Network


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

Hotspot


Dive into the research topics where Zofia Mazerska is active.

Publication


Featured researches published by Zofia Mazerska.


Biomacromolecules | 2013

Progress in Targeting Tumor Cells by Using Drug-Magnetic Nanoparticles Conjugate

Anna M. Nowicka; Agata Kowalczyk; Anita Jarzębińska; Mikolaj Donten; Paweł Krysiński; Zbigniew Stojek; Ewa Augustin; Zofia Mazerska

To limit cytotoxicity of anticancer drugs against healthy cells, an appropriate carrier should be synthesized to deliver the drug to the tumor tissue only. A good solution is to anchor a magnetic nanoparticle to the molecule of the drug and to use a properly directed external magnetic field. The synthesis of the conjugate of doxorubicin with magnetic nanoparticles (iron oxide) modified by us resulted in a substantial depression of the aggregation process of the nanoparticles and therefore allowed the correct examination of cytotoxicity of the modified drug. It has been shown, by performing the electrochemical microbalance measurements, that the use of magnetic field guaranteed the efficient delivery of the drug to the desired place. The change in the synthesis procedure led to an increase in the number of DOX molecules attached to one magnetic nanoparticle. The release of the drug took place at pH 5.8 (and below it), which pH characterizes the cancer cells. It has also been found that while the iron oxide magnetic nanoparticles were not cytotoxic toward human urinary bladder carcinoma cells UM-UC-3, the tumor cell sensitivity of the DOX-Np complex was slightly higher in comparison to the identical concentration of doxorubicin alone.


The FASEB Journal | 2016

Antitumor DNA-Damaging C-1748 is a New Inhibitor of Autophagy that Triggers Apoptosis in Human Pancreatic Cancer Cell Lines

Barbara Borowa-Mazgaj; Anna Skwarska; Ewa Augustin; Anna Radominska-Pandya; Zofia Mazerska

Despite the enormous progress that has been made over the past decades in diagnosis, treatment and prevention of many types of tumors, survival rates in pancreatic cancer still remain poor. Pancreatic cancer is one of the most malignant and chemoresistant tumors and the profound mechanism supporting these phenomena is the constitutively activated prosurvival autophagy. The antitumor 1-nitroacridine derivative C-1748 belongs to a new set of DNA-damaging agents developed in our laboratory and is currently ready for phase I clinical trials. Here, we have uncovered a new role for C-1748 as a potent inhibitor of autophagy in four tested pancreatic cancer cell lines: Panc-1, MiaPaCa-2, BxPC-3 and AsPC-1. C-1748 treatment at 0.1 – 1 μM significantly reduced the typical hallmarks of autophagy, such as conversion of soluble LC3-I protein to its lipid-bound LC3-II isoform or development of acidic vesicular organelles, and importantly shifted pancreatic cancer cells into apoptotic cell death. Co-treatment with C-1748 and wortmannin, had a synergistic effect on autophagy inhibition and potentiated C-1748-induced apoptosis. In particular, compared with exposure to C-1748 alone, the combination treatment increased time- and dose-dependent cytotoxic activity of C-1748 with the most profound effect observed in BxPC-3 cells. Moreover, along with decrease in autophagy, co-treatment with wortmannin significantly elevated dysfunction of mitochondria, as manifested by a drop in mitochondrial membrane potential, increased caspase-3 activation, PARP cleavage, DNA fragmentation into apoptotic bodies and accumulation of lipid droplets arising due to the interruption of autophagy. In all studied cell lines, the level of apoptotic cells after dual exposure with C-1748 and wortmannin increased by about 20% as compared to C-1748 alone. Surprisingly, simultaneous exposure of pancreatic cancer cells to C-1748 in combination with the late-stage autophagy inhibitor, chloroquine, neither significantly increased the cytotoxic activity of C-1748 nor ameliorated its apoptosis-inducing potential. The synergistic effect of C-1748 and wortmannin on the inhibition of autophagy and lack of such effect when C-1748 was co-administered with chlroquine, suggests that C-1748, in addition to its DNA-damaging properties may also affects early stages of autophagy.That bespoke subtitle is 10/278ths of Abraham Lincoln’s iconic words at Gettysburg, borrowedhere to say what is so right about The FASEB Journal. This is a publication that serves a community and has always endeavored to do so with an open mind and a spirit of egalitarianism. We seek to publish engagingwork fromawide collective of scholars, with emphasis on the cogency of the work and leaving its “significance” for the sages of the future. To paraphrase an ancient physicians’ oath, I have been entrusted with the care of this journal. I take the helm with both excitement and humility. My predecessor, Gerald Weissmann (known to everyone as Gerry) is someone so engaging that I have never looked down in his presence to see whether his shoe size is 9, 10, or 11—all I know is that his are, with regard to this journal, gigantic shoes to fill. He brought to the journal exquisite scientific taste in the handling of papers and also provided extraordinary intellectual breadth in his engaging editorials, often illustrating his erudition in the allied domains of belles lettres and art, in the latter benefitting from the elegant contributions of Ann Weissmann each month in a cover selection and nexus to an article within, done by them in joyful communion. I thank Ann for her important role in these striking covers and am delighted that Gerry has accepted my invitation to serve as our Book Review editor going forward, succeeding me and adding his consummate bibliophilic judgment to thus enrich our Up Front pages. The FASEB Journal and FASEB owe him an enormous debt. WHAT WILL BE NEW AND WHAT SHALL ENDURE


