41 results on '"Masaaki Sekine"'
Search Results
2. TP53 and PTEN mutations were shared in concurrent germ cell tumor and acute megakaryoblastic leukemia
- Author
-
Keiichi Akizuki, Masaaki Sekine, Yasunori Kogure, Takuro Kameda, Kotaro Shide, Junji Koya, Ayako Kamiunten, Yoko Kubuki, Yuki Tahira, Tomonori Hidaka, Takumi Kiwaki, Hiroyuki Tanaka, Yuichiro Sato, Hiroaki Kataoka, Keisuke Kataoka, and Kazuya Shimoda
- Subjects
Acute myeloid leukemia ,Germ cell tumor ,TP53 ,PTEN ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background The occurrence of a mediastinal germ cell tumor (GCT) and hematological malignancy in the same patient is very rare. Due to its rarity, there have been only two reports of the concurrent cases undergoing detailed genetic analysis with whole-exome sequencing (WES), and the possible clonal relationship between the both tumors remained not fully elucidated. Methods We performed whole-exome sequencing analysis of mediastinal GCT and acute myeloid leukemia (AML) samples obtained from one young Japanese male adult patient with concurrent both tumors, and investigated the possible clonal relationship between them. Results Sixteen somatic mutations were detected in the mediastinal GCT sample and 18 somatic mutations in the AML sample. Mutations in nine genes, including TP53 and PTEN both known as tumor suppressor genes, were shared in both tumors. Conclusions All in our case and in the previous two cases with concurrent mediastinal GCT and AML undergoing with whole-exome sequencing analysis, TP53 and PTEN mutations were commonly shared in both tumors. These data not only suggest that these tumors share a common founding clone, but also indicate that associated mediastinal GCT and AML harboring TP53 and PTEN mutations represent a unique biological entity.
- Published
- 2020
- Full Text
- View/download PDF
3. Prognosis of Indolent Adult T-Cell Leukemia/Lymphoma
- Author
-
Takuro Kameda, Kotaro Shide, Yuki Tahira, Masaaki Sekine, Seiichi Sato, Junzo Ishizaki, Masanori Takeuchi, Keiichi Akizuki, Ayako Kamiunten, Haruko Shimoda, Takanori Toyama, Kouichi Maeda, Kiyoshi Yamashita, Noriaki Kawano, Hiroshi Kawano, Tomonori Hidaka, Hideki Yamaguchi, Yoko Kubuki, Akira Kitanaka, Hitoshi Matsuoka, and Kazuya Shimoda
- Subjects
adult T-cell leukemia/lymphoma ,smoldering-type ,chronic-type ,iATL-PI ,Microbiology ,QR1-502 - Abstract
A retrospective chart survey of the clinical features of indolent adult T-cell leukemia/lymphoma (ATL) was conducted in the Miyazaki Prefecture, Japan. This study enrolled 24 smoldering-type ATLs, 10 favorable chronic-type ATLs, and 20 unfavorable chronic-type ATLs diagnosed between 2010 and 2018. Among them, 4, 3, and 10 progressed to acute-type ATLs during their clinical course. The median survival time (MST) in smoldering-type ATL and favorable chronic-type ATL was not reached, and their 4-year overall survival (OS) was 73% and 79%, respectively. Compared with this, the prognosis of unfavorable chronic-type ATL was poor. Its MST was 3.32 years, and the 4-year OS was 46% (p = 0.0095). In addition to the three features that determine the unfavorable characteristics of chronic-type ATL, namely, increased lactate dehydrogenase, increased blood urea nitrogen, and decreased albumin, the high-risk category by the indolent ATL-Prognostic Index, which was defined by an increment of soluble interleukin-2 receptor (sIL2-R) of >6000 U/mL, could explain the poor prognosis in indolent ATL patients. The level of sIL-2R might be an indicator of the initiation of therapy for indolent ATL.
- Published
- 2022
- Full Text
- View/download PDF
4. Relationship between CYP3A5 Polymorphism and Tacrolimus Blood Concentration Changes in Allogeneic Hematopoietic Stem Cell Transplant Recipients during Continuous Infusion
- Author
-
Naoki Yoshikawa, Hidemi Takeshima, Masaaki Sekine, Keiichi Akizuki, Tomonori Hidaka, Kazuya Shimoda, and Ryuji Ikeda
- Subjects
cytochrome P450 family 3 subfamily a member 5 ,tacrolimus ,gene polymorphism ,therapeutic drug monitoring ,hematopoietic stem cell transplantation ,Medicine ,Pharmacy and materia medica ,RS1-441 - Abstract
A polymorphism in the gene encoding the metabolic enzyme cytochrome P450 family 3 subfamily A member 5 (CYP3A5) is a particularly influential factor in the use of tacrolimus in Japanese patients. Those who are homozygotic for the *3 mutation lack CYP3A5 activity, which results in substantial individual differences in tacrolimus metabolism. The aim of this study was to analyze the relationship between individual differences in tacrolimus blood concentration changes and CYP3A5 polymorphisms in allogeneic hematopoietic stem cell transplantation recipients during the period of increasing blood concentration of the drug following treatment onset. This was a prospective observational cohort study, involving 20 patients administered tacrolimus by continuous infusion. The subjects were divided into the *1/*3 and *3/*3 groups based on CYP3A5 polymorphism analysis. The tacrolimus blood concentration/dose (C/D) ratio increased from day 1 and was largely stable on day 5, and a significant difference was observed between the *1/*3 and *3/*3 groups in the time course of the C/D ratio during this period (p < 0.05). This study reveals the effects of CYP3A5 polymorphism on continuous changes in tacrolimus blood concentration.
- Published
- 2021
- Full Text
- View/download PDF
5. Gene expression profiling of loss of TET2 and/or JAK2V617F mutant hematopoietic stem cells from mouse models of myeloproliferative neoplasms
- Author
-
Takuro Kameda, Kotaro Shide, Takumi Yamaji, Ayako Kamiunten, Masaaki Sekine, Tomonori Hidaka, Yoko Kubuki, Goro Sashida, Kazumasa Aoyama, Makoto Yoshimitsu, Hiroo Abe, Tadashi Miike, Hisayoshi Iwakiri, Yoshihiro Tahara, Shojiro Yamamoto, Satoru Hasuike, Kenji Nagata, Atsushi Iwama, Akira Kitanaka, and Kazuya Shimoda
- Subjects
Microarray profiling ,Loss of TET2 ,JAK2V617F ,Hematopoietic stem cell ,Myeloproliferative neoplasm ,Genetics ,QH426-470 - Abstract
Myeloproliferative neoplasms (MPNs) are clinically characterized by the chronic overproduction of differentiated peripheral blood cells and the gradual expansion of malignant intramedullary/extramedullary hematopoiesis. In MPNs mutations in JAK2 MPL or CALR are detected mutually exclusive in more than 90% of cases [1,2]. Mutations in them lead to the abnormal activation of JAK/STAT signaling and the autonomous growth of differentiated cells therefore they are considered as “driver” gene mutations. In addition to the above driver gene mutations mutations in epigenetic regulators such as TET2 DNMT3A ASXL1 EZH2 or IDH1/2 are detected in about 5%–30% of cases respectively [3]. Mutations in TET2 DNMT3A EZH2 or IDH1/2 commonly confer the increased self-renewal capacity on normal hematopoietic stem cells (HSCs) but they do not lead to the autonomous growth of differentiated cells and only exhibit subtle clinical phenotypes [4,6–8,5]. It was unclear how mutations in such epigenetic regulators influenced abnormal HSCs with driver gene mutations how they influenced the disease phenotype or whether a single driver gene mutation was sufficient for the initiation of human MPNs. Therefore we focused on JAK2V617F and loss of TET2—the former as a representative of driver gene mutations and the latter as a representative of mutations in epigenetic regulators—and examined the influence of single or double mutations on HSCs (Lineage−Sca-1+c-Kit+ cells (LSKs)) by functional analyses and microarray whole-genome expression analyses [9]. Gene expression profiling showed that the HSC fingerprint genes [10] was statistically equally enriched in TET2-knockdown-LSKs but negatively enriched in JAK2V617F–LSKs compared to that in wild-type-LSKs. Double-mutant-LSKs showed the same tendency as JAK2V617F–LSKs in terms of their HSC fingerprint genes but the expression of individual genes differed between the two groups. Among 245 HSC fingerprint genes 100 were more highly expressed in double-mutant-LSKs than in JAK2V617F–LSKs. These altered gene expressions might partly explain the mechanisms of initiation and progression of MPNs which was observed in the functional analyses [9]. Here we describe gene expression profiles deposited at the Gene Expression Omnibus (GEO) under the accession number GSE62302 including experimental methods and quality control analyses.
- Published
- 2015
- Full Text
- View/download PDF
6. Table S7 from Single-Cell Analysis of the Multicellular Ecosystem in Viral Carcinogenesis by HTLV-1
- Author
-
Keisuke Kataoka, Kazuya Shimoda, Seishi Ogawa, Yosuke Togashi, Atae Utsunomiya, Nobuaki Nakano, Akira Kitanaka, Tomonori Hidaka, Yoko Kubuki, Kotaro Shide, Masaaki Sekine, Ayako Kamiunten, Keiichi Akizuki, Yuki Tahira, Mariko Tabata, Sumito Shingaki, Marni B. McClure, Joji Nagasaki, Mitsuhiro Yuasa, Yasunori Kogure, Takuro Kameda, Yuki Saito, and Junji Koya
- Abstract
Candidate pathogenic mutations detected by WES or targeted-seq in ATL and AC samples
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Data from Single-Cell Analysis of the Multicellular Ecosystem in Viral Carcinogenesis by HTLV-1
- Author
-
Keisuke Kataoka, Kazuya Shimoda, Seishi Ogawa, Yosuke Togashi, Atae Utsunomiya, Nobuaki Nakano, Akira Kitanaka, Tomonori Hidaka, Yoko Kubuki, Kotaro Shide, Masaaki Sekine, Ayako Kamiunten, Keiichi Akizuki, Yuki Tahira, Mariko Tabata, Sumito Shingaki, Marni B. McClure, Joji Nagasaki, Mitsuhiro Yuasa, Yasunori Kogure, Takuro Kameda, Yuki Saito, and Junji Koya
- Abstract
Supplementary methods, Supplementary Figures, Supplementary Figure Legends, Supplementary Table Legends, Supplementary references, and Key resource table.
- Published
- 2023
- Full Text
- View/download PDF
8. Data from Single-Cell Analysis of the Multicellular Ecosystem in Viral Carcinogenesis by HTLV-1
- Author
-
Keisuke Kataoka, Kazuya Shimoda, Seishi Ogawa, Yosuke Togashi, Atae Utsunomiya, Nobuaki Nakano, Akira Kitanaka, Tomonori Hidaka, Yoko Kubuki, Kotaro Shide, Masaaki Sekine, Ayako Kamiunten, Keiichi Akizuki, Yuki Tahira, Mariko Tabata, Sumito Shingaki, Marni B. McClure, Joji Nagasaki, Mitsuhiro Yuasa, Yasunori Kogure, Takuro Kameda, Yuki Saito, and Junji Koya
- Abstract
Premalignant clonal expansion of human T-cell leukemia virus type-1 (HTLV-1)–infected cells occurs before viral carcinogenesis. Here we characterize premalignant cells and the multicellular ecosystem in HTLV-1 infection with and without adult T-cell leukemia/lymphoma (ATL) by genome sequencing and single-cell simultaneous transcriptome and T/B-cell receptor sequencing with surface protein analysis. We distinguish malignant phenotypes caused by HTLV-1 infection and leukemogenesis and dissect clonal evolution of malignant cells with different clinical behavior. Within HTLV-1–infected cells, a regulatory T-cell phenotype associates with premalignant clonal expansion. We also delineate differences between virus- and tumor-related changes in the nonmalignant hematopoietic pool, including tumor-specific myeloid propagation. In a newly generated conditional knockout mouse model recapitulating T-cell–restricted CD274 (encoding PD-L1) gene lesions found in ATL, we demonstrate that PD-L1 overexpressed by T cells is transferred to surrounding cells, leading to their PD-L1 upregulation. Our findings provide insights into clonal evolution and immune landscape of multistep virus carcinogenesis.Significance:Our multimodal single-cell analyses comprehensively dissect the cellular and molecular alterations of the peripheral blood in HTLV-1 infection, with and without progression to leukemia. This study not only sheds light on premalignant clonal expansion in viral carcinogenesis, but also helps to devise novel diagnostic and therapeutic strategies for HTLV-1–related disorders.This article is highlighted in the In This Issue feature, p. 403
- Published
- 2023
- Full Text
- View/download PDF
9. Whole-genome landscape of adult T-cell leukemia/lymphoma
- Author
-
Yoko Kubuki, Kengo Takeuchi, Seishi Ogawa, Yuki Tahira, Akifumi Takaori-Kondo, Kota Yoshifuji, Makoto Yoshimitsu, Kenichi Chiba, Masaaki Sekine, Ai Okada, Ana Acuna-Villaorduna, Yuichi Shiraishi, Yasushi Miyazaki, Tatsuhiro Shibata, Jun-ichirou Yasunaga, Tomonori Hidaka, Michihiro Hidaka, Mariko Tabata, Kisato Nosaka, Masao Matsuoka, Yasunori Kogure, Takuro Kameda, R. Alejandro Sica, Yuta Ito, B. Hilda Ye, Yoshitaka Imaizumi, Ayako Kamiunten, Sumito Shingaki, Keiichi Akizuki, Yuki Saito, Juan Carlos Ramos, Kazuya Shimoda, Junji Koya, Murali Janakiram, Marni B McClure, Urvi A Shah, Yasuhito Nannya, Kenji Ishitsuka, Mizuki Watanabe, Nobuaki Nakano, Satoru Miyano, Nobuyuki Kakiuchi, Atae Utsunomiya, Keisuke Kataoka, Kotaro Shide, and Akira Kitanaka
- Subjects
DNA Copy Number Variations ,Immunology ,Somatic hypermutation ,Biology ,Biochemistry ,Genome ,Adult T-cell leukemia/lymphoma ,Mice ,Exome Sequencing ,Biomarkers, Tumor ,medicine ,Animals ,Humans ,Leukemia-Lymphoma, Adult T-Cell ,IL-2 receptor ,Gene ,Ataxin-1 ,Genetics ,Genome, Human ,Germinal center ,FOXP3 ,Cell Biology ,Hematology ,Prognosis ,medicine.disease ,Proto-Oncogene Proteins c-rel ,Gene Expression Regulation, Neoplastic ,Mice, Inbred C57BL ,Repressor Proteins ,Survival Rate ,Leukemia ,Mutation ,Female - Abstract
Adult T-cell leukemia/lymphoma (ATL) is an aggressive neoplasm immunophenotypically resembling regulatory T cells, associated with human T-cell leukemia virus type-1. Here, we performed whole-genome sequencing (WGS) of 150 ATL cases to reveal the overarching landscape of genetic alterations in ATL. We discovered frequent (33%) loss-of-function alterations preferentially targeting the CIC long isoform, which were overlooked by previous exome-centric studies of various cancer types. Long but not short isoform–specific inactivation of Cic selectively increased CD4+CD25+Foxp3+ T cells in vivo. We also found recurrent (13%) 3′-truncations of REL, which induce transcriptional upregulation and generate gain-of-function proteins. More importantly, REL truncations are also common in diffuse large B-cell lymphoma, especially in germinal center B-cell–like subtype (12%). In the non-coding genome, we identified recurrent mutations in regulatory elements, particularly splice sites, of several driver genes. In addition, we characterized the different mutational processes operative in clustered hypermutation sites within and outside immunoglobulin/T-cell receptor genes and identified the mutational enrichment at the binding sites of host and viral transcription factors, suggesting their activities in ATL. By combining the analyses for coding and noncoding mutations, structural variations, and copy number alterations, we discovered 56 recurrently altered driver genes, including 11 novel ones. Finally, ATL cases were classified into 2 molecular groups with distinct clinical and genetic characteristics based on the driver alteration profile. Our findings not only help to improve diagnostic and therapeutic strategies in ATL, but also provide insights into T-cell biology and have implications for genome-wide cancer driver discovery.
- Published
- 2022
- Full Text
- View/download PDF
10. Higher average chemotherapy dose intensity improves prognosis in patients with aggressive adult T‐cell leukemia/lymphoma
- Author
-
Kiyoshi Yamashita, Junzo Ishizaki, Keiichi Akizuki, Hitoshi Matsuoka, Kotaro Shide, Akira Kitanaka, Kouichi Maeda, Seiichi Sato, Kazuya Shimoda, Takanori Toyama, Tomonori Hidaka, Hiroshi Kawano, Takuro Kameda, Haruko Shimoda, Ayako Kamiunten, Masaaki Sekine, Masanori Takeuchi, Yoko Kubuki, Toshimasa Kukita, Noriaki Kawano, and Yuki Tahira
- Subjects
medicine.medical_specialty ,medicine.medical_treatment ,Clinical Decision-Making ,Gastroenterology ,Adult T-cell leukemia/lymphoma ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Leukemia-Lymphoma, Adult T-Cell ,Medicine ,Neoplasm Staging ,Retrospective Studies ,Chemotherapy ,business.industry ,Medical record ,Organ dysfunction ,Disease Management ,Hematology ,General Medicine ,Prognosis ,medicine.disease ,Dose intensity ,Lymphoma ,Leukemia ,Treatment Outcome ,030220 oncology & carcinogenesis ,Disease Progression ,Neoplasm Grading ,medicine.symptom ,Outcomes research ,business ,030215 immunology - Abstract
OBJECTIVE AND METHOD Adult T-cell leukemia/lymphoma (ATL) is an aggressive peripheral T-cell lymphoma with poor prognosis. We retrospectively reviewed the medical records of 312 patients with aggressive ATL and analyzed the effect of chemotherapy dose intensity on prognosis in clinical practice. RESULT As first-line therapy, 62 patients underwent best supportive care (BSC) or single-agent chemotherapy, and 235 underwent intensive chemotherapy. The median survival time (MST) was 0.58 years in the 312 total patients, and 0.13 years and 0.75 years in the BSC/single-agent chemotherapy group and intensive chemotherapy group, respectively. The median average relative dose intensity (ARDI) of patients who received intensive chemotherapy was 60%. We divided patients into 3 groups according to ARDI. Those in the top tertile of ARDI (ARDI ≥ 75%, n = 82) had better overall survival compared with those in the intermediate tertile (45% ≤ ARDI
- Published
- 2020
- Full Text
- View/download PDF
11. The effect of CARD11 mutation and HBZ expression accelerated lymphoproliferative diseases and formed the pathological basis for adult T-cell leukemia/lymphoma
- Author
-
Kazuya Shimoda, Takuro Kameda, Kotaro Shide, Ayako Kamiunten, Yasunori Kogure, Daisuke Morishita, Junji Koya, Yuki Tahira, Keiichi Akizuki, Takako Yokomizo-Nakano, Sho Kubota, Kosuke Marutsuka, Masaaki Sekine, Tomonori Hidaka, Yoko Kubuki, Akinori Yoda, Takayuki Ohshima, Midori Sugiyama, Goro Sashida, Keisuke Kataoka, and Seishi Ogawa
- Subjects
hemic and lymphatic diseases - Abstract
Adult T-cell leukemia/lymphoma (ATL) is caused by human T-cell leukemia virus type 1 (HTLV-1). In addition to HTLV-1 bZIP factor (HBZ), a leukemogenic antisense transcript of HTLV-1, abnormalities of genes involved in TCR-NF-κB signaling such as CARD11 have been detected in about 90% of patients. Utilizing mouse models with T cell-specific CARD11(E626K) expression and/or CD4+ T cell-specific HBZ expression, namely CARD11(E626K)CD4-Cre mice, HBZ transgenic (Tg) mice, and CARD11(E626K)CD4-Cre;HBZ Tg compound mice, we clarified the effect of these genes on pathogenesis. Both CARD11(E626K)CD4-Cre and HBZ Tg mice developed lymphoproliferative disease, and the compound mice showed accelerated disease and increased CD4+ T cells in vivo. CARD11(E626K) and HBZ cooperatively activated the NF-κB non-canonical pathway, IRF4 targets, and cell proliferation. Most of the KEGG and HALLMARK gene sets, which are enriched in acute-type ATL, were also enriched in the compound mice, indicating that these genes cooperatively form the pathological basis for ATL development.
- Published
- 2022
- Full Text
- View/download PDF
12. Real-World Data on Clinical Features, Outcomes, and Prognostic Factors in Multiple Myeloma from Miyazaki Prefecture, Japan
- Author
-
Keiichi Akizuki, Seiichi Sato, Yuki Tahira, Kotaro Shide, Hitoshi Matsuoka, Yoko Kubuki, Noriaki Kawano, Ayako Kamiunten, Junzo Ishizaki, Masaaki Sekine, Kouichi Maeda, Kazuya Shimoda, Tomonori Hidaka, Hiroshi Kawano, Takuro Kameda, Haruko Shimoda, Kiyoshi Yamashita, Masanori Takeuchi, and Takanori Toyama
- Subjects
Oncology ,medicine.medical_specialty ,Multivariate analysis ,lcsh:Medicine ,Disease ,Article ,03 medical and health sciences ,0302 clinical medicine ,Autologous stem-cell transplantation ,Internal medicine ,medicine ,Multiple myeloma ,Lenalidomide ,Bortezomib ,business.industry ,lcsh:R ,prognostic factors ,General Medicine ,medicine.disease ,real-world outcomes ,international staging system ,Thalidomide ,Clinical trial ,multiple myeloma ,030220 oncology & carcinogenesis ,business ,030215 immunology ,medicine.drug - Abstract
The prognosis of multiple myeloma (MM) has improved with the introduction of novel agents. These data are largely derived from clinical trials and might not reflect real-world patient outcomes accurately. We surveyed real-world data from 284 patients newly diagnosed with MM between 2010 and 2018 in Miyazaki Prefecture. The median follow-up period was 32.8 months. The median age at diagnosis was 71 years, with 68% of patients aged >, 65 years. The International Staging System (ISS) stage at diagnosis was I in 18.4% of patients, II in 34.1%, and III in 47.5%. Bortezomib-containing regimens were preferred as initial treatment, they were used in 147 patients (51.8%). In total, 80% of patients were treated with one or more novel agents (thalidomide, lenalidomide, or bortezomib). Among 228 patients who were treated with novel agents as an initial treatment, the overall response rate (partial response (PR) or better) to initial treatment was 78.4%, and the median time to next treatment (TTNT) was 11.6 months. In the multivariate analysis, PR or better responses to initial treatment were independently favorable prognostic factors for TTNT. The median survival time after initial therapy for patients with novel agents was 56.4 months and 3-year overall survival (OS) was 70.4%. In multivariate analysis, ISS stage I/II disease and PR or better response to initial treatment, and autologous stem cell transplantation (ASCT) were identified as independent prognostic factors for overall survival (OS).
- Published
- 2021
13. Single-Cell Analysis of the Multicellular Ecosystem in Viral Carcinogenesis by HTLV-1
- Author
-
Yoko Kubuki, Yasunori Kogure, Ayako Kamiunten, Kazuya Shimoda, Akira Kitanaka, Sumito Shingaki, Tomonori Hidaka, Keiichi Akizuki, Marni B McClure, Takuro Kameda, Yuki Saito, Junji Koya, Masaaki Sekine, Mariko Tabata, Yosuke Togashi, Nobuaki Nakano, Atae Utsunomiya, Kotaro Shide, Keisuke Kataoka, Joji Nagasaki, Yuki Tahira, Mitsuhiro Yuasa, and Seishi Ogawa
- Subjects
Carcinogenesis ,animal diseases ,viruses ,chemical and pharmacologic phenomena ,Biology ,medicine.disease_cause ,Somatic evolution in cancer ,Transcriptome ,Mice ,Immune system ,hemic and lymphatic diseases ,Conditional gene knockout ,medicine ,Animals ,Leukemia-Lymphoma, Adult T-Cell ,Research Articles ,Ecosystem ,Viral Carcinogenesis ,Human T-lymphotropic virus 1 ,General Medicine ,biochemical phenomena, metabolism, and nutrition ,medicine.disease ,Haematopoiesis ,Leukemia ,Cancer research ,bacteria ,Single-Cell Analysis - Abstract
High-dimensional single-cell landscape of immune alterations during HTLV-1 infection and leukemogenesis identifies hallmarks of premalignant and malignant T-cell states and the accompanying shift of systemic immune state toward myeloid and immunosuppressive., Premalignant clonal expansion of human T-cell leukemia virus type-1 (HTLV-1)–infected cells occurs before viral carcinogenesis. Here we characterize premalignant cells and the multicellular ecosystem in HTLV-1 infection with and without adult T-cell leukemia/lymphoma (ATL) by genome sequencing and single-cell simultaneous transcriptome and T/B-cell receptor sequencing with surface protein analysis. We distinguish malignant phenotypes caused by HTLV-1 infection and leukemogenesis and dissect clonal evolution of malignant cells with different clinical behavior. Within HTLV-1–infected cells, a regulatory T-cell phenotype associates with premalignant clonal expansion. We also delineate differences between virus- and tumor-related changes in the nonmalignant hematopoietic pool, including tumor-specific myeloid propagation. In a newly generated conditional knockout mouse model recapitulating T-cell–restricted CD274 (encoding PD-L1) gene lesions found in ATL, we demonstrate that PD-L1 overexpressed by T cells is transferred to surrounding cells, leading to their PD-L1 upregulation. Our findings provide insights into clonal evolution and immune landscape of multistep virus carcinogenesis. Significance: Our multimodal single-cell analyses comprehensively dissect the cellular and molecular alterations of the peripheral blood in HTLV-1 infection, with and without progression to leukemia. This study not only sheds light on premalignant clonal expansion in viral carcinogenesis, but also helps to devise novel diagnostic and therapeutic strategies for HTLV-1–related disorders.
- Published
- 2021
14. Calreticulin haploinsufficiency augments stem cell activity and is required for onset of myeloproliferative neoplasms
- Author
-
Yoko Kubuki, Tomonori Hidaka, Takako Yokomizo-Nakano, Takuro Kameda, Yoshinori Ozono, Masaya Ono, Goro Sashida, Yuki Tahira, Sho Kubota, Ayako Kamiunten, Hisayoshi Iwakiri, Satoru Hasuike, Masaaki Sekine, Kazuya Shimoda, Kotaro Shide, Kenichi Nakamura, Kenji Nagata, Kazuhiko Ikeda, and Keiichi Akizuki
- Subjects
0301 basic medicine ,Genotype ,Immunology ,Mice, Transgenic ,Biochemistry ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Bone Marrow ,medicine ,Animals ,Erythropoiesis ,Cell Self Renewal ,Sequence Deletion ,Myeloproliferative Disorders ,biology ,Essential thrombocythemia ,Stem Cells ,Cell Biology ,Hematology ,Hematopoietic Stem Cells ,medicine.disease ,Extramedullary hematopoiesis ,Transplantation ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,Hematopoiesis, Extramedullary ,Neoplastic Stem Cells ,Cancer research ,biology.protein ,Bone marrow ,Calreticulin ,Transcriptome ,Haploinsufficiency ,030215 immunology - Abstract
Mutations in JAK2, myeloproliferative leukemia virus (MPL), or calreticulin (CALR) occur in hematopoietic stem cells (HSCs) and are detected in more than 80% of patients with myeloproliferative neoplasms (MPNs). They are thought to play a driver role in MPN pathogenesis via autosomal activation of the JAK-STAT signaling cascade. Mutant CALR binds to MPL, activates downstream MPL signaling cascades, and induces essential thrombocythemia in mice. However, embryonic lethality of Calr-deficient mice precludes determination of a role for CALR in hematopoiesis. To clarify the role of CALR in normal hematopoiesis and MPN pathogenesis, we generated hematopoietic cell-specific Calr-deficient mice. CALR deficiency had little effect on the leukocyte count, hemoglobin levels, or platelet count in peripheral blood. However, Calr-deficient mice showed some hematopoietic properties of MPN, including decreased erythropoiesis and increased myeloid progenitor cells in the bone marrow and extramedullary hematopoiesis in the spleen. Transplantation experiments revealed that Calr haploinsufficiency promoted the self-renewal capacity of HSCs. We generated CALRdel52 mutant transgenic mice with Calr haploinsufficiency as a model that mimics human MPN patients and found that Calr haploinsufficiency restored the self-renewal capacity of HSCs damaged by CALR mutations. Only recipient mice transplanted with Lineage-Sca1+c-kit+ cells harboring both CALR mutation and Calr haploinsufficiency developed MPN in competitive conditions, showing that CALR haploinsufficiency was necessary for the onset of CALR-mutated MPNs.
