Network


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

Hotspot


Dive into the research topics where Katja Dahlgaard is active.

Publication


Featured researches published by Katja Dahlgaard.


Developmental Cell | 2007

Capu and Spire assemble a cytoplasmic actin mesh that maintains microtubule organization in the Drosophila oocyte.

Katja Dahlgaard; Alexandre A.S.F. Raposo; Teresa Niccoli; Daniel St Johnston

Summary Mutants in the actin nucleators Cappuccino and Spire disrupt the polarized microtubule network in the Drosophila oocyte that defines the anterior-posterior axis, suggesting that microtubule organization depends on actin. Here, we show that Cappuccino and Spire organize an isotropic mesh of actin filaments in the oocyte cytoplasm. capu and spire mutants lack this mesh, whereas overexpressed truncated Cappuccino stabilizes the mesh in the presence of Latrunculin A and partially rescues spire mutants. Spire overexpression cannot rescue capu mutants, but prevents actin mesh disassembly at stage 10B and blocks late cytoplasmic streaming. We also show that the actin mesh regulates microtubules indirectly, by inhibiting kinesin-dependent cytoplasmic flows. Thus, the Capu pathway controls alternative states of the oocyte cytoplasm: when active, it assembles an actin mesh that suppresses kinesin motility to maintain a polarized microtubule cytoskeleton. When inactive, unrestrained kinesin movement generates flows that wash microtubules to the cortex.


Genetics | 2011

Anterior–Posterior Axis Specification in Drosophila Oocytes: Identification of Novel bicoid and oskar mRNA Localization Factors

Chin-Wen Chang; Dmitry Nashchekin; Lucy Wheatley; Uwe Irion; Katja Dahlgaard; Tessa G. Montague; Jacqueline Hall; Daniel St Johnston

The Drosophila melanogaster anterior–posterior axis is established during oogenesis by the localization of bicoid and oskar mRNAs to the anterior and posterior poles of the oocyte. Although genetic screens have identified some trans-acting factors required for the localization of these transcripts, other factors may have been missed because they also function at other stages of oogenesis. To circumvent this problem, we performed a screen for revertants and dominant suppressors of the bicaudal phenotype caused by expressing Miranda–GFP in the female germline. Miranda mislocalizes oskar mRNA/Staufen complexes to the oocyte anterior by coupling them to the bicoid localization pathway, resulting in the formation of an anterior abdomen in place of the head. In one class of revertants, Miranda still binds Staufen/oskar mRNA complexes, but does not localize to the anterior, identifying an anterior targeting domain at the N terminus of Miranda. This has an almost identical sequence to the N terminus of vertebrate RHAMM, which is also a large coiled-coil protein, suggesting that it may be a divergent Miranda ortholog. In addition, we recovered 30 dominant suppressors, including multiple alleles of the spectroplakin, short stop, a lethal complementation group that prevents oskar mRNA anchoring, and a female sterile complementation group that disrupts the anterior localization of bicoid mRNA in late oogenesis. One of the single allele suppressors proved to be a mutation in the actin nucleator, Cappuccino, revealing a previously unrecognized function of Cappuccino in pole plasm anchoring and the induction of actin filaments by Long Oskar protein.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Neurofibromatosis-like phenotype in Drosophila caused by lack of glucosylceramide extension

Katja Dahlgaard; Anita Jung; Klaus Qvortrup; Henrik Clausen; Ole Kjaerulff; Hans H. Wandall

Glycosphingolipids (GSLs) are of fundamental importance in the nervous system. However, the molecular details associated with GSL function are largely unknown, in part because of the complexity of GSL biosynthesis in vertebrates. In Drosophila, only one major GSL biosynthetic pathway exists, controlled by the glycosyltransferase Egghead (Egh). Here we discovered that loss of Egh causes overgrowth of peripheral nerves and attraction of immune cells to the nerves. This phenotype is reminiscent of the human disorder neurofibromatosis type 1, which is characterized by disfiguring nerve sheath tumors with mast cell infiltration, increased cancer risk, and learning deficits. Neurofibromatosis type 1 is due to a reduction of the tumor suppressor neurofibromin, a negative regulator of the small GTPase Ras. Enhanced Ras signaling promotes glial growth through activation of phosphatidylinositol 3-kinase (PI3K) and its downstream kinase Akt. We find that overgrowth of peripheral nerves in egh mutants is suppressed by down-regulation of the PI3K signaling pathway by expression of either dominant-negative PI3K, the tumor suppressor PTEN, or the transcription factor FOXO in the subperineurial glia. These results show that loss of the glycosyltransferase Egh affects membrane signaling and activation of PI3K signaling in glia of the peripheral nervous system, and suggest that glycosyltransferases may suppress proliferation.