Biochemical Pharmacology | 2003

Molecular mechanism of the enzymatic oxidation investigated for imidazoacridinone antitumor drug, C-1311.

Zofia Mazerska; Paweł Sowiński; Jerzy Konopa

The imidazoacridinone derivative, C-1311, is an antitumor agent that has been under phase I of clinical trial. The work presented here aims to elucidate the molecular mechanism of the enzymatic oxidative activation of this drug in such a model metabolic system, where the covalent binding to DNA was previously demonstrated. The oxidative activation of C-1311 was performed with HRP/H(2)O(2) and MPO/H(2)O(2) systems. The obtained final products of such transformations were separated and analysed by HPLC. The structures of the products were identified by means of ESI-MS and NMR. It was demonstrated that C-1311 was oxidised with HRP and MPO in the manner dependent on the drug:H(2)O(2) ratio and the drug was more susceptible to HRP oxidation than to MPO. Structural studies showed compounds C0 and C1 to be the result of dealkylation, which occurred in the amino groups of the side chain. The structures of C3 and C4 products were identified as dimers, whose monomers held the imidazoacridinone core. The activation of the imidazoacridinone ring system in position ortho to 8-hydroxyl group was necessary to form such dimers. We suggest that similar mechanism of C-1311 activation should occur in the presence of DNA when, instead of the dimer formation, the covalent binding to DNA, showed earlier for this drug, was formed. Since peroxidase-type enzymes are present in the cell nucleus of tumour cells the activation mechanisms of the C-1311 proposed here may be expected to take place in the cellular environment in vivo.


Drug Metabolism and Disposition | 2011

The imidazoacridinone antitumor drug, C-1311, is metabolized by flavin monooxygenases but not by cytochrome P450s.

Agnieszka Potęga; Dabrowska E; Niemira M; Kot-Wasik A; Sebastien Ronseaux; Colin J. Henderson; Charles Roland Wolf; Zofia Mazerska

5-Diethylaminoethylamino-8-hydroxyimidazoacridinone (C-1311) is an antitumor agent that is also active against autoimmune diseases. The intention of the present studies was to elucidate the role of selected liver enzymes in metabolism of C-1311 and the less active 8-methyl derivative, 5-diethylaminoethylamino-8-methoxyimidazoacridinone (C-1330). Compounds were incubated with rat liver microsomal fraction, with a set of 16 human liver protein samples, and with human recombinant isoenzymes of cytochrome P450, flavin monooxygenases (FMO), and UDP-glucuronosyltransferase (UGT). Our results showed that C-1311 and C-1330 were metabolized with human liver microsomal enzymes but not with any tested human recombinant cytochromes P450 (P450s). Two of these, CYP1A2 and CYP3A4, were inhibited by both compounds. In addition, results of C-1311 elimination from hepatic reductase-null mice, in which liver NADPH-P450 oxidoreductase has been deleted indicated that liver P450s were slightly engaged in drug transformation. In contrast, both compounds were good substrates for human recombinant FMO1 and FMO3 but not for FMO5. The product of FMO metabolism, PFMO, which is identified as an N-oxide derivative, was identical to P3R of liver microsomes. P3R was observed even in the presence of the P450 inhibitor, 1-aminobenzotriazole, and it disappeared after heating. Therefore, FMO enzymes could be responsible for microsomal metabolism to P3R = PFMO. Glucuronidation on the 8-hydroxyl group of C-1311 was observed with liver microsomes supported by UDP-glucuronic acid and with recombinant UGT1A1, but it was not the case with UGT2B7. Summing up, we showed that, whereas liver P450 isoenzymes were involved in the metabolism of C-1311 to a limited extent, FMO plays a significant role in the microsomal transformations of this compound, which is also a specific substrate of UGT1A1.


Critical Reviews in Analytical Chemistry | 2015

Revision of Biological Methods for Determination of EDC Presence and Their Endocrine Potential

Błażej Kudłak; Natalia Szczepańska; Katarzyna Owczarek; Zofia Mazerska; Jacek Namieśnik

Endocrine-disrupting compounds (EDC) are chemicals responsible for disturbances in the hormonal balance of organisms. This group of chemicals includes both egzogenic and endogenic substances or their mixtures that impact functioning of natural hormones in organisms. In the available literature one can find information on the application of chromatographic and related techniques in the analysis of environmental samples for detection, identification, and quantitation of a wide spectrum of chemicals posing endocrine properties. On the other hand, more and more biotests are being developed to determine endocrine potency of environmental samples due to development of genetic engineering methods and specific detection methods of cells’ response to the action of particular chemicals of interest. This article presents revisions of the most novel methods for this potency determination with application to biological elements.


Drug Metabolism and Disposition | 2012

Role of Human UDP-Glucuronosyltransferases in the Biotransformation of the Triazoloacridinone and Imidazoacridinone Antitumor Agents C-1305 and C-1311: Highly Selective Substrates for UGT1A10

Barbara Fedejko-Kap; Stacie M. Bratton; Moshe Finel; Anna Radominska-Pandya; Zofia Mazerska

5-Diethylaminoethylamino-8-hydroxyimidazoacridinone, C-1311 (NSC-645809), is an antitumor agent shown to be effective against breast cancer in phase II clinical trials. A similar compound, 5-dimethylaminopropylamino-8-hydroxytriazoloacridinone, C-1305, shows high activity against experimental tumors and is expected to have even more beneficial pharmacological properties than C-1311. Previously published studies showed that these compounds are not substrates for cytochrome P450s; however, they do contain functional groups that are common targets for glucuronidation. Therefore, the aim of this work was to identify the human UDP-glucuronosyltransferases (UGTs) able to glucuronidate these two compounds. High-performance liquid chromatography analysis was used to examine the activities of human recombinant UGT1A and UGT2B isoforms and microsomes from human liver [human liver microsomes (HLM)], whole human intestinal mucosa [human intestinal microsomes (HIM)], and seven isolated segments of human gastrointestinal tract. Recombinant extrahepatic UGT1A10 glucuronidated 8-hydroxyl groups with the highest catalytic efficiency compared with other recombinant UGTs, Vmax/Km = 27.2 and 8.8 μl · min−1 · mg protein−1, for C-1305 and C-1311, respectively. In human hepatic and intestinal microsomes (HLM and HIM, respectively), high variability in UGT activities was observed among donors and for different regions of intestinal tract. However, both compounds underwent UGT-mediated metabolism to 8-O-glucuronides by microsomes from both sources with comparable efficiency; Vmax/Km values were from 4.0 to 5.5 μl · min−1 · mg protein−1. In summary, these studies suggest that imid azoacridinone and triazoloacridinone drugs are glucuronidated in human liver and intestine in vivo and may form the basis for future translational studies of the potential role of UGTs in resistance to these drugs.


Xenobiotica | 2011

Flavin monooxygenases, FMO1 and FMO3, not cytochrome P450 isoenzymes, contribute to metabolism of anti-tumour triazoloacridinone, C-1305, in liver microsomes and HepG2 cells

Barbara Fedejko-Kap; Magdalena Niemira; Anna Radominska-Pandya; Zofia Mazerska

5-Dimethylaminopropylamino-8-hydroxytriazoloacridinone, C-1305, being the close structural analogue of the clinically tested imidazoacridinone anti-tumour agent, C-1311, expressed high activity against experimental tumours and is expected to have more advantageous pharmacological properties than C-1311. The aim of this study was to elucidate the role of selected liver enzymes in the metabolism of C-1305. We demonstrated that the studied triazoloacridinone was transformed with rat and human liver microsomes, HepG2 hepatoma cells and with human recombinant flavin-containing monooxygenases FMO1, FMO3 but not with CYPs. Furthermore, this compound was an effective inhibitor of CYP1A2 and CYP3A4. The product of FMO catalysed metabolism was shown to be identical to the main metabolite from liver microsomes and HepG2 cells. It was identified as an N-oxide derivative and, under hypoxia, it underwent retroreduction back to C-1305, what was extremely effective with participation of CYP3A4. In summary, this work revealed that the involvement of the P450 enzymatic system in microsomal and cellular metabolism of C-1305 was negligible, whereas this agent was an inhibitor of CYP1A2 and CYP3A4. In contrast, FMO1 and FMO3 were crucial for metabolism of C-1305 by liver microsomes and in HepG2 cells, which makes C-1305 an attractive potent anti-tumour agent.


Toxicology in Vitro | 2016

Improved cytotoxicity and preserved level of cell death induced in colon cancer cells by doxorubicin after its conjugation with iron-oxide magnetic nanoparticles

Ewa Augustin; Bartłomiej Czubek; Anna M. Nowicka; Agata Kowalczyk; Zbigniew Stojek; Zofia Mazerska

A promising strategy for overcoming the problem of limited efficacy in antitumor drug delivery and in drug release is the use of a nanoparticle-conjugated drug. Doxorubicin (Dox) anticancer chemotherapeutics has been widely studied in this respect, because of severe cardiotoxic side effects. Here, we investigated the cytotoxic effects, the uptake process, the changes in cell cycle progression and the cell death processes in the presence of iron-oxide magnetic nanoparticles (Nps) and doxorubicin conjugates (Dox-Nps) in human colon HT29 cells. The amount of Dox participated in biological action of Dox-Nps was determined by cyclic voltammetry and thermogravimetric measurements. The cytotoxicity of Dox-Nps was shown to be two/three times higher than free Dox, whereas Nps alone did not inhibit cell proliferation. Dox-Nps penetrated cancer cells with higher efficacy than free Dox, what could be a consequence of Dox-Nps aggregation with proteins in culture medium and/or with cell surface. The treatment of HT29 cells with Dox-Nps and Dox at IC50 concentration resulted in G2/M arrest followed by late apoptosis and necrosis. Summing up, the application of iron-oxide magnetic nanoparticles improved Dox-Nps cell penetration compared to free Dox and achieved the cellular response to Dox-Nps conjugates similar to that of Dox alone.


Chemical Research in Toxicology | 2014

Novel Resveratrol-Based Substrates for Human Hepatic, Renal, and Intestinal UDP-Glucuronosyltransferases

Aleksandra K. Greer; Nikhil Reddy Madadi; Stacie M. Bratton; Sarah Eddy; Zofia Mazerska; Howard P. Hendrickson; Peter A. Crooks; Anna Radominska-Pandya

Trans-Resveratrol (tRes) has been shown to have powerful antioxidant, anti-inflammatory, anticarcinogenic, and antiaging properties; however, its use as a therapeutic agent is limited by its rapid metabolism into its conjugated forms by UDP-glucuronosyltransferases (UGTs). The aim of the current study was to test the hypothesis that the limited bioavailability of tRes can be improved by modifying its structure to create analogs which would be glucuronidated at a lower rate than tRes itself. In this work, three synthetic stilbenoids, (E)-3-(3-hydroxy-4-methoxyphenyl)-2-(3,4,5-trimethoxyphenyl)acrylic acid (NI-12a), (E)-2,4-dimethoxy-6-(4-methoxystyryl)benzaldehyde oxime (NI-ST-05), and (E)-4-(3,5-dimethoxystyryl)-2,6-dinitrophenol (DNR-1), have been designed based on the structure of tRes and synthesized in our laboratory. UGTs recognize and glucuronidate tRes at each of the 3 hydroxyl groups attached to its aromatic rings. Therefore, each of the above compounds was designed with the majority of the hydroxyl groups blocked by methylation and the addition of other novel functional groups as part of a drug optimization program. The activities of recombinant human UGTs from the 1A and 2B families were examined for their capacity to metabolize these compounds. Glucuronide formation was identified using HPLC and verified by β-glucuronidase hydrolysis and LC–MS/MS analysis. NI-12a was glucuronidated at both the −COOH and −OH functions, NI-ST-05 formed a novel N–O-glucuronide, and no product was observed for DNR-1. NI-12a is primarily metabolized by the hepatic and renal enzyme UGT1A9, whereas NI-ST-05 is primarily metabolized by an extrahepatic enzyme, UGT1A10, with apparent Km values of 240 and 6.2 μM, respectively. The involvement of hepatic and intestinal UGTs in the metabolism of both compounds was further confirmed using a panel of human liver and intestinal microsomes, and high individual variation in activity was demonstrated between donors. In summary, these studies clearly establish that modified, tRes-based stilbenoids may be preferable alternatives to tRes itself due to increased bioavailability via altered conjugation.


Drug Metabolism and Disposition | 2013

Metabolic Transformation of Antitumor Acridinone C-1305 but Not C-1311 via Selective Cellular Expression of UGT1A10 Increases Cytotoxic Response: Implications for Clinical Use

Monika Pawłowska; Rong Chu; Barbara Fedejko-Kap; Ewa Augustin; Zofia Mazerska; Anna Radominska-Pandya; Timothy C. Chambers

The acridinone derivates 5-dimethylaminopropylamino-8-hydroxytriazoloacridinone (C-1305) and 5-diethylaminoethylamino-8-hydroxyimidazoacridinone (C-1311) are promising antitumor agents with high activity against several experimental cellular and tumor models and are under evaluation in preclinical and early phase clinical trials. Recent evidence from our laboratories has indicated that both compounds were conjugated by several uridine diphosphate-glucuronyltransferase (UGT) isoforms, the most active being extrahepatic UGT1A10. The present studies were designed to test the ability and selectivity of UGT1A10 in the glucuronidation of acridinone antitumor agents in a cellular context. We show that in KB-3 cells, a HeLa subline lacking expression of any UGT isoforms, both C-1305 and C-1311 undergo metabolic transformation to the glucuronidated forms on overexpression of UGT1A10. Furthermore, UGT1A10 overexpression significantly increased the cytotoxicity of C-1305, but not C-1311, suggesting that the glucuronide was more potent than the C-1305 parent compound. These responses were selective for UGT1A10 because documented overexpression of UGT2B4 failed to produce glucuronide products and failed to alter the cytotoxicity for both compounds. These findings contribute to our understanding of the mechanisms of action of these agents and are of particular significance because data for C-1305 contradict the dogma that glucuronidation typically plays a role in detoxification or deactivation. In summary, these studies suggest that extrahepatic UGT1A10 plays an important role in the metabolism and the bioactivation of C-1305 and constitutes the basis for further mechanistic studies on the mode of action of this drug, as well as translational studies on the role of this enzyme in regulation of C-1305 toxicity in cancer.

Collaboration


Dive into the Zofia Mazerska's collaboration.

Top Co-Authors

Avatar

Ewa Augustin

Gdańsk University of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Agnieszka Potęga

Gdańsk University of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anna Radominska-Pandya

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Anita Wiśniewska

Gdańsk University of Technology

View shared research outputs
Top Co-Authors

Avatar

Anna Mróz

Gdańsk University of Technology

View shared research outputs
Top Co-Authors

Avatar

Magdalena Niemira

Gdańsk University of Technology

View shared research outputs
Top Co-Authors

Avatar

Monika Pawłowska

Gdańsk University of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge