Network


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

Hotspot


Dive into the research topics where Chi Huu Nguyen is active.

Publication


Featured researches published by Chi Huu Nguyen.


British Journal of Cancer | 2016

Cancer cell-derived 12(S)-HETE signals via 12-HETE receptor, RHO, ROCK and MLC2 to induce lymph endothelial barrier breaching

Chi Huu Nguyen; Serena Stadler; Stefan Brenner; Nicole Huttary; Sigurd Krieger; Walter Jäger; Helmut Dolznig; Georg Krupitza

Background:The arachidonic acid metabolite 12(S)-HETE is suspected to enhance metastatic spread by inducing cancer cell- and lymph endothelial cell (LEC) motility. However, the molecular mechanisms leading to 12(S)-HETE-triggered cell migration are still elusive.Methods:To delineate the signalling pathways involved in 12(S)-HETE-mediated migration, inhibitors against RHO and ROCK, and specific siRNAs downregulating 12(S)-HETE receptor (12-HETER) and myosin light chain 2 (MLC2) were used. The breaching of the endothelial barrier was investigated by an assay measuring tumour spheroid-triggered ‘circular chemorepellent-induced defects’ (CCIDs), and respective signal transduction was elucidated by western blotting.Results:We provide evidence that 12(S)-HETE phosphorylated (and activated) MLC2, which regulates actin/myosin-based contraction. MLC2 activation was found to be essential for LEC retraction and CCID formation. Furthermore, we show that 12(S)-HETE activated a 12-HETER–RHO–ROCK–MYPT signalling cascade to induce MLC2 function.Conclusions:Signalling via this pathway is described for this metabolite for the first time. This may provide potential targets for the intervention of metastatic colonisation.


Cellular and Molecular Life Sciences | 2017

Colon cancer cell-derived 12(S)-HETE induces the retraction of cancer-associated fibroblast via MLC2, RHO/ROCK and Ca2+ signalling

Serena Stadler; Chi Huu Nguyen; Helga Schachner; Daniela Milovanovic; Silvio Holzner; Stefan Brenner; Julia Eichsteininger; Mira Stadler; Daniel Senfter; Liselotte Krenn; Wolfgang Schmidt; Nicole Huttary; Sigurd Krieger; Oskar Koperek; Zsuzsanna Bago-Horvath; Konstantin Alexander Brendel; Brigitte Marian; Olivier De Wever; Robert M. Mader; Benedikt Giessrigl; Walter Jäger; Helmut Dolznig; Georg Krupitza

Retraction of mesenchymal stromal cells supports the invasion of colorectal cancer cells (CRC) into the adjacent compartment. CRC-secreted 12(S)-HETE enhances the retraction of cancer-associated fibroblasts (CAFs) and therefore, 12(S)-HETE may enforce invasivity of CRC. Understanding the mechanisms of metastatic CRC is crucial for successful intervention. Therefore, we studied pro-invasive contributions of stromal cells in physiologically relevant three-dimensional in vitro assays consisting of CRC spheroids, CAFs, extracellular matrix and endothelial cells, as well as in reductionist models. In order to elucidate how CAFs support CRC invasion, tumour spheroid-induced CAF retraction and free intracellular Ca2+ levels were measured and pharmacological- or siRNA-based inhibition of selected signalling cascades was performed. CRC spheroids caused the retraction of CAFs, generating entry gates in the adjacent surrogate stroma. The responsible trigger factor 12(S)-HETE provoked a signal, which was transduced by PLC, IP3, free intracellular Ca2+, Ca2+-calmodulin-kinase-II, RHO/ROCK and MYLK which led to the activation of myosin light chain 2, and subsequent CAF mobility. RHO activity was observed downstream as well as upstream of Ca2+ release. Thus, Ca2+ signalling served as central signal amplifier. Treatment with the FDA-approved drugs carbamazepine, cinnarizine, nifedipine and bepridil HCl, which reportedly interfere with cellular calcium availability, inhibited CAF-retraction. The elucidation of signalling pathways and identification of approved inhibitory drugs warrant development of intervention strategies targeting tumour–stroma interaction.


Oncotarget | 2015

NF-κB contributes to MMP1 expression in breast cancer spheroids causing paracrine PAR1 activation and disintegrations in the lymph endothelial barrier in vitro

Chi Huu Nguyen; Daniel Senfter; Jose Basilio; Silvio Holzner; Serena Stadler; Sigurd Krieger; Nicole Huttary; Daniela Milovanovic; Katharina Viola; Ingrid Simonitsch-Klupp; Walter Jäger; Rainer de Martin; Georg Krupitza

RELA, RELB, CREL, NFKB1 and NFKB2, and the upstream regulators NEMO and NIK were knocked-down in lymph endothelial cells (LECs) and in MDA-MB231 breast cancer spheroids to study the contribution of NF-κB in vascular barrier breaching. Suppression of RELA, NFKB1 and NEMO inhibited “circular chemo-repellent induced defects” (CCIDs), which form when cancer cells cross the lymphatic vasculature, by ~20–30%. Suppression of RELB, NFKB2 and NIK inhibited CCIDs by only ~10–15%. In MDA-MB231 cells RELA and NFKB1 constituted MMP1 expression, which caused the activation of PAR1 in adjacent LECs. The knock-down of MMP1 in MDA-MB231 spheroids and pharmacological inhibition of PAR1 in LECs inhibited CCID formation by ~30%. Intracellular Ca2+ release in LECs, which was induced by recombinant MMP1, was suppressed by the PAR1 inhibitor SCH79797, thereby confirming a functional intercellular axis: RELA/NFKB1 – MMP1 (MDA-MB231) – PAR1 (LEC). Recombinant MMP1 induced PAR1-dependent phosphorylation of MLC2 and FAK in LECs, which is indicative for their activity and for directional cell migration such as observed during CCID formation. The combined knock-down of the NF-κB pathways in LECs and MDA-MB231 spheroids inhibited CCIDs significantly stronger than knock-down in either cell type alone. Also the knock-down of ICAM-1 in LECs (a NF-κB endpoint with relevance for CCID formation) and knock-down of MMP1 in MDA-MB231 augmented CCID inhibition. This evidences that in both cell types NF-κB significantly and independently contributes to tumour-mediated breaching of the lymphatic barrier. Hence, inflamed tumour tissue and/or vasculature pose an additional threat to cancer progression.


Human Molecular Genetics | 2016

AHR/CYP1A1 interplay triggers lymphatic barrier breaching in breast cancer spheroids by inducing 12(S)-HETE synthesis

Chi Huu Nguyen; Stefan Brenner; Nicole Huttary; Atanas G. Atanasov; Verena M. Dirsch; Waranya Chatuphonprasert; Sivio Holzner; Serena Stadler; Juliane Riha; Sigurd Krieger; Rainer de Martin; Zsuzsanna Bago-Horvath; Georg Krupitza; Walter Jäger

A causal link between overexpression of aryl hydrocarbon receptor (AHR) and its target cytochrome P450 1A1 (CYP1A1) and metastatic outgrowth of various cancer entities has been established. Nevertheless, the mechanism how AHR/CYP1A1 support metastasis formation is still little understood. In vitro we discovered a potential mechanism facilitating tumour dissemination based on the production of 12(S)-hydroxyeicosatetraenoic acid (12(S)-HETE). Utilising a three-dimensional lymph endothelial cell (LEC) monolayer & MDA-MB231 breast cancer cell spheroid co-culture model in combination with knock-down approach allowed elucidation of the molecular/biochemical basis of AHR/CYP1A1-induced tumour breaching through the LEC barrier. Enzyme immunoassay evidenced the potential of recombinant CYP1A1 to synthesise 12(S)-HETE in vitro and qPCR and Western blotting measured gene and protein expression in specific experimental settings. In detail, AHR induced CYP1A1 expression and 12(S)-HETE secretion in tumour spheroids, which caused LEC junction retraction thereby forming large discontinuities allowing transmigration of the tumour. This was enforced by the activating AHR ligand 6-formylindolo (3,3-b)carbazole (FICZ), or inhibited by the AHR antagonist 3,3’-diindolylmethane (DIM) as well as by siRNA against AHR and CYP1A1. AHR and NF-κB were negatively cross talking and therefore, the inhibition of AHR (but not CYP1A1) induced RELA, RELB, NFKB1, NFKB2 and the NF-κB target MMP1, which itself promotes tumour intravasation by a mechanism that is different from 12(S)-HETE. Conversely, the inhibition of NFKB2 induced AHR, CYP1A1 and 12(S)-HETE synthesis. The approved clinical drugs guanfacine and vinpocetine, which inhibit CYP1A1 and NF-κB, respectively, significantly inhibited LEC barrier breaching in vitro indicating an option to reduce metastatic dissemination.


Mutation Research | 2015

A eudesmane-type sesquiterpene isolated from Pluchea odorata (L.) Cass. combats three hallmarks of cancer cells: Unrestricted proliferation, escape from apoptosis and early metastatic outgrowth in vitro

Michael Blaschke; Ruxandra McKinnon; Chi Huu Nguyen; Silvio Holzner; Martin Zehl; Atanas G. Atanasov; Karin Schelch; Sigurd Krieger; Rene Diaz; Richard Frisch; Björn Feistel; Walter Jäger; Gerhard F. Ecker; Verena M. Dirsch; Michael Grusch; István Zupkó; Ernst Urban; Brigitte Kopp; Georg Krupitza

Pluchea odorata is ethno pharmaceutically used to treat inflammation-associated disorders. The dichloromethane extract (DME) was tested in the carrageenan-induced rat paw oedema assay investigating its effect on inflammation that was inhibited by 37%. Also an in vitro anti-neoplastic potential was reported. However, rather limited information about the bio-activity of purified compounds and their cellular mechanisms are available. Therefore, two of the most abundant eudesmanes in P. odorata were isolated and their anti-neoplastic and anti-intravasative activities were studied. HL-60 cells were treated with P. odorata compounds and metabolic activity, cell number reduction, cell cycle progression and apoptosis induction were correlated with relevant protein expression. Tumour cell intravasation through lymph endothelial monolayers was measured and potential causal mechanisms were analyzed by Western blotting. Compound PO-1 decreased the metabolic activity of HL-60 cells (IC50 = 8.9 μM after 72 h) and 10 μM PO-1 induced apoptosis, while PO-2 showed just weak anti-neoplastic activities at concentrations beyond 100 μM. PO-1 arrested the cell cycle in G1 and this correlated with induction of JunB expression. Independent of this mechanism 25 μM PO-1 decreased MCF-7 spheroid intravasation through the lymph endothelial barrier. Hence, PO-1 inhibits an early step of metastasis, impairs unrestricted proliferation and induces apoptosis at low micromolar concentrations. These results warrant further testing in vivo to challenge the potential of PO-1 as novel lead compound.


Frontiers in Pharmacology | 2018

APIGENIN AND LUTEOLIN ATTENUATE THE BREACHING OF MDA-MB231 BREAST CANCER SPHEROIDS THROUGH THE LYMPH ENDOTHELIAL BARRIER IN VITRO

Junli Hong; Adryan Fristiohady; Chi Huu Nguyen; Daniela Milovanovic; Nicole Huttary; Sigurd Krieger; Junqiang Hong; Silvana Geleff; Peter Birner; Walter Jäger; Ali Özmen; Liselotte Krenn; Georg Krupitza

Flavonoids, present in fruits, vegetables and traditional medicinal plants, show anticancer effects in experimental systems and are reportedly non-toxic. This is a favorable property for long term strategies for the attenuation of lymph node metastasis, which may effectively improve the prognostic states in breast cancer. Hence, we studied two flavonoids, apigenin and luteolin exhibiting strong bio-activity in various test systems in cancer research and are readily available on the market. This study has further advanced the mechanistic understanding of breast cancer intravasation through the lymphatic barrier. Apigenin and luteolin were tested in a three-dimensional (3-D) assay consisting of MDA-MB231 breast cancer spheroids and immortalized lymph endothelial cell (LEC) monolayers. The 3-D model faithfully resembles the intravasation of breast cancer emboli through the lymphatic vasculature. Western blot analysis, intracellular Ca2+ determination, EROD assay and siRNA transfection revealed insights into mechanisms of intravasation as well as the anti-intravasative outcome of flavonoid action. Both flavonoids suppressed pro-intravasative trigger factors in MDA-MB231 breast cancer cells, specifically MMP1 expression and CYP1A1 activity. A pro-intravasative contribution of FAK expression in LECs was established as FAK supported the retraction of the LEC monolayer upon contact with cancer cells thereby enabling them to cross the endothelial barrier. As mechanistic basis, MMP1 caused the phosphorylation (activation) of FAK at Tyr397 in LECs. Apigenin and luteolin prevented MMP1-induced FAK activation, but not constitutive FAK phosphorylation. Luteolin, unlike apigenin, inhibited MMP1-induced Ca2+ release. Free intracellular Ca2+ is a central signal amplifier triggering LEC retraction through activation of the mobility protein MLC2, thereby enhancing intravasation. FAK activity and Ca2+ levels did not correlate. This implicates that the pro-intravasative contribution of FAK and of Ca2+ release in LECs was independent of each other and explains the better anti-intravasative effects of luteolin in vitro. In specific formulations, flavonoid concentrations causing significant anti-intravasative effects, can certainly be achieved in vivo. As the therapeutic strategy has to be based on permanent flavonoid treatment both the beneficial and adverse effects have to be investigated in future studies.


Phytomedicine | 2015

The germacranolide sesquiterpene lactone neurolenin B of the medicinal plant Neurolaena lobata (L.) R.Br. ex Cass inhibits NPM/ALK-driven cell expansion and NF-κB-driven tumour intravasation

Christine Unger; Izabella Kiss; Andrea Vasas; Ildikó Lajter; Nina Kramer; Atanas G. Atanasov; Chi Huu Nguyen; Waranya Chatuphonprasert; Stefan Brenner; Sigurd Krieger; Ruxandra McKinnon; Andrea Peschel; Renate Kain; Philipp Saiko; Thomas Szekeres; Lukas Kenner; Melanie R. Hassler; Rene Diaz; Richard Frisch; Verena M. Dirsch; Walter Jäger; Rainer de Martin; Valery N. Bochkov; Claus M. Passreiter; Barbara Peter-Vörösmarty; Robert M. Mader; Michael Grusch; Helmut Dolznig; Brigitte Kopp; István Zupkó

BACKGROUND The t(2;5)(p23;q35) chromosomal translocation results in the expression of the fusion protein NPM/ALK that when expressed in T-lymphocytes gives rise to anaplastic large cell lymphomas (ALCL). In search of new therapy options the dichloromethane extract of the ethnomedicinal plant Neurolaena lobata (L.) R.Br. ex Cass was shown to inhibit NPM/ALK expression. PURPOSE Therefore, we analysed whether the active principles that were recently isolated and found to inhibit inflammatory responses specifically inhibit growth of NPM/ALK+ ALCL, leukaemia and breast cancer cells, but not of normal cells, and the intravasation through the lymphendothelial barrier. METHODS ALCL, leukaemia and breast cancer cells, and normal peripheral blood mononuclear cells (PBMCs) were treated with isolated sesquiterpene lactones and analysed for cell cycle progression, proliferation, mitochondrial activity, apoptosis, protein and mRNA expression, NF-κB and cytochrome P450 activity, 12(S)-HETE production and lymphendothelial intravasation. RESULTS In vitro treatment of ALCL by neurolenin B suppressed NPM/ALK, JunB and PDGF-Rβ expression, inhibited the growth of ALCL cells late in M phase, and induced apoptosis via caspase 3 without compromising mitochondrial activity (as a measure of general exogenic toxicity). Moreover, neurolenin B attenuated tumour spheroid intravasation probably through inhibition of NF-κB and CYP1A1. CONCLUSION Neurolenin B specifically decreased pro-carcinogenic NPM/ALK expression in ALK+ ALCL cells and, via the inhibition of NF-kB signalling, attenuated tumour intra/extravasation into the lymphatics. Hence, neurolenin B may open new options to treat ALCL and to manage early metastatic processes to which no other therapies exist.


Cancer Letters | 2016

12(S)-HETE increases intracellular Ca2+ in lymph-endothelial cells disrupting their barrier function in vitro; stabilization by clinical drugs impairing calcium supply

Chi Huu Nguyen; Stefan Brenner; Nicole Huttary; Yuanfang Li; Atanas G. Atanasov; Verena M. Dirsch; Silvio Holzner; Serena Stadler; Juliane Riha; Sigurd Krieger; Danijela Milovanovic; Adryan Fristiohardy; Ingrid Simonitsch-Klupp; Helmut Dolznig; Philipp Saiko; Thomas Szekeres; Benedikt Giessrigl; Walter Jäger; Georg Krupitza


Oncology Reports | 2016

Colorectal cancer cell-derived microRNA200 modulates the resistance of adjacent blood endothelial barriers in vitro.

Silvio Holzner; Daniel Senfter; Serena Stadler; Anna Staribacher; Chi Huu Nguyen; Anna Gaggl; Silvana Geleff; Nicole Huttary; Sigurd Krieger; Walter Jäger; Helmut Dolznig; Robert M. Mader; Georg Krupitza


International Journal of Oncology | 2017

Fenofibrate inhibits tumour intravasation by several independent mechanisms in a 3-dimensional co-culture model.

Chi Huu Nguyen; Nicole Huttary; Atanas G. Atanasov; Waranya Chatuphonprasert; Stefan Brenner; Adryan Fristiohady; Junli Hong; Serena Stadler; Silvio Holzner; Daniela Milovanovic; Verena M. Dirsch; Brigitte Kopp; Philipp Saiko; Liselotte Krenn; Walter Jäger; Georg Krupitza

Collaboration


Dive into the Chi Huu Nguyen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Georg Krupitza

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Sigurd Krieger

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Nicole Huttary

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Serena Stadler

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Silvio Holzner

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daniela Milovanovic

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Helmut Dolznig

Medical University of Vienna

View shared research outputs
Researchain Logo
Decentralizing Knowledge