Network


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

Hotspot


Dive into the research topics where Takashi Akasaka is active.

Publication


Featured researches published by Takashi Akasaka.


Blood | 2010

Presence of the P2RY8-CRLF2 rearrangement is associated with a poor prognosis in non–high-risk precursor B-cell acute lymphoblastic leukemia in children treated according to the ALL-BFM 2000 protocol

Gunnar Cario; Martin Zimmermann; Renja Romey; Stefan Gesk; Inga Vater; Jochen Harbott; André Schrauder; Anja Moericke; Shai Izraeli; Takashi Akasaka; Martin J. S. Dyer; Reiner Siebert; Martin Schrappe; Martin Stanulla

High-level expression of the cytokine receptor-like factor 2 gene, CRLF2, in precursor B-cell acute lymphoblastic leukemia (pB-ALL) was shown to be caused by a translocation involving the IGH@ locus or a deletion juxtaposing CRLF2 with the P2RY8 promoter. To assess its possible prognostic value, CRLF2 expression was analyzed in 555 childhood pB-ALL patients treated according to the Acute Lymphoblastic Leukemia Berlin-Frankfurt-Münster 2000 (ALL-BFM 2000) protocol. Besides CRLF2 rearrangements, high-level CRLF2 expression was seen in cases with supernumerary copies of the CRLF2 locus. On the basis of the detection of CRLF2 rearrangements, a CRLF2 high-expression group (n = 49) was defined. This group had a 6-year relapse incidence of 31% plus or minus 8% compared with 11% plus or minus 1% in the CRLF2 low-expression group (P = .006). This difference was mainly attributable to an extremely high incidence of relapse (71% +/- 19%) in non-high-risk patients with P2RY8-CRLF2 rearrangement. The assessment of CRLF2 aberrations may therefore serve as new stratification tool in Berlin-Frankfurt-Münster-based protocols by identifying additional high-risk patients who may benefit from an intensified and/or targeted treatment.


Blood | 2010

Immunoglobulin heavy chain locus chromosomal translocations in B-cell precursor acute lymphoblastic leukemia: rare clinical curios or potent genetic drivers?

Martin J. S. Dyer; Takashi Akasaka; Melania Capasso; P. Dusanjh; Y. F. Lee; E. L. Karran; Inga Nagel; Inga Vater; G. Cario; Reiner Siebert

Chromosomal translocations involving the immunoglobulin heavy chain (IGH) locus define common subgroups of B-cell lymphoma but are rare in B-cell precursor acute lymphoblastic leukemia (BCP-ALL). Recent fluorescent in situ hybridization and molecular cloning studies have identified several novel IGH translocations involving genes that play important roles in normal hemopoiesis, including the cytokine receptor genes CRLF2 and EPOR, all members of the CCAAT enhancer-binding protein gene family, as well as genes not normally expressed in hemopoietic cells including inhibitor of DNA binding 4. IGH translocation results in deregulated target gene expression because of juxtaposition with IGH transcriptional enhancers. However, many genes targeted by IGH translocations are also more commonly deregulated in BCP-ALL as a consequence of other genetic or epigenetic mechanisms. For example, interstitial genomic deletions also result in deregulated CRLF2 expression, whereas EPOR expression is deregulated as a consequence of the ETV6-RUNX1 fusion. The possible clinical importance of many of the various IGH translocations in BCP-ALL remains to be determined from prospective studies, but CRLF2 expression is associated with a poor prognosis. Despite their rarity, IGH chromosomal translocations in BCP-ALL therefore define not only new mechanisms of B-cell transformation but also clinically important subgroups of disease and suggest new targeted therapeutic approaches.


Journal of Neuropathology and Experimental Neurology | 2006

Chromosomal translocations fusing the BCL6 gene to different partner loci are recurrent in primary central nervous system lymphoma and may be associated with aberrant somatic hypermutation or defective class switch recombination.

Heinrich Schwindt; Takashi Akasaka; Reina Zühlke-Jenisch; Volkmar Hans; Carlo Schaller; Wolfram Klapper; Martin J. S. Dyer; Reiner Siebert; Martina Deckert

Abstract Primary central nervous system lymphomas (PCNSLs) are diffuse large B cell lymphomas confined to the brain. Only minimal data exist on chromosomal aberrations underlying PCNSLs. We studied 41 PCNSLs by fluorescence in situ hybridization for breakpoints affecting the BCL6 locus in chromosomal band 3q27. Of 37 cases evaluable, 14 (38%) carried a breakpoint in the BCL6 locus. Two of these showed juxtaposition of BCL6 to the IGH locus. In 4 cases, the BCL6 breakpoints were cloned using long-distance inverse polymerase chain reaction. All breakpoints were located within the BCL6 major translocation cluster. The translocation partners were the IGH gene in 14q32.33, the IGL gene in 22q11.22, and the histone 1 H4I gene in 6p22.1. In the fourth case, a deletion in 3q leads to loss of an 837-kb fragment extending from the first intron of BCL6 to the third intron of the lipoma-preferred partner (LPP) gene. This deletion may bring the BCL6 gene under the control of regulatory elements of the LPP gene or the miRNA-28 gene located in intron 4 of LPP. DNA sequence analysis of the junctional sequences provided evidence that aberrant class switch recombination or somatic hypermutation may be involved in the generation of BCL6 translocations.


Blood | 2012

t(X;14)(p11;q32) in MALT lymphoma involving GPR34 reveals a role for GPR34 in tumor cell growth

Stephen M. Ansell; Takashi Akasaka; Ellen D. McPhail; Michelle K. Manske; Esteban Braggio; Tammy Price-Troska; Steven C. Ziesmer; Frank J. Secreto; Rafael Fonseca; Mamta Gupta; Mark E. Law; Thomas E. Witzig; Martin J. S. Dyer; Ahmet Dogan; James R. Cerhan; Anne J. Novak

Genetic aberrations, including trisomies 3 and 18, and well-defined IGH translocations, have been described in marginal zone lymphomas (MZLs); however, these known genetic events are present in only a subset of cases. Here, we report the cloning of an IGH translocation partner on chromosome X, t(X;14)(p11.4;q32) that deregulates expression of an poorly characterized orphan G-protein-coupled receptor, GPR34. Elevated GPR34 gene expression was detected independent of the translocation in multiple subtypes of non-Hodgkin lymphoma and distinguished a unique molecular subtype of MZL. Increased expression of GPR34 was also detected in tissue from brain tumors and surface expression of GPR34 was detected on human MZL tumor cells and normal immune cells. Overexpression of GPR34 in lymphoma and HeLa cells resulted in phosphorylation of ERK, PKC, and CREB; induced CRE, AP1, and NF-κB-mediated gene transcription; and increased cell proliferation. In summary, these results are the first to identify a role for a GPR34 in lymphoma cell growth, provide insight into GPR34-mediated signaling, identify a genetically unique subset of MZLs that express high levels of GPR34, and suggest that MEK inhibitors may be useful for treatment of GPR34-expressing tumors.


Blood | 2010

Deregulation of the telomerase reverse transcriptase (TERT) gene by chromosomal translocations in B-cell malignancies

Inga Nagel; Monika Szczepanowski; José I. Martín-Subero; Lana Harder; Takashi Akasaka; Ole Ammerpohl; Evelyne Callet-Bauchu; Randy D. Gascoyne; Stefan Gesk; Douglas E. Horsman; Wolfram Klapper; Aneela Majid; Jose A. Martinez-Climent; Stephan Stilgenbauer; Holger Tönnies; Martin J. S. Dyer; Reiner Siebert

Sequence variants at the TERT-CLPTM1L locus in chromosome 5p have been recently associated with disposition for various cancers. Here we show that this locus including the gene encoding the telomerase reverse-transcriptase TERT at 5p13.33 is rarely but recurrently targeted by somatic chromosomal translocations to IGH and non-IG loci in B-cell neoplasms, including acute lymphoblastic leukemia, chronic lymphocytic leukemia, mantle cell lymphoma and splenic marginal zone lymphoma. In addition, cases with genomic amplification of TERT locus were identified. Tumors bearing chromosomal aberrations involving TERT showed higher TERT transcriptional expression and increased telomerase activity. These data suggest that deregulation of TERT gene by chromosomal abnormalities leading to increased telomerase activity might contribute to B-cell lymphomagenesis.


Genes, Chromosomes and Cancer | 2012

The CBFA2T3/ACSF3 locus is recurrently involved in IGH chromosomal translocation t(14;16)(q32;q24) in pediatric B-cell lymphoma with germinal center phenotype

Itziar Salaverria; Takashi Akasaka; Stefan Gesk; Monika Szczepanowski; Birgit Burkhardt; Lana Harder; Christine Damm-Welk; Ilske Oschlies; Wolfram Klapper; Martin J. S. Dyer; Reiner Siebert

Translocations involving immunoglobulin (IG) loci are the hallmarks of several subtypes of B‐cell lymphoma. Common to these translocations is that cellular proto‐oncogenes come under the influence of IG regulatory elements leading to deregulated expression. In case of a breakpoint in the IGH switch region, oncogene activation can take place on both derivative chromosomes, which means that in principle one translocation can result in concurrent activation of two genes. By fluorescence in situ hybridization (FISH), we identified a case of leukemic B‐cell lymphoma in a child with an IGH break and unknown partner. Subsequent long‐distance inverse PCR revealed fusion of IGH Sμ in 14q32 and the 5′ region of CBFA2T3 in 16q24.3, suggesting presence of the t(14;16)(q32;q24.3). Candidate oncogenes targeted through this translocation are CBFA2T3 and ACSF3, which could be activated on der(16) and der(14), respectively. FISH screening of a population‐based cohort of B‐cell lymphomas from a prospective trial for the treatment of lymphoma in childhood (BFM‐NHL) identified additionally a follicular lymphoma Grade 3/diffuse large B‐cell lymphoma with IGH‐CBFA2T3/ACSF3 juxtaposition. Both lymphomas shared expression of CD10 and CD20 in the absence of TdT, suggesting a germinal center (GC) B‐cell origin. Our data indicate that the CBFA2T3/ACSF3 locus is a novel recurrent oncogenic target of IGH translocations, which might contribute to the pathogenesis of pediatric GC‐derived B‐cell lymphoma.


Nature Communications | 2016

Homeobox NKX2-3 promotes marginal-zone lymphomagenesis by activating B-cell receptor signalling and shaping lymphocyte dynamics

Eloy F. Robles; Maria Mena-Varas; Laura Barrio; Sara V. Merino-Cortes; Péter Balogh; Ming-Qing Du; Takashi Akasaka; Anton Parker; Sergio Roa; Carlos Panizo; Idoia Martin-Guerrero; Reiner Siebert; Victor Segura; Xabier Agirre; Laura Macri-Pellizeri; Beatriz Aldaz; Amaia Vilas-Zornoza; Shaowei Zhang; Sarah Moody; María José Calasanz; Thomas Tousseyn; Cyril Broccardo; Pierre Brousset; Elena Campos-Sanchez; Cesar Cobaleda; Isidro Sánchez-García; Jose Luis Fernandez-Luna; Ricardo García-Muñoz; Esther Pena; Beatriz Bellosillo

NKX2 homeobox family proteins have a role in cancer development. Here we show that NKX2-3 is overexpressed in tumour cells from a subset of patients with marginal-zone lymphomas, but not with other B-cell malignancies. While Nkx2-3-deficient mice exhibit the absence of marginal-zone B cells, transgenic mice with expression of NKX2-3 in B cells show marginal-zone expansion that leads to the development of tumours, faithfully recapitulating the principal clinical and biological features of human marginal-zone lymphomas. NKX2-3 induces B-cell receptor signalling by phosphorylating Lyn/Syk kinases, which in turn activate multiple integrins (LFA-1, VLA-4), adhesion molecules (ICAM-1, MadCAM-1) and the chemokine receptor CXCR4. These molecules enhance migration, polarization and homing of B cells to splenic and extranodal tissues, eventually driving malignant transformation through triggering NF-κB and PI3K-AKT pathways. This study implicates oncogenic NKX2-3 in lymphomagenesis, and provides a valid experimental mouse model for studying the biology and therapy of human marginal-zone B-cell lymphomas.


Blood | 2011

Homeobox NKX2-3 Is Over-Expressed in Human B-Cell Lymphomas and Drives Marginal Zone B-Cell Lymphomagenesis in Mice

Eloy F. Robles; Beatriz Aldaz; Takashi Akasaka; Laura Macri Pellizzeri; Eduardo Martínez-Ansó; Xavier Aguirre; Felipe Prosper; Idoia Martin-Guerrero; Victor Segura; María José Calasanz; Isidro Sánchez-García; Elena Campos-Sanchez; César Cobaleda; Charlotte Cresson; Cyril Broccardo; Reiner Siebert; Xavier Sagaert; Martin J. S. Dyer; Jose A. Martinez-Climent


Blood | 2008

Elevated Expression of GPR34 and Its Association with a Novel Translocation T(X;14)(p11;q32) Involving IGHS and GPR34 in MALT Lymphoma.

Anne Novak; Takashi Akasaka; Michelle Manske; Mamta Gupta; Thomas Witzig; Martin J. S. Dyer; Ahmet Dogan; Ellen Remstein; Stephen Ansell


Blood | 2009

Elevated Expression of GPR34 in Mucosa-Associated Lymphoid Tissue (MALT) Lymphoma and Its Association with Increased Cell Growth, Erk Activation, and AP-1 and CRE-Mediated Transcription.

Anne J. Novak; Takashi Akasaka; Michelle K. Manske; Tammy Price-Troska; Mamta Gupta; Thomas E. Witzig; Martin J. S. Dyer; Ahmet Dogan; Stephen M. Ansell

Collaboration


Dive into the Takashi Akasaka's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ahmet Dogan

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge