Network


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

Hotspot


Dive into the research topics where Julia Höbaus is active.

Publication


Featured researches published by Julia Höbaus.


Frontiers in Physiology | 2014

Vitamin D and the epigenome

Irfete S. Fetahu; Julia Höbaus; Enikő Kállay

Epigenetic mechanisms play a crucial role in regulating gene expression. The main mechanisms involve methylation of DNA and covalent modifications of histones by methylation, acetylation, phosphorylation, or ubiquitination. The complex interplay of different epigenetic mechanisms is mediated by enzymes acting in the nucleus. Modifications in DNA methylation are performed mainly by DNA methyltransferases (DNMTs) and ten-eleven translocation (TET) proteins, while a plethora of enzymes, such as histone acetyltransferases (HATs), histone deacetylases (HDACs), histone methyltransferases (HMTs), and histone demethylases (HDMs) regulate covalent histone modifications. In many diseases, such as cancer, the epigenetic regulatory system is often disturbed. Vitamin D interacts with the epigenome on multiple levels. Firstly, critical genes in the vitamin D signaling system, such as those coding for vitamin D receptor (VDR) and the enzymes 25-hydroxylase (CYP2R1), 1α-hydroxylase (CYP27B1), and 24-hydroxylase (CYP24A1) have large CpG islands in their promoter regions and therefore can be silenced by DNA methylation. Secondly, VDR protein physically interacts with coactivator and corepressor proteins, which in turn are in contact with chromatin modifiers, such as HATs, HDACs, HMTs, and with chromatin remodelers. Thirdly, a number of genes encoding for chromatin modifiers and remodelers, such as HDMs of the Jumonji C (JmjC)-domain containing proteins and lysine-specific demethylase (LSD) families are primary targets of VDR and its ligands. Finally, there is evidence that certain VDR ligands have DNA demethylating effects. In this review we will discuss regulation of the vitamin D system by epigenetic modifications and how vitamin D contributes to the maintenance of the epigenome, and evaluate its impact in health and disease.


Anti-cancer Agents in Medicinal Chemistry | 2012

Role of Calcium, Vitamin D, and the Extrarenal Vitamin D Hydroxylases in Carcinogenesis

Julia Höbaus; Ursula Thiem; Doris M. Hummel; Enikö Kállay

Vitamin D deficiency and low calcium intake are considered risk factors for several cancers. Vitamin D, synthesized in the skin or ingested through the diet, is transformed through two hydroxylation steps to the active metabolite, 1α,25-dihydroxyvitamin D3 (1,25-D3). 25-hydroxylases in the liver are responsible for the first hydroxylation step. The ultimate activation is performed by the renal 25-hydroxyvitamin D 1α-hydroxylase (CYP27B1), while the 1,25-dihydroxyvitamin D 24-hydroxylase (CYP24A1) in the kidneys degrades the active metabolite. These two renal vitamin D hydroxylases control the endocrine serum 1,25-D3 levels, and are responsible for maintaining mineral homeostasis. In addition, the active vitamin D hormone 1,25-D3 regulates cellular proliferation, differentiation, and apoptosis in multiple tissues in a paracrine/autocrine manner. Interestingly, it is the low serum level of the precursor 25- hydroxyvitamin D3 (25-D3) that predisposes to numerous cancers and other chronic diseases, and not the serum concentration of the active vitamin D hormone. The extra-renal autocrine/paracrine vitamin D system is able to synthesize and degrade locally the active 1,25- D3 necessary to maintain normal cell growth and to counteract mitogenic stimuli. Thus, vitamin D hydroxylases play a prominent role in this process. The present review describes the role of the vitamin D hydroxylases in cancer pathogenesis and the cross-talk between the extra-renal autocrine/paracrine vitamin D system and calcium in cancer prevention.


International Journal of Cancer | 2013

Increased copy-number and not DNA hypomethylation causes overexpression of the candidate proto-oncogene CYP24A1 in colorectal cancer

Julia Höbaus; Doris M. Hummel; Ursula Thiem; Irfete S. Fetahu; Abhishek Aggarwal; Leonhard Müllauer; Gerwin Heller; Gerda Egger; Ildiko Mesteri; Sabina Baumgartner-Parzer; Enikö Kállay

In colorectal cancer (CRC) the vitamin D catabolizing enzyme 1,25‐dihydroxyvitamin D 24‐hydroxylase (CYP24A1) is overexpressed with a potentially significant, positive impact on the catabolism of 1,25‐dihydroxyvitamin D3 (1,25‐D3). However, the underlying mechanism of CYP24A1 overexpression is poorly understood. In the present study, we investigated possible causes including hypomethylation of the CYP24A1 promoter, amplification of the CYP24A1 gene locus (20q13.2), and altered expression of CYP24A1‐specific transcription factors. We quantified CYP24A1 gene copy‐number, performed bisulfite sequencing of the CYP24A1 promoter to assess DNA methylation, and measured mRNA expression of CYP24A1, 25‐hydroxyvitamin D 1α‐hydroxylase (CYP27B1), vitamin D receptor (VDR) and retinoid X receptor (RXR). We found that 77 (60%) out of 127 colorectal tumors showed increased CYP24A1 gene copy‐number and that more than 6 copies of CYP24A1 correlated positively with CYP24A1 mRNA expression suggestive of a causal relationship. No differences in CYP24A1 promoter methylation were found between tumor tissue and adjacent mucosa from the same patient or between tissues with high or low mRNA expression, thus excluding DNA hypomethylation as a possible cause of CYP24A1 overexpression in CRC. Furthermore, mRNA expression of several factors involved in replication licensing positively correlated with CYP24A1 mRNA expression, raising the possibility that CYP24A1 overexpression might favor increased proliferation in tumors by suppressing local 1,25‐D3 levels. We conclude that high copy‐number gain is a key determinant of CYP24A1 overexpression in CRC. Other postulated causes of CYP24A1 overexpression including promoter hypomethylation and enhanced VDR and/or RXR expression do not appear to be involved.


The Journal of Steroid Biochemistry and Molecular Biology | 2013

Prevention of preneoplastic lesions by dietary vitamin D in a mouse model of colorectal carcinogenesis

Doris M. Hummel; Ursula Thiem; Julia Höbaus; Ildiko Mesteri; Lukas Gober; Caroline Stremnitzer; Joao Graca; Barbara Obermayer-Pietsch; Enikö Kállay

Highlights ► High dietary vitamin D was able to prevent premalignant lesions caused by AOM/DSS. ► Increasing vitamin D intake raised serum 25-D3 levels reaching a plateau ≥1000 IU/kg. ► Serum 25-D3 levels over 30 ng/ml are needed to prevent tumorigenesis.


Biochimica et Biophysica Acta | 2015

The calcium-sensing receptor: A promising target for prevention of colorectal cancer.

Abhishek Aggarwal; Maximilian Prinz-Wohlgenannt; Samawansha Tennakoon; Julia Höbaus; C. Boudot; Romuald Mentaverri; Edward M. Brown; Sabina Baumgartner-Parzer; Enikö Kállay

The inverse correlation between dietary calcium intake and the risk of colorectal cancer (CRC) is well known, but poorly understood. Expression of the calcium-sensing receptor (CaSR), a calcium-binding G protein-coupled receptor is downregulated in CRC leading us to hypothesize that the CaSR has tumor suppressive roles in the colon. The aim of this study was to understand whether restoration of CaSR expression could reduce the malignant phenotype in CRC. In human colorectal tumors, expression of the CaSR negatively correlated with proliferation markers whereas loss of CaSR correlated with poor tumor differentiation and reduced apoptotic potential. In vivo, dearth of CaSR significantly increased expression of proliferation markers and decreased levels of differentiation and apoptotic markers in the colons of CaSR/PTH double knock-out mice confirming the tumor suppressive functions of CaSR. In vitro CRC cells stably overexpressing wild-type CaSR showed significant reduction in proliferation, as well as increased differentiation and apoptotic potential. The positive allosteric modulator of CaSR, NPS R-568 further enhanced these effects, whereas treatment with the negative allosteric modulator, NPS 2143 inhibited these functions. Interestingly, the dominant-negative mutant (R185Q) was able to abrogate these effects. Our results demonstrate a critical tumor suppressive role of CaSR in the colon. Restoration of CaSR expression and function is linked to regulation of the balance between proliferation, differentiation, and apoptosis and provides a rationale for novel strategies in CRC therapy.


International Journal of Cancer | 2014

Calcium-sensing receptor silencing in colorectal cancer is associated with promoter hypermethylation and loss of acetylation on histone 3

Irfete S. Fetahu; Julia Höbaus; Abhishek Aggarwal; Doris M. Hummel; Samawansha Tennakoon; Ildiko Mesteri; Sabina Baumgartner-Parzer; Enikő Kállay

The calcium‐sensing receptor (CaSR) is suggested to mediate the antiproliferative effects of calcium in colon. However, in colorectal cancer (CRC) the expression of the CaSR is silenced and the underlying mechanisms leading to its loss are poorly understood. We investigated whether loss of the CaSR expression in colorectal tumors is caused by DNA hypermethylation and imbalance of transcriptionally permissive/repressive histone alterations. We observed significantly lower CaSR mRNA expression (n = 65, p < 0.001) in colorectal tumors compared with the adjacent mucosa from the same patient. Immunofluorescence staining confirmed downregulation of the CaSR protein also. The CaSR promoter was methylated to a greater extent in tumors compared with adjacent mucosa as determined by bisulfite sequencing (n = 20, p < 0.01) and by pyrosequencing (n = 45, p < 0.001), and methylation correlated inversely with mRNA expression (n = 20, ρ = −0.310, p < 0.05 and n = 45, ρ = −0.588, p < 0.001). Treatments with 5‐aza‐2′‐deoxycytidine (DAC), a DNA methyltransferase inhibitor and/or with two different histone deacetylase inhibitors, trichostatin A (TSA) or suberoylanilide hydroxamic acid (SAHA) restored the expression of CaSR in colon cancer cells. Restored CaSR expression in Coga1A and HT29 cells was functional. Inhibition of lysine‐specific demethylase 1 (LSD1) to prevent demethylation of mono‐ and dimethylated H3K4, increased CaSR expression only marginally. Our data show that hypermethylation of the CaSR promoter and H3K9 deacetylation, but not H3K4me2 demethylation are important factors that cause silencing of the CaSR in colorectal cancer.


The Journal of Steroid Biochemistry and Molecular Biology | 2013

Epigenetic regulation of the 1,25-dihydroxyvitamin D3 24-hydroxylase (CYP24A1) in colon cancer cells

Julia Höbaus; Irfete S. Fetahu; Maya Khorchide; Teresa Manhardt; Enikö Kállay

Highlights ► Basal and calcitriol-induced expression of CYP24A1 is cell line dependent. ► Histone deacetylase and methyltransferase inhibitors increase CYP24A1 expression. ► Cell line specific drug response does not correlate with promoter methylation. ► Drug treatment may activate genes upstream of CYP24A1 and cause indirect induction.


International Journal of Cancer | 2016

Impact of CYP24A1 overexpression on growth of colorectal tumour xenografts in mice fed with vitamin D and soy

Julia Höbaus; Samawansha Tennakoon; Petra Heffeter; Charlotte Groeschel; Abhishek Aggarwal; Doris M. Hummel; Ursula Thiem; Rodrig Marculescu; Walter Berger; Enikö Kállay

Our previous studies showed that the 1,25‐dihydroxyvitamin D (1,25‐D3) catabolizing enzyme, 1,25‐dihydoxyvitamin D 24 hydroxylase (CYP24A1) was overexpressed in colorectal tumours and its level correlated with increased proliferation. We hypothesised that cells overexpressing CYP24A1 have growth advantage and a diet rich in vitamin D and soy would restore sensitivity to the anti‐tumourigenic effects of vitamin D. Soy contains genistein, a natural CYP24A1 inhibitor. To determine causality between CYP24A1 and tumour growth, we established xenografts in male SCID mice with HT29 cells stably overexpressing either GFP‐tagged CYP24A1 or GFP. Mice were fed with either high (2500 IU D3/kg) or low vitamin D (100 IU D3/kg) diet in the presence or absence of soy (20% diet). In vitro, cells overexpressing CYP24A1 grew faster than controls. 1,25‐D3, the active vitamin D metabolite, reduced cell number only in the presence of the CYP24A1 inhibitor VID400. Regardless of the amount of vitamin D in the diet, xenografts overexpressing CYP24A1 grew faster, were heavier and more aggressive. Soy reduced tumour volume only in the control xenografts, while the tumours overexpressing CYP24A1 were larger in the presence of dietary soy. In conclusion, we demonstrate that CYP24A1 overexpression results in increased aggressiveness and proliferative potential of colorectal tumours. Irrespective of the dietary vitamin D3, dietary soy is able to increase tumour volume when tumours overexpress CYP24A1, suggesting that combination of vitamin D3 and soy could have an anti‐tumourigenic effect only if CYP24A1 levels are normal.


Cancer Research | 2013

Abstract 5383: Overexpression of the vitamin D catabolizing enzyme CYP24A1 is caused by gene amplification and results in highly proliferative colorectal tumors.

Julia Höbaus; Abhishek Aggarwal; Doris M. Hummel; Ursula Thiem; Irfete S. Fetahu; Ildiko Mesteri; Enikö Kállay

Vitamin D insufficiency increases risk of colorectal cancer. Vitamin D is produced photochemically in the skin, thus, low sunlight exposure results in vitamin D insufficiency. Vitamin D is further processed in the liver to its storage form calcidiol (25-OH vitamin D3). Calcidiol can be activated by 1α-hydroxylation to the secosteroid hormone calcitriol (1α,25-OH vitamin D3). Although systemic levels of bioactive calcitriol are regulated by the kidneys, almost every tissue can synthesize and degrade calcitriol. Tissue calcitriol acts in an autocrine/paracrine manner and controls proliferation, apoptosis, and differentiation. In colorectal cancer, the calcidiol and calcitriol-degrading enzyme CYP24A1 is substantially overexpressed both on mRNA and protein level. High CYP24A1 levels markedly reduce the half-life of vitamin D metabolites, likely reducing the anti-tumorigenic effects of calcitriol in the tumor. The causes and consequences of this overexpression are not fully understood. Here, we investigated gene amplification of the CYP24A1 locus (20q13.2) as a possible cause of CYP24A1 overexpression and increased proliferation as a consequence thereof. Quantitative real time PCR assays showed that approximately 60% of colorectal tumors carry CYP24A1 gene amplification (n=127). This gene amplification correlated with increased mRNA expression (ρ=0.38, p In conclusion, our data suggest that CYP24A1 gene amplification results in increased mRNA expression in colorectal tumors. Further, high CYP24A1 expression correlates with increased proliferation, possibly caused by an inhibition of the anti-proliferative effects of calcitriol. Tumor specific inhibition of CYP24A1 may provide a future strategy to restore local vitamin D levels and its anti-tumorigenic activities. Citation Format: Julia Hobaus, Abhishek Aggarwal, Doris M. Hummel, Ursula Thiem, Irfete Fetahu, Ildiko Mesteri, Eniko Kallay. Overexpression of the vitamin D catabolizing enzyme CYP24A1 is caused by gene amplification and results in highly proliferative colorectal tumors. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 5383. doi:10.1158/1538-7445.AM2013-5383


Cancer Research | 2013

Abstract 5314: Calcium mediated growth control in normal and transformed colon cells: A possible role of the calcium sensing receptor.

Abhishek Aggarwal; Julia Höbaus; Irfete S. Fetahu; Ildiko Mesteri; Enikö Kállay

Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC Introduction: Calcium (Ca2+) plays an important role in regulating proliferation of colonic epithelium. Epidemiological studies suggest an inverse correlation between dietary Ca2+ intake and colorectal cancer (CRC) risk. However in CRC, transformed cells lose their sensitivity to the tumour inhibiting effects of Ca2+. We have shown that the extracellular calcium-sensing receptor (CaSR) expression is lost in CRC and therefore hypothesized that the CaSR mediates the effect of Ca2+ in regulating proliferation. Methods: We investigated the expression of the CaSR and of a cluster of genes responsible for ‘triggering off’ proliferation (CDT1, CDC6, CDC45, MCM2-7, Geminin, known as replication licensing genes) at mRNA level by qRT-PCR in CRC samples and in their respective ‘normal’ adjacent mucosa (n=57). To study the effect of Ca2+ in normal colon physiology, we fed male and female C57BL/6 mice with a diet containing high (0.9%) or low (0.1%) levels of Ca2+ and assessed the influence of dietary Ca2+ on proliferation (immunohistochemistry staining and mRNA expression) and CaSR mRNA expression. Results and Discussion: In tumours we found the expression of CaSR significantly downregulated when compared with respective adjacent mucosa (p<0.001). All genes involved in replication licensing are significantly higher expressed (p<0.001) in these samples. Our data suggest that the loss of CaSR gives the tumour cells growth advantage. In the tumour samples we found negative correlation between CaSR and licensing factors (CDC45 (p<0.05, ρ=−0.265) and MCM6 (p<0.05, ρ=−0.262) reaching significance). Surprisingly, CaSR showed positive correlation with the licensing factors in the ‘normal’ mucosa (CDT1 (p<0.05, ρ=0.269), MCM2 (p<0.003, ρ=0.382) and MCM5 (p<0.001, ρ=0.490) reaching significance). These results are similar to our observation in the mouse study, where low dietary Ca2+ enlarged the proliferative zone of the crypts with no significant change in CaSR expression. Conclusion: We conclude that the CaSR might regulate the calcium-dependent inhibition of proliferation in normal colonocytes. Upregulation of CaSR is probably a defence mechanism of the normal colon to control epithelial growth and counteract hyper-proliferative signals. This control mechanism is lost in cancer where CaSR expression is downregulated. Citation Format: Abhishek Aggarwal, Julia Hobaus, Irfete Sh. Fetahu, Ildiko Mesteri, Eniko Kallay. Calcium mediated growth control in normal and transformed colon cells: A possible role of the calcium sensing receptor. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 5314. doi:10.1158/1538-7445.AM2013-5314

Collaboration


Dive into the Julia Höbaus's collaboration.

Top Co-Authors

Avatar

Enikö Kállay

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Ildiko Mesteri

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Doris M. Hummel

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Irfete S. Fetahu

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Samawansha Tennakoon

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Ursula Thiem

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Enikő Kállay

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

I.Sh. Fetahu

Medical University of Vienna

View shared research outputs
Researchain Logo
Decentralizing Knowledge