Network


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

Hotspot


Dive into the research topics where Aparna Raval is active.

Publication


Featured researches published by Aparna Raval.


Cell | 2007

Downregulation of Death-Associated Protein Kinase 1 (DAPK1) in Chronic Lymphocytic Leukemia

Aparna Raval; Stephan M. Tanner; John C. Byrd; Elizabeth Angerman; James D. Perko; Shih Shih Chen; Björn Hackanson; Michael R. Grever; David M. Lucas; Jennifer J. Matkovic; Thomas S. Lin; Thomas J. Kipps; Fiona Murray; Dennis D. Weisenburger; Warren G. Sanger; Jane F. Lynch; Patrice Watson; Mary Jansen; Yuko Yoshinaga; Richard Rosenquist; Pieter J. de Jong; Penny Coggill; Stephan Beck; Henry T. Lynch; Albert de la Chapelle; Christoph Plass

Summary The heritability of B cell chronic lymphocytic leukemia (CLL) is relatively high; however, no predisposing mutation has been convincingly identified. We show that loss or reduced expression of death-associated protein kinase 1 (DAPK1) underlies cases of heritable predisposition to CLL and the majority of sporadic CLL. Epigenetic silencing of DAPK1 by promoter methylation occurs in almost all sporadic CLL cases. Furthermore, we defined a disease haplotype, which segregates with the CLL phenotype in a large family. DAPK1 expression of the CLL allele is downregulated by 75% in germline cells due to increased HOXB7 binding. In the blood cells from affected family members, promoter methylation results in additional loss of DAPK1 expression. Thus, reduced expression of DAPK1 can result from germline predisposition, as well as epigenetic or somatic events causing or contributing to the CLL phenotype.


Nature Genetics | 2005

Global assessment of promoter methylation in a mouse model of cancer identifies ID4 as a putative tumor-suppressor gene in human leukemia

Li Yu; Chunhui Liu; Jeff Vandeusen; Brian Becknell; Zunyan Dai; Yue Zhong Wu; Aparna Raval; Te Hui Liu; Wei Ding; Charlene Mao; Shujun Liu; Laura T. Smith; Stephen C. Lee; Laura Z. Rassenti; Guido Marcucci; John C. Byrd; Michael A. Caligiuri; Christoph Plass

DNA methylation is associated with malignant transformation, but limitations imposed by genetic variability, tumor heterogeneity, availability of paired normal tissues and methodologies for global assessment of DNA methylation have limited progress in understanding the extent of epigenetic events in the initiation and progression of human cancer and in identifying genes that undergo methylation during cancer. We developed a mouse model of T/natural killer acute lymphoblastic leukemia that is always preceded by polyclonal lymphocyte expansion to determine how aberrant promoter DNA methylation and consequent gene silencing might be contributing to leukemic transformation. We used restriction landmark genomic scanning with this mouse model of preleukemia reproducibly progressing to leukemia to show that specific genomic methylation is associated with only the leukemic phase and is not random. We also identified Idb4 as a putative tumor-suppressor gene that is methylated in most mouse and human leukemias but in only a minority of other human cancers.


Cancer Research | 2004

Epigenetic Profiling in Chronic Lymphocytic Leukemia Reveals Novel Methylation Targets

Laura J. Rush; Aparna Raval; Pauline Funchain; Amy J. Johnson; Lisa L. Smith; David M. Lucas; Melania Bembea; Te Hui Liu; Nyla A. Heerema; Laura Z. Rassenti; Sandya Liyanarachchi; Ramana V. Davuluri; John C. Byrd; Christoph Plass

CpG island methylation is an epigenetic alteration that contributes to tumorigenesis by transcriptional inactivation of genes. Little is known about the overall levels of CpG island methylation in chronic lymphocytic leukemia (CLL). To provide a baseline estimate of global aberrant methylation and identify target sequences for additional investigation, we performed Restriction Landmark Genomic Scanning on 10 CLL samples. Two methylation-sensitive landmark enzymes were used (NotI and AscI), allowing assessment of over 3000 CpG islands in each sample. Tumor-derived Restriction Landmark Genomic Scanning profiles were compared with profiles from CD19-selected B cells from normal volunteers and matched normal neutrophils from 4 CLL patients. We found 2.5–8.1% (mean 4.8%) of the CpG islands in CLL samples were aberrantly methylated compared with controls, and the methylation events had a nonrandom distribution (P < 0.0001). Furthermore, we identified 193 aberrantly methylated sequences, of which 93% have CpG island characteristics and 90% have homology to genes or expressed sequences. One such gene, the G protein-coupled metabotropic glutamate receptor 7 (GRM7), possibly inhibits cyclic AMP signaling in the induction of apoptosis. Bisulfite sequencing of GRM7 confirmed extensive CpG island methylation, and treatment with 5-aza-2′-deoxycytidine (decitabine) resulted in up-regulated expression of several genes in vitro with concurrent cellular depletion of DNMT1 protein. Our dual-enzyme global methylation study shows that CLL is characterized by widespread nonrandom CpG island methylation similar to other tumors and provides a panel of novel methylation targets that can be used in larger studies designed to assess impact on disease progression and survival.


Journal of Clinical Oncology | 2005

TWIST2 Demonstrates Differential Methylation in Immunoglobulin Variable Heavy Chain Mutated and Unmutated Chronic Lymphocytic Leukemia

Aparna Raval; David M. Lucas; Jennifer J. Matkovic; Kristi L. Bennett; Sandya Liyanarachchi; Donn C. Young; Laura Z. Rassenti; Thomas J. Kipps; Michael R. Grever; John C. Byrd; Christoph Plass

PURPOSE Chronic lymphocytic leukemia (CLL) is a clinically heterogeneous disease for which natural history can be predicted based on the presence or absence of immunoglobulin (Ig) variable heavy chain (V(H)) gene mutations. Herein we report selective epigenetic silencing of the transcription factor TWIST2 (DERMO1) in Ig V(H) mutated CLL and describe a semiquantitative assay to study promoter methylation of this gene in primary tumor cells. MATERIALS AND METHODS TWIST2 promoter methylation was identified by restriction landmark genome scanning. Southern blot (SB), bisulfite sequencing, and combined bisulfite restriction analysis (COBRA), and quantitative SB-COBRA was performed to study methylation of the TWIST2 promoter. Reverse transcription polymerase chain reaction assays were used to study TWIST2 expression in CLL cells. RESULTS Following identification and confirmation of TWIST2 methylation in CLL patients, we demonstrated that expression of this transcription factor is related to the degree of promoter methylation. Expression of TWIST2 in a CLL cell line in which the promoter is methylated was increased following decitabine treatment. We next studied 53 patients by COBRA and demonstrated that 72% of patient samples with mutated Ig V(H) show TWIST2 methylation, while only 16% of patient samples with unmutated Ig V(H) were methylated (P < .001). In a subset of patients, methylation of TWIST2 correlated with mRNA expression. CONCLUSION TWIST2 is differentially methylated in CLL cells relative to Ig V(H) mutational status and can be quantitatively monitored by SB-COBRA. Based on the known role of TWIST2 in silencing p53 function in other malignancies, future studies should focus on the role of TWIST2 in CLL and related lymphoproliferative diseases.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Epigenetic changes during disease progression in a murine model of human chronic lymphocytic leukemia

Shih-Shih Chen; Aparna Raval; Amy J. Johnson; Erin Hertlein; Te-Hui Liu; Victor X. Jin; Mara H. Sherman; Shujun Liu; David W. Dawson; Katie Williams; Mark C. Lanasa; Sandya Liyanarachchi; Thomas S. Lin; Guido Marcucci; Yuri Pekarsky; Ramana V. Davuluri; Carlo M. Croce; Denis C. Guttridge; Michael A. Teitell; John C. Byrd; Christoph Plass

Epigenetic alterations, including gain or loss of DNA methylation, are a hallmark of nearly every malignancy. Changes in DNA methylation can impact expression of cancer-related genes including apoptosis regulators and tumor suppressors. Because such epigenetic changes are reversible, they are being aggressively investigated as potential therapeutic targets. Here we use the Eμ-TCL1 transgenic mouse model of chronic lymphocytic leukemia (CLL) to determine the timing and patterns of aberrant DNA methylation, and to investigate the mechanisms that lead to aberrant DNA methylation. We show that CLL cells from Eμ-TCL1 mice at various stages recapitulate epigenetic alterations seen in human CLL. Aberrant methylation of promoter sequences is observed as early as 3 months of age in these animals, well before disease onset. Abnormally methylated promoter regions include binding sites for the transcription factor FOXD3. We show that loss of Foxd3 expression due to an NF-κB p50/p50:HDAC1 repressor complex occurs in TCL1-positive B cells before methylation. Therefore, specific transcriptional repression is an early event leading to epigenetic silencing of target genes in murine and human CLL. These results provide strong rationale for the development of strategies to target NF-κB components in CLL and potentially other B-cell malignancies.


Nucleic Acids Research | 2006

Accurate quantification of DNA methylation using combined bisulfite restriction analysis coupled with the Agilent 2100 Bioanalyzer platform

Romulo M. Brena; Herbert Auer; Karl Kornacker; Björn Hackanson; Aparna Raval; John C. Byrd; Christoph Plass

DNA methylation is the best-studied epigenetic modification and describes the conversion of cytosine to 5-methylcytosine. The importance of this phenomenon is that aberrant promoter hypermethylation is a common occurrence in cancer and is frequently associated with gene silencing. Various techniques are currently available for the analysis of DNA methylation. However, accurate and reproducible quantification of DNA methylation remains challenging. In this report, we describe Bio-COBRA (combined bisulfite restriction analysis coupled with the Agilent 2100 Bioanalyzer platform), as a novel approach to quantitative DNA methylation analysis. The combination of a well-established method, COBRA, which interrogates DNA methylation via the restriction enzyme analysis of PCR-amplified bisulfite treated DNAs, with the Bioanalyzer platform allows for the rapid and quantitative assessment of DNA methylation patterns in large sample sets. The sensitivity and reproducibility of Bio-COBRA make it a valuable tool for the analysis of DNA methylation in clinical samples, which could aid in the development of diagnostic and prognostic parameters with respect to disease detection and management.


PLOS Genetics | 2005

CpG island methylation in a mouse model of lymphoma is driven by the genetic configuration of tumor cells

Rene Opavsky; Shu Huei Wang; Prashant Trikha; Aparna Raval; Yuan Huang; Yue-Zhong Wu; Benjamin Rodriguez; Benjamin Keller; Sandya Liyanarachchi; Guo Wei; Ramana V. Davuluri; Michael Weinstein; Dean W. Felsher; Michael C. Ostrowski; Gustavo Leone; Christoph Plass

Hypermethylation of CpG islands is a common epigenetic alteration associated with cancer. Global patterns of hypermethylation are tumor-type specific and nonrandom. The biological significance and the underlying mechanisms of tumor-specific aberrant promoter methylation remain unclear, but some evidence suggests that this specificity involves differential sequence susceptibilities, the targeting of DNA methylation activity to specific promoter sequences, or the selection of rare DNA methylation events during disease progression. Using restriction landmark genomic scanning on samples derived from tissue culture and in vivo models of T cell lymphomas, we found that MYC overexpression gave rise to a specific signature of CpG island hypermethylation. This signature reflected gene transcription profiles and was detected only in advanced stages of disease. The further inactivation of the Pten, p53, and E2f2 tumor suppressors in MYC-induced lymphomas resulted in distinct and diagnostic CpG island methylation signatures. Our data suggest that tumor-specific DNA methylation in lymphomas arises as a result of the selection of rare DNA methylation events during the course of tumor development. This selection appears to be driven by the genetic configuration of tumor cells, providing experimental evidence for a causal role of DNA hypermethylation in tumor progression and an explanation for the tremendous epigenetic heterogeneity observed in the evolution of human cancers. The ability to predict genome-wide epigenetic silencing based on relatively few genetic alterations will allow for a more complete classification of tumors and understanding of tumor cell biology.


Oncogene | 2004

Bone morphogenetic protein 3B silencing in non-small-cell lung cancer

Zunyan Dai; Anthony P. Popkie; Wei Guo Zhu; Cynthia Timmers; Aparna Raval; Sarah Tannehill-Gregg; Carl Morrison; Herbert Auer; Robert A. Kratzke; Gloria A. Niehans; Stefan Amatschek; Wolfgang Sommergruber; Gustavo Leone; Thomas J. Rosol; Gregory A. Otterson; Christoph Plass

Bone morphogenetic protein 3B (BMP3B) is a member of the TGF-β superfamily. The BMP3B promoter sequence was previously identified as a target for aberrant DNA methylation in non-small-cell lung cancer (NSCLC). Aberrant DNA hypermethylation in the BMP3B promoter is associated with downregulation of BMP3B transcription in both primary human lung cancers as well as lung cancer cell lines. In order to understand the mechanisms of BMP3B silencing in lung cancer, a sample set of 91 primary NSCLCs was used to detect aberrant BMP3B promoter methylation, mutations in the coding sequence of BMP3B, and loss of heterozygosity (LOH). Our results showed that 45 of 91 (or 49.5%) tested primary NSCLCs exhibited increased promoter methylation, and 40% demonstrated LOH in at least one of the flanking microsatellite markers sJRH and D10S196 (63 kb upstream or 3.338 Mbp downstream of BMP3B). The lung cancer cell line A549, a type II alveolar epithelial human lung cancer cell line, is characterized by aberrant DNA promoter methylation. We used retroviral vector constructs containing the BMP3B cDNA to re-express the gene in A549 cells and to investigate the effects on cell growth. No change in the cell growth rate was observed after BMP3B re-expression, as compared to the vector controls. Although the number of colonies formed in anchorage-dependent assays was only slightly decreased, the colony-forming ability of A549 cells after BMP3B expression in anchorage-independent assays in soft agar was significantly reduced to 10% (P<0.005, t-test). Moreover, the in vivo tumorigenicity assay in nude mice indicated that cells re-expressing BMP3B grew significantly slower than cells not expressing BMP3B (P<0.05, t-test). In conclusion, this study provides evidence that BMP3B expression is repressed by different mechanisms in lung cancer, and that the silencing of BMP3B promotes lung tumor development.


British Journal of Haematology | 2007

Molecular profiling of chronic lymphocytic leukaemia : genetics meets epigenetics to identify predisposing genes

Christoph Plass; John C. Byrd; Aparna Raval; Stephan M. Tanner; Albert de la Chapelle

Molecular profiling may lead to a better understanding of a disease. This knowledge is especially important in malignancies, where multiple alterations are required during the progression from premalignant to malignant stages. Such information can be useful for the development of novel biomarkers that allow the prediction of a clinical course, response to treatment or early detection. Molecular data is also utilized to develop targeted therapies. Moreover, gene defects identified in profiling studies will help to understand the molecular pathways disrupted in the disease. This review provides an overview of molecular profiling approaches in chronic lymphocytic leukaemia (CLL). We will describe our current understanding of genetic alterations in CLL, the use of familial CLL for the identification of predisposing mutations, and the search for epigenetic alterations in CLL.


Hematological Oncology | 2011

Impact of TET2 mutations on mRNA expression and clinical outcomes in MDS patients treated with DNA methyltransferase inhibitors

Daniel A. Pollyea; Aparna Raval; Brenda Kusler; Jason Gotlib; Ash A. Alizadeh; Beverly S. Mitchell

Despite a common lineage derivation, the clonal myeloid disorders acute myeloid leukemia (AML), myelodysplastic syndrome (MDS) and myeloproliferative neoplasms (MPN) are phenotypically distinct, with few known shared recurrent genetic abnormalities. The recent recognition that the ten-eleven translocation 2 (TET2) gene located on the long arm of chromosome four is mutated with high frequency across all myeloid malignancies is therefore a compelling unifying feature [1]. As many as 30% of MDS patients have a variety of TET2 abnormalities including deletions, insertions and nonsense or missense mutations throughout the nine coding exons of the 150 kB gene, many of which are predicted to result in truncated translation [2]. Based on analyses from a small number of patient samples, TET2 mutations may confer a favourable outcome in MDS patients [3], but a worse outcome in those with chronic myelomonocytic leukemia (CMML) [4] and AML [5]. TET2 belongs to a three-member family that also includes TET1 and TET3; TET1 was originally identified as a partner for the MLL gene within t(10;11)(p12;q23) translocations in AML [6]. All three paralogs share two highly conserved regions, BOX1 and BOX2 (Figure 1A). A highly homologous catalytic domain shared among the three proteins catalyzes the conversion of 5-methylcytosine to 5-hydroxymethylcytosine in TET1 [7], which could epigenetically regulate gene expression by altering methylation-driven gene silencing. Given its homology to TET1, TET2 is hypothesized to act as a tumour suppressor gene (TSG) by similarly regulating DNA methylation and epigenetic control of gene expression at critical loci important for myelopoeisis and leukemogenesis [8]. The DNA methyltransferase inhibitors (DNMTIs) 5-azacytidine and decitabine, approved agents with activity in MDS, are incorporated into DNA and/or RNA, inhibiting DNA methyltransferase and preventing promoter methylation and TSG silencing. The aim of this study was to determine whether the presence of TET2 mutations is associated with prognosis in MDS patients treated with DNMTIs. Bone marrow aspirates from 12 patients with MDS who received DNMTIs and had documented clinical follow up were obtained with informed consent in accordance with the Declaration of Helsinki and approval by the Institutional Review Board at the Stanford University School of Medicine (SUMC). DNA and RNA were extracted using a Qiagen AllPrep DNA/RNA Mini Kit (Qiagen, Valencia, CA). All patients were diagnosed according to World Health Organization criteria and confirmed by the clinical pathology department at SUMC.

Collaboration


Dive into the Aparna Raval'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

Li Yu

Ohio State University

View shared research outputs
Researchain Logo
Decentralizing Knowledge