Network


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

Hotspot


Dive into the research topics where Taras Stasyk is active.

Publication


Featured researches published by Taras Stasyk.


Nature | 2015

SLC38A9 is a component of the lysosomal amino acid sensing machinery that controls mTORC1

Manuele Rebsamen; Lorena Pochini; Taras Stasyk; Mariana E. G. de Araujo; Michele Galluccio; Richard K. Kandasamy; Berend Snijder; Astrid Fauster; Elena L. Rudashevskaya; Manuela Bruckner; Stefania Scorzoni; Przemyslaw A. Filipek; Kilian Huber; Johannes W. Bigenzahn; Leonhard X. Heinz; Claudine Kraft; Keiryn L. Bennett; Cesare Indiveri; Lukas A. Huber; Giulio Superti-Furga

Cell growth and proliferation are tightly linked to nutrient availability. The mechanistic target of rapamycin complex 1 (mTORC1) integrates the presence of growth factors, energy levels, glucose and amino acids to modulate metabolic status and cellular responses. mTORC1 is activated at the surface of lysosomes by the RAG GTPases and the Ragulator complex through a not fully understood mechanism monitoring amino acid availability in the lysosomal lumen and involving the vacuolar H+-ATPase. Here we describe the uncharacterized human member 9 of the solute carrier family 38 (SLC38A9) as a lysosomal membrane-resident protein competent in amino acid transport. Extensive functional proteomic analysis established SLC38A9 as an integral part of the Ragulator–RAG GTPases machinery. Gain of SLC38A9 function rendered cells resistant to amino acid withdrawal, whereas loss of SLC38A9 expression impaired amino-acid-induced mTORC1 activation. Thus SLC38A9 is a physical and functional component of the amino acid sensing machinery that controls the activation of mTOR.


Proteomics | 2008

Intracellular signaling pathways regulating radioresistance of human prostate carcinoma cells

Ira Skvortsova; Sergej Skvortsov; Taras Stasyk; Uma Raju; Bela Andre Popper; Bernhard Schiestl; Elisabeth von Guggenberg; Andreas Neher; Guenther K. Bonn; Lukas A. Huber; Peter Lukas

Radiation therapy plays an important role in the management of prostate carcinoma. However, the problem of radioresistance and molecular mechanisms by which prostate carcinoma cells overcome cytotoxic effects of radiation therapy remains to be elucidated. In order to investigate possible intracellular mechanisms underlying the prostate carcinoma recurrences after radiotherapy, we have established three radiation‐resistant prostate cancer cell lines, LNCaP‐IRR, PC3‐IRR, and Du145‐IRR derived from the parental LNCaP, PC3, and Du145 prostate cancer cells by repetitive exposure to ionizing radiation. LNCaP‐IRR, PC3‐IRR, and Du145‐IRR cells (prostate carcinoma cells recurred after radiation exposure (IRR cells)) showed higher radioresistance and cell motility than parental cell lines. IRR cells exhibited higher levels of androgen and epidermal growth factor (EGF) receptors and activation of their downstream pathways, such as Ras‐mitogen‐activated protein kinase (MAPK) and phosphatidyl inositol 3‐kinase (PI3K)‐Akt and Jak‐STAT. In order to define additional mechanisms involved in the radioresistance development, we determined differences in the proteome profile of parental and IRR cells using 2‐D DIGE followed by computational image analysis and MS. Twenty‐seven proteins were found to be modulated in all three radioresistant cell lines compared to parental cells. Identified proteins revealed capacity to interact with EGF and androgen receptors related signal transduction pathways and were involved in the regulation of intracellular routs providing cell survival, increased motility, mutagenesis, and DNA repair. Our data suggest that radioresistance development is accompanied by multiple mechanisms, including activation of cell receptors and related downstream signal transduction pathways. Identified proteins regulated in the radioresistant prostate carcinoma cells can significantly intensify activation of intracellular signaling that govern cell survival, growth, proliferation, invasion, motility, and DNA repair. In addition, such analyses may be utilized in predicting cellular response to radiotherapy.


Proteomics | 2008

Quantitative proteomics and phosphoproteomics reveal novel insights into complexity and dynamics of the EGFR signaling network.

Sandra Morandell; Taras Stasyk; Sergej Skvortsov; Stefan Ascher; Lukas A. Huber

The epidermal growth factor receptor (EGFR/ErbB1/Her1) belongs to the ErbB family of receptor tyrosine kinases (RTKs) and is a key player in the regulation of cell proliferation, differentiation, survival, and migration. Overexpression and mutational changes of EGFR have been identified in a variety of human cancers and the regulation of EGFR signaling plays a critical role in tumor development and progression. Due to its biological significance the EGFR signaling network is a widely used model system for the development of analytical techniques. Novel quantitative proteomics and phosphoproteomics approaches play an important role in the characterization of signaling pathways in a time and stimulus dependent manner. Recent studies discussed in this review provide new insights into different aspects of EGFR signal transduction, such as regulation and dynamics of its phosphorylation sites, association with interaction partners and identification of regulated phosphoproteins. Correlation of data from functional proteomics studies with results from other fields of signal transduction research by systems biology will be necessary to integrate and translate these findings into successful clinical applications.


Journal of Proteome Research | 2011

Proteomics profiling of microdissected low- and high-grade prostate tumors identifies Lamin A as a discriminatory biomarker.

Sergej Skvortsov; Georg Schäfer; Taras Stasyk; Christian Fuchsberger; Guenther K. Bonn; Georg Bartsch; Helmut Klocker; Lukas A. Huber

Proteomics screening methods for the identification of diagnostic and prognostic biomarkers in cancer are still lagging behind DNA- or RNA-based analysis. We used two-dimensional differential gel electrophoresis (2D-DIGE) in combination with laser capture microdissection (LCM) and MALDI-TOF/TOF mass spectrometry to determine differentially abundant proteins and candidate biomarkers in prostate cancer. Paired (benign and tumor) samples were isolated from 23 Gleason Score 6 (GS 6) and 23 Gleason Score 8 and higher (GS 8+) radical prostatectomy specimens and subjected to 2D-DIGE analysis. Minimal fluorescent dye labeling was applied and electrophoresis performed with triple samples (paired benign and tumor; internal control) for each case of tumor. Nineteen differently abundant proteins were identified by mass spectrometry and further validated. One half of them were associated with glycolysis and the Warburg effect; these were upregulated in tumors. The upregulation correlated with tumor dedifferentiation and might be relevant for selection of therapeutic strategies. Among the other proteins, heat shock protein 60 (HSP60) was significantly upregulated in tumor tissue compared to its benign counterpart. Furthermore, lamin A was statistically highly discriminatory between low and high Gleason score tumors and might serve as a new biomarker of tumor differentiation and prognosis.


Radiotherapy and Oncology | 2010

Epithelial-to-mesenchymal transition and c-myc expression are the determinants of cetuximab-induced enhancement of squamous cell carcinoma radioresponse

Ira Skvortsova; Sergej Skvortsov; Uma Raju; Taras Stasyk; Oliver Riesterer; Eva Maria Schottdorf; Bela Andre Popper; Bernhard Schiestl; Paul Eichberger; Paul Debbage; Andreas Neher; Guenther K. Bonn; Lukas A. Huber; Luka Milas; Peter Lukas

PURPOSE Radiation therapy cures malignant tumors of the head and neck region more effectively when it is combined with application of the anti-EGFR monoclonal antibody cetuximab. Despite the successes achieved, we still do not know how to select patients who will respond to this combination of anti-EGFR monoclonal antibody and radiation. This study was conducted to elucidate possible mechanisms which cause the combined treatment with cetuximab and irradiation to fail in some cases of squamous cell carcinomas. METHODS AND MATERIALS Mice bearing FaDu and A431 squamous cell carcinoma xenograft tumors were treated with cetuximab (total dose 3 mg, intraperitoneally), irradiation (10 Gy) or their combination at the same doses. Treatment was applied when tumors reached 8mm in size. To collect samples for further protein analysis (two-dimensional differential gel electrophoresis (2-D DIGE), mass spectrometry MALDI-TOF/TOF, Western blot analysis, and ELISA), mice from each group were sacrificed on the 8th day after the first injection of cetuximab. Other mice were subjected to tumor growth delay assay. RESULTS In FaDu xenografts, treatment with cetuximab alone was nearly as effective as cetuximab combined with ionizing radiation, whereas A431 tumors responded to the combined treatment with significantly enhanced delay in tumor growth. Tumors extracted from the untreated FaDu and A431 xenografts were analysed for protein expression, and 34 proteins that were differently expressed in the two tumor types were identified. The majority of these proteins are closely related to intratumoral angiogenesis, cell adhesion, motility, differentiation, epithelial-to-mesenchymal transition (EMT), c-myc signaling and DNA repair. CONCLUSIONS The failure of cetuximab to enhance radiation response in FaDu xenografts was associated with the initiation of the program of EMT and with c-myc up-regulation in the carcinoma cells. For this reason, c-myc and EMT-related proteins (E-cadherin, vimentin) may be considered as potential biomarkers to predict squamous cell carcinoma response after treatment with cetuximab in combination with radiation.


Molecular & Cellular Proteomics | 2007

Identification of Endosomal Epidermal Growth Factor Receptor Signaling Targets by Functional Organelle Proteomics

Taras Stasyk; Natalia Schiefermeier; Sergej Skvortsov; Heinz Zwierzina; Johan Peränen; Guenther K. Bonn; Lukas A. Huber

Epidermal growth factor (EGF) receptor (EGFR) signal transduction is organized by scaffold and adaptor proteins, which have specific subcellular distribution. On a way from the plasma membrane to the lysosome EGFRs are still in their active state and can signal from distinct subcellular locations. To identify organelle-specific targets of EGF receptor signaling on endosomes a combination of subcellular fractionation, two-dimensional DIGE, fluorescence labeling of phosphoproteins, and MALDI-TOF/TOF mass spectrometry was applied. All together 23 EGF-regulated (phospho)proteins were identified as being differentially associated with endosomal fractions by functional organelle proteomics; among them were proteins known to be involved in endosomal trafficking and cytoskeleton rearrangement (Alix, myosin-9, myosin regulatory light chain, Trap1, moesin, cytokeratin 8, septins 2 and 11, and CapZβ). Interestingly R-Ras, a small GTPase of the Ras family that regulates cell survival and integrin activity, was associated with endosomes in a ligand-dependent manner. EGF-dependent association of R-Ras with late endosomes was confirmed by confocal laser scanning immunofluorescence microscopy and Western blotting of endosomal fractions. EGFR tyrosine kinase inhibitor gefitinib was used to confirm EGF-dependent regulation of all identified proteins. EGF-dependent association of signaling molecules, such as R-Ras, with late endosomes suggests signaling specification through intracellular organelles.


Methods of Molecular Biology | 2008

Isolation of Endocitic Organelles by Density Gradient Centrifugation

MarianaEÇaGuimarães Araùjo; Lukas Alfons Hube; Taras Stasyk

Advanced prefractionation strategies, in combination with highly sensitive and accurate mass spectrometers provide powerful means to detect and analyze low abundant proteins on the subcellular and organelle-specific level. Among enrichment techniques, subcellular fractionation has become the most commonly used. Its application gives access to less complex subproteomes and organelle constituents, facilitating downstream analysis. Furthermore, subcellular fractionation allows the identification of proteins that shuttle between different subcellular compartments in a stimulus dependent manner. As a paradigm of subcellular organelle isolation, we describe here endosomal purification protocols, based on differential centrifugation in continuous and discontinuous sucrose gradients. Described methods can be easily modified to isolate other organelles and are compatible with subsequent organelle- and functional organelle proteome analyses by, e.g., two-dimensional gel electrophoresis.


