Network


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

Hotspot


Dive into the research topics where Anna Vähärautio is active.

Publication


Featured researches published by Anna Vähärautio.


Nature Methods | 2012

Counting absolute numbers of molecules using unique molecular identifiers

Teemu Kivioja; Anna Vähärautio; Kasper Karlsson; Martin Bonke; Martin Enge; Sten Linnarsson; Jussi Taipale

Counting individual RNA or DNA molecules is difficult because they are hard to copy quantitatively for detection. To overcome this limitation, we applied unique molecular identifiers (UMIs), which make each molecule in a population distinct, to genome-scale human karyotyping and mRNA sequencing in Drosophila melanogaster. Use of this method can improve accuracy of almost any next-generation sequencing method, including chromatin immunoprecipitation–sequencing, genome assembly, diagnostics and manufacturing-process control and monitoring.


Science | 2012

Mice lacking a Myc enhancer that includes human SNP rs6983267 are resistant to intestinal tumors.

Inderpreet Sur; Outi Hallikas; Anna Vähärautio; Jian Yan; Mikko P. Turunen; Martin Enge; Minna Taipale; Auli Karhu; Lauri A. Aaltonen; Jussi Taipale

From Man to Mouse Genome-wide association studies of humans have identified single-nucleotide polymorphisms (SNPs) that increase an individuals risk of developing common diseases like cancer. Most of these SNPs have only a modest effect on risk, and many map to noncoding regions of the genome. Sur et al. (p. 1360, published online 1 November; see the Perspective by Lewis and Tomlinson) used a mouse model to study the functional impact of a particular SNP that resides 300 kilobases upstream of the MYC oncogene on human chromosome 8q24 and has been linked to cancer risk in humans. When a sequence encompassing this SNP was deleted in mice that were predisposed to develop intestinal tumors, the mice displayed fewer tumors than control mice. This SNP may thus play a causal role in human cancer, presumably through altered regulation of MYC. A human genetic variant, identified in genome-wide association studies as increasing cancer risk, alters tumorigenesis in mice. Multiple cancer-associated single-nucleotide polymorphisms (SNPs) have been mapped to conserved sequences within a 500-kilobase region upstream of the MYC oncogene on human chromosome 8q24. These SNPs may affect cancer development through altered regulation of MYC expression, but this hypothesis has been difficult to confirm. We generated mice deficient in Myc-335, a putative MYC regulatory element that contains rs6983267, a SNP accounting for more human cancer-related morbidity than any other genetic variant or mutation. In Myc-335 null mice, Myc transcripts were expressed in the intestinal crypts in a pattern similar to that in wild-type mice but at modestly reduced levels. The mutant mice displayed no overt phenotype but were markedly resistant to intestinal tumorigenesis induced by the APCmin mutation. These results establish that a cancer-associated SNP identified in human genome-wide association studies has a functional effect in vivo.


Nature Communications | 2014

The Glanville fritillary genome retains an ancient karyotype and reveals selective chromosomal fusions in Lepidoptera

Virpi Ahola; Rainer Lehtonen; Panu Somervuo; Leena Salmela; Patrik Koskinen; Pasi Rastas; Niko Välimäki; Lars Paulin; Jouni Kvist; Niklas Wahlberg; Jaakko Tanskanen; Emily A. Hornett; Laura Ferguson; Shiqi Luo; Zijuan Cao; Maaike de Jong; Anne Duplouy; Olli-Pekka Smolander; Heiko Vogel; Rajiv C. McCoy; Kui Qian; Wong Swee Chong; Qin Zhang; Freed Ahmad; Jani K. Haukka; Aruj Joshi; Jarkko Salojärvi; Christopher W. Wheat; Ewald Grosse-Wilde; Daniel C. Hughes

Previous studies have reported that chromosome synteny in Lepidoptera has been well conserved, yet the number of haploid chromosomes varies widely from 5 to 223. Here we report the genome (393 Mb) of the Glanville fritillary butterfly (Melitaea cinxia; Nymphalidae), a widely recognized model species in metapopulation biology and eco-evolutionary research, which has the putative ancestral karyotype of n=31. Using a phylogenetic analyses of Nymphalidae and of other Lepidoptera, combined with orthologue-level comparisons of chromosomes, we conclude that the ancestral lepidopteran karyotype has been n=31 for at least 140 My. We show that fusion chromosomes have retained the ancestral chromosome segments and very few rearrangements have occurred across the fusion sites. The same, shortest ancestral chromosomes have independently participated in fusion events in species with smaller karyotypes. The short chromosomes have higher rearrangement rate than long ones. These characteristics highlight distinctive features of the evolutionary dynamics of butterflies and moths.


Cell Reports | 2014

Uterine Leiomyoma-Linked MED12 Mutations Disrupt Mediator-Associated CDK Activity

Mikko P. Turunen; Jason M. Spaeth; Salla Keskitalo; Min Ju Park; Teemu Kivioja; Alison D. Clark; Netta Mäkinen; Fangjian Gao; Kimmo Palin; Helka Nurkkala; Anna Vähärautio; Mervi Aavikko; Kati Kämpjärvi; Pia Vahteristo; Chongwoo A. Kim; Lauri A. Aaltonen; Markku Varjosalo; Jussi Taipale; Thomas G. Boyer

Somatic mutations in exon 2 of the RNA polymerase II transcriptional Mediator subunit MED12 occur at very high frequency (∼70%) in uterine leiomyomas. However, the influence of these mutations on Mediator function and the molecular basis for their tumorigenic potential remain unknown. To clarify the impact of these mutations, we used affinity-purification mass spectrometry to establish the global protein-protein interaction profiles for both wild-type and mutant MED12. We found that uterine leiomyoma-linked mutations in MED12 led to a highly specific decrease in its association with Cyclin C-CDK8/CDK19 and loss of Mediator-associated CDK activity. Mechanistically, this occurs through disruption of a MED12-Cyclin C binding interface that we also show is required for MED12-mediated stimulation of Cyclin C-dependent CDK8 kinase activity. These findings indicate that uterine leiomyoma-linked mutations in MED12 uncouple Cyclin C-CDK8/19 from core Mediator and further identify the MED12/Cyclin C interface as a prospective therapeutic target in CDK8-driven cancers.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Integrated data analysis reveals uterine leiomyoma subtypes with distinct driver pathways and biomarkers.

Miika Mehine; Eevi Kaasinen; Hanna-Riikka Heinonen; Netta Mäkinen; Kati Kämpjärvi; Nanna Sarvilinna; Mervi Aavikko; Anna Vähärautio; Annukka Pasanen; Ralf Bützow; Oskari Heikinheimo; Jari Sjöberg; Esa Pitkänen; Pia Vahteristo; Lauri A. Aaltonen

Significance The clinical and scientific community widely regards uterine leiomyomas as a single entity, although evidence of genetic heterogeneity exists. The aim of this study was to explore transcriptional differences between leiomyomas harboring different genetic alterations, including high mobility group AT-hook 2 rearrangements, mediator complex subunit 12 mutations, biallelic inactivation of fumarate hydratase, and collagen, type IV, alpha 5-collagen, type IV, alpha 6 deletions. The evidence presented herein strongly suggests that specific driver mutations are the major determinants of expression changes in leiomyomas. Here we highlight subtype-specific expression differences in key driver pathways and emphasize the utility of stratification in leiomyoma research. Finally, we offer a set of candidate biomarkers that will facilitate the molecular classification of leiomyomas. Uterine leiomyomas are common benign smooth muscle tumors that impose a major burden on women’s health. Recent sequencing studies have revealed recurrent and mutually exclusive mutations in leiomyomas, suggesting the involvement of molecularly distinct pathways. In this study, we explored transcriptional differences among leiomyomas harboring different genetic drivers, including high mobility group AT-hook 2 (HMGA2) rearrangements, mediator complex subunit 12 (MED12) mutations, biallelic inactivation of fumarate hydratase (FH), and collagen, type IV, alpha 5 and collagen, type IV, alpha 6 (COL4A5-COL4A6) deletions. We also explored the transcriptional consequences of 7q22, 22q, and 1p deletions, aiming to identify possible target genes. We investigated 94 leiomyomas and 60 corresponding myometrial tissues using exon arrays, whole genome sequencing, and SNP arrays. This integrative approach revealed subtype-specific expression changes in key driver pathways, including Wnt/β-catenin, Prolactin, and insulin-like growth factor (IGF)1 signaling. Leiomyomas with HMGA2 aberrations displayed highly significant up-regulation of the proto-oncogene pleomorphic adenoma gene 1 (PLAG1), suggesting that HMGA2 promotes tumorigenesis through PLAG1 activation. This was supported by the identification of genetic PLAG1 alterations resulting in expression signatures as seen in leiomyomas with HMGA2 aberrations. RAD51 paralog B (RAD51B), the preferential translocation partner of HMGA2, was up-regulated in MED12 mutant lesions, suggesting a role for this gene in the genesis of leiomyomas. FH-deficient leiomyomas were uniquely characterized by activation of nuclear factor erythroid 2-related factor 2 (NRF2) target genes, supporting the hypothesis that accumulation of fumarate leads to activation of the oncogenic transcription factor NRF2. This study emphasizes the need for molecular stratification in leiomyoma research and possibly in clinical practice as well. Further research is needed to determine whether the candidate biomarkers presented herein can provide guidance for managing the millions of patients affected by these lesions.


G3: Genes, Genomes, Genetics | 2013

Transcriptional Networks Controlling the Cell Cycle

Martin Bonke; Mikko P. Turunen; Maria Sokolova; Anna Vähärautio; Teemu Kivioja; Minna Taipale; Mikael Björklund; Jussi Taipale

In this work, we map the transcriptional targets of 107 previously identified Drosophila genes whose loss caused the strongest cell-cycle phenotypes in a genome-wide RNA interference screen and mine the resulting data computationally. Besides confirming existing knowledge, the analysis revealed several regulatory systems, among which were two highly-specific and interconnected feedback circuits, one between the ribosome and the proteasome that controls overall protein homeostasis, and the other between the ribosome and Myc/Max that regulates the protein synthesis capacity of cells. We also identified a set of genes that alter the timing of mitosis without affecting gene expression, indicating that the cyclic transcriptional program that produces the components required for cell division can be partially uncoupled from the cell division process itself. These genes all have a function in a pathway that regulates the phosphorylation state of Cdk1. We provide evidence showing that this pathway is involved in regulation of cell size, indicating that a Cdk1-regulated cell size checkpoint exists in metazoans.


Science | 2014

Cancer by super-enhancer

Anna Vähärautio; Jussi Taipale

Tiny changes in our genomes can enhance oncogene expression and contribute to tumorigenesis [Also see Report by Mansour et al.] Most recurring somatic mutations in cancer affect protein-coding regions, either through activating oncogenes or inactivating tumor suppressors (1). However, several classes of mutations have been identified that affect the much larger noncoding regions of the genome, leading to changes in gene expression. These include large-scale genomic rearrangements that bring a strongly active promoter next to an oncogene, or place oncogene promoters under the influence of strong transcriptional enhancers (see the figure, panel A). On page 1373 of this issue, Mansour et al. (2) identify a very small mutation in ∼5% of T cell acute lymphoblastic leukemias (T-ALL; see the figure, panel B) that generates a large, powerful enhancer capable of driving tumorigenesis.


Cell Cycle | 2017

Genome-wide screen of cell-cycle regulators in normal and tumor cells identifies a differential response to nucleosome depletion

Maria Sokolova; Mikko P. Turunen; Oliver Mortusewicz; Teemu Kivioja; Patrick Herr; Anna Vähärautio; Mikael Björklund; Minna Taipale; Thomas Helleday; Jussi Taipale

ABSTRACT To identify cell cycle regulators that enable cancer cells to replicate DNA and divide in an unrestricted manner, we performed a parallel genome-wide RNAi screen in normal and cancer cell lines. In addition to many shared regulators, we found that tumor and normal cells are differentially sensitive to loss of the histone genes transcriptional regulator CASP8AP2. In cancer cells, loss of CASP8AP2 leads to a failure to synthesize sufficient amount of histones in the S-phase of the cell cycle, resulting in slowing of individual replication forks. Despite this, DNA replication fails to arrest, and tumor cells progress in an elongated S-phase that lasts several days, finally resulting in death of most of the affected cells. In contrast, depletion of CASP8AP2 in normal cells triggers a response that arrests viable cells in S-phase. The arrest is dependent on p53, and preceded by accumulation of markers of DNA damage, indicating that nucleosome depletion is sensed in normal cells via a DNA-damage -like response that is defective in tumor cells.


Nature Communications | 2018

Contribution of allelic imbalance to colorectal cancer

Kimmo Palin; Esa Pitkänen; Mikko P. Turunen; Biswajyoti Sahu; Päivi Pihlajamaa; Teemu Kivioja; Eevi Kaasinen; Niko Välimäki; Ulrika A. Hänninen; Tatiana Cajuso; Mervi Aavikko; Sari Tuupanen; Outi Kilpivaara; Linda van den Berg; Johanna Kondelin; Tomas Tanskanen; Riku Katainen; Marta Grau; Heli Rauanheimo; Roosa-Maria Plaketti; Aurora Taira; Päivi Sulo; Tuomo Hartonen; Kashyap Dave; Bernhard Schmierer; Sandeep Kumar Botla; Maria Sokolova; Anna Vähärautio; Kornelia Gladysz; Halit Ongen

Point mutations in cancer have been extensively studied but chromosomal gains and losses have been more challenging to interpret due to their unspecific nature. Here we examine high-resolution allelic imbalance (AI) landscape in 1699 colorectal cancers, 256 of which have been whole-genome sequenced (WGSed). The imbalances pinpoint 38 genes as plausible AI targets based on previous knowledge. Unbiased CRISPR-Cas9 knockout and activation screens identified in total 79 genes within AI peaks regulating cell growth. Genetic and functional data implicate loss of TP53 as a sufficient driver of AI. The WGS highlights an influence of copy number aberrations on the rate of detected somatic point mutations. Importantly, the data reveal several associations between AI target genes, suggesting a role for a network of lineage-determining transcription factors in colorectal tumorigenesis. Overall, the results unravel the contribution of AI in colorectal cancer and provide a plausible explanation why so few genes are commonly affected by point mutations in cancers.In this study the authors examine the allelic imbalance (AI) landscape of colorectal cancer, reporting loss of TP53 as a driver of AI. They use CRISPR-Cas9 screens to identify 79 genes (within AI regions) regulating cell growth and identify a network of transcription factors that may contribute to colorectal tumorigenesis.


Clinical Cancer Research | 2018

A functional homologous recombination assay predicts primary chemotherapy response and long-term survival in ovarian cancer patients

Manuela Tumiati; Sakari Hietanen; Johanna Hynninen; Elina Pietilä; Anniina Färkkilä; Katja Kaipio; Pia Roering; Kaisa Huhtinen; Amjad Alkodsi; Yilin Li; Rainer Lehtonen; Erdogan Pekcan Erkan; Minna Tuominen; Kaisa Lehti; Sampsa Hautaniemi; Anna Vähärautio; Seija Grénman; Olli Carpén; Liisa Kauppi

Purpose: Homologous recombination deficiency (HRD) correlates with platinum sensitivity in patients with ovarian cancer, which clinically is the most useful predictor of sensitivity to PARPi. To date, there are no reliable diagnostic tools to anticipate response to platinum-based chemotherapy, thus we aimed to develop an ex vivo functional HRD detection test that could predict both platinum-sensitivity and patient eligibility to targeted drug treatments. Experimental Design: We obtained a functional HR score by quantifying homologous recombination (HR) repair after ionizing radiation-induced DNA damage in primary ovarian cancer samples (n = 32). Samples clustered in 3 categories: HR-deficient, HR-low, and HR-proficient. We analyzed the HR score association with platinum sensitivity and treatment response, platinum-free interval (PFI) and overall survival (OS), and compared it with other clinical parameters. In parallel, we performed DNA-sequencing of HR genes to assess if functional HRD can be predicted by currently offered genetic screening. Results: Low HR scores predicted primary platinum sensitivity with high statistical significance (P = 0.0103), associated with longer PFI (HR-deficient vs. HR-proficient: 531 vs. 53 days), and significantly correlated with improved OS (HR score <35 vs. ≥35, hazard ratio = 0.08, P = 0.0116). At the genomic level, we identified a few unclear mutations in HR genes and the mutational signature associated with HRD, but, overall, genetic screening failed to predict functional HRD. Conclusions: We developed an ex vivo assay that detects tumor functional HRD and an HR score able to predict platinum sensitivity, which holds the clinically relevant potential to become the routine companion diagnostic in the management of patients with ovarian cancer. Clin Cancer Res; 24(18); 4482–93. ©2018 AACR.

Collaboration


Dive into the Anna Vähärautio's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge