Network


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

Hotspot


Dive into the research topics where Ayana Kon is active.

Publication


Featured researches published by Ayana Kon.


Nature | 2011

Frequent pathway mutations of splicing machinery in myelodysplasia.

Kenichi Yoshida; Masashi Sanada; Yuichi Shiraishi; Daniel Nowak; Yasunobu Nagata; Ryo Yamamoto; Yusuke Sato; Aiko Sato-Otsubo; Ayana Kon; Masao Nagasaki; George Chalkidis; Yutaka Suzuki; Masashi Shiosaka; Ryoichiro Kawahata; Tomoyuki Yamaguchi; Makoto Otsu; Naoshi Obara; Mamiko Sakata-Yanagimoto; Ken Ishiyama; Hiraku Mori; Florian Nolte; Wolf-Karsten Hofmann; Shuichi Miyawaki; Sumio Sugano; Claudia Haferlach; H. Phillip Koeffler; Lee-Yung Shih; Torsten Haferlach; Shigeru Chiba; Hiromitsu Nakauchi

Myelodysplastic syndromes and related disorders (myelodysplasia) are a heterogeneous group of myeloid neoplasms showing deregulated blood cell production with evidence of myeloid dysplasia and a predisposition to acute myeloid leukaemia, whose pathogenesis is only incompletely understood. Here we report whole-exome sequencing of 29 myelodysplasia specimens, which unexpectedly revealed novel pathway mutations involving multiple components of the RNA splicing machinery, including U2AF35, ZRSR2, SRSF2 and SF3B1. In a large series analysis, these splicing pathway mutations were frequent (∼45 to ∼85%) in, and highly specific to, myeloid neoplasms showing features of myelodysplasia. Conspicuously, most of the mutations, which occurred in a mutually exclusive manner, affected genes involved in the 3′-splice site recognition during pre-mRNA processing, inducing abnormal RNA splicing and compromised haematopoiesis. Our results provide the first evidence indicating that genetic alterations of the major splicing components could be involved in human pathogenesis, also implicating a novel therapeutic possibility for myelodysplasia.


Leukemia | 2014

Landscape of genetic lesions in 944 patients with myelodysplastic syndromes

Torsten Haferlach; Yasunobu Nagata; Vera Grossmann; Yusuke Okuno; Ulrike Bacher; Genta Nagae; Susanne Schnittger; Masashi Sanada; Ayana Kon; Tamara Alpermann; Kenichi Yoshida; Andreas Roller; Niroshan Nadarajah; Yuichi Shiraishi; Yusuke Shiozawa; Kenichi Chiba; Hidenori Tanaka; Koeffler Hp; H-U Klein; Martin Dugas; Hiroyuki Aburatani; Alexander Kohlmann; Satoru Miyano; Claudia Haferlach; Wolfgang Kern; Seishi Ogawa

High-throughput DNA sequencing significantly contributed to diagnosis and prognostication in patients with myelodysplastic syndromes (MDS). We determined the biological and prognostic significance of genetic aberrations in MDS. In total, 944 patients with various MDS subtypes were screened for known/putative mutations/deletions in 104 genes using targeted deep sequencing and array-based genomic hybridization. In total, 845/944 patients (89.5%) harbored at least one mutation (median, 3 per patient; range, 0–12). Forty-seven genes were significantly mutated with TET2, SF3B1, ASXL1, SRSF2, DNMT3A, and RUNX1 mutated in >10% of cases. Many mutations were associated with higher risk groups and/or blast elevation. Survival was investigated in 875 patients. By univariate analysis, 25/48 genes (resulting from 47 genes tested significantly plus PRPF8) affected survival (P<0.05). The status of 14 genes combined with conventional factors revealed a novel prognostic model (‘Model-1’) separating patients into four risk groups (‘low’, ‘intermediate’, ‘high’, ‘very high risk’) with 3-year survival of 95.2, 69.3, 32.8, and 5.3% (P<0.001). Subsequently, a ‘gene-only model’ (‘Model-2’) was constructed based on 14 genes also yielding four significant risk groups (P<0.001). Both models were reproducible in the validation cohort (n=175 patients; P<0.001 each). Thus, large-scale genetic and molecular profiling of multiple target genes is invaluable for subclassification and prognostication in MDS patients.


Nature Genetics | 2013

Integrated molecular analysis of clear-cell renal cell carcinoma

Yusuke Sato; Tetsuichi Yoshizato; Yuichi Shiraishi; Shigekatsu Maekawa; Yusuke Okuno; Takumi Kamura; Teppei Shimamura; Aiko Sato-Otsubo; Genta Nagae; Hiromichi Suzuki; Yasunobu Nagata; Kenichi Yoshida; Ayana Kon; Yutaka Suzuki; Kenichi Chiba; Hiroko Tanaka; Atsushi Niida; Akihiro Fujimoto; Tatsuhiko Tsunoda; Teppei Morikawa; Daichi Maeda; Haruki Kume; Sumio Sugano; Masashi Fukayama; Hiroyuki Aburatani; Masashi Sanada; Satoru Miyano; Yukio Homma; Seishi Ogawa

Clear-cell renal cell carcinoma (ccRCC) is the most prevalent kidney cancer and its molecular pathogenesis is incompletely understood. Here we report an integrated molecular study of ccRCC in which ≥100 ccRCC cases were fully analyzed by whole-genome and/or whole-exome and RNA sequencing as well as by array-based gene expression, copy number and/or methylation analyses. We identified a full spectrum of genetic lesions and analyzed gene expression and DNA methylation signatures and determined their impact on tumor behavior. Defective VHL-mediated proteolysis was a common feature of ccRCC, which was caused not only by VHL inactivation but also by new hotspot TCEB1 mutations, which abolished Elongin C–VHL binding, leading to HIF accumulation. Other newly identified pathways and components recurrently mutated in ccRCC included PI3K-AKT-mTOR signaling, the KEAP1-NRF2-CUL3 apparatus, DNA methylation, p53-related pathways and mRNA processing. This integrated molecular analysis unmasked new correlations between DNA methylation, gene mutation and/or gene expression and copy number profiles, enabling the stratification of clinical risks for patients with ccRCC.


Nature Genetics | 2013

Recurrent mutations in multiple components of the cohesin complex in myeloid neoplasms

Ayana Kon; Lee-Yung Shih; Masashi Minamino; Masashi Sanada; Yuichi Shiraishi; Yasunobu Nagata; Kenichi Yoshida; Yusuke Okuno; Masashige Bando; Ryuichiro Nakato; Shumpei Ishikawa; Aiko Sato-Otsubo; Genta Nagae; Aiko Nishimoto; Claudia Haferlach; Daniel Nowak; Yusuke Sato; Tamara Alpermann; Masao Nagasaki; Teppei Shimamura; Hiroko Tanaka; Kenichi Chiba; Ryo Yamamoto; Tomoyuki Yamaguchi; Makoto Otsu; Naoshi Obara; Mamiko Sakata-Yanagimoto; Tsuyoshi Nakamaki; Ken Ishiyama; Florian Nolte

Cohesin is a multimeric protein complex that is involved in the cohesion of sister chromatids, post-replicative DNA repair and transcriptional regulation. Here we report recurrent mutations and deletions involving multiple components of the cohesin complex, including STAG2, RAD21, SMC1A and SMC3, in different myeloid neoplasms. These mutations and deletions were mostly mutually exclusive and occurred in 12.1% (19/157) of acute myeloid leukemia, 8.0% (18/224) of myelodysplastic syndromes, 10.2% (9/88) of chronic myelomonocytic leukemia, 6.3% (4/64) of chronic myelogenous leukemia and 1.3% (1/77) of classical myeloproliferative neoplasms. Cohesin-mutated leukemic cells showed reduced amounts of chromatin-bound cohesin components, suggesting a substantial loss of cohesin binding sites on chromatin. The growth of leukemic cell lines harboring a mutation in RAD21 (Kasumi-1 cells) or having severely reduced expression of RAD21 and STAG2 (MOLM-13 cells) was suppressed by forced expression of wild-type RAD21 and wild-type RAD21 and STAG2, respectively. These findings suggest a role for compromised cohesin functions in myeloid leukemogenesis.


The New England Journal of Medicine | 2015

Somatic Mutations and Clonal Hematopoiesis in Aplastic Anemia.

Tetsuichi Yoshizato; Bogdan Dumitriu; Kohei Hosokawa; Hideki Makishima; Kenichi Yoshida; Danielle M. Townsley; Aiko Sato-Otsubo; Yusuke Sato; Delong Liu; Hiromichi Suzuki; Colin O. Wu; Yuichi Shiraishi; Michael J. Clemente; Keisuke Kataoka; Yusuke Shiozawa; Yusuke Okuno; Kenichi Chiba; Hiroko Tanaka; Yasunobu Nagata; Takamasa Katagiri; Ayana Kon; Masashi Sanada; Phillip Scheinberg; Satoru Miyano; Jaroslaw P. Maciejewski; Shinji Nakao; Neal S. Young; Seishi Ogawa

BACKGROUND In patients with acquired aplastic anemia, destruction of hematopoietic cells by the immune system leads to pancytopenia. Patients have a response to immunosuppressive therapy, but myelodysplastic syndromes and acute myeloid leukemia develop in about 15% of the patients, usually many months to years after the diagnosis of aplastic anemia. METHODS We performed next-generation sequencing and array-based karyotyping using 668 blood samples obtained from 439 patients with aplastic anemia. We analyzed serial samples obtained from 82 patients. RESULTS Somatic mutations in myeloid cancer candidate genes were present in one third of the patients, in a limited number of genes and at low initial variant allele frequency. Clonal hematopoiesis was detected in 47% of the patients, most frequently as acquired mutations. The prevalence of the mutations increased with age, and mutations had an age-related signature. DNMT3A-mutated and ASXL1-mutated clones tended to increase in size over time; the size of BCOR- and BCORL1-mutated and PIGA-mutated clones decreased or remained stable. Mutations in PIGA and BCOR and BCORL1 correlated with a better response to immunosuppressive therapy and longer and a higher rate of overall and progression-free survival; mutations in a subgroup of genes that included DNMT3A and ASXL1 were associated with worse outcomes. However, clonal dynamics were highly variable and might not necessarily have predicted the response to therapy and long-term survival among individual patients. CONCLUSIONS Clonal hematopoiesis was prevalent in aplastic anemia. Some mutations were related to clinical outcomes. A highly biased set of mutations is evidence of Darwinian selection in the failed bone marrow environment. The pattern of somatic clones in individual patients over time was variable and frequently unpredictable. (Funded by Grant-in-Aid for Scientific Research and others.).


Nature Genetics | 2013

The landscape of somatic mutations in Down syndrome–related myeloid disorders

Kenichi Yoshida; Tsutomu Toki; Yusuke Okuno; Rika Kanezaki; Yuichi Shiraishi; Aiko Sato-Otsubo; Masashi Sanada; Myoung-ja Park; Kiminori Terui; Hiromichi Suzuki; Ayana Kon; Yasunobu Nagata; Yusuke Sato; Ru Nan Wang; Norio Shiba; Kenichi Chiba; Hiroko Tanaka; Asahito Hama; Hideki Muramatsu; Daisuke Hasegawa; Kazuhiro Nakamura; Hirokazu Kanegane; Keiko Tsukamoto; Souichi Adachi; Kiyoshi Kawakami; Koji Kato; Ryosei Nishimura; Shai Izraeli; Yasuhide Hayashi; Satoru Miyano

Transient abnormal myelopoiesis (TAM) is a myeloid proliferation resembling acute megakaryoblastic leukemia (AMKL), mostly affecting perinatal infants with Down syndrome. Although self-limiting in a majority of cases, TAM may evolve as non-self-limiting AMKL after spontaneous remission (DS-AMKL). Pathogenesis of these Down syndrome–related myeloid disorders is poorly understood, except for GATA1 mutations found in most cases. Here we report genomic profiling of 41 TAM, 49 DS-AMKL and 19 non-DS-AMKL samples, including whole-genome and/or whole-exome sequencing of 15 TAM and 14 DS-AMKL samples. TAM appears to be caused by a single GATA1 mutation and constitutive trisomy 21. Subsequent AMKL evolves from a pre-existing TAM clone through the acquisition of additional mutations, with major mutational targets including multiple cohesin components (53%), CTCF (20%), and EZH2, KANSL1 and other epigenetic regulators (45%), as well as common signaling pathways, such as the JAK family kinases, MPL, SH2B3 (LNK) and multiple RAS pathway genes (47%).


Nature Genetics | 2013

Exome sequencing identifies secondary mutations of SETBP1 and JAK3 in juvenile myelomonocytic leukemia

Hirotoshi Sakaguchi; Yusuke Okuno; Hideki Muramatsu; Kenichi Yoshida; Yuichi Shiraishi; Mariko Takahashi; Ayana Kon; Masashi Sanada; Kenichi Chiba; Hiroko Tanaka; Hideki Makishima; Xinan Wang; Yinyan Xu; Sayoko Doisaki; Asahito Hama; Koji Nakanishi; Yoshiyuki Takahashi; Nao Yoshida; Jaroslaw P. Maciejewski; Satoru Miyano; Seishi Ogawa; Seiji Kojima

Juvenile myelomonocytic leukemia (JMML) is an intractable pediatric leukemia with poor prognosis whose molecular pathogenesis is poorly understood, except for somatic or germline mutations of RAS pathway genes, including PTPN11, NF1, NRAS, KRAS and CBL, in the majority of cases. To obtain a complete registry of gene mutations in JMML, whole-exome sequencing was performed for paired tumor-normal DNA from 13 individuals with JMML (cases), which was followed by deep sequencing of 8 target genes in 92 tumor samples. JMML was characterized by a paucity of gene mutations (0.85 non-silent mutations per sample) with somatic or germline RAS pathway involvement in 82 cases (89%). The SETBP1 and JAK3 genes were among common targets for secondary mutations. Mutations in the latter were often subclonal and may be involved in the progression rather than the initiation of leukemia, and these mutations associated with poor clinical outcome. Our findings provide new insights into the pathogenesis and progression of JMML.


Science | 2014

Recurrent somatic mutations underlie corticotropin-independent Cushing’s syndrome

Yusuke Sato; Shigekatsu Maekawa; Ryohei Ishii; Masashi Sanada; Teppei Morikawa; Yuichi Shiraishi; Kenichi Yoshida; Yasunobu Nagata; Aiko Sato-Otsubo; Tetsuichi Yoshizato; Hiromichi Suzuki; Yusuke Shiozawa; Keisuke Kataoka; Ayana Kon; Kosuke Aoki; Kenichi Chiba; Hiroko Tanaka; Haruki Kume; Satoru Miyano; Masashi Fukayama; Osamu Nureki; Yukio Homma; Seishi Ogawa

Cushing’s syndrome is caused by excess cortisol production from the adrenocortical gland. In corticotropin-independent Cushing’s syndrome, the excess cortisol production is primarily attributed to an adrenocortical adenoma, in which the underlying molecular pathogenesis has been poorly understood. We report a hotspot mutation (L206R) in PRKACA, which encodes the catalytic subunit of cyclic adenosine monophosphate (cAMP)–dependent protein kinase (PKA), in more than 50% of cases with adrenocortical adenomas associated with corticotropin-independent Cushing’s syndrome. The L206R PRKACA mutant abolished its binding to the regulatory subunit of PKA (PRKAR1A) that inhibits catalytic activity of PRKACA, leading to constitutive, cAMP-independent PKA activation. These results highlight the major role of cAMP-independent activation of cAMP/PKA signaling by somatic mutations in corticotropin-independent Cushing’s syndrome, providing insights into the diagnosis and therapeutics of this syndrome. Adrenal Cushing’s syndrome involves recurrent mutations in a key signal transduction pathway [Also see Perspective by Kirschner] Candidate Cushings culprit identified Cushings syndrome is a rare condition resulting from the excess production of cortisol. About 15% of Cushings syndrome cases are associated with an adrenocortical tumor. However, the genetic etiology of these adrenocortical tumors is ill defined (see the Perspective by Kirschner). Cao et al. and Sato et al. both performed whole-exome sequencing of tumors from individuals with adrenal Cushings syndrome and compared it with the patients own matched non-tumor DNA and identified recurrent mutations in the protein kinase A catalytic subunit alpha (PRKACA) gene, as well as less frequent mutations in other putative pathological genes. The most common recurrent mutation activated the kinase, which may suggest a potential therapeutic target. Science, this issue p. 913, p. 917; see also p. 804


Blood | 2016

Variegated RHOA mutations in adult T-cell leukemia/lymphoma

Yasunobu Nagata; Kenji Kontani; Terukazu Enami; Keisuke Kataoka; Ryohei Ishii; Yasushi Totoki; Tatsuki R. Kataoka; Masahiro Hirata; Kazuhiro Aoki; Kazumi Nakano; Akira Kitanaka; Mamiko Sakata-Yanagimoto; Sachiko Egami; Yuichi Shiraishi; Kenichi Chiba; Hiroko Tanaka; Yusuke Shiozawa; Tetsuichi Yoshizato; Hiromichi Suzuki; Ayana Kon; Kenichi Yoshida; Yusuke Sato; Aiko Sato-Otsubo; Masashi Sanada; Wataru Munakata; Hiromi Nakamura; Natsuko Hama; Satoru Miyano; Osamu Nureki; Tatsuhiro Shibata

Adult T-cell leukemia/lymphoma (ATLL) is a distinct form of peripheral T-cell lymphoma with poor prognosis, which is caused by the human T-lymphotropic virus type 1 (HTLV-1). In contrast to the unequivocal importance of HTLV-1 infection in the pathogenesis of ATLL, the role of acquired mutations in HTLV-1 infected T cells has not been fully elucidated, with a handful of genes known to be recurrently mutated. In this study, we identified unique RHOA mutations in ATLL through whole genome sequencing of an index case, followed by deep sequencing of 203 ATLL samples. RHOA mutations showed distinct distribution and function from those found in other cancers. Involving 15% (30/203) of ATLL cases, RHOA mutations were widely distributed across the entire coding sequence but almost invariably located at the guanosine triphosphate (GTP)-binding pocket, with Cys16Arg being most frequently observed. Unexpectedly, depending on mutation types and positions, these RHOA mutants showed different or even opposite functional consequences in terms of GTP/guanosine diphosphate (GDP)-binding kinetics, regulation of actin fibers, and transcriptional activation. The Gly17Val mutant did not bind GTP/GDP and act as a dominant negative molecule, whereas other mutants (Cys16Arg and Ala161Pro) showed fast GTP/GDP cycling with enhanced transcriptional activation. These findings suggest that both loss- and gain-of-RHOA functions could be involved in ATLL leukemogenesis. In summary, our study not only provides a novel insight into the molecular pathogenesis of ATLL but also highlights a unique role of variegation of heterologous RHOA mutations in human cancers.


Nature Genetics | 2017

Recurrent SPI1 (PU.1) fusions in high-risk pediatric T cell acute lymphoblastic leukemia

Masafumi Seki; Shunsuke Kimura; Tomoya Isobe; Kenichi Yoshida; Hiroo Ueno; Yaeko Nakajima-Takagi; Changshan Wang; Lin Lin; Ayana Kon; Hiromichi Suzuki; Yusuke Shiozawa; Keisuke Kataoka; Yoichi Fujii; Yuichi Shiraishi; Kenichi Chiba; Hiroko Tanaka; Teppei Shimamura; Kyoko Masuda; Hiroshi Kawamoto; Kentaro Ohki; Motohiro Kato; Yuki Arakawa; Katsuyoshi Koh; Ryoji Hanada; Hiroshi Moritake; Masaharu Akiyama; Ryoji Kobayashi; Takao Deguchi; Yoshiko Hashii; Toshihiko Imamura

The outcome of treatment-refractory and/or relapsed pediatric T cell acute lymphoblastic leukemia (T-ALL) is extremely poor, and the genetic basis for this is not well understood. Here we report comprehensive profiling of 121 cases of pediatric T-ALL using transcriptome and/or targeted capture sequencing, through which we identified new recurrent gene fusions involving SPI1 (STMN1-SPI1 and TCF7-SPI1). Cases positive for fusions involving SPI1 (encoding PU.1), accounting for 3.9% (7/181) of the examined pediatric T-ALL cases, showed a double-negative (DN; CD4−CD8−) or CD8+ single-positive (SP) phenotype and had uniformly poor overall survival. These cases represent a subset of pediatric T-ALL distinguishable from the known T-ALL subsets in terms of expression of genes involved in T cell precommitment, establishment of T cell identity, and post-β-selection maturation and with respect to mutational profile. PU.1 fusion proteins retained transcriptional activity and, when constitutively expressed in mouse stem/progenitor cells, induced cell proliferation and resulted in a maturation block. Our findings highlight a unique role of SPI1 fusions in high-risk pediatric T-ALL.

Collaboration


Dive into the Ayana Kon's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge