13 research outputs found

    Monocarboxylate transporter 8 modulates the viability and invasive capacity of human placental cells and fetoplacental growth in mice

    Get PDF
    Monocarboxylate transporter 8 (MCT8) is a well-established thyroid hormone (TH) transporter. In humans, MCT8 mutations result in changes in circulating TH concentrations and X-linked severe global neurodevelopmental delay. MCT8 is expressed in the human placenta throughout gestation, with increased expression in trophoblast cells from growth-restricted pregnancies. We postulate that MCT8 plays an important role in placental development and transplacental TH transport. We investigated the effect of altering MCT8 expression in human trophoblast in vitro and in a Mct8 knockout mouse model. Silencing of endogenous MCT8 reduced T3 uptake into human extravillous trophoblast-like cells (SGHPL-4; 40%, P<0.05) and primary cytotrophoblast (15%, P<0.05). MCT8 over-expression transiently increased T3 uptake (SGHPL-4∶30%, P<0.05; cytotrophoblast: 15%, P<0.05). Silencing MCT8 did not significantly affect SGHPL-4 invasion, but with MCT8 over-expression T3 treatment promoted invasion compared with no T3 (3.3-fold; P<0.05). Furthermore, MCT8 silencing increased cytotrophoblast viability (∼20%, P<0.05) and MCT8 over-expression reduced cytotrophoblast viability independently of T3 (∼20%, P<0.05). In vivo, Mct8 knockout reduced fetal:placental weight ratios compared with wild-type controls at gestational day 18 (25%, P<0.05) but absolute fetal and placental weights were not significantly different. The volume fraction of the labyrinthine zone of the placenta, which facilitates maternal-fetal exchange, was reduced in Mct8 knockout placentae (10%, P<0.05). However, there was no effect on mouse placental cell proliferation in vivo. We conclude that MCT8 makes a significant contribution to T3 uptake into human trophoblast cells and has a role in modulating human trophoblast cell invasion and viability. In mice, Mct8 knockout has subtle effects upon fetoplacental growth and does not significantly affect placental cell viability probably due to compensatory mechanisms in vivo

    The maternal microbiome during pregnancy and allergic disease in the offspring

    Get PDF
    There is substantial epidemiological and mechanistic evidence that the increase in allergic disease and asthma in many parts of the world in part relates to changes in microbial exposures and diet acting via the composition and metabolic products of the intestinal microbiome. The majority of research in this field has focused on the gut microbiome during infancy, but it is increasingly clear that the maternal microbiome during pregnancy also has a key role in preventing an allergy-prone immune phenotype in the offspring. The mechanisms by which the maternal microbiome influences the developing fetal immune system include alignment between the maternal and infant regulatory immune status and transplacental passage of microbial metabolites and IgG. Interplay between microbial stimulatory factors such as lipopolysaccharides and regulatory factors such as short-chain fatty acids may also influence on fetal immune development. However, our understanding of these pathways is at an early stage and further mechanistic studies are needed. There are also no data from human studies relating the composition and metabolic activity of the maternal microbiome during pregnancy to the offspring's immune status at birth and risk of allergic disease. Improved knowledge of these pathways may inform novel strategies for tackling the increase in allergic disorders in the modern world

    MINocyclinE to Reduce inflammation and blood brain barrier leakage in small Vessel diseAse (MINERVA) trial study protocol.

    No full text
    BACKGROUND: Cerebral small vessel disease (SVD) is a common cause of stroke and cognitive impairment. Recent data has implicated neuroinflammation and increased blood-brain barrier (BBB) permeability in its pathogenesis, but whether such processes are causal and can be therapeutically modified is uncertain. In a rodent model of SVD, minocycline was associated with reduced white matter lesions, inflammation and BBB permeability. AIMS: To determine whether blood-brain barrier permeability (measured using dynamic contrast-enhanced MRI) and microglial activation (measured by positron emission tomography using the radioligand 11C-PK11195) can be modified in SVD. DESIGN: Phase II randomised double blind, placebo-controlled trial of minocycline 100 mg twice daily for 3 months in 44 participants with moderate to severe SVD defined as a clinical lacunar stroke and confluent white matter hyperintensities. OUTCOMES: Primary outcome measures are volume and intensity of focal increases of blood-brain barrier permeability and microglial activation determined using PET-MRI imaging. Secondary outcome measures include inflammatory biomarkers in serum, and change in conventional MRI markers and cognitive performance over 1 year follow up. DISCUSSION: The MINERVA trial aims to test whether minocycline can influence novel pathological processes thought to be involved in SVD progression, and will provide insights into whether central nervous system inflammation in SVD can be therapeutically modulated

    Stereohistological analysis of male wild-type (+/y) and male Mct8 knockout (−/y) E18.5 placentae.

    No full text
    <p><b>A</b>: Representative picture of a placental section (wild-type), stained with hematoxylin and eosin, and photographed using a 2× objective lens. The different regions of the mouse placenta are annotated: db = decidua basalis, Lz = labyrinthine zone, Jz = junctional zone and c = chorionic plate. <b>B–C</b>: Volume fractions (B) of the placental regions relative to the whole placenta and absolute volumes (C) of the placental regions in wild-type and litter-matched Mct8 knockout samples. Six placentae were assessed per genotype. The bars represent the mean ±SEM for each group. Significant differences between groups are indicated by *P<0.05.</p

    T3 uptake in primary cytotrophoblast cells following MCT8 silencing (A,C) or MCT8 over-expression (B,D).

    No full text
    <p><b>A and B</b>: T3 uptake time course from one representative experiment. Cytotrophoblast cells were incubated for 0 to 30 minutes with [<sup>125</sup>I]-T3. The amount of intracellular radioactivity was quantified and expressed as a percentage of the total radioactivity added. <b>C and D</b>: Relative T3 uptake following incubation for 10 minutes with [<sup>125</sup>I]-T3. Bars represent average of four experiments +SEM. Within each experiment, each condition was performed in duplicate. The mean uptake for the control within each experiment has been assigned the value of 100%. Statisticallysignificant differences are indicated by *P<0.05 and **P<0.01.</p

    Effect of MCT8 silencing (A,C,E) and MCT8 over-expression (B,D,F) on the viability of the EVT-like SGHPL-4 cells.

    No full text
    <p>assessed by MTT assay (A–B), and apoptosis by quantification of Caspase 3/7 activity (C–D) and counting the number of apoptotic cells by time lapse microscopy (E–F). The assays were performed 72 hours following silencing of endogenous MCT8 or transfection with human wild-type MCT8, and 48 hours post-treatment with 0 or 10 nM T3. <b>A and B</b>: The absorbance for each sample was normalized to the average absorbance measured in cells transfected with control (control siRNA or VO) and treated with 0 nM T3. The bars represent average of five experiments +SEM. Four replicates were performed per condition within each experiment. <b>C and D</b>: Results were normalized to the MTT results in order to correct for cell numbers. Bars represent average of three experiments +SEM. Within each experiment, each condition was performed in triplicate. <b>E and F:</b> Bars represent average of three experiments +SEM. Within each experiment 20 cells were counted per condition.</p
    corecore