32 results on '"Kanezaki R"'
Search Results
2. Physical association of the patient-specific GATA1 mutants with RUNX1 in acute megakaryoblastic leukemia accompanying Down syndrome
- Author
-
Xu, G, Kanezaki, R, Toki, T, Watanabe, S, Takahashi, Y, Terui, K, Kitabayashi, I, and Ito, E
- Published
- 2006
- Full Text
- View/download PDF
3. The key role of stem cell factor/KIT signaling in the proliferation of blast cells from Down syndrome-related leukemia
- Author
-
Toki, T, primary, Kanezaki, R, additional, Adachi, S, additional, Fujino, H, additional, Xu, G, additional, Sato, T, additional, Suzuki, K, additional, Tauchi, H, additional, Endo, M, additional, and Ito, E, additional
- Published
- 2008
- Full Text
- View/download PDF
4. Landscape of driver mutations and their clinical effects on Down syndrome-related myeloid neoplasms.
- Author
-
Sato T, Yoshida K, Toki T, Kanezaki R, Terui K, Saiki R, Ojima M, Ochi Y, Mizuno S, Yoshihara M, Uechi T, Kenmochi N, Tanaka S, Matsubayashi J, Kisai K, Kudo K, Yuzawa K, Takahashi Y, Tanaka T, Yamamoto Y, Kobayashi A, Kamio T, Sasaki S, Shiraishi Y, Chiba K, Tanaka H, Muramatsu H, Hama A, Hasegawa D, Sato A, Koh K, Karakawa S, Kobayashi M, Hara J, Taneyama Y, Imai C, Hasegawa D, Fujita N, Yoshitomi M, Iwamoto S, Yamato G, Saida S, Kiyokawa N, Deguchi T, Ito M, Matsuo H, Adachi S, Hayashi Y, Taga T, Saito AM, Horibe K, Watanabe K, Tomizawa D, Miyano S, Takahashi S, Ogawa S, and Ito E
- Subjects
- Humans, Male, Female, Leukemoid Reaction genetics, Infant, Child, Preschool, Exome Sequencing, Prognosis, Leukemia, Myeloid genetics, Infant, Newborn, Child, Core Binding Factor Alpha 2 Subunit genetics, Down Syndrome genetics, Down Syndrome complications, Mutation
- Abstract
Abstract: Transient abnormal myelopoiesis (TAM) is a common complication in newborns with Down syndrome (DS). It commonly progresses to myeloid leukemia (ML-DS) after spontaneous regression. In contrast to the favorable prognosis of primary ML-DS, patients with refractory/relapsed ML-DS have poor outcomes. However, the molecular basis for refractoriness and relapse and the full spectrum of driver mutations in ML-DS remain largely unknown. We conducted a genomic profiling study of 143 TAM, 204 ML-DS, and 34 non-DS acute megakaryoblastic leukemia cases, including 39 ML-DS cases analyzed by exome sequencing. Sixteen novel mutational targets were identified in ML-DS samples. Of these, inactivations of IRX1 (16.2%) and ZBTB7A (13.2%) were commonly implicated in the upregulation of the MYC pathway and were potential targets for ML-DS treatment with bromodomain-containing protein 4 inhibitors. Partial tandem duplications of RUNX1 on chromosome 21 were also found, specifically in ML-DS samples (13.7%), presenting its essential role in DS leukemia progression. Finally, in 177 patients with ML-DS treated following the same ML-DS protocol (the Japanese Pediatric Leukemia and Lymphoma Study Group acute myeloid leukemia -D05/D11), CDKN2A, TP53, ZBTB7A, and JAK2 alterations were associated with a poor prognosis. Patients with CDKN2A deletions (n = 7) or TP53 mutations (n = 4) had substantially lower 3-year event-free survival (28.6% vs 90.5%; P < .001; 25.0% vs 89.5%; P < .001) than those without these mutations. These findings considerably change the mutational landscape of ML-DS, provide new insights into the mechanisms of progression from TAM to ML-DS, and help identify new therapeutic targets and strategies for ML-DS., (© 2024 American Society of Hematology. Published by Elsevier Inc. All rights are reserved, including those for text and data mining, AI training, and similar technologies.)
- Published
- 2024
- Full Text
- View/download PDF
5. Antileukemic effect of azacitidine, a DNA methyltransferase inhibitor, on cell lines of myeloid leukemia associated with Down syndrome.
- Author
-
Tanaka T, Kudo K, Kanezaki R, Yuzawa K, Toki T, Okuse R, Kobayashi A, Sato T, Kamio T, Terui K, and Ito E
- Subjects
- Humans, Cell Line, DNA, Methyltransferases, Azacitidine pharmacology, Azacitidine therapeutic use, Down Syndrome complications, Down Syndrome drug therapy, Down Syndrome genetics, Enzyme Inhibitors pharmacology, Enzyme Inhibitors therapeutic use, Interferon Type I, Leukemia, Myeloid, Acute complications, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics
- Abstract
Myeloid leukemia associated with Down syndrome (ML-DS) responds well to chemotherapy and has a favorable prognosis, but the clinical outcome of patients with refractory or relapsed ML-DS is dismal. We recently reported a case of relapsed ML-DS with an effective response to a DNA methyltransferase inhibitor, azacitidine (AZA). However, the efficacy of AZA for refractory or relapsed ML-DS remains uncertain. Here, we investigated the effects and mechanism of action of AZA on three ML-DS cell lines derived from relapsed cases. AZA inhibited the proliferation of all examined ML-DS cell lines to the same extent as that of AZA-sensitive acute myeloid leukemia non-Down syndrome cell lines. Transient low-dose AZA treatment exerted durable antileukemic effects on ML-DS cells. The inhibitory effect included cell cycle arrest, apoptosis, and reduction of aldehyde dehydrogenase activity. Comprehensive differential gene expression analysis showed that AZA induced megakaryocytic differentiation in all ML-DS cell lines examined. Furthermore, AZA induced activation of type I interferon-stimulated genes, primarily involved in antiproliferation signaling, without stimulation of the interferon receptor-mediated autocrine system. Activation of the type I interferon pathway by stimulation with interferon-α exerted antiproliferative effects on ML-DS cells, suggesting that AZA exerts its antileukemic effects on ML-DS cells at least partially through the type I interferon pathway. Moreover, the effect of AZA on normal hematopoiesis did not differ significantly between individuals with non-Down syndrome and Down syndrome. In summary, this study suggests that AZA is a potentially effective treatment option for ML-DS disease control, including relapsed cases, and has reduced side effects., Competing Interests: Conflict of Interest Disclosure The authors do not have any conflicts of interest to declare in relation to this work., (Copyright © 2024 ISEH -- Society for Hematology and Stem Cells. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
6. Mechanism of KIT gene regulation by GATA1 lacking the N-terminal domain in Down syndrome-related myeloid disorders.
- Author
-
Kanezaki R, Toki T, Terui K, Sato T, Kobayashi A, Kudo K, Kamio T, Sasaki S, Kawaguchi K, Watanabe K, and Ito E
- Subjects
- Child, Humans, Cell Line, Tumor, GATA1 Transcription Factor genetics, Gene Expression Regulation, Oncogenes, Down Syndrome genetics
- Abstract
Children with Down syndrome (DS) are at high risk of transient abnormal myelopoiesis (TAM) and myeloid leukemia of DS (ML-DS). GATA1 mutations are detected in almost all TAM and ML-DS samples, with exclusive expression of short GATA1 protein (GATA1s) lacking the N-terminal domain (NTD). However, it remains to be clarified how GATA1s is involved with both disorders. Here, we established the K562 GATA1s (K562-G1s) clones expressing only GATA1s by CRISPR/Cas9 genome editing. The K562-G1s clones expressed KIT at significantly higher levels compared to the wild type of K562 (K562-WT). Chromatin immunoprecipitation studies identified the GATA1-bound regulatory sites upstream of KIT in K562-WT, K562-G1s clones and two ML-DS cell lines; KPAM1 and CMK11-5. Sonication-based chromosome conformation capture (3C) assay demonstrated that in K562-WT, the - 87 kb enhancer region of KIT was proximal to the - 115 kb, - 109 kb and + 1 kb region, while in a K562-G1s clone, CMK11-5 and primary TAM cells, the - 87 kb region was more proximal to the KIT transcriptional start site. These results suggest that the NTD of GATA1 is essential for proper genomic conformation and regulation of KIT gene expression, and that perturbation of this function might be involved in the pathogenesis of TAM and ML-DS., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
7. BRAF V600E-positive cells as molecular markers of bone marrow disease in pediatric Langerhans cell histiocytosis.
- Author
-
Kudo K, Toki T, Kanezaki R, Tanaka T, Kamio T, Sato T, Sasaki S, Imamura M, Imai C, Ando K, Kakuda H, Doi T, Kawaguchi H, Irie M, Sasahara Y, Tamura A, Hasegawa D, Itakura Y, Watanabe K, Sakamoto K, Shioda Y, Kato M, Kudo K, Fukano R, Sato A, Yagasaki H, Kanegane H, Kato I, Umeda K, Adachi S, Kataoka T, Kurose A, Nakazawa A, Terui K, and Ito E
- Subjects
- Biomarkers, Child, Humans, Mutation, Proto-Oncogene Proteins B-raf genetics, Bone Marrow Diseases, Histiocytosis, Langerhans-Cell diagnosis, Histiocytosis, Langerhans-Cell genetics
- Published
- 2022
- Full Text
- View/download PDF
8. Childhood acute myeloid leukemia with 5q deletion and HNRNPH1-MLLT10 fusion: the first case report.
- Author
-
Kudo K, Kubota Y, Toki T, Kanezaki R, Kobayashi A, Sato T, Kamio T, Sasaki S, Shiba N, Tomizawa D, Adachi S, Yoshida K, Ogawa S, Seki M, Takita J, Ito E, and Terui K
- Subjects
- Child, Gene Fusion, Humans, Transcription Factors genetics, Chromosome Aberrations, Leukemia, Myeloid, Acute genetics
- Published
- 2022
- Full Text
- View/download PDF
9. Dyserythropoietic anaemia with an intronic GATA1 splicing mutation in patients suspected to have Diamond-Blackfan anaemia.
- Author
-
Kobayashi A, Ohtaka R, Toki T, Hara J, Muramatsu H, Kanezaki R, Takahashi Y, Sato T, Kamio T, Kudo K, Sasaki S, Yoshida T, Utsugisawa T, Kanno H, Yoshida K, Nannya Y, Takahashi Y, Kojima S, Miyano S, Ogawa S, Terui K, and Ito E
- Abstract
Diamond-Blackfan anaemia (DBA) shares clinical features with two recently reported sporadic cases of dyserythropoietic anaemia with a cryptic GATA1 splicing mutation (c.871-24 C>T). We hypothesized that some patients clinically diagnosed with DBA but whose causative genes were unknown may carry the intronic GATA1 mutation. Here, we examined 79 patients in our DBA cohort, who had no detectable causative genes. The intronic GATA1 mutation was identified in two male patients sharing the same pedigree that included multiple cases with anaemia. Cosegregation of this mutation and disease in multiple family members provide evidence to support the pathogenicity of the intronic GATA1 mutation., Competing Interests: The authors declare no conflict of interest., (© 2022 The Authors. eJHaem published by British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
10. Correction: Reduced-intensity conditioning is effective for hematopoietic stem cell transplantation in young pediatric patients with Diamond-Blackfan anemia.
- Author
-
Koyamaishi S, Kamio T, Kobayashi A, Sato T, Kudo K, Sasaki S, Kanezaki R, Hasegawa D, Muramatsu H, Takahashi Y, Sasahara Y, Hiramatsu H, Kakuda H, Tanaka M, Ishimura M, Nishi M, Ishiguro A, Yabe H, Sarashina T, Yamamoto M, Yuza Y, Hyakuna N, Yoshida K, Kanno H, Ohga S, Ohara A, Kojima S, Miyano S, Ogawa S, Toki T, Terui K, and Ito E
- Published
- 2021
- Full Text
- View/download PDF
11. Reduced-intensity conditioning is effective for hematopoietic stem cell transplantation in young pediatric patients with Diamond-Blackfan anemia.
- Author
-
Koyamaishi S, Kamio T, Kobayashi A, Sato T, Kudo K, Sasaki S, Kanezaki R, Hasegawa D, Muramatsu H, Takahashi Y, Sasahara Y, Hiramatsu H, Kakuda H, Tanaka M, Ishimura M, Nishi M, Ishiguro A, Yabe H, Sarashina T, Yamamoto M, Yuza Y, Hyakuna N, Yoshida K, Kanno H, Ohga S, Ohara A, Kojima S, Miyano S, Ogawa S, Toki T, Terui K, and Ito E
- Subjects
- Child, Humans, Retrospective Studies, Siblings, Transplantation Conditioning, Anemia, Diamond-Blackfan therapy, Graft vs Host Disease, Hematopoietic Stem Cell Transplantation
- Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is the only curative therapy for the hematologic manifestations of Diamond-Blackfan anemia (DBA). However, data regarding the optimal conditioning regimen for DBA patients are limited. We retrospectively compared the outcomes of DBA patients who underwent HSCT using either myeloablative conditioning (MAC) or reduced-intensity conditioning (RIC) regimens. The patients belonged to a cohort treated at our hospitals between 2000 and 2018. HSCT was performed in 27 of 165 patients (16.4%). The median age at the time of HSCT was 3.6 years. Stem cell sources included bone marrow for 25 patients (HLA-matched sibling donors, n = 5; HLA-mismatched related donors, n = 2; HLA-matched/mismatched unrelated donors, n = 18) or cord blood for 2 patients. MAC or RIC regimens were used in 12 and 15 patients, respectively. Engraftment was successful in all 27 patients who underwent HSCT. Three patients who underwent HSCT using MAC regimens developed sinusoidal obstruction syndrome. The 3-year overall survival (OS) and failure-free survival rates (FFS) post-transplantations were 95.2% and 88.4%, respectively, with no significant differences between MAC and RIC regimens. Our data suggest that HSCTs using RIC regimens are effective and obtain engraftment with excellent OS and FFS for young DBA patients.
- Published
- 2021
- Full Text
- View/download PDF
12. Clinical, cytogenetic, and molecular analyses of 17 neonates with transient abnormal myelopoiesis and nonconstitutional trisomy 21.
- Author
-
Yuzawa K, Terui K, Toki T, Kanezaki R, Kobayashi A, Sato T, Kamio T, Kudo K, Sasaki S, Endo M, Ozono S, Nomura K, and Ito E
- Subjects
- Disease-Free Survival, Female, Humans, Infant, Infant, Newborn, Male, Survival Rate, Chromosomes, Human, Pair 21 genetics, Down Syndrome genetics, Down Syndrome mortality, Down Syndrome pathology, GATA1 Transcription Factor genetics, Mutation, Myelopoiesis genetics
- Abstract
Background: Transient abnormal myelopoiesis (TAM) is a unique myeloproliferative disorder that occurs in neonates with constitutional trisomy 21/Down syndrome (DS). Although TAM also develops in neonates without constitutional trisomy 21, the clinical, cytogenetic, and molecular characteristics of those patients are not fully understood., Procedure: We retrospectively evaluated the clinical and cytogenetic findings and GATA1 mutation status of 17 neonates with TAM and nonconstitutional trisomy 21 tested for GATA1 mutations at our institute, and compared the findings with those of 64 neonates with TAM and constitutional trisomy 21/DS., Results: DS clinical features were observed in five of the 17 (29%) patients. In all patients, both trisomy 21 and GATA1 mutations were detected in diagnostic samples. Over a median follow-up of 33 (range, 0-139) months, early death (< 6 months of age) occurred in four patients (24%). Overall and event-free survivals were not significantly different between the patients with TAM and nonconstitutional trisomy 21 and those with TAM and constitutional trisomy 21/DS (five-year overall survival: 76% ± 10% vs 53% ± 13%, P = 0.40; five-year event-free survival: 55% ± 13% vs 48% ± 12%, P = 0.90). The five-year cumulative incidence of progression to myeloid leukemia of DS was also similar between the groups (21% vs 24%, P = 0.80)., Conclusions: Patients with TAM and nonconstitutional trisomy 21 exhibited similar biology and outcomes to those with TAM and constitutional trisomy 21/DS. The possibility of TAM should be considered even in phenotypically normal neonates with TAM symptoms, for appropriate management., (© 2020 Wiley Periodicals, Inc.)
- Published
- 2020
- Full Text
- View/download PDF
13. Highly sensitive detection of GATA1 mutations in patients with myeloid leukemia associated with Down syndrome by combining Sanger and targeted next generation sequencing.
- Author
-
Terui K, Toki T, Taga T, Iwamoto S, Miyamura T, Hasegawa D, Moritake H, Hama A, Nakashima K, Kanezaki R, Kudo K, Saito AM, Horibe K, Adachi S, Tomizawa D, and Ito E
- Subjects
- Adolescent, Adult, Alleles, Amino Acid Substitution, Biopsy, Bone Marrow pathology, Child, DNA Mutational Analysis, Down Syndrome diagnosis, Female, Genetic Association Studies, Genotype, High-Throughput Nucleotide Sequencing, Humans, Japan, Leukemia, Megakaryoblastic, Acute diagnosis, Male, Young Adult, Down Syndrome complications, Down Syndrome genetics, GATA1 Transcription Factor genetics, Genetic Predisposition to Disease, Leukemia, Megakaryoblastic, Acute complications, Leukemia, Megakaryoblastic, Acute genetics, Mutation
- Abstract
Myeloid leukemia associated with Down syndrome (ML-DS) is characterized by a predominance of acute megakaryoblastic leukemia, the presence of GATA1 mutations and a favorable outcome. Because DS children can also develop conventional acute myeloid leukemia with unfavorable outcome, detection of GATA1 mutations is important for diagnosis of ML-DS. However, myelofibrosis and the significant frequency of dry taps have hampered practical screening of GATA1 mutations using bone marrow (BM) samples. In response to those problems, 82 patients were enrolled in the Japanese Pediatric Leukemia/Lymphoma Study Group AML-D11 study. GATA1 mutations were analyzed by Sanger sequencing (SS) using genomic DNA (gDNA) from BM and cDNA from peripheral blood (PB) followed by targeted next-generation sequencing (NGS) using pooled diagnostic samples. BM and PB samples were obtained from 71 (87%) and 82 (100%) patients, respectively. GATA1 mutations were detected in 46 (56%) and 58 (71%) patients by SS using BM gDNA and PB cDNA, respectively. Collectively, GATA1 mutations were identified in 73/82 (89%) patients by SS. Targeted NGS detected GATA1 mutations in 74/82 (90%) patients. Finally, combining the results of SS with those of targeted NGS, GATA1 mutations were identified in 80/82 (98%) patients. These results indicate that SS using BM gDNA and PB cDNA is a rapid and useful method for screening for GATA1 mutations in ML-DS patients. Thus, a combination of SS and targeted NGS is a sensitive and useful method to evaluate the actual incidence and clinical significance of GATA1 mutations in ML-DS patients., (© 2019 Wiley Periodicals, Inc.)
- Published
- 2020
- Full Text
- View/download PDF
14. Two siblings with familial neuroblastoma with distinct clinical phenotypes harboring an ALK germline mutation.
- Author
-
Kudo K, Ueno H, Sato T, Kubo K, Kanezaki R, Kobayashi A, Kamio T, Sasaki S, Terui K, Kurose A, Yoshida K, Shiozawa Y, Toki T, Ogawa S, and Ito E
- Subjects
- Adrenal Gland Neoplasms genetics, Child, Preschool, Exome, Female, Humans, Infant, Liver Neoplasms genetics, Male, Mediastinal Neoplasms genetics, Pedigree, Phenotype, Point Mutation, Spinal Neoplasms genetics, Anaplastic Lymphoma Kinase genetics, Germ-Line Mutation, Neoplasms, Multiple Primary genetics, Neuroblastoma genetics
- Abstract
The authors report two siblings with familial neuroblastoma with a germline R1275Q mutation of the tyrosine kinase domain of ALK. Whole exome sequencing and copy number variation assay were performed to investigate genetic alterations in the two cases. No common somatic mutations or gene polymorphisms related to the tumorigenesis of neuroblastoma were detected. A distinct pattern involving both segmental chromosomal alteration and MYCN amplification was detected. The diversity of biological behavior of familial neuroblastoma harboring a germline ALK mutation may depend on conventional prognostic factors, such as segmental chromosomal alterations and MYCN amplification, rather than additional acquired mutations., (© 2018 Wiley Periodicals, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
15. De Novo Mutations Activating Germline TP53 in an Inherited Bone-Marrow-Failure Syndrome.
- Author
-
Toki T, Yoshida K, Wang R, Nakamura S, Maekawa T, Goi K, Katoh MC, Mizuno S, Sugiyama F, Kanezaki R, Uechi T, Nakajima Y, Sato Y, Okuno Y, Sato-Otsubo A, Shiozawa Y, Kataoka K, Shiraishi Y, Sanada M, Chiba K, Tanaka H, Terui K, Sato T, Kamio T, Sakaguchi H, Ohga S, Kuramitsu M, Hamaguchi I, Ohara A, Kanno H, Miyano S, Kojima S, Ishiguro A, Sugita K, Kenmochi N, Takahashi S, Eto K, Ogawa S, and Ito E
- Subjects
- Adolescent, Adult, Agammaglobulinemia genetics, Anemia, Diamond-Blackfan genetics, Animals, Child, Preschool, Erythrocytes pathology, Female, Growth Disorders genetics, Humans, Induced Pluripotent Stem Cells pathology, Infant, Infant, Newborn, Male, Mice, Middle Aged, Young Adult, Zebrafish, Bone Marrow pathology, Bone Marrow Diseases genetics, Germ Cells pathology, Mutation genetics, Tumor Suppressor Protein p53 genetics
- Abstract
Inherited bone-marrow-failure syndromes (IBMFSs) include heterogeneous genetic disorders characterized by bone-marrow failure, congenital anomalies, and an increased risk of malignancy. Many lines of evidence have suggested that p53 activation might be central to the pathogenesis of IBMFSs, including Diamond-Blackfan anemia (DBA) and dyskeratosis congenita (DC). However, the exact role of p53 activation in each clinical feature remains unknown. Here, we report unique de novo TP53 germline variants found in two individuals with an IBMFS accompanied by hypogammaglobulinemia, growth retardation, and microcephaly mimicking DBA and DC. TP53 is a tumor-suppressor gene most frequently mutated in human cancers, and occasional germline variants occur in Li-Fraumeni cancer-predisposition syndrome. Most of these mutations affect the core DNA-binding domain, leading to compromised transcriptional activities. In contrast, the variants found in the two individuals studied here caused the same truncation of the protein, resulting in the loss of 32 residues from the C-terminal domain (CTD). Unexpectedly, the p53 mutant had augmented transcriptional activities, an observation not previously described in humans. When we expressed this mutant in zebrafish and human-induced pluripotent stem cells, we observed impaired erythrocyte production. These findings together with close similarities to published knock-in mouse models of TP53 lacking the CTD demonstrate that the CTD-truncation mutations of TP53 cause IBMFS, providing important insights into the previously postulated connection between p53 and IBMFSs., (Copyright © 2018 American Society of Human Genetics. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
16. Exome sequencing identified RPS15A as a novel causative gene for Diamond-Blackfan anemia.
- Author
-
Ikeda F, Yoshida K, Toki T, Uechi T, Ishida S, Nakajima Y, Sasahara Y, Okuno Y, Kanezaki R, Terui K, Kamio T, Kobayashi A, Fujita T, Sato-Otsubo A, Shiraishi Y, Tanaka H, Chiba K, Muramatsu H, Kanno H, Ohga S, Ohara A, Kojima S, Kenmochi N, Miyano S, Ogawa S, and Ito E
- Subjects
- Adult, Alternative Splicing, Anemia, Diamond-Blackfan metabolism, Anemia, Diamond-Blackfan pathology, Animals, CRISPR-Cas Systems, Cell Proliferation, Child, Preschool, Embryo, Nonmammalian, Exons, Female, Gene Editing, Gene Expression, Humans, Infant, K562 Cells, Pedigree, RNA, Ribosomal, 18S metabolism, Ribosomal Proteins deficiency, Siblings, Exome Sequencing, Zebrafish, Anemia, Diamond-Blackfan genetics, Exome, Haploinsufficiency, Point Mutation, RNA, Ribosomal, 18S genetics, Ribosomal Proteins genetics
- Published
- 2017
- Full Text
- View/download PDF
17. ALDH2 polymorphism in patients with Diamond-Blackfan anemia in Japan.
- Author
-
Ikeda F, Toki T, Kanezaki R, Terui K, Yoshida K, Kanno H, Ohga S, Ohara A, Kojima S, Ogawa S, and Ito E
- Subjects
- Humans, Aldehyde Dehydrogenase genetics, Bone Marrow Diseases genetics, Fanconi Anemia genetics, Genetic Variation
- Published
- 2016
- Full Text
- View/download PDF
18. Loss of function mutations in RPL27 and RPS27 identified by whole-exome sequencing in Diamond-Blackfan anaemia.
- Author
-
Wang R, Yoshida K, Toki T, Sawada T, Uechi T, Okuno Y, Sato-Otsubo A, Kudo K, Kamimaki I, Kanezaki R, Shiraishi Y, Chiba K, Tanaka H, Terui K, Sato T, Iribe Y, Ohga S, Kuramitsu M, Hamaguchi I, Ohara A, Hara J, Goi K, Matsubara K, Koike K, Ishiguro A, Okamoto Y, Watanabe K, Kanno H, Kojima S, Miyano S, Kenmochi N, Ogawa S, and Ito E
- Subjects
- Anemia, Diamond-Blackfan physiopathology, Animals, Child, Preschool, DNA Mutational Analysis methods, Erythropoiesis genetics, Exome genetics, Female, Humans, Infant, Infant, Newborn, Male, Pedigree, RNA, Ribosomal genetics, Zebrafish, Anemia, Diamond-Blackfan genetics, Germ-Line Mutation, Metalloproteins genetics, Nuclear Proteins genetics, RNA-Binding Proteins genetics, Ribosomal Proteins genetics
- Abstract
Diamond-Blackfan anaemia is a congenital bone marrow failure syndrome that is characterized by red blood cell aplasia. The disease has been associated with mutations or large deletions in 11 ribosomal protein genes including RPS7, RPS10, RPS17, RPS19, RPS24, RPS26, RPS29, RPL5, RPL11, RPL26 and RPL35A as well as GATA1 in more than 50% of patients. However, the molecular aetiology of many Diamond-Blackfan anaemia cases remains to be uncovered. To identify new mutations responsible for Diamond-Blackfan anaemia, we performed whole-exome sequencing analysis of 48 patients with no documented mutations/deletions involving known Diamond-Blackfan anaemia genes except for RPS7, RPL26, RPS29 and GATA1. Here, we identified a de novo splicing error mutation in RPL27 and frameshift deletion in RPS27 in sporadic patients with Diamond-Blackfan anaemia. In vitro knockdown of gene expression disturbed pre-ribosomal RNA processing. Zebrafish models of rpl27 and rps27 mutations showed impairments of erythrocyte production and tail and/or brain development. Additional novel mutations were found in eight patients, including RPL3L, RPL6, RPL7L1T, RPL8, RPL13, RPL14, RPL18A and RPL31. In conclusion, we identified novel germline mutations of two ribosomal protein genes responsible for Diamond-Blackfan anaemia, further confirming the concept that mutations in ribosomal protein genes lead to Diamond-Blackfan anaemia., (© 2014 John Wiley & Sons Ltd.)
- Published
- 2015
- Full Text
- View/download PDF
19. Gene alterations involving the CRLF2-JAK pathway and recurrent gene deletions in Down syndrome-associated acute lymphoblastic leukemia in Japan.
- Author
-
Hanada I, Terui K, Ikeda F, Toki T, Kanezaki R, Sato T, Kamio T, Kudo K, Sasaki S, Takahashi Y, Hayashi Y, Inukai T, Kojima S, Koike K, Kosaka Y, Kobayashi M, Imaizumi M, Mitsui T, Hori H, Hara J, Horibe K, Nagai J, Goto H, and Ito E
- Subjects
- Adolescent, Asian People, Child, Child, Preschool, Down Syndrome complications, Down Syndrome ethnology, Female, Gene Deletion, Gene Dosage, Humans, Japan, Male, Mutation, Precursor Cell Lymphoblastic Leukemia-Lymphoma complications, Precursor Cell Lymphoblastic Leukemia-Lymphoma ethnology, Receptors, Purinergic P2Y genetics, Signal Transduction, Young Adult, Down Syndrome genetics, Janus Kinase 2 genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Receptors, Cytokine genetics
- Abstract
In Western countries, gene alterations involving the CRLF2-JAK signaling pathway are identified in approximately 50-60% of patients with Down syndrome-associated acute lymphoblastic leukemia (DS-ALL), and this pathway is considered a potential therapeutic target. The frequency of BTG1 deletions in DS-ALL is controversial. IKZF1 deletions, found in 20-30% of DS-ALL patients, are associated with a poor outcome and EBF1 deletions are very rare (∼2%). We analyzed 38 patients to determine the frequencies and clinical implications of CRLF2-JAK pathway genetic alterations and recurrent gene deletions in Japanese DS-ALL patients. We confirmed a high incidence of P2RY8-CRLF2 (29%) and JAK2 mutations (16%), though the frequency of P2RY8-CRLF2 was slightly lower than that in Western countries (∼50%). BTG1 deletions were common in our cohort (25%). IKZF1 deletions were detected in 25% of patients and associated with shorter overall survival (OS). EBF1 deletions were found at an unexpectedly high frequency (16%), and at a significantly higher level in P2RY8-CRLF2-positive patients than in P2RY8-CRLF2-negative patients (44% vs. 4%, P=0.015). Deletions of CDKN2A/B and PAX5 were common in P2RY8-CRLF2-negative patients (48 and 39%, respectively) but not in P2RY8-CRLF2-positive patients (11% each). Associations between these genetic alterations and clinical characteristics were not observed except for inferior OS in patients with IKZF1 deletions. These results suggest that differences exist between the genetic profiles of DS-ALL patients in Japan and in Western countries, and that P2RY8-CRLF2 and EBF1 deletions may cooperate in leukemogenesis in a subset of Japanese DS-ALL patients., (© 2014 Wiley Periodicals, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
20. The landscape of somatic mutations in Down syndrome-related myeloid disorders.
- Author
-
Yoshida K, Toki T, Okuno Y, Kanezaki R, Shiraishi Y, Sato-Otsubo A, Sanada M, Park MJ, Terui K, Suzuki H, Kon A, Nagata Y, Sato Y, Wang R, Shiba N, Chiba K, Tanaka H, Hama A, Muramatsu H, Hasegawa D, Nakamura K, Kanegane H, Tsukamoto K, Adachi S, Kawakami K, Kato K, Nishimura R, Izraeli S, Hayashi Y, Miyano S, Kojima S, Ito E, and Ogawa S
- Subjects
- Base Sequence, CCCTC-Binding Factor, Cell Cycle Proteins genetics, Cell Proliferation, Chromosomal Proteins, Non-Histone genetics, Chromosomes, Human, Pair 21 genetics, Enhancer of Zeste Homolog 2 Protein, GATA1 Transcription Factor genetics, Gene Expression Profiling, Humans, Myeloid Cells, Myeloproliferative Disorders genetics, Nuclear Proteins genetics, Polycomb Repressive Complex 2 genetics, Repressor Proteins genetics, Sequence Analysis, DNA, Cohesins, Down Syndrome genetics, Down Syndrome immunology, Leukemia, Megakaryoblastic, Acute genetics, Leukemoid Reaction genetics
- Abstract
Transient abnormal myelopoiesis (TAM) is a myeloid proliferation resembling acute megakaryoblastic leukemia (AMKL), mostly affecting perinatal infants with Down syndrome. Although self-limiting in a majority of cases, TAM may evolve as non-self-limiting AMKL after spontaneous remission (DS-AMKL). Pathogenesis of these Down syndrome-related myeloid disorders is poorly understood, except for GATA1 mutations found in most cases. Here we report genomic profiling of 41 TAM, 49 DS-AMKL and 19 non-DS-AMKL samples, including whole-genome and/or whole-exome sequencing of 15 TAM and 14 DS-AMKL samples. TAM appears to be caused by a single GATA1 mutation and constitutive trisomy 21. Subsequent AMKL evolves from a pre-existing TAM clone through the acquisition of additional mutations, with major mutational targets including multiple cohesin components (53%), CTCF (20%), and EZH2, KANSL1 and other epigenetic regulators (45%), as well as common signaling pathways, such as the JAK family kinases, MPL, SH2B3 (LNK) and multiple RAS pathway genes (47%).
- Published
- 2013
- Full Text
- View/download PDF
21. Naturally occurring oncogenic GATA1 mutants with internal deletions in transient abnormal myelopoiesis in Down syndrome.
- Author
-
Toki T, Kanezaki R, Kobayashi E, Kaneko H, Suzuki M, Wang R, Terui K, Kanegane H, Maeda M, Endo M, Mizuochi T, Adachi S, Hayashi Y, Yamamoto M, Shimizu R, and Ito E
- Subjects
- Cell Proliferation, Child, Down Syndrome pathology, Humans, Leukemoid Reaction pathology, Megakaryocytes metabolism, Megakaryocytes pathology, Down Syndrome blood, Down Syndrome complications, Down Syndrome genetics, GATA1 Transcription Factor genetics, Leukemoid Reaction complications, Leukemoid Reaction genetics, Sequence Deletion
- Abstract
Children with Down syndrome have an increased incidence of transient abnormal myelopoiesis (TAM) and acute megakaryoblastic leukemia. The majority of these cases harbor somatic mutations in the GATA1 gene, which results in the loss of full-length GATA1. Only a truncated isoform of GATA1 that lacks the N-terminal 83 amino acids (GATA1-S) remains. We found through genetic studies of 106 patients with TAM that internally deleted GATA1 proteins (GATA1-IDs) lacking amino acid residues 77-119 or 74-88 (created by splicing mutations) contributed to the genesis of TAM in 6 patients. Analyses of GATA1-deficient embryonic megakaryocytic progenitors revealed that the GATA1 function in growth restriction was disrupted in GATA1-IDs. In contrast, GATA1-S promoted megakaryocyte proliferation more profoundly than that induced by GATA1 deficiency. These results indicate that the internally deleted regions play important roles in megakaryocyte proliferation and that perturbation of this mechanism is involved in the pathogenesis of TAM.
- Published
- 2013
- Full Text
- View/download PDF
22. Identification of TRIB1 R107L gain-of-function mutation in human acute megakaryocytic leukemia.
- Author
-
Yokoyama T, Toki T, Aoki Y, Kanezaki R, Park MJ, Kanno Y, Takahara T, Yamazaki Y, Ito E, Hayashi Y, and Nakamura T
- Subjects
- Animals, Bone Marrow Cells cytology, Bone Marrow Cells metabolism, CCAAT-Enhancer-Binding Protein-alpha genetics, CCAAT-Enhancer-Binding Protein-alpha metabolism, Cell Differentiation, Down Syndrome complications, Extracellular Signal-Regulated MAP Kinases genetics, Extracellular Signal-Regulated MAP Kinases metabolism, Female, Humans, Immunoblotting, Leukemia, Megakaryoblastic, Acute pathology, Mice, Mice, Inbred C57BL, Phosphorylation, RNA, Messenger genetics, Real-Time Polymerase Chain Reaction, Down Syndrome genetics, Intracellular Signaling Peptides and Proteins genetics, Leukemia, Megakaryoblastic, Acute etiology, Mutation genetics, Oncogenes genetics, Protein Serine-Threonine Kinases genetics
- Abstract
Trib1 has been identified as a myeloid oncogene in a murine leukemia model. Here we identified a TRIB1 somatic mutation in a human case of Down syndrome-related acute megakaryocytic leukemia. The mutation was observed at well-conserved arginine 107 residue in the pseudokinase domain. This R107L mutation remained in leukocytes of the remission stage in which GATA1 mutation disappeared, suggesting the TRIB1 mutation is an earlier genetic event in leukemogenesis. The bone marrow transfer experiment showed that acute myeloid leukemia development was accelerated by transducing murine bone marrow cells with the R107L mutant in which enhancement of ERK phosphorylation and C/EBPα degradation by Trib1 expression was even greater than in those expressing wild-type. These results suggest that TRIB1 may be a novel important oncogene for Down syndrome-related acute megakaryocytic leukemia.
- Published
- 2012
- Full Text
- View/download PDF
23. Down syndrome and GATA1 mutations in transient abnormal myeloproliferative disorder: mutation classes correlate with progression to myeloid leukemia.
- Author
-
Kanezaki R, Toki T, Terui K, Xu G, Wang R, Shimada A, Hama A, Kanegane H, Kawakami K, Endo M, Hasegawa D, Kogawa K, Adachi S, Ikeda Y, Iwamoto S, Taga T, Kosaka Y, Kojima S, Hayashi Y, and Ito E
- Subjects
- Alternative Splicing, Down Syndrome genetics, Female, Gene Expression, Humans, Infant, Newborn, Leukemia, Myeloid genetics, Male, Myeloproliferative Disorders genetics, Phenotype, Down Syndrome complications, GATA1 Transcription Factor genetics, Leukemia, Myeloid etiology, Mutation, Myeloproliferative Disorders complications
- Abstract
Twenty percent to 30% of transient abnormal myelopoiesis (TAM) observed in newborns with Down syndrome (DS) develop myeloid leukemia of DS (ML-DS). Most cases of TAM carry somatic GATA1 mutations resulting in the exclusive expression of a truncated protein (GATA1s). However, there are no reports on the expression levels of GATA1s in TAM blasts, and the risk factors for the progression to ML-DS are unidentified. To test whether the spectrum of transcripts derived from the mutant GATA1 genes affects the expression levels, we classified the mutations according to the types of transcripts, and investigated the modalities of expression by in vitro transfection experiments using GATA1 expression constructs harboring mutations. We show here that the mutations affected the amount of mutant protein. Based on our estimates of GATA1s protein expression, the mutations were classified into GATA1s high and low groups. Phenotypic analyses of 66 TAM patients with GATA1 mutations revealed that GATA1s low mutations were significantly associated with a risk of progression to ML-DS (P < .001) and lower white blood cell counts (P = .004). Our study indicates that quantitative differences in mutant protein levels have significant effects on the phenotype of TAM and warrants further investigation in a prospective study.
- Published
- 2010
- Full Text
- View/download PDF
24. Platelet-derived growth factor may be associated with fibrosis in a Down syndrome patient with transient myeloproliferative disorder.
- Author
-
Ogawa J, Kanegane H, Tsuneyama K, Kanezaki R, Futatani T, Nomura K, Ishizawa S, Sasahara M, Ito E, and Miyawaki T
- Subjects
- Humans, Infant, Newborn, Male, Receptor, Platelet-Derived Growth Factor alpha, Receptor, Platelet-Derived Growth Factor beta, Down Syndrome complications, Fibrosis etiology, Myeloproliferative Disorders pathology, Platelet-Derived Growth Factor physiology, Receptors, Platelet-Derived Growth Factor analysis
- Abstract
Transient myeloproliferative disorder (TMD) is experienced by approximately 10% of neonates with Down syndrome (DS). Most TMD is asymptomatic and the patients undergo spontaneous remission within a few months. However, some cases are fatal because of systemic organ dysfunctions including hepatic fibrosis. Some cytokines such as platelet-derived growth factor (PDGF) may be involved in the development of hepatic fibrosis in TMD. The report describes a fatal case of TMD accompanying DS. The patient presented with pulmonary hypertension and hepatic failure. An autopsy disclosed severe fibrosis in the lung, liver, kidney and pancreas. Immunohistochemical analysis revealed high expression of PDGF receptor beta in the severe fibrotic areas of the fibrotic tissues. A real-time polymerase chain reaction (PCR) analysis demonstrated the expression of PDGFalpha and PDGFbeta in the peripheral blood samples of the patient. The finding indicates that the PDGF pathway may play an important role in the fibrosis of several organs in patients with TMD.
- Published
- 2008
- Full Text
- View/download PDF
25. Functional analysis of JAK3 mutations in transient myeloproliferative disorder and acute megakaryoblastic leukaemia accompanying Down syndrome.
- Author
-
Sato T, Toki T, Kanezaki R, Xu G, Terui K, Kanegane H, Miura M, Adachi S, Migita M, Morinaga S, Nakano T, Endo M, Kojima S, Kiyoi H, Mano H, and Ito E
- Subjects
- Base Sequence, Cell Proliferation drug effects, Dose-Response Relationship, Drug, Down Syndrome metabolism, Down Syndrome pathology, Female, Humans, Janus Kinase 3 antagonists & inhibitors, Janus Kinase 3 metabolism, Leukemia, Megakaryoblastic, Acute metabolism, Leukemia, Megakaryoblastic, Acute pathology, Male, Molecular Sequence Data, Myeloproliferative Disorders metabolism, Myeloproliferative Disorders pathology, Neoplasm Proteins antagonists & inhibitors, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Quinazolines pharmacology, Signal Transduction genetics, Tumor Cells, Cultured, Down Syndrome genetics, Janus Kinase 3 genetics, Leukemia, Megakaryoblastic, Acute genetics, Mutation, Myeloproliferative Disorders genetics
- Abstract
JAK3 mutations have been reported in transient myeloproliferative disorder (TMD) as well as in acute megakaryoblastic leukaemia of Down syndrome (DS-AMKL). However, functional consequences of the JAK3 mutations in TMD patients remain undetermined. To further understand how JAK3 mutations are involved in the development and/or progression of leukaemia in Down syndrome, additional TMD patients and the DS-AMKL cell line MGS were screened for JAK3 mutations, and we examined whether each JAK3 mutation is an activating mutation. JAK3 mutations were not detected in 10 TMD samples that had not previously been studied. Together with our previous report we detected JAK3 mutations in one in 11 TMD patients. Furthermore, this study showed for the first time that a TMD patient-derived JAK3 mutation (JAK3(I87T)), as well as two novel JAK3 mutations (JAK3(Q501H) and JAK3(R657Q)) identified in an MGS cell line, were activating mutations. Treatment of MGS cells and Ba/F3 cells expressing the JAK3 mutants with JAK3 inhibitors significantly decreased their growth and viability. These results suggest that the JAK3 activating mutation is an early event during leukaemogenesis in Down syndrome, and they provide proof-of-principle evidence that JAK3 inhibitors would have therapeutic effects on TMD and DS-AMKL patients carrying activating JAK3 mutations.
- Published
- 2008
- Full Text
- View/download PDF
26. Cloning and characterization of the novel chimeric gene p53/FXR2 in the acute megakaryoblastic leukemia cell line CMK11-5.
- Author
-
Kanezaki R, Toki T, Xu G, Narayanan R, and Ito E
- Subjects
- Animals, Base Sequence, COS Cells, Cell Line, Tumor, Cell Transformation, Neoplastic, Chlorocebus aethiops, Cloning, Molecular, Cyclin-Dependent Kinase Inhibitor p21 metabolism, Etoposide pharmacology, Gene Expression Profiling, Humans, Mice, Molecular Sequence Data, NIH 3T3 Cells, Topoisomerase II Inhibitors, Genes, p53 genetics, Leukemia, Megakaryoblastic, Acute genetics, Mutant Chimeric Proteins genetics, RNA-Binding Proteins genetics
- Abstract
The loss of p53 function is a key event in tumorigenesis. Inactivation of p53 in primary tumors and cell lines is mediated by several molecular mechanisms, including deletions and rearrangements. However, generation of a p53 fusion gene has not yet been reported. Here we report a novel p53/an autosomal homolog of the fragile X mental retardation (FXR2) chimeric gene generated by an interstitial deletion. Western blot analyses have shown that the p53/FXR2 protein is indeed expressed in a Down syndrome-related acute megakaryoblastic leukemia cell line, CMK11-5 cells. To investigate the properties of the p53/FXR2 protein, we observed its subcellular localization. Flag-tagged expression vectors were transfected into COS-7 cells and the proteins were stained with an anti-Flag antibody. The p53/FXR2 protein was expressed at high levels in the cytoplasm, whereas wild-type p53 and FXR2 were localized primarily in the nucleus and in the periphery of the nucleus, respectively. Treatment with a topoisomerase II inhibitor, VP16, failed to induce expression of a p53 target gene, the cyclin-dependent kinase inhibitor p21(WAF-1/CIP1), in CMK11-5 cells, and transient transfection analysis showed that the p53/FXR2 protein failed to transactivate the p21(WAF-1/CIP1) promoter. These results suggest that the p53/FXR2 fusion protein lacks the ability of wild-type p53 to function as a transcription factor. The p53/FXR2 gene is the first reported p53 fusion gene.
- Published
- 2006
- Full Text
- View/download PDF
27. Transgenic expression of BACH1 transcription factor results in megakaryocytic impairment.
- Author
-
Toki T, Katsuoka F, Kanezaki R, Xu G, Kurotaki H, Sun J, Kamio T, Watanabe S, Tandai S, Terui K, Yagihashi S, Komatsu N, Igarashi K, Yamamoto M, and Ito E
- Subjects
- Animals, Basic-Leucine Zipper Transcription Factors, Cells, Cultured, DNA, Complementary, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Enhancer Elements, Genetic physiology, Erythroid-Specific DNA-Binding Factors, Fanconi Anemia Complementation Group Proteins, GATA1 Transcription Factor, Gene Expression Regulation, Humans, Megakaryocytes ultrastructure, Mice, Mice, Transgenic, Microscopy, Electron, NF-E2 Transcription Factor, p45 Subunit, Thrombocytopenia pathology, Megakaryocytes physiology, Thrombocytopenia physiopathology, Transcription Factors genetics, Transcription Factors metabolism
- Abstract
Both nuclear factor erythroid 2 45 kDa subunit (p45) and BTB and CNC homolog 1 (Bach) transcription factors can form dimers with one of the small Maf proteins, and these heterodimers bind to the musculoaponeurotic fibrosarcoma oncogene (Maf) recognition element (MARE). MARE is known to act as a critical cis-regulatory element of erythroid and megakaryocytic genes. Although detailed analyses of p45-null mutant mice and small maf compound mutant mice revealed that these factors are both critical for platelet production, the functional contributions of Bach1 and the relationship or redundancy between Bach1 and p45 in megakaryocytes remain to be clarified. To address these issues, we generated transgenic lines of mice bearing human BACH1 cDNA under the control of the GATA-1 locus hematopoietic regulatory domain. The transgenic mouse lines showed significant thrombocytopenia associated with impaired maturation of the megakaryocytes, and they developed myelofibrosis. The megakaryocytes in the transgenic mice exhibited reduced proplatelet formation, and the modal ploidy class of megakaryocytes was 2N, indicating the impairment of endomitosis. Transcription of the p45 target genes was down-regulated and we indeed found that BACH1 binds to the thromboxane synthase gene, one of the target genes for p45 in megakaryocytes. These findings thus provide evidence that BACH1 acts as a transcriptional repressor in the regulation of MARE-dependent genes in megakaryocytes.
- Published
- 2005
- Full Text
- View/download PDF
28. Three novel mutations of the fibrillin-1 gene and ten single nucleotide polymorphisms of the fibrillin-3 gene in Marfan syndrome patients.
- Author
-
Uyeda T, Takahashi T, Eto S, Sato T, Xu G, Kanezaki R, Toki T, Yonesaka S, and Ito E
- Subjects
- Asian People genetics, Base Sequence, DNA Mutational Analysis, DNA Primers, Fibrillin-1, Fibrillins, Heteroduplex Analysis, Humans, Molecular Sequence Data, Point Mutation, Marfan Syndrome genetics, Microfilament Proteins genetics, Mutation, Polymorphism, Single Nucleotide
- Abstract
Marfan syndrome (MFS) is an autosomal dominant disorder of the extracellular matrix. Allelic variations in the gene for fibrillin-1 ( FBN1) have been shown to cause MFS. To date, over 550 mutations have been identified in patients with MFS and related connective tissue diseases. However, about a half of MFS cases do not possess mutations in the FBN1 gene. These findings raise the possibility that variants located in other genes cause or modify MFS. To explore this possibility, firstly we analyzed FBN1 allelic variants in 12 Japanese patients with MFS, and secondly we analyzed fibrillin-3 gene ( FBN3) in patients without FBN1 mutations using conformation sensitive gel electrophoresis (CSGE) and direct sequencing analysis. We identified three novel FBN1 mutations and ten FBN3 single nucleotide polymorphisms (SNPs). In this report, we could not detect a responsible mutation of the FBN3 gene for MFS. Although the number of the cases in this report is small, at least these results suggest that disease-causing mutations in exon regions of the FBN3 gene are very rare in MFS.
- Published
- 2004
- Full Text
- View/download PDF
29. B-cell-specific transcription factor BACH2 modifies the cytotoxic effects of anticancer drugs.
- Author
-
Kamio T, Toki T, Kanezaki R, Sasaki S, Tandai S, Terui K, Ikebe D, Igarashi K, and Ito E
- Subjects
- Active Transport, Cell Nucleus drug effects, Antineoplastic Agents toxicity, Antioxidants pharmacology, B-Lymphocytes chemistry, Basic-Leucine Zipper Transcription Factors, Benzamides, Cell Death drug effects, Cell Line, Tumor, Gene Expression Regulation drug effects, Humans, Imatinib Mesylate, Oxidative Stress genetics, Piperazines pharmacology, Pyrimidines pharmacology, Reactive Oxygen Species analysis, Reactive Oxygen Species metabolism, Transcription Factors biosynthesis, Transcription Factors metabolism, Antineoplastic Agents pharmacology, Transcription Factors physiology
- Abstract
The transcription factor Bach2, a member of the CNC family of proteins, binds to the Maf recognition element (MARE) by forming homodimers or dimerizing with small Maf transcription factors. Bach2-expressing cells show reduced proliferation and undergo spontaneous cell death. The inhibition of BCR/ABL tyrosine kinase activity by STI571 in chronic myeloid leukemia (CML) cell lines and CD34+ cells from patients with CML in lymphoid crisis results in induction of BACH2 expression. We show here that BACH2 modifies the in vitro cytotoxicity of anticancer drugs. The cytotoxic effects of commonly used anticancer agents were studied by overexpression of BACH2 in RAJI lymphoid cells, a cell line that does not express endogenous BACH2. Cell growth inhibition was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT) assay. Clones overexpressing BACH2 were more sensitive to etoposide, doxorubicin, and cytarabine than control RAJI cells, whereas there were no significant differences in the sensitivity of either cells to methotrexate or vincristine. Interestingly, we found that the former drugs were oxidative stressors that induced the nuclear accumulation of BACH2. In contrast, methotrexate or vincristine did not induce production of intracellular reactive oxygen species (ROS) and nuclear accumulation of BACH2. These results, coupled with our previous data showing that BACH2 promotes oxidative stress-induced cell death, suggest that combination chemotherapy involving STI571 and anticancer drugs that produce ROS may be of benefit in the treatment of Philadelphia chromosome 1 (Ph1)-positive leukemia.
- Published
- 2003
- Full Text
- View/download PDF
30. Frequent mutations in the GATA-1 gene in the transient myeloproliferative disorder of Down syndrome.
- Author
-
Xu G, Nagano M, Kanezaki R, Toki T, Hayashi Y, Taketani T, Taki T, Mitui T, Koike K, Kato K, Imaizumi M, Sekine I, Ikeda Y, Hanada R, Sako M, Kudo K, Kojima S, Ohneda O, Yamamoto M, and Ito E
- Subjects
- Age Factors, Cell Differentiation, Cell Line, Cell Lineage, DNA, Complementary metabolism, Down Syndrome complications, Erythroid-Specific DNA-Binding Factors, Exons, Female, Flow Cytometry, GATA1 Transcription Factor, Genetic Vectors, Humans, Immunoblotting, Infant, Newborn, K562 Cells, Leukemia, Megakaryoblastic, Acute genetics, Male, Models, Genetic, Protein Structure, Tertiary, Retroviridae genetics, Reverse Transcriptase Polymerase Chain Reaction, Sequence Analysis, DNA, DNA-Binding Proteins genetics, Down Syndrome genetics, Mutation, Myeloproliferative Disorders genetics, Transcription Factors genetics
- Abstract
Transient myeloproliferative disorder (TMD) is a leukemoid reaction occurring occasionally in Down syndrome newborn infants. Acute megakaryocytic leukemia (AMKL) develops in approximately 20% to 30% of the cases with TMD. Recently, acquired mutations in the N-terminal activation domain of the GATA-1 gene, encoding the erythroid/megakaryocytic transcription factor GATA-1, have been reported in Down syndrome-related AMKL (DS-AMKL). To understand the multistep leukemogenesis in Down syndrome, GATA-1 mutations were investigated in patients with TMD. We show here that mutations in the GATA-1 gene were detected in 21 of 22 cases with TMD. Most of the mutations in TMD were located in the regions including exon 2 and were essentially identical to those observed in DS-AMKL. In the DS-AMKL cell line, MGS, which itself expresses only a truncated mutant of GATA-1, expression of full-length GATA-1 induced the differentiation toward the erythroid lineage. However, expression of the short form of GATA-1 did not induce erythroid differentiation. These results indicate that expression of GATA-1 with a defective N-terminal activation domain contributes to the expansion of TMD blast cells and that other genetic changes contribute to the development of AMKL in Down syndrome.
- Published
- 2003
- Full Text
- View/download PDF
31. Transcription factor BACH1 is recruited to the nucleus by its novel alternative spliced isoform.
- Author
-
Kanezaki R, Toki T, Yokoyama M, Yomogida K, Sugiyama K, Yamamoto M, Igarashi K, and Ito E
- Subjects
- Base Sequence, Basic-Leucine Zipper Transcription Factors, Binding Sites, Blotting, Northern, Cell Line, Cytoplasm metabolism, DNA, Complementary metabolism, Dimerization, Exons, Fanconi Anemia Complementation Group Proteins, Gene Library, Genes, Reporter, Green Fluorescent Proteins, Humans, Luminescent Proteins metabolism, Male, Microscopy, Fluorescence, Models, Genetic, Molecular Sequence Data, Plasmids metabolism, Precipitin Tests, Promoter Regions, Genetic, Protein Binding, Protein Isoforms, Protein Structure, Tertiary, Protein Transport, Recombinant Fusion Proteins metabolism, Reverse Transcriptase Polymerase Chain Reaction, Sequence Homology, Nucleic Acid, Testis metabolism, Tissue Distribution, Transcription Factors genetics, Transcription, Genetic, Transfection, Zinc Fingers, Alternative Splicing, Cell Nucleus metabolism, Transcription Factors chemistry, Transcription Factors metabolism
- Abstract
The transcription factor Bach1 is a member of a novel family of broad complex, tramtrack, bric-a-brac/poxvirus and zinc finger (BTB/POZ) basic region leucine zipper factors. Bach1 forms a heterodimer with MafK, a member of the small Maf protein family (MafF, MafG, and MafK), which recognizes the NF-E2/Maf recognition element, a cis-regulatory motif containing a 12-O-tetradecanoylphorbol-13-acetate-responsive element. Here we describe the gene structure of human BACH1, including a newly identified promoter and an alternatively RNA-spliced truncated form of BACH1, designated BACH1t, abundantly transcribed in human testis. The alternate splicing originated from the usage of a novel exon located 5.6 kilobase pairs downstream of the exon encoding the leucine zipper domain, and produced a protein that contained the conserved BTB/POZ, Cap'n collar, and basic region domains, but lacked the leucine zipper domain essential for NF-E2/Maf recognition element binding. Subcellular localization studies using green fluorescent protein as a reporter showed that full-length BACH1 localized to the cytoplasm, whereas BACH1t accumulated in the nucleus. Interestingly, coexpression of BACH1 and BACH1t demonstrated interaction between the molecules and the induction of nuclear import of BACH1. These results suggested that BACH1t recruits BACH1 to the nucleus through BTB domain-mediated interaction.
- Published
- 2001
- Full Text
- View/download PDF
32. Cloning and expression of human B cell-specific transcription factor BACH2 mapped to chromosome 6q15.
- Author
-
Sasaki S, Ito E, Toki T, Maekawa T, Kanezaki R, Umenai T, Muto A, Nagai H, Kinoshita T, Yamamoto M, Inazawa J, Taketo MM, Nakahata T, Igarashi K, and Yokoyama M
- Subjects
- Adult, Amino Acid Sequence, Animals, B-Lymphocytes cytology, Base Sequence, Basic-Leucine Zipper Transcription Factors, Cells, Cultured, Chromosome Mapping, Cloning, Molecular, DNA, Complementary, Gene Expression, Gene Frequency, Humans, K562 Cells, Loss of Heterozygosity, Lymphoma, B-Cell pathology, Lymphoma, Non-Hodgkin pathology, Mice, Molecular Sequence Data, Tumor Cells, Cultured, B-Lymphocytes metabolism, Chromosomes, Human, Pair 6, Leucine Zippers, Lymphoma, B-Cell genetics, Lymphoma, Non-Hodgkin genetics, Transcription Factors genetics
- Abstract
The transcription factor Bach2, a member of the BTB-basic region leucine zipper (bZip) factor family, binds to a 12-O-tetradecanoylphorbol-13-acetate (TPA)-responsive element and the related Maf-recognition element (MARE) by forming homodimers or heterodimers with Maf-related transcription factors. Bach2 regulates transcription by binding to these elements. To understand the function in hematopoiesis, we isolated a cDNA clone for human Bach2 (BACH2) encoding a protein of 841 amino acid residues with a deduced amino acid sequence having 89.5% identity to mouse homolog. Among human hematopoietic cell lines, BACH2 is expressed abundantly only in some B-lymphocytic cell lines. RT-PCR analysis of hematopoietic cells revealed that BACH2 mRNA is expressed in primary B-cells. Enforced expression of BACH2 in a human Burkitt cell line, RAJI that does not express endogenous BACH2, resulted in marked reduction of clonogenic activity, indicating that BACH2 possesses an inhibitory effect on cell proliferation. By fluorescent in situ hybridization, the BACH2 gene was localized to chromosome 6q15. Because deletion of the long arm of chromosome 6 (6q) is one of the commonest chromosomal alterations in human B-cell lymphoma, we examined for the loss of heterozygosity (LOH) of the BACH2 gene in human B-cell non-Hodgkin's lymphomas (NHL). Among 25 informative cases, five (20%) showed LOH. These results indicate that BACH2 plays important roles in regulation of B cell development.
- Published
- 2000
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.