Network


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

Hotspot


Dive into the research topics where Yoshiki Akatsuka is active.

Publication


Featured researches published by Yoshiki Akatsuka.


Nature | 2009

Frequent inactivation of A20 in B-cell lymphomas

Motohiro Kato; Masashi Sanada; Itaru Kato; Yasuharu Sato; Junko Takita; Kengo Takeuchi; Akira Niwa; Yuyan Chen; Kumi Nakazaki; Junko Nomoto; Yoshitaka Asakura; Satsuki Muto; Azusa Tamura; Mitsuru Iio; Yoshiki Akatsuka; Yasuhide Hayashi; Hiraku Mori; Takashi Igarashi; Mineo Kurokawa; Shigeru Chiba; Shigeo Mori; Yuichi Ishikawa; Koji Okamoto; Kensei Tobinai; Hitoshi Nakagama; Tatsutoshi Nakahata; Tadashi Yoshino; Yukio Kobayashi; Seishi Ogawa

A20 is a negative regulator of the NF-κB pathway and was initially identified as being rapidly induced after tumour-necrosis factor-α stimulation. It has a pivotal role in regulation of the immune response and prevents excessive activation of NF-κB in response to a variety of external stimuli; recent genetic studies have disclosed putative associations of polymorphic A20 (also called TNFAIP3) alleles with autoimmune disease risk. However, the involvement of A20 in the development of human cancers is unknown. Here we show, using a genome-wide analysis of genetic lesions in 238 B-cell lymphomas, that A20 is a common genetic target in B-lineage lymphomas. A20 is frequently inactivated by somatic mutations and/or deletions in mucosa-associated tissue lymphoma (18 out of 87; 21.8%) and Hodgkin’s lymphoma of nodular sclerosis histology (5 out of 15; 33.3%), and, to a lesser extent, in other B-lineage lymphomas. When re-expressed in a lymphoma-derived cell line with no functional A20 alleles, wild-type A20, but not mutant A20, resulted in suppression of cell growth and induction of apoptosis, accompanied by downregulation of NF-κB activation. The A20-deficient cells stably generated tumours in immunodeficient mice, whereas the tumorigenicity was effectively suppressed by re-expression of A20. In A20-deficient cells, suppression of both cell growth and NF-κB activity due to re-expression of A20 depended, at least partly, on cell-surface-receptor signalling, including the tumour-necrosis factor receptor. Considering the physiological function of A20 in the negative modulation of NF-κB activation induced by multiple upstream stimuli, our findings indicate that uncontrolled signalling of NF-κB caused by loss of A20 function is involved in the pathogenesis of subsets of B-lineage lymphomas.


Journal of Experimental Medicine | 2003

Restoration of CD28 Expression in CD28- CD8+ Memory Effector T Cells Reconstitutes Antigen-induced IL-2 Production

Max S. Topp; Stanley R. Riddell; Yoshiki Akatsuka; Michael C. Jensen; Joseph N. Blattman; Philip D. Greenberg

The control of many persistent viral infections by Ag-specific cytolytic CD8+ T cells requires a concurrent virus-specific CD4+ Th cell response. This reflects in part a requirement of activated effector CD8+ T cells for paracrine IL-2 production as a growth and survival factor. In human CMV and HIV infection, the majority of differentiated virus-specific CD8+ T cells notably lose the ability to produce IL-2 but also lose expression of CD28, a costimulatory molecule. Analysis of the fraction of memory CD8+ T cells that continue to express CD28 revealed these cells retain the ability to produce IL-2. Therefore, we examined if IL-2 production by CD28− CD8+ T cells could be restored by introduction of a constitutively expressed CD28 gene. Expression of CD28 in CD28− CD8+ CMV- and HIV-specific CD8+ T cells reconstituted the ability to produce IL-2, which could sustain an autocrine proliferative response after Ag recognition. These results suggest that the loss of CD28 expression during differentiation of memory/effector CD8+ T cells represents a decisive step in establishing regulation of responding CD8+ T cells, increasing the dependence on CD4+ Th for proliferation after target recognition, and has implications for the treatment of viral disease with adoptively transferred CD8+ T cells.


Clinical Cancer Research | 2004

The CC Chemokine Receptor 4 as a Novel Specific Molecular Target for Immunotherapy in Adult T-Cell Leukemia/Lymphoma

Takashi Ishida; Shinsuke Iida; Yoshiki Akatsuka; Toshihiko Ishii; Mikinori Miyazaki; Hirokazu Komatsu; Hiroshi Inagaki; Noriko Okada; Teizo Fujita; Kenya Shitara; Shiro Akinaga; Toshitada Takahashi; Atae Utsunomiya; Ryuzo Ueda

Adult T-cell leukemia/lymphoma (ATLL) is a peripheral T-cell neoplasm with dismal prognosis, and no optimal therapy has been developed. We tested the defucosylated chimeric anti-CC chemokine receptor 4 (CCR4) monoclonal antibody, KM2760, to develop a novel immunotherapy for this refractory tumor. In the presence of peripheral blood mononuclear cells (PBMCs) from healthy adult donors, KM2760 induced CCR4-specific antibody-dependent cellular cytotoxicity (ADCC) against CCR4-positive ATLL cell lines and primary tumor cells obtained from ATLL patients. We next examined the KM2760-induced ADCC against primary ATLL cells in an autologous setting. Antibody-dependent cellular cytotoxicity mediated by autologous effector cells was generally lower than that mediated by allogeneic control effector cells. However, a robust ADCC activity was induced in some cases, which was comparable with that mediated by allogeneic effector cells. It suggests that the ATLL patients’ PBMCs retain substantial ADCC-effector function, although the optimal conditions for maximal effect have not yet been determined. In addition, we also found a high expression of FoxP3 mRNA and protein, a hallmark of regulatory T cells, in ATLL cells, indicating the possibility that ATLL cells originated from regulatory T cells. KM2760 reduced FoxP3 mRNA expression in normal PBMCs along with CCR4 mRNA by lysis of CCR4+ T cells in vitro. Our data suggest not only that the CCR4 molecule could be a suitable target for the novel antibody-based therapy for patients with ATLL but also that KM2760 may induce effective tumor immunity by reducing the number of regulatory T cells.


Journal of Immunology | 2002

Efficient Generation of Antigen-Specific Cytotoxic T Cells Using Retrovirally Transduced CD40-Activated B Cells

Eisei Kondo; Max S. Topp; Hans Peter Kiem; Yuichi Obata; Yasuo Morishima; Kiyotaka Kuzushima; Mitsune Tanimoto; Mine Harada; Toshitada Takahashi; Yoshiki Akatsuka

The development of rapid, efficient, and safe methods for generating Ag-specific T cells is necessary for the clinical application of adoptive immunotherapy. We show that B cells stimulated with CD40 ligand and IL-4 (CD40-B cells) can be efficiently transduced with retroviral vectors encoding a model Ag, CMV tegument protein pp65 gene, and maintain high levels of costimulatory molecules after gene transfer. CTL lines specific for pp65 were readily generated in all four healthy CMV-seropositive donors by stimulating autologous CD8+ T cells with these transduced CD40-B cells, both of which were derived from 10 ml peripheral blood. ELISPOT assays revealed that the CTL lines used multiple HLA alleles as restricting elements. Thus, CD40-B cells transduced retrovirally with Ag-encoding cDNA can be potent APC and facilitate to generate Ag-specific CTL in vitro.


Blood | 2010

Relapse of leukemia with loss of mismatched HLA resulting from uniparental disomy after haploidentical hematopoietic stem cell transplantation.

Itzel Bustos Villalobos; Yoshiyuki Takahashi; Yoshiki Akatsuka; Hideki Muramatsu; Nobuhiro Nishio; Asahito Hama; Hiroshi Yagasaki; Hiroh Saji; Motohiro Kato; Seishi Ogawa; Seiji Kojima

We investigated human leukocyte antigen (HLA) expression on leukemic cells derived from patients at diagnosis and relapse after hematopoietic stem cell transplantation (HSCT) using flow cytometry with locus-specific antibodies. Two of 3 patients who relapsed after HLA-haploidentical HSCT demonstrated loss of HLA alleles in leukemic cells at relapse; on the other hand, no loss of HLA alleles was seen in 6 patients who relapsed after HLA-identical HSCT. Single-nucleotide polymorphism array analyses of sorted leukemic cells further revealed the copy number-neutral loss of heterozygosity, namely, acquired uniparental disomy on the short arm of chromosome 6, resulting in the total loss of the mismatched HLA haplotype. These results suggest that the escape from immunosurveillance by the loss of mismatched HLA alleles may be a crucial mechanism of relapse after HLA-haploidentical HSCT. Accordingly, the status of mismatched HLA on relapsed leukemic cells should be checked before donor lymphocyte infusion.


Journal of Immunology | 2004

A Novel HLA-A*3303-Restricted Minor Histocompatibility Antigen Encoded by an Unconventional Open Reading Frame of Human TMSB4Y Gene

Hiroki Torikai; Yoshiki Akatsuka; Mikinori Miyazaki; Edus H. Warren; Taku Oba; Kunio Tsujimura; Kazuo Motoyoshi; Yasuo Morishima; Yoshihisa Kodera; Kiyotaka Kuzushima; Toshitada Takahashi

Female-to-male hemopoietic stem cell transplantation (HSCT) elicits T cell responses against male-specific minor histocompatibility (H-Y) Ags encoded by the Y chromosome. All previously identified H-Y Ags are encoded by conventional open reading frames, but we report in this study the identification of a novel H-Y Ag encoded in the 5′-untranslated region of the TMSB4Y gene. An HLA-A*3303-restricted CD8+ CTL clone was isolated from a male patient after an HSCT from his HLA-identical sister. Using a panel of cell lines carrying Y chromosome terminal deletions, a narrow region controlling the susceptibility of these target cells to CTL recognition was localized. Minigene transfection and epitope reconstitution assays identified an 11-mer peptide, EVLLRPGLHFR, designated TMSB4Y/A33, whose first amino acid was located 405 bp upstream of the TMSB4Y initiation codon. Analysis of the precursor frequency of CTL specific for recipient minor histocompatibility Ags in post-HSCT peripheral blood T cells revealed that a significant fraction of the total donor CTL response in this patient was directed against the TMSB4Y epitope. Tetramer analysis continued to detect TMSB4Y/A33-specific CD8+ T cells at least up to 700 days post-HSCT. This finding underscores the in vivo immunological relevance of minor histocompatibility Ags derived from unconventional open reading frame products.


Immunology | 1999

Role of mitogen-activated protein kinases in activation-induced apoptosis of T cells

L Zhu; X Yu; Yoshiki Akatsuka; J A Cooper; Claudio Anasetti

A member of the mitogen‐activated protein (MAP) kinase family, Jun N‐terminal kinase (JNK), has been implicated in regulating apoptosis in various cell types. We have investigated the requirement for another type of MAP kinase, extracellular signal‐regulated protein kinase (ERK) in activation‐induced cell death (AICD) of T cells. AICD is the process by which recently activated T cells undergo apoptosis when restimulated through the T‐cell antigen receptor. Here we show that both JNK and ERK are activated rapidly upon T‐cell receptor (TCR) ligation prior to the onset of AICD. A chemical inhibitor of ERK activation, PD 098059, inhibits ERK activation and apoptosis, while JNK activation is not inhibited. This suggests that JNK activation is not sufficient for apoptosis. TCR cross‐linking induces expression of the apoptosis‐inducing factor, Fas ligand (FasL), and its expression correlates with ERK activation. In addition, apoptosis induced by direct ligation of the Fas receptor by anti‐Fas antibody is not associated with ERK activation and is not inhibited by PD 098059. These data suggest that ERK activation is an early event during T‐cell apoptosis induced by antigen–receptor ligation, and is not involved in apoptosis per se but in the expression of FasL. MAP kinase family members may be similarly involved in inducing apoptosis signals in other cell types.


British Journal of Haematology | 2003

Disparity for a newly identified minor histocompatibility antigen, HA‐8, correlates with acute graft‐versus‐host disease after haematopoietic stem cell transplantation from an HLA‐identical sibling

Yoshiki Akatsuka; Edus H. Warren; Ted Gooley; Anthony G. Brickner; Ming Tseh Lin; John A. Hansen; Paul J. Martin; David K. Madtes; Victor H. Engelhard; Toshitada Takahashi; Stanley R. Riddell

Summary.  We recently identified a new minor histocompatibility antigen, termed HA‐8, which is presented by human leucocyte antigen (HLA)‐A*0201 or HLA‐A*0202 and expressed ubiquitously among tissues. A retrospective analysis of 577 Caucasian patients with HLA‐A*0201 or A*0202 who had received a haematopoietic stem cell transplant from a human leucocyte antigen (HLA)‐identical sibling was conducted to determine whether HA‐8 disparity correlated with clinical outcome. HA‐8 disparity was detected in 72 recipients, and grades II–IV graft‐versus‐host disease (GVHD) occurred in 46 (64%), compared with 251 (50%) of the 503 patients without HA‐8 disparity. After adjusting for known risk factors for acute GVHD, this difference was statistically significant (odds ratio, 1·8; 95% confidence interval, 1·0–3·1; P = 0·04). However, the hazards of clinical extensive chronic GVHD, overall mortality and recurrent malignancy were not statistically significantly different between the two groups. These data suggest that the increased risk of acute GVHD associated with recipient HA‐8 disparity was not sufficient to change other clinical outcomes.


Blood | 2008

DDX3Y encodes a class I MHC–restricted H-Y antigen that is expressed in leukemic stem cells

Kellie V. Rosinski; Nobuharu Fujii; Jeffrey K. Mito; Kevin K. W. Koo; Suzanne M. Xuereb; Olga Sala-Torra; James S. Gibbs; Jerald P. Radich; Yoshiki Akatsuka; Benoît Van den Eynde; Stanley R. Riddell; Edus H. Warren

The Y chromosome encodes male-specific minor histocompatibility (H-Y) antigens that stimulate T- and B-lymphocyte responses after sex-mismatched allogeneic hematopoietic cell transplantation (HCT). A CD8(+) cytotoxic T lymphocyte (CTL) clone that recognizes a novel HLA-B*2705-restricted H-Y antigen encoded by the DDX3Y gene was isolated from a male who had received a hematopoietic cell graft from his human leukocyte antigen (HLA)-identical sister. The antigenic peptide is a decamer that differs from the homologous DDX3X-encoded peptide at 4 positions. Expression of DDX3Y and of the H-Y epitope that it encodes was examined by quantitative polymerase chain reaction (PCR) and by CTL recognition assays. Expression of DDX3Y is detected in all myeloid and lymphoid leukemic cells that carry an intact Y chromosome. Moreover, the DDX3Y-encoded H-Y epitope is presented on the surface of both myeloid and lymphoid leukemic cells from male HLA-B*2705(+) patients. DDX3Y-specific CTLs prevent engraftment of human acute leukemia in nonobese diabetic/severe combined immune deficient mice, demonstrating that the DDX3Y-encoded H-Y antigen is also expressed in leukemic stem cells. These results demonstrate that CD8(+) T-cell responses against DDX3Y have the potential to contribute to graft-versus-leukemia (GVL) activity after female into male allogeneic HCT. This study is registered at http://clinicaltrials.gov as NCT00107354.


Blood | 2009

HapMap scanning of novel human minor histocompatibility antigens

Michi Kamei; Yasuhito Nannya; Hiroki Torikai; Takakazu Kawase; Kenjiro Taura; Yoshihiro Inamoto; Taro Takahashi; Makoto Yazaki; Satoko Morishima; Kunio Tsujimura; Koichi Miyamura; Tetsuya Ito; Hajime Togari; Stanley R. Riddell; Yoshihisa Kodera; Yasuo Morishima; Toshitada Takahashi; Kiyotaka Kuzushima; Seishi Ogawa; Yoshiki Akatsuka

Minor histocompatibility antigens (mHags) are molecular targets of allo-immunity associated with hematopoietic stem cell transplantation (HSCT) and involved in graft-versus-host disease, but they also have beneficial antitumor activity. mHags are typically defined by host SNPs that are not shared by the donor and are immunologically recognized by cytotoxic T cells isolated from post-HSCT patients. However, the number of molecularly identified mHags is still too small to allow prospective studies of their clinical importance in transplantation medicine, mostly due to the lack of an efficient method for isolation. Here we show that when combined with conventional immunologic assays, the large data set from the International HapMap Project can be directly used for genetic mapping of novel mHags. Based on the immunologically determined mHag status in HapMap panels, a target mHag locus can be uniquely mapped through whole genome association scanning taking advantage of the unprecedented resolution and power obtained with more than 3 000 000 markers. The feasibility of our approach could be supported by extensive simulations and further confirmed by actually isolating 2 novel mHags as well as 1 previously identified example. The HapMap data set represents an invaluable resource for investigating human variation, with obvious applications in genetic mapping of clinically relevant human traits.

Collaboration


Dive into the Yoshiki Akatsuka's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yasuo Morishima

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nobuhiko Emi

Fujita Health University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stanley R. Riddell

Fred Hutchinson Cancer Research Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge