Network


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

Hotspot


Dive into the research topics where Katrin Rapakko is active.

Publication


Featured researches published by Katrin Rapakko.


Nature | 2007

A recurrent mutation in PALB2 in Finnish cancer families

Hannele Erkko; Bing Xia; Jenni Nikkilä; Johanna Schleutker; Kirsi Syrjäkoski; Arto Mannermaa; Anne Kallioniemi; Katri Pylkäs; Sanna Maria Karppinen; Katrin Rapakko; Alexander Miron; Qing Sheng; Guilan Li; Henna Mattila; Daphne W. Bell; Daniel A. Haber; Mervi Grip; Mervi Reiman; Arja Jukkola-Vuorinen; Aki Mustonen; Juha Kere; Lauri A. Aaltonen; Veli-Matti Kosma; Vesa Kataja; Ylermi Soini; Ronny Drapkin; David M. Livingston; Robert Winqvist

BRCA1, BRCA2 and other known susceptibility genes account for less than half of the detectable hereditary predisposition to breast cancer. Other relevant genes therefore remain to be discovered. Recently a new BRCA2-binding protein, PALB2, was identified. The BRCA2–PALB2 interaction is crucial for certain key BRCA2 DNA damage response functions as well as its tumour suppression activity. Here we show, by screening for PALB2 mutations in Finland that a frameshift mutation, c.1592delT, is present at significantly elevated frequency in familial breast cancer cases compared with ancestry-matched population controls. The truncated PALB2 protein caused by this mutation retained little BRCA2-binding capacity and was deficient in homologous recombination and crosslink repair. Further screening of c.1592delT in unselected breast cancer individuals revealed a roughly fourfold enrichment of this mutation in patients compared with controls. Most of the mutation-positive unselected cases had a familial pattern of disease development. In addition, one multigenerational prostate cancer family that segregated the c.1592delT truncation allele was observed. These results indicate that PALB2 is a breast cancer susceptibility gene that, in a suitably mutant form, may also contribute to familial prostate cancer development.


European Journal of Human Genetics | 2000

Multiple founder effects and geographical clustering of BRCA1 and BRCA2 families in Finland

Laura Sarantaus; Pia Huusko; Hannaleena Eerola; Virpi Launonen; Paula Vehmanen; Katrin Rapakko; Elizabeth M. Gillanders; Kirsi Syrjäkoski; Tommi Kainu; Pia Vahteristo; Ralf Krahe; Kati Pääkkönen; Jaana M. Hartikainen; Carl Blomqvist; Tuija Löppönen; Kaija Holli; Markku Ryynänen; Ralf Bützow; Åke Borg; Brita Arver; Eva Holmberg; Arto Mannermaa; Juha Kere; Olli Kallioniemi; Robert Winqvist; Heli Nevanlinna

In the Finnish breast and ovarian cancer families six BRCA1 and five BRCA2 mutations have been found recurrently. Some of these recurrent mutations have also been seen elsewhere in the world, while others are exclusively of Finnish origin. A haplotype analysis of 26xa0Finnish families carrying a BRCA1 mutation and 20xa0families with a BRCA2 mutation indicated that the carriers of each recurrent mutation have common ancestors. The common ancestors were estimated to trace back to 7–36xa0generations (150–800xa0years). The time estimates and the geographical clustering of these founder mutations in Finland are in concordance with the population history of this country. Analysis of the cancer phenotypes showed differential ovarian cancer expression in families carrying mutations in the 5′ and 3′ ends of the BRCA1 gene, and earlier age of ovarian cancer onset in families with BRCA1 mutations compared with families with BRCA2 mutations. The identification of prominent and regional BRCA1 and BRCA2 founder mutations in Finland will have significant impact on diagnostics in Finnish breast and ovarian cancer families. An isolated population with known history and multiple local founder effects in multigenic disease may offer distinct advantages also for mapping novel predisposing genes.


Journal of Medical Genetics | 2004

Mutation screening of the BARD1 gene: evidence for involvement of the Cys557Ser allele in hereditary susceptibility to breast cancer.

Sanna-Maria Karppinen; Heikkinen K; Katrin Rapakko; Robert Winqvist

Approximately 5–10% of all breast and ovarian cancers are thought to arise from a hereditary predisposition to the disease,1 BRCA1 and BRCA2 being the most important susceptibility genes.2,3 Genomic alterations in BRCA1 are found in 40–50% of families with a high incidence of breast cancer (six or more cases), and in a majority (75–80%) of the families that display both breast and ovarian cancers.4,5 However, a significant portion of genetic aberrations predisposing to these cancers, especially in relatively small risk families, still remains unexplained.6,7 BRCA1 interacts with a variety of proteins and is involved in multiple cellular processes including DNA repair, transcription, and checkpoint control.8–10 In attempts to identify new breast and ovarian cancer susceptibility genes, much research has focused on BRCA1 associated proteins.nnBARD1 was originally identified through its interaction with BRCA1, with which it has a closely related domain structure.11 Both proteins possess an N-terminal RING finger motif and two BRCA1 C-terminal (BRCT) domains present in numerous proteins involved in DNA repair and cell cycle regulation.11 The functionally important BARD1/BRCA1 heterodimer formation is mediated by the RING finger motifs and has also been shown to markedly increase the stability of both proteins.11–13 The finding of breast cancer associated mutations within the RING finger domain of BRCA1 , disrupting BRCA1/BARD1 interaction,11,14 and the occurrence of BARD1 missense mutations in breast cancer patients,15–17 implies participation of BARD1 in BRCA1 mediated tumour suppression. BARD1, unlike BRCA1, also contains a centrally located sequence of three ankyrin repeats11 that are found in many proteins involved in transcriptional regulation.18nnColocalisation of BARD1 with BRCA1 and RAD51 in response to DNA damage indicates a role in DNA repair,19,20 which is supported by the recent …


Journal of Medical Genetics | 2006

Nordic collaborative study of the BARD1 Cys557Ser allele in 3956 patients with cancer: enrichment in familial BRCA1/BRCA2 mutation-negative breast cancer but not in other malignancies

Sanna-Maria Karppinen; Rosa B. Barkardottir; Katja Backenhorn; Thomas Sydenham; Kirsi Syrjäkoski; Johanna Schleutker; Tarja Ikonen; Katri Pylkäs; Katrin Rapakko; Hannele Erkko; Gudrun Johannesdottir; Anne-Marie Gerdes; Mads Thomassen; Bjarni A. Agnarsson; Mervi Grip; Anne Kallioniemi; Juha Kere; Lauri A. Aaltonen; Adalgeir Arason; Pål Møller; Torben A. Kruse; Åke Borg; Robert Winqvist

Background: BARD1 was originally identified as a BRCA1-interacting protein but has also been described in tumour-suppressive functions independent of BRCA1. Several studies have indicated that the BARD1 gene is a potential target for germline changes predisposing to breast and ovarian cancer. The C-terminal Cys557Ser change has previously been uncovered to associate with an increased risk of breast cancer and was recently shown to result in defective apoptotic activities. Aim and methods: Conformation-sensitive gel electrophoresis, minisequencing, TaqMan assays, denaturing high-performance liquid chromatography analysis and DNA sequencing were used to investigate the prevalence of the Cys557Ser allele in a large Nordic case–control study cohort consisting of 2906 patients with breast or ovarian cancer, 734 with prostate cancer, 188 with colorectal cancer, 128 men with breast cancer, and 3591 controls from Finland, Iceland, Denmark, Sweden and Norway. Results: The frequency of the BARD1 Cys557Ser variant seemed to increase among patients from families with breast or ovarian cancer lacking BRCA1 or BRCA2 mutations: a significant difference was obtained compared with controls (6.8% v 2.7%; p<0.001; odds ratio (OR) 2.6; 95% confidence interval (CI) 1.7 to 4.0) and with patients from BRCA1/BRCA2 mutation-positive families (6.8% v 2.2%; pu200a=u200a0.01; OR 3.2; 95% CI 1.2 to 8.3). In contrast, no major association with male breast, ovarian, colorectal or prostate cancer was observed. Additionally, a novel BARD1 allele resulting in Ser558Pro was identified in familial breast cancer cases. Conclusion: These results provide further evidence that BARD1 Cys557Ser confers a slightly increased risk of breast cancer in women.


European Journal of Human Genetics | 2004

Genome-wide scanning for linkage in Finnish breast cancer families.

Pia Huusko; Suh-Hang Hank Juo; Elizabeth Gillanders; Laura Sarantaus; Tommi Kainu; Pia Vahteristo; Minna Allinen; MaryPat Jones; Katrin Rapakko; Hannaleena Eerola; Carol Markey; Paula Vehmanen; Derek Gildea; Diane Freas-Lutz; Carl Blomqvist; Jaakko Leisti; Guillermo Blanco; Ulla Puistola; Jeffrey Trent; Joan Bailey-Wilson; Robert Winqvist; Heli Nevanlinna; Olli-P. Kallioniemi

Only a proportion of breast cancer families has germline mutations in the BRCA1 or BRCA2 genes, suggesting the presence of additional susceptibility genes. Finding such genes by linkage analysis has turned out to be difficult due to the genetic heterogeneity of the disease, phenocopies and incomplete penetrance of the mutations. Isolated populations may be helpful in reducing the level of genetic heterogeneity and in providing useful starting points for further genetic analyses. Here, we report results from a genome-wide linkage analysis of 14 high-risk breast cancer families from Finland. These families tested negative for BRCA1 and BRCA2 germline mutations and showed no linkage to the 13q21 region, recently proposed as an additional susceptibility locus. Suggestive linkage was seen at marker D2S364 (2q32) with a parametric two-point LOD score of 1.61 (θ=0), and an LOD score of 2.49 in nonparametric analyses. Additional genotyping of a 40u2009cM chromosomal region surrounding the region of interest yielded a maximum parametric two-point LOD score of 1.80 (θ=0) at D2S2262 and a nonparametric LOD score of 3.11 at an adjacent novel marker 11291M1 in BAC RP11-67G7. A nonparametric multipoint LOD score of 3.20 was seen at 11291M1 under the assumption of dominant inheritance. While not providing proof of linkage considering the small number of families and large number of laboratory and statistical analyses performed, these results warrant further studies of the 2q32 chromosomal region as a candidate breast cancer susceptibility locus. Both linkage and association studies are likely to be useful, particularly in other isolated populations.


Nature Communications | 2013

Heterozygous mutations in PALB2 cause DNA replication and damage response defects

Jenni Nikkilä; Ann Christin Parplys; Katri Pylkäs; Muthiah Bose; Yanying Huo; Kerstin Borgmann; Katrin Rapakko; Pentti Nieminen; Bing Xia; Helmut Pospiech; Robert Winqvist

Besides mutations in BRCA1/BRCA2, heterozygous defects in PALB2 are important in breast cancer predisposition. PALB2 heterozygosity increases the risk of malignancy about sixfold. PALB2 interacts with BRCA1 and BRCA2 to regulate homologous recombination and mediate DNA damage response. Here we show, by analysing lymphoblastoid cell lines from heterozygous female PALB2 mutation carriers, that PALB2 haploinsufficiency causes aberrant DNA replication/damage response. Mutation carrier cells show increased origin firing and shorter distance between consecutive replication forks. Carrier cell lines also show elevated ATR protein, but not phosphorylation levels, and a majority of them display aberrant Chk1-/Chk2-mediated DNA damage response. Elevated chromosome instability is observed in primary blood lymphocytes of PALB2 mutation carriers, indicating that the described mechanisms of genome destabilization operate also at the organism level. These findings provide a new mechanism for early stages of breast cancer development that may also apply to other heterozygous homologous recombination signalling pathway gene mutations in hereditary cancer predisposition.


Cancer Letters | 2011

Screening for large genomic rearrangements in the FANCA gene reveals extensive deletion in a Finnish breast cancer family

Szilvia Solyom; Robert Winqvist; Jenni Nikkilä; Katrin Rapakko; Pasi Hirvikoski; Hannaleena Kokkonen; Katri Pylkäs

A portion of familial breast cancer cases are caused by mutations in the same genes that are inactivated in the downstream part of Fanconi anemia (FA) signaling pathway. Here we have assessed the FANCA gene for breast cancer susceptibility by examining blood DNA for aberrations from 100 Northern Finnish breast cancer families using the MLPA method. We identified a novel heterozygous deletion, removing the promoter and 12 exons of the gene in one family. This allele was absent from 124 controls. We conclude that FANCA deletions might contribute to breast cancer susceptibility, potentially in combination with other germline mutations. To our knowledge, this is the first study reporting a large deletion in an upstream FA gene in familial breast cancer.


PLOS Genetics | 2016

Targeted Next-Generation Sequencing Identifies a Recurrent Mutation in MCPH1 Associating with Hereditary Breast Cancer Susceptibility.

Tuomo Mantere; Robert Winqvist; Saila Kauppila; Mervi Grip; Arja Jukkola-Vuorinen; Anna Tervasmäki; Katrin Rapakko; Katri Pylkäs

Breast cancer is strongly influenced by hereditary risk factors, a majority of which still remain unknown. Here, we performed a targeted next-generation sequencing of 796 genes implicated in DNA repair in 189 Finnish breast cancer cases with indication of hereditary disease susceptibility and focused the analysis on protein truncating mutations. A recurrent heterozygous mutation (c.904_916del, p.Arg304ValfsTer3) was identified in early DNA damage response gene, MCPH1, significantly associating with breast cancer susceptibility both in familial (5/145, 3.4%, P = 0.003, OR 8.3) and unselected cases (16/1150, 1.4%, P = 0.016, OR 3.3). A total of 21 mutation positive families were identified, of which one-third exhibited also brain tumors and/or sarcomas (P = 0.0007). Mutation carriers exhibited significant increase in genomic instability assessed by cytogenetic analysis for spontaneous chromosomal rearrangements in peripheral blood lymphocytes (P = 0.0007), suggesting an effect for MCPH1 haploinsufficiency on cancer susceptibility. Furthermore, 40% of the mutation carrier tumors exhibited loss of the wild-type allele. These findings collectively provide strong evidence for MCHP1 being a novel breast cancer susceptibility gene, which warrants further investigations in other populations.


Scientific Reports | 2017

Case-control analysis of truncating mutations in DNA damage response genes connects TEX15 and FANCD2 with hereditary breast cancer susceptibility

Tuomo Mantere; Anna Tervasmäki; Anna Nurmi; Katrin Rapakko; Saila Kauppila; Jiangbo Tang; Johanna Schleutker; Anne Kallioniemi; Jaana M. Hartikainen; Arto Mannermaa; Pentti Nieminen; Riitta Hanhisalo; Sini Lehto; Maija Suvanto; Mervi Grip; Arja Jukkola-Vuorinen; Maria Tengström; Päivi Auvinen; Anders Kvist; Åke Borg; Carl Blomqvist; Kristiina Aittomäki; Roger A. Greenberg; Robert Winqvist; Heli Nevanlinna; Katri Pylkäs

Several known breast cancer susceptibility genes encode proteins involved in DNA damage response (DDR) and are characterized by rare loss-of-function mutations. However, these explain less than half of the familial cases. To identify novel susceptibility factors, 39 rare truncating mutations, identified in 189 Northern Finnish hereditary breast cancer patients in parallel sequencing of 796 DDR genes, were studied for disease association. Mutation screening was performed for Northern Finnish breast cancer cases (nu2009=u2009578–1565) and controls (nu2009=u2009337–1228). Mutations showing potential cancer association were analyzed in additional Finnish cohorts. c.7253dupT in TEX15, encoding a DDR factor important in meiosis, associated with hereditary breast cancer (pu2009=u20090.018) and likely represents a Northern Finnish founder mutation. A deleterious c.2715u2009+u20091Gu2009>u2009A mutation in the Fanconi anemia gene, FANCD2, was over two times more common in the combined Finnish hereditary cohort compared to controls. A deletion (c.640_644del5) in RNF168, causative for recessive RIDDLE syndrome, had high prevalence in majority of the analyzed cohorts, but did not associate with breast cancer. In conclusion, truncating variants in TEX15 and FANCD2 are potential breast cancer risk factors, warranting further investigations in other populations. Furthermore, high frequency of RNF168 c.640_644del5 indicates the need for its testing in Finnish patients with RIDDLE syndrome symptoms.


Scientific Reports | 2018

Mutation of TP53, translocation analysis and immunohistochemical expression of MYC, BCL-2 and BCL-6 in patients with DLBCL treated with R-CHOP

Pekka Peroja; Mette Pedersen; Tuomo Mantere; Peter Nørgaard; Jenni Peltonen; Kirsi-Maria Haapasaari; Jan Böhm; Esa Jantunen; Taina Turpeenniemi-Hujanen; Katrin Rapakko; Peeter Karihtala; Ylermi Soini; Kaija Vasala; Outi Kuittinen

Diffuse large B-cell lymphoma (DLBCL) is an aggressive lymphoma with diverse outcomes. Concurrent translocation of MYC and BCL-2 and/or BCL-6, and concurrent immunohistochemical (IHC) high expression of MYC and BCL-2, have been linked to unfavorable treatment responses. TP53-mutated DLBCL has also been linked to worse outcome. Our aim was to evaluate the aforementioned issues in a cohort of 155 patients uniformly treated with R-CHOP-like therapies. We performed direct sequencing of TP53 exons 5, 6, 7 and 8 as well as fluorescence in-situ hybridization (FISH) of MYC, BCL-2 and BCL-6, and IHC of MYC, BCL-2 and BCL-6. In multivariate analysis, TP53 mutations in L3 and loop-sheet helix (LSH) associated with a risk ratio (RR) of disease-specific survival (DSS) of 8.779 (pu2009=u20090.022) and a RR of disease-free survival (DFS) of 10.498 (pu2009=u20090.011). In IHC analysis BCL-2 overexpression was associated with inferior DFS (pu2009=u20090.002) and DSS (pu2009=u20090.002). DLBCL with BCL-2 and MYC overexpression conferred inferior survival in all patients (DSS, pu2009=u20090.038 and DFS, pu2009=u20090.011) and in patients with non-GC phenotype (DSS (pu2009=u20090.013) and DFS (pu2009=u20090.010). Our results imply that in DLBCL, the location of TP53 mutations and IHC analysis of BCL-2 and MYC might have a role in the assessment of prognosis.

Collaboration


Dive into the Katrin Rapakko's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Arto Mannermaa

University of Eastern Finland

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mervi Grip

Oulu University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Juha Kere

Karolinska Institutet

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge