Network


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

Hotspot


Dive into the research topics where Magdalena Winiarska is active.

Publication


Featured researches published by Magdalena Winiarska.


PLOS Medicine | 2008

Statins impair antitumor effects of rituximab by inducing conformational changes of CD20.

Magdalena Winiarska; Jacek Bil; Ewa Wilczek; Grzegorz M. Wilczynski; Malgorzata Lekka; Patrick J. Engelberts; Wendy J.M. Mackus; Elżbieta Górska; Lukasz Bojarski; Tomasz Stoklosa; Dominika Nowis; Zuzanna Kurzaj; Marcin Makowski; Eliza Glodkowska; Tadeusz Issat; Piotr Mrowka; Witold Lasek; Anna Dabrowska-Iwanicka; Grzegorz W. Basak; Maria Wasik; Krzysztof Warzocha; Maciej Siński; Zbigniew Gaciong; Marek Jakóbisiak; Paul Parren; Jakub Golab

Background Rituximab is used in the treatment of CD20+ B cell lymphomas and other B cell lymphoproliferative disorders. Its clinical efficacy might be further improved by combinations with other drugs such as statins that inhibit cholesterol synthesis and show promising antilymphoma effects. The objective of this study was to evaluate the influence of statins on rituximab-induced killing of B cell lymphomas. Methods and Findings Complement-dependent cytotoxicity (CDC) was assessed by MTT and Alamar blue assays as well as trypan blue staining, and antibody-dependent cellular cytotoxicity (ADCC) was assessed by a 51Cr release assay. Statins were found to significantly decrease rituximab-mediated CDC and ADCC of B cell lymphoma cells. Incubation of B cell lymphoma cells with statins decreased CD20 immunostaining in flow cytometry studies but did not affect total cellular levels of CD20 as measured with RT-PCR and Western blotting. Similar effects are exerted by other cholesterol-depleting agents (methyl-β-cyclodextrin and berberine), but not filipin III, indicating that the presence of plasma membrane cholesterol and not lipid rafts is required for rituximab-mediated CDC. Immunofluorescence microscopy using double staining with monoclonal antibodies (mAbs) directed against a conformational epitope and a linear cytoplasmic epitope revealed that CD20 is present in the plasma membrane in comparable amounts in control and statin-treated cells. Atomic force microscopy and limited proteolysis indicated that statins, through cholesterol depletion, induce conformational changes in CD20 that result in impaired binding of anti-CD20 mAb. An in vivo reduction of cholesterol induced by short-term treatment of five patients with hypercholesterolemia with atorvastatin resulted in reduced anti-CD20 binding to freshly isolated B cells. Conclusions Statins were shown to interfere with both detection of CD20 and antilymphoma activity of rituximab. These studies have significant clinical implications, as impaired binding of mAbs to conformational epitopes of CD20 elicited by statins could delay diagnosis, postpone effective treatment, or impair anti-lymphoma activity of rituximab.


Molecules | 2011

Aminolevulinic Acid (ALA) as a Prodrug in Photodynamic Therapy of Cancer

Malgorzata Wachowska; Angelika Muchowicz; Malgorzata Firczuk; Magdalena Gabrysiak; Magdalena Winiarska; Malgorzata Wanczyk; Kamil Bojarczuk; Jakub Golab

Aminolevulinic acid (ALA) is an endogenous metabolite normally formed in the mitochondria from succinyl-CoA and glycine. Conjugation of eight ALA molecules yields protoporphyrin IX (PpIX) and finally leads to formation of heme. Conversion of PpIX to its downstream substrates requires the activity of a rate-limiting enzyme ferrochelatase. When ALA is administered externally the abundantly produced PpIX cannot be quickly converted to its final product - heme by ferrochelatase and therefore accumulates within cells. Since PpIX is a potent photosensitizer this metabolic pathway can be exploited in photodynamic therapy (PDT). This is an already approved therapeutic strategy making ALA one of the most successful prodrugs used in cancer treatment.


American Journal of Pathology | 2010

Cardiotoxicity of the Anticancer Therapeutic Agent Bortezomib

Dominika Nowis; Michał Mączewski; Urszula Mackiewicz; Marek Kujawa; Anna Ratajska; Mariusz R. Wieckowski; Grzegorz M. Wilczynski; Monika Malinowska; Jacek Bil; Pawel Salwa; Marek Bugajski; Cezary Wójcik; Maciej Siński; Piotr Abramczyk; Magdalena Winiarska; Anna Dąbrowska-Iwanicka; Jerzy Duszyński; Marek Jakóbisiak; Jakub Golab

Recent case reports provided alarming signals that treatment with bortezomib might be associated with cardiac events. In all reported cases, patients experiencing cardiac problems were previously or concomitantly treated with other chemotherapeutics including cardiotoxic anthracyclines. Therefore, it is difficult to distinguish which components of the therapeutic regimens contribute to cardiotoxicity. Here, we addressed the influence of bortezomib on cardiac function in rats that were not treated with other drugs. Rats were treated with bortezomib at a dose of 0.2 mg/kg thrice weekly. Echocardiography, histopathology, and electron microscopy were used to evaluate cardiac function and structural changes. Respiration of the rat heart mitochondria was measured polarographically. Cell culture experiments were used to determine the influence of bortezomib on cardiomyocyte survival, contractility, Ca(2+) fluxes, induction of endoplasmic reticulum stress, and autophagy. Our findings indicate that bortezomib treatment leads to left ventricular contractile dysfunction manifested by a significant drop in left ventricle ejection fraction. Dramatic ultrastructural abnormalities of cardiomyocytes, especially within mitochondria, were accompanied by decreased ATP synthesis and decreased cardiomyocyte contractility. Monitoring of cardiac function in bortezomib-treated patients should be implemented to evaluate how frequently cardiotoxicity develops especially in patients with pre-existing cardiac conditions, as well as when using additional cardiotoxic drugs.


Frontiers in Bioscience | 2011

HDACi--going through the mechanisms.

Malgorzata Wanczyk; Katarzyna Roszczenko; Marcinkiewicz K; Kamil Bojarczuk; Kowara M; Magdalena Winiarska

Histone deacetylases inhibitors (HDACi) have recently emerged as potent antitumor treatment modality. They are currently tested in many phase I, II and III clinical trials as single agents as wells as in combination schemes. They have demonstrated promising antitumor activity and favorable clinical outcome. Histone deacetylases (HDACs) are involved in the process of epigenetic regulation of gene expression. Epigenetic changes are believed to be crucial for the onset and progression of cancer and have recently gained remarkable attention. Since epigenetic regulation of gene expression is a reversible process, targeting histone deacetylases provides a good rationale for anticancer therapy. The acetylation status of histones regulates the organization of chromatin and the access of transcription factors. Moreover, functions of many non-histone proteins are controlled by acetylation. The broad and complicated influences of HDACi on various molecular processes may account for the observed pleiotropic effects. In this review we summarize recent advances in the understanding of biology of HDACs and mechanism of action of their inhibitors.


BMC Cancer | 2008

Zinc protoporphyrin IX, a heme oxygenase-1 inhibitor, demonstrates potent antitumor effects but is unable to potentiate antitumor effects of chemotherapeutics in mice

Dominika Nowis; Marek Bugajski; Magdalena Winiarska; Jacek Bil; Angelika Szokalska; Pawel Salwa; Tadeusz Issat; Halina Was; Alicja Jozkowicz; Jozef Dulak; Tomasz Stoklosa; Jakub Golab

BackgroundHO-1 participates in the degradation of heme. Its products can exert unique cytoprotective effects. Numerous tumors express high levels of HO-1 indicating that this enzyme might be a potential therapeutic target. In this study we decided to evaluate potential cytostatic/cytotoxic effects of zinc protoporphyrin IX (Zn(II)PPIX), a selective HO-1 inhibitor and to evaluate its antitumor activity in combination with chemotherapeutics.MethodsCytostatic/cytotoxic effects of Zn(II)PPIX were evaluated with crystal violet staining and clonogenic assay. Western blotting was used for the evaluation of protein expression. Flow cytometry was used to evaluate the influence of Zn(II)PPIX on the induction of apoptosis and generation of reactive oxygen species. Knock-down of HO-1 expression was achieved with siRNA. Antitumor effects of Zn(II)PPIX alone or in combination with chemotherapeutics were measured in transplantation tumor models.ResultsZn(II)PPIX induced significant accumulation of reactive oxygen species in tumor cells. This effect was partly reversed by administration of exogenous bilirubin. Moreover, Zn(II)PPIX exerted potent cytostatic/cytotoxic effects against human and murine tumor cell lines. Despite a significant time and dose-dependent decrease in cyclin D expression in Zn(II)PPIX-treated cells no accumulation of tumor cells in G1 phase of the cell cycle was observed. However, incubation of C-26 cells with Zn(II)PPIX increased the percentage of cells in sub-G1 phase of the cells cycle. Flow cytometry studies with propidium iodide and annexin V staining as well as detection of cleaved caspase 3 by Western blotting revealed that Zn(II)PPIX can induce apoptosis of tumor cells. B16F10 melanoma cells overexpressing HO-1 and transplanted into syngeneic mice were resistant to either Zn(II)PPIX or antitumor effects of cisplatin. Zn(II)PPIX was unable to potentiate antitumor effects of 5-fluorouracil, cisplatin or doxorubicin in three different tumor models, but significantly potentiated toxicity of 5-FU and cisplatin.ConclusionInhibition of HO-1 exerts antitumor effects but should not be used to potentiate antitumor effects of cancer chemotherapeutics unless procedures of selective tumor targeting of HO-1 inhibitors are developed.


Frontiers in Bioscience | 2011

Approaches to improve photodynamic therapy of cancer.

Malgorzata Firczuk; Magdalena Winiarska; Angelika Szokalska; Malgorzata Jodlowska; Marta Swiech; Kamil Bojarczuk; Pawel Salwa; Dominika Nowis

Photodynamic therapy (PDT) is a clinically approved method of tumor treatment. Its unique mechanism of action results from minimal invasiveness and high selectivity towards transformed cells. However, visible light used to excite most photosensitizers has rather limited ability to penetrate tissues resulting in insufficient destruction of deeply seated malignant cells. Therefore, novel strategies for further potentiation of the anticancer effectiveness of PDT have been developed. These include combined treatments with surgery, chemo- and radiotherapy, strategies targeting cytoprotective mechanisms induced in PDT-treated cells, as well as attempts aimed at enhancement of PDT-mediated antitumor immune response. Moreover, new photosensitizers and novel light sources are being developed. Impressive progress in nanotechnology and understanding of tumor cell biology rise hopes for further improvements in this elegant and promising method of cancer treatment.


Leukemia | 2014

B-cell receptor pathway inhibitors affect CD20 levels and impair antitumor activity of anti-CD20 monoclonal antibodies

Kamil Bojarczuk; Marta Siernicka; Michal Dwojak; Malgorzata Bobrowicz; Beata Pyrzynska; P Gaj; Marta Karp; Krzysztof Giannopoulos; Dimitar G. Efremov; C Fauriat; Jakub Golab; Magdalena Winiarska

B-cell receptor pathway inhibitors affect CD20 levels and impair antitumor activity of anti-CD20 monoclonal antibodies


Frontiers in Bioscience | 2011

Molecular mechanisms of the antitumor effects of anti-CD20 antibodies.

Magdalena Winiarska; Eliza Glodkowska-Mrowka; Jacek Bil; Jakub Golab

Anti-CD20 monoclonal antibodies (mAbs) have become the mainstay in the treatment of non-Hodgkins lymphomas and have shown significant activity in patients with B-cell chronic lymphocytic leukemia. Antitumor action of these antibodies results from triggering of indirect effector mechanisms of the immune system that include activation of complement-dependent cytotoxicity (CDC), antibody-dependent cellular cytotoxicity (ADCC), or phagocytosis. Moreover, some studies indicate direct influence of anti-CD20 mAbs on tumor cells that leads to induction of various types of cell death. Despite the wealth of data on the mechanisms of cytotoxicity that accumulated over the last two decades their relative contribution to the therapeutic outcome is still difficult to predict in individual patients. Elucidation of molecular mechanisms of anti-CD20 mAbs action is necessary to deliver their maximal activity in rationally designed combinations with other therapeutic approaches and to design next generation anti-CD20 mAb with improved ability to eliminate tumor cells.


Blood | 2010

Bortezomib modulates surface CD20 in B-cell malignancies and affects rituximab-mediated complement-dependent cytotoxicity

Jacek Bil; Magdalena Winiarska; Dominika Nowis; Kamil Bojarczuk; Anna Dabrowska-Iwanicka; Grzegorz W. Basak; Sułek K; Marek Jakóbisiak; Jakub Golab

