34 results on '"Yongkai Mo"'
Search Results
2. Widespread hypomethylation occurs early and synergizes with gene amplification during esophageal carcinogenesis.
- Author
-
Hector Alvarez, Joanna Opalinska, Li Zhou, Davendra Sohal, Melissa J Fazzari, Yiting Yu, Christina Montagna, Elizabeth A Montgomery, Marcia Canto, Kerry B Dunbar, Jean Wang, Juan Carlos Roa, Yongkai Mo, Tushar Bhagat, K H Ramesh, Linda Cannizzaro, J Mollenhauer, Reid F Thompson, Masako Suzuki, Stephen J Meltzer, Ari Melnick, John M Greally, Anirban Maitra, and Amit Verma
- Subjects
Genetics ,QH426-470 - Abstract
Although a combination of genomic and epigenetic alterations are implicated in the multistep transformation of normal squamous esophageal epithelium to Barrett esophagus, dysplasia, and adenocarcinoma, the combinatorial effect of these changes is unknown. By integrating genome-wide DNA methylation, copy number, and transcriptomic datasets obtained from endoscopic biopsies of neoplastic progression within the same individual, we are uniquely able to define the molecular events associated progression of Barrett esophagus. We find that the previously reported global hypomethylation phenomenon in cancer has its origins at the earliest stages of epithelial carcinogenesis. Promoter hypomethylation synergizes with gene amplification and leads to significant upregulation of a chr4q21 chemokine cluster and other transcripts during Barrett neoplasia. In contrast, gene-specific hypermethylation is observed at a restricted number of loci and, in combination with hemi-allelic deletions, leads to downregulatation of selected transcripts during multistep progression. We also observe that epigenetic regulation during epithelial carcinogenesis is not restricted to traditionally defined "CpG islands," but may also occur through a mechanism of differential methylation outside of these regions. Finally, validation of novel upregulated targets (CXCL1 and 3, GATA6, and DMBT1) in a larger independent panel of samples confirms the utility of integrative analysis in cancer biomarker discovery.
- Published
- 2011
- Full Text
- View/download PDF
3. Meta-analysis of microarray studies reveals a novel hematopoietic progenitor cell signature and demonstrates feasibility of inter-platform data integration.
- Author
-
Davendra Sohal, Andrew Yeatts, Kenny Ye, Andrea Pellagatti, Li Zhou, Perry Pahanish, Yongkai Mo, Tushar Bhagat, John Mariadason, Jacqueline Boultwood, Ari Melnick, John Greally, and Amit Verma
- Subjects
Medicine ,Science - Abstract
Microarray-based studies of global gene expression (GE) have resulted in a large amount of data that can be mined for further insights into disease and physiology. Meta-analysis of these data is hampered by technical limitations due to many different platforms, gene annotations and probes used in different studies. We tested the feasibility of conducting a meta-analysis of GE studies to determine a transcriptional signature of hematopoietic progenitor and stem cells. Data from studies that used normal bone marrow-derived hematopoietic progenitors was integrated using both RefSeq and UniGene identifiers. We observed that in spite of variability introduced by experimental conditions and different microarray platforms, our meta-analytical approach can distinguish biologically distinct normal tissues by clustering them based on their cell of origin. When studied in terms of disease states, GE studies of leukemias and myelodysplasia progenitors tend to cluster with normal progenitors and remain distinct from other normal tissues, further validating the discriminatory power of this meta-analysis. Furthermore, analysis of 57 normal hematopoietic stem and progenitor cell GE samples was used to determine a gene expression signature characteristic of these cells. Genes that were most uniformly expressed in progenitors and at the same time differentially expressed when compared to other normal tissues were found to be involved in important biological processes such as cell cycle regulation and hematopoiesis. Validation studies using a different microarray platform demonstrated the enrichment of several genes such as SMARCE, Septin 6 and others not previously implicated in hematopoiesis. Most interestingly, alpha-integrin, the only common stemness gene discovered in a recent comparative murine analysis (Science 302(5644):393) was also enriched in our dataset, demonstrating the usefulness of this analytical approach.
- Published
- 2008
- Full Text
- View/download PDF
4. Supplementary Figures 1-4 from Methylome Profiling Reveals Distinct Alterations in Phenotypic and Mutational Subgroups of Myeloproliferative Neoplasms
- Author
-
Amit Verma, Alison Moliterno, Ulrich Steidl, John M. Greally, Ari M. Melnick, Jaroslaw P. Maciejewski, Michael McDevitt, Animesh Pardanani, Masako Suzuki, Yongkai Mo, Britta Will, Davendra Sohal, Tushar D. Bhagat, Li Zhou, Yiting Yu, Maximilian Christopeit, Sanchari Bhattacharyya, and Sangeeta Nischal
- Abstract
PDF file - 646K, Supp Fig 1: Pathways involved by hypermethylated genes in PV / ET. Supp Fig 2: Validation of HELP assay findings by MassArray bisulfite analysis Supp Fig 3: Pathways involved by genes that are overexpressed and hypomethylated at promoters by the Jak2V617F mutant. Supp Fig 4: Jak2V617F mutation does not have significant effects on global DNA methylation profiles
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Tables 1-7 from Methylome Profiling Reveals Distinct Alterations in Phenotypic and Mutational Subgroups of Myeloproliferative Neoplasms
- Author
-
Amit Verma, Alison Moliterno, Ulrich Steidl, John M. Greally, Ari M. Melnick, Jaroslaw P. Maciejewski, Michael McDevitt, Animesh Pardanani, Masako Suzuki, Yongkai Mo, Britta Will, Davendra Sohal, Tushar D. Bhagat, Li Zhou, Yiting Yu, Maximilian Christopeit, Sanchari Bhattacharyya, and Sangeeta Nischal
- Abstract
PDF file - 846K, Supplementary Table 1: Genes hypermethylated in PV / ET Supplementary Table 2: Genes hypermethylated in PMF Supplementary Table 3: Genes hypomethylated in PMF Supplementary Table 4: Genes hypermethylated in PMF cases with ASXL1 mutations / deletions Supp Table 5: Biological pathways affected by hypermethylated genes in ASXL1 mutated/deleted cases of PMF Supplementary Table 6: Genes hypermethylated in MPN cases with TET2 mutations Supp Table 7: Biological pathways affected by hypermethylated genes in TET2 mutated cases of MPNs
- Published
- 2023
- Full Text
- View/download PDF
6. The RUNX1/IL-34/CSF-1R axis is an autocrinally regulated modulator of resistance to BRAF-V600E inhibition in melanoma
- Author
-
Emily Bernstein, Hoa Nguyen, Gaston Habets, Gregg B. Fields, Chao Zhang, Tushar D. Bhagat, Elizabeth A. Burton, Yiting Yu, Evripidis Gavathiotis, Amit Verma, Veronika Polishchuk, Chiara Vardabasso, John M. Greally, Kith Pradhan, Paraic A. Kenny, Douglas B. Johnson, Matthias Bartenstein, E. Richard Stanley, Gideon Bollag, Yongkai Mo, Brian L. West, Kimberly B. Dahlman, Bernice Matusow, James Tsai, Rafe Shellooe, Orsolya Giricz, Xiomaris M. Cotto-Rios, and Jeffrey A. Sosman
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Proto-Oncogene Proteins B-raf ,MAP Kinase Signaling System ,THP-1 Cells ,Transplantation, Heterologous ,Antineoplastic Agents ,Biology ,03 medical and health sciences ,Mice ,Downregulation and upregulation ,RNA interference ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Epigenetics ,neoplasms ,Melanoma ,Mitogen-Activated Protein Kinase Kinases ,Mice, Inbred BALB C ,Kinase ,Interleukins ,Drug Synergism ,General Medicine ,Methylation ,U937 Cells ,DNA Methylation ,medicine.disease ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Drug Resistance, Neoplasm ,Receptors, Granulocyte-Macrophage Colony-Stimulating Factor ,DNA methylation ,Core Binding Factor Alpha 2 Subunit ,Mutation ,Cancer research ,Female ,Research Article - Abstract
Resistance to current therapies still impacts a significant number of melanoma patients and can be regulated by epigenetic alterations. Analysis of global cytosine methylation in a cohort of primary melanomas revealed a pattern of early demethylation associated with overexpression of oncogenic transcripts. Loss of methylation and associated overexpression of the CSF 1 receptor (CSF1R) was seen in a majority of tumors and was driven by an alternative, endogenous viral promoter in a subset of samples. CSF1R was particularly elevated in melanomas with BRAF and other MAPK activating mutations. Furthermore, rebound ERK activation after BRAF inhibition was associated with RUNX1-mediated further upregulation of CSF-1R and its ligand IL-34. Importantly, increased CSF-1R and IL-34 overexpression were detected in an independent cohort of resistant melanomas. Inhibition of CSF-1R kinase or decreased CSF-1R expression by RNAi reduced 3-D growth and invasiveness of melanoma cells. Coinhibition of CSF-1R and BRAF resulted in synergistic efficacy in vivo. To our knowledge, our data unveil a previously unknown role for the autocrine-regulated CSF-1R in BRAF V600E resistance and provide a preclinical rationale for targeting this pathway in melanoma.
- Published
- 2018
7. Unusual interaction of RNA polymerase with the bacteriophage Mu middle promoter Pm in the absence of its activator protein Mor
- Author
-
Martha M. Howe and Yongkai Mo
- Subjects
Transcription, Genetic ,Biology ,Cleavage (embryo) ,Microbiology ,prokaryotic transcription ,Bacteriophage mu ,chemistry.chemical_compound ,Transcription (biology) ,Bacterial transcription ,Genes, Reporter ,RNA polymerase ,Mu middle promoter Pm ,Promoter Regions, Genetic ,Original Research ,Mor activator protein ,Gene Expression Profiling ,Temperature ,Promoter ,RNAP-promoter interactions ,DNA-Directed RNA Polymerases ,beta-Galactosidase ,Molecular biology ,In vitro ,enzymes and coenzymes (carbohydrates) ,chemistry ,Trans-Activators ,bacteria ,Bacteriophage Mu ,DNA ,Protein Binding - Abstract
The bacteriophage Mu Mor activator protein is absolutely required for transcription from the Mu middle promoter P(m). However, when RNA polymerase (RNAP) was incubated with P(m) DNA in the absence of Mor, a band at promoter position -51 was hypersensitive to DNase I cleavage, demonstrating an interaction of RNAP with the promoter DNA. The hypersensitivity was similar at four different lengths of P(m) DNA assayed from -62 to +10, -62 to +46, -96 to +10, and -96 to +46. The hypersensitivity occurred equally well at 5 °C, 15 °C, and 30 °C, indicating that it did not require open complex formation, which only occurred at 30 °C. The -51 hypersensitivity at 5 °C and 15 °C was eliminated by the addition of heparin, consistent with the possibility that it arose by formation of unstable closed complexes of RNAP bound to P(m) DNA. Generation of the hypersensitive band required the complete RNAP with its αCTDs, but neither the αCTD nor intact α were sufficient for the interaction and resulting hypersensitivity. There was no correlation between the level of hypersensitivity observed in vitro and the level of Pm activity in vivo, as assayed by the Mor-dependent production of β-galactosidase from a P(m)-lacZ fusion. In an "order of addition" experiment, preincubation of P(m) DNA with Mor followed by addition of RNAP led to the fastest open complex formation, whereas preincubation of P(m) DNA with RNAP gave the slowest. These results support the conclusion that Mor recruits RNAP to P(m) rather than reposition a prebound RNAP, as occurs for C-dependent repositioning of RNAP at the Mu late promoter Pmom .
- Published
- 2014
8. Abstract 2515: Aberrant expression of CSF1R in melanoma is driven through an endogenous viral promoter and it contributes to malignant growth and BRAF-inhibitor resistance
- Author
-
E. Richard Stanley, Gideon Bollag, Orsolya Giricz, Kimberly B. Dahlman, Bernice Matusow, Xiomaris M. Cotto-Rios, Brian L. West, Elizabeth M. Burton, Veronika Polishchuck-Lee, Yiting Yu, Kith Pradhan, Yongkai Mo, Hoa Nguyen, John M. Greally, Matthias Bartenstein, Tushar B. Bhagat, Emily Bernstein, Gaston Habets, Jeffrey A. Sosman, Douglas B. Johnson, Paraic A. Kenny, Evripidis Gavathiotis, Rafe Shellooe, Amit Verma, and Chiara Vardabasso
- Subjects
Cancer Research ,Cell growth ,Melanoma ,Cancer ,Biology ,medicine.disease ,medicine.disease_cause ,Metastasis ,Oncology ,medicine ,Cancer research ,Receptor ,Autocrine signalling ,Carcinogenesis ,V600E - Abstract
Epigenetic changes in cancer are thought to contribute to the regulation of invasion and metastasis. To study this at a genome-wide level in melanoma, we analyzed the methylome of 44 cases of malignant melanoma. We saw widespread demethylation occurring preferentially outside of CpG islands. Comparison of primary and metastatic lesions showed demethylation occurs early during carcinogenesis with few additional alterations in advanced tumors. The colony stimulating factor-1 receptor was aberrantly expressed and hypomethylated in nearly all cases. Its expression was validated by IHC and RNA-FISH on primary tumors and by qPCR, Western blotting and FACS in BRAF mutant and WT cell lines. CSF1R can be aberrantly expressed via an upstream LTR element in Hodgkin's lymphoma. After analyzing our patient samples and cell lines, we have found this aberrant transcript may be the dominant form in melanoma as well. Expression of one of its ligands IL34 was also shown in the cell lines by both ELISA and qPCR pointing to a potential autocrine regulatory loop. The effects of a small molecule inhibitor, PLX3397 as well as shRNA-mediated knockdown of the receptor were investigated in 2D and 3D cell culture. We saw inhibition of cell growth, smaller colony size, increased apoptosis and decreased invasiveness suggesting a functional role for CSF-1R in melanoma. Treatment of melanoma with BRAF-V600E inhibitors is effective for a time, but resistance invariably develops. The feedback activation of EGFR, BRAF amplification, BRAF splice variants and others are known to aid in the acquisition of resistance and the rebound activation of the MAPK-pathway. We are suggesting a role for CSF1R in this process. In Western experiments, the rebound of phospho-ERK after BRAF inhibitor treatment was accelerated with the addition of CSF1R ligands, or delayed with PLX3397, also attenuating AKT phosphorylation. Melanoma cells stably expressing shRNA against CSF1R recapitulated the effects of the inhibitor. Assaying the cells at different time points during a long-term V600E inhibitory experiment, we saw increasing levels of the transcription factor RUNX1, followed by increasing levels of IL34 and of the receptor, as well as its maturation, and presentation on the cell surface. shRNA-mediated knockdown of RUNX1 resulted in lower levels of the CSF1R and IL34 transcripts and delayed the rebound. Analysis of primary RNA-Seq data showed an increase in RUNX1, CSF1R and IL34 expression in resistant tumors. Co-inhibition of CSF1R and BRAF was also tested and resulted in synergistic blockade of cell growth in vitro and xenograft growth in vivo.The CSF1R inhibitor, PLX3397 is currently in clinical trials for glioblastoma, prostate, breast cancers and other cancers. These data present a preclinical rationale for its study in malignant melanoma. Citation Format: Orsolya Giricz, Yongkai Mo, Kimberly B. Dahlman, Xiomaris M. Cotto-Rios, Chiara Vardabasso, Hoa Nguyen, Bernice Matusow, Matthias Bartenstein, Veronika Polishchuck-Lee, Douglas B. Johnson, Tushar B. Bhagat, Rafe Shellooe, Elizabeth Burton, Gaston Habets, John M. Greally, Yiting Yu, Gideon Bollag, Paraic A. Kenny, Kith Pradhan, E. Richard Stanley, Emily Bernstein, Evripidis Gavathiotis, Brian L. West, Jeffrey A. Sosman, Amit Verma. Aberrant expression of CSF1R in melanoma is driven through an endogenous viral promoter and it contributes to malignant growth and BRAF-inhibitor resistance [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 2515.
- Published
- 2018
- Full Text
- View/download PDF
9. High-resolution genome-wide cytosine methylation profiling with simultaneous copy number analysis and optimization for limited cell numbers
- Author
-
Reid F. Thompson, Masako Suzuki, Jacob L. Glass, Mayumi Oda, Maria E. Figueroa, Roland Green, Eli Hatchwell, Amit Verma, Xinmin Zhang, Luke Dannenberg, Emmanuel Olivier, Yongkai Mo, Todd Richmond, Eric E. Bouhassira, Ari Melnick, John M. Greally, and Rebecca R. Selzer
- Subjects
Genetics ,0303 health sciences ,HpaII ,Genome, Human ,Copy number analysis ,DNA ,Biology ,Gene Regulation, Chromatin and Epigenetics ,DNA Methylation ,Genome ,Polymerase Chain Reaction ,Deoxyribonuclease HpaII ,03 medical and health sciences ,Cytosine ,0302 clinical medicine ,CpG site ,030220 oncology & carcinogenesis ,DNA methylation ,Illumina Methylation Assay ,Humans ,Human genome ,Cells, Cultured ,030304 developmental biology ,Epigenomics - Abstract
Many genome-wide assays involve the generation of a subset (or representation) of the genome following restriction enzyme digestion. The use of enzymes sensitive to cytosine methylation allows high-throughput analysis of this epigenetic regulatory process. We show that the use of a dual-adapter approach allows us to generate genomic representations that includes fragments of
- Published
- 2009
10. Inhibition of the TGF-β receptor I kinase promotes hematopoiesis in MDS
- Author
-
Leonidas C. Platanias, Josh Hayman, Satyanarayana Medicherla, Linda S. Higgins, Jing Ying Ma, Bhaskar Das, Andrea Pellagatti, Perry Pahanish, Krishna Gundabolu, Markus Bitzer, Ying Zhang, Tony A. Navas, Davendra Sohal, Aaron N. Nguyen, Amittha Wickrema, Alan F. List, Suman Kambhampati, Adam Chubak, Tushar D. Bhagat, Simrit Parmar, Ira Braunchweig, Amit Verma, Li Zhou, Jacqueline Boultwood, Yongkai Mo, and Ann M. Kapoun
- Subjects
Male ,Stromal cell ,Immunology ,Receptor, Transforming Growth Factor-beta Type I ,Antigens, CD34 ,Mice, Transgenic ,Protein Serine-Threonine Kinases ,Biology ,Models, Biological ,Biochemistry ,Mice ,Bone Marrow ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Aged ,Aged, 80 and over ,Ineffective Hematopoiesis ,Regulation of gene expression ,Pteridines ,Lentivirus ,Cell Biology ,Hematology ,Middle Aged ,Hematopoiesis ,Haematopoiesis ,medicine.anatomical_structure ,Myelodysplastic Syndromes ,Luspatercept ,Cancer research ,Female ,Bone marrow ,Signal transduction ,Receptors, Transforming Growth Factor beta ,Transforming growth factor - Abstract
MDS is characterized by ineffective hematopoiesis that leads to peripheral cytopenias. Development of effective treatments has been impeded by limited insight into pathogenic pathways governing dysplastic growth of hematopoietic progenitors. We demonstrate that smad2, a downstream mediator of transforming growth factor–β (TGF-β) receptor I kinase (TBRI) activation, is constitutively activated in MDS bone marrow (BM) precursors and is overexpressed in gene expression profiles of MDS CD34+ cells, providing direct evidence of overactivation of TGF-β pathway in this disease. Suppression of the TGF-β signaling by lentiviral shRNA-mediated down-regulation of TBRI leads to in vitro enhancement of hematopoiesis in MDS progenitors. Pharmacologic inhibition of TBRI (alk5) kinase by a small molecule inhibitor, SD-208, inhibits smad2 activation in hematopoietic progenitors, suppresses TGF-β–mediated gene activation in BM stromal cells, and reverses TGF-β–mediated cell-cycle arrest in BM CD34+ cells. Furthermore, SD-208 treatment alleviates anemia and stimulates hematopoiesis in vivo in a novel murine model of bone marrow failure generated by constitutive hepatic expression of TGF-β1. Moreover, in vitro pharmacologic inhibition of TBRI kinase leads to enhancement of hematopoiesis in varied morphologic MDS subtypes. These data directly implicate TGF-β signaling in the pathobiology of ineffective hematopoiesis and identify TBRI as a potential therapeutic target in low-risk MDS.
- Published
- 2008
- Full Text
- View/download PDF
11. Genetic Alteration ofMycobacterium smegmatisTo Improve Mycobacterium-Mediated Transfer of Plasmid DNA into Mammalian Cells and DNA Immunization
- Author
-
Natalie M. Quanquin, Lydia Tesfa, Yongkai Mo, William R. Jacobs, Keith M. Derbyshire, William R. Bourn, Glenn J. Fennelly, Norman L. Letvin, William H. Vecino, and Uma Devi K. Ranganathan
- Subjects
Mycobacterium smegmatis ,Immunology ,CD8-Positive T-Lymphocytes ,HIV Envelope Protein gp120 ,Biology ,Microbiology ,Cell Line ,DNA vaccination ,Mice ,Transformation, Genetic ,Plasmid ,Vaccines, DNA ,Animals ,Humans ,Replicon ,Gene ,Mice, Inbred BALB C ,Vaccination ,Genetic transfer ,biology.organism_classification ,Mycobacterium bovis ,Molecular biology ,Bacterial vaccine ,Transformation (genetics) ,Infectious Diseases ,Bacterial Vaccines ,Microbial Immunity and Vaccines ,Parasitology ,Plasmids - Abstract
Mycobacteria target and persist within phagocytic monocytes and are strong adjuvants, making them attractive candidate vectors for DNA vaccines. We characterized the ability of mycobacteria to deliver transgenes to mammalian cells and the effects of various bacterial chromosomal mutations on the efficiency of transfer in vivo and in vitro. First, we observed green fluorescent protein expression via microscopy and fluorescence-activated cell sorting analysis after infection of phagocytic and nonphagocytic cell lines byMycobacterium smegmatisorM. bovisBCG harboring a plasmid encoding the fluorescence gene under the control of a eukaryotic promoter. Next, we compared the efficiencies of gene transfer usingM. smegmatisor BCG containing chromosomal insertions or deletions that cause early lysis, hyperconjugation, or an increased plasmid copy number. We observed a significant—albeit only 1.7-fold—increase in the level of plasmid transfer to eukaryotic cells infected withM. smegmatishyperconjugation mutants.M. smegmatisstrains that overexpressed replication proteins (Rep) of pAL5000, a plasmid whose replicon is incorporated in many mycobacterial constructs, generated a 10-fold increase in plasmid copy number and 3.5-fold and 3-fold increases in gene transfer efficiency to HeLa cells and J774 cells, respectively. Although BCG strains overexpressing Rep could not be recovered, BCG harboring a plasmid with a copy-up mutation inoriMresulted in a threefold increase in gene transfer to J774 cells. Moreover,M. smegmatisstrains overexpressing Rep enhanced gene transfer in vivo compared with a wild-type control. Immunization of mice with mycobacteria harboring a plasmid (pgp120hE) encoding human immunodeficiency virus gp120 elicited gp120-specific CD8 T-cell responses among splenocytes and peripheral blood mononuclear cells that were up to twofold (P< 0.05) and threefold (P< 0.001) higher, respectively, in strains supporting higher copy numbers. The magnitude of these responses was approximately one-half of that observed after intramuscular immunization with pgp120hE.M. smegmatisand other nonpathogenic mycobacteria are promising candidate vectors for DNA vaccine delivery.
- Published
- 2007
- Full Text
- View/download PDF
12. Abstract 3332: Aberrant expression of CSF1R in melanoma is driven through an endogenous viral promoter and it contributes to malignant growth and the acquisition of resistance against BRAF inhibition
- Author
-
Veronika Polishchuck, Yiting Yu, Paraic A. Kenny, Brian L. West, Rafe Shellooe, Tushar D. Bhagat, Orsolya Giricz, Caroline Hu, Yongkai Mo, Kith Pradhan, Gideon Bollag, Jeffrey A. Sosman, John M. Greally, Kimberly B. Dahlman, Bernice Matusow, Matthias Bartenstein, Amit Verma, Elizabeth M. Burton, Nandini Ramachandra, and Hoa Nguyen
- Subjects
Cancer Research ,Cell growth ,Melanoma ,Cancer ,Biology ,medicine.disease_cause ,medicine.disease ,Molecular biology ,Metastasis ,Small hairpin RNA ,Oncology ,medicine ,Cancer research ,Autocrine signalling ,Carcinogenesis ,V600E - Abstract
Epigenetic changes in cancer are thought to contribute to regulation of invasion and metastasis. To study this at a genome-wide level in melanoma we analyzed the methylome of 44 cases of malignant melanoma. We saw widespread demethylation in melanoma occurring preferentially outside of CpG islands. Comparison of primary and metastatic lesions showed demethylation occurs early during carcinogenesis with few additional alterations in advanced tumors. The colony stimulating factor-1 receptor was aberrantly expressed and hypomethylated in nearly all cases. The expression of CSF1R was validated by IHC on primary tumors and by qPCR and Western blotting in BRAF mutant and WT cell lines. CSF1R can be aberrantly expressed via an upstream LTR element in Hodgkin’s lymphoma. After analyzing our patient samples and the cell lines, we have found this aberrant transcript may be the dominant form in melanoma as well. Expression of one of its ligands IL34 was also shown in the cell lines by both ELISA and qPCR pointing to a potential autocrine regulatory loop. The effects of a small molecule inhibitor, PLX3397 as well as shRNA-mediated knockdown of the receptor were investigated in 2D and 3D cell culture. We saw inhibition of cell growth, smaller colony size, increased apoptosis and decreased invasiveness - suggesting a functional role for CSF1R in melanoma. Treatment of melanoma with small molecule inhibitors of BRAF V600E is effective for a time, but resistance invariably develops. The feedback activation of EGFR, BRAF amplification, BRAF splice variants and others are known to aid in the acquisition of resistance and the rebound activation of the MAPK-pathway. We are suggesting a role for CSF1R in this process. In Western experiments, the rebound of phospho-ERK after BRAF inhibitor treatment was accelerated with the addition of CSF1R ligands, or delayed with PLX3397, also attenuating AKT phosphorylation. Melanoma cells stably expressing shRNA against CSF1R recapitulated the effects of the inhibitor. Assaying the cells at different time points during a long-term V600E inhibitory experiment, we saw increasing levels of the transcription factor RUNX1, followed by increasing levels of IL34 and of the receptor, as well as its maturation, evidenced by the appearance of the high MW form. Utilizing shRNA-mediated knockdown of RUNX1 resulted in lower levels of the CSF1R and IL34 transcripts and delayed the rebound. Analysis of primary RNA-Seq data showed an increase in RUNX1, CSF1R and IL34 expression in resistant tumors. Co-inhibition of CSF1R and BRAF was also tested and resulted in synergistic blockade of cell growth in vitro and xenograft growth in vivo. The CSF1R inhibitor, PLX3397 is currently in clinical trials for glioblastoma, prostate, breast cancers and other cancers. These data present a preclinical rationale for its study in malignant melanoma. Citation Format: Orsolya Giricz, Yongkai Mo, Caroline Y. Hu, Yiting Yu, Kith Pradhan, Matthias Bartenstein, Nandini Ramachandra, Veronika Polishchuck, Kimberly B. Dahlman, Tushar Bhagat, Hoa Nguyen, Bernice Matusow, Rafe Shellooe, Elizabeth Burton, Paraic Kenny, John Greally, Jeffrey Sosman, Gideon Bollag, Brian West, Amit Verma. Aberrant expression of CSF1R in melanoma is driven through an endogenous viral promoter and it contributes to malignant growth and the acquisition of resistance against BRAF inhibition [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 3332. doi:10.1158/1538-7445.AM2017-3332
- Published
- 2017
- Full Text
- View/download PDF
13. HSC commitment-associated epigenetic signature is prognostic in acute myeloid leukemia
- Author
-
Ujunwa C. Okoye-Okafor, Yongkai Mo, Boris Bartholdy, Tushar D. Bhagat, Amit Verma, Ross L. Levine, Laura Barreyro, Yiting Yu, Maximilian Christopeit, Tihomira I. Todorova, Ari Melnick, Britta Will, Ulrich Steidl, and John M. Greally
- Subjects
Myeloid ,Cellular differentiation ,Kaplan-Meier Estimate ,Biology ,Epigenesis, Genetic ,hemic and lymphatic diseases ,medicine ,Humans ,Epigenetics ,Progenitor cell ,Proportional Hazards Models ,Myeloid leukemia ,Cell Differentiation ,General Medicine ,Methylation ,DNA Methylation ,medicine.disease ,Hematopoietic Stem Cells ,Prognosis ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,DNA methylation ,Immunology ,Cancer research ,Transcriptome ,Research Article - Abstract
Acute myeloid leukemia (AML) is characterized by disruption of HSC and progenitor cell differentiation. Frequently, AML is associated with mutations in genes encoding epigenetic modifiers. We hypothesized that analysis of alterations in DNA methylation patterns during healthy HSC commitment and differentiation would yield epigenetic signatures that could be used to identify stage-specific prognostic subgroups of AML. We performed a nano HpaII-tiny-fragment-enrichment-by-ligation-mediated-PCR (nanoHELP) assay to compare genome-wide cytosine methylation profiles between highly purified human long-term HSC, short-term HSC, common myeloid progenitors, and megakaryocyte-erythrocyte progenitors. We observed that the most striking epigenetic changes occurred during the commitment of short-term HSC to common myeloid progenitors and these alterations were predominantly characterized by loss of methylation. We developed a metric of the HSC commitment–associated methylation pattern that proved to be highly prognostic of overall survival in 3 independent large AML patient cohorts, regardless of patient treatment and epigenetic mutations. Application of the epigenetic signature metric for AML prognosis was superior to evaluation of commitment-based gene expression signatures. Together, our data define a stem cell commitment–associated methylome that is independently prognostic of poorer overall survival in AML.
- Published
- 2013
14. High resolution methylome analysis reveals widespread functional hypomethylation during adult human erythropoiesis
- Author
-
Kathleen Bathon, John M. Greally, Sangeeta Nischal, David L. Ebenezer, Arthur I. Skoultchi, Bennett Caces, Sriram Sundaravel, Andrew S. Artz, Lucy A. Godley, Orsolya Giricz, Jessy Cartier, Yongkai Mo, Oleg Gligich, Amit Verma, Sandeep N. Wontakal, Ulrich Steidl, Amittha Wickrema, Yiting Yu, Hui Liu, Masako Suzuki, Shahina Maqbool, Tushar D. Bhagat, and Sanchari Bhattacharyya
- Subjects
Epigenomics ,Erythrocytes ,Genomics and Proteomics ,Cellular differentiation ,Antigens, CD34 ,Biology ,Biochemistry ,Methylation ,Epigenesis, Genetic ,Humans ,Erythropoiesis ,Epigenetics ,skin and connective tissue diseases ,Molecular Biology ,Oligonucleotide Array Sequence Analysis ,Genetics ,Regulation of gene expression ,Binding Sites ,Genome, Human ,Gene Expression Profiling ,Stem Cells ,GATA2 ,Cell Differentiation ,Cell Biology ,Genomics ,DNA Methylation ,Flow Cytometry ,Introns ,Gene expression profiling ,Gene Expression Regulation ,DNA methylation ,CpG Islands ,sense organs - Abstract
Differentiation of hematopoietic stem cells to red cells requires coordinated expression of numerous erythroid genes and is characterized by nuclear condensation and extrusion during terminal development. To understand the regulatory mechanisms governing these widespread phenotypic changes, we conducted a high resolution methylomic and transcriptomic analysis of six major stages of human erythroid differentiation. We observed widespread epigenetic differences between early and late stages of erythropoiesis with progressive loss of methylation being the dominant change during differentiation. Gene bodies, intergenic regions, and CpG shores were preferentially demethylated during erythropoiesis. Epigenetic changes at transcription factor binding sites correlated significantly with changes in gene expression and were enriched for binding motifs for SCL, MYB, GATA, and other factors not previously implicated in erythropoiesis. Demethylation at gene promoters was associated with increased expression of genes, whereas epigenetic changes at gene bodies correlated inversely with gene expression. Important gene networks encoding erythrocyte membrane proteins, surface receptors, and heme synthesis proteins were found to be regulated by DNA methylation. Furthermore, integrative analysis enabled us to identify novel, potential regulatory areas of the genome as evident by epigenetic changes in a predicted PU.1 binding site in intron 1 of the GATA1 gene. This intronic site was found to be conserved across species and was validated to be a novel PU.1 binding site by quantitative ChIP in erythroid cells. Altogether, our study provides a comprehensive analysis of methylomic and transcriptomic changes during erythroid differentiation and demonstrates that human terminal erythropoiesis is surprisingly associated with hypomethylation of the genome.
- Published
- 2013
15. Abstract 1885: Integrated epigenomic profiling reveals widespread demethylation in melanoma and points to the role of CSF1R-RUNX1 axis in resistance against BRAF inhibition
- Author
-
Gideon Bollag, Kimberly B. Dahlman, Bernice Matusow, Gaston Habets, Elizabeth A. Burton, Jeffrey A. Sosman, Tushar D. Bhagat, Caroline H. Hu, Brian L. West, Paraic A. Kenny, Yiting Yu, Orsolya Giricz, Kith Pradhan, Nandini Ramachandra, Hoa Nguyen, John M. Greally, Matthias Bartenstein, Yongkai Mo, Rafe Shellooe, and Amit Verma
- Subjects
Cancer Research ,Gene knockdown ,Cell growth ,Melanoma ,Biology ,medicine.disease_cause ,medicine.disease ,Molecular biology ,Metastasis ,Oncology ,DNA methylation ,medicine ,Receptor ,Carcinogenesis ,V600E - Abstract
Epigenetic changes in cancer are thought to contribute to regulation of invasion and metastasis. To study this at a genome-wide level in melanoma we analyzed the methylome of 44 cases of malignant melanoma with the HELP (HpaII tiny fragment enriched by LM-PCR) assay and compared it to melanocyte controls. We saw widespread demethylation in melanoma occurring preferentially outside of CpG islands. Comparison of primary and metastatic lesions demonstrated that demethylation occurs early during carcinogenesis with few additional alterations in advanced tumors. Parallel transcriptomic analysis revealed many known and novel oncogenic pathways aberrantly expressed and regulated by loss of DNA methylation. The colony stimulating factor-1 receptor (CSF1R) was aberrantly expressed and hypomethylated in nearly all cases. The expression of CSF1R was validated by immunohistochemistry on primary tumors and by Western blotting in BRAF V600E mutant and WT melanoma cell lines. Expression of its ligand IL34, but not of CSF1 was also shown in the melanoma cells by both ELISA and qPCR. The effects of a small molecule inhibitor, PLX3397 as well as shRNA-mediated knockdown of the receptor were investigated in traditional and 3D cell culture. We saw inhibition of cell growth, smaller colony size, increased apoptosis and decreased invasiveness - suggesting a functional role for CSF1R in melanoma. Treatment of melanoma with small molecule inhibitors of BRAF V600E is effective for a time, but resistance invariably develops. The feedback activation of EGFR, BRAF amplification, BRAF splice variants and others are known to aid in the acquisition of resistance and lead to rebound activation of the MAPK-pathway. In Western blotting experiments, the rebound of ERK phosphorylation after BRAF inhibitor treatment was accelerated with the addition of the CSF1R ligands CSF1 and IL34, or delayed with PLX3397, also attenuating AKT phosphorylation. Melanoma cells stably expressing CSF1R shRNA recapitulated the effects of the inhibitor. Assaying the cells at different time points during a long-term V600E inhibitory experiment, we saw increasing levels of the transcription factor RUNX1, followed by increasing levels of IL34 and of the CSF1R protein, as well as its maturation, evidenced by the appearance of the high MW form. Utilizing shRNA-mediated knockdown of RUNX1 resulted in lower levels of the CSF1R and IL34 transcripts and delayed the rebound. Analysis of primary RNA-Seq data showed an increase in RUNX1, CSF1R and IL34 expression as resistance was acquired. Co-inhibition of CSF1R and BRAF was also tested and resulted in synergistic blockade of cell growth in vitro and xenograft growth in vivo. The CSF1R inhibitor, PLX3397, is in clinical trials for breast and other cancers, and these data present a preclinical rationale for its study in malignant melanoma. Citation Format: Orsolya Giricz, Yongkai Mo, Caroline H. Hu, Kimberly Dahlman, Nandini Ramachandra, Matthias Bartenstein, Kith Pradhan, Tushar Bhagat, Yiting Yu, Hoa Nguyen, Elizabeth Burton, Bernice Matusow, Gaston Habets, Rafe Shellooe, Gideon Bollag, Brian West, John Greally, Jeffrey Sosman, Paraic Kenny, Amit Verma. Integrated epigenomic profiling reveals widespread demethylation in melanoma and points to the role of CSF1R-RUNX1 axis in resistance against BRAF inhibition. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 1885.
- Published
- 2016
- Full Text
- View/download PDF
16. Methylome profiling reveals distinct alterations in phenotypic and mutational subgroups of myeloproliferative neoplasms
- Author
-
Masako Suzuki, Davendra Sohal, Sangeeta Nischal, Alison R. Moliterno, Ulrich Steidl, Animesh Pardanani, John M. Greally, Yiting Yu, Tushar D. Bhagat, Ari Melnick, Maximilian Christopeit, Li Zhou, Amit Verma, McDevitt Michael McDevitt, Jaroslaw P. Maciejewski, Britta Will, Yongkai Mo, and Sanchari Bhattacharyya
- Subjects
Adult ,Male ,Cancer Research ,Biology ,Decitabine ,Article ,Dioxygenases ,Polycythemia vera ,hemic and lymphatic diseases ,Cell Line, Tumor ,Proto-Oncogene Proteins ,medicine ,Humans ,Epigenetics ,Myelofibrosis ,Polycythemia Vera ,Aged ,Genetics ,Aged, 80 and over ,Thrombocytosis ,GATA1 ,Methylation ,Epigenome ,DNA Methylation ,Janus Kinase 2 ,Middle Aged ,medicine.disease ,DNA-Binding Proteins ,Repressor Proteins ,Oncology ,Primary Myelofibrosis ,DNA methylation ,Mutation ,Azacitidine ,Female ,Thrombocythemia, Essential - Abstract
Even though mutations in epigenetic regulators frequently occur in myeloproliferative neoplasms, their effects on the epigenome have not been well studied. Furthermore, even though primary myelofibrosis (PMF) has a markedly worse prognosis than essential thrombocytosis or polycythemia vera, the molecular distinctions between these subgroups are not well elucidated. We conducted the HELP (HpaII tiny fragment enriched by LM-PCR) assay to study genome-wide methylation in polycythemia vera, essential thrombocytosis, and PMF samples compared with healthy controls. We determined that polycythemia vera and essential thrombocytosis are characterized by aberrant promoter hypermethylation, whereas PMF is an epigenetically distinct subgroup characterized by both aberrant hyper- and hypomethylation. Aberrant hypomethylation in PMF was seen to occur in non-CpG island loci, showing further qualitative differences between the disease subgroups. The differentially methylated genes in polycythemia vera and essential thrombocytosis were involved predominantly in cell signaling pathways and were enriched for binding sites of GATA1 and other transcription factors. In contrast, aberrantly methylated genes in PMF were involved in inflammatory pathways and were enriched for NF1, LEF1, and other transcription factors. Within the PMF subgroup, cases with ASXL1 disruptions formed an epigenetically distinct subgroup with relatively increased methylation. Cases of myeloproliferative neoplasms (MPN) with TET2 mutations showed decreased levels of hydroxymethylation and distinct set of hypermethylated genes. In contrast, the JAK2V617F mutation did not drive epigenetic clustering within MPNs. Finally, the significance of aberrant methylation was shown by sensitivity of MPN-derived cell lines to decitabine. These results show epigenetic differences between PMF and polycythemia vera/essential thrombocytosis and reveal methylomic signatures of ASXL1 and TET2 mutations. Cancer Res; 73(3); 1076–85. ©2012 AACR.
- Published
- 2012
17. Aberrant Epigenetic and Genetic Marks Are Seen in Myelodysplastic Leukocytes and Reveal Dock4 as a Candidate Pathogenic Gene on Chromosome 7q*
- Author
-
Jinghang Zhang, Andrea Pellagatti, Cristina Alencar, Christine McMahon, Yongkai Mo, Melissa Fazzari, Davendra Sohal, Vijay Yajnik, Ross L. Levine, Maria E. Figueroa, John M. Greally, Suman Kambhampati, Ari Melnick, Ellen Freidman, Steven D. Gore, Ulrich Steidl, Amit Vermaa, Yogen Sauthararajah, Li Zhou, Tushar D. Bhagat, Simrit Parmar, Leonidas C. Platanias, Joanna Opalinska, Masako Suzuki, Christoph Hueck, Omar Abdel-Wahab, Jacqueline Boultwood, Amittha Wickrema, Yiting Yu, and Sangeeta Nischal
- Subjects
Genetic Markers ,Male ,Genomics and Proteomics ,Apoptosis ,Bone Marrow Cells ,Biology ,Biochemistry ,Epigenesis, Genetic ,medicine ,Leukocytes ,Humans ,Epigenetics ,Molecular Biology ,Myelodysplastic syndromes ,Stem Cells ,GTPase-Activating Proteins ,Bone marrow failure ,Bone Marrow Stem Cell ,Cell Biology ,DNA Methylation ,medicine.disease ,Molecular biology ,medicine.anatomical_structure ,Myelodysplastic Syndromes ,Chromosomal region ,DNA methylation ,Cancer research ,CpG Islands ,Female ,Bone marrow ,Stem cell ,Chromosome Deletion ,Chromosomes, Human, Pair 7 - Abstract
Myelodysplastic syndromes (MDS) are characterized by abnormal and dysplastic maturation of all blood lineages. Even though epigenetic alterations have been seen in MDS marrow progenitors, very little is known about the molecular alterations in dysplastic peripheral blood cells. We analyzed the methylome of MDS leukocytes by the HELP assay and determined that it was globally distinct from age-matched controls and was characterized by numerous novel, aberrant hypermethylated marks that were located mainly outside of CpG islands and preferentially affected GTPase regulators and other cancer-related pathways. Additionally, array comparative genomic hybridization revealed that novel as well as previously characterized deletions and amplifications could also be visualized in peripheral blood leukocytes, thus potentially reducing the need for bone marrow samples for future studies. Using integrative analysis, potentially pathogenic genes silenced by genetic deletions and aberrant hypermethylation in different patients were identified. DOCK4, a GTPase regulator located in the commonly deleted 7q31 region, was identified by this unbiased approach. Significant hypermethylation and reduced expression of DOCK4 in MDS bone marrow stem cells was observed in two large independent datasets, providing further validation of our findings. Finally, DOCK4 knockdown in primary marrow CD34(+) stem cells led to decreased erythroid colony formation and increased apoptosis, thus recapitulating the bone marrow failure seen in MDS. These findings reveal widespread novel epigenetic alterations in myelodysplastic leukocytes and implicate DOCK4 as a pathogenic gene located on the 7q chromosomal region.
- Published
- 2011
18. Design and synthesis of novel derivatives of all-trans retinoic acid demonstrate the combined importance of acid moiety and conjugated double bonds in its binding to PML-RAR-alpha oncogene in acute promyelocytic leukemia
- Author
-
Yongkai Mo, Robert E. Gallagher, Amit Verma, George W. Kabalka, Li Zhou, Tushar D. Bhagat, Carolina Schinke, Swati Goel, Leonidas C. Platanias, and Bhaskar Das
- Subjects
Acute promyelocytic leukemia ,Cancer Research ,Oncogene Proteins, Fusion ,medicine.drug_class ,Recombinant Fusion Proteins ,Retinoic acid ,Apoptosis ,Tretinoin ,Conjugated system ,Biology ,Response Elements ,Transfection ,Article ,chemistry.chemical_compound ,Leukemia, Promyelocytic, Acute ,Amide ,Cell Line, Tumor ,medicine ,Moiety ,Humans ,Retinoid ,Luciferases ,neoplasms ,Cell Proliferation ,Molecular Structure ,CD11 Antigens ,organic chemicals ,Infant ,Cell Differentiation ,Hematology ,medicine.disease ,Polyene ,Flow Cytometry ,Oncology ,chemistry ,Biochemistry ,Gene Expression Regulation ,Drug Design ,medicine.drug - Abstract
The binding of all-trans retinoic acid (ATRA) to retinoid receptor-alpha (RAR-alpha) relieves transcriptional repression induced by the promyelocytic leukemia-retinoic acid receptor (PML-RAR) oncoprotein. The ATRA molecule contains a cyclohexenyl ring, a polyene chain containing conjugated double alkene bonds, and a terminal carboxyl group. To determine the contributions of these structural components of ATRA to its clinical efficacy, we synthesized three novel retinoids. These consisted of either a modified conjugated alkene backbone with an intact acid moiety (13a) or a modified conjugated alkene backbone and conversion of the acid group to either an ester (13b) or an aromatic amide (13c). Reporter assays demonstrated that compound 13a successfully relieved transcriptional repression by RAR-alpha, while 13b and 13c could not, demonstrating the critical role of the acid moiety in this binding. However, only ATRA was able to significantly inhibit the proliferation of APL cells while 13a, 13b, or 13c was not. Furthermore, only 13a led to partial non-significant differentiation of NB4 cells, demonstrating the importance of C9-C10 double bonds in differentiation induced CD11 expression. Our results demonstrate that both the acid moiety and conjugated double bonds present in the ATRA molecule are important for its biological activity in APL and have important implications for the design of future novel retinoids.
- Published
- 2010
19. Aberrant DNA methylation in malignant melanoma
- Author
-
Kathy Amiri, John M. Greally, Yongkai Mo, Carolina Schinke, Amit Verma, Jeffrey A. Sosman, and Yiting Yu
- Subjects
Cancer Research ,Skin Neoplasms ,medicine.medical_treatment ,Melanoma ,Decitabine ,Dermatology ,Disease ,Methylation ,Biology ,DNA Methylation ,medicine.disease ,Article ,Targeted therapy ,Epigenesis, Genetic ,Gene Expression Regulation, Neoplastic ,Oncology ,DNA methylation ,medicine ,Cancer research ,Gene silencing ,Humans ,Epigenetics ,Gene Silencing ,medicine.drug - Abstract
Malignant melanoma remains one of the most deadly human cancers with no effective cures for metastatic disease. The poor efficacy of current therapy in advanced melanoma highlights the need for better understanding of molecular mechanisms contributing to the disease. Recent work has shown that epigenetic changes, including aberrant DNA methylation, lead to alterations in gene expression and are as important in the development of malignant melanoma as the specific and well-characterized genetic events. Reversion of these methylation patterns could thus lead to a more targeted therapy and are currently under clinical investigation. The purpose of this review is to compile recent information on aberrant DNA methylation of melanoma, to highlight key genes and molecular pathways in melanoma development, which have been found to be epigenetically altered and to provide insight as to how DNA methylation might serve as targeted treatment option as well as a molecular and prognostic marker in malignant melanoma.
- Published
- 2010
20. Abstract PR06: Integrated epigenomic profiling reveals widespread demethylation in melanoma and reveals CSF-1 Receptor as an aberrant regulator of malignant growth and invasion
- Author
-
Orsolya Giricz, Yongkai Mo, Caroline Hu, Kimberly Dahlman, Sanchari Bhattacharyya, Hoa Nguyen, Bernice Matusow, Tushar Bhagat, Yiting Yu, Rafe Shellooe, Elizabeth Burton, Gaston Habets, John Greally, Kenny Paraic, Jeffrey Sosman, Gideon Bollag, Brian West, and Amit Verma
- Subjects
Cancer Research ,Oncology - Abstract
Epigenetic changes in cancer are thought to contribute to regulation of tumor invasion and metastasis, but this previously has not been studied at a genome wide level in melanoma. We analyzed the methylome of 44 cases of malignant melanoma with the HELP (HpaII tiny fragment enriched by LM-PCR) assay and compared it with healthy melanocyte controls. We observed widespread demethylation in malignant melanoma, preferentially outside of CpG islands. The epigenomic loss of methylation was independent of mutational status of BRAF, RAS and Kit. Comparison of primary and metastatic lesions demonstrated that demethylation occurs early during carcinogenesis with very few additional alterations in advanced tumors. Parallel transcriptomic analysis revealed many known and novel oncogenic pathways that were aberrantly expressed and regulated by loss of DNA methylation. Strikingly, the colony stimulating factor-1 receptor (CSF1R, c-fms) was aberrantly expressed and hypomethylated in nearly all cases. CSF1R is a transmembrane tyrosine kinase receptor that predominantly regulates macrophages, osteoclasts, and microglia, but is known to sometimes be aberrantly expressed by malignant cells in Hodgkins lymphoma. The expression of CSF1R on malignant melanocytes was validated by immunohistochemical analysis of primary tumors. In several melanoma cell lines (A2058, WM-266-4, SK-MEL-2, M14c#5) we found through PCR sequencing of the cDNA 5′ untranslated region that the CSF1R can be expressed through an aberrant promoter, as has been described for Hodgkin lymphoma. A custom Taqman assay was developed for this unique transcript, and then used to detect the transcript in 4 of 40 samples in a panel of melanoma biopsies, suggesting that aberrant CSF1R expression in melanoma is not uncommon. Expression of CSF1R protein in the cell lines was confirmed by FACS using anti-CD115 antibodies, and by Western blot using antibodies directed to the C-terminus. Expression of the ligand CSF-1 was also found in the melanoma cells by both ELISA and Taqman assays. Inhibition of in vitro cell growth by PLX3397, a clinically relevant small molecule inhibitor of CSF1R kinase, could be observed in 3D cell culture, indicating that under some conditions an autocrine stimulation of growth occurs. shRNA mediated knockdown of CSF1R also demonstrated decreased colony size and increased apoptosis in 3D culture conditions. The invasiveness of melanoma cells was decreased after treatment with PLX3397 or anti-CSF1 antibodies, suggesting a role for melanoma cancer cell expression of CSF1R in metastasis. Since three of cell lines possess an oncogenic BRAF mutation, co-inhibition of CSF1R and BRAF was tested and resulted in synergistic blockade of cell growth in vitro and A2058 xenograft growth in vivo. The CSF1R inhibitor, PLX3397, is under investigation in clinical trials for breast, glioma, and other cancers, and these data present a preclinical rationale for its study in malignant melanoma. This abstract is also being presented as Poster A06. Citation Format: Orsolya Giricz, Yongkai Mo, Caroline Hu, Kimberly Dahlman, Sanchari Bhattacharyya, Hoa Nguyen, Bernice Matusow, Tushar Bhagat, Yiting Yu, Rafe Shellooe, Elizabeth Burton, Gaston Habets, John Greally, Kenny Paraic, Jeffrey Sosman, Gideon Bollag, Brian West, Amit Verma. Integrated epigenomic profiling reveals widespread demethylation in melanoma and reveals CSF-1 Receptor as an aberrant regulator of malignant growth and invasion. [abstract]. In: Proceedings of the AACR Special Conference on Advances in Melanoma: From Biology to Therapy; Sep 20-23, 2014; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(14 Suppl):Abstract nr PR06.
- Published
- 2015
- Full Text
- View/download PDF
21. Abstract 4781: Integrated epigenomic profiling reveals widespread demethylation in melanoma and reveals CSF-1 receptor as an aberrant regulator of malignant growth and invasion
- Author
-
Caroline H. Hu, Paraic A. Kenny, James Tsai, Amit Verma, Orsolya Giricz, Tushar D. Bhagat, Yu Shyr, Jeffrey M. Trent, Sanchari Bhattacharyya, Yongkai Mo, Yiting Yu, Richard E. Stanley, Jeffrey A. Sosman, Hoa Nguyen, Chao Zhang, Gaston Habets, Gideon Bollag, Kimberly B. Dahlman, Bernice Matusow, Elizabeth M. Burton, John M. Greally, Rafe Shellooe, and Brian L. West
- Subjects
Genome instability ,Cancer Research ,Melanoma ,Biology ,medicine.disease ,medicine.disease_cause ,Molecular biology ,Transcriptome ,Oncology ,CpG site ,DNA methylation ,medicine ,Epigenetics ,Carcinogenesis ,Epigenomics - Abstract
Epigenetic alterations can direct carcinogenesis by leading to transcriptional changes and inducing genomic instability. We analyzed the methylome of malignant melanoma and observed widespread loss of DNA methylation that was found to preferentially occur outside of CpG islands. Demethylation was seen to occur early during carcinogenesis, was independent of mutational status and correlated with genomic instability. Parallel transcriptomic analyses revealed that various immune and cancer associated pathways were overexpressed and were associated with promoter demethylation. The CSF1-receptor (CSF1R) was aberrantly overexpressed and hypomethylated in nearly all cases and was strikingly expressed via an aberrant upstream promoter in 10% of melanomas. shRNA mediated knockdown and inhibition of CSF1R kinase via a clinically relevant inhibitor, PLX3397, led to decreased 3D growth and invasiveness. Co-inhibition of CSF1R and BRAF resulted in synergistic blockade of BRAF-mutant melanoma xenograft growth. Thus, widespread epigenetic changes are seen in melanoma and CSF1R is a potential therapeutic target in this disease. Citation Format: Yongkai Mo, Orsolya Giricz, Caroline H. Hu, Kimberly B. Dahlman, Sanchari Bhattacharyya, Hoa Nguyen, Bernice Matusow, Tushar Bhagat, Rafe Shellooe, Elizabeth Burton, James Tsai, Chao Zhang, Gaston Habets, Yu Shyr, John Greally, Yiting Yu, Gideon E. Bollag, Richard Stanley, Jeffrey Trent, Paraic A. Kenny, Brian L. West, Jeffrey Sosman, Amit K. Verma. Integrated epigenomic profiling reveals widespread demethylation in melanoma and reveals CSF-1 receptor as an aberrant regulator of malignant growth and invasion. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 4781. doi:10.1158/1538-7445.AM2014-4781
- Published
- 2014
- Full Text
- View/download PDF
22. Abstract A26: Integrated epigenomic profiling reveals widespread demethylation in melanoma, and reveals aberrant CSF-1 receptor expression as a regulator of malignant growth and invasion inhibited by PLX3397
- Author
-
Brian L. West, Yongkai Mo, Sanchari Bhattacharyya, Caroline Hu, Orsolya Giricz, Rafe Shellooe, John M. Greally, Tushar D. Bhagat, Amit Verma, Yiting Yu, Jeffrey A. Sosman, Hoa Nguyen, Gaston Habets, Gideon Bollag, Kimberly B. Dahlman, Bernice Matusow, Paraic A. Kenny, and Elizabeth A. Burton
- Subjects
Cancer Research ,biology ,Melanoma ,Receptor expression ,medicine.disease ,medicine.disease_cause ,Molecular biology ,Receptor tyrosine kinase ,Metastasis ,Oncology ,DNA methylation ,medicine ,biology.protein ,Autocrine signalling ,Carcinogenesis ,Epigenomics - Abstract
Epigenetic changes in cancer are thought to contribute to regulation of tumor invasion and metastasis, but this previously has not been studied at a genome wide level in melanoma. We analyzed the methylome of 44 cases of malignant melanoma with the HELP (HpaII tiny fragment enriched by LM-PCR) assay and compared it with healthy melanocyte controls. We observed widespread demethylation in malignant melanoma, preferentially outside of CpG islands. The epigenomic loss of methylation was independent of mutational status of BRAF, RAS and Kit. Comparison of primary and metastatic lesions demonstrated that demethylation occurs early during carcinogenesis with very few additional alterations in advanced tumors. Parallel transcriptomic analysis revealed many known and novel oncogenic pathways that were aberrantly expressed and regulated by loss of DNA methylation. Strikingly, the colony stimulating factor-1 receptor (CSF1R, c-fms) was aberrantly expressed and hypomethylated in nearly all cases. CSF1R is a transmembrane tyrosine kinase receptor that predominantly regulates macrophages, osteoclasts, and microglia, but is known to sometimes be aberrantly expressed by malignant cells in Hodgkins lymphoma. The expression of CSF1R on malignant melanocytes was validated by immunohistochemical analysis of primary tumors. In a melanoma cell line (A2058) we found through PCR sequencing of the cDNA 5' untranslated region that the CSF1R can be expressed through an aberrant promoter, as has been described for Hodgkin lymphoma. A custom Taqman assay was developed for this unique transcript, and then used to detect the transcript in 4 of 40 samples in a panel of melanoma biopsies, suggesting that aberrant CSF1R expression in melanoma is not uncommon. Expression of CSF1R protein in A2058 cells was confirmed by FACS using anti-CD115 antibodies, and by Western blot using antibodies directed to the C-terminus. Expression of the ligand CSF-1 was also found in A2058 cells by both ELISA and Taqman assays. Inhibition of A2058 cell growth by PLX3397, a clinically relevant small molecule inhibitor of CSF1R kinase, could be observed in 3D cell culture, indicating that under some conditions an autocrine stimulation of growth occurs. shRNA mediated knockdown of CSF1R also demonstrated decreased colony size and increased apoptosis in 3D culture conditions. The invasiveness of A2058 cells was decreased after treatment with PLX3397 or anti-CSF1 antibodies, suggesting a role for melanoma cancer cell expression of CSF1R in metastasis. Since A2058 cells possess an oncogenic BRAF mutation, co-inhibition of CSF1R and BRAF was tested and resulted in synergistic blockade of xenograft growth. The CSF1R inhibitor, PLX3397, is under investigation in clinical trials for breast, glioma, and other cancers, and these data present a preclinical rationale for its study in malignant melanoma. Citation Format: Yongkai Mo, Orsolya Giricz, Caroline Hu, Kimberly Dahlman, Sanchari Bhattacharyya, Hoa Nguyen, Bernice Matusow, Tushar Bhagat, Yiting Yu, Rafe Shellooe, Elizabeth Burton, Gaston Habets, John Greally, Paraic Kenny, Jeffrey Sosman, Gideon Bollag, Brian L. West, Amit Verma. Integrated epigenomic profiling reveals widespread demethylation in melanoma, and reveals aberrant CSF-1 receptor expression as a regulator of malignant growth and invasion inhibited by PLX3397. [abstract]. In: Proceedings of the AACR Special Conference on Tumor Invasion and Metastasis; Jan 20-23, 2013; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2013;73(3 Suppl):Abstract nr A26.
- Published
- 2013
- Full Text
- View/download PDF
23. Epigenomic Profiling of Myeloproliferative Diseases Reveal Idiopathic Myelofibrosis as An Epigenetically Distinct Subgroup and Highlights the Epigenetic Effects of Jak2V617F Mutation
- Author
-
Shahina Maqbool, Sangeeta Nischal, Christoph Heuck, Tushar D. Bhagat, John M. Greally, Masako Suzuki, Li Zhou, Yongkai Mo, Alison R. Moliterno, Amit Verma, Animesh Pardanani, and Yiting Yu
- Subjects
Genome instability ,Genetics ,HpaII ,Immunology ,Luma ,Cell Biology ,Hematology ,Methylation ,Biology ,Biochemistry ,HELP assay ,DNA methylation ,Epigenetics ,Epigenomics - Abstract
Abstract 627 Myeloproliferative diseases (MPD) are clonal hematologic disorders that present with increased numbers of functional, mature, terminally differentiated myeloid elements. Even though genetic, biochemical and functional studies have provided important insights into the pathogenesis of MPDs, the role of epigenetic changes in disease pathobiology is not well elucidated. We performed the HELP assay to study genome-wide methylation patterns in cases of Polycythemia Vera (PV), Essential Thrombocytosis (ET) and Idiopathic Myelofibrosis (IMF) and compared it with normal matched controls. The HELP assay uses differential methylation specific digestion by HpaII and MspI followed by amplification, two color labeling and hybridization to quantitatively determine individual promoter CpG methylation of 25636 loci. Analysis of 26 MPD neutrophil samples comprising 9 cases of ET, 6 cases of PV and 11 Cases of IMF was performed and compared to normal healthy controls. Unsupervised clustering based on global methylation profiles showed that IMF cases formed a distinct epigenetic cluster, while PV and ET cases were more similar to the normal controls. Further analysis of epigenetic differences between these groups showed that PV and ET samples were characterized by aberrant hypermethylation when compared to controls and had 143 genes that were uniformly hypermethylated. These genes are involved in pathways regulated by the NF-Kb and HNF-4alpha transcription factors. IMF on the other hand was characterized by both aberrantly hyper (n=162) and hypomethylated (n=95) loci when compared to controls. The pathways affected by hypomethylated genes were involved in cytokine cell signaling and MAP kinases. We subsequently validated these observations in an independent set of 8 IMF cases compared to 8 age matched controls. Methylation profiles obtained from whole blood by the HELP assay were able to clearly separate IMF cases from controls demonstrating the validity of our observations. These changes were quantified on a whole genome level by the Luminometric methylation assay (LUMA) that also revealed significantly more hypomethylation in IMF when compared to PV and ET cases (49% hypomethylation in IMF vs. 38% in PV/ET, p We next wanted to determine the epigenomic effects of Jak2V617F mutation and compared methylation profiles of MPD cases with and without the mutation. We observed that cases with Jak2 mutation had a higher number of differentially hypomethylated loci. This striking difference was seen by the LUMA assay also with 67% hypomethylation seen in mutant cases when compared to 48% in those without the mutation (p=0.02). This observation was validated in vitro in a cell line (FDCP) that expressed wither WT or mutant Jak2 kinase. We observed that expression of the mutant kinase led to global hypomethylation. This observation builds on recent data demonstrating nuclear binding of the mutant Jak2 kinase and its effects on the histone epigenetic machinery. In conclusion, we report that MPDs are characterized by various novel epigenetic alterations that affect important functional pathways and IMF is grossly epigenetically distinct from ET and PV. The Jak2 mutation also affects the methylome and leads to global hypomethylation that potentially contributes to the genomic instability and disease pathobiology. Disclosures: No relevant conflicts of interest to declare.
- Published
- 2010
- Full Text
- View/download PDF
24. Dysregulation of TGF-Beta Stimulated Smad Signaling Is Seen in Myelodysplasia and Points to the Potential Therapeutic Efficacy of TGF-Beta Receptor I Kinase Inhibition in Low Grade Disease
- Author
-
Tushar D. Bhagat, Pearlie K Burnette, Jm Yingling, Amittha Wickrema, Yiting Yu, Chun Ng, Amit Verma, Ellen W. Friedman, Davendra Sohal, Yongkai Mo, Christina Alencar, Alan F. List, Krishna Gundabolu, Li Zhou, Ulrich Steidl, Lubomir Sokol, Markus Bitzer, Lei Yan, Michael Lahn, Christine McMahon, Melissa Fazzari, and G. Kong
- Subjects
Ineffective Hematopoiesis ,medicine.medical_treatment ,Immunology ,Cell Biology ,Hematology ,SMAD ,Biology ,Biochemistry ,Haematopoiesis ,medicine.anatomical_structure ,Cytokine ,Downregulation and upregulation ,TGF beta signaling pathway ,medicine ,Bone marrow ,Signal transduction - Abstract
Abstract 737 Myelodysplastic syndromes (MDS) are characterized by ineffective hematopoiesis that leads to peripheral cytopenias. TGF-beta is a hematopoietic inhibitory cytokine that has been indirectly linked to the pathogenesis of some subsets of MDS and acute leukemias. We have shown (Blood, 112(8):3434; 2008) that smad2, a component of the TGF-beta signaling pathway, is constitutively activated and upregulated in MDS progenitors. Since there is conflicting data about upregulation of TGF-beta levels in MDS, we next sought to determine the molecular basis of TGF-beta receptor-I (TBRI) overactivation and subsequent smad2 phosphorylation / activation in this disease. We observed that smad-7, a negative regulator of TBRI kinase, is markedly down regulated in a meta-analysis of gene expression studies from 89 MDS bone marrow derived CD34+ cells when compared to 61 normal controls (Adjusted P Disclosures: No relevant conflicts of interest to declare.
- Published
- 2009
- Full Text
- View/download PDF
25. Corrigendum for Rosenquist Letter
- Author
-
Leonidas C. Platanias, Linda S. Higgins, T. Cao, Yongkai Mo, Mani Mohindru, Li Zhou, Myka Estes, Tony Navas, Amit Verma, A. List, Perry Pahanish, Aaron N. Nguyen, and Edwin Haghnazari
- Subjects
MAPK/ERK pathway ,Cancer Research ,Leukemia lymphoma ,medicine.anatomical_structure ,Oncology ,business.industry ,Immunology ,medicine ,Hematology ,Bone marrow ,business ,Pathological ,Proinflammatory cytokine - Published
- 2009
- Full Text
- View/download PDF
26. Design and Synthesis of Novel Derivatives of ATRA Demonstrate the Combined Importance of Acid Moiety and Conjugated Double Bonds in Its Binding to PMLRAR-Alpha Oncogene in Acute Promyelocytic Leukemia
- Author
-
Jaime Anguiano, Amit Verma, Christina Alencar, Perry Pahanish, Carolina Schinke, Li Zhou, Bhaskar C. Das, Yongkai Mo, Christoph Heuck, Tushar D. Bhagat, George W. Kabalka, Leonidas C. Platanias, and Swati Goel
- Subjects
Acute promyelocytic leukemia ,organic chemicals ,Immunology ,Wild type ,Retinoic acid ,Cell Biology ,Hematology ,Biology ,Conjugated system ,medicine.disease ,Biochemistry ,biological factors ,chemistry.chemical_compound ,chemistry ,Cell culture ,medicine ,Moiety ,Luciferase ,neoplasms ,Psychological repression - Abstract
All trans retinoic acid (ATRA) is the treatment of choice for Acute Promyelocytic Leukemia (APL) and binds to the transcriptional repressor PML-RAR-alpha oncogene. The binding of ATRA to RAR-alpha relieves transcriptional repression and leads to induction of genes important for myeloid differentiation. ATRA consists of an acid moiety at position 15 and is also characterized by conjugated double alkene bonds (Fig1A). The exact contributions of these structural components of ATRA in its clinically important binding to RAR-alpha receptor are not very well studied. In an effort to improve the therapeutic index and to study its binding mechanism, we synthesized novel retinoic acid analogs (retinoids). Our objective was to construct retinoids to test these structure functional correlates. 3 novel retinoids created were synthesized as follows: Compound 13a (Fig1B): consisting of modifying conjugated alkene backbone while keeping acid moiety intact. Compound 13b (Fig1C): consisting of modifying conjugated alkene backbone and converting acid to ester moiety. Compound 13c (Fig1D): consisting of modifying conjugated alkene and cycloalkene ring backbone and keeping acid moiety intact. We tested the ability of these newer retinoids to bind to RAR-alpha as well as inhibit the proliferation of PML-RAR-alpha containing acute promyelocytic leukemic cell line NB4. Reporter assays using a RAR- luciferase demonstrated that Compound 13a successfully relieved transcriptional repression by RAR-alpha and led to downstream gene induction. This was significantly less compared to wild type ATRA demonstrating the importance of C9–C10 double bonds in this binding. Compounds 13b and 13c could not relieve RAR mediated repression of gene induction demonstrating that the acid moiety plays a important role in this binding. Functional studies were consistent with these observations and showed that both ATRA and 13a were able to inhibit the proliferation of APL cells while 13b, 13c were not. Flow cytometry showed that 13a led to partial differentiation of NB4 cells, as evident from CD11b expression after five days of culture. 13b and 13c did not lead to any differentiation, while ATRA led to the greatest expression of CD11b. Our results demonstrate that both the acid moiety and conjugated double bonds are important in binding to RAR-alpha and the subsequent transcriptional repression by retinoic acid. These insights will be important in future efforts to improve the stability and efficacy of ATRA and also have implication in efforts focused on treating ATRA resistant cases of APL. Figure Figure
- Published
- 2008
- Full Text
- View/download PDF
27. Integrative Genomic Analysis Reveals Aberrant Epigenetic Marks in MDS That Can Be Seen in Peripheral Blood Leucocytes
- Author
-
Lawrence Cytryn, Perry Pahanish, Suman Kambhampati, Christina Alencar, Maria E. Figueroa, Li Zhou, Ellen Friedman, Chun Ng, Carolina Schinke, Ivette Vigoda, Swati Goel, Yongkai Mo, Amittha Wickrema, Amit Verma, Davendra Sohal, Christoph Heuck, Simrit Parmar, John M. Greally, and Joanna Opalinska
- Subjects
HpaII ,Cellular differentiation ,Immunology ,Cell Biology ,Hematology ,Methylation ,Biology ,Biochemistry ,Molecular biology ,HELP assay ,CpG site ,DNA methylation ,Epigenetics ,Gene - Abstract
Myelodysplasia (MDS) is a clonal hematopoietic disorder that leads to ineffective hematopoiesis and peripheral cytopenias. DNMT inhibitors such as azacytidine have led to clinical responses in patients, though global epigenetic alterations in MDS have not been well described. The transmission of these epigenetic marks during hematopoietic differentiation and their role in disease pathophysiology is also unknown. We first compared global methylation profiles of 8 bone marrow samples with peripheral leucocytes by using a recently described novel method, the HELP assay (HpaII tiny fragment Enrichment by Ligation-mediated PCR; Khulan et al, Genome Res. 2006 Aug;16(8)) that uses differential methylation-specific digestion by HpaII and MspI followed by amplification, two color labeling and hybridization to quantitatively determine individual promoter CpG island methylation. A whole genome human promoter array (Nimblegen) was used to determine the level of methylation of 25626 gene promoters by calculating HpaII/MspI cut fragment intensity ratio. We observed a high correlation (r=0.89–0.96) of epigenetic marks between bone marrow and peripheral blood samples suggesting that a majority of epigenetic marks can be also be seen in differentiated cells. We subsequently compared peripheral blood leucocytes from 20 patients with MDS with 10 age-matched normal and anemic controls. Parallel gene expression analysis was performed using 37K oligo maskless arrays on cDNA from the same samples. Analysis showed that whole genome methylation profiling has greater discriminatory power in separating clusters of MDS samples from normal and anemic controls when compared to gene expression analysis. Epigenetic profiling demonstrated two clusters of MDS based on similarity of aberrant epigenetic changes. Overall, there was a trend towards hypermethylation in MDS, albeit not statistically significant given the large number of relatively unchanged genes. Detailed analysis revealed several novel differentially methylated genes that had corresponding changes in gene expression, when MDS samples were compared to the controls with a low false discovery rate of analysis. Interesting genes getting hypomethylated and overexpressed included TNF superfamily member 9, granulocyte pep A, microsomal glutathione S-transferase, homeo box B4, mitochondrial RPL11, and others. Similarly, the set of genes that were getting hypermethylated with associated decrease in gene expression included Evi-1, DAPK, HOXB3, Protein Phosphatase 1, CEBPB, mutated in colorectal cancer (MCC), myeloid-lymphoid or mixed-lineage leukemia 5 (MLL5), plasminogen-related protein B, ovarian cancer related protein 1 (ORP1), and others. In addition, we did array-based comparative genome hybridization (aCGH) to look at exact genome copy number changes in these samples. We found changes that were not detectable by conventional karyotyping in all samples. Commonly seen alterations were del(14q11), del(20q11), del(5q13), del(8p23), amp(1q42), amp(5q11), amp(17q12), amp(19q13) and amp(7q22). Integrative analysis revealed sets of genes that were either silenced by methylation or deletion in different patients. Thus, our data demonstrates that promoter DNA methylation changes are an important phenomenon in MDS evolution, and are associated with changes in expression of genes playing important roles in cancer development and/or progression. We also show that previously unrecognizable changes in copy number exist in most patients with MDS. In addition, our work shows that whole genome methylation assays, even when done on peripheral blood leukocytes, can be used for potential biomarker studies in the diagnosis of MDS.
- Published
- 2008
- Full Text
- View/download PDF
28. MDS Marrow Stroma Is Characterized by Distinct Epigenetic Alterations
- Author
-
John M. Greally, Amit Verma, Reid F. Thompson, Emily Spaulding, A. Mario Q. Marcondes, Davendra Sohal, Tushar D. Bhagat, Yongkai Mo, H. Joachim Deeg, and Li Zhou
- Subjects
Stromal cell ,Immunology ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Molecular biology ,Haematopoiesis ,medicine.anatomical_structure ,HELP assay ,Stroma ,DNA methylation ,medicine ,Bone marrow ,Epigenetics ,Epigenomics - Abstract
The bone marrow microenvironment plays an important role in the pathogenesis and perpetuation of stem cell defects in Myelodysplastic Syndrome (MDS). However, while distinct cytogenetic alterations have been described in the stem cell compartment in MDS, the bone marrow stroma has never been shown to be part of the clone. Thus, aberrant epigenetic alterations may be responsible for altered function of bone marrow stroma in MDS. DNA methyl transferase (DNMT) inhibitors, which are therapeutically effective in MDS, affect both hematopoietic cells and the stroma, providing further rationale for studying DNA methylation profiles of bone marrow stroma in this disease. To accomplish this aim, bone marrow mononuclear cells from MDS patients and controls were grown to form adherent cell layers and then depleted for hematopoietic elements by immunomagnetic CD45 negative selection. CD45 negative adherent cells were subsequently expanded and then used for whole genome methylation studies using a recently described novel method, the HELP assay (HpaII tiny fragment Enrichment by Ligation-mediated PCR; Khulan et al, Genome Res. 2006 Aug;16(8)) which uses differential methylation-specific digestion by HpaII and MspI followed by amplification, two color labeling and hybridization to quantify individual promoter CpG island methylation. A custom whole genome human promoter array (Roche-Nimblegen) was used to determine the level of methylation of 25626 gene promoters by calculating HpaII/MspI cut fragment intensity ratio. Global epigenetic profiling revealed that MDS stroma (n=6) was epigenetically distinct from normal bone marrow stroma (n=4) (ANOVA, P In subsequent studies, we profiled stroma from another set of MDS patients who had been treated with the DNMT inhibitor, 5-Azacytidine (n=4). In contrast to untreated MDS patients, there were no significant epigenetic differences between these 5-Azacytidine treated MDS patients and healthy controls (p = NS). These 5-Azacytidine exposed stroma cells did not demonstrate global hypomethylation (as hypothesized after DNMT inhibitor treatment) and were characterized by both hyper- and hypo-methylated loci similar to healthy controls. Thus our results reveal that MDS is characterized by widespread aberrant epigenetic changes in the bone marrow microenvironment. Our results also demonstrate that DNMT inhibitors can alter the epigenomic profiles of stromal cells, and we hypothesize that those stroma effects contribute in part to their clinical efficacy. Overall, these studies underscore the importance of studying the entire bone marrow, including the microenvironment, if we are to improve our understanding of the pathophysiology of MDS and further improve therapy.
- Published
- 2008
- Full Text
- View/download PDF
29. Downregulation of Ribosomal Proteins Is Seen in Non 5q- MDS
- Author
-
John M. Greally, Jacqueline Boultwood, Joanna Opalinska, Jonathan R. Warner, Li Zhou, Christoph Heuck, Ellen Friedman, Yongkai Mo, Davendra Sohal, Amit Verma, Amittha Wickrema, Andrea Pellagati, Christina Alencar, and Benjamin L. Ebert
- Subjects
Genetics ,Tiling array ,HpaII ,Immunology ,UniGene ,Cell Biology ,Hematology ,Ribosomal RNA ,Biology ,Biochemistry ,Gene expression profiling ,HELP assay ,Ribosomal protein ,Gene - Abstract
Recent work (Ebert et al, Nature 2008, Jan 17:335-9) has shown that ribosomal protein S14 (RPS14) is underexpressed in myelodysplasia (MDS) with deletion of chromosome 5q and plays a role in its pathogenesis. The role of ribosomal proteins in other more common subtypes of MDS is unknown. We conducted a meta-analytical comparison of gene expression profiles of 60 cases of non 5q- MDS CD34+ cells with 52 normal CD34+ profiles. These datasets were obtained from seven independent studies from NCBI’s GEO database. The data was integrated based on UniGene IDs and were quantile normalized to ensure cross-study comparability. (Based on our previous approach; Sohal et al PLOS One, 2008, Zhou et al, Blood, 2008). Using significance analysis of microarrays (SAM) and a false discovery rate (FDR) of just 0.04%, we found that ribosomal protein were the class of genes that were most significantly altered in MDS. We observed that RPL35a, RPS9, RPL10, RPL22, RPS14, RPS10, RPS15a, RPS24, RPL24, RPL36, RPL21, RPL23 were strikingly downregulated in non-5q- MDS CD34+ cells. To determine if these alterations were a result of changes in DNA copy numbers of these genes, we examined 20 MDS samples by high resolution array comparative genome hybridization (aCGH) performed on Nimblegen whole genome tiling arrays. aCGH at 6kb resolution revealed deletions in RPL14, RPL22, RPL36, RPS10, RPS5 and even RPS14 in distinct selected cases of non 5q- MDS. These small deletions, which were not identifiable by traditional karyotyping methods, may be putative mechanisms of ribosomal protein downregulation and ultimately, in MDS development and/or progression. Since MDS is also characterized by aberrant epigenetic silencing of genes, we next examined the methylation status of ribosomal gene promoters by high resolution global DNA methylation profiling by using the HELP assay (HpaII tiny fragment Enrichment by Ligation-mediated PCR; Khulan et al, Genome Res. 2006 Aug;16(8)). This assay uses differential methylation-specific digestion by HpaII and MspI followed by amplification, two color labeling and hybridization to quantitatively determine individual promoter CpG island methylation. While there were sporadic changes in some patients, we did not observe any consistently significant changes in methylation of these ribosomal gene promoters on comparison with normal anemic controls. In summary, we show novel widespread alterations in ribosomal protein expression in MDS, at least some of which are associated with genomic deletions. These findings illustrate the applicability of meta-analytical genomic approaches in a heterogeneous disease such as MDS. Most importantly, our data points to the dramatic role of alteration in the protein translational machinery in pathogenesis of MDS.
- Published
- 2008
- Full Text
- View/download PDF
30. High Resolution Methylome Analysis Reveals Widespread Functional Hypomethylation during Adult Human Erythropoiesis.
- Author
-
Yiting Yu, Yongkai Mo, Ebenezer, David, Bhattacharyya, Sanchari, Hui Liu, Sundaravel, Sriram, Giricz, Orsolya, Wontakal, Sandeep, Cartier, Jessy, Caces, Bennett, Artz, Andrew, Nischal, Sangeeta, Bhagat, Tushar, Bathon, Kathleen, Maqbool, Shahina, Gligich, Oleg, Suzuki, Masako, Steidl, Ulrich, Godley, Lucy, and Skoultchi, Art
- Subjects
- *
ERYTHROPOIESIS , *HEMATOPOIETIC stem cells , *MEMBRANE proteins , *TRANSCRIPTION factors , *BONE marrow cells , *GENE expression - Abstract
Differentiation of hematopoietic stem cells to red cells requires coordinated expression of numerous erythroid genes and is characterized by nuclear condensation and extrusion during terminal development. To understand the regulatory mechanisms governing these widespread phenotypic changes, we conducted a high resolution methylomic and transcriptomic analysis of six major stages of human erythroid differentiation. We observed widespread epigenetic differences between early and late stages of erythropoiesis with progressive loss of methylation being the dominant change during differentiation. Gene bodies, intergenic regions, and CpG shores were preferentially demethylated during erythropoiesis. Epigenetic changes at transcription factor binding sites correlated significantly with changes in gene expression and were enriched for binding motifs for SCL, MYB, GATA, and other factors not previously implicated in erythropoiesis. Demethylation at gene promoters was associated with increased expression of genes, whereas epigenetic changes at gene bodies correlated inversely with gene expression. Important gene networks encoding erythrocyte membrane proteins, surface receptors, and heme synthesis proteins were found to be regulated by DNA methylation. Furthermore, integrative analysis enabled us to identify novel, potential regulatory areas of the genome as evident by epigenetic changes in a predicted PU.1 binding site in intron 1 of the GATA1 gene. This intronic site was found to be conserved across species and was validated to be a novel PU.1 binding site by quantitative ChIP in erythroid cells. Altogether, our study provides a comprehensive analysis of methylomic and transcriptomic changes during erythroid differentiation and demonstrates that human terminal erythropoiesis is surprisingly associated with hypomethylation of the genome. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
31. Widespread Hypomethylation Occurs Early and Synergizes with Gene Amplification during Esophageal Carcinogenesis.
- Author
-
Alvarez, Hector, Opalinska, Joanna, Li Zhou, Sohal, Davendra, Fazzari, Melissa J., Yiting Yu, Montagna, Christina, Montgomery, Elizabeth A., Canto, Marcia, Dunbar, Kerry B., Wang, Jean, Roa, Juan Carlos, Yongkai Mo, Bhagat, Tushar, Ramesh, K. H., Cannizzaro, Linda, Mollenhauer, J., Thompson, Reid F., Suzuki, Masako, and Meltzer, Stephen
- Subjects
METHYLATION ,GENE amplification ,ESOPHAGEAL cancer ,GENOMICS ,ADENOCARCINOMA ,DYSPLASIA ,BIOMARKERS ,DISEASE progression - Published
- 2011
- Full Text
- View/download PDF
32. Meta-Analysis of Microarray Studies Reveals a Novel Hematopoietic Progenitor Cell Signature and Demonstrates Feasibility of Inter-Platform Data Integration.
- Author
-
Sohal, Davendra, Yeatts, Andrew, Ye, Kenny, Pellagatti, Andrea, Li Zhou, Pahanish, Perry, Yongkai Mo, Bhagat, Tushar, Mariadason, John, Boultwood, Jacqueline, Melnick, Ari, Greally, John, and Verma, Amit
- Subjects
META-analysis ,FEASIBILITY studies ,HEMATOPOIETIC growth factors ,IMMUNE system ,LEUKEMIA ,HEMATOPOIESIS ,CELL cycle regulation ,SEPTINS ,TRANSCRIPTION factors - Abstract
Microarray-based studies of global gene expression (GE) have resulted in a large amount of data that can be mined for further insights into disease and physiology. Meta-analysis of these data is hampered by technical limitations due to many different platforms, gene annotations and probes used in different studies. We tested the feasibility of conducting a meta-analysis of GE studies to determine a transcriptional signature of hematopoietic progenitor and stem cells. Data from studies that used normal bone marrow-derived hematopoietic progenitors was integrated using both RefSeq and UniGene identifiers. We observed that in spite of variability introduced by experimental conditions and different microarray platforms, our meta-analytical approach can distinguish biologically distinct normal tissues by clustering them based on their cell of origin. When studied in terms of disease states, GE studies of leukemias and myelodysplasia progenitors tend to cluster with normal progenitors and remain distinct from other normal tissues, further validating the discriminatory power of this meta-analysis. Furthermore, analysis of 57 normal hematopoietic stem and progenitor cell GE samples was used to determine a gene expression signature characteristic of these cells. Genes that were most uniformly expressed in progenitors and at the same time differentially expressed when compared to other normal tissues were found to be involved in important biological processes such as cell cycle regulation and hematopoiesis. Validation studies using a different microarray platform demonstrated the enrichment of several genes such as SMARCE, Septin 6 and others not previously implicated in hematopoiesis. Most interestingly, alpha-integrin, the only common stemness gene discovered in a recent comparative murine analysis (Science 302(5644):393) was also enriched in our dataset, demonstrating the usefulness of this analytical approach. [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
33. HSC commitment--associated epigenetic signature is prognostic in acute myeloid leukemia.
- Author
-
Bartholdy, Boris, Christopeit, Maximilian, Will, Britta, Yongkai Mo, Barreyro, Laura, Yiting Yu, Bhagat, Tushar D., Okoye-Okafor, Ujunwa C., Todorova, Tihomira I., Greally, John M., Levine, Ross L., Melnick, Ari, Verma, Amit, and Steidl, Ulrich
- Subjects
- *
ACUTE myeloid leukemia , *DNA methylation , *PROGENITOR cells , *EPIGENETICS , *MEGAKARYOCYTES , *ERYTHROCYTES - Abstract
Acute myeloid leukemia (AML) is characterized by disruption of HSC and progenitor cell differentiation. Frequently, AML is associated with mutations in genes encoding epigenetic modifiers. We hypothesized that analysis of alterations in DNA methylation patterns during healthy HSC commitment and differentiation would yield epigenetic signatures that could be used to identify stage-specific prognostic subgroups of AML. We performed a nano HpaII-tiny-fragment-enrichment-by-ligation-mediated-PCR (nanoHELP) assay to compare genome-wide cytosine methylation profiles between highly purified human long-term HSC, short-term HSC, common myeloid progenitors, and megakaryocyte-erythrocyte progenitors. We observed that the most striking epigenetic changes occurred during the commitment of short-term HSC to common myeloid progenitors and these alterations were predominantly characterized by loss of methylation. We developed a metric of the HSC commitment--associated methylation pattern that proved to be highly prognostic of overall survival in 3 independent large AML patient cohorts, regardless of patient treatment and epigenetic mutations. Application of the epigenetic signature metric for AML prognosis was superior to evaluation of commitment-based gene expression signatures. Together, our data define a stem cell commitment--associated methylome that is independently prognostic of poorer overall survival in AML. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
34. Aberrant Epigenetic and Genetic Marks Are Seen in Myelodysplastic Leukocytes and Reveal Dock4 as a Candidate Pathogenic Gene on Chromosome 7q.
- Author
-
Li Zhou, Opalinska, Joanna, Sohal, Davendra, Yiting Yu, Yongkai Mo, Bhagat, Tushar, Abdel-Wahab, Omar, Fazzari, Melissa, Figueroa, Maria, Alencar, Cristina, Jinghang Zhang, Kambhampati, Suman, Parmar, Simrit, Nischal, Sangeeta, Hueck, Christoph, Suzuki, Masako, Freidman, Ellen, Pellagatti, Andrea, Boultwood, Jacqueline, and Steidl, Ulrich
- Subjects
- *
GENETIC markers , *LEUCOCYTES , *CELL nuclei , *BLOOD cells , *GENE silencing - Abstract
Myelodysplastic syndromes (MDS) are characterized by abnormal and dysplastic maturation of all blood lineages. Even though epigenetic alterations have been seen in MDS marrow progenitors, very little is known about the molecular alterations in dysplastic peripheral blood cells. We analyzed the methylome of MDS leukocytes by the HELP assay and determined that it was globally distinct from age-matched controls and was characterized by numerous novel, aberrant hypermethylated marks that were located mainly outside of CpG islands and preferentially affected GTPase regulators and other cancer-related pathways. Additionally, array comparative genomic hybridization revealed that novel as well as previously characterized deletions and amplifications could also be visualized in peripheral blood leukocytes, thus potentially reducing the need for bone marrow samples for future studies. Using integrative analysis, potentially pathogenic genes silenced by genetic deletions and aberrant hypermethylation in different patients were identffied. DOCK4, a GTPase regulator located in the commonly deleted 7q31 region, was identified by this unbiased approach. Significant hypermethylation and reduced expression of DOCK4 in MDS bone marrow stem cells was observed in two large independent datasets, providing further validation of our findings. Finally, DOCK4 knockdown in primary marrow CD34+ stem cells led to decreased erythroid colony formation and increased apoptosis, thus recapitulating the bone marrow failure seen in MDS. These findings reveal widespread novel epigenetic alterations in myelodysplastic leukocytes and implicate DOCK4 as a pathogenic gene located on the 7q chromosomal region. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.