Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Birgit Hirschmugl is active.

Publication


Featured researches published by Birgit Hirschmugl.


Circulation Research | 2009

Human Endothelial Cells of the Placental Barrier Efficiently Deliver Cholesterol to the Fetal Circulation via ABCA1 and ABCG1

Jasminka Stefulj; Ute Panzenboeck; Tatjana Becker; Birgit Hirschmugl; Cornelia Schweinzer; Ingrid Lang; Gunther Marsche; Anton Sadjak; U Lang; Gernot Desoye; Christian Wadsack

Although maternal–fetal cholesterol transfer may serve to compensate for insufficient fetal cholesterol biosynthesis under pathological conditions, it may have detrimental consequences under conditions of maternal hypercholesterolemia leading to preatherosclerotic lesion development in fetal aortas. Maternal cholesterol may enter fetal circulation by traversing syncytiotrophoblast and endothelial layers of the placenta. We hypothesized that endothelial cells (ECs) of the fetoplacental vasculature display a high and tightly regulated capacity for cholesterol release. Using ECs isolated from human term placenta (HPECs), we investigated cholesterol release capacity and examined transporters involved in cholesterol efflux pathways controlled by liver-X-receptors (LXRs). HPECs demonstrated 2.5-fold higher cholesterol release to lipid-free apolipoprotein (apo)A-I than human umbilical vein ECs (HUVECs), whereas both cell types showed similar cholesterol efflux to high-density lipoproteins (HDLs). Interestingly, treatment of HPECs with LXR activators increased cholesterol efflux to both types of acceptors, whereas no such response could be observed for HUVECs. In line with enhanced cholesterol efflux, LXR activation in HPECs increased expression of ATP-binding cassette transporters ABCA1 and ABCG1, while not altering expression of ABCG4 and scavenger receptor class B type I (SR-BI). Inhibition of ABCA1 or silencing of ABCG1 decreased cholesterol efflux to apoA-I (−70%) and HDL3 (−57%), respectively. Immunohistochemistry localized both transporters predominantly to the apical membranes of placental ECs in situ. Thus, ECs of human term placenta exhibit unique, efficient and LXR-regulated cholesterol efflux mechanisms. We propose a sequential pathway mediated by ABCA1 and ABCG1, respectively, by which HPECs participate in forming mature HDL in the fetal blood.


Journal of Lipid Research | 2017

The influence of placental metabolism on fatty acid transfer to the fetus

Simone Perazzolo; Birgit Hirschmugl; Christian Wadsack; Gernot Desoye; Rohan M. Lewis; Bram G. Sengers

The factors determining fatty acid transfer across the placenta are not fully understood. This study used a combined experimental and computational modeling approach to explore placental transfer of nonesterified fatty acids and identify the rate-determining processes. Isolated perfused human placenta was used to study the uptake and transfer of 13C-fatty acids and the release of endogenous fatty acids. Only 6.2 ± 0.8% of the maternal 13C-fatty acids taken up by the placenta was delivered to the fetal circulation. Of the unlabeled fatty acids released from endogenous lipid pools, 78 ± 5% was recovered in the maternal circulation and 22 ± 5% in the fetal circulation. Computational modeling indicated that fatty acid metabolism was necessary to explain the discrepancy between uptake and delivery of 13C-fatty acids. Without metabolism, the model overpredicts the fetal delivery of 13C-fatty acids 15-fold. Metabolic rate was predicted to be the main determinant of uptake from the maternal circulation. The microvillous membrane had a greater fatty acid transport capacity than the basal membrane. This study suggests that incorporation of fatty acids into placental lipid pools may modulate their transfer to the fetus. Future work needs to focus on the factors regulating fatty acid incorporation into lipid pools.


International Journal of Obesity | 2017

Maternal obesity modulates intracellular lipid turnover in the human term placenta

Birgit Hirschmugl; Gernot Desoye; Patrick M. Catalano; I Klymiuk; H Scharnagl; S Payr; E Kitzinger; C Schliefsteiner; U Lang; Christian Wadsack; S Hauguel-de Mouzon

Background:Obesity before pregnancy is associated with impaired metabolic status of the mother and the offspring later in life. These adverse effects have been attributed to epigenetic changes in utero, but little is known about the role of placental metabolism and its contribution to fetal development.Objectives:We examined the impact of maternal pre-pregnancy obesity on the expression of genes involved in placental lipid metabolism in lean and obese women.Subjects/Methods:Seventy-three lean and obese women with healthy pregnancy were recruited at term elective cesarean delivery. Metabolic parameters were measured on maternal venous blood samples. Expression of 88 genes involved in lipid metabolism was measured in whole placenta tissue. Proteins of genes differently expressed in response to maternal obesity were quantified, correlated with maternal parameters and immunolocalized in placenta sections. Isolated primary trophoblasts were used for in vitro assays.Results:Triglyceride (TG) content was increased in placental tissue of obese (1.10, CI 1.04–1.24 mg g−1, P<0.05) vs lean (0.84, CI 0.72–1.02 mg g−1) women. Among target genes examined, six showed positive correlation (P<0.05) with maternal pre-pregnancy BMI, namely ATGL (PNPLA2), FATP1 (SLC27A1), FATP3 (SLC27A3), PLIN2, PPARG and CGI-58 (ABHD5). CGI-58 protein abundance was twofold higher (P<0.001) in placentas of obese vs lean women. CGI-58 protein levels correlated positively with maternal insulin levels and pre-pregnancy body mass index (R=0.63, P<0.001 and R=0.64, P<0.001, respectively). CGI-58 and PLIN2 were primarily located in the syncytiotrophoblast and, were upregulated (1.38- and 500-fold, respectively) upon oleic acid and insulin treatment of cultured trophoblast cells.Conclusion:Pre-gravid obesity significantly modifies the expression of placental genes related to transport and storage of neutral lipids. We propose that the upregulation of CGI-58, a master regulator of TG hydrolysis, contributes to the turnover of intracellular lipids in placenta of obese women, and is tightly regulated by metabolic factors of the mother.


Clinical Endocrinology | 2014

Cord blood chemerin: differential effects of gestational diabetes mellitus and maternal obesity

Mireille N. M. van Poppel; Willibald Zeck; Daniela Ulrich; Eva-Christina Schest; Birgit Hirschmugl; U Lang; Christian Wadsack; Gernot Desoye

Chemerin is a novel adipokine implicated in inflammation and obesity. We hypothesized that foetal chemerin would be elevated in gestational diabetes mellitus (GDM) and correlate with foetal and maternal adiposity.


PLOS ONE | 2016

TASK-1 Regulates Apoptosis and Proliferation in a Subset of Non-Small Cell Lung Cancers.

Katharina Leithner; Birgit Hirschmugl; Yingji Li; Bi Tang; Rita Papp; Chandran Nagaraj; Elvira Stacher; Philipp Stiegler; Jörg Lindenmann; Andrea Olschewski; Horst Olschewski; Andelko Hrzenjak

Lung cancer is the leading cause of cancer deaths worldwide; survival times are poor despite therapy. The role of the two-pore domain K+ (K2P) channel TASK-1 (KCNK3) in lung cancer is at present unknown. We found that TASK-1 is expressed in non-small cell lung cancer (NSCLC) cell lines at variable levels. In a highly TASK-1 expressing NSCLC cell line, A549, a characteristic pH- and hypoxia-sensitive non-inactivating K+ current was measured, indicating the presence of functional TASK-1 channels. Inhibition of TASK-1 led to significant depolarization in these cells. Knockdown of TASK-1 by siRNA significantly enhanced apoptosis and reduced proliferation in A549 cells, but not in weakly TASK-1 expressing NCI-H358 cells. Na+-coupled nutrient transport across the cell membrane is functionally coupled to the efflux of K+ via K+ channels, thus TASK-1 may potentially influence Na+-coupled nutrient transport. In contrast to TASK-1, which was not differentially expressed in lung cancer vs. normal lung tissue, we found the Na+-coupled nutrient transporters, SLC5A3, SLC5A6, and SLC38A1, transporters for myo-inositol, biotin and glutamine, respectively, to be significantly overexpressed in lung adenocarcinomas. In summary, we show for the first time that the TASK-1 channel regulates apoptosis and proliferation in a subset of NSCLC.


international conference of the ieee engineering in medicine and biology society | 2015

Computational modelling of fatty acid transport in the human placenta

Simone Perazzolo; Birgit Hirschmugl; Christian Wadsack; Gernot Desoye; Rohan M. Lewis; Bram G. Sengers

Fatty acids are critical for normal fetal growth and development. The placenta mediates the transfer of fatty acids from the maternal to the fetal circulation. Yet, the mechanisms of fatty acid transport are not fully understood. The development of a computational model alongside experiments will test our understanding of the transfer mechanisms. Modelling experimental data suggest the presence of a metabolic pool within placental tissue that could represent the rate-limiting factor for fatty acid transfer. In addition the model suggests a slower flux capacity of the fetal-side of the placenta compared with the maternal-side. The model provides key insights into placental fatty acid transfer which will form the basis for future experimentation.


Scientific Reports | 2018

Endothelial indoleamine 2,3-dioxygenase-1 regulates the placental vascular tone and is deficient in intrauterine growth restriction and pre-eclampsia

Pablo Zardoya-Laguardia; Astrid Blaschitz; Birgit Hirschmugl; Ingrid Lang; Sereina A. Herzog; Liudmila Nikitina; Martin Gauster; M Häusler; M Cervar-Zivkovic; Eva Karpf; Ghassan J. Maghzal; Christopher Stanley; Roland Stocker; Christian Wadsack; Saša Frank; Peter Sedlmayr

Indoleamine 2,3-dioxygenase-1 (IDO1) mediates the degradation of L-tryptophan (L-Trp) and is constitutively expressed in the chorionic vascular endothelium of the human placenta with highest levels in the microvasculature. Given that endothelial expression of IDO1 has been shown to regulate vascular tone and blood pressure in mice under the condition of systemic inflammation, we asked whether IDO1 is also involved in the regulation of placental blood flow and if yes, whether this function is potentially impaired in intrauterine growth restriction (IUGR) and pre-eclampsia (PE). In the large arteries of the chorionic plate L-Trp induced relaxation only after upregulation of IDO1 using interferon gamma and tumor necrosis factor alpha. However, ex vivo placental perfusion of pre-constricted cotyledonic vasculature with L-Trp decreases the vessel back pressure without prior IDO1 induction. Further to this finding, IDO1 protein expression and activity is reduced in IUGR and PE when compared to gestational age–matched control tissue. These data suggest that L-Trp catabolism plays a role in the regulation of placental vascular tone, a finding which is potentially linked to placental and fetal growth. In this context our data suggest that IDO1 deficiency is related to the pathogenesis of IUGR and PE.


International Journal of Obesity | 2018

Relation of placental alkaline phosphatase expression in human term placenta with maternal and offspring fat mass

Birgit Hirschmugl; Sarah Crozier; Nina Matthews; Eva Kitzinger; Ingeborg Klymiuk; Hazel Inskip; Nicholas C. Harvey; C Cooper; Colin P. Sibley; Jocelyn D. Glazier; Christian Wadsack; Keith M. Godfrey; Gernot Desoye; Rohan M. Lewis

Introduction:Alkaline phosphatase is implicated in intestinal lipid transport and in the development of obesity. Placental alkaline phosphatase is localised to the microvillous plasma membrane of the placental syncytiotrophoblast at the maternal–fetal interface, but its role is unclear. We investigated the relations of placental alkaline phosphatase activity and mRNA expression with maternal body composition and offspring fat mass in humans.Methods:Term human placentas from the UK Birthright cohort (n = 52) and the Southampton Women’s Survey (SWS) (n = 95) were studied. In the Birthright cohort, alkaline phosphatase activity was measured in placental microvillous plasma membrane vesicles. In the SWS, alkaline phosphatase mRNA was measured using Nanostring. Alkaline phosphatase gene expression was compared to other lipid-related genes.Results:In Birthright samples placental microvillous plasma membrane alkaline phosphatase activity was positively associated with maternal triceps skinfold thickness and BMI (β = 0.04 (95% CI: 0.01–0.06) and β = 0.02 (0.00–0.03) µmol/mg protein/min per SD, P = 0.002 and P = 0.05, respectively) after adjusting for potential confounders. In SWS samples placental alkaline phosphatase mRNA expression in term placenta was positively associated with maternal triceps skinfold (β = 0.24 (0.04, 0.44) SD/SD, P = 0.02), had no association with neonatal %fat mass (β = 0.01 (−0.20 to 0.21) SD/SD, P = 0.93) and was negatively correlated with %fat mass at ages 4 (β = −0.28 (−0.52 to −0.04) SD/SD, P = 0.02), 6–7 (β = −0.25 (−0.49 to −0.02) SD/SD, P = 0.03) years. When compared with placental expression of other genes, alkaline phosphatase expression was positively related to genes including the lysophosphatidylcholine transporter MFSD2A (major facilitator superfamily domain containing 2A, P < 0.001) and negatively related to genes including the fatty acid transport proteins 2 and 3 (P = 0.001, P < 0.001).Conclusions:Our findings suggest relationships between placental alkaline phosphatase and both maternal and childhood adiposity. The inverse relationship between placental alkaline phosphatase gene expression and childhood %fat mass suggests that placental alkaline phosphatase may help to protect the foetus from the adverse effects of maternal obesity.


Scientific Reports | 2017

Maternal Gestational Diabetes Mellitus increases placental and foetal lipoprotein-associated Phospholipase A2 which might exert protective functions against oxidative stress

Carolin Schliefsteiner; Birgit Hirschmugl; Susanne Kopp; Sanja Curcic; Eva Bernhart; Gunther Marsche; U Lang; Gernot Desoye; Christian Wadsack

Increased Lipoprotein associated phospholipase A2 (LpPLA2) has been associated with inflammatory pathologies, including Type 2 Diabetes. Studies on LpPLA2 and Gestational Diabetes Mellitus (GDM) are rare, and have focused mostly on maternal outcome. In the present study, we investigated whether LpPLA2 activity on foetal lipoproteins is altered by maternal GDM and/or obesity (a major risk factor for GDM), thereby contributing to changes in lipoprotein functionality. We identified HDL as the major carrier of LpPLA2 activity in the foetus, which is in contrast to adults. We observed marked expression of LpPLA2 in placental macrophages (Hofbauer cells; HBCs) and found that LpPLA2 activity in these cells was increased by insulin, leptin, and pro-inflammatory cytokines. These regulators were also increased in plasma of children born from GDM pregnancies. Our results suggest that insulin, leptin, and pro-inflammatory cytokines are positive regulators of LpPLA2 activity in the foeto-placental unit. Of particular interest, functional assays using a specific LpPLA2 inhibitor suggest that high-density lipoprotein (HDL)-associated LpPLA2 exerts anti-oxidative, athero-protective functions on placental endothelium and foetus. Our results therefore raise the possibility that foetal HDL-associated LpPLA2 might act as an anti-inflammatory enzyme improving vascular barrier function.


FEBS Journal | 2003

Trophoblast‐like human choriocarcinoma cells serve as a suitable in vitro model for selective cholesteryl ester uptake from high density lipoproteins

Christian Wadsack; Andelko Hrzenjak; Astrid Hammer; Birgit Hirschmugl; Sanja Levak-Frank; Gernot Desoye; Wolfgang Sattler; Ernst Malle

Collaboration


Dive into the Birgit Hirschmugl's collaboration.

Top Co-Authors

Avatar

Christian Wadsack

Medical University of Graz

View shared research outputs
Top Co-Authors

Avatar

Gernot Desoye

Medical University of Graz

View shared research outputs
Top Co-Authors

Avatar

Ingrid Lang

Medical University of Graz

View shared research outputs
Top Co-Authors

Avatar

U Lang

Medical University of Graz

View shared research outputs
Top Co-Authors

Avatar

Astrid Blaschitz

Medical University of Graz

View shared research outputs
Top Co-Authors

Avatar

Eva Karpf

Medical University of Graz

View shared research outputs
Top Co-Authors

Avatar

M Cervar-Zivkovic

Medical University of Graz

View shared research outputs
Top Co-Authors

Avatar

M Häusler

Medical University of Graz

View shared research outputs
Top Co-Authors

Avatar

Martin Gauster

Medical University of Graz

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge