Network


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

Hotspot


Dive into the research topics where Aleksandra Glogowska is active.

Publication


Featured researches published by Aleksandra Glogowska.


Journal of Materials Chemistry B | 2013

Polymeric mesoporous silica nanoparticles as a pH-responsive switch to control doxorubicin intracellular delivery

Ye Tian; Aleksandra Glogowska; Wen Zhong; Thomas Klonisch; Malcolm Xing

Cancer is a leading cause of death. Mesoporous nanomaterials with stimuli sensitivity have received increasing interest as efficient anti-cancer drug carriers. Here, we report hybrid mesoporous nanoparticles of PEGylated silica-poly[2-(dimethylamino)ethyl acrylate] (PEGylated MSN-g-PDMAEA) that can deliver and release the anti-cancer drug doxorubicin (DOX) to tumor cells in a pH dependent switch on/off status. The reversible pH sensitivity resulted in nanoparticles with enhanced functionality through controllable release of the anticancer drug. Confocal laser scanning microscopy (CLSM) and three dimensional (3D) image capture of the intracellular localization of nanoparticles revealed the fast and efficient drug delivery in the human hepatocellular carcinoma cell line. The results highlight the potential of these pH sensitive silica nanoparticles as a novel system for the delivery and controlled release of hydrophobic anti-cancer drugs in the treatment of solid tumors.


Journal of Alzheimer's Disease | 2014

Three-Dimensional Quantitative Imaging of Telomeres in Buccal Cells Identifies Mild, Moderate, and Severe Alzheimer's Disease Patients

Shubha Mathur; Aleksandra Glogowska; Elizabeth McAvoy; Christiaan H. Righolt; Jaclyn Rutherford; Cornelia Willing; Upama Banik; Myuri Ruthirakuhan; Sabine Mai; Angeles Garcia

Using three-dimensional (3D) telomeric analysis of buccal cells of 82 Alzheimers disease (AD) patients and cognitively normal age and gender-matched controls, we have for the first time examined changes in the 3D nuclear telomeric architecture of buccal cells among levels of AD severity based on five 3D parameters: i) telomere length, ii) telomere number, iii) telomere aggregation, iv) nuclear volume, and v) a/c ratio, a measure of spatial telomere distribution. Our data indicate that matched controls have significantly different 3D telomere profiles compared to mild, moderate, and severe AD patients (p < 0.0001). Distinct profiles were also evident for each AD severity group. An increase in telomere number and aggregation concomitant with a decrease in telomere length from normal to severe AD defines the individual stages of the disease (p < 0.0001).


The Journal of Pathology | 2013

C1q-tumour necrosis factor-related protein 8 (CTRP8) is a novel interaction partner of relaxin receptor RXFP1 in human brain cancer cells

Aleksandra Glogowska; Usakorn Kunanuvat; Jörg Stetefeld; Trushar R. Patel; Thatchawan Thanasupawat; Jerry Krcek; Ekkehard Weber; G William Wong; Marc R. Del Bigio; Cuong Hoang-Vu; Sabine Hombach-Klonisch; Thomas Klonisch

We report a novel ligand–receptor system composed of the leucine‐rich G‐protein‐coupled relaxin receptor, RXFP1, and the C1q‐tumour necrosis factor‐related protein 8 (CTRP8) in human primary brain cancer, a tumour entity devoid of the classical RXFP1 ligands, RLN1–3. In structural homology studies and computational docking experiments we delineated the N‐terminal region of the globular C1q region of CTRP8 and the leucine‐rich repeat units 7 and 8 of RXFP1 to mediate this new ligand–receptor interaction. CTRP8 secreted from HEK293T cells, recombinant human (rh) CTRP8, and short synthetic peptides derived from the C1q globular domain of human CTRP8 caused the activation of RXFP1 as determined by elevated intracellular cAMP levels and the induction of a marked pro‐migratory phenotype in established glioblastoma (GB) cell lines and primary cells from GB patients. Employing a small competitor peptide, we were able to disrupt the CTRP8–RXFP1‐induced increased GB motility. The CTRP8–RXFP1‐mediated migration in GB cells involves the activation of PI3K and specific protein kinase C pathways and the increased production/secretion of the potent lysosomal protease cathepsin B (cathB), a known prognostic marker of GB. Specific inhibition of CTRP8‐induced cathB activity effectively blocked the ability of primary GB to invade laminin matrices. Finally, co‐immunoprecipitation studies revealed the direct interaction of human CTRP8 with RXFP1. Our results support a therapeutic approach in GB aimed at targeting multiple steps of the CTRP8–RXFP1 signalling pathway by a combined inhibitor and peptide‐based strategy to block GB dissemination within the brain. Copyright


Scientific Reports | 2017

Mevalonate Cascade Inhibition by Simvastatin Induces the Intrinsic Apoptosis Pathway via Depletion of Isoprenoids in Tumor Cells

Javad Alizadeh; Amir A. Zeki; Nima Mirzaei; Sandipan Tewary; Adel Rezaei Moghadam; Aleksandra Glogowska; Pandian Nagakannan; Eftekhar Eftekharpour; Emilia Wiechec; Joseph W. Gordon; Fred Y. Xu; Jared T. Field; Ken Y. Yoneda; Nicholas J. Kenyon; Mohammad Hashemi; Grant M. Hatch; Sabine Hombach-Klonisch; Thomas Klonisch; Saeid Ghavami

The mevalonate (MEV) cascade is responsible for cholesterol biosynthesis and the formation of the intermediate metabolites geranylgeranylpyrophosphate (GGPP) and farnesylpyrophosphate (FPP) used in the prenylation of proteins. Here we show that the MEV cascade inhibitor simvastatin induced significant cell death in a wide range of human tumor cell lines, including glioblastoma, astrocytoma, neuroblastoma, lung adenocarcinoma, and breast cancer. Simvastatin induced apoptotic cell death via the intrinsic apoptotic pathway. In all cancer cell types tested, simvastatin-induced cell death was not rescued by cholesterol, but was dependent on GGPP- and FPP-depletion. We confirmed that simvastatin caused the translocation of the small Rho GTPases RhoA, Cdc42, and Rac1/2/3 from cell membranes to the cytosol in U251 (glioblastoma), A549 (lung adenocarcinoma) and MDA-MB-231(breast cancer). Simvastatin-induced Rho-GTP loading significantly increased in U251 cells which were reversed with MEV, FPP, GGPP. In contrast, simvastatin did not change Rho-GTP loading in A549 and MDA-MB-231. Inhibition of geranylgeranyltransferase I by GGTi-298, but not farnesyltransferase by FTi-277, induced significant cell death in U251, A549, and MDA-MB-231. These results indicate that MEV cascade inhibition by simvastatin induced the intrinsic apoptosis pathway via inhibition of Rho family prenylation and depletion of GGPP, in a variety of different human cancer cell lines.


Cancer Research | 2005

Cytoplasmic Domain of proEGF Affects Distribution and Post-Translational Modification of Microtubuli and Increases Microtubule-Associated Proteins 1b and 2 Production in Human Thyroid Carcinoma Cells

Janette Pyka; Aleksandra Glogowska; Henning Dralle; Cuong Hoang-Vu; Thomas Klonisch

We studied the distribution of transcripts encoding the cytoplasmic domain of the membrane-anchored precursor epidermal growth factor (proEGFcyt) and a novel cytoplasmic proEGF splice isoform with a deleted exon 23 and an out-of-frame fusion of exon 24 (proEGFdel23) in human normal and neoplastic thyroid tissues. In papillary thyroid carcinoma (PTC), coexpression of transcripts encoding for both proEGFcyt and proEGFdel23 correlated with poor differentiation of PTC. To determine potential roles of the cytoplasmic proEGF domain in human thyroid cells, we generated stable transfectants of the human follicular thyroid carcinoma cell line FTC-133 overexpressing the normal cytoplasmic domain proEGFcyt, a truncated proEGFcyt composed of the peptide sequence encoded by exons 22 and 23 (proEGF22.23) and proEGFdel23. The proEGFcyt and proEGF22.23 transfectants displayed significantly reduced proliferation rates, an enlarged cellular phenotype, and alterations in the distribution and post-translational modification of the microtubular system. These transfectants also displayed increased production of microtubule-associated proteins 1b and 2c, which was absent in FTC-133-proEGFdel23 or FTC-133-empty plasmid transfectants. This is the first evidence of an involvement of proEGF cytoplasmic domain in microtubular stability in the human thyroid carcinoma cell line FTC-133 and may suggest a specific role for the cytoplasmic domain of membrane-anchored proEGF, particularly exon 23, in thyroid carcinoma. The up-regulation of proEGFdel23 in poorly differentiated PTC and the exclusive detection of both proEGF isoforms in undifferentiated thyroid carcinoma may indicate an involvement of this novel truncated proEGFdel23 cytoplasmic domain during dedifferentiation processes of human thyroid cells.


FEBS Letters | 2009

The C-terminal cytoplasmic domain of human proEGF is a negative modulator of body and organ weights in transgenic mice

Thomas Klonisch; Aleksandra Glogowska; Ana A. Gratao; Marjeta Grzech; Andreea Nistor; Mark G. Torchia; Ekkehard Weber; Martin Hrabé de Angelis; Birgit Rathkolb; Cuong Hoang-Vu; Eckhard Wolf; Marlon R. Schneider

We generated transgenic mice to study the in vivo role of the cytoplasmic domain of human proEGF (proEGFcyt). Post‐pubertal proEGFcyt transgenic (tg) mice displayed an up to 15% reduction in body weight, including smaller kidney and brain weights as compared to control littermates. Renal histology, gene expression profiles, and functional parameters were normal. In both sexes, serum levels of IGFBP‐3 were reduced. Circulating IGF‐I/IGF‐II levels were unchanged. Histomorphological analysis revealed isolated foci of liver necrosis specific to proEGFcyt tg mice. In conclusion, we identified proEGF cytoplasmic domain as a novel modulator of whole body and organ‐specific growth in mice.


Biochimica et Biophysica Acta | 2018

Autophagy modulates transforming growth factor beta 1 induced epithelial to mesenchymal transition in non-small cell lung cancer cells

Javad Alizadeh; Aleksandra Glogowska; James A. Thliveris; Forouh Kalantari; Shahla Shojaei; Sabine Hombach-Klonisch; Thomas Klonisch; Saeid Ghavami

Lung cancer is considered one of the most frequent causes of cancer-related death worldwide and Non-Small Cell Lung Cancer (NSCLC) accounts for 80% of all lung cancer cases. Autophagy is a cellular process responsible for the recycling of damaged organelles and protein aggregates. Transforming growth factor beta-1 (TGFβ1) is involved in Epithelial to Mesenchymal Transition (EMT) and autophagy induction in different cancer models and plays an important role in the pathogenesis of NSCLC. It is not clear how autophagy can regulate EMT in NSCLC cells. In the present study, we have investigated the regulatory role of autophagy in EMT induction in NSCLC and show that TGFβ1 can simultaneously induce both autophagy and EMT in the NSCL lines A549 and H1975. Upon chemical inhibition of autophagy using Bafilomycin-A1, the expression of the mesenchymal marker vimentin and N-cadherin was reduced. Immunoblotting and immunocytochemistry (ICC) showed that the mesenchymal marker vimentin was significantly downregulated upon TGFβ1 treatment in ATG7 knockdown cells when compared to corresponding cells treated with scramble shRNA (negative control), while E-cadherin was unchanged. Furthermore, autophagy inhibition (Bafilomycin A1 and ATG7 knockdown) decreased two important mesenchymal functions, migration and contraction, of NSCLC cells upon TGFβ1 treatment. This study identified a crucial role of autophagy as a potential positive regulator of TGFβ1-induced EMT in NSCLC cells and identifies inhibitors of autophagy as promising new drugs in antagonizing the role of EMT inducers, like TGFβ1, in the clinical progression of NSCLC.


Molecular Oncology | 2017

Dovitinib enhances temozolomide efficacy in glioblastoma cells

Thatchawan Thanasupawat; Suchitra Natarajan; Amy Rommel; Aleksandra Glogowska; Hugo Bergen; Jerry Krcek; Marshall W. Pitz; Jason Beiko; Sherry Krawitz; Inder M. Verma; Saeid Ghavami; Thomas Klonisch; Sabine Hombach-Klonisch

The multikinase inhibitor and FDA‐approved drug dovitinib (Dov) crosses the blood–brain barrier and was recently used as single drug application in clinical trials for GB patients with recurrent disease. The Dov‐mediated molecular mechanisms in GB cells are unknown. We used GB patient cells and cell lines to show that Dov downregulated the stem cell protein Lin28 and its target high‐mobility group protein A2 (HMGA2). The Dov‐induced reduction in pSTAT3Tyr705 phosphorylation demonstrated that Dov negatively affects the STAT3/LIN28/Let‐7/HMGA2 regulatory axis in GB cells. Consistent with the known function of LIN28 and HMGA2 in GB self‐renewal, Dov reduced GB tumor sphere formation. Dov treatment also caused the downregulation of key base excision repair factors and O6‐methylguanine‐DNA‐methyltransferase (MGMT), which are known to have important roles in the repair of temozolomide (TMZ)‐induced alkylating DNA damage. Combined Dov/TMZ treatment enhanced TMZ‐induced DNA damage as quantified by nuclear γH2AX foci and comet assays, and increased GB cell apoptosis. Pretreatment of GB cells with Dov (‘Dov priming’) prior to TMZ treatment reduced GB cell viability independent of p53 status. Sequential treatment involving ‘Dov priming’ and alternating treatment cycles with TMZ and Dov substantially reduced long‐term GB cell survival in MGMT+ patient GB cells. Our results may have immediate clinical implications to improve TMZ response in patients with LIN28+/HMGA2+ GB, independent of their MGMT methylation status.


British Journal of Pharmacology | 2017

Structural commonality of C1q TNF‐related proteins and their potential to activate relaxin/insulin‐like family peptide receptor 1 signalling pathways in cancer cells

Thomas Klonisch; Aleksandra Glogowska; Thatchawan Thanasupawat; Maxwell Burg; Jerry Krcek; Marshall W. Pitz; Appalaraju Jaggupilli; Prashen Chelikani; G William Wong; Sabine Hombach-Klonisch

We established the role of the GPCR relaxin/insulin‐like family peptide receptor 1 (RXFP1 receptor) as a novel active receptor in human glioblastoma (GB), a fatal brain tumour. We identified C1q/TNF‐related protein 8 (CTRP8) as a novel agonist of the RXFP1 receptor. CTRP8 enhanced the motility and matrix invasion of GB, and this involved PKC‐mediated up‐regulation of cathepsin B, a marker for poor prognosis in GB patients. We conclude that the absence of relaxin isoforms does not preclude the activation of the RXFP1 receptor, as the least known member of the CTRP family, CTRP8, can effectively target and activate RXFP1 receptors.


British Journal of Pharmacology | 2016

Structural commonality of C1q Tumor Necrosis Factor‐related proteins and their potential to activate RXFP1 signaling pathways in cancer cells

Thomas Klonisch; Aleksandra Glogowska; Thatchawan Thanasupawat; Maxwell Burg; Jerry Krcek; Marshall W. Pitz; Appalaraju Jaggupilli; Prashen Chelikani; G William Wong; Sabine Hombach-Klonisch

We established the role of the GPCR relaxin/insulin‐like family peptide receptor 1 (RXFP1 receptor) as a novel active receptor in human glioblastoma (GB), a fatal brain tumour. We identified C1q/TNF‐related protein 8 (CTRP8) as a novel agonist of the RXFP1 receptor. CTRP8 enhanced the motility and matrix invasion of GB, and this involved PKC‐mediated up‐regulation of cathepsin B, a marker for poor prognosis in GB patients. We conclude that the absence of relaxin isoforms does not preclude the activation of the RXFP1 receptor, as the least known member of the CTRP family, CTRP8, can effectively target and activate RXFP1 receptors.

Collaboration


Dive into the Aleksandra Glogowska's collaboration.

Top Co-Authors

Avatar

Thomas Klonisch

Martin Luther University of Halle-Wittenberg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jerry Krcek

University of Manitoba

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

G William Wong

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Marek Los

Linköping University

View shared research outputs
Researchain Logo
Decentralizing Knowledge