- Published
- 2020
- Full Text
- View/download PDF
15. Neoplastic fibrocytes play an essential role in bone marrow fibrosis in Jak2V617F-induced primary myelofibrosis mice
- Author
-
Yoshinori Ozono, Kazuya Shimoda, Mitsue Sueta, Akira Sawaguchi, Kenichi Nakamura, Shojiro Yamamoto, Kenji Nagata, Yoko Kubuki, Ayako Kamiunten, Keiichi Akizuki, Hisayoshi Iwakiri, Tomonori Hidaka, Satoru Hasuike, Takuro Kameda, Yuki Tahira, Masaaki Sekine, and Kotaro Shide
- Subjects
Cancer Research ,Bone marrow transplantation ,Mice, Transgenic ,Article ,Transforming Growth Factor beta1 ,Mice ,Fibrosis ,Myeloproliferation ,medicine ,Animals ,Myelofibrosis ,Cancer models ,Myeloproliferative neoplasm ,Cell Proliferation ,business.industry ,Monocyte ,Neoplastic Monocyte ,Cell Differentiation ,Hematology ,Fibroblasts ,Janus Kinase 2 ,medicine.disease ,Haematopoiesis ,medicine.anatomical_structure ,Oncology ,Primary Myelofibrosis ,Mutation ,Splenomegaly ,Cancer research ,Bone marrow ,business ,Megakaryocytes - Abstract
Primary myelofibrosis (PMF) is a myeloproliferative neoplasm (MPN) characterized by clonal myeloproliferation, progressive bone marrow (BM) fibrosis, splenomegaly, and anemia. BM fibrosis was previously thought to be a reactive phenomenon induced by mesenchymal stromal cells that are stimulated by the overproduction of cytokines such as transforming growth factor (TGF)-β1. However, the involvement of neoplastic fibrocytes in BM fibrosis was recently reported. In this study, we showed that the vast majority of collagen- and fibronectin-producing cells in the BM and spleens of Jak2V617F-induced myelofibrosis (MF) mice were fibrocytes derived from neoplastic hematopoietic cells. Neoplastic monocyte depletion eliminated collagen- and fibronectin-producing fibrocytes in BM and spleen, and ameliorated most characteristic MF features in Jak2V617F transgenic mice, including BM fibrosis, anemia, and splenomegaly, while had little effect on the elevated numbers of megakaryocytes and stem cells in BM, and leukothrombocytosis in peripheral blood. TGF-β1, which was produced by hematopoietic cells including fibrocytes, promoted the differentiation of neoplastic monocytes to fibrocytes, and elevated plasma TGF-β1 levels were normalized by monocyte depletion. Collectively, our data suggest that neoplastic fibrocytes are the major contributor to BM fibrosis in PMF, and TGF-β1 is required for their differentiation.
- Published
- 2020
16. Outcome of allogeneic hematopoietic cell transplantation in patients with adult T-cell leukemia
- Author
-
Keiichi Akizuki, Koji Kato, Yuki Tahira, Haruko Shimoda, Yoko Kubuki, Kazuya Shimoda, Tomonori Hidaka, Masaaki Sekine, Takuro Kameda, Kotaro Shide, and Ayako Kamiunten
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,T-cell leukemia ,chemotherapy ,Adult T-cell leukemia/lymphoma ,03 medical and health sciences ,0302 clinical medicine ,immune system diseases ,Original Research Articles ,hemic and lymphatic diseases ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,adult T‐cell leukemia/lymphoma ,Humans ,Leukemia-Lymphoma, Adult T-Cell ,Cumulative incidence ,Original Research Article ,Aged ,Retrospective Studies ,Chemotherapy ,business.industry ,Hematopoietic Stem Cell Transplantation ,Hematology ,General Medicine ,allogeneic hematopoietic stem‐cell transplantation ,Middle Aged ,medicine.disease ,Lymphoma ,Survival Rate ,Transplantation ,Leukemia ,Treatment Outcome ,surgical procedures, operative ,030220 oncology & carcinogenesis ,Cord blood ,Female ,business ,030215 immunology - Abstract
Adult T‐cell leukemia/lymphoma (ATL) is an aggressive peripheral T‐cell neoplasm, and the outcome of patients with ATL after chemotherapy is poor. Allogeneic hematopoietic stem‐cell transplantation (allo‐HSCT) is a curative treatment modality for ATL, and four factors, namely, age > 50 years, male recipient, lack of complete remission at transplantation, and transplantation of cord blood, were previously shown to be associated with poor survival. We retrospectively analyzed the outcome of 21 patients with ATL who had undergone allo‐HSCT at our hospital during a 3‐year period. Of 21 patients, all had at least one of the above risk factors, and 18 had two or more. With a median follow‐up of 19.7 months for living patients, the 1‐ and 2‐year overall survival (OS) rates after transplantation were 34% and 27%, respectively. All relapse/progression events occurred within 1 year after allo‐HSCT, and the cumulative incidence of relapse/progression at 1 year after allo‐HSCT was 46.9%. The 100‐day and 1‐year nonrelapse mortality (NRM) rates were 19% and 42%, respectively. No significant difference in OS was observed between myeloablative and reduced‐intensity conditioning regimens. The 3‐year OS (27%) of ATL patients who received allo‐HSCT and who had at least one adverse factor was somewhat poorer than the 3‐year OS of 33% identified in a nationwide study of allo‐HSCT in ATL patients in Japan. The high relapse/progression and NRM rates are major problems to be solved to achieve better outcome.
- Published
- 2018
- Full Text
- View/download PDF
17. TET2 mutation in diffuse large B-cell lymphoma
- Author
-
Ayako Kamiunten, Tomonori Hidaka, Kenichi Nakamura, Yoshihiro Tahara, Takuro Kameda, Takumi Yamaji, Hiroo Abe, Yoko Kubuki, Tadashi Miike, Kotaro Shide, Keiichi Akizuki, Mitsue Sueta, Yuuki Tahira, Kazuya Shimoda, Shojiro Yamamoto, Masaaki Sekine, Akira Kitanaka, Kenji Nagata, Hisayoshi Iwakiri, Satoru Hasuike, and Haruko Shimoda
- Subjects
0301 basic medicine ,Myeloid ,business.industry ,General Medicine ,Disease ,medicine.disease ,Lymphoma ,Frameshift mutation ,Pathogenesis ,03 medical and health sciences ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,hemic and lymphatic diseases ,Mutation (genetic algorithm) ,medicine ,Cancer research ,business ,Diffuse large B-cell lymphoma - Abstract
Ten-eleven translocation-2 (TET2) mutation is frequently observed in myeloid malignancies, and loss-of-function of TET2 is essential for the initiation of malignant hematopoiesis. TET2 mutation presents across disease entities and was reported in lymphoid malignancies. We investigated TET2 mutations in 27 diffuse large B-cell lymphoma (DLBCL) patients and found a frameshift mutation in 1 case (3.7%). TET2 mutation occurred in some populations of DLBCL patients and was likely involved in the pathogenesis of their malignancies.
- Published
- 2017
- Full Text
- View/download PDF
18. CARD11 Mutation Induces Oligoclonal Expansion of T-Cells, and Accelerates ATL Development in Combination with HBZ
- Author
-
Midori Sugiyama, Tahira Yuki, Yoko Kubuki, Daisuke Morishita, Kazuya Shimoda, Masaaki Sekine, Kotaro Shide, Tomonori Hidaka, Ayako Kamiunten, Takuro Kameda, and Keiichi Akizuki
- Subjects
medicine.medical_specialty ,Hematology ,CD3 ,Immunology ,Alveolar septum ,Spleen ,Cell Biology ,Gene mutation ,Biology ,medicine.disease ,Biochemistry ,Molecular biology ,Lymphoma ,Leukemia ,medicine.anatomical_structure ,Internal medicine ,medicine ,biology.protein ,Lymph node - Abstract
Adult T-cell leukemia/lymphoma (ATL) is an aggressive peripheral T-cell lymphoma that develops in about 5% of human T-cell leukemia/lymphoma virus 1 (HTLV-1) carriers. In addition to viral oncogenes, namely tax and HTLV-1 bZIP factor (HBZ), gene mutations, highly enriched for T-cell receptor (TCR)-NF-kB signaling, should be involved in the development of ATL. Among gene mutations, mutation in CARD11, a cytoplasmic scaffolding protein required for TCR-induced NF-kB activation, was detected in 24% of ATL patients. Here we generated a mouse model for conditional expression of a human ATL-derived CARD11(E626K) gain-of-function mutant, and demonstrated CARD11 activation induced oligoclonal expansion of T-cell and infiltration to many organs. We also showed that expression of HBZ accelerated mutant CARD11-induced lymphoproliferative diseases. We introduced a human Card11(E626K) into the mouse genome at the ROSA26 locus. After crossing with CD4-Cre Tg, CARD11(E626K)CD4-Cre mice was obtained. In CD4+ cells from CARD11(E626K)CD4-Cre, the amount of cleaved BCL-10 and NF-kBp65 increased compared with those in WT CD4+cells, confirming the activation of NF-kB. About half of CARD11(E626K)CD4-Cre mice died on or after 6 months after birth. At 6 months, leukocytosis was observed in CARD11(E626K)CD4-Cre, and accordingly the number of CD4+ cells cells was about 1.43 times greater than those in WT mice. The most affected organ in CARD11(E626K)CD4-Cre mice was lung. Alveolar septum was thickened by infiltrated cells at 6 months, and worsened subsequently. CD3+ T-cell accumulated around capillary blood vessels, and had high proliferation indices (>50%), as assayed by Ki-67 staining. CARD11(E626K)CD4-Cre mice developed lymphadenopathy (4/8 mice (50%) at 6M and 4/6 mice (66.7%) at 12M). Normal lymph node (LN) architecture was barely preserved, and medullary sinus was expanded with CD3+ T-cell. Some of them were positive for FoxP3, and had moderate proliferation indices (25%-50%). Among CD4+ T-cell, the proportion of naive T-cell (Tn) decreased, and that of effector/memory T-cell (Tem) and regulatory T-cell (Treg) increased compared to WT LNs. The proportion of Treg in CD4+ T-cell from LN of 6M- and 12M-old CARD11(E626K)CD4-Cre mice was 25% and 40%, respectively, which values were much larger than the normal range as 10-15%. We next examined the clonality of CD4+ cells in spleen and swollen LNs from CARD11(E626K)CD4-Cre mice. The clonality of the TCR repertoires of 20 individual Vb gene famines from Vb1-20 was assessed by a PCR. The clonality assay using the TCR repertoires exhibited an oligoclonal pattern in 4 of 5 splenic CD4+ cells, and 1 of 2 LN CD4+ cells from CARD11(E626K)CD4-Cre mice. To assess the effect of HBZ constant expression on CARD11(E626K)CD4-Cre mice, we generated HBZ Tg, in which HBZ cDNA was expressed under the CD4 promoter. Similar to the previous report (by Satou et al.), our HBZ Tg showed increased number of Tem, destroyed architecture of lung such as thickened alveolar septum by infiltrated cells and decreased alveolar space by edema, and the lymphadenopathy after 12M (66.7%). We then crossed CARD11(E626K)CD4-Creand HBZ Tg, and obtained the compound mice. The compound mice caused more aggressive lymphoproliferative diseases compared with CARD11(E626K)CD4-Cre mice. Most of compound mice died within 8M. At 6M, architecture of lung, kidney, spleen, and LN was destroyed. In lung, alveolar space of lung was scarcely observed caused of T-cell invasion. Alveolar septum was thickened with infiltrated cells, and CD3+ cells accumulated around capillary blood vessel. Some of them were positive for FoxP3, and indicated moderate proliferation indices (25-50%). T-cell invasion was also observed in kidney. Lymphadenopathy was detected in 6 of 9 (66.7%) with completely destroyed architecture, increment of the proportion of Fox3+ cells, and moderate proliferation indices (25-50%). The clonality assay using the TCR repertoires exhibited an oligoclonal pattern in 4 of 4 splenic CD4+ cells, and 2 of 2 LNs CD4+ cells from compound mice. These results suggest that CARD11 mutant-induced NFkB activation and constant HBZ expression may have similar effects, such as T-cell infiltration into organs and LN adenopathy, and that the simultaneous occurrence of both may have additive effects. Disclosures Sugiyama: Chordia Therapeutics, Japan.: Current Employment. Morishita:Chordia Therapeutics Inc.: Current Employment, Current equity holder in private company. Shimoda:Perseus Proteomics: Research Funding; PharmaEssentia Japan: Research Funding; AbbVie Inc.: Research Funding; Astellas Pharma: Research Funding; Merck & Co.: Research Funding; CHUGAI PHARMACEUTICAL CO., LTD.: Research Funding; Kyowa Hakko Kirin Co., Ltd.: Research Funding; Pfizer Inc.: Research Funding; Otsuka Pharmaceutical: Research Funding; Asahi Kasei Medical: Research Funding; Japanese Society of Hematology: Research Funding; The Shinnihon Foundation of Advanced Medical Treatment Research: Research Funding; Celgene: Honoraria; Shire plc: Honoraria; Novartis: Honoraria, Research Funding; Takeda Pharmaceutical Company: Honoraria; Bristol-Myers Squibb: Honoraria.
- Published
- 2020
- Full Text
- View/download PDF
19. TP53 and PTEN Mutations Were Shared in Concurrent Germ Cell Tumor and Acute Megakaryoblastic Leukemia
- Author
-
Yoko Kubuki, Takumi Kiwaki, Yuichiro Sato, Keiichi Akizuki, Masaaki Sekine, Hiroyuki Tanaka, Junji Koya, Kazuya Shimoda, Kotaro Shide, Keisuke Kataoka, Hiroaki Kataoka, Yasunori Kogure, Tomonori Hidaka, Yuki Tahira, Takuro Kameda, and Ayako Kamiunten
- Subjects
Adult ,Male ,0301 basic medicine ,Cancer Research ,PTEN ,Mediastinal germ cell tumor ,Somatic cell ,Biopsy ,Clone (cell biology) ,Biology ,lcsh:RC254-282 ,Clonal Evolution ,03 medical and health sciences ,Acute megakaryoblastic leukemia ,0302 clinical medicine ,Japan ,Bone Marrow ,Leukemia, Megakaryoblastic, Acute ,Surgical oncology ,Exome Sequencing ,Genetics ,medicine ,Germ cell tumor ,Humans ,TP53 ,In Situ Hybridization, Fluorescence ,Acute myeloid leukemia ,PTEN Phosphohydrolase ,Myeloid leukemia ,Neoplasms, Germ Cell and Embryonal ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cytogenetic Analysis ,Cancer research ,biology.protein ,Radiography, Thoracic ,Tumor Suppressor Protein p53 ,Tomography, X-Ray Computed ,Germ cell ,Research Article - Abstract
Background The occurrence of a mediastinal germ cell tumor (GCT) and hematological malignancy in the same patient is very rare. Due to its rarity, there have been only two reports of the concurrent cases undergoing detailed genetic analysis with whole-exome sequencing (WES), and the possible clonal relationship between the both tumors remained not fully elucidated. Methods We performed whole-exome sequencing analysis of mediastinal GCT and acute myeloid leukemia (AML) samples obtained from one young Japanese male adult patient with concurrent both tumors, and investigated the possible clonal relationship between them. Results Sixteen somatic mutations were detected in the mediastinal GCT sample and 18 somatic mutations in the AML sample. Mutations in nine genes, including TP53 and PTEN both known as tumor suppressor genes, were shared in both tumors. Conclusions All in our case and in the previous two cases with concurrent mediastinal GCT and AML undergoing with whole-exome sequencing analysis, TP53 and PTEN mutations were commonly shared in both tumors. These data not only suggest that these tumors share a common founding clone, but also indicate that associated mediastinal GCT and AML harboring TP53 and PTEN mutations represent a unique biological entity.
- Published
- 2019
- Full Text
- View/download PDF
20. Relationship between CYP3A5 Polymorphism and Tacrolimus Blood Concentration Changes in Allogeneic Hematopoietic Stem Cell Transplant Recipients during Continuous Infusion
- Author
-
Kazuya Shimoda, Masaaki Sekine, Hidemi Takeshima, Ryuji Ikeda, Naoki Yoshikawa, Keiichi Akizuki, and Tomonori Hidaka
- Subjects
Drug ,medicine.medical_specialty ,gene polymorphism ,therapeutic drug monitoring ,media_common.quotation_subject ,medicine.medical_treatment ,lcsh:Medicine ,lcsh:RS1-441 ,Pharmaceutical Science ,Hematopoietic stem cell transplantation ,030226 pharmacology & pharmacy ,Article ,lcsh:Pharmacy and materia medica ,03 medical and health sciences ,0302 clinical medicine ,Polymorphism (computer science) ,Internal medicine ,Drug Discovery ,Medicine ,tacrolimus ,CYP3A5 ,media_common ,medicine.diagnostic_test ,business.industry ,lcsh:R ,Tacrolimus ,surgical procedures, operative ,Endocrinology ,cytochrome P450 family 3 subfamily a member 5 ,Therapeutic drug monitoring ,030220 oncology & carcinogenesis ,hematopoietic stem cell transplantation ,Molecular Medicine ,Gene polymorphism ,business ,Cohort study - Abstract
A polymorphism in the gene encoding the metabolic enzyme cytochrome P450 family 3 subfamily A member 5 (CYP3A5) is a particularly influential factor in the use of tacrolimus in Japanese patients. Those who are homozygotic for the *3 mutation lack CYP3A5 activity, which results in substantial individual differences in tacrolimus metabolism. The aim of this study was to analyze the relationship between individual differences in tacrolimus blood concentration changes and CYP3A5 polymorphisms in allogeneic hematopoietic stem cell transplantation recipients during the period of increasing blood concentration of the drug following treatment onset. This was a prospective observational cohort study, involving 20 patients administered tacrolimus by continuous infusion. The subjects were divided into the *1/*3 and *3/*3 groups based on CYP3A5 polymorphism analysis. The tacrolimus blood concentration/dose (C/D) ratio increased from day 1 and was largely stable on day 5, and a significant difference was observed between the *1/*3 and *3/*3 groups in the time course of the C/D ratio during this period (p <, 0.05). This study reveals the effects of CYP3A5 polymorphism on continuous changes in tacrolimus blood concentration.
- Published
- 2021
- Full Text
- View/download PDF
21. Calreticulin mutant mice develop essential thrombocythemia that is ameliorated by the JAK inhibitor ruxolitinib
- Author
-
Hisayoshi Iwakiri, Satoru Hasuike, Haruko Shimoda, Kenji Nagata, Shojiro Yamamoto, Kenichi Nakamura, Kazuya Shimoda, N Inada, Akira Kitanaka, Hiroo Abe, Arata Honda, Akira Sawaguchi, Mitsue Sueta, Yoko Kubuki, Takumi Yamaji, Yoshihiro Tahara, Tadashi Miike, Keiichi Akizuki, Ayako Kamiunten, Kotaro Shide, Masaaki Sekine, Tomonori Hidaka, and Takuro Kameda
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Ruxolitinib ,Transgene ,Mutant ,Mice, Transgenic ,03 medical and health sciences ,Mice ,0302 clinical medicine ,hemic and lymphatic diseases ,Internal medicine ,Nitriles ,medicine ,STAT5 Transcription Factor ,Animals ,Humans ,Cell Self Renewal ,Protein Kinase Inhibitors ,Thrombopoietin ,Janus Kinases ,Hematology ,Myeloproliferative Disorders ,biology ,Essential thrombocythemia ,medicine.disease ,Hematopoietic Stem Cells ,Leukemia ,030104 developmental biology ,HEK293 Cells ,Pyrimidines ,Oncology ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Pyrazoles ,Original Article ,Calreticulin ,Receptors, Thrombopoietin ,medicine.drug ,Thrombocythemia, Essential - Abstract
Mutations of calreticulin (CALR) are detected in 25-30% of patients with essential thrombocythemia (ET) or primary myelofibrosis and cause frameshifts that result in proteins with a novel C-terminal. We demonstrate that CALR mutations activated signal transducer and activator of transcription 5 (STAT5) in 293T cells in the presence of thrombopoietin receptor (MPL). Human megakaryocytic CMK11-5 cells and erythroleukemic F-36P-MPL cells with knocked-in CALR mutations showed increased growth and acquisition of cytokine-independent growth, respectively, accompanied by STAT5 phosphorylation. Transgenic mice expressing a human CALR mutation with a 52 bp deletion (CALRdel52-transgenic mice (TG)) developed ET, with an increase in platelet count, but not hemoglobin level or white blood cell count, in association with an increase in bone marrow (BM) mature megakaryocytes. CALRdel52 BM cells did not drive away wild-type (WT) BM cells in in vivo competitive serial transplantation assays, suggesting that the self-renewal capacity of CALRdel52 hematopoietic stem cells (HSCs) was comparable to that of WT HSCs. Therapy with the Janus kinase (JAK) inhibitor ruxolitinib ameliorated the thrombocytosis in TG mice and attenuated the increase in number of BM megakaryocytes and HSCs. Taken together, our study provides a model showing that the C-terminal of mutant CALR activated JAK-STAT signaling specifically downstream of MPL and may have a central role in CALR-induced myeloproliferative neoplasms.
- Published
- 2016
22. Early/prefibrotic primary myelofibrosis in patients who were initially diagnosed with essential thrombocythemia
- Author
-
Yuki Tahira, Kouichi Maeda, Yoko Kubuki, Hitoshi Matsuoka, Kousuke Marutsuka, Seiichi Sato, Junzo Ishizaki, Noriaki Kawano, Ayako Kamiunten, Tomonori Hidaka, Takuro Kameda, Kazuya Shimoda, Haruko Shimoda, Masafumi Ito, Kiyoshi Yamashita, Masaaki Sekine, Keiichi Akizuki, Kotaro Shide, Takanori Toyama, Masanori Takeuchi, and Hiroshi Kawano
- Subjects
Adult ,Male ,medicine.medical_specialty ,Myeloid ,Anemia ,World Health Organization ,Gastroenterology ,Diagnosis, Differential ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Biopsy ,medicine ,Humans ,Leukocytosis ,Myelofibrosis ,Aged ,Hematology ,Myeloproliferative Disorders ,medicine.diagnostic_test ,Essential thrombocythemia ,business.industry ,Bone Marrow Examination ,Middle Aged ,medicine.disease ,Survival Analysis ,medicine.anatomical_structure ,Primary Myelofibrosis ,030220 oncology & carcinogenesis ,Splenomegaly ,Female ,Bone marrow ,medicine.symptom ,business ,030215 immunology ,Thrombocythemia, Essential - Abstract
A new entity, namely early/prefibrotic primary myelofibrosis (PMF), was introduced as a subtype of PMF in the 2016 revised World Health Organization (WHO) criteria for myeloproliferative neoplasms (MPN). It was diagnosed based on histopathological features of bone marrow (BM) biopsy specimens together with clinical parameters [leukocytosis, anemia, elevated lactate dehydrogenase (LDH) values, and splenomegaly]. The aim of this study was to evaluate the prevalence of early/prefibrotic PMF in patients who were previously diagnosed with ET, and to compare clinical features at diagnosis and outcomes between early/prefibrotic PMF and essential thrombocythemia (ET) patients. BM biopsy samples obtained at the time of ET diagnosis were available in 42 patients. Sample reevaluation according to the 2016 revised WHO criteria revealed that early/prefibrotic PMF accounted for 14% of patients who were previously diagnosed with ET, which was comparable to the rates in previous reports. Compared to patients with ET, patients with early/prefibrotic PMF had higher LDH values and higher frequencies of splenomegaly. Overall, myelofibrosis-free and acute myeloid leukemia-free survivals were comparable between the 2 groups. Accurate diagnosis is required to clarify the clinical features of Japanese ET patients.
- Published
- 2018
23. Gene expression profiling of loss of TET2 and/or JAK2V617F mutant hematopoietic stem cells from mouse models of myeloproliferative neoplasms
- Author
-
Kenji Nagata, Atsushi Iwama, Akira Kitanaka, Takumi Yamaji, Yoko Kubuki, Hisayoshi Iwakiri, Satoru Hasuike, Makoto Yoshimitsu, Shojiro Yamamoto, Yoshihiro Tahara, Tadashi Miike, Masaaki Sekine, Kazumasa Aoyama, Kotaro Shide, Tomonori Hidaka, Takuro Kameda, Kazuya Shimoda, Ayako Kamiunten, Goro Sashida, and Hiroo Abe
- Subjects
Genetics ,Lineage (genetic) ,Loss of TET2 ,lcsh:QH426-470 ,Cellular differentiation ,Mutant ,Microarray profiling ,Myeloproliferative neoplasm ,Data in Brief Article ,Biology ,Gene mutation ,Biochemistry ,Phenotype ,Hematopoietic stem cell ,Gene expression profiling ,lcsh:Genetics ,Cancer research ,Molecular Medicine ,Epigenetics ,JAK2V617F ,Gene ,Biotechnology - Abstract
Myeloproliferative neoplasms (MPNs) are clinically characterized by the chronic overproduction of differentiated peripheral blood cells and the gradual expansion of malignant intramedullary/extramedullary hematopoiesis. In MPNs mutations in JAK2 MPL or CALR are detected mutually exclusive in more than 90% of cases [1], [2]. Mutations in them lead to the abnormal activation of JAK/STAT signaling and the autonomous growth of differentiated cells therefore they are considered as “driver” gene mutations. In addition to the above driver gene mutations mutations in epigenetic regulators such as TET2 DNMT3A ASXL1 EZH2 or IDH1/2 are detected in about 5%–30% of cases respectively [3]. Mutations in TET2 DNMT3A EZH2 or IDH1/2 commonly confer the increased self-renewal capacity on normal hematopoietic stem cells (HSCs) but they do not lead to the autonomous growth of differentiated cells and only exhibit subtle clinical phenotypes [[4], [6], [7], [8],5]. It was unclear how mutations in such epigenetic regulators influenced abnormal HSCs with driver gene mutations how they influenced the disease phenotype or whether a single driver gene mutation was sufficient for the initiation of human MPNs. Therefore we focused on JAK2V617F and loss of TET2—the former as a representative of driver gene mutations and the latter as a representative of mutations in epigenetic regulators—and examined the influence of single or double mutations on HSCs (Lineage−Sca-1+c-Kit+ cells (LSKs)) by functional analyses and microarray whole-genome expression analyses [9]. Gene expression profiling showed that the HSC fingerprint genes [10] was statistically equally enriched in TET2-knockdown-LSKs but negatively enriched in JAK2V617F–LSKs compared to that in wild-type-LSKs. Double-mutant-LSKs showed the same tendency as JAK2V617F–LSKs in terms of their HSC fingerprint genes but the expression of individual genes differed between the two groups. Among 245 HSC fingerprint genes 100 were more highly expressed in double-mutant-LSKs than in JAK2V617F–LSKs. These altered gene expressions might partly explain the mechanisms of initiation and progression of MPNs which was observed in the functional analyses [9]. Here we describe gene expression profiles deposited at the Gene Expression Omnibus (GEO) under the accession number GSE62302 including experimental methods and quality control analyses.
- Published
- 2015
- Full Text
- View/download PDF
24. Loss of TET2 has dual roles in murine myeloproliferative neoplasms: disease sustainer and disease accelerator
- Author
-
Taku Harada, Kazuya Shimoda, Kazumasa Aoyama, Kousuke Marutsuka, Kotaro Shide, Makoto Yoshimitsu, Ayako Kamiunten, Masaaki Sekine, Hisayoshi Iwakiri, Satoru Hasuike, Hiroo Abe, Tomonori Hidaka, Mitsue Sueta, Takuro Kameda, Tadashi Miike, Shojiro Yamamoto, Kenji Nagata, Yasuhiro Taniguchi, Goro Sashida, Takumi Yamaji, Yoshihiro Tahara, Akira Kitanaka, Yoko Kubuki, Haruko Shimoda, and Atsushi Iwama
- Subjects
Transgene ,Immunology ,Mice, Transgenic ,Disease ,Biochemistry ,Dioxygenases ,Flow cytometry ,Mice ,Proto-Oncogene Proteins ,medicine ,Animals ,Leukocytosis ,Gene ,Oligonucleotide Array Sequence Analysis ,Myeloproliferative Disorders ,medicine.diagnostic_test ,business.industry ,Hematopoietic stem cell ,Cell Biology ,Hematology ,Janus Kinase 2 ,Flow Cytometry ,medicine.disease ,Extramedullary hematopoiesis ,DNA-Binding Proteins ,Mice, Inbred C57BL ,Transplantation ,medicine.anatomical_structure ,medicine.symptom ,business - Abstract
Acquired mutations of JAK2 and TET2 are frequent in myeloproliferative neoplasms (MPNs). We examined the individual and cooperative effects of these mutations on MPN development. Recipients of JAK2V617F cells developed primary myelofibrosis-like features; the addition of loss of TET2 worsened this JAK2V617F-induced disease, causing prolonged leukocytosis, splenomegaly, extramedullary hematopoiesis, and modestly shorter survival. Double-mutant (JAK2V617F plus loss of TET2) myeloid cells were more likely to be in a proliferative state than JAK2V617F single-mutant myeloid cells. In a serial competitive transplantation assay, JAK2V617F cells resulted in decreased chimerism in the second recipients, which did not develop MPNs. In marked contrast, cooperation between JAK2V617F and loss of TET2 developed and maintained MPNs in the second recipients by compensating for impaired hematopoietic stem cell (HSC) functioning. In-vitro sequential colony formation assays also supported the observation that JAK2V617F did not maintain HSC functioning over the long-term, but concurrent loss of TET2 mutation restored it. Transcriptional profiling revealed that loss of TET2 affected the expression of many HSC signature genes. We conclude that loss of TET2 has two different roles in MPNs: disease accelerator and disease initiator and sustainer in combination with JAK2V617F.
- Published
- 2015
- Full Text
- View/download PDF
25. Nasopharyngeal Carcinoma with Bone Marrow Metastasis: Positive Response to Weekly Paclitaxel Chemotherapy
- Author
-
Yoko Kubuki, Sae Miyaushiro, Tomonori Hidaka, Kazuya Shimoda, Takuro Kameda, Yoriko Ishiguro, Masaaki Sekine, Akira Kitanaka, Kotaro Shide, Takayuki Kawabata, Ayako Kamiunten, and Yoshiko Umekita
- Subjects
Male ,Oncology ,medicine.medical_specialty ,Paclitaxel ,medicine.medical_treatment ,Neutropenia ,chemistry.chemical_compound ,Internal medicine ,Internal Medicine ,medicine ,Carcinoma ,Humans ,Disseminated intravascular coagulation ,Chemotherapy ,Nasopharyngeal Carcinoma ,Performance status ,business.industry ,Nasopharyngeal Neoplasms ,General Medicine ,Middle Aged ,medicine.disease ,chemistry ,Nasopharyngeal carcinoma ,Radiology ,Neoplasm Recurrence, Local ,Bone Marrow Neoplasms ,business ,Chemoradiotherapy - Abstract
A 51-year-old man with nasopharyngeal carcinoma underwent chemoradiotherapy with cisplatin and 5-fluorouracil, followed by a left cervical lymphadenectomy. Distant metastatic disease was excluded using fluoro-deoxyglucose positron emission tomography. Seven months later, bone marrow metastasis and disseminated intravascular coagulation were diagnosed. The patient received weekly paclitaxel therapy and maintained a good performance status for seven months. During the treatment period, the patient developed no severe organ toxicity except for neutropenia. Weekly paclitaxel may therefore be considered as the treatment of choice in patients with advanced or recurrent nasopharyngeal carcinoma with bone marrow metastasis.
- Published
- 2015
- Full Text
- View/download PDF
26. Thrombohemorrhagic events, disease progression, and survival in polycythemia vera and essential thrombocythemia: a retrospective survey in Miyazaki prefecture, Japan
- Author
-
Noriaki Kawano, Tomonori Hidaka, Kiyoshi Yamashita, Takuro Kameda, Yuki Tahira, Keiichi Akizuki, Kotaro Shide, Kousuke Marutsuka, Ayako Kamiunten, Masanori Takeuchi, Hitoshi Matsuoka, Masafumi Ito, Masaaki Sekine, Seiichi Sato, Kazuya Shimoda, Takanori Toyama, Junzo Ishizaki, Kouichi Maeda, Hiroshi Kawano, Yoko Kubuki, and Haruko Shimoda
- Subjects
Adult ,Male ,medicine.medical_specialty ,Adolescent ,Hemorrhage ,03 medical and health sciences ,Leukocyte Count ,Young Adult ,0302 clinical medicine ,Polycythemia vera ,Japan ,Retrospective survey ,Predictive Value of Tests ,Risk Factors ,Internal medicine ,medicine ,Humans ,Polycythemia Vera ,Aged ,Retrospective Studies ,Aged, 80 and over ,Hematology ,business.industry ,Essential thrombocythemia ,Incidence ,Disease progression ,Thrombosis ,Middle Aged ,medicine.disease ,Survival Rate ,030220 oncology & carcinogenesis ,Risk stratification ,Disease Progression ,Observational study ,Female ,business ,030215 immunology ,Thrombocythemia, Essential - Abstract
Polycythemia vera (PV) and essential thrombocythemia (ET) are associated with life-threatening thrombohemorrhagic events, and disease progression and development of non-hematological malignancies also reduce long-term survival. We retrospectively surveyed thrombohemorrhagic events and overall survival (OS) in 62 PV and 117 ET patients. The cumulative incidences of thrombohemorrhagic events in PV and ET patients were 11.3 and 10.3%, and the incidence rates were 2.42 and 1.85 per 100 person-years. The combined incidence rates of disease progression and development of non-hematological malignancies in PV and ET patients were 1.73 and 1.69 per 100 person-years. The incidence rates of thrombohemorrhagic events in our Japanese PV/ET patients were lower than those reported by most Western studies, but were comparable to those in the largest prospective observational study in ET patients. The combined incidence rates of disease progression and development of non-hematological malignancies were similar between Japanese and Western PV/ET patients. In ET patients, the conventional risk stratification model based on the presence of advanced age or history of thrombosis was useful to predict thrombosis risk, and both the conventional model and the International Prognostic Score of thrombosis in ET based on the above 2 risk factors plus increased leukocyte count could predict poor survival.
- Published
- 2017
27. Loss of Tyrosine Kinase 2 Does Not Affect the Severity of
- Author
-
Takumi, Yamaji, Kotaro, Shide, Takuro, Kameda, Masaaki, Sekine, Ayako, Kamiunten, Tomonori, Hidaka, Yoko, Kubuki, Haruko, Shimoda, Hiroo, Abe, Tadashi, Miike, Hisayoshi, Iwakiri, Yoshihiro, Tahara, Mitsue, Sueta, Shojiro, Yamamoto, Satoru, Hasuike, Kenji, Nagata, and Kazuya, Shimoda
- Subjects
TYK2 Kinase ,Mice ,Myeloproliferative Disorders ,Liver ,Mutation ,Animals ,Mice, Transgenic ,Janus Kinase 2 ,Lung ,Protein Kinase Inhibitors ,Spleen - Abstract
In myeloproliferative neoplasms (MPN), Janus kinase 2 (JAK2) is activated by mutations including JAK2V617F (JAK2VF). It is unclear whether JAK kinases [i.e. JAK1, JAK2, JAK3, or tyrosine kinase 2 (TYK2)] other than JAK2 have cooperative actions such as enhancement or suppression of JAK2. If other kinases enhance activation, therapies that co-target them could have a therapeutic efficacy. We examined the role of TYK2 in Jak2VF-induced murine MPN.We crossed Jak2VF transgenic mice and Tyk2-knockout (Tyk2KO) mice to generate Jak2VF/Tyk2KO mice. The disease severity and treatment effect with a JAK2 inhibitor was compared between Jak2VF and Jak2VF/Tyk2KO mice.Both types of mice developed MPN, and there were no differences in peripheral blood counts, spleen weight, or survival period. Upon JAK2 inhibitor therapy, both types of mice had equally improved leukocytosis and splenomegaly.TYK2 does not have cooperative effects with JAK2VF upon MPN onset nor in the presence of a JAK2 inhibitor.
- Published
- 2017
28. TET2 mutation in diffuse large B-cell lymphoma
- Author
-
Yoko, Kubuki, Takumi, Yamaji, Tomonori, Hidaka, Takuro, Kameda, Kotaro, Shide, Masaaki, Sekine, Ayako, Kamiunten, Keiichi, Akizuki, Haruko, Shimoda, Yuuki, Tahira, Kenichi, Nakamura, Hiroo, Abe, Tadashi, Miike, Hisayoshi, Iwakiri, Yoshihiro, Tahara, Mitsue, Sueta, Shojiro, Yamamoto, Satoru, Hasuike, Kenji, Nagata, Akira, Kitanaka, and Kazuya, Shimoda
- Subjects
Male ,Middle Aged ,Dioxygenases ,Hematopoiesis ,DNA-Binding Proteins ,Leukemia, Myeloid ,hemic and lymphatic diseases ,Hematologic Neoplasms ,Proto-Oncogene Proteins ,Humans ,Female ,Original Article ,Lymphoma, Large B-Cell, Diffuse ,Frameshift Mutation ,Aged - Abstract
Ten-eleven translocation-2 (TET2) mutation is frequently observed in myeloid malignancies, and loss-of-function of TET2 is essential for the initiation of malignant hematopoiesis. TET2 mutation presents across disease entities and was reported in lymphoid malignancies. We investigated TET2 mutations in 27 diffuse large B-cell lymphoma (DLBCL) patients and found a frameshift mutation in 1 case (3.7%). TET2 mutation occurred in some populations of DLBCL patients and was likely involved in the pathogenesis of their malignancies.
- Published
- 2017
29. Effects of mogamulizumab in adult T-cell leukemia/lymphoma in clinical practice
- Author
-
Kiyoshi Yamashita, Junzo Ishizaki, Tomonori Hidaka, Takuro Kameda, Seiichi Sato, Haruko Shimoda, Yoko Kubuki, Takanori Toyama, Kouichi Maeda, Ayako Kamiunten, Keiichi Akizuki, Masanori Takeuchi, Yuki Tahira, Hitoshi Matsuoka, Noriaki Kawano, Hiroshi Kawano, Kazuya Shimoda, Akira Kitanaka, Masaaki Sekine, and Kotaro Shide
- Subjects
Oncology ,Adult ,Male ,medicine.medical_specialty ,Phases of clinical research ,Antibodies, Monoclonal, Humanized ,Adult T-cell leukemia/lymphoma ,03 medical and health sciences ,0302 clinical medicine ,Antineoplastic Agents, Immunological ,Refractory ,Recurrence ,hemic and lymphatic diseases ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Mogamulizumab ,Humans ,Leukemia-Lymphoma, Adult T-Cell ,Adverse effect ,Aged ,Retrospective Studies ,Aged, 80 and over ,business.industry ,Hematology ,General Medicine ,Middle Aged ,medicine.disease ,Rash ,Survival Analysis ,Lymphoma ,Clinical trial ,Treatment Outcome ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Retreatment ,Female ,medicine.symptom ,business ,030215 immunology ,medicine.drug - Abstract
Objective The efficacy of mogamulizumab in adult T-cell leukemia/lymphoma (ATLL) was reported in a previous phase 2 study. Compared with patients in clinical trials, however, most patients in real-life settings have demonstrated worse outcomes. Method We retrospectively analyzed 96 patients with relapsed/refractory ATLL who received mogamulizumab treatment. Results Relapsed/refractory ATLL patients with a median age of 70 years received a median of five courses of mogamulizumab. Hematologic toxicity and skin rash were the most common adverse events, and both were manageable. Of 96 patients, 87 were evaluable for efficacy. The overall response rate was 36%, and the median progression-free survival (PFS) and overall survival (OS) from the start of mogamulizumab therapy were 1.8 and 4.0 months, respectively. Of the original 96 patients, only 25 fulfilled the inclusion criteria of the phase 2 study. Those who met the criteria demonstrated longer median PFS and OS durations of 2.7 and 8.5 months, respectively. The median OS from diagnosis in relapsed/refractory ATLL patients receiving mogamulizumab was 12 months, longer than the 5.8 months in a historical cohort without mogamulizumab. Conclusion In clinical practice, mogamulizumab exhibited antitumor activity in patients with relapsed/refractory ATLL, with an acceptable toxicity profile. Mogamulizumab therapy improved the OS of ATLL patients.
- Published
- 2017
30. Depletion of Neoplastic CD11b Positive Cells in Jak2V617F Mutant Mice Reduced Fibrocytes in Bone Marrow and Improved Myelofibrosis
- Author
-
Fumiyo Toyoshima, Kotaro Shide, Kenichi Nakamura, Yuki Tahira, Kazuya Shimoda, Yoshinori Ozono, Keiichi Akizuki, Akira Sawaguchi, Yoko Kubuki, Hisayoshi Iwakiri, Satoru Hasuike, Kenji Nagata, Masaaki Sekine, Ayako Kamiunten, Tomonori Hidaka, and Takuro Kameda
- Subjects
Chemistry ,Monocyte ,Immunology ,Neoplastic Monocyte ,Spleen ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Molecular biology ,medicine.anatomical_structure ,Fibrocyte ,medicine ,CD90 ,Bone marrow ,Myelofibrosis ,Fibroblast - Abstract
Myelofibrosis (MF) associated with myeloproliferative neoplasms (MPN) has been considered to be a reactive phenomenon caused by mesenchymal stromal cells (MSCs) stimulated by cytokines such as TGFb-1 overproduced by neoplastic megakaryocytes (MKs) and platelets. TGFb-1 stimulates non-neoplastic mesenchymal cells to produce collagen and fibronectin and to induces bone marrow (BM) fibrosis. However, the involvement of neoplastic fibrocyte in MF has recently been reported (Verstovsek et al. JEM 2016), and among blood cells, monocytes in particular are considered to be the main source of neoplastic fibrocytes. In this study, we assesed the role of neoplastic fibrocytes using a mouse model of MPN induced by Jak2V617F (Shide et al. Leukemia 2008). First, the distribution of neoplastic fibrocyte in the BM of Jak2V617F transgenic (TG) mice was examined. We transplanted wild-type (WT) or Jak2V617F TG cells (B6-CD45.2), together with WT BM cells (B6-CD45.1) into irradiated WT recipient mice (B6-CD45.1). Only recipient mice transplanted with a mixture of Jak2V617F cells and WT cells developed BM fibrosis. In immunofluorescent staining of fibrotic BM, cells expressing the fibrocyte marker CD45/Collagen-1(Col-1) were observed much more than cells expressing the fibroblast marker CD90(usually positive for MSCs)/Col-1. As for CD45/Col-1 positive cells, cells expressing CD45.2/Col-1 were much more than cells expressing CD45.1/Col-1, clearly indicating that these cells were derived from Jak2V617F mutant blood cells. On the other hand, in the BM of recipient mice transplanted with control WT cells, few cells expressing CD45/Col-1 or CD90/Col-1 were present. To examine the differentiation ability of Jak2V617F blood cells to fibrocytes directly, peripheral blood (PB) mononuclear cells (MNC) of Jak2V617F mice or WT mice were cultured in vitro. After 5 days of culture, PB MNCs from Jak2V617F mice differentiated into mature fibrocytes exhibiting a long spindle shape with Col-1 expression. On the other hand, there were very few fibrocytes differentiated from PB MNC from WT mice. Next, we depleted monocytes, the main source of fibrocytes, and observed its effects on BM fibrosis in vivo. Jak2V617F TG mice were mated with CD11b-diphtheria toxin receptor (DTR) TG mice (Duffield et al. JCI 2005) to obtain Jak2V617F/CD11b-DTR double TG mice. Mice transplanted with BM cells from Jak2V617F/CD11b-DTR double TG mice (hereinafter called Jak2V617F/CD11b-DTR mice) exhibit leukocytosis, thrombocytosis, anemia, splenomegaly, and BM fibrosis with increased megakaryocytes. Jak2V617F/CD11b-DTR mice was administered diphtheria toxin (DT) intraperitoneally to deplete monocytes. One day after DT administration, the number of PB monocytes (CD11b+/F4/80+) drastically decreased in Jak2V617F/CD11b-DTR mice, and the reduction of monocyte was maintained by every-other-day DT administration. After 8 weeks DT treatment, mice were sacrificed and analyzed. As a control group, Jak2V617F/CD11b-DTR mice treated with PBS were examined. DT treatment drastically decreased the number of neoplastic fibrocytes expressing CD45.2/Col-1 in BM and spleen of Jak2V617F/CD11b-DTR mice compared with control mice treated with PBS. Consistently, reticulin fibers were eliminated almost completely and collagen fibers almost fully disappeared in BM, which led to a reversal of the decrease in BM cellularity, although the number of MKs was not affected. Similar findings were observed in the spleen, although not completely. Plasma TGF-b1 level were about 2-fold higher in Jak2V617F/CD11b-DTR mice than in WT mice. Neoplastic monocyte depletion significantly decreased TGF-b1 level. Since MK numbers did not change, this indicates that fibrocytes are one of the main sources of TGF-b1. In other features of MF in Jak2V617F/CD11b-DTR mice, splenomegaly was ameliorated by DT treatment. Microscopic analysis revealed an improvement in the damaged spleen architecture and the disappearance of splenic fibrosis. In summary, most collagen-producing cells in BM were neoplastic fibrocytes in Jak2V617F-induced MPN, indicating that neoplastic fibrocytes played an essential role and mesenchymal fibroblasts had a minor contribution in fibrosis in MPN. Depletion of neoplastic monocytes also improved splenomegaly as well as BM fibrosis in mice, and this cell fraction could be a promising therapeutic target. Disclosures No relevant conflicts of interest to declare.
- Published
- 2019
- Full Text
- View/download PDF
31. The Role of Calreticulin in Normal Hematopoiesis and Neoplastic Hematopoiesis of Myeloproliferative Neoplasms
- Author
-
Yoko Kubuki, Yoshinori Ozono, Masaya Ono, Kazuya Shimoda, Keiichi Akizuki, Takako Yokomizo, Goro Sashida, Tomonori Hidaka, Sho Kubota, Takuro Kameda, Ayako Kamiunten, Yuki Tahira, Kotaro Shide, and Masaaki Sekine
- Subjects
Myeloid ,CD40 ,biology ,Chemistry ,Immunology ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Molecular biology ,Haematopoiesis ,Leukemia ,medicine.anatomical_structure ,medicine ,biology.protein ,Erythropoiesis ,Bone marrow ,Myelofibrosis ,Calreticulin - Abstract
Mutations in the Calreticulin (CALR) gene were identified in cases of myeloproriferative neoplasms (MPNs), and various functions of the CALR mutant protein are being elucidated. On the other hand, few data are available on the role of CALR in the hematopoietic system. The knockout (KO) mice of Calr are impaired in expression of transcription factors necessary for cardiac development and are embryonic lethal. To clarify the role of CALR in normal hematopoiesis and MPN pathogenesis, we generated hematopoietic cell-specific Calr KO mice. Mice carrying floxed allele targeted exons 4-7 of Calr (Calrf/+ mice) and Calr heterozygous KO mice (Calr+/− mice) (Tokuhiro et al., Sci Rep 2015) were crossed with Mx1-Cre transgenic mice and obtained mice with three genotypes; Mx1-cre;Calr+/+, Mx1-cre;Calr+/−, and Mx1-cre;Calrf/−. Floxed alleles were then deleted by intraperitoneal injection with polyinosinic: polycytidilic acid. No differences were found in the peripheral blood (PB) leukocyte count, hemoglobin levels, or platelet count among the three genotypes of mice. The proportions of Mac1+ or Gr1+ myeloid lineage cells, B220+ B cells, and CD3+ T cells among the three groups were comparable. In the bone marrow (BM), cell pellet from Mx1-cre;Calrf/− mice appeared anemic and the proportion of CD71+/Ter119+ erythroid cells and the number of CFU-E were significantly lower in Mx1-cre;Calrf/− BM cells compared to the other two genotypes of BM cells. On the other hand, no difference was found in other mature cells such as myeloid, T, B, or CD41+ megakaryocytes (Mks) in BM. As for HSCs and progenitors, Mx1-cre;Calrf/− mice exhibited a higher proportion of MPP and GMP compared to the other two genotypes of mice. We found no difference in other progenitor compartments including long- and short-term HSCs among the three groups. In contrast to the minor effect on BM and PB cells, The spleen weight in Mx1-cre;Calrf/− mice was about 2-fold heavier than that in Mx1-cre;Calr+/+ mice. In spleens from Mx1-cre;Calrf/− mice, the border between the white and red pulp was obscured. Mks and maturing myeloid cells had markedly infiltrated into the red pulp. In FACS analysis, mature myeloid cells, erythroid cells, and Mks were significantly increased in spleens from Mx1-cre;Calrf/− mice compared to those from the other two genotypes of mice. HSCs and most types of myeloid progenitors were also increased in the spleen. Hematopoiesis in the spleen may compensate for the reduced erythropoiesis in the BM, as no anemia was seen in Mx1-cre;Calrf/− mice. No onset of leukemia or myelofibrosis was observed, and no difference in survival was seen among the three groups following observation for 2 years. As CALR plays a role as a chaperone in the endoplasmic reticulum (ER) during protein synthesis, we searched for proteins with expression that was reduced by Calr deficiency. The quantitative differences of proteins in Mac1+/Gr1+ BM cells between Mx1-cre;Calrf/− and Mx1-cre;Calr+/+ mice were compared using proteomics analysis by 2DICAL, a shotgun proteomics analysis system (Ono et al. Cellular Proteomics 2006). We found that only a few proteins, including myeloperoxidase and CALR itself, were reduced, and conversely, many proteins were increased by the absence of CALR. List of differentially upregulated proteins higher than those in WT samples were analyzed using Metascape (http://metascape.org) to determine enriched pathways. The most enriched cluster was "protein processing in the ER", and 19 out of 76 upregulated (>1.8-fold) proteins were included in this cluster. In real-time PCR analysis, we confirmed that ER chaperon genes (BiP, Grp94, Hsp40, Pdia3, Pdia4, Pdia6) and genes involved in ER-related degradation (Trap, Bap31, Derl1, p97) were significantly upregulated in Mx1-cre;Calrf/− myeloid cells. These observations suggested that chaperone dysfunction due to Calr deficiency is compensated by upregulation of unfolded protein response (UPR) pathway. In summary, Calr deficiency induced erythroid hypoplasia in the BM, and induced splenomegaly and extramedullary hematopoiesis. This suggests that the amount of WT CALR expression remaining in CALR mutant cells may modify the phenotype of MPN patients. Absence of myeloperoxidase protein and upregulation of UPR pathway in myeloid cells from Calr KO mice indicates that CALR plays a significant role as a chaperone also in hematopoiesis and that CALR deficient cells are exposed to ER stress. Disclosures No relevant conflicts of interest to declare.
- Published
- 2019
- Full Text
- View/download PDF
32. Mice with Calr mutations homologous to human CALR mutations only exhibit mild thrombocytosis
- Author
-
Tomonori Hidaka, Yoshihiro Tahara, Takuro Kameda, Kazuya Shimoda, Mitsue Sueta, Masaaki Sekine, Akira Kitanaka, Hiroo Abe, Yoko Kubuki, Arata Honda, Shojiro Yamamoto, Asami Oji, Yuki Tahira, Kenichi Nakamura, Ayako Kamiunten, Hisayoshi Iwakiri, Tadashi Miike, Satoru Hasuike, Kotaro Shide, Kenji Nagata, Keiichi Akizuki, Yoshinori Ozono, and Masahito Ikawa
- Subjects
Mutant ,medicine.disease_cause ,lcsh:RC254-282 ,Article ,Frameshift mutation ,Mice ,Exon ,medicine ,Animals ,Humans ,KDEL Motif ,Myelofibrosis ,Thrombocytosis ,Mutation ,biology ,Essential thrombocythemia ,Chemistry ,Hematology ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Molecular biology ,Oncology ,biology.protein ,Calreticulin - Abstract
Shide, K., Kameda, T., Kamiunten, A. et al. Mice with Calr mutations homologous to human CALR mutations only exhibit mild thrombocytosis. Blood Cancer J. 9, 42 (2019). https://doi.org/10.1038/s41408-019-0202-z, Calreticulin (CALR) exon 9 frameshift mutations, commonly detected in essential thrombocythemia (ET) and primary myelofibrosis patients, activate signal transducer and activator of transcription (STAT) proteins in the presence of Myeloproliferative Leukemia Virus (MPL) and induce ET in vivo. Loss of the KDEL motif, an endoplasmic reticulum retention signal, and generation of many positively charged amino acids (AAs) in the mutated C-terminus are thought to be important for disease induction. To test this hypothesis, we generated mice harboring a Calr frameshift mutation using the CRISPR/Cas9 system. Deletion of 19-base pairs in exon 9 (c.1099-1117del), designated the del19 mutation, induced loss of the KDEL motif and generated many positively charged AAs, similar to human mutants. Calr del19 mice exhibited mild thrombocytosis, slightly increased megakaryocytes, and mild splenomegaly. In vitro experiments revealed that the murine CALR del19 mutant had a weaker ability to combine with murine MPL than the human CALR del52 mutant. Consequently, STAT5 activation was also very weak downstream of the murine mutant and murine MPL, and may be the reason for the mild disease severity. In summary, loss of the KDEL motif and positively charged AAs in the C-terminus of CALR is insufficient for MPL binding and ET development.
- Published
- 2019
33. Differences in Hematological and Clinical Features Between Essential Thrombocythemia Cases With JAK2- or CALR-Mutations
- Author
-
Yoshihiro Tahara, Ayako Kamiunten, Kenichi Nakamura, Yoko Kubuki, Tomonori Hidaka, Takuro Kameda, Yuki Tahira, Kanna Hashimoto, Hisayoshi Iwakiri, Satoru Hasuike, Mitsue Sueta, Hiroo Abe, Akira Kitanaka, Kenji Nagata, Haruko Shimoda, Tadashi Miike, Keiichi Akizuki, Masaaki Sekine, Shojiro Yamamoto, Kotaro Shide, Kazuya Shimoda, and Takumi Yamaji
- Subjects
Adult ,Male ,Adolescent ,Clinical Biochemistry ,Molecular Sequence Data ,Polymorphism, Single Nucleotide ,03 medical and health sciences ,Exon ,chemistry.chemical_compound ,Young Adult ,0302 clinical medicine ,Text mining ,Sex Factors ,Sex factors ,Medicine ,Humans ,Amino Acid Sequence ,Child ,Peptide sequence ,Letter to the Editor ,Aged ,Genetics ,Aged, 80 and over ,Janus kinase 2 ,biology ,business.industry ,Essential thrombocythemia ,Biochemistry (medical) ,Age Factors ,General Medicine ,DNA ,Exons ,Sequence Analysis, DNA ,Janus Kinase 2 ,Middle Aged ,medicine.disease ,Diagnostic Hematology ,chemistry ,030220 oncology & carcinogenesis ,biology.protein ,Female ,business ,Calreticulin ,Receptors, Thrombopoietin ,030215 immunology ,Thrombocythemia, Essential - Published
- 2016
34. TET2 Mutation in Adult T-Cell Leukemia/Lymphoma
- Author
-
Akira Kitanaka, Yoko Kubuki, Kazuya Shimoda, Mitsue Sueta, Hiroo Abe, Hisayoshi Iwakiri, Satoru Hasuike, Kotaro Shide, Tadashi Miike, Keiichi Akizuki, Tomonori Hidaka, Takuro Kameda, Ayako Kamiunten, Takumi Yamaji, Shojiro Yamamoto, Masaaki Sekine, Kenji Nagata, Haruko Shimoda, Yoshihiro Tahara, and Kenichi Nakamura
- Subjects
Male ,Myeloid ,Genotype ,Biology ,Adult T-cell leukemia/lymphoma ,Dioxygenases ,immune system diseases ,hemic and lymphatic diseases ,Proto-Oncogene Proteins ,medicine ,Missense mutation ,Humans ,Leukemia-Lymphoma, Adult T-Cell ,Genetic Association Studies ,Aged ,Aged, 80 and over ,General Medicine ,Middle Aged ,medicine.disease ,Immunohistochemistry ,Lymphoma ,DNA-Binding Proteins ,Haematopoiesis ,Leukemia ,medicine.anatomical_structure ,Cell Transformation, Neoplastic ,Immunology ,Mutation ,Cancer research ,Female ,Stem cell - Abstract
Loss-of-function of ten-eleven translocation-2 (TET2) is a common event in myeloid malignancies, and plays pleiotropic roles, including augmenting stem cell self-renewal and skewing hematopoietic cells to the myeloid lineage. TET2 mutation has also been reported in lymphoid malignancies; 5.7~12% of diffuse large B-cell lymphomas and 18~83% of angioimmunoblastic T-cell lymphomas had TET2 mutations. We investigated TET2 mutations in 22 adult T-cell leukemia/lymphoma (ATLL) patients and identified a missense mutation in 3 cases (14%). TET2 mutation occurred in a number of ATLL patients and was likely involved in their leukemogenesis.
- Published
- 2016
35. Haploinsufficiency of CALR Confers Hematopoietic Stem Cells (HSCs) with a Clonal Advantage over Wild-Type Cells, and, in Setting of Myeloproliferative Neoplasms, Compensates for the Functions of HSCs Impaired By the Calr Mutation
- Author
-
Yoshinori Ozono, Yuki Tahira, Masaaki Sekine, Keiichi Akizuki, Yoko Kubuki, Ayako Kamiunten, Kotaro Shide, Tomonori Hidaka, Kazuya Shimoda, Takuro Kameda, Takako Yokomizo, and Goro Sashida
- Subjects
Immunology ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,Molecular biology ,Transplantation ,03 medical and health sciences ,Leukemia ,Haematopoiesis ,0302 clinical medicine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,medicine ,Bone marrow ,Progenitor cell ,Stem cell ,Haploinsufficiency ,Myelofibrosis ,030215 immunology - Abstract
CALR exon9 frameshift mutations function as driver mutations in essential thrombocythemia (ET) and primary myelofibrosis patients with non-mutated JAK2 or MPL. The mutations augment signal transducer and activator of transcription activity in the presence of MPL, induce increased cell proliferation and growth factor independence in cell lines, and cause ET-like myeloproliferative neoplasms (MPN) in mice. In tumor cells bearing the CALR mutation, mutant CALR protein expression occurs while wild-type (WT) CALR expression is decreased by half. Although the biological activity of mutant CALR has been elucidated in detail, the significance of CALR haploinsufficiency is unclear. The purpose of this study was therefore to clarify the influence of CALR haploinsufficiency on hematopoiesis in normal and CALR-mutated MPN in mice. First, we analyzed the effect of CALR haploinsufficiency on hematopoiesis using CALR heterozygous knockout (CALR-hKO) mice (Tokuhiro et al. Sci Rep. 2015). Blood cell counts, liver and spleen weight, histology, cell fraction in bone marrow (BM) and spleen, and survival of CALR-hKO mice were all equivalent to that observed in WT mice. In the analysis of progenitor cells by fluorescence-activated cell sorting and colony formation assays, no difference was observed in the amount of progenitor cells and in colony replating capacity between WT mice and CALR-hKO mice. Interestingly, in competitive serial transplantation experiments using whole BM cells in primary and secondary transplanted mice, CALR-hKO cells showed higher levels of chimerism than WT cells. Next, the effect of CALR haploinsufficiency on hematopoiesis in mutant CALR-del52 overexpressing mice was analyzed. We compared three groups of mice, WT mice, CALR-del52 transgenic (TG) mice (Shide et al. Leukemia 2017) and CALR-del52 TG/CALR-hKO double-mutant (TG-hKO) mice. Both TG mice and TG-hKO mice developed ET-like MPN. Compared to WT mice, increases in megakaryocytes, platelets and hematopoietic progenitor cells (including HSCs) were observed in these mice. However, no differences were observed between TG mice and TG-hKO mice. Finally, 4000 LSK cells sorted from WT mice, TG mice, and TG-hKO mice (B6-Ly5.2) and B6-Ly5.1 competitor cells (1 × 106 WT BM cells) were mixed and injected into lethally irradiated B6-Ly5.1 recipient mice, and the percent chimerism of donor cells was followed for 1 year after transplantation. From 12 weeks after transplantation, the chimerism of TG cells was significantly lower than that of control WT cells, suggesting that the CALR mutation has a negative influence on clonal expansion of HSCs. The recipient mice transplanted with TG LSK cells showed very mild thrombocythemia. On the other hand, chimerism in TG-hKO cells was significantly higher than that in control WT cells up to 12 weeks after transplantation. Chimerism at 20 weeks after transplantation was equivalent to that in control WT cells, and the recipient mice transplanted with TG-hKO LSK cells showed severe thrombocythemia. These findings show that CALR haploinsufficiency compensates for HSCs functioning, which has been impaired by the CALR mutation, enhancing the ability of HSCs to develop ET. Mice experiments have showed that HSCs with the JAK2V617F mutation are fragile, and it is considered that mutations in DNMT3A or TET2 often occurred prior to mutations in JAK2 to compensate for the defect in self renewal capacity. Conversely, since the CALR mutation is not typically preceded by any mutations, it is considered that the CALR mutation may not require any preceding mutations. Our results showed that, like JAK2 V617F mutants, overexpression of the CALR-del52 mutant impairs HSC functioning. Furthermore, we found that CALR haploinsufficiency restores the functions of impaired HSCs to the same extent as that found in WT HSCs. When an HSC acquires the CALR mutation, "defect" and "recovery" states thus occur simultaneously in the cell. This finding may explain why mutant CALR clones expand without needing to undergo any preceding mutation. Disclosures No relevant conflicts of interest to declare.
- Published
- 2018
- Full Text
- View/download PDF
36. TET2 Mutation Associated with Organ Infiltrations in ATLL
- Author
-
Seishi Ogawa, Yoko Kubuki, Michihiro Hidaka, Atae Utsunomiya, Takayuki Ohshima, Keisuke Kataoka, Keiichi Akizuki, Masaaki Sekine, Yuki Tahira, Ayako Kamiunten, Kazuya Shimoda, Kotaro Shide, Tomonori Hidaka, and Takuro Kameda
- Subjects
Pathology ,medicine.medical_specialty ,Lung ,biology ,business.industry ,Immunology ,Disease progression ,Spleen ,Cell Biology ,Hematology ,biology.organism_classification ,medicine.disease ,Biochemistry ,Lymphoma ,Leukemia ,medicine.anatomical_structure ,Human T-lymphotropic virus 1 ,Mutation (genetic algorithm) ,Medicine ,business ,Organ weight - Abstract
Introduction: Adult T-cell leukemia/lymphoma (ATLL) is a peripheral T-cell lymphoma that is caused by HTLV-1. The prognosis of acute and lymphomatous variants of ATLL is poor, ranging from 2 weeks to >1 year. Compared to other types of malignant lymphomas, the organ infiltration is frequently observed in ATLL (Yamada et al. Leuk Lymphoma 1997). We previously reported the landscape of genetic mutations in ATLL, and showed that various mutations occurred in the TCR-NFκB pathway in more than 90% of ATLL cases (Kataoka et al. Nat Genet 2015). These somatic mutations are thought to develop ATLL in combination with viral genes such as HTLV-1 bZIP factor (HBZ). Among them, mutations in TET2, an epigenetic regulator, was observed in about 10% of ATLL cases. Higher frequencies inTET2 mutation was reported in other types of peripheral T-cell lymphoma (PTCL); it was observed in about 80% of angioimmunoblastic T-cell lymphoma (AITL) and in about 50% of PTCL, not otherwise specified. In PTCL, it has been reported that additional mutations in lymphoid progenitors derived from TET2 mutated hematopoietic stem cells cause increased cell proliferation and anti-apoptosis, leading to the disease progression. In ATLL, the role of TET2 mutation in disease progression is still unknown. In this study, we investigated the role of TET2 mutation in ATLL using mouse model and acute and lymphomatous variant ATLL cohort. Materials and methods: As an animal model of HTLV-1 infection or ATLL, transgenic mice expressing HBZ under the control of the mouse CD4 promoter (HBZ-Tg) were generated with C57BL/6 background. Heterozygous TET2 knock-down mice (TET2KD) were generated with C57BL/6 background by gene trapping (Tang et al. Transgenic Res 2008; Shide et al. Leukemia 2012). HBZ-Tg/TET2KD compound mice (double mutant) were generated by crossing them. HBZ-Tg, TET2KD, and double mutant mice were investigated by cell counts, organ weight, FACS analysis, pathological analysis, and survival analysis. The relationship between the TET2 mutation status and the clinical feature was investigated using our acute and lymphomatous variant ATLL cohort (n=115). Result: At 12 months, compared to wild type mouse (WT), sporadic splenomegaly and lymphadenopathy were observed in HBZ-Tg. No significant increase was observed in peripheral blood (PB) leukocyte and mononuclear cell (MNC) of BM and spleen, but an increase was observed in the estimated whole body MNC (Femur x 100/6 + spleen) (WT vs. HBZ-Tg; estimated whole body MNC (x106 cells/body), 416±162 vs. 621±147, p=0.01). In FACS analysis, the frequency of CD4+ T-cell was increased in PB, spleen, and BM (WT vs. HBZ-Tg; PB-CD4+ T-cell%, 4.9±0.9 vs. 28.2±22.8, p Next, to elucidate the role of TET2 mutation in ATLL, the double mutant was analyzed. At six months, compared to HBZ-Tg, no increase was observed in the number of PB leukocyte, spleen-MNC, and BM-MNC, and also in the frequency and the number of CD4+ T-cells in PB, spleen and BM. However, in pathological and survival analysis, the double mutant showed severe cell infiltration in lung and liver and demonstrated inferior OS (median OS (month), 11.1 vs. 6.0, p Conclusion: In both mice model and human cohort, TET2 mutation exacerbated organ infiltration of ATLL cells. Disclosures No relevant conflicts of interest to declare.
- Published
- 2018
- Full Text
- View/download PDF
37. Physiological Expression of Calr Mutant Increases Cell Growth and Cytokine Independency in Human Cell Lines Expressing Mpl, and Develops Essential Thrombocythemia in Mice
- Author
-
Kotaro Shide, Arata Honda, Masaaki Sekine, Akira Sawaguchi, Yoko Kubuki, Akira Kitanaka, Kazuya Shimoda, Yuki Tahira, Tomonori Hidaka, Keiichi Akizuki, Takuro Kameda, and Ayako Kamiunten
- Subjects
Cloning ,Cell growth ,Essential thrombocythemia ,medicine.medical_treatment ,Immunology ,Mutant ,Spleen ,Cell Biology ,Hematology ,Biology ,Colony-stimulating factor ,medicine.disease ,Biochemistry ,Cell biology ,medicine.anatomical_structure ,Cytokine ,medicine ,Bone marrow - Abstract
Calreticulin (CALR) exon 9 mutations were reported in about two-thirds of JAK2 or MPL mutation negative ET and PMF patients. The mutations cause frameshifts that result in proteins with novel C-terminus.Retrovirus-mediated gene transfer into cell lines and mouse bone marrow (BM) cells is a common technique, but the expression level is very high compared to the physiological expression.We investigated the effects of physiological expression of mutant CALR using CRISPR/Cas9 gene editing techniques for cell lines, and as for the mouse model, we generated a transgenic mice (TG) expressing human CALR del52 mutant. We used two human cell lines expressing MPL: human acute megakaryoblastic leukemia cell line CMK11-5 which expressed endogenous MPL, and F-36P-MPL cell line which was generated by introducing MPL to GM-CSF-dependent erythroleukemia cell line F-36P. Plasmids coexpressing hCas9 and single-guide RNA were prepared by ligating oligonucleotides (5'-CACCGACAAGAAACGCAAAGAGGAGG-3', 5'-AAACCCTCCTCTTTGCGTTTCTTGTC-3') for the target sequence of human CALR exon 9 into pX330. The plasmids were introduced with a electroporator to each of the cell lines. After limiting dilution cloning, we identified cell lines which have indel mutation at the target site. We produced two types of CMK11-5 subline knocked in a CALR mutation, namely CALR del25 CMK cells and CALR del25/del17 CMK cells, respectively. The former lacks 25 bases in one CALR allele, causing a frameshift that results in a protein resembling human CALR mutant, while the latter lacks an additional 17 bases in another allele in CALR exon 9 and induces a frameshift that causes a deletion in CALR exon 9. Both kinds of CALR mutant CMK11-5 cells showed increased cell proliferation compared to WT cells. We also produced one type of F-36P-MPL subline, CALR del1/ins1 F-36P-MPL cells which had 1 base deletion in one CALR allele resembling human mutation and 1 base insertion in another allele. Though the growth of this subline in the presence of GM-CSF was comparable to WT cells, it showed GM-CSF independent autonomous cell growth. We generated TG mice expressing human CALR del52 mutant driven by the murine H2Kb promoter. We compared the expression level of human CALR mRNA in TG BM cells with the expression of endogenous WT CALR in human cell lines (CMK11-5, F-36P-MPL, CHRF288) using Rn18s as an endogenous control. The expression of human CALR in TG BM was approximately 0.6 times that of endogenous WT CALR in human cell lines, and the physiological expression level was obtained. They exhibited thrombocytosis, with platelet (PLT) counts as high as 2,000 x 109/L. Leukocyte number and the proportion of granulocytes and T and B lymphocytes, were comparable to WT mice. CALR mutation had no impact on Hb level or spleen weight. There was a striking difference in the number of megakaryocytes (Mgks), which was 2-fold higher in BM from TG mice than in WT mice. The TG Mgks were also more mature, with larger diameter, and contained higher number of alpha-granules compared to WT cells. In one year of observation, there is no fibrosis in BM. These observations showed that TG developed human ET-like disease. The survival of TG mice was comparable to that of WT mice. The disease phenotype was transplantable into WT recipient mice. To characterize in detail the impact of MPNs induced by the CALR del52 mutant, we evaluated the frequencies of HSCs and progenitors in BM. The frequency of both LT-HSC and ST-HSC in BM was higher inTG mice compared to WT mice. The frequencies of progenitors (CMP, MEP, and MKP) were also greater in BM from TG mice than from WT mice. However, BM cells did not have enhanced replating capacity. We next examined whether or not ruxolitinib (RUX) treatment ameliorated thrombocytosis in TG mice. Either 90 mg/kg bid of RUX or vehicle was administrated to TG mice for 4 weeks.TG mice treated with vehicle showed a mean 16% increase in PLT count during the treatment period, probably due to the disease progression. RUX treatment attenuated the increase in the number of PLTs in TG mice by a mean of 22%, but the overall count was still higher than that in WT mice. BM sections showed that RUX reduced the Mgks number in TG. In summary, physiological expression of CALR mutant increases cell growth and cytokine independency in human cell lines expressing MPL, and develops ET in mice. RUX therapy attenuated the increased numbers of peripheral blood PLTs and BM Mgks, and ameliorated CALR mutation-induced ET. Disclosures No relevant conflicts of interest to declare.
- Published
- 2016
- Full Text
- View/download PDF
38. Mogamulizumab for ATLL in Clinical Practice
- Author
-
Masaki Takeuchi, Kazuya Shimoda, Akira Kitanaka, Takanori Toyama, Kiyoshi Yamashita, Tomonori Hidaka, Takuro Kameda, Junzo Ishizaki, Masaaki Sekine, Seiichi Sato, Akizuki Keiichi, Kotaro Shide, Ayako Kamiunten, Yoko Kubuki, and Kouichi Maeda
- Subjects
medicine.medical_specialty ,Combination therapy ,business.industry ,Immunology ,Phases of clinical research ,Cell Biology ,Hematology ,CHOP ,medicine.disease ,Biochemistry ,Chemotherapy regimen ,Rash ,Surgery ,Lymphoma ,030207 dermatology & venereal diseases ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Mogamulizumab ,medicine.symptom ,business ,Adverse effect ,medicine.drug - Abstract
Introduction Adult T-cell leukemia/lymphoma (ATLL) is an aggressive peripheral T cell neoplasm that is resistant to conventional chemotherapy and carries a poor prognosis. The effect of mogamulizumab, an immunoglobulin (Ig) G1 monoclonal antibody targeting CCR4 for ATLL cells, was reported in a previous phase 2 study in which mogamulizumab monotherapy was evaluated in relapsed ATLL patients. The overall response rate (ORR), median progression-free survival (PFS) and median overall survival (OS) were 50%, 5.2 and 13.7 months, respectively. It was not stated whether these values were derived in the real world or in clinical practice. Here we evaluate the clinical impact of mogamulizumab treatment in CCR-4-positive aggressive ATLL patients in clinical practice. Patients and methods We retrospectively analyzed 101 CCR-4-positive ATLL patients who received at least one cycle of mogamulizumab infusion between March, 2012 and April, 2016 in 7 facilities in Miyazaki prefecture, an HTLV-1 endemic area in Southwestern Japan. The ORR, PFS, OS and adverse effects (AEs) were evaluated. We next compared OS in patients with at least one course of mogamulizumab therapy with that in historical control patients without mogamulizumab therapy. Results Of the 101 patients, 92 were evaluable for treatment response, survival and AEs. The median age was 70 years old (range; 45 to 90), and 52 patients (51%) were more than 70 years old. According to Shimoyama's criteria, 66 patients were classified as acute type, 32 as lymphoma type, and 3 as chronic type. All 3 chronic-type ATLL patients had at least one unfavorable risk factor. Of the 101 patients, 96 had refractory or relapsed ATLL when mogamulizumab treatment was started, and the prior treatments consisted of VCAP-AMP-VECP, CHOP, DeVIC or CHASE therapy, with an average of 2 courses. In the 5 remaining cases, mogamulizumab was administered as the initial therapy for ATLL. Mogamulizumab was administered as monotherapy in 87 cases (86%), and as combination therapy with other drugs in 14 cases (14%). The ORR was 37%, including a complete remission rate of 19%. The median PFS and OS were 1.8 and 4.2 months, respectively. Among the 101 patients treated with mogamulizumab, only 26 (26%) fulfilled the inclusion criteria of the phase 2 clinical study. Among patients who met those inclusion criteria, the median PFS and OS were 6.0 and 8.4 months, respectively. The use of mogamulizumab improved OS in clinical practice. The median OS of patients receiving mogamulizumab therapy was 12 months, whereas that of patients who did not receive mogamulizumab in the historical cohort was 8.4 months. Hematologic toxicity and skin rash were the most common AEs, and both were manageable Conclusion Mogamulizumab therapy showed clinically meaningful activity in ATLL patients, with an acceptable toxicity in clinical practice. Disclosures No relevant conflicts of interest to declare.
- Published
- 2016
- Full Text
- View/download PDF
39. Therapies Targeting the MAPK Pathway Improve Bone Marrow (BM) Fibrosis Induced By JAK2V617F
- Author
-
Tomonori Hidaka, Takuro Kameda, Yoko Kubuki, Haruko Shimoda, Masaaki Sekine, Akira Kitanaka, Keiichi Akizuki, Kotaro Shide, Ayako Kamiunten, and Kazuya Shimoda
- Subjects
MAPK/ERK pathway ,medicine.medical_specialty ,business.industry ,MEK inhibitor ,medicine.medical_treatment ,Immunology ,Stimulation ,Cell Biology ,Hematology ,Pharmacology ,medicine.disease ,Biochemistry ,Erythropoietin receptor ,Granulocyte colony-stimulating factor ,Endocrinology ,Cytokine ,Fibrosis ,Internal medicine ,medicine ,Myelofibrosis ,business - Abstract
In primary myelofibrosis patients, somatic mutations such as JAK2V617F(JAKVF) and MPLW515 that activate JAK-STAT signaling are often seen. Small-molecule JAK2 inhibitors are effective for organomegaly and constitutional symptoms, but the drugs have little effect on BM fibrosis. To clarify the mechanism by which MPN cells with JAK2 mutations cause BM fibrosis, we compared the gene expression patterns of Lin−Sca1+ BM cells in JAK2VF transgenic mice (JAK2VF-TG), which develop myelofibrosis (MF), with that in WT mice. We found that TGFb1 and HOXB4, the target genes of transcription factor USF1 were highly expressed. TGFβ1, which is secreted by hematopoietic cells, is essential for fibrotic development in a murine model of MF (Chagraoui et al. Blood 2002), and increased expression of HOXB4 enhances human megakaryocytic development (Zhong et al. BBRC 2010). To investigate the mechanism of the high expression of these genes downstream of JAK2 signaling, USF1 and a cytokine receptor gene (MPL, EPOR or CSF3R) were co-transfected into 293T cells along with either a TGF-β1/HOXB4 promoter-driven or a STAT5 response element-driven luciferase reporter. Stimulation of MPL with TPO enhanced USF1 transcriptional activity about 3 fold, but stimulation of EPOR with EPO or of CSF3R with G-CSF did not change this activity. However, stimulation with any of the 3 types of cytokines enhanced STAT5 transcriptional activity. JAK2VF upregulated USF1 and STAT5 much more highly than JAK2WT without TPO stimulation. This USF1 upregulation specifically to TPO/MPL signaling was suppressed by a dominant negative mutant of USF1, JAK2 inhibitors (AG490, NS-018) or MEK inhibitors (U0126, PD325901). Inhibition of PI3K or p38MAPK did not affect the USF1 activation. Co-treatment with JAK2 and MEK inhibitors showed a synergistic effect in blocking both USF1 upregulation and STAT5 activation induced by JAK2VF. Next, we tested the MEK inhibitor, PD325901, in combination with the JAK2 inhibitor, NS-018, in the JAK2VF-TG mice. After disease was established 12 weeks after birth, JAK2VF-TG mice were divided into the following 4 groups: vehicle control; PD325901 monotherapy; NS-018 monotherapy; and combined therapy. PD325901 (5 mg/kg) and NS-018 (50 mg/kg) were orally administered once and twice daily, respectively. After 12 weeks of treatment, we evaluated the effect on BM fibrosis. The grading of MF in each group (n = 5-6) was as follows: vehicle control (MF-0: 0/6, MF-1 or 2: 6/6); PD325901 monotherapy (MF-0: 4/5, MF-1 or 2: 1/5); NS-018 monotherapy (MF-0: 0/6, MF-1 or 2: 6/6); and combined therapy (MF-0: 3/6, MF-1 or 2: 3/6). In the 2 groups treated with PD325901, 50~80% of mice showed MF-0. In contrast, in vehicle-treated or NS-018 monotherapy groups, all mice showed MF-1 or 2. Consistent with the MF grading, BM cellularity was significantly increased in the PD325901 monotherapy or combined therapy groups compared with the vehicle-treated group. A significant reduction was seen in the plasma TGFβ1 concentration in the PD325901 monotherapy and combined therapy groups compared with the vehicle-treated group (9.7 ng/ml, 8.1 ng/ml vs. 18.2 ng/ml, respectively). The TGFβ1 concentration in the extracellular fluid of BM (Wagner et al blood 2007) was also significantly reduced (5.6 ng/ml, 6.8 ng/ml vs. 9.1 ng/ml, respectively). BM cellularity and the TGFβ1 concentration in the NS-018 monotherapy group were comparable to those in the vehicle-treated group. Interestingly, megakaryocytes in the PD325901 monotherapy and combined therapy groups were decreased in number and were smaller than those in the vehicle-treated or NS-018 monotherapy groups. Regarding the effect on splenomegaly, spleen weight was significantly reduced in the NS-018 monotherapy and combined therapy groups compared with the vehicle-treated group (0.83 g, 0.69 g vs. 1.18 g, respectively). PD325901 monotherapy had little effect on splenomegaly. It is known that MEK-ERK1/2 pathway is critical in normal megakaryocyte development. In vitro data suggest that JAK2VF activates this pathway downstream of MPL and may contribute to TGFβ1 overproduction and dysmegakaryopoiesis, causing BM fibrosis via transcriptional enhancement of USF1. In vivo data suggest that MEK inhibition has the potential to improve dysmegakaryopoiesis and BM fibrosis. The combined therapy of JAK2 inhibitors with MEK inhibitors might be a promising therapy for improving both splenomegaly and BM fibrosis. Disclosures No relevant conflicts of interest to declare.
- Published
- 2014
- Full Text
- View/download PDF
40. TET2 Mutation in Adult T-Cell Leukemia/Lymphoma.
- Author
-
Kazuya Shimoda, Kotaro Shide, Takuro Kameda, Tomonori Hidaka, Yoko Kubuki, Ayako Kamiunten, Masaaki Sekine, Keiichi Akizuki, Haruko Shimoda, Takumi Yamaji, Kenichi Nakamura, Hiroo Abe, Tadashi Miike, Hisayoshi Iwakiri, Yoshihiro Tahara, Mitsue Sueta, Shojiro Yamamoto, Satoru Hasuike, Kenji Nagata, and Akira Kitanaka
- Published
- 2015
- Full Text
- View/download PDF
41. Differences in Hematological and Clinical Features Between Essential Thrombocythemia Cases With JAK2- or CALR-Mutations.
- Author
-
Yoko Kubuki, Kotaro Shide, Takuro Kameda, Takumi Yamaji, Masaaki Sekine, Ayako Kamiunten, Keiichi Akizuki, Haruko Shimoda, Yuki Tahira, Kenichi Nakamura, Hiroo Abe, Tadashi Miike, Hisayoshi Iwakiri, Yoshihiro Tahara, Mitsue Sueta, Kanna Hashimoto, Shojiro Yamamoto, Satoru Hasuike, Tomonori Hidaka, and Kenji Nagata
- Subjects
THROMBOCYTOSIS ,HEMATOLOGICAL manifestations of general diseases ,JANUS kinases ,CALRETICULIN ,DISEASE prevalence ,GENETIC mutation ,GENETICS - Abstract
The article offers information on the essential thrombocythemia (ET). Topics discussed include background information about ET including its characteristics and the prevalence of the disease, provides details on a research study conducted to determine the hematological and clinical differences between Janus kinase 2 and calreticulin mutations
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.