GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
Filter
  • American Society of Hematology  (18)
  • Inaba, Toshiya  (18)
Material
Publisher
  • American Society of Hematology  (18)
Language
Subjects(RVK)
  • 1
    Online Resource
    Online Resource
    American Society of Hematology ; 2012
    In:  Blood Vol. 120, No. 21 ( 2012-11-16), p. 1297-1297
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 1297-1297
    Abstract: Abstract 1297 Centrosomes acquire dense microtubule nucleation sites in the beginning of mitosis. Failure of this process (called centrosome maturation) impairs the function of mitotic centrosomes to create robust mitotic spindles, resulting in lagging and scattered chromosomes that subsequently cause abnormal nuclear morphology such as bi- tri- or multiple-nuclei with or without small nucleus, as seen routinely in MDS. We previously reported that the Miki (LOC253012) gene, located in 7q21.3, is frequently deleted in MDS patients, and that low levels of Miki are tightly associated with abnormal mitosis and nuclear morphology (BBRC 2009). Here we demonstrate that Miki plays critical roles in the formation of robust spindles required for the prompt movement of chromosomes in a poly(ADP) ribosylation (PARsylation)-dependent manner (a part of data was published in Mol. Cell 2012). While Miki was localized in the Golgi apparatus during interphase, it was relocated to centrosomes at the beggining of mitosis. Treatment of cells with Miki-specific siRNA induced ‘pseudometaphase’ condition, in which lagging chromosomes juxtaposed to, or even situated behind, spindle poles. Pseudometaphase was followed by apoptosis or abnormal exit from mitosis that creates cells with abnormal nuclear morphology. This phenotype of Miki-downregulation was caused by the reduced robustness of mitotic spindles. α -tubulin staining of siRNA-treated cells revealed curling and disorganized spindles, with an occasional chaotic centrosome at only one side of the alignment. In addition, Miki-downregulation reduced γ-tubulin signals in mitotic centrosomes and markedly inhibited microtubule nucleation, shown by the impaired accumulation of the EB1 microtubule tip-binding protein at centrosomes. In immnunoblot analysis of lysate extracted from isolated spindles/centrosomes using Miki antibody, we detected a dense 125 kDa band in addition to the expected 50 kDa band and found that the 125 kDa band represents PARsylated Miki. A recent report indicated that tankyrase-1, a PAR polymerase (PARP), is required for the progression of prometaphase. We found that the downregulation of tankyrase-1 prevents Miki from localizing to mitotic centrosomes. In addition, immunoblot analysis of immunoprecipitation revealed that Miki is a substrate for tankyrase-1. These data suggested that tankyrase-1 PARsylates and translocates Miki from the Golgi apparatus to mitotic centrosomes/spindles during the short period from late G2 to prophase. We also found that PARsylated Miki promotes CG-NAP, a major component of microtubule nucleation sites, to concentrate in mitotic centrosomes. Interestingly, the CG-NAP gene resides 1.2Mb centromeric to Miki in band 7q21, and approximately 20% of MDS patients lose one allele of both Miki and CG-NAP genes. This indicates that loss of 7q results in low expression of two crucial factors in the tankyrase-1/Miki-dependent system for centrosome maturation, and that this may cause miotic/nuclear abnormalities and chromosome instability characteristic of 7q- MDS. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2012
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 108, No. 11 ( 2006-11-16), p. 390-390
    Abstract: To isolate myeloid tumor suppressor genes from 7q, we tried to detect microdeletions ( 〈 100 Kb) that might be present in MDS/AML cells carrying apparently normal chromosome 7. For this purpose, we developed our original microarray-based CGH technology. In this system, instead of BAC clones generally used as probes, we applied short (3–5 Kb) genomic DNA fragments containing strictly no repetitive sequences. We made 235 probes in a region spanning 21.7 Mb within 7q21.3–7q31.1. Although we selected MDS/AML patients whose marrow did not show cytogenetically visible 7q deletions, gross copy number changes frequently observed in adult patients prevented us from identification of common microdeletions. By investigation of 21 childhood myeloid leukemia patients with normal karyotype, we successfully identified a common microdeletion spanning approximately 120 Kb. Eight (38%) patients shared this microdeletion, which was not detected in normal individuals. Database search revealed that this region contains three hypothetical genes. Only vertebrates have these genes that likely evolved from one common ancestral gene of fish. Real-time quantitative PCR revealed that 9 (29%) out of 31 adult MDS/AML harbors microdeletions in at least one of these three genes. None of these genes had been well characterized nor has known motifs that would suggest function of the gene products. We named them Miki, Titan and Kasumi. Immunoblot analysis revealed expression of all three genes at high levels in most lymphoid leukemia cell lines, while half of myeloid cell lines lacked at least one of their expression. In leukemia cells carrying monosomy 7, expression levels were generally low. Miki, a heavily glycosylated protein, co-localized with centrosomes and spindles in the mitotic phase. To test the function of Miki, we used si-RNAs to downregulate Miki expression in HeLa and K562 cells, both of which show basically normal metaphase and nuclear morphology. Cells expressing Miki at reduced levels showed small and fragmented centrosomes, loss of spindle tension, tripolar mitosis or even completely disturbed spindle formation. As a result, anaphase lagging, colchicine-mitosis (C-mitosis), premature chromosome decondensation and chromatid bridges were observed in virtually all cells in the mitotic phase. In the interphase, bi- or tri-nuclear or even multinuclear cells with micronuclei, all of which are characteristic to MDS, were frequently observed. On the other hand, proteomic analysis revealed that Titan and Kasumi bind to the DNA-PK complex, which plays critical roles in the non-homologous end joining (NHEJ) of double stranded DNA (dsDNA) breaks. Indeed, these proteins were translocated from cytoplasm to nucleus by ionizing radiation (IR) or by treatment of drugs that yield dsDNA breaks. Cells expressing Kasumi at reduced levels by si-RNA showed increased radiosensitivity, sister chromatid exchange, and number of background-level phosphorylated histone H2AX foci (i.e., foci formed without IR), which are co-localized with dsDNA breaks. These results indicate that the genes we isolated are promising candidates for anti-leukemic genes located in 7q, because downregulation of these gene products by 7q deletions would cause the abnormal morphology of MDS and genetic instability.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2006
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Society of Hematology ; 2018
    In:  Blood Vol. 131, No. 26 ( 2018-06-28), p. 2891-2898
    In: Blood, American Society of Hematology, Vol. 131, No. 26 ( 2018-06-28), p. 2891-2898
    Abstract: Since a report of some 50 years ago describing refractory anemia associated with group C monosomy, monosomy 7 (−7) and interstitial deletions of chromosome 7 (del(7q)) have been established as one of the most frequent chromosomal aberrations found in essentially all types of myeloid tumors regardless of patient age and disease etiology. In the last century, researchers sought recessive myeloid tumor-suppressor genes by attempting to determine commonly deleted regions (CDRs) in del(7q) patients. However, these efforts were not successful. Today, tumor suppressors located in 7q are believed to act in a haploinsufficient fashion, and powerful new technologies such as microarray comparative genomic hybridization and high-throughput sequencing allow comprehensive searches throughout the genes encoded on 7q. Among those proposed as promising candidates, 4 have been validated by gene targeting in mouse models. SAMD9 (sterile α motif domain 9) and SAMD9L (SAMD9-like) encode related endosomal proteins, mutations of which cause hereditary diseases with strong propensity to infantile myelodysplastic syndrome (MDS) harboring monosomy 7. Because MDS develops in SAMD9L-deficient mice over their lifetime, SAMD9/SAMD9L are likely responsible for sporadic MDS with −7/del(7q) as the sole anomaly. EZH2 (enhancer of zeste homolog 2) and MLL3 (mixed lineage leukemia 3) encode histone-modifying enzymes; loss-of-function mutations of these are detected in some myeloid tumors at high frequencies. In contrast to SAMD9/SAMD9L, loss of EZH2 or MLL3 likely contributes to myeloid tumorigenesis in cooperation with additional specific gene alterations such as of TET2 or genes involved in the p53/Ras pathway, respectively. Distinctive roles with different significance of the loss of multiple responsible genes render the complex nature of myeloid tumors carrying −7/del(7q).
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2018
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Society of Hematology ; 2008
    In:  Blood Vol. 112, No. 11 ( 2008-11-16), p. 1793-1793
    In: Blood, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. 1793-1793
    Abstract: We previously reported a candidate myeloid tumor-suppressor gene Miki (mitotic kinetic regulator, LOC253012) isolated from a common microdeletion cluster in chromosome subband 7q21.2 that was identified by microarray-based CGH analyses of JMML (ASH Annual Meeting, 2006 and 2007). Deletion of one Miki gene was also detected in 28 % of adult MDS/AML patients by copy-number assessment using real-time quantitative PCR (qPCR). Miki encodes a centrosomal protein: downregulation of Miki by siRNA disturbs the maturation and positioning of centrosomes, as well as spindle formation in mitotic cells, resulting in severe mitotic defects, such as chromosome lagging and proanaphase arrest. This causes abnormal nuclear morphology. When Miki expression levels were constitutively reduced by short-hairpin (sh) RNA in K562 cells, morphology of cells changed drastically: bi- tri- or multiple-nuclear cells with or without micronuclei appeared frequently that strikingly resembled the bone marrow picture of MDS. In addition, FISH analysis revealed widely varying chromosome numbers in these cells, suggesting that Miki-downregulation induces chromosome instability. Six myeloid leukemia cell lines derived from MDS/AML patients with monosomy 7 generally expressed Miki protein at very low (but detectable) levels, suggesting that haploinsufficient effects and/or epigenetic mechanisms reduce Miki expression levels. These six lines harboring monosomy 7 showed severe abnormal mitosis and nuclear morphology, and induction of Miki to these cells using a retroviral vector restored normal mitosis. These findings suggested that loss of Miki gene contributes to myelodysplasia and chromosome instability, which are characteristic of -7/7q- MDS/AML. To elucidate molecular mechanism through which Miki plays roles in centrosomal maturation and spindle formation, we tried to identify proteins associated with Miki using yeast two hybrid assay or mass spectrometry and found that a centrosomal giant scaffold protein CG-NAP (also known as AKAP9/AKAP450/yotiao) binds to Miki in a G2/M specific manner. Intriguingly, CG-NAP gene locates to subband 7q21.2, 1.2 Mb centromeric to Miki gene. qPCR revealed that CG-NAP was also frequently deleted in adult MDS/AML (36 %) and protein expression levels were very low (but detectable) in six cell lines derived from MDS/AML with monosomy 7, raising a possibility that CG-NAP is another candidate for the responsible genes of 7q deletion. CG-NAP is known to promote microtubule nucleation in centrosomes. As expected, downregulation of CG-NAP by siRNA showed abnormal spindle formation and mitotic disturbance (proanaphase arrest and chromosome lagging) similar to those by Miki-downregulation. Moreover, constitutive downregulation of CG-NAP by shRNA transformed K562 cells to bi- tri- or multiple-nuclear with or without micronuclei, indicating that the Miki/CG-NAP protein complex is responsible for the mitotic disturbance and abnormal nuclear morphology. Finally, we found that CG-NAP does not localize to mitotic centrosomes when Miki expression is downregulated by siRNA, suggesting that Miki contributes to organized progression of mitosis by transporting and/or anchoring CG-NAP to mitotic centrosomes. Our data indicate that Miki and CG-NAP in subband 7q21.2 encode centrosomal proteins, which play critical roles in mitosis. Loss of one 7q allele would cause marked reduction of these two gene products, resulting in myelodysplasia and chromosome instability.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2008
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 106, No. 11 ( 2005-11-16), p. 2988-2988
    Abstract: The E2A-HLF fusion transcription factor generated by the t(17;19)(q22;p13) translocation is found in a small population of pro-B cell ALL. Patients associated with this chimera share distinct clinical features such as hypercalcemia, coagulopathy and very poor prognosis due to resistance to intensive chemotherapy including aggressive conditioning for BMT, all of which are unusual for this type of ALL. We have previously demonstrated that inhibition of the trans-activation potential of the E2A-HLF chimera by the dominant negative mutant results in apoptosis in t(17;19)+ ALL cells but does not affect cell cycle. Moreover, E2A-HLF blocks apoptosis induced by cytokine deprivation in IL-3-dependent cells, suggesting that this fusion protein contributes to leukemogenesis by substituting for the anti-apoptotic function of cytokines. The present study shows that survivin is a downstream target molecule of E2A-HLF. Four t(17;19)+ ALL cell lines expressed survivin at high levels and down-regulation of E2A-HLF function by the dominant negative mutant suppressed survivin expression. In addition, forced expression of E2A-HLF in Nalm-6, a t(17;19)− ALL cell line, up-regulated survivin expression. Survivin is known to be expressed predominantly in the G2/M phase. Indeed, separation of the fractions enriched for in each phase of the cell cycle using a counterflow centrifugal elutriator revealed G2/M phase-dominant survivin expression in t(17;19) − ALL cells including Nalm-6. In t(17;19)+ ALL cells, however, survivin was expressed throughout the cell cycle. Moreover, Nalm-6 cells forced to express E2A-HLF showed cell cycle-independent survivin expression. Reporter assay revealed that E2A-HLF induced luciferase activity by transfecting with each reporter construct containing the survivin promoter at a different length from the initial ATG, suggesting that E2A-HLF induces survivin expression at the transcriptional level, but not by direct binding of E2A-HLF to the survivin promoter. To test whether survivin plays anti-apoptotic roles in t(17:19)+ cells, we used a survivin mutant lacking a phosphorylation site (T34A-survivin) and considered to inhibit survivin function in a dominant negative manner. T34A-survivin induced massive apoptosis throughout the cell cycle in t(17;19)+ cells. In contrast, T34A-survivin in t(17;19) − cells induced cell death in only a small population in G2/M phase. In addition to caspase-dependent pathways, T34A-survivin induced apoptosis in t(17;19)+ ALL cells through caspase-independent pathways, in which apoptosis-inducing factor (AIF) translocated from cytoplasm to the nucleus. These results indicate that cell cycle-independent up-regulation of survivin by the E2A-HLF chimera is indispensable for the survival of t(17;19)+ ALL cells, and that inhibition of survivin may offer an effective therapeutic strategy against this refractory ALL.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2005
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 1081-1081
    Abstract: Abstract 1081 α IIbβ3 integrin mutations that result in the complete loss of expression of this molecule on the platelet surface cause Glanzmann's thrombasthenia. This is usually an autosomal recessive, while other mutations are known to cause dominantly inherited macrothrombocytopenia (although such cases are rare). Here, we report a 4-generation pedigree including 10 individuals affected by dominantly inherited macrothrombocytopenia. Six individuals, whose detailed clinical and laboratory data are available, carry a non-synonymous ITGB3gene T2231C alteration resulting in the substitution of leucine at 718 for proline (L718P) in the integrin β3 protein. The patient was 4-year old Japanese girl, who presented with mild bleeding tendency. Her platelet count was 49–72 x109/L with a mean platelet volume of 6.7–10.4 fl. WBC and RBC numbers were normal and there were no morphological abnormalities including inclusions in neutrophils. In the patient, marked platelet anisocytosis and giant platelets were observed in peripheral blood samples. Platelet aggregation induced by ADP and collagen was markedly reduced, but agglutination induced by ristocetin was within the normal range. The α IIbβ3 expression level determined by flow cytometry in the patient was 50–60% of the healthy control. A total of six of her relatives were subsequently found to have low platelet counts and were referred to our institute for further investigation. To isolate a candidate gene alteration responsible for the macrothrombocytopenia, whole exome sequencing analysis was performed using genomic DNA obtained from four affected individuals of the pedigree including the patient. Among the 90 non-synonymous alterations commonly found in the affected individuals, we focused on the heterozygous integrin β3-L718P mutation, because this was recently reported as a candidate mutation responsible for macrothrombocytopenia (Jayo et al, 2010). As far as we could determine, no other non-synonymous gene alterations previously reported to cause thrombocytopenia or defective platelet function were present in the affected individuals of the pedigree. Resting platelets from affected individuals showed a mild but significant increase of a ligand-mimicking PAC-1 binding relative to healthy individuals. However, in ADP-treated platelets carrying the mutation, only a small increase of affinity to PAC-1 was observed. These findings suggest that α IIbβ3-L718P is partially activated in the absence of inside-out signals such as ADP, but nevertheless cannot be fully activated in the presence of stimulating signals. As previously reported by others, CHO cells expressing α IIbβ3-L718P formed long proplatelet-like protrusions on fibrinogen-coated dishes. This was reported to be mediated by the downregulation of RhoA activity, which is initiated by the binding of c-Src to the C-terminal tail of integrin β3. Indeed, we found that the formation of long cell protrusion was inhibited, when a constitutively-active form of RhoA (Q63L) was introduced into α IIbβ3-L718P-expressing cells. In addition, CHO cells expressing α IIbβ3-L718P (del. 759) mutant, which lacks the C-terminal c-Src binding site of integrin β3, did not form any proplatelet-like protrusions. However, because the enforced expression of a dominant negative form of RhoA (T19N) in α IIbβ3-WT expressing cells did not show typical proplatelet-like protrusions, it is suggested that downregulation of RhoA was required but not sufficient for the formation of proplatelet-like protrusions induced by integrin β3-L718P. In summary, identification of a pedigree showing autosomal dominant inheritance leads to a model whereby the integrin β3-L718P mutation contributes to macrothrombocytopenia most likely through gain-of-function mechanisms. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2012
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 227-227
    Abstract: Monosomy 7 is a common chromosomal abnormality found frequently in MDS and AML. We previously identified a common microdeletion cluster in 7q21.3 in juvenile myelomonocytic leukemia patients. This cluster contains three poorly characterized genes: sterile alpha motif (SAM) domain-9 (SAMD9) which is absent in mice, samd9-like (SAMD9L) and Miki (LOC253012). Although mutations have rarely been seen in these genes, the genes reside proximal to the 7q22 band that is deleted in single copy in nearly 25% of AML and MDS patients. We recently reported that Miki, a centrosomal protein that promotes alignment of chromosomes at metaphase, is a candidate gene responsible for mitotic/nuclear abnormalities observed in MDS patients (Mol Cell 2012). In this study, we established and characterized SAMD9L-deficient mice, along with the analysis of molecular function of SAMD9L protein. Among SAMD9L-/- (n=15) and SAMD9L+/- (n=15) littermates, 13 mice developed myeloid dysplasia, 2 mice developed myeloid leukemia and one mouse developed myeloproliferative disease after the age of 18 months, while all but one SAMD9L+/+ mice (n=23) maintained normal hematopoiesis throughout the 24-month observation period. Infection of MOL4070A retrovirus into newborn mice developed myeloid leukemia within 15 months preferentially in SAMD9L-deficient genetic background with Evi1 and Fbxl10 (encoding a H3K36 demethylase) genes as common virus integration sites. While bone marrow (BM) cells from SAMD9L+/+ mice (12 weeks old) formed fewer colonies by the third replating, cells from SAMD9L-deficient mice continued to form similar numbers and sizes of well-differentiated colonies beyond the 7th plating. The excess number of colonies formed was reduced by retrovirus-mediated forced expression of Samd9L. These data suggested enhanced self-renewal and/or delays in differentiation of SAMD9L-deficient stem cells. In addition, enhanced reconstitution ability of SAMD9L-deficient stem cells was demonstrated by competitive repopulation assay using the Ly5 congenic mouse system, where irradiated Ly5.1 mice were transplanted with long term-LSK cells from SAMD9L+/+ or SAMD9L-deficient Ly5.2 mice (10 weeks old) together with BM cells from SAMD9L+/+Ly5.1 mice. This was confirmed by limiting-dilution transplants, results of which showed a higher frequency of multi-lineage repopulating cells at 8 weeks in SAMD9L-/- donor BM. Moreover, growth advantage in the presence of cytokines was evident in liquid cultures of SAMD9L-deficient BM progenitor cells. Hypersensitivity of SAMD9L-deficient BM progenitors to cytokines was also shown in in vivo experiments, in which SAMD9L-deficient mice injected with cyclophosphamide (day 0) and G-CSF (days 1-4) showed significantly higher WBC counts than SAMD9L+/+ mice at the nadir (day 3). These findings suggested that SAMD9L-deficiency sensitizes hematopoietic progenitors to cytokines. Immunostaining using SAMD9L antibody showed a vesicular pattern of SAMD9L localization in approximately 15% of BM progenitor cells that overlapped with the localization of EEA1, an early endosomal protein. In SAMD9L-/- fibroblasts, while rapid endocytosis of PDGF-receptor (PDGFR) by PDGF stimulation occurred in a time-course similar to that in SAMD9L+/+ cells, homotypic fusion of endosomes containing PDGFR delayed. Inhibition of endosome fusion in SAMD9L-/- cells lead to the accumulation of PDGFR that were remained to be phosphorylated in early endosome, resulting in the prolonged activation of cytokine signals. Accumulation of cytokine receptors in early endosome and persistent cytokine signals were also found in BM progenitors obtained from SAMD9L-deficient mice. These observations suggest that SAMD9L is a crucial component of a protein complex that facilitates the degradation of cytokine receptors through the homotypic fusion of endosomes. Collectively, our study suggests the contribution of haploinsufficiency of SAMD9L to the pathogenesis of myeloid diseases harboring -7/7q- through the prolonged activation of cytokine signals that results in the enhancement of stem cell self-renewal and/or delay in differentiation of early progenitors. Deletion of other haploinsufficient tumor-suppressor genes that reside in 7q would collaborate with the deficiency of SAMD9L for myeloid leukemogenesis. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 2963-2963
    Abstract: Abstract 2963 Poster Board II-939 From a common microdeletion cluster located in chromosome subband 7q21.3, we identified three candidate responsible genes (Kasumi=Samd9, Titan=Samd9L and Miki=LOC253012) which encode myeloid-tumor suppressors (BBRC 2009). As we presented previously (ASH annual meeting, 2008), Miki encodes a centrosomal protein and is involved in myelodysplasia and chromosomal instability. On the other hand, Kasumi and Titan, that encode 60% identical proteins, are poorly characterized. Recent reports revealed that bi-allelic point mutations in Kasumi gene cause a rare fatal skin disease, Normophosphatemic Familial Tumoral Calcinosis (NFTC). To identify the contributions of these genes to leukemogenesis, we initially generated Titan deficient mice (mouse genome contains only Titan and lacks Kasumi gene). Although heterozygous (titan+/−) and homozygous (titan-/-) mice were born and grown normally without hematological abnormalities, they naturally developed AML at high frequency after they reached 20 months old. This unusually long latency suggests that additional gene alterations are required for leukemia development. Thus we attempted to accelerate the onset of leukemia by retroviral insertional mutagenesis. Virus infection induced various myeloid leukemias after 10 to 12 months in almost all titan+/− and titan-/- mice. Inverse PCR detected two common virus integration sites specific for titan+/− and titan-/- mice, which induced deregulated expression of a zinc finger transcription factor, Evi1, and a histone H3K36 demethylase, Fbxl10. We next performed mouse BMT using titan-/- and +/+ bone marrow cells transduced with Evi1 retrovirally. Mice transplanted with titan(+/+)/Evi1 overexpressing cells developed MDS or AML after 7 months. By contrast, most mice transplanted with titan(-/-)/Evi1 overexpressing cells developed AML within 6 months after BMT, confirming co-operation between loss of Titan and Evi1 overexpression in myeloid leukemogenesis. Both Evi1 and Fbxl10 are reported to downregulate p15Ink4b tumor suppressor gene. Moreover, in human secondary MDS, DNA methylation in the promoter region of p15Ink4b is closely associated with 7q deletion. Thus we compared Fbxl10 and p15Ink4b expressions between AML/MDS samples with or without 7q deletion. We found that 7q deletion was correlated with higher Fbxl10 and with lower p15Ink4b levels, suggesting that silencing of p15ink4b through transcriptional and epigenetical mechanisms would be involved in leukemia with 7q deletion. To elucidate the function of Kasumi and Titan, we firstly immunoprecipitated Titan binding proteins from FLAG-Titan expressing cells and identified two specific bands around 150 and 70KDa. Mass spectrometry analysis showed that they correspond to Flightless1 (Fli1) and Scinderin (Scin), respectively. Because both Fli1 and Scin belong to the gelsolin superfamily proteins that bind to and sever actin filaments, we speculated that Titan is involved in cell movement via actin remodeling. To analyze this, we established Titan-knockdown (K/D) mouse fibloblasts by introducing Titan-specific shRNA-expressing vectors and observed their migration under time-lapse microscopy. In wound-healing assay, Titan-K/D cells migrated slower towards wound edge with loss of polarity. Each cell moved restlessly by quickly changing the direction. In these cells, lamellipodial protrusions rapidly formed and retracted. In accordance with this phenotype, activity of Rac1, a Rho GTPase, increased in Titan-K/D cells. This abnormality in cell migration is likely involved in pathogenesis of NFTC, i.e., severe inflammations in skin and mucosae. Moreover, since recent reports revealed the contribution of hyperactivated Rac1 to transformation of hematopoietic stem cells through abnormal actin remodeling, our findings suggest that deletion of Kasumi or Titan is a cue to cause AML/MDS through aberrant Rac1 activation. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2009
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 110, No. 11 ( 2007-11-16), p. 2638-2638
    Abstract: To isolate myeloid tumor suppressor gene(s) in 7q, we searched microdeletions in a region spanning 21.7 Mb within 7q21.2–7q31.1 using a microarray-based CGH system. By investigation of 21 childhood myeloid leukemia patients with normal karyotype, we identified a common microdeletion cluster spanning approximately 120 Kb in the 7q21.3 subband. Eight (38%) patients shared this microdeletion, which was not detected in normal individuals. Real-time quantitative PCR revealed that this region is also deleted in 9 (29%) out of 31 adult RAEB and AML patients. Database search revealed that this region contains three hypothetical genes. Among them, we chose one previously uncharacterized gene for further investigation and named Miki (mitotic kinetics regulator) for the function of its gene product, described below. Immunoblot analysis revealed high levels of Miki expression in most lymphoid leukemia cell lines, while half of myeloid leukemia cell lines expressed Miki at reduced levels. In six leukemia lines carrying monosomy 7, expression levels were generally low. Miki co-localized with the Golgi apparatus in the interphase and with centrosomes and spindles in the mitotic phase. To test the function of Miki, we used si-RNAs to downregulate Miki expression in HeLa and K562 cells, both of which show basically normal metaphase and nuclear morphology. Cells expressing Miki at reduced levels showed insufficient maturation and disturbed positioning of centrosomes, resulting in unorganized spindles including loss of spindle tension, curled and fragile spindles, or even completely disturbed spindle formation. Time-lapse observation revealed prometaphase and/or metaphase delay with unaligned or even totally scattered chromosomes in prometaphase in virtually all cells in the mitotic phase. As a result, cells underwent pre-anaphase arrest and exited mitosis in the absence of chromosome segregation or terminated mitosis by cell death. In the interphase, there were many cells with chromatid bridges and/or bi- or tri-nuclear or even multinuclear cells with micronuclei that resembled pathological cells routinely observed in the bone marrow pictures of MDS patients. Interestingly, myeloid cell lines with low Miki expression, including those with monosomy 7, generally showed abnormal mitosis such as scattered chromosomes and abnormal nuclear morphologies (multi-nuclear cells with small nuclei) at higher frequency than cell lines expressing Miki at high levels. Moreover, induction of Miki restores normal mitosis in leukemia cells with monosomy 7. Miki was poly(ADP-ribosyl)ated (PARsylated) in late G2 to M phase by tankyrase-1, one of PAR polymerase (PARP), and tankyrase-1 activity was required for the binding of Miki to spindles and centrosomes. These data suggest that loss of Miki gene contributes to the development and progression of MDS by disturbing mitosis.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2007
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood Advances, American Society of Hematology, Vol. 4, No. 5 ( 2020-03-10), p. 845-854
    Abstract: Loss-of-function mutations in ten-eleven translocation-2 (TET2) are recurrent events in acute myeloid leukemia (AML) as well as in preleukemic hematopoietic stem cells (HSCs) of age-related clonal hematopoiesis. TET3 mutations are infrequent in AML, but the level of TET3 expression in HSCs has been found to decline with age. We examined the impact of gradual decrease of TET function in AML development by generating mice with Tet deficiency at various degrees. Tet2f/f and Tet3f/f mice were crossed with mice expressing Mx1-Cre to generate Tet2f/wtTet3f/fMx-Cre+ (T2ΔT3), Tet2f/fTet3f/wtMx-Cre+ (ΔT2T3), and Tet2f/fTet3f/fMx-Cre+ (ΔT2ΔT3) mice. All ΔT2ΔT3 mice died of aggressive AML at a median survival of 10.7 weeks. By comparison, T2ΔT3 and ΔT2T3 mice developed AML at longer latencies, with a median survival of ∼27 weeks. Remarkably, all 9 T2ΔT3 and 8 ΔT2T3 mice with AML showed inactivation of the remaining nontargeted Tet2 or Tet3 allele, respectively, owing to exonic loss in either gene or stop-gain mutations in Tet3. Recurrent mutations other than Tet3 were not noted in any mice by whole-exome sequencing. Spontaneous inactivation of residual Tet2 or Tet3 alleles is a recurrent genetic event during the development of AML with Tet insufficiency.
    Type of Medium: Online Resource
    ISSN: 2473-9529 , 2473-9537
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2020
    detail.hit.zdb_id: 2876449-3
    detail.hit.zdb_id: 2915908-8
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...