sep.
Article in English | MEDLINE | ID: mdl-31368388

ABSTRACT

Cytochrome c has been used as first-aid in the clinic for organs which are lacking oxygen. But recent report show cytochrome c injection destroys dendritic cells (DCs) which play a pivotal role in feto-maternal tolerance. However, it is not clear whether cytochrome c injection causes abortion. The cytochrome c was injected by tail vein of mice at the Day 5.5 of pregnancy (E5.5) after mating with male BALB/c mice. The total number of implantations and resorption sites was recorded at the E12.5 in pregnant mice. Expression of interferon-γ, tumor necrosis-α interleukin (IL)-4, IL-10, IL-12 and transforming growth factor-ß in the mouse endometrium was measured by ELISA. Injection of cytochrome c via tail vein at the E5.5 induced fetal resorption at E12.5, and evoked an immune imbalance at the maternal-fetal interface. Notably, injection of mouse bone marrow-derived DCs (BM-DCs) rescued the cytochrome c-evoked embryo resorption. The present study suggests cytochrome c injection causes embryo resorption in mice, hinting caution regarding the use of cytochrome c in pregnant women. In addition, it may provide an easy and novel way to establish a mouse model of abortion.HighlightsCytochrome c injection induced fetal rejection.Cytochrome c injection leads to a T helper 1/T helper 2 imbalance at the maternal-fetal interface.A mouse model of abortion was established by injecting tail vein with cytochrome c.


Subject(s)
Cytochromes c/toxicity , Cytokines/metabolism , Embryo Loss/chemically induced , Immune Tolerance/immunology , Animals , Cytochromes c/administration & dosage , Disease Models, Animal , Embryo Loss/immunology , Female , Horses , Male , Mice , Mice, Inbred BALB C , Pregnancy
8.
Bull Exp Biol Med ; 168(6): 757-760, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32328935

ABSTRACT

Spontaneous proliferative activity of splenocytes in female CBA mice and the response of these cells to antigens of allogeneic male BALB/c and DBA/2 mice in a mixed splenocyte culture were evaluated by 3H-thymidine incorporation in different pregnancy models. ♀CBA×♂BALB/c mating was used for modeling physiological pregnancy. Spontaneous abortions were reproduced by abortion-prone ♀CBA×♂DBA/2 mating. In order to simulate immunostimulant-induced and immunostimulant-potentiated abortions, 0.83 mg/kg muramyl dipeptide ß-heptylglycoside was intraperitoneally injected to CBA females mated with BALB/c or DBA/2 males, respectively, on gestation days 5 and 7. The increase in the rate of embryo resorption in the models of spontaneous, induced, and potentiated abortions occurred against the background of an increase in the level of spontaneous proliferation of splenocytes and a decrease in their reactivity to paternal antigens on gestation day 9.


Subject(s)
Abortion, Spontaneous/immunology , Cell Proliferation/drug effects , Embryo Loss/immunology , Glycopeptides/pharmacology , Lymphocytes/drug effects , Spleen/drug effects , Abortion, Induced/methods , Abortion, Spontaneous/chemically induced , Abortion, Spontaneous/pathology , Animals , Coculture Techniques , Crosses, Genetic , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/pathology , Embryo Loss/chemically induced , Embryo Loss/pathology , Female , Gestational Age , Injections, Intraperitoneal , Lymphocytes/immunology , Lymphocytes/pathology , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred CBA , Mice, Inbred DBA , Pregnancy , Primary Cell Culture , Spleen/immunology , Spleen/pathology , Thymidine/metabolism , Tritium
9.
Gynecol Endocrinol ; 35(sup1): 5-10, 2019.
Article in English | MEDLINE | ID: mdl-31532308

ABSTRACT

The trial objective was to determine the peripheral blood NK cells cytotoxic activity effect on trophoblast cells at recurrent pregnancy loss (RPL). The investigation involved non-pregnant women with PRL in proliferating and secretory menstrual cycle phases (PMCPh and SMCPh, respectively); women of 6-7 weeks pregnancy with RPL in past medical history; healthy fertile non-pregnant women in PMCPh and SMCPh, women of 6-7 weeks physiological pregnancy, nulliparity healthy women with regular menstrual function in PMCPh and SMCPh. NK cells cytotoxic activity was determined using peripheral blood mononuclear cells. The target cells were JEG-3 line trophoblasts. It has been established that NK cells cytotoxic activity effect on trophoblasts is lower in SMCPh than in PMCPh in non-pregnant fertile women. The NK cells cytotoxic activity was higher in SMCPh than in PMCPh in non-pregnant women with PRL and also higher than the same value in SMCPh in non-pregnant fertile women. The increased NK cells cytotoxic activity values in SMCPh in women with RPL may be the reason for miscarriage.


Subject(s)
Abortion, Habitual/blood , Cell Communication , Killer Cells, Natural/physiology , Trophoblasts/physiology , Abortion, Habitual/immunology , Adult , Case-Control Studies , Cells, Cultured , Cohort Studies , Embryo Loss/blood , Embryo Loss/immunology , Female , Humans , Leukocytes, Mononuclear/physiology , Pregnancy , Pregnancy Trimester, First , Trophoblasts/immunology , Trophoblasts/pathology
10.
Int Immunopharmacol ; 68: 1-6, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30597415

ABSTRACT

Innate lymphoid cells (ILCs) are newly identified members of the innate lymphocyte family, which can function as adaptive T cells and act as critical modulators of inflammatory processes within different tissues and immune diseases. The role of uterine ILCs (uILCs) has recently been elucidated alongside changes associated with normal pregnancy. However, the proportions of uterine ILCs and their role in unsuccessful pregnancy remain unclear. We analyzed the characterization of uILC subsets and the expression of signature cytokines associated with ILCs in a mouse model of unsuccessful pregnancy induced by LPS, and we describe the dynamic changes they undergo during this process. We found that mice exposed to LPS display significantly higher levels of uNK cells, and uILC3s. However, a lower proportion of uILC2s and uILC1s were detected in abortion mice. In addition, we found that abortion mice display markedly higher expression of IFN-γ and IL-A17, and lower levels of IL-5. No significant differences in the expression of IL-13 and IL-22 were observed. The findings suggest that uILCs play distinct non-redundant roles during pregnancy, and uILCs may affect maternal-fetal tolerance via IL-17A, IL-5, and IFN-γ production.


Subject(s)
Embryo Loss/immunology , Lymphocytes/immunology , Uterus/immunology , Animals , Cytokines/immunology , Disease Models, Animal , Female , Immunity, Innate , Lipopolysaccharides , Mice, Inbred BALB C , Mice, Inbred C57BL , Pregnancy
11.
J Reprod Immunol ; 131: 30-35, 2019 02.
Article in English | MEDLINE | ID: mdl-30634133

ABSTRACT

OBJECTIVE: To investigate whether epigenetic modification of CD4+CD25- T-cells in vitro can make up for the inadequacy of CD4+CD25+Foxp3+ Treg in animal model of spontaneous abortion and prevent immune response-mediated spontaneous abortion. METHODS: Trichostatin A (TSA) was applied to inhibit histone deacetylases (HDACs) and thereby to epigenetically modify the special location of Foxp3 gene in CD4+CD25- T-cells of CBA/J mice. The expressions of CD25, Foxp3, CTLA-4 and PD-1 of CD4+ T cells isolated from spleen of mice were characterized by flow cytometric analysis. Concentrations of transforming growth factor- ß (TGF-ß) and IL-10 in the supernatants of cultured Treg were measured using ELISA. The purified CD4+ T cells treated with different reagents were injected into pregnant CBA/J mice mated with DBA/2J males on Day 1 and 4 of pregnancy, respectively. The embryo resorption rate was assessed on Day 14 of pregnancy. RESULTS: TSA treatment significantly increased the population of CD4+CD25+Foxp3+ iTreg. Those TSA induced Treg expressed high levels of PD-1 and CTLA-4, and secreted high levels of TGF-ß and IL-10. Adoptive transfer of those iTreg at both early stage and implantation of stage of pregnancy significantly increased population of CD4+CD25+Foxp3+ Treg in spleens of recipient miscarriage prone mice and significantly reduced resorption in those mice. CONCLUSION: Epigenetic regulation of Foxp3 can generate functional regulatory T-cells. Adoptive transfer of TSA- induced CD4+CD25+Foxp3+ Treg at an early stage of pregnancy can induce maternal-fetal immune tolerance and reduce embryo resorption in miscarriage prone mice.


Subject(s)
Adoptive Transfer , Embryo Loss/prevention & control , Hydroxamic Acids/pharmacology , T-Lymphocytes, Regulatory/transplantation , Animals , Antigens, Differentiation/immunology , Embryo Loss/immunology , Embryo Loss/pathology , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/immunology , Female , Male , Mice , Mice, Inbred BALB C , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
12.
J Reprod Immunol ; 125: 80-88, 2018 02.
Article in English | MEDLINE | ID: mdl-29306096

ABSTRACT

Cytokines in the reproductive tract environment at conception mediate a dialogue between the embryo and maternal tissues to profoundly influence embryo development and implantation success. Through effects on gene expression and the cell stress response, cytokines elicit an epigenetic impact with consequences for placental development and fetal growth, which in turn affect metabolic phenotype and long-term health of offspring. There is substantial evidence demonstrating that pro-survival cytokines, such as GM-CSF, CSF1, LIF, HB-EGF and IGFII, support embryos to develop optimally. Less attention has been paid to cytokines that adversely impact embryo development, including the pro-inflammatory cytokines TNF, TRAIL and IFNG. These agents elicit cell stress, impair cell survival and retard blastocyst development, and at sufficiently high concentrations, can cause embryo demise. Experiments in mice suggest these so-called 'embryotoxic' cytokines can harm embryos through pro-apoptotic and adverse programming effects, as well as indirectly suppressing uterine receptivity through the maternal immune response. Embryotrophic factors may mitigate against and protect from these adverse effects. Thus, the balance between embryotrophic and embryotoxic cytokines can impart effects on embryo development and implantation, and has the potential to contribute to endometrial 'biosensor' function to mediate embryo selection. Embryotoxic cytokines can be elevated in plasma and reproductive tract tissues in inflammatory conditions including infection, diabetes, obesity, PCOS and endometriosis. Studies are therefore warranted to investigate whether excessive embryotoxic cytokines contribute to infertility and recurrent implantation failure in women, and compromised reproductive performance in livestock animals.


Subject(s)
Cytokines/immunology , Embryo Loss/immunology , Embryonic Development/immunology , Endometrium/immunology , Fetal Development/immunology , Animals , Cytokines/blood , Cytokines/metabolism , Embryo Loss/blood , Embryo, Mammalian/immunology , Endometrium/metabolism , Female , Fetal Growth Retardation/immunology , Humans , Infertility/immunology , Metabolic Diseases/immunology , Placentation/immunology , Pregnancy , Prenatal Exposure Delayed Effects/immunology
13.
Am J Reprod Immunol ; 77(3)2017 Mar.
Article in English | MEDLINE | ID: mdl-28045211

ABSTRACT

PROBLEM: Increasing evidence demonstrates that inflammatory cytokines are involved in LPS-induced adverse pregnant outcomes including early embryo loss. Vitamin D3 (VitD3) has anti-inflammatory activity. We aimed to investigate the effects of vitamin D3 (VitD3) on LPS-induced early embryo loss in mice. METHOD OF STUDY: All pregnant mice except controls were intraperitoneally (ip) injected with LPS on GD7. In VitD3 alone and LPS+VitD3 groups, pregnant mice were pretreated with VitD3 by gavage daily from GD5 to GD7. RESULTS: LPS caused 62.5% pregnant mice with early embryo loss. Interestingly, the rate of abortion dropped to 14.3% when pregnant mice were pretreated with VitD3. Additional experiment showed that VitD3 significantly attenuated LPS-evoked elevation on TNF-α, IFN-γ, MIP-2, and nitrate plus nitrite in maternal serum. In addition, VitD3 alleviated LPS-induced COX-2 expression in the decidua and attenuated the elevation of PGF2α in maternal serum. Although VitD3 had no effect on IL-10 in maternal serum, it induced further elevation of serum IL-10 level in LPS-treated mice. Further analysis showed that VitD3 activated VDR signaling, simultaneously inhibited LPS-induced nuclear translocation of NF-κB p65 subunits in the decidua. CONCLUSIONS: VitD3 protects mice from LPS-induced early embryo loss at least partially through its anti-inflammatory effects.


Subject(s)
Anti-Inflammatory Agents/immunology , Cholecalciferol/immunology , Embryo Loss/prevention & control , Inflammation/immunology , Administration, Oral , Animals , Cyclooxygenase 2/metabolism , Cytokines/blood , Decidua/metabolism , Dinoprost/blood , Embryo Loss/immunology , Female , Humans , Inflammation Mediators/blood , Lipopolysaccharides/immunology , Mice , Mice, Inbred ICR , NF-kappa B/metabolism , Pregnancy
14.
Reprod Fertil Dev ; 30(1): 50-55, 2017 Jan.
Article in English | MEDLINE | ID: mdl-29539302

ABSTRACT

The question of 'how does the allogeneic fetus survive gestation in the face of the maternal immune system?' has yet to be definitively answered. Several acceptable mechanisms exist to facilitate survival of the semi-allogeneic fetus in various species; paramount is the immunological separation of maternal and fetal tissues during gestation. However, keen observation of the maternal immune system during pregnancy has noted maternal immune tolerance to paternal-specific antigens. A mechanism by which the maternal immune system tolerates specific paternal antigens expressed on the fetus would be far more beneficial than the previously proposed immune indolence that would leave the mother susceptible to infection. In species like human or rodent, implantation occurs days after fertilisation and, as such, the mechanisms to establish antigen-specific tolerance must be initiated very early during pregnancy. We and others propose that these mechanisms are initiated at the time of insemination when paternal antigens are first introduced to the maternal immune system. Indeed, a new paradigm demonstrating the importance of paternal-maternal communication at the time of insemination is becoming evident as it relates to maternal tolerance to fetal antigen and ultimately pregnancy success.


Subject(s)
Fertilization/immunology , Immune Tolerance/physiology , Pregnancy Outcome , Semen/immunology , Sperm-Ovum Interactions/immunology , Animals , Embryo Loss/immunology , Female , Fertilization/physiology , Humans , Male , Pregnancy , Sperm-Ovum Interactions/physiology
15.
Placenta ; 47: 29-36, 2016 11.
Article in English | MEDLINE | ID: mdl-27780537

ABSTRACT

INTRODUCTION: The successful pregnancy depends on maternal immune tolerance against the fetus. It has been reported that MSCs (mesenchymal stem cells) could play a regulatory role on immune cells such as CD4+T cells, macrophages and NK cells, but their effect on recurrent miscarriage is unknown. STUDY DESIGN: In a prospective study, the abortion-prone (CBA/J × DBA/2) H-2d × H-2k mice were utilized. Female CBA/J mice (8-10 weeks old) were injected with vehicle or MSCs via tail vein or uterine horns, and 14 days later, they were mated with DBA/2 males for the following experiments. RESULTS: Comparing with the control group, the embryo resorption rate in MSCs-horn injection group was dramatically decreased. MSCs were mainly located at the maternal-fetal interface, indicating that the reduction of resorption rate was due to MSCs' local effect. No matter which treatment was given, there was no significant difference in the levels of IL-4, IL-10, TNF-α and IFN-γ in CD4+T cells and IL-10 and IL-12 in macrophages in spleens among each group. However, in contrast to other groups, the levels of IL-4 and IL-10 in CD4+T cells localized at the maternal-fetal interface in MSCs-horn injection group were dramatically increased, and TNF-α and IFN-γ levels were notably decreased. While IL-10 expressed in macrophages was obviously higher than other groups and IL-12 in macrophages was significantly lower than other groups. CONCLUSIONS: The findings indicate that MSCs injection through uterine horns could decrease embryo resorption rate.


Subject(s)
Abortion, Habitual/immunology , Bone Marrow Cells/immunology , Embryo Loss/immunology , Immune Tolerance/physiology , Mesenchymal Stem Cells/immunology , Animals , Decidua/immunology , Female , Mice , Pregnancy , Pregnancy Outcome
16.
Am J Reprod Immunol ; 76(1): 38-49, 2016 07.
Article in English | MEDLINE | ID: mdl-27094728

ABSTRACT

PROBLEM: CpG oligodeoxynucleotides (ODNs) can induce immunological changes in non-obese diabetic (NOD) mice and increase embryo loss, but little is known about the mechanism. This study aimed to determine the role of adiponectin in CpG ODN-induced pregnancy failure. METHOD OF STUDY: Oligodeoxynucleotide 1826 was intraperitoneally injected to NOD mice, and ODN 2216, ODN 2006, and ODN 2395 were used to stimulate human trophoblast cell lines to investigate adiponectin expression patterns and its possible effects on trophoblast function. RESULTS: CpG ODNs downregulated adiponectin via the cJun N-terminal kinase signaling pathway and led to increased embryo loss (from 6.9 to 33.3%). ODN 2006 impaired human trophoblast cell migration, which was successfully rescued by adiponectin treatment. CONCLUSION: CpG ODNs decreased placental adiponectin expression in NOD mice and impaired human trophoblast function and was associated with increased embryo loss. Adiponectin may therefore play an important protective role in the prevention of bacteria-induced pregnancy failure.


Subject(s)
Adiponectin/immunology , Down-Regulation/drug effects , Embryo Loss , Oligodeoxyribonucleotides/adverse effects , Placenta/immunology , Animals , Down-Regulation/immunology , Embryo Loss/chemically induced , Embryo Loss/immunology , Female , Humans , Mice , Mice, Inbred NOD , Oligodeoxyribonucleotides/pharmacology , Pregnancy
17.
Mol Immunol ; 71: 87-97, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26874827

ABSTRACT

Surfactant associated protein D (SFTPD, also known as SP-D), a pattern recognition molecule, is an integral component of the mucosal immune system of female reproductive tract (FRT). In addition to host defense functions in the FRT, recent evidences indicate immunomodulatory role of SFTPD in parturition and pre-term labor. Regulation of SFTPD expression by ovarian hormones in the mouse uterus implicates SFTPD of FRT in pregnancy establishment and maintenance. In the current study, we attempted to decipher the functional relevance of SFTPD in FRT by characterizing the fertility parameters of surfactant associated protein D knockout (Sftpd(tm1Jhf)/Sftpd(tm1Jhf)) female mice. Knockout female mice exhibited extended estrous cycle with altered serum profile of ovarian hormones. We also demonstrate altered expression of ovarian hormone receptors and hormone responsive genes ITGB1, LIF and HOXA10 in uteri of these mice. Knockout females mated with wild type males had significantly smaller litter size due to increased pre-implantation embryo loss. We also observed an altered immune profile in knockout mice uteri with elevated levels of inflammatory cytokines, increased numbers of pro-inflammatory monocytes/macrophages and lower FOXP3 levels during the pre-implantation period. LPS administration to pregnant knockout mice did not result in any increase in embryo implantation loss and was associated with a blunted uterine pro-inflammatory response, plausibly due to higher levels of serum progesterone. Taken together, our results demonstrate that SFTPD deficiency affects female fertility, highlighting roles for SFTPD in ovarian and uterine physiology.


Subject(s)
Embryo Loss/immunology , Fertility/immunology , Gonadal Hormones/immunology , Immunity, Mucosal/immunology , Pulmonary Surfactant-Associated Protein D/immunology , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Immunoblotting , Mice , Mice, Knockout , Ovary/immunology , Pregnancy , Real-Time Polymerase Chain Reaction , Uterus/immunology
18.
Biochem Biophys Res Commun ; 469(3): 704-10, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26686416

ABSTRACT

OBJECTIVE: To elucidate the mechanism by which embryo-resorption was enhanced by pathogenic CpG ODN motif in abortion-prone CBA/J × DBA/2 model and to develop a counter strategy for normal pregnancy outcome. METHODS: This is an animal model-based study. Abortion-prone model is established by CBA/J × DBA/2. An infection was mimicked by CpG ODN injection. RESULTS: Embryo-resorption was readily induced by CpG ODN in low doses of CpG ODN (∼25 µg/dam) when intraperitoneally (IP) injected on gestational day(gd) 6.5 in male DBA/2 mated CBA/J female mice. A more modest decline in Progesterone(P4), but not Estrogen(E2) was observed after exposure to CpG ODN in the model. P4 supplement fail to improve pregnancy outcomes, even at pharmocology dose. CpG ODN-induced fetal resorption is prevented by the treatment of anti-F4/80 or by that of anti-TNFα.In the implantation sites, the treatment of anti-F4/80 inhibits the increase both of F4/80(+) macrophage proportion and TNF-αexpression level which are induced by CpG ODN. The anti-TNFαtreatment also recovers CpG ODN-induced reduction of CD4(+)Foxp3(+) T cells. CONCLUSION: Circulating P4 is not responsible for the process by which CpG ODN-induced embryonic resorption in an abortion-prone mice. Macrophage depletion and TNF-α inhibition are really noteworthy for CpG ODN-induced pregnancy disruption.


Subject(s)
CpG Islands , Disease Models, Animal , Embryo Loss/immunology , Embryo Loss/prevention & control , Macrophages/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Embryo Loss/chemically induced , Female , Macrophages/pathology , Male , Mice , Mice, Inbred CBA , Mice, Inbred DBA , Oligodeoxyribonucleotides , Pregnancy , Pregnancy Outcome , Pregnancy, Animal
19.
Bull Exp Biol Med ; 159(1): 53-7, 2015 May.
Article in English | MEDLINE | ID: mdl-26033590

ABSTRACT

Muramyl dipeptide ß-heptylglycoside (C7MDP) was administered to non-pregnant CBA female mice and pregnant mice after non-abortion-prone mating (CBA×BALB/c) and mating associated with a high rate of spontaneous abortion (CBA×DBA/2). In non-pregnant females, C7MDP increased the production of IL-2, IL-4, IL-5, IL-6, IL-17, IFNγ, TNFα, and GM-CSF at constant production of IL-1α and IL-10. C7MDP increased the production of IL-10 and IL-17 and suppressed the production of IFNγ on day 8 of gestation in non-abortion-prone mouse couples and stimulated the synthesis of IL-4 and IFNγ, reduced IL-5 production, and slightly increased IL-1α secretion after abortion-prone mating. On day 14 of gestation, C7MDP elevated the yield of IL-2, IL-4, IFNγ, TNFα, and GM-CSF in CBA×BALB/c and CBA×DBA/2 couples and IL-17 in the fi rst variant of mating.


Subject(s)
Adjuvants, Immunologic/toxicity , Cytokines/biosynthesis , Embryo Loss/chemically induced , Glycopeptides/toxicity , Lymphocyte Subsets/drug effects , Abortion, Spontaneous/genetics , Abortion, Spontaneous/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Cells, Cultured , Crosses, Genetic , Cytokines/genetics , Disease Models, Animal , Embryo Loss/genetics , Embryo Loss/immunology , Female , Glycopeptides/administration & dosage , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Injections, Intraperitoneal , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interleukins/biosynthesis , Interleukins/genetics , Lymphocyte Subsets/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred CBA , Mice, Inbred DBA , Pregnancy , Spleen/cytology , Spleen/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
20.
Hum Reprod Update ; 21(3): 378-87, 2015.
Article in English | MEDLINE | ID: mdl-25634660

ABSTRACT

BACKGROUND: Thyroid hormone disorders and thyroid peroxidase autoantibodies (TPO-Ab) in women are associated with subfertility and early pregnancy loss. Here, we aim to provide a comprehensive overview of the literature on the pathophysiology of these associations. METHODS: A review of the literature in the English language was carried out. Relevant studies were identified by searching Medline, EMBASE and the Cochrane Controlled Trials Register from 1975 until March 2014. RESULTS: From a total of 6108 primary selected articles from the literature search, 105 articles were selected for critical appraisal. Observational data indicate that altered thyroid hormone levels are associated with disturbed folliculogenesis, spermatogenesis, lower fertilization rates and lower embryo quality. Triiodothyronine (T3) in combination with FSH enhances granulosa cell proliferation and inhibits granulosa cell apoptosis by the PI3K/Akt pathway. T3 is considered a biological amplifier of the stimulatory action of gonadotrophins on granulosa cell function. T3 increases the expression of matrix metalloproteinases (MMP), MMP-2, MMP-3, fetal fibronectin and integrin α5ß1T3 in early placental extravillous trophoblasts. Thyroid hormone transporters and receptors are expressed in the ovary, early embryo, endometrium, uterus and placenta. No other data explaining the associations could be retrieved from the literature. The presence of TPO-Ab is negatively associated with spermatogenesis, fertilization and embryo quality, but no data are available on the potential pathophysiological mechanisms. CONCLUSIONS: Thyroid hormone disorders and TPO-Ab are associated with disturbed folliculogenesis, spermatogenesis, fertilization and embryogenesis. The pathophysiology of these associations remains largely unknown, as evidence is limited and includes studies using small sample sizes, and often restricted to animal models. There are no studies on the pathophysiology underlying the association between TPO-Ab and reproduction. The available evidence, although limited, supports a role of thyroid hormone in fertility and early pregnancy. This justifies clinical intervention studies on the effects of thyroid hormone supplementation in women with subclinical hypothyroidism and in women prone to develop hypothyroidism due to the presence of TPO-Ab. In addition, more research is needed to identify the underlying mechanisms. This would be of particular interest in women undergoing IVF to pinpoint the effects of thyroid hormone on different parameters of reproduction.


Subject(s)
Autoantibodies/immunology , Embryonic Development/physiology , Hypothyroidism/pathology , Iodide Peroxidase/immunology , Triiodothyronine/metabolism , Apoptosis/immunology , Cell Proliferation/physiology , Embryo Loss/immunology , Female , Follicle Stimulating Hormone/metabolism , Granulosa Cells/cytology , Humans , Models, Animal , Ovarian Follicle/cytology , Ovarian Follicle/immunology , Phosphatidylinositol 3-Kinases , Placenta/physiology , Pregnancy , Reproduction/immunology , Reproduction/physiology , Spermatogenesis/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...