Network


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

Hotspot


Dive into the research topics where Jana Opavska is active.

Publication


Featured researches published by Jana Opavska.


Nature | 2008

Mouse development with a single E2F activator

Shih-Yin Tsai; Rene Opavsky; Nidhi Sharma; Lizhao Wu; Shan Naidu; Eric Nolan; Enrique Feria-Arias; Cynthia Timmers; Jana Opavska; Alain de Bruin; Jean Leon Chong; Prashant Trikha; Soledad Fernandez; Paul C. Stromberg; Thomas J. Rosol; Gustavo Leone

The E2F family is conserved from Caenorhabditis elegans to mammals, with some family members having transcription activation functions and others having repressor functions. Whereas C. elegans and Drosophila melanogaster have a single E2F activator protein and repressor protein, mammals have at least three activator and five repressor proteins. Why such genetic complexity evolved in mammals is not known. To begin to evaluate this genetic complexity, we targeted the inactivation of the entire subset of activators, E2f1, E2f2, E2f3a and E2f3b, singly or in combination in mice. We demonstrate that E2f3a is sufficient to support mouse embryonic and postnatal development. Remarkably, expression of E2f3b or E2f1 from the E2f3a locus (E2f3a3bki or E2f3a1ki, respectively) suppressed all the postnatal phenotypes associated with the inactivation of E2f3a. We conclude that there is significant functional redundancy among activators and that the specific requirement for E2f3a during postnatal development is dictated by regulatory sequences governing its selective spatiotemporal expression and not by its intrinsic protein functions. These findings provide a molecular basis for the observed specificity among E2F activators during development.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Specific tumor suppressor function for E2F2 in Myc-induced T cell lymphomagenesis

Rene Opavsky; Shih-Yin Tsai; Martin Guimond; Anjulie Arora; Jana Opavska; Brian Becknell; Michael Kaufmann; Nathaniel A. Walton; Julie A. Stephens; Soledad Fernandez; Natarajan Muthusamy; Dean W. Felsher; Pierluigi Porcu; Michael A. Caligiuri; Gustavo Leone

Deregulation of the Myc pathway and deregulation of the Rb pathway are two of the most common abnormalities in human malignancies. Recent in vitro experiments suggest a complex cross-regulatory relationship between Myc and Rb that is mediated through the control of E2F. To evaluate the functional connection between Myc and E2Fs in vivo, we used a bitransgenic mouse model of Myc-induced T cell lymphomagenesis and analyzed tumor progression in mice deficient for E2f1, E2f2, or E2f3. Whereas the targeted inactivation of E2f1 or E2f3 had no significant effect on tumor progression, loss of E2f2 accelerated lymphomagenesis. Interestingly, loss of a single copy of E2f2 also accelerated tumorigenesis, albeit to a lesser extent, suggesting a haploinsufficient function for this locus. The combined ablation of E2f1 or E2f3, along with E2f2, did not further accelerate tumorigenesis. Myc-overexpressing T cells were more resistant to apoptosis in the absence of E2f2, and the reintroduction of E2F2 into these tumor cells resulted in an increase of apoptosis and inhibition of tumorigenesis. These results identify the E2f2 locus as a tumor suppressor through its ability to modulate apoptosis.


Journal of Clinical Investigation | 2012

Loss of Dnmt3b function upregulates the tumor modifier Ment and accelerates mouse lymphomagenesis

Ryan A. Hlady; Slavomira Novakova; Jana Opavska; David Klinkebiel; Staci L. Peters; Juraj Bies; Jay Hannah; Javeed Iqbal; Kristi M. Anderson; Hollie M. Siebler; Lynette M. Smith; Timothy C. Greiner; Dhundy Bastola; Shantaram S. Joshi; Oksana Lockridge; Melanie A. Simpson; Dean W. Felsher; Kay Uwe Wagner; Wing C. Chan; Judith K. Christman; Rene Opavsky

DNA methyltransferase 3B (Dnmt3b) belongs to a family of enzymes responsible for methylation of cytosine residues in mammals. DNA methylation contributes to the epigenetic control of gene transcription and is deregulated in virtually all human tumors. To better understand the generation of cancer-specific methylation patterns, we genetically inactivated Dnmt3b in a mouse model of MYC-induced lymphomagenesis. Ablation of Dnmt3b function using a conditional knockout in T cells accelerated lymphomagenesis by increasing cellular proliferation, which suggests that Dnmt3b functions as a tumor suppressor. Global methylation profiling revealed numerous gene promoters as potential targets of Dnmt3b activity, the majority of which were demethylated in Dnmt3b-/- lymphomas, but not in Dnmt3b-/- pretumor thymocytes, implicating Dnmt3b in maintenance of cytosine methylation in cancer. Functional analysis identified the gene Gm128 (which we termed herein methylated in normal thymocytes [Ment]) as a target of Dnmt3b activity. We found that Ment was gradually demethylated and overexpressed during tumor progression in Dnmt3b-/- lymphomas. Similarly, MENT was overexpressed in 67% of human lymphomas, and its transcription inversely correlated with methylation and levels of DNMT3B. Importantly, knockdown of Ment inhibited growth of mouse and human cells, whereas overexpression of Ment provided Dnmt3b+/+ cells with a proliferative advantage. Our findings identify Ment as an enhancer of lymphomagenesis that contributes to the tumor suppressor function of Dnmt3b and suggest it could be a potential target for anticancer therapies.


Leukemia | 2014

Tumor suppressor functions of Dnmt3a and Dnmt3b in the prevention of malignant mouse lymphopoiesis

Staci L. Peters; Ryan A. Hlady; Jana Opavska; David Klinkebiel; Samuel J. Pirruccello; G A Talmon; J G Sharp; Lizhao Wu; R Jaenisch; Melanie A. Simpson; Adam R. Karpf; Rene Opavsky

Tumor suppressor functions of Dnmt3a and Dnmt3b in the prevention of malignant mouse lymphopoiesis


Molecular and Cellular Biology | 2013

Essential Role for Dnmt1 in the Prevention and Maintenance of MYC-Induced T-Cell Lymphomas

Staci L. Peters; Ryan A. Hlady; Jana Opavska; David Klinkebiel; Slavomira Novakova; Lynette M. Smith; Robert E. Lewis; Adam R. Karpf; Melanie A. Simpson; Lizhao Wu; Rene Opavsky

ABSTRACT DNA cytosine methylation is an epigenetic modification involved in the transcriptional repression of genes controlling a variety of physiological processes, including hematopoiesis. DNA methyltransferase 1 (Dnmt1) is a key enzyme involved in the somatic inheritance of DNA methylation and thus plays a critical role in epigenomic stability. Aberrant methylation contributes to the pathogenesis of human cancer and of hematologic malignancies in particular. To gain deeper insight into the function of Dnmt1 in lymphoid malignancies, we genetically inactivated Dnmt1 in a mouse model of MYC-induced T-cell lymphomagenesis. We show that loss of Dnmt1 delays lymphomagenesis by suppressing normal hematopoiesis and impairing tumor cell proliferation. Acute inactivation of Dnmt1 in primary lymphoma cells rapidly induced apoptosis, indicating that Dnmt1 is required to sustain T-cell lymphomas. Using high-resolution genome-wide profiling, we identified differentially methylated regions between control and Dnmt1-deficient lymphomas, demonstrating a locus-specific function for Dnmt1 in both maintenance and de novo promoter methylation. Dnmt1 activity is independent of the presence of Dnmt3a or Dnmt3b in de novo promoter methylation of the H2-Ab1 gene. Collectively, these data show for the first time that Dnmt1 is critical for the prevention and maintenance of T-cell lymphomas and contributes to aberrant methylation by both de novo and maintenance methylation.


Oncogene | 2015

Methylation-independent repression of Dnmt3b contributes to oncogenic activity of Dnmt3a in mouse MYC-induced T-cell lymphomagenesis

Staci L. Haney; Ryan A. Hlady; Jana Opavska; David Klinkebiel; Samuel J. Pirruccello; Samikshan Dutta; Kaustubh Datta; Melanie A. Simpson; Lizhao Wu; Rene Opavsky

DNA methyltransferase 3A (DNMT3A) catalyzes cytosine methylation of mammalian genomic DNA. In addition to myeloid malignancies, mutations in DNMT3A have been recently reported in T-cell lymphoma and leukemia, implying a possible involvement in the pathogenesis of human diseases. However, the role of Dnmt3a in T-cell transformation in vivo is poorly understood. Here we analyzed the functional consequences of Dnmt3a inactivation in a mouse model of MYC-induced T-cell lymphomagenesis (MTCL). Loss of Dnmt3a delayed tumorigenesis by suppressing cellular proliferation during disease progression. Gene expression profiling and pathway analysis identified upregulation of 17 putative tumor suppressor genes, including DNA methyltransferase Dnmt3b, in Dnmt3a-deficient lymphomas as molecular events potentially responsible for the delayed lymphomagenesis in Dnmt3aΔ/Δ mice. Interestingly, promoter and gene body methylation of these genes was not substantially changed between control and Dnmt3a-deficient lymphomas, suggesting that Dnmt3a may inhibit their expression in a methylation-independent manner. Re-expression of both wild type and catalytically inactive Dnmt3a in Dnmt3aΔ/Δ lymphoma cells in vitro inhibited Dnmt3b expression, indicating that Dnmt3b upregulation may be directly repressed by Dnmt3a. Importantly, genetic inactivation of Dnmt3b accelerated lymphomagenesis in Dnmt3aΔ/Δ mice, demonstrating that upregulation of Dnmt3b is a relevant molecular change in Dnmt3a-deficient lymphomas that inhibits disease progression. Collectively, our data demonstrate an unexpected oncogenic role for Dnmt3a in MTCL through methylation-independent repression of Dnmt3b and possibly other tumor suppressor genes.


PLOS Genetics | 2016

Dnmt3a Is a Haploinsufficient Tumor Suppressor in CD8+ Peripheral T Cell Lymphoma

Staci L. Haney; G. Michael Upchurch; Jana Opavska; David Klinkebiel; Ryan A. Hlady; Sohini Roy; Samikshan Dutta; Kaustubh Datta; Rene Opavsky

DNA methyltransferase 3A (DNMT3A) is an enzyme involved in DNA methylation that is frequently mutated in human hematologic malignancies. We have previously shown that inactivation of Dnmt3a in hematopoietic cells results in chronic lymphocytic leukemia in mice. Here we show that 12% of Dnmt3a-deficient mice develop CD8+ mature peripheral T cell lymphomas (PTCL) and 29% of mice are affected by both diseases. 10% of Dnmt3a+/- mice develop lymphomas, suggesting that Dnmt3a is a haploinsufficient tumor suppressor in PTCL. DNA methylation was deregulated genome-wide with 10-fold more hypo- than hypermethylated promoters and enhancers, demonstrating that hypomethylation is a major event in the development of PTCL. Hypomethylated promoters were enriched for binding sites of transcription factors AML1, NF-κB and OCT1, implying the transcription factors potential involvement in Dnmt3a-associated methylation. Whereas 71 hypomethylated genes showed an increased expression in PTCL, only 3 hypermethylated genes were silenced, suggesting that cancer-specific hypomethylation has broader effects on the transcriptome of cancer cells than hypermethylation. Interestingly, transcriptomes of Dnmt3a+/- and Dnmt3aΔ/Δ lymphomas were largely conserved and significantly overlapped with those of human tumors. Importantly, we observed downregulation of tumor suppressor p53 in Dnmt3a+/- and Dnmt3aΔ/Δ lymphomas as well as in pre-tumor thymocytes from 9 months old but not 6 weeks old Dnmt3a+/- tumor-free mice, suggesting that p53 downregulation is chronologically an intermediate event in tumorigenesis. Decrease in p53 is likely an important event in tumorigenesis because its overexpression inhibited proliferation in mouse PTCL cell lines, suggesting that low levels of p53 are important for tumor maintenance. Altogether, our data link the haploinsufficient tumor suppressor function of Dnmt3a in the prevention of mouse mature CD8+ PTCL indirectly to a bona fide tumor suppressor of T cell malignancies p53.


Scientific Reports | 2016

Loss of Dnmt3a induces CLL and PTCL with distinct methylomes and transcriptomes in mice.

Staci L. Haney; Garland Michael Upchurch; Jana Opavska; David Klinkebiel; Adams Kusi Appiah; Lynette M. Smith; Tayla Heavican; Javeed Iqbal; Shantaram S. Joshi; Rene Opavsky

Cytosine methylation of DNA is an epigenetic modification involved in the repression of genes that affect biological processes including hematopoiesis. It is catalyzed by DNA methyltransferases, one of which -DNMT3A- is frequently mutated in human hematologic malignancies. We have previously reported that Dnmt3a inactivation in hematopoietic stem cells results in chronic lymphocytic leukemia (CLL) and CD8-positive peripheral T cell lymphomas (PTCL) in EμSRα-tTA;Teto-Cre;Dnmt3afl/fl; Rosa26LOXPEGFP/EGFP (Dnmt3aΔ/Δ) mice. The extent to which molecular changes overlap between these diseases is not clear. Using high resolution global methylation and expression analysis we show that whereas patterns of methylation and transcription in normal B-1a cells and CD8-positive T cells are similar, methylomes and transcriptomes in malignant B-1a and CD8+ T cells are remarkably distinct, suggesting a cell-type specific function for Dnmt3a in cellular transformation. Promoter hypomethylation in tumors was 10 times more frequent than hypermethylation, three times more frequent in CLL than PTCL and correlated better with gene expression than hypermethylation. Cross-species molecular comparison of mouse and human CLL and PTCL reveals significant overlaps and identifies putative oncogenic drivers of disease. Thus, Dnmt3aΔ/Δ mice can serve as a new mouse model to study CLL and PTCL in relevant physiological settings.


Nature | 2003

Extra-embryonic function of Rb is essential for embryonic development and viability

Lizhao Wu; Alain de Bruin; Harold I. Saavedra; Maja Starovic; Anthony J. Trimboli; Ying Yang; Jana Opavska; Pamela Wilson; John C. Thompson; Michael C. Ostrowski; Thomas J. Rosol; Laura A. Woollett; Michael Weinstein; James C. Cross; Michael L. Robinson; Gustavo Leone


Cell Reports | 2016

Promoter Hypomethylation and Expression Is Conserved in Mouse Chronic Lymphocytic Leukemia Induced by Decreased or Inactivated Dnmt3a

Staci L. Haney; G. Michael Upchurch; Jana Opavska; David Klinkebiel; Ryan A. Hlady; Abhinav Suresh; Samuel J. Pirruccello; Vipul Shukla; Runqing Lu; Stefan Costinean; Angie Rizzino; Adam R. Karpf; Shantaram S. Joshi; Patrick C. Swanson; Rene Opavsky

Collaboration


Dive into the Jana Opavska's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

David Klinkebiel

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lizhao Wu

Ohio State University

View shared research outputs
Top Co-Authors

Avatar

Melanie A. Simpson

University of Nebraska–Lincoln

View shared research outputs
Top Co-Authors

Avatar

Adam R. Karpf

Eppley Institute for Research in Cancer and Allied Diseases

View shared research outputs
Top Co-Authors

Avatar

Lynette M. Smith

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Staci L. Haney

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Samuel J. Pirruccello

University of Nebraska Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge