16 research outputs found

    Probing the potential of mucus permeability to signify preterm birth risk

    Get PDF
    Preterm birth is the leading cause of neonatal mortality, and is frequently associated with intra-amniotic infection hypothesized to arise from bacterial ascension across a dysfunctional cervical mucus plug. To study this dysfunction, we assessed the permeability of cervical mucus from non-pregnant ovulating (n = 20) and high-(n = 9) and low-risk (n = 16) pregnant women to probes of varying sizes and surface chemistries. We found that the motion of negatively charged, carboxylated microspheres in mucus from pregnant patients was significantly restricted compared to ovulating patients, but not significantly different between high-A nd low-risk pregnant women. In contrast, charged peptide probes small enough to avoid steric interactions, but sensitive to the biochemical modifications of mucus components exhibited significantly different transport profiles through mucus from high-A nd low-risk patients. Thus, although both microstructural rearrangements of the components of mucus as well as biochemical modifications to their adhesiveness may alter the overall permeability of the cervical mucus plug, our findings suggest that the latter mechanism plays a dominant role in the impairment of the function of this barrier during preterm birth. We expect that these probes may be readily adapted to study the mechanisms underlying disease progression on all mucosal epithelia, including those in the mouth, lungs, and gut.National Science Foundation (U.S.) (Award DMR-0819762)National Science Foundation (U.S.) (Award 1122374

    Differential expression of collectins in human placenta and role in inflammation during spontaneous Labor.

    Get PDF
    © 2014 Yadav et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.Collectins, collagen-containing Ca2+ dependent C-type lectins and a class of secretory proteins including SP-A, SP-D and MBL, are integral to immunomodulation and innate immune defense. In the present study, we aimed to investigate their placental transcript synthesis, labor associated differential expression and localization at feto-maternal interface, and their functional implication in spontaneous labor. The study involved using feto-maternal interface (placental/decidual tissues) from two groups of healthy pregnant women at term (≥37 weeks of gestation), undergoing either elective C-section with no labor ('NLc' group, n = 5), or normal vaginal delivery with spontaneous labor ('SLv' group, n = 5). The immune function of SP-D, on term placental explants, was analyzed for cytokine profile using multiplexed cytokine array. SP-A, SP-D and MBL transcripts were observed in the term placenta. The 'SLv' group showed significant up-regulation of SP-D (p = 0.001), and down-regulation of SP-A (p = 0.005), transcripts and protein compared to the 'NLc' group. Significant increase in 43 kDa and 50 kDa SP-D forms in placental and decidual tissues was associated with the spontaneous labor (p<0.05). In addition, the MMP-9-cleaved form of SP-D (25 kDa) was significantly higher in the placentae of 'SLv' group compared to the 'NLc' group (p = 0.002). Labor associated cytokines IL-1α, IL-1β, IL-6, IL-8, IL-10, TNF-α and MCP-1 showed significant increase (p<0.05) in a dose dependent manner in the placental explants treated with nSP-D and rhSP-D. In conclusion, the study emphasizes that SP-A and SP-D proteins associate with the spontaneous labor and SP-D plausibly contributes to the pro-inflammatory immune milieu of feto-maternal tissues.Funding provided by BT/PR15227/BRB/10/906/2011) Department of Biotechnology (DBT), Government of India http://dbtindia.nic.in/index.asp (TM) and Indian Council of Medical Research (ICMR) Junior Research Fellowship (JRF)/Senior Research Fellowship (SRF), Government of India, www.icmr.nic.in (AKY)

    Primate-specific evolution of noncoding element insertion into PLA2G4C and human preterm birth

    Get PDF
    Background The onset of birth in humans, like other apes, differs from non-primate mammals in its endocrine physiology. We hypothesize that higher primate-specific gene evolution may lead to these differences and target genes involved in human preterm birth, an area of global health significance. Methods We performed a comparative genomics screen of highly conserved noncoding elements and identified PLA2G4C, a phospholipase A isoform involved in prostaglandin biosynthesis as human accelerated. To examine whether this gene demonstrating primate-specific evolution was associated with birth timing, we genotyped and analyzed 8 common single nucleotide polymorphisms (SNPs) in PLA2G4C in US Hispanic (n = 73 preterm, 292 control), US White (n = 147 preterm, 157 control) and US Black (n = 79 preterm, 166 control) mothers. Results Detailed structural and phylogenic analysis of PLA2G4C suggested a short genomic element within the gene duplicated from a paralogous highly conserved element on chromosome 1 specifically in primates. SNPs rs8110925 and rs2307276 in US Hispanics and rs11564620 in US Whites were significant after correcting for multiple tests (p < 0.006). Additionally, rs11564620 (Thr360Pro) was associated with increased metabolite levels of the prostaglandin thromboxane in healthy individuals (p = 0.02), suggesting this variant may affect PLA2G4C activity. Conclusions Our findings suggest that variation in PLA2G4C may influence preterm birth risk by increasing levels of prostaglandins, which are known to regulate labor.Children’s Discovery InstituteMarch of Dimes Birth Defects FoundationNational Institute of General Medical Sciences (U.S.) (grant T32 GM081739)Washington University (Saint Louis, Mo.) (Mr. and Mrs. Spencer T. Olin Fellowship for Women in Graduate Study)Sigrid Jusélius FoundationSigne and Anne Gyllenberg FoundationAcademy of FinlandVanderbilt University (Turner-Hazinski grant award

    Ovarian cancer stem cells: still an elusive entity?

    Full text link

    Role of GnRH-GnRH receptor signaling at the maternal-fetal interface

    No full text
    Objective: To investigate the expression and function of GnRH and GnRH receptor (GnRHR) subtypes at the maternal-fetal interface. Design: In vitro experiments using freshly isolated human trophoblast cells, decidual stromal cells (DSCs), and immortalized cell lines. Setting: University teaching hospital. Patient(S): Placenta-fetal membranes from term deliveries. Intervention(s): Human trophoblast and DSCs were isolated, purified, and cultured. Main Outcome Measure(s): Expression of GnRH-I, GnRH-II, and GnRHR-I mRNA and protein in human trophoblast cell lines and tissues were evaluated by reverse-transcription polymerase chain reaction and Western blot. The effect of GnRH-I and -II on the production of select cytokines (hCG, interleukin [IL] 8, IL-6, matrix metalloproteinase 3, monocyte chemoattractant protein 1, vascular endothelial growth factor, soluble Fms-like tyrosine kinase 1, urokinase-type plasminogen activator, and plasminogen activator inhibitor 1) were measured by ELISA and normalized for protein content. Result(s): GnRH-I, GnRH-II, and GnRHR-I mRNA and protein were identified in trophoblasts and decidua. GnRH-I and -II stimulated hCG production by trophoblast and trophoblast-derived cell lines in a dose-dependent fashion (e. g., 2.8-fold, from 2.5 +/- 0.5 to 7.0 +/- 0.4 ng/mg protein per 24 h, for 1,000 nmol/L GnRH-I and 2.4-fold, from 2.5 +/- 0.5 to 6.1 +/- 0.6 ng/mg protein per 24 h, for 1,000 nmol/L GnRH-II) without affecting the production of other cytokines. Conclusion(s): Trophoblasts and decidua express GnRH-I, GnRH-II, and GnRHR-I mRNA and protein. GnRH-I and -II selectively stimulate hCG production by trophoblast cells without altering the production of select cytokines by trophoblasts or decidua. The role of GnRH-GnRHR signaling at the maternal-fetal interface therefore appears to be limited to the regulation of trophoblast hCG production. (Fertil Steril (R) 2010;94:2680-7. (C) 2010 by American Society for Reproductive Medicine.)Snegovskikh VV, 2009, REPROD SCI, V16, P767, DOI 10.1177/1933719109336623Fest S, 2008, AM J REPROD IMMUNOL, V59, P75, DOI 10.1111/j.1600-0897.2007.00557.xMeade ES, 2007, PLACENTA, V28, P1012, DOI 10.1016/j.placenta.2607.04.005Lu ZL, 2007, J BIOL CHEM, V282, P17921, DOI 10.1074/jbc.M610413200Lockwood CJ, 2007, AM J PATHOL, V170, P1398, DOI 10.2353/ajpath.2007.060465NORWITZ ER, 2007, AM J OBSTET GYNECOL, V196, pNI523Elustondo PA, 2006, BIOL REPROD, V74, P959, DOI 10.1095/biolreprod.105.050419Wang HX, 2006, J CELL PHYSIOL, V206, P616, DOI 10.1002/jcp.20508Lu ZL, 2005, J BIOL CHEM, V280, P29796, DOI 10.1074/jbc.M413520200Lockwood CJ, 2005, J CLIN ENDOCR METAB, V90, P4710, DOI 10.1210/jc.2004-2528Pawson AJ, 2005, ENDOCRINOLOGY, V146, P2639, DOI 10.1210/en.2005-0133Mackenzie AP, 2004, AM J OBSTET GYNECOL, V191, P1996, DOI 10.1016/j.ajog.2004.08.003Rama S, 2004, MOL CELL ENDOCRINOL, V218, P79, DOI 10.1016/j.mce.2003.12.016Koopman LA, 2003, J EXP MED, V198, P1201, DOI 10.1084/jem.20030305Chou CS, 2003, J CLIN ENDOCR METAB, V88, P4781, DOI 10.1210/jc.2003-030659Chou CS, 2003, J CLIN ENDOCR METAB, V88, P3806, DOI 10.1210/jc.2002-021955Morgan K, 2003, ENDOCRINOLOGY, V144, P423, DOI 10.1210/en.2002-220622Chou CS, 2003, J CLIN ENDOCR METAB, V88, P680, DOI 10.1210/jc.2002.021277Millar RP, 2003, TRENDS ENDOCRIN MET, V14, P35Chou CS, 2002, J CLIN ENDOCR METAB, V87, P5594, DOI 10.1210/jc.2002-020883van Biljon W, 2002, BIOL REPROD, V67, P1741, DOI 10.1095/bioreprod.101.002808Pfleger KDG, 2002, MOL ENDOCRINOL, V16, P2155, DOI 10.1210/me.2002-0159Grundker C, 2002, J CLIN ENDOCR METAB, V87, P1427Neill JD, 2002, ENDOCRINOLOGY, V143, P737ROSEN T, 2002, J MATERN-FETAL NEO M, V11, P11Faurholm B, 2001, GENOMICS, V78, P15Lala PK, 2001, PLACENTA, V22, P889Millar R, 2001, P NATL ACAD SCI USA, V98, P9636Siler-Khodr TM, 2001, J SOC GYNECOL INVEST, V8, P233Neill JD, 2001, BIOCHEM BIOPH RES CO, V282, P1012Siler-Khodr TM, 2001, J CLIN ENDOCR METAB, V86, P804Wang L, 2001, P NATL ACAD SCI USA, V98, P361Islami D, 2001, MOL HUM REPROD, V7, P3Nathwani PS, 2000, ENDOCRINOLOGY, V141, P1754Di Simone N, 2000, ARTHRITIS RHEUM, V43, P140Millar R, 1999, J ENDOCRINOL, V162, P117Raga F, 1999, J CLIN ENDOCR METAB, V84, P636White RB, 1998, P NATL ACAD SCI USA, V95, P305Emons G, 1997, TRENDS ENDOCRIN MET, V8, P355KAKAR SS, 1994, MOL CELL ENDOCRINOL, V106, P145GULLER S, 1993, J STEROID BIOCHEM, V46, P1GRAHAMCH, 1993, EXP CELL RES, V206, P204BRAMLEY TA, 1992, PLACENTA, V13, P555BELISLE S, 1989, J CLIN ENDOCR METAB, V69, P117KIM SJ, 1987, PLACENTA, V8, P257KLIMAN HJ, 1986, ENDOCRINOLOGY, V118, P1567KAIHO T, 1984, NIPPON SANKA FUJINHA, V36, P1801POPKIN R, 1983, BIOCHEM BIOPH RES CO, V114, P750MIYAKE A, 1982, OBSTET GYNECOL, V60, P444BUTZOW R, 1982, INT J CANCER, V15, P9CURRIE AJ, 1981, BIOCHEM BIOPH RES CO, V99, P332KHODR GS, 1980, SCIENCE, V207, P315SILERKHODR TM, 1978, AM J OBSTET GYNECOL, V130, P216JAFFE RB, 1969, J CLIN ENDOCR METAB, V29, P1281

    Intra-amniotic Infection Upregulates Neutrophil Gelatinase-Associated Lipocalin (NGAL) Expression at the Maternal-Fetal Interface at Term

    No full text
    Objective: Neutrophil gelatinase-associated lipocalin (NGAL) is a ubiquitous lipocalin that serves as a critical component of innate immunity and a transport shuttle for numerous substances (retinoids, arachidonic acid, prostaglandins, fatty acids, steroids, iron, and MMPs). Despite the well-documented association between intra-amniotic infection/inflammation (IAI) and preterm birth, NGAL expression in the uterus has not previously been examined. This study investigates NGAL expression at the maternal-fetal interface in vivo and in vitro. Methods: Neutrophil gelatinase-associated lipocalin expression in term placenta with/without IAI was examined by immunohistochemistry. Trophoblast and decidual stromal cells were retrieved from elective cesarean, purified, and depleted of leukocytes. On days 1 (cytotrophoblast cells) and 4 (syncytiotrophoblast), cells were stimulated with/without interleukin 1β (IL-1β; 1 ng/mL), tumor necrosis factor α (TNF-α; 1 ng/mL), or lipopolysaccharide (LPS; 1 μg/mL). Neutrophil gelatinase-associated lipocalin messenger RNA (mRNA) and protein expression were measured by immunocytochemistry/Western blot and RT-qPCR, respectively. Results: Under basal conditions, NGAL is expressed in trophoblast, but not decidua. Trophoblast NGAL is significantly upregulated in tissues with evidence of IAI vs controls. NGAL expression was increased after stimulation with all 3 pro-inflammatory mediators in day 1 (cytotrophoblast) but not day 4 cells (syncytiotrophoblast). IL-1β and TNF-α (not LPS) upregulated NGAL gene expression in cytotrophoblast (not syncytiotrophoblast) cells. Conclusions: Intra-amniotic infection/inflammation is associated with increased expression of NGAL in trophoblast tissues in vivo. IL-1β, TNF-α, and LPS stimulated NGAL in cytotrophoblast cells (not syncytiotrophoblast and decidua) in vitro. These data suggest that, in keeping with its role as a mediator of innate immunity, NGAL may have a central role to play in IAI-induced preterm birth
    corecore