Network


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

Hotspot


Dive into the research topics where Piotr Rieske is active.

Publication


Featured researches published by Piotr Rieske.


British Journal of Cancer | 2011

Glioma cells showing IDH1 mutation cannot be propagated in standard cell culture conditions.

Sylwester Piaskowski; Michal Bienkowski; Ewelina Stoczynska-Fidelus; Robert Stawski; Monika Sieruta; Malgorzata Szybka; Wielisław Papierz; M Wolanczyk; Liberski Pp; Piotr Rieske

Background:It has recently been reported by several sources that original (i.e., present in vivo) glioma cell phenotypes or genotypes cannot be maintained in vitro. For example, glioblastoma cell lines presenting EGFR amplification cannot be established.Methods and results:IDH1 sequencing and loss of heterozygosity analysis was performed for 15 surgery samples of astrocytoma and early and late passages of cells derived from those and for 11 archival samples. We were not able to culture tumour cells presenting IDH1 mutations originating from currently proceeded 10 tumours; the same results were observed in 7 samples of archival material.Conclusion:The IDH1 mutation is expected to be almost mutually exclusive with EGFR amplification, so glioma cells with IDH1 mutations seem to represent a new group of tumour cells, which cannot be readily analysed in vitro because of their elimination. The reasons for this intriguing phenomenon should be investigated since its understanding can help to define a new therapeutic approach based on simulating in vivo conditions, responsible for tumour cells elimination in vitro. Moreover, a new model for culturing glioma cells in vitro should be designed since the current one does not provide conditions corresponding to in vivo growth.


European Journal of Neuroscience | 2007

A population of human brain parenchymal cells express markers of glial, neuronal and early neural cells and differentiate into cells of neuronal and glial lineages.

Piotr Rieske; S. Ausim Azizi; Brian J. Augelli; John P. Gaughan; Barbara Krynska

Glial fibrillary acidic protein (GFAP)‐positive cells derived from the neurogenic areas of the brain can be stem/progenitor cells and give rise to new neurons in vitro and in vivo. We report here that a population of GFAP‐positive cells derived from fetal human brain parenchyma coexpress markers of early neural and neuronal cells, and have neural progenitor cell characteristics. We used a monolayer culture system to expend and differentiate these cells. During the initial proliferative phase, all cells expressed GFAP, nestin and low levels of βIII‐tubulin. When these cells were cultured in serum and then basic fibroblast growth factor, they generated two distinct progenies: (i) βIII‐tubulin‐ and nestin‐positive cells and (ii) GFAP‐ and nestin‐positive cells. These cells, when subsequently cultured in serum‐free media without growth factors, ceased to proliferate and differentiated into two major neural cell classes, neurons and glia. In the cells of neuronal lineage, nestin expression was down‐regulated and βIII‐tubulin expression became robust. Cells of glial lineage differentiated by down‐regulating nestin expression and up‐regulating GFAP expression. These data suggest that populations of parenchymal brain cells, initially expressing both glial and neuronal markers, are capable of differentiating into single neuronal and glial lineages through asymmetric regulation of gene expression in these cells, rather than acquiring markers through differentiation.


BMC Cancer | 2009

Arrested neural and advanced mesenchymal differentiation of glioblastoma cells-comparative study with neural progenitors

Piotr Rieske; Ewa Golanska; Magdalena Zakrzewska; Sylwester Piaskowski; Krystyna Hulas-Bigoszewska; M Wolanczyk; Malgorzata Szybka; Monika Witusik-Perkowska; Krzysztof Zakrzewski; Wojciech Biernat; Barbara Krynska; Pawel P. Liberski

BackgroundAlthough features of variable differentiation in glioblastoma cell cultures have been reported, a comparative analysis of differentiation properties of normal neural GFAP positive progenitors, and those shown by glioblastoma cells, has not been performed.MethodsFollowing methods were used to compare glioblastoma cells and GFAP+NNP (NHA): exposure to neural differentiation medium, exposure to adipogenic and osteogenic medium, western blot analysis, immunocytochemistry, single cell assay, BrdU incorporation assay. To characterize glioblastoma cells EGFR amplification analysis, LOH/MSI analysis, and P53 nucleotide sequence analysis were performed.ResultsIn vitro differentiation of cancer cells derived from eight glioblastomas was compared with GFAP-positive normal neural progenitors (GFAP+NNP). Prior to exposure to differentiation medium, both types of cells showed similar multilineage phenotype (CD44+/MAP2+/GFAP+/Vimentin+/Beta III-tubulin+/Fibronectin+) and were positive for SOX-2 and Nestin. In contrast to GFAP+NNP, an efficient differentiation arrest was observed in all cell lines isolated from glioblastomas. Nevertheless, a subpopulation of cells isolated from four glioblastomas differentiated after serum-starvation with varying efficiency into derivatives indistinguishable from the neural derivatives of GFAP+NNP. Moreover, the cells derived from a majority of glioblastomas (7 out of 8), as well as GFAP+NNP, showed features of mesenchymal differentiation when exposed to medium with serum.ConclusionOur results showed that stable co-expression of multilineage markers by glioblastoma cells resulted from differentiation arrest. According to our data up to 95% of glioblastoma cells can present in vitro multilineage phenotype. The mesenchymal differentiation of glioblastoma cells is advanced and similar to mesenchymal differentiation of normal neural progenitors GFAP+NNP.


Cancer Genetics and Cytogenetics | 2009

High incidence of MGMT promoter methylation in primary glioblastomas without correlation with TP53 gene mutations

Emilia Jesień-Lewandowicz; Dorota Jesionek-Kupnicka; Izabela Zawlik; Malgorzata Szybka; Dominika Kulczycka-Wojdala; Piotr Rieske; Monika Sieruta; Waldemar Och; Wiesław Skowronski; Beata Sikorska; Piotr Potemski; Wielisław Papierz; Pawel P. Liberski; Radzisław Kordek

O(6)-methylguanine DNA methyltransferase (MGMT) reduces cytotoxicity of therapeutic or environmental alkylating agents. MGMT promoter methylation has been associated with TP53 G: C to A:T transition mutations in various types of cancers, and with poor prognosis in patients who did not receive chemotherapy. Mutations of TP53 are more frequent in secondary than in primary glioblastoma, thus the expected MGMT promoter methylation was low in primary glioblastoma. Glioblastoma patients with MGMT promoter methylation showed better response to chemotherapy based on alkylating agents and longer survival than patients without MGMT methylation. We examined 32 primary glioblastomas, treated with radiotherapy and surgery, for TP53 mutation by direct sequencing and MGMT promoter methylation by methylation-specific PCR. MGMT promoter methylation and TP53 mutations were detected in 72% and 31% of primary glioblastoma, respectively. Although not statistically significant, the frequency of TP53 G:C to A:T mutations were higher in cases with (26%) than without (11%) MGMT promoter methylation (p=0.376). MGMT promoter methylation had no impact on patient survival. Our data indicate that MGMT promoter methylation occurs frequently in primary glioblastoma, but does not lead to G:C to A:T TP53 mutations, has no independent prognostic value and is not a predictive marker unless glioblastoma patients are treated with chemotherapy.


Stem Cell Research & Therapy | 2015

Generation of human iPSCs from cells of fibroblastic and epithelial origin by means of the oriP/EBNA-1 episomal reprogramming system

Anna M. Drozd; Maciej P. Walczak; Sylwester Piaskowski; Ewelina Stoczynska-Fidelus; Piotr Rieske; Dawid P. Grzela

IntroductionThe prospect of therapeutic applications of the induced pluripotent stem cells (iPSCs) is based on their ability to generate virtually any cell type present in human body. Generation of iPSCs from somatic cells has opened up new possibilities to investigate stem cell biology, to better understand pathophysiology of human diseases, and to design new therapy approaches in the field of regenerative medicine. In this study, we focus on the ability of the episomal system, a non-viral and integration-free reprogramming method to derive iPSCs from somatic cells of various origin.MethodsCells originating from neonatal and adult tissue, renal epithelium, and amniotic fluid were reprogrammed by using origin of replication/Epstein-Barr virus nuclear antigen-1 (oriP/EBNA-1)-based episomal vectors carrying defined factors. The iPSC colony formation was evaluated by using immunocytochemistry and alkaline phosphatase assay and by investigating gene expression profiles. The trilineage formation potential of generated pluripotent cells was assessed by embryoid body-mediated differentiation. The impact of additionally introduced factors on episome-based reprogramming was also investigated.ResultsReprogramming efficiencies were significantly higher for the epithelial cells compared with fibroblasts. The presence of additional factor miR 302/367 in episomal system enhanced reprogramming efficiencies in fibroblasts and epithelial cells, whereas the downregulation of Mbd3 expression increased iPSC colony-forming efficiency in fibroblasts solely.ConclusionsIn this study, we performed a side-by-side comparison of iPSC colony-forming efficiencies in fibroblasts and epithelial cells transiently transfected with episomal plasmids and demonstrated that iPSC generation efficiency was highest when donor samples were derived from epithelial cells. We determined that reprogramming efficiency of episomal system could be further improved. Considering results obtained in the course of this study, we believe that episomal reprogramming provides a simple, reproducible, and efficient tool for generating clinically relevant pluripotent cells.


Neuroscience Letters | 2005

CYP46 : A risk factor for Alzheimer's disease or a coincidence?

Ewa Golanska; Krystyna Hulas-Bigoszewska; Izabela Wojcik; Piotr Rieske; Maria Styczyńska; Beata Peplonska; Anna Pfeffer; Elzbieta Luczywek; B. Wasiak; Tomasz Gabryelewicz; Dorota Religa; Malgorzata Chodakowska-Zebrowska; Maria Barcikowska; Tomasz Sobow; Pawel P. Liberski

Excess cholesterol is removed from the brain via hydroxylation mediated by cholesterol 24S-hydroxylase (CYP46). Although serum and cerebrospinal fluid (CSF) concentrations of 24S-hydroxycholesterol are altered during the progress of Alzheimers disease, studies carried out to date in different populations on the association of CYP46 gene polymorphisms and risk of AD have been inconclusive. In this report, we analyzed CYP46 polymorphisms in 215 Polish AD cases and 173 healthy individuals. A fragment of CYP46 intron 2 was amplified by PCR reaction and sequenced. We discovered a new single nucleotide substitution in CYP46 intron 2, but found no difference in particular genotype or allele frequencies between AD patients and controls. However, the GG genotype of the known rs754203 polymorphic site might be a risk factor for AD, especially in APOE varepsilon4 carriers. Interestingly, in AD patients the rs754203 G allele was more frequent in males than in females. However, considering the extreme divergence of results obtained by different authors, a clear connection between the CYP46 gene and AD is questionable.


Neuroscience Letters | 2006

Regulation of PrPC expression: nerve growth factor (NGF) activates the prion gene promoter through the MEK1 pathway in PC12 cells.

Izabela Zawlik; Monika Witusik; Krystyna Hulas-Bigoszewska; Sylwester Piaskowski; Malgorzata Szybka; Ewa Golanska; Pawel P. Liberski; Piotr Rieske

A high expression of PrP(C) in cells is one factor that increases the risk of conversion to the misfolded, disease-associated form (PrP(Sc)) characteristic of transmissible spongiform encephalopathies. Thus, developing a method to control the level of PrP(C) expression in cells could be one way to delay or prevent the onset of clinical signs of these diseases. In this study the mechanisms controlling the expression of the Prnp gene in PC12 cells and in rat brain were examined. We observed a slight activation of a cloned fragment of the human PRNP gene promoter using the luciferase reporter system in PC12 cells stimulated with nerve growth factor (NGF). The activating effect of NGF was enhanced by mitogen-activated protein kinase (MEK1) and suppressed by myristylated serine/threonine kinase (myrAKT). These results suggest that MEK1 is a positive activator of the PRNP promoter that inhibits the AKT pathway. Independent experiments suggested that high expression of PrP(C) in the brain depends on the rate of translation and/or the efficiency of PrP(C) stabilization. We also investigated the epigenic status of the Prnp promoter. We observed no increase of PrP(C) or Prnp mRNA levels in PC12 cells after treatment with the DNA-demethylating agent. The Prnp promoter did not display methylation either in NGF-treated and untreated PC12 cells, or in the rat brain. These results improve the understanding of the regulation of the Prnp gene promoter, a DNA regulatory element controlling the expression of PrP(C), a protein involved in several neurological diseases.


Journal of Neuro-oncology | 2005

Atypical molecular background of glioblastoma and meningioma developed in a patient with Li–Fraumeni syndrome

Piotr Rieske; Magdalena Zakrzewska; Wojciech Biernat; Jacek Bartkowiak; Arthur Zimmermann; Pawel P. Liberski

We observed three neoplasms with completely different histologies: malignant fibrous histiocytoma (MFH), atypical meningioma (AM), and glioblastoma (GB), developing in a patient with Li–Fraumeni syndrome. By using a combined molecular approach we performed molecular characterization of all three tumours. Data obtained showed an interesting molecular background of the AM and GB. AM showed TP53mutations and a 22q loss of heterozygosity (LOH). GB showed epidermal growth factor receptor (EGFR) amplification and TP53 mutations, whereas P16, PTEN, Rbwere intact in terms of LOH and/or multiplex PCR (polymerase chain reaction) analysis. Additionally, GB has a 1q LOH, which is an extremely rare alteration in glioblastomas. Identical 1q LOH was also observed in MFH.


Leukemia & Lymphoma | 2004

Multiple myeloma in a patient with systemic lupus erythematosus, myasthenia gravis and non-familial diffuse palmoplantar keratoderma.

Halina Urbańska-Ryś; Ewa Robak; Radzisław Kordek; Jacek Bartkowiak; Piotr Rieske; Anna Woźniacka; Piotr Smolewski; Tadeusz Robak

The coexistence of autoimmune diseases and malignancies including lymphoproliferative diseases is often reported in the literature. Here we report an unusual case with two autoimmune diseases--myasthenia gravis (MG) and systemic lupus erythematosus (SLE) associated with unique palmoplantar keratoderma (PK) which preceded the development of multiple myeloma (MM) for twenty and seven years respectively. MG associated with non-malignant thymoma developed in 1981 and was successfully treated with thymectomy and physostigmine. Thirteen years later SLE was diagnosed and until now it is also accompanied by skin lesions corresponding to non-familial, diffuse palmoplantar keratoderma which is resistant to treatment. In 2001 the patient revealed inguinal and abdominal lymphadenopathy first diagnosed as extramedullary plasmacytoma and then as multiple myeloma on the basis of bone marrow infiltration and monoclonal gammopathy. Therapy with VAD regimen achieved complete remission of the MM and significant improvement of the skin changes lasting for six months. We failed to collect sufficient numbers of CD 34+ cells for peripheral blood stem cell transplantation. Now the malignancy is in partial remission after CHOP therapy and the skin lesions have returned to their initial status. To our knowledge, this is the first case to be reported with coexistence of these four diseases.


British Journal of Cancer | 2008

Elimination of wild-type P53 mRNA in glioblastomas showing heterozygous mutations of P53

Malgorzata Szybka; Izabela Zawlik; Kulczycka D; Ewa Golanska; E Jesien; Kupnicka D; Robert Stawski; Sylwester Piaskowski; Ewa Bieniek; Magdalena Zakrzewska; Radzisław Kordek; Pawel P. Liberski; Piotr Rieske

We screened 50 glioblastomas for P53 mutations. Five glioblastomas showed heterozygous mutations, while three were putatively heterozygous. Six of these eight glioblastomas showed elimination of wild-type P53 mRNA. These results strongly suggest that some sort of mechanism(s) favouring mutated over wild-type P53 mRNA exists in glioblastoma cells with heterozygous mutations of this gene.

Collaboration


Dive into the Piotr Rieske's collaboration.

Top Co-Authors

Avatar

Sylwester Piaskowski

Medical University of Łódź

View shared research outputs
Top Co-Authors

Avatar

Pawel P. Liberski

Medical University of Łódź

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Malgorzata Szybka

Medical University of Łódź

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Magdalena Zakrzewska

Medical University of Łódź

View shared research outputs
Top Co-Authors

Avatar

Michal Bienkowski

Medical University of Łódź

View shared research outputs
Top Co-Authors

Avatar

Ewa Golanska

Medical University of Łódź

View shared research outputs
Top Co-Authors

Avatar

Mateusz Banaszczyk

Medical University of Łódź

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge