Małgorzata Opydo-Chanek
Jagiellonian University
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Małgorzata Opydo-Chanek.
Anti-Cancer Drugs | 2011
Lidia Mazur; Małgorzata Opydo-Chanek; Marta Stojak
Glufosfamide (&bgr;-D-glucose-isophosphoramide mustard, D-19575) belongs to the oxazaphosphorine class. Glufosfamide is a novel glucose conjugate of ifosfamide in which isophosphoramide mustard, the alkylating metabolite of ifosfamide, is glycosidically linked to the &bgr;-D-glucose molecule. Glufosfamide represents an attractive new agent for cancer therapy. Its mode of action on normal and pathological cells is still under experimental and clinical investigations. An assessment of the anticancer potential of glufosfamide is of key importance in therapy. The researchers reviewed the current knowledge available on glufosfamide tested in the preclinical studies/clinical trials, based on a collection of the original papers and conference abstracts published and relevant articles searched in the SCOPUS and MEDLINE database and websites.
Neuroscience Letters | 2011
Małgorzata Opydo-Chanek; Zbigniew Dąbrowski
The aim of the present study was to investigate the effects of intracerebral and intravenous administration of bone marrow stromal cells (BMSCs) on cellular activity in the injured brain. Female Wistar rats were subjected to cerebral cortex injury followed by the injection of BMSCs or saline, directly to the injured site or to the tail vein. Lectin histochemistry and glial fibrillary acidic protein immunohistochemistry were used to analyze the number of microglia/macrophages and astrocytes in the injured cerebral cortex, respectively. BMSC treatment affected cell response to brain injury. The effects of BMSC action were dependent on the site of their administration. The intravenous injection of BMSCs noticeably increased the number of microglia/macrophages in the injured brain. Intracerebral transplantation of BMSCs significantly enhanced the number of astrocytes, and in a less degree caused changes in the number of microglia/macrophages. The results suggest that BMSCs can affect the restorative processes in the injured brain through stimulation of the cellular response to injury.
Biochemical Pharmacology | 2017
Małgorzata Opydo-Chanek; Oscar Gonzalo; Isabel Marzo
BH3 mimetics are a novel class of anticancer agents designed to specifically target pro-survival proteins of the Bcl-2 family. Like endogenous BH3-only proteins, BH3 mimetics competitively bind to surface hydrophobic grooves of pro-survival Bcl-2 family members, counteracting their protective effects and thus facilitating apoptosis in cancer cells. Among the small-molecule BH3 mimetics identified, ABT-737 and its analogs, obatoclax as well as gossypol derivatives are the best characterized. The anticancer potential of these compounds applied as a single agent or in combination with chemotherapeutic drugs is currently being evaluated in preclinical studies and in clinical trials. In spite of promising results, the actual mechanisms of their anticancer action remain to be identified. Findings from preclinical studies point to additional activities of BH3 mimetics in cancer cells that are not connected with apoptosis induction. These off-target effects involve induction of autophagy and necrotic cell death as well as modulation of the cell cycle and multiple cell signaling pathways. For the optimization and clinical implementation of BH3 mimetics, a detailed understanding of their role as inhibitors of the pro-survival Bcl-2 proteins, but also of their possible additional effects is required. This review summarizes the most representative BH3 mimetic compounds with emphasis on their off-target effects. Based on the present knowledge on the multifaceted effects of BH3 mimetics on cancer cells, the commentary outlines the potential pitfalls and highlights the considerable promise for cancer treatment with BH3 mimetics.
Tissue & Cell | 2018
Małgorzata Kotula-Balak; Agnieszka Milon; Piotr Pawlicki; Małgorzata Opydo-Chanek; Anna Pacwa; Klaudia Lesniak; Małgorzata Sekuła; Marta Zarzycka; Monika Bubka; Waclaw Tworzydlo; Barbara Bilińska; Anna Hejmej
In this study, we demonstrate, for the first time, estrogen-related receptor (ERR) regulation of the physiological and biochemical status of testicular tumor Leydig cells. In a mouse tumor Leydig cells, ERRs (α, β, and γ) were silenced via siRNA. Cell morphology and cell physiology (proliferation and observation of monolayer formation) were performed by inverted phase-contrast microscope. Leydig cell functional markers (steroid receptors and signaling molecules) were examined by immunofluorescence and Western blotting. Additionally, progesterone secretion was assessed. Mitochondrial mass and membrane potential were analyzed by flow-cytometry while cGMP and Ca2+ concentrations were analyzed using immunoenzymatic and colorimetric assays, respectively. These results revealed, ERRs indirectly regulate Leydig cell proliferation while ERRα and β affect cell monolayer formation. ERRs interact with canonical and membrane estrogen receptors (ERα, ERβ, and GPER), androgen receptor, metalloproteinase (MMP 9), protein kinase A (PKA), extracellular-regulated kinase (ERK), and neurogenic locus notch homolog protein 2 (Notch2). Depending on the type of ERR knocked down, coupled with estradiol treatment, changes in progesterone concentration and cGMP and Ca2+ concentrations constitute a microenvironment that may effect tumor Leydig cell characteristics. ERRs should be considered important factors in developing of innovating approaches that target pathological processes of testicular Leydig cells.
Toxicology in Vitro | 2017
Małgorzata Opydo-Chanek; Agnieszka Rak; Agnieszka Cierniak; Lidia Mazur
ABT-737 belongs to a new class of anticancer agents named BH3 mimetics. ABT-737 competitively binds to surface hydrophobic grooves of anti-apoptotic proteins of Bcl-2 family, counteracting their protective effect. Resveratrol is a natural polyphenol that has been shown to inhibit the proliferation and/or induce apoptosis in a number of different types of cancer cells. The present study was designed to analyze the combined effects of ABT-737 and resveratrol on human acute lymphoblastic leukemia cells. The in vitro cytotoxic activity of these agents against MOLT-4 leukemia cells was determined using the Coulter electrical impedance method, comet assay, and flow cytometry, light microscopy and western blot techniques. The results are the first data showing that ABT-737 combined with resveratrol markedly decreased the cell viability, increased DNA damage, caused the cell cycle perturbation, and synergistically enhanced apoptosis in MOLT-4 cells, when compared to the data obtained after application of the single agent. Moreover, the simultaneous treatment of leukemia cells with ABT-737 and resveratrol resulted in a reduction in mitochondrial membrane potential, an increase of p53 protein level and up-regulation of the Bax/Bcl-2 ratio. The obtained data indicate that the combination of ABT-737 and resveratrol is a promising approach for acute lymphoblastic leukemia treatment that should be further explored.
Cell and Tissue Research | 2017
Agnieszka Milon; Małgorzata Opydo-Chanek; Waclaw Tworzydlo; Jerzy Galas; Laura Pardyak; Alicja Kamińska; Anna Ptak; Małgorzata Kotula-Balak
Folia Biologica | 2013
Lidia Mazur; Małgorzata Opydo-Chanek; Marta Stojak; Ulf Niemeyer
Folia Biologica | 2009
Lidia Mazur; Małgorzata Opydo-Chanek; Marta Stojak; Jaroslaw Baran; Ulf Niemeyer
Indian Journal of Experimental Biology | 2013
Małgorzata Opydo-Chanek; Lidia Mazur; Marta Stojak
Acta Biologica Cracoviensia. Series Botanica. Supplement | 2009
Lidia Mazur; Marta Stojak; Małgorzata Opydo-Chanek; Ulf Niemeyer