Recent observations indicate that rituximab-resistant lymphoma cells exhibit upregulation of components of the ubiquitin-proteasome system (UPS). Therefore, proteasome inhibitors including the clinically approved bortezomib might influence the levels of CD20, a rituximab target antigen. We observed that incubation of tumor cells with rituximab leads to increased levels of ubiquitinated CD20. However, inhibition of the UPS is not associated with upregulation, but rather with a counterintuitive downregulation of surface CD20 levels that increases resistance of tumor cells to rituximab-mediated cytotoxicity. Although preliminary observations indicate that CD20 might be a substrate for two proteolytic systems, the mechanisms as well as significance of these findings require further studies.Unresponsiveness to rituximab treatment develops in many patients prompting elucidation of underlying molecular pathways. It was recently observed that rituximab-resistant lymphoma cells exhibit up-regulation of components of the ubiquitin-proteasome system (UPS). Therefore, we investigated in more detail the role of this system in the regulation of CD20 levels and the influence of proteasome inhibitors on rituximab-mediated complement-dependent cytotoxicity (R-CDC). We observed that incubation of Raji cells with rituximab leads to increased levels of ubiquitinated CD20. However, inhibition of the UPS was not associated with up-regulation of surface CD20 levels, although it significantly increased its ubiquitination. Short-term (24 hours) incubation of Raji cells with 10 or 20 nM bortezomib did not change surface CD20 levels, but sensitized CD20(+) lymphoma cells to R-CDC. Prolonged (48 hours) incubation with 20 nM bortezomib, or incubation with 50 nM bortezomib for 24 hours led to a significant decrease in surface CD20 levels as well as R-CDC. These effects were partly reversed by bafilomycin A1, an inhibitor of lysosomal/autophagosomal pathway of protein degradation. These studies indicate that CD20 levels are regulated by 2 proteolytic systems and that the use of proteasome inhibitors may be associated with unexpected negative influence on R-CDC.


Oncotarget | 2016

Dimeric peroxiredoxins are druggable targets in human Burkitt lymphoma

Anna Trzeciecka; Szymon Klossowski; Malgorzata Bajor; Radoslaw Zagozdzon; Pawel Gaj; Angelika Muchowicz; Agata Malinowska; Anna Czerwoniec; Joanna Barankiewicz; Antoni Domagala; Justyna Chlebowska; Monika Prochorec-Sobieszek; Magdalena Winiarska; Ryszard Ostaszewski; Iwonna Gwizdalska; Jakub Golab; Dominika Nowis; Malgorzata Firczuk

Burkitt lymphoma is a fast-growing tumor derived from germinal center B cells. It is mainly treated with aggressive chemotherapy, therefore novel therapeutic approaches are needed due to treatment toxicity and developing resistance. Disturbance of red-ox homeostasis has recently emerged as an efficient antitumor strategy. Peroxiredoxins (PRDXs) are thioredoxin-family antioxidant enzymes that scavenge cellular peroxides and contribute to red-ox homeostasis. PRDXs are robustly expressed in various malignancies and critically involved in cell proliferation, differentiation and apoptosis. To elucidate potential role of PRDXs in lymphoma, we studied their expression level in B cell-derived primary lymphoma cells as well as in cell lines. We found that PRDX1 and PRDX2 are upregulated in tumor B cells as compared with normal counterparts. Concomitant knockdown of PRDX1 and PRDX2 significantly attenuated the growth rate of lymphoma cells. Furthermore, in human Burkitt lymphoma cell lines, we isolated dimeric 2-cysteine peroxiredoxins as targets for SK053, a novel thiol-specific small-molecule peptidomimetic with antitumor activity. We observed that treatment of lymphoma cells with SK053 triggers formation of covalent PRDX dimers, accumulation of intracellular reactive oxygen species, phosphorylation of ERK1/2 and AKT and leads to cell cycle arrest and apoptosis. Based on site-directed mutagenesis and modeling studies, we propose a mechanism of SK053-mediated PRDX crosslinking, involving double thioalkylation of active site cysteine residues. Altogether, our results suggest that peroxiredoxins are novel therapeutic targets in Burkitt lymphoma and provide the basis for new approaches to the treatment of this disease.

Collaboration


Dive into the Magdalena Winiarska's collaboration.

Top Co-Authors

Avatar

Jakub Golab

Medical University of Warsaw

View shared research outputs
Top Co-Authors

Avatar

Kamil Bojarczuk

Medical University of Warsaw

View shared research outputs
Top Co-Authors

Avatar

Dominika Nowis

Medical University of Warsaw

View shared research outputs
Top Co-Authors

Avatar

Michal Dwojak

Medical University of Warsaw

View shared research outputs
Top Co-Authors

Avatar

Malgorzata Bobrowicz

Medical University of Warsaw

View shared research outputs
Top Co-Authors

Avatar

Marta Siernicka

Medical University of Warsaw

View shared research outputs
Top Co-Authors

Avatar

Malgorzata Firczuk

Medical University of Warsaw

View shared research outputs
Top Co-Authors

Avatar

Beata Pyrzynska

Medical University of Warsaw

View shared research outputs
Top Co-Authors

Avatar

Angelika Muchowicz

Medical University of Warsaw

View shared research outputs
Top Co-Authors

Avatar

Nina Miazek

Medical University of Warsaw

View shared research outputs
Researchain Logo
Decentralizing Knowledge