Network


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

Hotspot


Dive into the research topics where Adelheid Elbe-Bürger is active.

Publication


Featured researches published by Adelheid Elbe-Bürger.


Anatomy and Embryology | 1999

Recruitment of bone-marrow-derived cells by skeletal and cardiac muscle in adult dystrophic mdx mice.

Reginald E. Bittner; Christian Schöfer; Klara Weipoltshammer; Silva Ivanova; Berthold Streubel; Erwin Hauser; Michael Freilinger; H. Höger; Adelheid Elbe-Bürger; F. Wachtler

 It is commonly accepted, that regenerative capacity of striated muscle is confined to skeletal muscle by activation of satellite cells that normally reside quiescent between the plasmalemma and the basement membrane of muscle fibers. Muscular dystrophies are characterized by repetitive cycles of de- and regeneration of skeletal muscle fibers and by the frequent involvement of the cardiac muscle. Since during the longstanding course of muscular dystrophies there is a permanent demand of myogenic progenitors we hypothesized that this may necessitate a recruitment of additional myogenic precursors from an undifferentiated, permanently renewed cell pool, such as bone marrow (BM) cells. To this end normal and dystrophic (mdx) female mice received bone marrow transplantation (BMT) from normal congenic male donor mice. After 70 days, histological sections of skeletal and cardiac muscle from BMT mice were probed for the donor-derived Y chromosomes. In normal BMT recipients, no Y chromosome-containing myonuclei were detected, either in skeletal or in cardiac muscle. However, in all samples from dystrophic mdx skeletal muscles Y chromosome-specific signals were detected within muscle fiber nuclei, which additionally were found to express the myoregulatory proteins myogenin and myf-5. Moreover, in the hearts of BMT-mdx mice single cardiomyocytes with donor derived nuclei were identified, indicating, that even cardiac muscle cells are able to regenerate by recruitment of circulating BM-derived progenitors. Our findings suggest that further characterization and identification of the BM cells capable of undergoing myogenic differentiation may have an outstanding impact on therapeutic strategies for diseases of skeletal and cardiac muscle.


Journal of Investigative Dermatology | 2012

Human Dermis Harbors Distinct Mesenchymal Stromal Cell Subsets

Christine Vaculik; Christopher Schuster; Wolfgang Bauer; Nousheen Iram; Karin Pfisterer; Gero Kramer; Andreas Reinisch; Dirk Strunk; Adelheid Elbe-Bürger

Multipotent mesenchymal stromal cells (MSCs) are found in a variety of adult tissues including human dermis. These MSCs are morphologically similar to bone marrow–derived MSCs, but are of unclear phenotype. To shed light on the characteristics of human dermal MSCs, this study was designed to identify and isolate dermal MSCs by a specific marker expression profile, and subsequently rate their mesenchymal differentiation potential. Immunohistochemical staining showed that MSC markers CD73/CD90/CD105, as well as CD271 and SSEA-4, are expressed on dermal cells in situ. Flow cytometric analysis revealed a phenotype similar to bone marrow–derived MSCs. Human dermal cells isolated by plastic adherence had a lower differentiation capacity as compared with bone marrow–derived MSCs. To distinguish dermal MSCs from differentiated fibroblasts, we immunoselected CD271+ and SSEA-4+ cells from adherent dermal cells and investigated their mesenchymal differentiation capacity. This revealed that cells with increased adipogenic, osteogenic, and chondrogenic potential were enriched in the dermal CD271+ population. The differentiation potential of dermal SSEA-4+ cells, in contrast, appeared to be limited to adipogenesis. These results indicate that specific cell populations with variable mesenchymal differentiation potential can be isolated from human dermis. Moreover, we identified three different subsets of dermal mesenchymal progenitor cells.


Journal of Experimental Medicine | 2009

HLA-DR+ leukocytes acquire CD1 antigens in embryonic and fetal human skin and contain functional antigen-presenting cells

Christopher Schuster; Christine Vaculik; Christian Fiala; Simone Meindl; Oliver Brandt; Martin Imhof; Georg Stingl; Wolfgang Eppel; Adelheid Elbe-Bürger

Adequate numbers and functional maturity are needed for leukocytes to exhibit a protective role in host defense. During intrauterine life, the skin immune system has to acquire these prerequisites to protect the newborn from infection in the hostile external environment after birth. We investigated the quantitative, phenotypic, and functional development of skin leukocytes and analyzed the factors controlling their proliferation and trafficking during skin development. We show that CD45+ leukocytes are scattered in embryonic human skin and that their numbers continuously increase as the developing skin generates an environment that promotes proliferation of skin resident leukocytes as well as the influx of leukocytes from the circulation. We also found that CD45+HLA-DRhighCD1c+ dendritic cells (DCs) are already present in the epidermis and dermis at 9 wk estimated gestational age (EGA) and that transforming growth factor β1 production precedes Langerin and CD1a expression on CD45+CD1c+ Langerhans cell (LC) precursors. Functionally, embryonic antigen-presenting cells (APCs) are able to phagocytose antigen, to up-regulate costimulatory molecules upon culture, and to efficiently stimulate T cells in a mixed lymphocyte reaction. Collectively, our data provide insight into skin DC biology and the mechanisms through which skin DCs presumably populate the skin during development.


British Journal of Dermatology | 2003

Pimecrolimus does not affect Langerhans cells in murine epidermis

J.G. Meingassner; E. Kowalsky; H. Schwendinger; Adelheid Elbe-Bürger; A. Stütz

Background  Langerhans cells (LCs) function as specialized antigen‐presenting cells in the epidermis, and therefore play a critical role in cutaneous immunological reactions. Topical treatment with corticosteroids is associated with a decrease in epidermal LC number and antigen‐presenting capacity in laboratory animals and humans.


Journal of Experimental Medicine | 2013

Identification of bone morphogenetic protein 7 (BMP7) as an instructive factor for human epidermal Langerhans cell differentiation

Nighat Yasmin; Thomas Bauer; Madhura Modak; Karin Wagner; Christopher Schuster; René Köffel; Maria Seyerl; Johannes Stöckl; Adelheid Elbe-Bürger; Daniel Graf; Herbert Strobl

Bone morphogenetic protein 7 (BMP7) promotes the differentiation of Langerhans cells in the epidermis during prenatal development.


Nature | 2017

Human fetal dendritic cells promote prenatal T-cell immune suppression through arginase-2

Naomi McGovern; Amanda Shin; Gillian Low; Donovan Low; Kaibo Duan; Leong Jing Yao; Rasha Msallam; Ivy Low; Nurhidaya Binte Shadan; Hermi Rizal Bin Sumatoh; Erin Soon; Josephine Lum; Esther Wing Hei Mok; Sandra Hubert; Peter See; Edwin Huang Kunxiang; Yie Hou Lee; Baptiste Janela; Mahesh Choolani; Citra Nurfarah Zaini Mattar; Yiping Fan; Tony Kiat Hon Lim; Dedrick Kok Hong Chan; Ker-Kan Tan; John Tam; Christopher Schuster; Adelheid Elbe-Bürger; Xiao-Nong Wang; Venetia Bigley; Matthew Collin

During gestation the developing human fetus is exposed to a diverse range of potentially immune-stimulatory molecules including semi-allogeneic antigens from maternal cells, substances from ingested amniotic fluid, food antigens, and microbes. Yet the capacity of the fetal immune system, including antigen-presenting cells, to detect and respond to such stimuli remains unclear. In particular, dendritic cells, which are crucial for effective immunity and tolerance, remain poorly characterized in the developing fetus. Here we show that subsets of antigen-presenting cells can be identified in fetal tissues and are related to adult populations of antigen-presenting cells. Similar to adult dendritic cells, fetal dendritic cells migrate to lymph nodes and respond to toll-like receptor ligation; however, they differ markedly in their response to allogeneic antigens, strongly promoting regulatory T-cell induction and inhibiting T-cell tumour-necrosis factor-α production through arginase-2 activity. Our results reveal a previously unappreciated role of dendritic cells within the developing fetus and indicate that they mediate homeostatic immune-suppressive responses during gestation.


Journal of Leukocyte Biology | 2005

Fetal and neonatal murine skin harbors Langerhans cell precursors.

Souyet Chang-Rodriguez; Wolfram Hoetzenecker; C. Schwärzler; T. Biedermann; Sem Saeland; Adelheid Elbe-Bürger

Resident epidermal Langerhans cells (LC) in adult mice express ADPase, major histocompatibility complex (MHC) class II, and CD205 and CD207 molecules, while the first dendritic leukocytes that colonize the fetal and newborn epidermis are only ADPase+. In this study, we tested whether dendritic epidermal leukocytes (DEL) are end‐stage cells or represent LC precursors. In epidermal sheets of fetal and neonatal mice, we found no apoptotic leukocytes, suggesting that these cells do not die in situ. To address whether DEL can give rise to LC, sorted DEL from murine newborn skin were cultured with cytokines used to generate LC from human CD34+ precursors. After 7–14 days, DEL proliferated and acquired the morphology and phenotype of cells reminiscent of LC. In concordance with this finding, we show that neonatal epidermis harbors 10–20 times the number of cycling MHC class II+ leukocytes as adult tissue. To test whether LC can differentiate from skin precursors in vivo, we developed a transplantation model. As it was impossible to transplant fetal epidermis, whole fetal skin was grafted onto adult severe combined immunodeficient mice. As opposed to the uniform absence of donor LC at the time of transplantation, examination of the epidermis from the grafts after 2–4 weeks revealed MHC class II+ donor cells, which had acquired CD205 and CD207, thus qualifying them as LC. Finally, we present evidence that endogenous LC persist in skin grafts for the observation period of 45 days. These studies show that hematopoietic precursors seed the skin during embryonic life and can give rise to LC.


Development | 2012

Age-related changes in expression and function of Toll-like receptors in human skin

Nousheen Iram; Michael Mildner; Marion Prior; Peter Petzelbauer; Christian Fiala; Stefan Hacker; Alice Schöppl; Erwin Tschachler; Adelheid Elbe-Bürger

Toll-like receptors (TLRs) initiate innate immune responses and direct subsequent adaptive immunity. They play a major role in cutaneous host defense against micro-organisms and in the pathophysiology of several inflammatory skin diseases. To understand the role of TLRs in the acquisition of immunological competence, we conducted a comprehensive study to evaluate TLR expression and function in the developing human skin before and after birth and compared it with adults. We found that prenatal skin already expresses the same spectrum of TLRs as adult skin. Strikingly, many TLRs were significantly higher expressed in prenatal (TLRs 1-5) and infant and child (TLRs 1 and 3) skin than in adult skin. Surprisingly, neither dendritic cell precursors in prenatal skin nor epidermal Langerhans cells and dermal dendritic cells in adult skin expressed TLRs 3 and 6, whereas the staining pattern and intensity of both TLRs in fetal basal keratinocytes was almost comparable to those of adults. Stimulation of primary human keratinocytes from fetal, neonatal and adult donors with selected TLR agonists revealed that the synthetic TLR3 ligand poly (I:C) specifically, mimicking viral double-stranded RNA, induced a significantly enhanced secretion of CXCL8/IL8, CXCL10/IP-10 and TNFα in fetal and neonatal keratinocytes compared with adult keratinocytes. This study demonstrates quantitative age-specific modifications in TLR expression and innate skin immune reactivity in response to TLR activation. Thus, antiviral innate immunity already in prenatal skin may contribute to protect the developing human body from viral infections in utero in a scenario where the adaptive immune system is not yet fully functional.


Blood | 2009

Reciprocal role of GATA-1 and vitamin D receptor in human myeloid dendritic cell differentiation

Florian Göbel; Sabine Taschner; Jennifer Jurkin; Sabine Konradi; Christine Vaculik; Susanne Richter; Doris Kneidinger; Christina Mühlbacher; Christian Bieglmayer; Adelheid Elbe-Bürger; Herbert Strobl

Two major pathways of human myeloid dendritic cell (DC) subset differentiation have previously been delineated. Langerhans cells (LCs) reside in epithelia in the steady state, whereas monocytes can provide dendritic cells (DCs) on demand in response to inflammatory signals. Both DC subset pathways arise from shared CD14+ monocyte precursors, which in turn develop from myeloid committed progenitor cells. However, the underlying hematopoietic mechanisms still remain poorly defined. Here, we demonstrate that the vitamin D(3) receptor (VDR) is induced by transforming growth factor beta1 during LC lineage commitment and exerts a positive role during LC generation. In contrast, VDR is repressed during interleukin-4 (IL-4)-dependent monocyte-derived DC (moDC) differentiation. We identified GATA-1 as a repressor of VDR. GATA-1 is induced by IL-4 in moDCs. Forced inducible expression of GATA-1 mimics IL-4 in redirecting moDC differentiation and vice versa, GATA-1 knockdown arrests moDC differentiation at the monocyte stage. Moreover, ectopic GATA-1 expression stabilizes the moDC phenotype under monocyte-promoting conditions in the presence of vitamin D3 (VD3). In summary, human myeloid DC subset differentiation is inversely regulated by GATA-1 and VDR. GATA-1 mediates the repression of VDR and enables IL-4-dependent moDC differentiation. Conversely, VDR is induced downstream of transforming growth factor beta1 and is functionally involved in promoting LC differentiation.


Journal of Investigative Dermatology | 2012

Phenotypic Characterization of Leukocytes in Prenatal Human Dermis

Christopher Schuster; Christine Vaculik; Marion Prior; Christian Fiala; Michael Mildner; Wolfgang Eppel; Georg Stingl; Adelheid Elbe-Bürger

The adult human skin harbors a variety of leukocytes providing immune surveillance and host defense, but knowledge about their ontogeny is scarce. In this study we investigated the number and phenotype of leukocytes in prenatal human skin (dermal dendritic cells (DDCs), macrophages, T cells (including FoxP3+ regulatory T cells), and mast cells) to unravel their derivation and to get a clue as to their putative function in utero. By flow cytometry and immunofluorescence, we found a distinction between CD206+CD1c+CD11c+ DDCs and CD206+CD209+CD1c− skin macrophages by 9 weeks estimated gestational age (EGA). T cells appear at the end of the first trimester, expressing CD3 intracytoplasmatically. During midgestation, CD3+FoxP3− and CD3+FoxP3+ cells can exclusively be found in the dermis. Similarly, other leukocytes such as CD117+ (c-kit) mast cells were not identified before 12–14 weeks EGA and only slowly acquire a mature phenotype during gestation. Our data show at which time point during gestation antigen-presenting cells, T cells, and mast cells populate the human dermis and provide a step forward to a better understanding of the development of the human skin immune system.

Collaboration


Dive into the Adelheid Elbe-Bürger's collaboration.

Top Co-Authors

Avatar

Georg Stingl

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael Mildner

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Christine Vaculik

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Maria Gschwandtner

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Marion Prior

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Wolfgang Bauer

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Wolfgang Eppel

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Wolfram Hoetzenecker

Medical University of Vienna

View shared research outputs
Researchain Logo
Decentralizing Knowledge