DNA Research | 2014

Identification of TNF-α-responsive promoters and enhancers in the intestinal epithelial cell model Caco-2.

Mette Boyd; Mehmet Coskun; Berit Lilje; Robin Andersson; Ilka Hoof; Jette Bornholdt; Katja Dahlgaard; Jørgen Olsen; Morana Vitezic; Jacob Tveiten Bjerrum; Jakob Benedict Seidelin; Ole Haagen Nielsen; Jesper T. Troelsen; Albin Sandelin

The Caco-2 cell line is one of the most important in vitro models for enterocytes, and is used to study drug absorption and disease, including inflammatory bowel disease and cancer. In order to use the model optimally, it is necessary to map its functional entities. In this study, we have generated genome-wide maps of active transcription start sites (TSSs), and active enhancers in Caco-2 cells with or without tumour necrosis factor (TNF)-α stimulation to mimic an inflammatory state. We found 520 promoters that significantly changed their usage level upon TNF-α stimulation; of these, 52% are not annotated. A subset of these has the potential to confer change in protein function due to protein domain exclusion. Moreover, we locate 890 transcribed enhancer candidates, where ∼50% are changing in usage after TNF-α stimulation. These enhancers share motif enrichments with similarly responding gene promoters. As a case example, we characterize an enhancer regulating the laminin-5 γ2-chain (LAMC2) gene by nuclear factor (NF)-κB binding. This report is the first to present comprehensive TSS and enhancer maps over Caco-2 cells, and highlights many novel inflammation-specific promoters and enhancers.


Journal of Cellular Biochemistry | 2017

Regulation of Laminin γ2 Expression by CDX2 in Colonic Epithelial Cells Is Impaired During Active Inflammation.

Mehmet Coskun; Christoffer Soendergaard; Steffen Joergensen; Katja Dahlgaard; Lene Riis; Ole Haagen Nielsen; Albin Sandelin; Jesper T. Troelsen

The expression of Caudal‐related homeobox transcription factor 2 (CDX2) is impaired by tumor necrosis factor‐α (TNF‐α)‐mediated activation of nuclear factor‐κB (NF‐κB) in ulcerative colitis (UC). Laminin subunit γ2 (LAMC2) is an epithelial basement membrane protein implicated in cell migration, proliferation, differentiation, as well as tumor invasion and intestinal inflammation, and its expression is enhanced by TNF‐α in a NF‐κB‐dependent regulation of the recently identified LAMC2 enhancer. The aim was to determine whether CDX2 is involved in the basal regulation of LAMC2 in epithelial cells and to assess the influence of inflammation. Transcriptional regulation of LAMC2 was examined by reporter gene assays, overexpression, and shRNA‐mediated knock‐down of CDX2. CDX2‐DNA interactions were assessed by chromatin immunoprecipitation on Caco‐2 cells without or with TNF‐α, as well as in purified colonic human epithelial cells. Immunohistochemical staining and quantitative reverse‐transcription polymerase chain reaction analyses were used to measure the expression of CDX2 and LAMC2 in colonic biopsies from healthy controls and patients with UC. These data indicate that CDX2 directly regulates LAMC2 gene expression through interaction with elements in the LAMC2 promoter region. We further revealed an inverse effect of inflammation on CDX2 and LAMC2. The data presented provide a novel insight into how CDX2 is implicated in the transcriptional regulation of LAMC2 in intestinal epithelial cells, a function that is impaired during mucosal inflammation where a high level of TNF‐α is present. J. Cell. Biochem. 118: 298–307, 2017.


Scientific Reports | 2018

Intestinal regulation of suppression of tumorigenicity 14 (ST14) and serine peptidase inhibitor, Kunitz type -1 (SPINT1) by transcription factor CDX2

E. Thomas Danielsen; Anders Krüger Olsen; Mehmet Coskun; Annika W. Nonboe; Sylvester Larsen; Katja Dahlgaard; Eric P. Bennett; Cathy Mitchelmore; Lotte K. Vogel; Jesper T. Troelsen

The type II membrane-anchored serine protease, matriptase, encoded by suppression of tumorgenicity-14 (ST14) regulates the integrity of the intestinal epithelial barrier in concert with its inhibitor, HAI-1 encoded by serine peptidase inhibitor, Kunitz type -1 (SPINT1). The balance of the protease/inhibitor gene expression ratio is vital in preventing the oncogenic potential of matriptase. The intestinal cell lineage is regulated by a transcriptional regulatory network where the tumor suppressor, Caudal homeobox 2 (CDX2) is considered to be an intestinal master transcription factor. In this study, we show that CDX2 has a dual function in regulating both ST14 and SPINT1, gene expression in intestinal cells. We find that CDX2 is not required for the basal ST14 and SPINT1 gene expression; however changes in CDX2 expression affects the ST14/SPINT1 mRNA ratio. Exploring CDX2 ChIP-seq data from intestinal cell lines, we identified genomic CDX2-enriched enhancer elements for both ST14 and SPINT1, which regulate their corresponding gene promoter activity. We show that CDX2 displays both repressive and enhancing regulatory abilities in a cell specific manner. Together, these data reveal new insight into transcriptional mechanisms controlling the intestinal matriptase/inhibitor balance.


PLOS ONE | 2018

The VTI1A-TCF4 colon cancer fusion protein is a dominant negative regulator of Wnt signaling and is transcriptionally regulated by intestinal homeodomain factor CDX2

Johanne Davidsen; Sylvester Larsen; Mehmet Coskun; Ismail Gögenur; Katja Dahlgaard; Eric P. Bennett; Jesper T. Troelsen

Sequencing of primary colorectal tumors has identified a gene fusion in approximately 3% of colorectal cancer patients of the VTI1A and TCF7L2 genes, encoding a VTI1A-TCF4 fusion protein containing a truncated TCF4. As dysregulation of the Wnt signaling pathway is associated with colorectal cancer development and progression, the functional properties and transcriptional regulation of the VTI1A-TCF4 fusion protein may also play a role in these processes. Functional characteristics of the VTI1A-TCF4 fusion protein in Wnt signaling were analyzed in NCI-H508 and LS174T colon cancer cell lines. The NCI-H508 cell line, containing the VTI1A-TCF7L2 fusion gene, showed no active Wnt signaling, and overexpression of the VTI1A-TCF4 fusion protein in LS174T cells along with a Wnt signaling luciferase reporter plasmid showed inhibition of activity. The transcriptional regulation of the VTI1A-TCF4 fusion gene was investigated in LS174T cells where the activity of the VTI1A promoter was compared to that of the TCF7L2 promoter, and the transcription factor CDX2 was analyzed for gene regulatory activity of the VTI1A promoter through luciferase reporter gene assay using colon cancer cell lines as a model. Transfection of LS174T cells showed that the VTI1A promoter is highly active compared to the TCF7L2 promoter, and that CDX2 activates transcription of VTI1A. These results suggest that the VTI1A-TCF4 fusion protein is a dominant negative regulator of the Wnt signaling pathway, and that transcription of VTI1A is activated by CDX2.


Archive | 2018

Identification and Functional Analysis of Gene Regulatory Sequences Interacting with Colorectal Tumor Suppressors

Katja Dahlgaard; Jesper T. Troelsen


Nature Communications | 2018

Characterization of the enhancer and promoter landscape of inflammatory bowel disease from human colon biopsies

Mette Boyd; Malte Thodberg; Morana Vitezic; Jette Bornholdt; Kristoffer Vitting-Seerup; Yun Chen; Mehmet Coskun; Yuan Li; Bobby Zhao Sheng Lo; Pia Klausen; Pawel J. Schweiger; Anders Gorm Pedersen; Nicolas Rapin; Kerstin Skovgaard; Katja Dahlgaard; Robin Andersson; Thilde Terkelsen; Berit Lilje; Jesper T. Troelsen; Andreas Petersen; Kim B. Jensen; Ismail Gögenur; Peter Thielsen; Jakob Benedict Seidelin; Ole Haagen Nielsen; Jacob Tveiten Bjerrum; Albin Sandelin


Teaching for Active Learning | 2016

Problembaseret undervisning i forskning – med eksplicit fokus på professional adfærd

Katja Dahlgaard; Tinne Hoff Kjeldsen

Collaboration


Dive into the Katja Dahlgaard's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mehmet Coskun

University of Copenhagen

View shared research outputs
Top Co-Authors

Avatar

Albin Sandelin

University of Copenhagen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Berit Lilje

University of Copenhagen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge