Network


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

Hotspot


Dive into the research topics where Agnieszka Dansonka-Mieszkowska is active.

Publication


Featured researches published by Agnieszka Dansonka-Mieszkowska.


BMC Medical Genetics | 2010

A novel germline PALB2 deletion in Polish breast and ovarian cancer patients

Agnieszka Dansonka-Mieszkowska; Anna Kluska; Joanna Moes; Michalina Dabrowska; Dorota Nowakowska; Anna Niwińska; Pawel Derlatka; Krzysztof Cendrowski; Jolanta Kupryjanczyk

BackgroundPALB2 protein was recently identified as a partner of BRCA1 and BRCA2 which determines their proper function in DNA repair.MethodsInitially, the entire coding sequence of the PALB2 gene with exon/intron boundaries was evaluated by the PCR-SSCP and direct sequencing methods on 70 ovarian carcinomas. Sequence variants of interest were further studied on enlarged groups of ovarian carcinomas (total 339 non-consecutive ovarian carcinomas), blood samples from 334 consecutive sporadic and 648 consecutive familial breast cancer patients, and 1310 healthy controls from central Poland.ResultsTen types of sequence variants were detected, and among them four novel polymorphisms: c.2996+58T>C in intron 9; c.505C>A (p.L169I), c.618T>G (p.L206L), both in exon 4; and c.2135C>T (A712V) in exon 5 of the PALB2 gene. Another two polymorphisms, c.212-58A>C and c.2014G>C (E672Q) were always detected together, both in cancer (7.5% of patients) and control samples (4.9% of controls, p = 0.2). A novel germline truncating mutation, c.509_510delGA (p.R170fs) was found in exon 4: in 2 of 339 (0.6%) unrelated ovarian cancer patients, in 4 of 648 (0.6%) unrelated familial breast cancer patients, and in 1 of 1310 controls (0.08%, p = 0.1, p = 0.044, respectively). One ovarian cancer patient with the PALB2 mutation had also a germline nonsense mutation of the BRCA2 gene.ConclusionsThe c.509_510delGA is a novel PALB2 mutation that increases the risk of familial breast cancer. Occurrence of the same PALB2 alteration in seven unrelated women suggests that c.509_510delGA (p.R170fs) is a recurrent mutation for Polish population.


Cancer Biology & Therapy | 2009

PIK3CA amplification associates with resistance to chemotherapy in ovarian cancer patients.

Iwona Kolasa; Alina Rembiszewska; Anna Felisiak; Izabela Ziółkowska-Seta; Magdalena Murawska; Joanna Moes; Agnieszka Timorek; Agnieszka Dansonka-Mieszkowska; Jolanta Kupryjanczyk

PI3K/AKT signalling pathway controls important cellular processes such as the cell proliferation and apoptosis. PIK3CA gene encoding a catalytic subunit of the PI3K is mutated and/or amplified in various neoplasms, including ovarian cancer. We aimed to evaluate PIK3CA alterations and their clinical importance in ovarian cancer patients. Molecular analysis was performed on 117 ovarian carcinomas with the use of qPCR, SSCP and sequencing. In a group of 98 patients with complete clinical data, 62 patients were treated with standard taxane-platinum regimens and 36 patients with platinum-cyclophosphamide regimens. A multivariate analysis was performed by the Cox’s and logistic regression models. PIK3CA mutations occurred in 5/117 (4.3%) carcinomas, exclusively in the endometrioid and clear cell types (P=0.0002); they were also associated with low FIGO stage (P=0.0003), low tumor grade (P=0.045) and early patient’s age at diagnosis (P=0.0005). The PIK3CA amplification (predominantly a low-level) was found in 28/117 (24%) ovarian carcinomas. It was more frequent in TP53 mutant tumors (P=0.012) and tended to associate with high pAKT expression (P=0.061). The PIK3CA amplification strongly diminished odds of complete remission (OR=0.25, P=0.033) and platinum sensitive response (PS, OR=0.12, P=0.004) in the taxane-platinum treated patients. The odds of PS were also much lower in all patients with the PIK3CA amplification evaluated together, regardless of the treatment applied (OR=0.18, P=0.001). Our results suggest that PIK3CA amplification may be a marker predicting ovarian cancer response to chemotherapy.


Cancer Epidemiology, Biomarkers & Prevention | 2013

Analysis of Over 10,000 Cases Finds No Association between Previously Reported Candidate Polymorphisms and Ovarian Cancer Outcome

Kristin L. White; Robert A. Vierkant; Zachary C. Fogarty; Bridget Charbonneau; Matthew S. Block; Paul Pharoah; Georgia Chenevix-Trench; Mary Anne Rossing; Daniel W. Cramer; Celeste Leigh Pearce; Joellen M. Schildkraut; Usha Menon; Susanne K. Kjaer; Douglas A. Levine; Jacek Gronwald; Hoda Anton Culver; Alice S. Whittemore; Beth Y. Karlan; Diether Lambrechts; Nicolas Wentzensen; Jolanta Kupryjanczyk; Jenny Chang-Claude; Elisa V. Bandera; Estrid Høgdall; Florian Heitz; Stanley B. Kaye; Peter A. Fasching; Ian G. Campbell; Marc T. Goodman; Tanja Pejovic

Background: Ovarian cancer is a leading cause of cancer-related death among women. In an effort to understand contributors to disease outcome, we evaluated single-nucleotide polymorphisms (SNP) previously associated with ovarian cancer recurrence or survival, specifically in angiogenesis, inflammation, mitosis, and drug disposition genes. Methods: Twenty-seven SNPs in VHL, HGF, IL18, PRKACB, ABCB1, CYP2C8, ERCC2, and ERCC1 previously associated with ovarian cancer outcome were genotyped in 10,084 invasive cases from 28 studies from the Ovarian Cancer Association Consortium with over 37,000-observed person-years and 4,478 deaths. Cox proportional hazards models were used to examine the association between candidate SNPs and ovarian cancer recurrence or survival with and without adjustment for key covariates. Results: We observed no association between genotype and ovarian cancer recurrence or survival for any of the SNPs examined. Conclusions: These results refute prior associations between these SNPs and ovarian cancer outcome and underscore the importance of maximally powered genetic association studies. Impact: These variants should not be used in prognostic models. Alternate approaches to uncovering inherited prognostic factors, if they exist, are needed. Cancer Epidemiol Biomarkers Prev; 22(5); 987–. ©2013 AACR.


Clinical Cancer Research | 2015

Genome-wide Analysis Identifies Novel Loci Associated with Ovarian Cancer Outcomes: Findings from the Ovarian Cancer Association Consortium

Sharon E. Johnatty; Jonathan Tyrer; Siddhartha Kar; Jonathan Beesley; Yi Lu; Bo Gao; Peter A. Fasching; Alexander Hein; Arif B. Ekici; Matthias W. Beckmann; Diether Lambrechts; Els Van Nieuwenhuysen; Ignace Vergote; Sandrina Lambrechts; Mary Anne Rossing; Jennifer A. Doherty; Jenny Chang-Claude; Francesmary Modugno; Roberta B. Ness; Kirsten B. Moysich; Douglas A. Levine; Lambertus A. Kiemeney; Leon F.A.G. Massuger; Jacek Gronwald; Jan Lubinski; Louise A. Brinton; Jolanta Lissowska; Nicolas Wentzensen; Honglin Song; Valerie Rhenius

Purpose: Chemotherapy resistance remains a major challenge in the treatment of ovarian cancer. We hypothesize that germline polymorphisms might be associated with clinical outcome. Experimental Design: We analyzed approximately 2.8 million genotyped and imputed SNPs from the iCOGS experiment for progression-free survival (PFS) and overall survival (OS) in 2,901 European epithelial ovarian cancer (EOC) patients who underwent first-line treatment of cytoreductive surgery and chemotherapy regardless of regimen, and in a subset of 1,098 patients treated with ≥4 cycles of paclitaxel and carboplatin at standard doses. We evaluated the top SNPs in 4,434 EOC patients, including patients from The Cancer Genome Atlas. In addition, we conducted pathway analysis of all intragenic SNPs and tested their association with PFS and OS using gene set enrichment analysis. Results: Five SNPs were significantly associated (P ≤ 1.0 × 10−5) with poorer outcomes in at least one of the four analyses, three of which, rs4910232 (11p15.3), rs2549714 (16q23), and rs6674079 (1q22), were located in long noncoding RNAs (lncRNAs) RP11-179A10.1, RP11-314O13.1, and RP11-284F21.8, respectively (P ≤ 7.1 × 10−6). ENCODE ChIP-seq data at 1q22 for normal ovary show evidence of histone modification around RP11-284F21.8, and rs6674079 is perfectly correlated with another SNP within the super-enhancer MEF2D, expression levels of which were reportedly associated with prognosis in another solid tumor. YAP1- and WWTR1 (TAZ)-stimulated gene expression and high-density lipoprotein (HDL)-mediated lipid transport pathways were associated with PFS and OS, respectively, in the cohort who had standard chemotherapy (pGSEA ≤6 × 10−3). Conclusions: We have identified SNPs in three lncRNAs that might be important targets for novel EOC therapies. Clin Cancer Res; 21(23); 5264–76. ©2015 AACR.


PLOS ONE | 2015

The Novel Gene CRNDE Encodes a Nuclear Peptide (CRNDEP) Which Is Overexpressed in Highly Proliferating Tissues

Lukasz Szafron; Anna Balcerak; Ewa Grzybowska; Barbara Pienkowska-Grela; Anna Felisiak-Golabek; Agnieszka Podgorska; Magdalena Kulesza; Natalia Nowak; Paweł Pomorski; Juliusz Wysocki; Tymon Rubel; Agnieszka Dansonka-Mieszkowska; Bozena Konopka; Martyna Lukasik; Jolanta Kupryjanczyk

CRNDE, recently described as the lncRNA-coding gene, is overexpressed at RNA level in human malignancies. Its role in gametogenesis, cellular differentiation and pluripotency has been suggested as well. Herein, we aimed to verify our hypothesis that the CRNDE gene may encode a protein product, CRNDEP. By using bioinformatics methods, we identified the 84-amino acid ORF encoded by one of two CRNDE transcripts, previously described by our research team. This ORF was cloned into two expression vectors, subsequently utilized in localization studies in HeLa cells. We also developed a polyclonal antibody against CRNDEP. Its specificity was confirmed in immunohistochemical, cellular localization, Western blot and immunoprecipitation experiments, as well as by showing a statistically significant decrease of endogenous CRNDEP expression in the cells with transient shRNA-mediated knockdown of CRNDE. Endogenous CRNDEP localizes predominantly to the nucleus and its expression seems to be elevated in highly proliferating tissues, like the parabasal layer of the squamous epithelium, intestinal crypts or spermatocytes. After its artificial overexpression in HeLa cells, in a fusion with either the EGFP or DsRed Monomer fluorescent tag, CRNDEP seems to stimulate the formation of stress granules and localize to them. Although the exact role of CRNDEP is unknown, our preliminary results suggest that it may be involved in the regulation of the cell proliferation. Possibly, CRNDEP also participates in oxygen metabolism, considering our in silico results, and the correlation between its enforced overexpression and the formation of stress granules. This is the first report showing the existence of a peptide encoded by the CRNDE gene.


Orphanet Journal of Rare Diseases | 2015

Germline SMARCA4 mutations in patients with ovarian small cell carcinoma of hypercalcemic type.

Joanna Moes-Sosnowska; Lukasz Szafron; Dorota Nowakowska; Agnieszka Dansonka-Mieszkowska; Agnieszka Budzilowska; Bozena Konopka; Joanna Plisiecka-Halasa; Agnieszka Podgorska; Iwona K. Rzepecka; Jolanta Kupryjanczyk

BackgroundSMARCA4 mutations have recently been identified as driving lesions of the ovarian small cell carcinoma of hypercalcemic type (SCCHT). Familial occurrence of this neoplasm was described previously.MethodsWe looked for germline SMARCA4 alterations in eight patients with the SCCHT. DNA was extracted from probands’ and their relatives’ blood. The SMARCA4 coding sequence, previously found altered in all the tumors, was PCR amplified and sequenced in the germline DNA.ResultsTwo patients carried a heterozygous germline SMARCA4 alteration: c.3760G > T and c.2352insG, respectively. The analysis of the probands’ next of kins revealed that the c.3760G > T mutation was inherited by the proband and her sister from their father, and the sisters’ four children also carried the mutation. The proband’s sister was diagnosed with a carcinoma of the parotid gland at age 2. A brother of the other proband was tested negative.ConclusionsOur study suggests that some women develop the ovarian SCCHT due to the inherited or possibly de novo-occurring germline alterations in the SMARCA4 gene, however, its penetrance appears limited. Nevertheless, because of high aggressiveness of the SCCHT, a molecular diagnostics of the SMARCA4 gene and careful follow-up should be offered to patients with this cancer and their families.


Cancer Epidemiology, Biomarkers & Prevention | 2014

Variation in NF-κB Signaling Pathways and Survival in Invasive Epithelial Ovarian Cancer

Matthew S. Block; Bridget Charbonneau; Robert A. Vierkant; Zachary C. Fogarty; William R. Bamlet; Paul Pharoah; Georgia Chenevix-Trench; Mary Anne Rossing; Daniel W. Cramer; Celeste Leigh Pearce; Joellen M. Schildkraut; Usha Menon; Susanne K. Kjaer; Douglas A. Levine; Jacek Gronwald; Hoda Anton Culver; Alice S. Whittemore; Beth Y. Karlan; Diether Lambrechts; Nicolas Wentzensen; Jolanta Kupryjanczyk; Jenny Chang-Claude; Elisa V. Bandera; Estrid Høgdall; Florian Heitz; Stanley B. Kaye; Peter A. Fasching; Ian G. Campbell; Marc T. Goodman; Tanja Pejovic

Survival in epithelial ovarian cancer (EOC) is influenced by the host immune response, yet the key genetic determinants of inflammation and immunity that affect prognosis are not known. The nuclear factor-κB (NF-κB) transcription factor family plays an important role in many immune and inflammatory responses, including the response to cancer. We studied common inherited variation in 210 genes in the NF-κB family in 10,084 patients with invasive EOC (5,248 high-grade serous, 1,452 endometrioid, 795 clear cell, and 661 mucinous) from the Ovarian Cancer Association Consortium. Associations between genotype and overall survival were assessed using Cox regression for all patients and by major histology, adjusting for known prognostic factors and correcting for multiple testing (threshold for statistical significance, P < 2.5 × 10−5). Results were statistically significant when assessed for patients of a single histology. Key associations were with caspase recruitment domain family, member 11 (CARD11) rs41324349 in patients with mucinous EOC [HR, 1.82; 95% confidence interval (CI), 1.41–2.35; P = 4.13 × 10−6] and tumor necrosis factor receptor superfamily, member 13B (TNFRSF13B) rs7501462 in patients with endometrioid EOC (HR, 0.68; 95% CI, 0.56–0.82; P = 2.33 × 10−5). Other associations of note included TNF receptor–associated factor 2 (TRAF2) rs17250239 in patients with high-grade serous EOC (HR, 0.84; 95% CI, 0.77–0.92; P = 6.49 × 10−5) and phospholipase C, gamma 1 (PLCG1) rs11696662 in patients with clear cell EOC (HR, 0.43; 95% CI, 0.26–0.73; P = 4.56 × 10−4). These associations highlight the potential importance of genes associated with host inflammation and immunity in modulating clinical outcomes in distinct EOC histologies. Cancer Epidemiol Biomarkers Prev; 23(7); 1421–7. ©2014 AACR.


Journal of Cancer Research and Clinical Oncology | 2016

Unsupervised analysis reveals two molecular subgroups of serous ovarian cancer with distinct gene expression profiles and survival

Katarzyna M. Lisowska; Magdalena Olbryt; Sebastian Student; Katarzyna Kujawa; Alexander J. Cortez; Krzysztof Simek; Agnieszka Dansonka-Mieszkowska; Iwona K. Rzepecka; Patrycja Tudrej; Jolanta Kupryjanczyk

PurposeOvarian cancer is typically diagnosed at late stages, and thus, patients’ prognosis is poor. Improvement in treatment outcomes depends, at least partly, on better understanding of ovarian cancer biology and finding new molecular markers and therapeutic targets.MethodsAn unsupervised method of data analysis, singular value decomposition, was applied to analyze microarray data from 101 ovarian cancer samples; then, selected genes were validated by quantitative PCR.ResultsWe found that the major factor influencing gene expression in ovarian cancer was tumor histological type. The next major source of variability was traced to a set of genes mainly associated with extracellular matrix, cell motility, adhesion, and immunological response. Hierarchical clustering based on the expression of these genes revealed two clusters of ovarian cancers with different molecular profiles and distinct overall survival (OS). Patients with higher expression of these genes had shorter OS than those with lower expression. The two clusters did not derive from high- versus low-grade serous carcinomas and were unrelated to histological (ovarian vs. fallopian) origin. Interestingly, there was considerable overlap between identified prognostic signature and a recently described invasion-associated signature related to stromal desmoplastic reaction. Several genes from this signature were validated by quantitative PCR; two of them—DSPG3 and LOX—were validated both in the initial and independent sets of samples and were significantly associated with OS and disease-free survival.ConclusionsWe distinguished two molecular subgroups of serous ovarian cancers characterized by distinct OS. Among differentially expressed genes, some may potentially be used as prognostic markers. In our opinion, unsupervised methods of microarray data analysis are more effective than supervised methods in identifying intrinsic, biologically sound sources of variability. Moreover, as histological type of the tumor is the greatest source of variability in ovarian cancer and may interfere with analyses of other features, it seems reasonable to use histologically homogeneous groups of tumors in microarray experiments.


Oncotarget | 2016

Inherited variants affecting RNA editing may contribute to ovarian cancer susceptibility: results from a large-scale collaboration

Jennifer B. Permuth; Brett M. Reid; Madalene Earp; Y. Ann Chen; Alvaro N.A. Monteiro; Zhihua Chen; Georgia Chenevix-Trench; Peter A. Fasching; Matthias W. Beckmann; Diether Lambrechts; Adriaan Vanderstichele; Els Van Niewenhuyse; Ignace Vergote; Mary Anne Rossing; Jennifer A. Doherty; Jenny Chang-Claude; Kirsten B. Moysich; Kunle Odunsi; Marc T. Goodman; Yurii B. Shvetsov; Lynne R. Wilkens; Pamela J. Thompson; Thilo Dörk; Natalia Bogdanova; Ralf Bützow; Heli Nevanlinna; Liisa M. Pelttari; Arto Leminen; Francesmary Modugno; Robert P. Edwards

RNA editing in mammals is a form of post-transcriptional modification in which adenosine is converted to inosine by the adenosine deaminases acting on RNA (ADAR) family of enzymes. Based on evidence of altered ADAR expression in epithelial ovarian cancers (EOC), we hypothesized that single nucleotide polymorphisms (SNPs) in ADAR genes modify EOC susceptibility, potentially by altering ovarian tissue gene expression. Using directly genotyped and imputed data from 10,891 invasive EOC cases and 21,693 controls, we evaluated the associations of 5,303 SNPs in ADAD1, ADAR, ADAR2, ADAR3, and SND1. Unconditional logistic regression was used to estimate odds ratios (OR) and 95% confidence intervals (CI), with adjustment for European ancestry. We conducted gene-level analyses using the Admixture Maximum Likelihood (AML) test and the Sequence-Kernel Association test for common and rare variants (SKAT-CR). Association analysis revealed top risk-associated SNP rs77027562 (OR (95% CI)= 1.39 (1.17-1.64), P=1.0×10−4) in ADAR3 and rs185455523 in SND1 (OR (95% CI)= 0.68 (0.56-0.83), P=2.0×10−4). When restricting to serous histology (n=6,500), the magnitude of association strengthened for rs185455523 (OR=0.60, P=1.0×10−4). Gene-level analyses revealed that variation in ADAR was associated (P<0.05) with EOC susceptibility, with PAML=0.022 and PSKAT-CR=0.020. Expression quantitative trait locus analysis in EOC tissue revealed significant associations (P<0.05) with ADAR expression for several SNPs in ADAR, including rs1127313 (G/A), a SNP in the 3′ untranslated region. In summary, germline variation involving RNA editing genes may influence EOC susceptibility, warranting further investigation of inherited and acquired alterations affecting RNA editing.


Cancer Biology & Therapy | 2013

p19INK4d mRNA and protein expression as new prognostic factors in ovarian cancer patients

Anna Felisiak-Golabek; Agnieszka Dansonka-Mieszkowska; Iwona K. Rzepecka; Lukasz Szafron; Ewa Kwiatkowska; Bozena Konopka; Agnieszka Podgorska; Alina Rembiszewska; Jolanta Kupryjanczyk

p19INK4d (CDKN2D) is a negative regulator of the cell cycle. Little is known of its role in cancer development and prognosis. We aimed to evaluate the clinical significance of p19INK4d expression in ovarian carcinomas with respect to the TP53 accumulation status, as well as the frequency of CDKN2D mutations. p19INK4d and TP53 expression was evaluated immunohistochemically in 445 ovarian carcinomas: 246 patients were treated with platinum–cyclophosphamide (PC/PAC), while 199 were treated with taxane–platinum agents (TP). CDKN2D gene expression (mRNA) was examined in 106 carcinomas, while CDKN2D mutations in 68 tumors. Uni- and multivariate statistical analyses (logistic regression and the Cox proportional hazards model) were performed for patient groups divided according to the chemotherapeutic regimen administered, and in subgroups with and without TP53 accumulation. High p19INK4d expression increased the risk of death, but only in patients with the TP53-negative carcinomas (HR 1.61, P = 0.049 for PC/PAC-treated patients, HR 2.00, P = 0.015 for TP-treated patients). This result was confirmed by the mRNA analysis (HR 4.24, P = 0.001 for TP-treated group). High p19INK4d protein expression associated with adverse clinicopathological factors. We found no alterations in the CDKN2D gene; the c.90C>G (p.R30R; rs1968445) polymorphism was detected in 10% of tumors. Our results suggest that p19INK4d expression is a poor prognostic factor in ovarian cancer patients. Analyses of tumor groups according to the TP53 accumulation status facilitate the identification of cancer biomarkers.

Collaboration


Dive into the Agnieszka Dansonka-Mieszkowska's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jenny Chang-Claude

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter A. Fasching

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marc T. Goodman

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Georgia Chenevix-Trench

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Ian G. Campbell

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Diether Lambrechts

Katholieke Universiteit Leuven

View shared research outputs
Researchain Logo
Decentralizing Knowledge