Journal of Cell Biology | 2014

The late endosomal p14–MP1 (LAMTOR2/3) complex regulates focal adhesion dynamics during cell migration

Natalia Schiefermeier; Julia M. Scheffler; Mariana E. G. de Araujo; Taras Stasyk; Teodor Yordanov; Hannes L. Ebner; Martin Offterdinger; Sebastian Munck; Michael W. Hess; Sara A. Wickström; Anika Lange; Winfried Wunderlich; Reinhard Fässler; David Teis; Lukas A. Huber

Late endosomes locally regulate cell migration by transporting the p14–MP1 scaffold complex to the vicinity of focal adhesions.


RNA | 2010

Identification of novel ribonucleo-protein complexes from the brain-specific snoRNA MBII-52

Yuuichi Soeno; Yuji Taya; Taras Stasyk; Lukas A. Huber; Takaaki Aoba; Alexander Hüttenhofer

Small nucleolar RNAs (snoRNAs) guide nucleotide modifications within ribosomal RNAs or spliceosomal RNAs by base-pairing to complementary regions within their RNA targets. The brain-specific snoRNA MBII-52 lacks such a complementarity to rRNAs or snRNAs, but instead has been reported to target the serotonin receptor 2C pre-mRNA, thereby regulating pre-mRNA editing and/or alternative splicing. To understand how the MBII-52 snoRNA might be involved in these regulatory processes, we isolated the MBII-52 snoRNP from total mouse brain by an antisense RNA affinity purification approach. Surprisingly, by mass spectrometry we identified 17 novel candidates for MBII-52 snoRNA binding proteins, which previously had not been reported to be associated with canonical snoRNAs. Among these, Nucleolin and ELAVL1 proteins were confirmed to independently and directly interact with the MBII-52 snoRNA by coimmunoprecipitation. Our findings suggest that the MBII-52 snoRNA assembles into novel RNA-protein complexes, distinct from canonical snoRNPs.


Trends in Molecular Medicine | 2012

Mapping in vivo signal transduction defects by phosphoproteomics

Taras Stasyk; Lukas A. Huber

Abnormal protein phosphorylation is implicated in a variety of diseases, but until recently the complexity of tissue material, technical limitations, and the substantial volume of required data processing did not allow large-scale phosphoproteomic analysis of patient material, despite tremendous progress in developing mass spectrometry technologies. Phosphoproteomic approaches were primarily developed using model systems such as transformed cell lines, but technological advances in proteomics now make it feasible to analyze thousands of phosphorylation sites in a quantitative manner in patient materials or complex animal and cellular model systems to identify signaling abnormalities. This review summarizes very recent phosphoproteomic studies on complex tissue material, including tissue samples in biobanks, to complement recent reviews that focus primarily on technical advances in instrumentation and methods. Several successful examples reviewed here suggest it is now possible to apply phosphoproteomic techniques to address more challenging medical questions such as mapping within patient samples signal transduction defects that are relevant for diagnosis and individualized treatment development.

Collaboration


Dive into the Taras Stasyk's collaboration.

Top Co-Authors

Avatar

Lukas A. Huber

Innsbruck Medical University

View shared research outputs
Top Co-Authors

Avatar

Guenther K. Bonn

Innsbruck Medical University

View shared research outputs
Top Co-Authors

Avatar

Sandra Morandell

Innsbruck Medical University

View shared research outputs
Top Co-Authors

Avatar

Sergej Skvortsov

Innsbruck Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Isabel Feuerstein

Innsbruck Medical University

View shared research outputs
Top Co-Authors

Avatar

Herbert Lindner

Innsbruck Medical University

View shared research outputs
Top Co-Authors

Avatar

Hong-Lei Huang

Innsbruck Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge