Network


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

Hotspot


Dive into the research topics where Jumpei Yamazaki is active.

Publication


Featured researches published by Jumpei Yamazaki.


Epigenetics | 2012

Effects of TET2 mutations on DNA methylation in chronic myelomonocytic leukemia

Jumpei Yamazaki; Rodolphe Taby; Aparna Vasanthakumar; Trisha Macrae; Kelly R. Ostler; Lanlan Shen; Hagop M. Kantarjian; Marcos R. Estecio; Jaroslav Jelinek; Lucy A. Godley; Jean-Pierre Issa

TET2 enzymatically converts 5-methyl-cytosine to 5-hydroxymethyl-cytosine, possibly leading to loss of DNA methylation. TET2 mutations are common in myeloid leukemia and were proposed to contribute to leukemogenesis through DNA methylation. To expand on this concept, we studied chronic myelomonocytic leukemia (CMML) samples. TET2 missense or nonsense mutations were detected in 53% (16/30) of patients. In contrast, only 1/30 patient had a mutation in IDH1 or IDH2, and none of them had a mutation in DNMT3A in the sites most frequently mutated in leukemia. Using bisulfite pyrosequencing, global methylation measured by the LINE-1 assay and DNA methylation levels of 10 promoter CpG islands frequently abnormal in myeloid leukemia were not different between TET2 mutants and wild-type CMML cases. This was also true for 9 out of 11 gene promoters reported by others as differentially methylated by TET2 mutations. We found that two non-CpG island promoters, AIM2 and SP140, were hypermethylated in patients with mutant TET2. These were the only two gene promoters (out of 14,475 genes) previously found to be hypermethylated in TET2 mutant cases. However, total 5-methyl-cytosine levels in TET2 mutant cases were significantly higher than TET2 wild-type cases (median = 14.0% and 9.8%, respectively) (p = 0.016). Thus, TET2 mutations affect global methylation in CMML but most of the changes are likely to be outside gene promoters.


Genome Biology | 2013

Architecture of epigenetic reprogramming following Twist1-mediated epithelial-mesenchymal transition

Gabriel G. Malouf; Joseph H. Taube; Yue Lu; Tapasree Roysarkar; Shoghag Panjarian; Marcos R. Estecio; Jaroslav Jelinek; Jumpei Yamazaki; Noël J.-M. Raynal; Hai Long; Tomomitsu Tahara; Agata A. Tinnirello; Xiu Ying Zhang; Shoudan Liang; Sendurai A. Mani; Jean-Pierre Issa

BackgroundEpithelial-mesenchymal transition (EMT) is known to impart metastasis and stemness characteristics in breast cancer. To characterize the epigenetic reprogramming following Twist1-induced EMT, we characterized the epigenetic and transcriptome landscapes using whole-genome transcriptome analysis by RNA-seq, DNA methylation by digital restriction enzyme analysis of methylation (DREAM) and histone modifications by CHIP-seq of H3K4me3 and H3K27me3 in immortalized human mammary epithelial cells relative to cells induced to undergo EMT by Twist1.ResultsEMT is accompanied by focal hypermethylation and widespread global DNA hypomethylation, predominantly within transcriptionally repressed gene bodies. At the chromatin level, the number of gene promoters marked by H3K4me3 increases by more than one fifth; H3K27me3 undergoes dynamic genomic redistribution characterized by loss at half of gene promoters and overall reduction of peak size by almost half. This is paralleled by increased phosphorylation of EZH2 at serine 21. Among genes with highly altered mRNA expression, 23.1% switch between H3K4me3 and H3K27me3 marks, and those point to the master EMT targets and regulators CDH1, PDGFRα and ESRP1. Strikingly, Twist1 increases the number of bivalent genes by more than two fold. Inhibition of the H3K27 methyltransferases EZH2 and EZH1, which form part of the Polycomb repressive complex 2 (PRC2), blocks EMT and stemness properties.ConclusionsOur findings demonstrate that the EMT program requires epigenetic remodeling by the Polycomb and Trithorax complexes leading to increased cellular plasticity. This suggests that inhibiting epigenetic remodeling and thus decrease plasticity will prevent EMT, and the associated breast cancer metastasis.


Cancer Research | 2015

TET2 mutations affect non-CpG island DNA methylation at enhancers and transcription factor binding sites in chronic myelomonocytic leukemia

Jumpei Yamazaki; Jaroslav Jelinek; Yue Lu; Matteo Cesaroni; Jozef Madzo; Frank Neumann; Rong He; Rodolphe Taby; Aparna Vasanthakumar; Trisha Macrae; Kelly R. Ostler; Hagop M. Kantarjian; Shoudan Liang; Marcos R. Estecio; Lucy A. Godley; Jean-Pierre Issa

TET2 enzymatically converts 5-methylcytosine to 5-hydroxymethylcytosine as well as other covalently modified cytosines and its mutations are common in myeloid leukemia. However, the exact mechanism and the extent to which TET2 mutations affect DNA methylation remain in question. Here, we report on DNA methylomes in TET2 wild-type (TET2-WT) and mutant (TET2-MT) cases of chronic myelomonocytic leukemia (CMML). We analyzed 85,134 CpG sites [28,114 sites in CpG islands (CGI) and 57,020 in non-CpG islands (NCGI)]. TET2 mutations do not explain genome-wide differences in DNA methylation in CMML, and we found few and inconsistent differences at CGIs between TET2-WT and TET2-MT cases. In contrast, we identified 409 (0.71%) TET2-specific differentially methylated CpGs (tet2-DMCs) in NCGIs, 86% of which were hypermethylated in TET2-MT cases, suggesting a strikingly different biology of the effects of TET2 mutations at CGIs and NCGIs. DNA methylation of tet2-DMCs at promoters and nonpromoters repressed gene expression. Tet2-DMCs showed significant enrichment at hematopoietic-specific enhancers marked by H3K4me1 and at binding sites for the transcription factor p300. Tet2-DMCs showed significantly lower 5-hydroxymethylcytosine in TET2-MT cases. We conclude that leukemia-associated TET2 mutations affect DNA methylation at NCGI regions containing hematopoietic-specific enhancers and transcription factor-binding sites.


Epigenetics | 2013

The epigenome of AML stem and progenitor cells

Jumpei Yamazaki; Marcos R. Estecio; Yue Lu; Hai Long; Gabriel G. Malouf; David Graber; Yujia Huo; Louis S. Ramagli; Shoudan Liang; Steven M. Kornblau; Jaroslav Jelinek; Jean-Pierre Issa

Acute myeloid leukemia (AML) is sustained by a population of cancer stem cells (CSCs or cancer-initiating cell). The mechanisms underlying switches from CSCs to non-CSCs in vivo remain to be understood. We address this issue in AML from the aspect of epigenetics using genome-wide screening for DNA methylation and selected histone modifications. We found no major differences in DNA methylation, especially in promoter CpG islands, between CSCs and non-CSCs. By contrast, we found thousands of genes that change H3K4me3 and/or H3K27me3 status between stem and progenitor cells as well as between progenitor and mature cells. Stem cell related pathways and proliferation or metabolism related pathways characterize genes differentially enriched for H3K4me3/H3K27me3 in stem and progenitor populations. Bivalent genes in stem cells are more plastic during differentiation and are more likely to lose H3K4me3 than to lose H3K27me3, consistent with increasingly closed chromatin state with differentiation. Our data indicates that histone modifications but not promoter DNA methylation are involved in switches from CSCs to non-CSCs in AML.


International Journal of Hematology | 2013

Epigenetic aspects of MDS and its molecular targeted therapy

Jumpei Yamazaki; Jean-Pierre Issa

The term “epigenetics” refers to clonally inherited stable variability in gene expression without underlying genetic changes. There are two well-known molecular mechanisms for epigenetic information: DNA methylation and histone modifications. Epigenetic changes have been recognized in the past decade as critical factors for physiological phenomena such as embryogenesis and the differentiation of normal cells. There is recent interest regarding the involvement of aberrant DNA methylation and histone modifications in mediating altered physiology in cancer. MDS is characterized by epigenetic changes, mutations in epigenetic regulators, and response to DNA methylation inhibitors, suggesting that epigenetic changes are unique features of MDS patients. In this article, recent progress in the understanding of MDS epigenetics and epigenetics-based therapies is reviewed.


Leukemia | 2017

A CpG island methylator phenotype in acute myeloid leukemia independent of IDH mutations and associated with a favorable outcome

Andrew D. Kelly; Heike Kroeger; Jumpei Yamazaki; Rodolphe Taby; Frank Neumann; S. Yu; Justin T. Lee; B. Patel; Y. Li; Rong He; Shoudan Liang; Yue Lu; Matteo Cesaroni; Sherry Pierce; Steven M. Kornblau; Carlos E. Bueso-Ramos; Farhad Ravandi; Hagop M. Kantarjian; Jaroslav Jelinek; Jean-Pierre Issa

Genetic changes are infrequent in acute myeloid leukemia (AML) compared with other malignancies and often involve epigenetic regulators, suggesting that an altered epigenome may underlie AML biology and outcomes. In 96 AML cases including 65 pilot samples selected for cured/not-cured, we found higher CpG island (CGI) promoter methylation in cured patients. Expanded genome-wide digital restriction enzyme analysis of methylation data revealed a CGI methylator phenotype independent of IDH1/2 mutations we term AML-CGI methylator phenotype (CIMP) (A-CIMP+). A-CIMP was associated with longer overall survival (OS) in this data set (median OS, years: A-CIMP+=not reached, CIMP-=1.17; P=0.08). For validation we used 194 samples from The Cancer Genome Atlas interrogated with Illumina 450k methylation arrays where we confirmed longer OS in A-CIMP (median OS, years: A-CIMP+=2.34, A-CIMP-=1.00; P=0.01). Hypermethylation in A-CIMP+ favored CGIs (OR: CGI/non-CGI=5.21), and while A-CIMP+ was enriched in CEBPA (P=0.002) and WT1 mutations (P=0.02), 70% of cases lacked either mutation. Hypermethylated genes in A-CIMP+ function in pluripotency maintenance, and a gene expression signature of A-CIMP was associated with outcomes in multiple data sets. We conclude that CIMP in AML cannot be explained solely by gene mutations (for example, IDH1/2, TET2), and that curability in A-CIMP+ AML should be validated prospectively.


Epigenetics | 2015

Methylome sequencing for fibrolamellar hepatocellular carcinoma depicts distinctive features

Gabriel G. Malouf; Tomomitsu Tahara; Valérie Paradis; Monique Fabre; Catherine Guettier; Jumpei Yamazaki; Hi Long; Yue Lu; Noël J.-M. Raynal; Jaroslav Jelinek; Roger Mouawad; David Khayat; Laurence Brugières; Eric Raymond; Jean-Pierre Issa

With the goal of studying epigenetic alterations in fibrolamellar hepatocellular carcinoma (FLC) and establish an associated DNA methylation signature, we analyzed LINE-1 methylation in a cohort of FLC and performed next-generation sequencing of DNA methylation in a training set of pure-FLCs and non-cirrhotic hepatocellular carcinomas (nc-HCC). DNA methylation was correlated with gene expression. Furthermore, we established and validated an epigenetic signature differentiating pure-FLC from other HCCs. LINE-1 methylation correlated with shorter recurrence-free survival and overall survival in resected pure-FLC patients. Unsupervised clustering using CG sites located in islands distinguished pure-FLC from nc-HCC. Major DNA methylation changes occurred outside promoters, mainly in gene bodies and intergenic regions located in the vicinity of liver developmental genes (i.e., SMARCA4 and RXRA). Partially methylated domains were more prone to DNA methylation changes. Furthermore, we identified several putative tumor suppressor genes (e.g., DLEU7) and oncogenes (e.g., DUSP4). While ∼70% of identified gene promoters gaining methylation were marked by bivalent histone marks (H3K4me3/H3K27me3) in embryonic stem cells, ∼70% of those losing methylation were marked by H3K4me3. Finally, we established a pure FLC DNA methylation signature and validated it in an independent dataset. Our analysis reveals a distinct epigenetic signature of pure FLC as compared to nc-HCC, with DNA methylation changes occurring in the vicinity of liver developmental genes. These data suggest new options for targeting FLC based on cancer epigenome aberrations.


Oncotarget | 2016

Mutation spectrum of TP53 gene predicts clinicopathological features and survival of gastric cancer

Tomomitsu Tahara; Tomoyuki Shibata; Yasuyuki Okamoto; Jumpei Yamazaki; Tomohiko Kawamura; Noriyuki Horiguchi; Masaaki Okubo; Naoko Nakano; Takamitsu Ishizuka; Mitsuo Nagasaka; Yoshihito Nakagawa; Naoki Ohmiya

Background and aim TP53 gene is frequently mutated in gastric cancer (GC), but the relationship with clinicopathological features and prognosis is conflicting. Here, we screened TP53 mutation spectrum of 214 GC patients in relation to their clinicopathological features and prognosis. Results TP53 nonsilent mutations were detected in 80 cases (37.4%), being frequently occurred as C:G to T:A single nucleotide transitions at 5′-CpG-3′ sites. TP53 mutations occurred more frequently in differentiated histologic type than in undifferentiated type in the early stage (48.6% vs. 7%, P=0.0006), while the mutations correlated with venous invasion among advanced stage (47.7% vs. 20.7%, P=0.04). Subset of GC with TP53 hot spot mutations (R175, G245, R248, R273, R282) presented significantly worse overall survival and recurrence free survival compared to others (both P=0.001). Methods Matched biopsies from GC and adjacent tissues from 214 patients were used for the experiment. All coding regions of TP53 gene (exon2 to exon11) were examined using Sanger sequencing. Conclusion Our data suggest that GC with TP53 mutations seems to develop as differentiated histologic type and show aggressive biological behavior such as venous invasion. Moreover, our data emphasizes the importance of discriminating TP53 hot spot mutations (R175, G245, R248, R273, R282) to predict worse overall survival and recurrence free survival of GC patients.


Journal of the National Cancer Institute | 2016

Hypomethylation of TET2 Target Genes Identifies a Curable Subset of Acute Myeloid Leukemia

Jumpei Yamazaki; Rodolphe Taby; Jaroslav Jelinek; Noël J.-M. Raynal; Matteo Cesaroni; Sherry Pierce; Steven M. Kornblau; Carlos E. Bueso-Ramos; Farhad Ravandi; Hagop M. Kantarjian; Jean-Pierre Issa

BACKGROUND Acute myeloid leukemia (AML) is curable in a subset of cases. The DNA methylation regulator TET2 is frequently mutated in AML, and we hypothesized that studying TET2-specific differentially methylated CpGs (tet2-DMCs) improves AML classification. METHODS We used bisulfite pyrosequencing to analyze the methylation status of four tet2-DMCs (SP140, MCCC1, EHMT1, and MTSS1) in a test group of 94 consecutive patients and a validation group of 92 consecutive patients treated with cytarabine-based chemotherapy. Data were analyzed with hierarchical clustering, Cox proportional hazards regression, and Kaplan-Meier analyses. All statistical tests were two-sided. RESULTS In the test cohort, hierarchical clustering analysis identified low levels of tet2-DMC methylation in 31 of 94 (33%) cases, and these had markedly longer overall survival (median survival 72+ vs 14 months, P = .002). Similar results were seen in the validation cohort. tet2-DMC-low status was shown to be an independent predictor of overall survival (hazard ratio = 0.29, P = .0002). In The Cancer Genome Atlas (TCGA) dataset where DNA methylation was analyzed by a different platform, tet2-DMC-low methylation was also associated with improved outcome (median survival = 55 vs 15 months, P = .0003) and was a better predictor of survival than mutations in TET2, IDH1, or IDH2, individually or combined. CONCLUSIONS Low levels of tet2-DMC methylation define a subgroup of AML that is highly curable and cannot be identified solely by genetic and cytogenetic analyses.


Clinical Cancer Research | 2015

Abstract B22: Genome-wide methylation analysis reveals an independently validated CpG island methylator phenotype associated with favorable prognosis in acute myeloid leukemia.

Andrew D. Kelly; Heike Kroeger; Jumpei Yamazaki; Rodolphe Taby; Frank Neumann; Justin T. Lee; Rong He; Shoudan Liang; Yue Lu; Matteo Cesaroni; Sherry Pierce; Steven M. Kornblau; Carlos E. Bueso-Ramos; Farhad Ravandi; Hagop M. Kantarjian; Jean-Pierre Issa; Jaroslav Jelinek

Background: Acute myeloid leukemia (AML) accounts for the most leukemia-related deaths in the United States and its incidence has been rising as the population ages. Although certain molecular aberrations are prognostic and have come into mainstream clinical practice, the genetic and epigenetic determinants of curability in AML remain incompletely understood. Our study examines the role of DNA methylation patterns in AML prognosis and expands on our preliminary work showing DNA hypermethylation may associate with improved overall survival. Methods: To quantitatively interrogate genome-wide CpG methylation we used Digital Restriction Enzyme Analysis of Methylation (DREAM) on a cohort of 102 AML patient samples and 25 normal control samples. We validated our findings using DNA methylation data from 194 patient samples from The Cancer Genome Atlas (TCGA) on the Illumina Infinium HumanMethylation450 platform. Statistical analysis was done using R. Results: Preliminary analysis by our group of DNA methylation levels at promoter CpG islands (CGI) of OSCP1, NPM2, OLIG2, SCGB3A1, and SLC26A4 showed significant hypermethylation in a small group of long-surviving AML patients compared to a short-surviving cohort (median OS = 2,694 days vs. 207 days). We expanded on this observation using DREAM to measure genome-wide DNA methylation in clinical AML samples and found that hierarchical clustering based on 2,537 CpG sites with a standard deviation above 20% stratified patients into three groups with significant differences in overall survival. The hypermethylated cluster had the best prognosis and seemed to be defined by hypermethylation at promoter CGIs, suggesting that a CGI methylator phenotype (CIMP) in AML may be a favorable prognostic factor (median OS: CIMP = 5,110 days, Cluster 2 = 380 days, Cluster 3 = 555 days; log-rank p=0.0162). We then validated these findings using TCGA data from AML patient samples. Hierarchical clustering on the basis of CGI promoter sites revealed three distinct groups with the CIMP cluster having significantly improved overall survival compared to the other clusters (median OS: CIMP = 761 days, Cluster 2 = 306 days, Cluster 3 = 365 days; log-rank p=0.0013). There was also a trend in overall survival when non-CGI non-promoter sites were used to cluster samples (median OS: CIMP = 593 days, Cluster 2 = 245 days, Cluster 3 = 456 days; log-rank p=0.1530). Consistent with our DREAM data, combining CGI promoter sites with non-CGI, non-promoter sites revealed a hierarchical clustering pattern of three major clusters with significant differences in overall survival (median OS: CIMP = 822 days, Cluster 2 = 365 days, Cluster 3 = 365 days; log-rank p=0.0295). Despite technical differences between platforms, there was significant overlap in the genes most proximal to differentially methylated sites between the DREAM and TCGA analyses. These common genes were significantly enriched in transcription factors, pyrimidine metabolism genes, and development genes. Interestingly, the presence of IDH1 R140 mutations was significantly greater in the CIMP clusters in both the DREAM and TCGA analyses (p Conclusions: We propose that the CIMP methylation pattern is associated with favorable prognosis in AML. We have identified a subset of methylation sites that, when interrogated, predict overall survival independent of other clinical factors. Citation Format: Andrew D. Kelly, Heike Kroeger, Jumpei Yamazaki, Rodolphe Taby, Frank Neumann, Justin T. Lee, Rong He, Shoudan Liang, Yue Lu, Matteo Cesaroni, Sherry A. Pierce, Steven M. Kornblau, Carlos E. Bueso-Ramos, Farhad Ravandi, Hagop M. Kantarjian, Jean-Pierre J. Issa, Jaroslav Jelinek. Genome-wide methylation analysis reveals an independently validated CpG island methylator phenotype associated with favorable prognosis in acute myeloid leukemia. [abstract]. In: Proceedings of the AACR Special Conference on Hematologic Malignancies: Translating Discoveries to Novel Therapies; Sep 20-23, 2014; Philadelphia, PA. Philadelphia (PA): AACR; Clin Cancer Res 2015;21(17 Suppl):Abstract nr B22.

Collaboration


Dive into the Jumpei Yamazaki's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yue Lu

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Shoudan Liang

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Rodolphe Taby

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Hagop M. Kantarjian

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Steven M. Kornblau

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Marcos R. Estecio

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Carlos E. Bueso-Ramos

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge