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  (33)
  • 1
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 110, No. 11 ( 2007-11-16), p. 2362-2362
    Abstract: Rituximab has greatly improved the prognosis of B cell malignancies (BCM). However, several resistance mechanisms have been reported, including the inhibition of apoptosis, complement-dependent cytotoxicity (CDC) and antibody-dependent cellular cytotoxicity (ADCC) as well as modulation of CD20 antigen and increased complement-related cytotoxicity inhibition factor (CD55). The calicheamicin-conjugated anti-CD22 monoclonal antibody (mAb), CMC544, is one of the new promising agents, and effective on several types of BCM in vitro, especially when used in combination with rituximab. However, details regarding the cytotoxic effects of CMC544 and mechanisms triggering cell death, particularly when it is combined with rituximab, have not been elucidated. We studied cell cycle features of CMC544 on BCM used alone or in combination with rituximab, and analyzed quantitative alteration of target molecules such as CD20, CD22 and CD55.(Materials and Methods) The cell lines used in this study were CD22-positive Daudi and Raji cells, and CD22-negative Jurkat and NB4 cells. Cells obtained from 8 patients with BCM were also used. CMC544, unconjugated anti-CD22 mAb (G5/44), and free NAC-calicheamicin DMH were provided by Wyeth Pharmaceutical Co., Ltd. The effects of CMC544 were analyzed by incubating the cells at the concentrations of 0–100μg/ml for 0–96h, then by measuring cell count, cell viability, 3H-thymidine incorporation and cell cycle distribution on flow cytometry (FCM) as well as by video microscopic observation. The amounts of CD20, CD22 and CD55 antigens were analyzed by FCM, laser scanning microscope. The process of CMC544-induced cell death was assessed by Apocyto®. Three possible mechanisms of the combined effect of CMC544 and rituximab were investigated separately by direct effect of the mAb, CDC and ADCC. (Results) Analysis of the cell cycle distribution indicated that CD22-positive cells were temporally arrested at the G2/M phase after 6–12h incubation with CMC544, and the hypodiploid proportion increased after 24–72h. CD22-positive cells enlarged after 12–24h incubation with CMC544. Apoptotic morphological changes such as bleb formation and cell shrinkage were observed in 35–57% of the cells. These changes were not observed after incubation with G5/44. Apocyto® stain showed a continuous pattern from early to late apoptosis. Similar effects were observed in clinical samples that expressed CD22. The amounts of CD22 and CD55 were significantly reduced 3h after incubation with CMC544, while the CD20 expression was maintained 24h after incubation with CMC544. Incubation of the cells for 12–24h with CMC544 and then for 12hr with rituximab induced 1.2–2.1 times more anti-proliferation, apoptosis, CDC and ADCC than simultaneous incubation.(Conclusion) The effect of CMC544 was clearly observed as G2/M arrest following increase of the hypodiploid proportion in the cell cycle distribution, and this was a useful method to investigate the effect of CMC544. Combination of CMC544 and rituximab enhanced the cytotoxic effect, and sequential administration was more effective. The reduction of CD55 and continued expression of CD20 after the incubation with CMC544 supported the above observation. Further investigations are required to find the most effective treatment regimen to overcome drug resistance of B cell malignancies.
    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 ...
  • 3
    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 ...
  • 4
    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 ...
  • 5
    In: Blood, American Society of Hematology, Vol. 121, No. 17 ( 2013-04-25), p. 3434-3446
    Abstract: BMI1 overexpression is one of the second hit partner genes of RUNX1 mutations that contribute to the development of MDSs.
    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 ...
  • 6
    In: Blood, American Society of Hematology, Vol. 132, No. 23 ( 2018-12-06), p. 2470-2483
    Abstract: BCOR, encoding BCL-6 corepressor (BCOR), is X-linked and targeted by somatic mutations in various hematological malignancies including myelodysplastic syndrome (MDS). We previously reported that mice lacking Bcor exon 4 (BcorΔE4/y) in the hematopoietic compartment developed NOTCH-dependent acute T-cell lymphoblastic leukemia (T-ALL). Here, we analyzed mice lacking Bcor exons 9 and 10 (BcorΔE9-10/y), which express a carboxyl-terminal truncated BCOR that fails to interact with core effector components of polycomb repressive complex 1.1. BcorΔE9-10/y mice developed lethal T-ALL in a similar manner to BcorΔE4/y mice, whereas BcorΔE9-10/y hematopoietic cells showed a growth advantage in the myeloid compartment that was further enhanced by the concurrent deletion of Tet2. Tet2Δ/ΔBcorΔE9-10/y mice developed lethal MDS with progressive anemia and leukocytopenia, inefficient hematopoiesis, and the morphological dysplasia of blood cells. Tet2Δ/ΔBcorΔE9-10/y MDS cells reproduced MDS or evolved into lethal MDS/myeloproliferative neoplasms in secondary recipients. Transcriptional profiling revealed the derepression of myeloid regulator genes of the Cebp family and Hoxa cluster genes in BcorΔE9-10/y progenitor cells and the activation of p53 target genes specifically in MDS erythroblasts where massive apoptosis occurred. Our results reveal a tumor suppressor function of BCOR in myeloid malignancies and highlight the impact of Bcor insufficiency on the initiation and progression of MDS.
    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 ...
  • 7
    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 ...
  • 8
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 525-525
    Abstract: Mutations in a variety of genes have been identified in MDS patients. Among them, mutations of additional sex combs-like 1 (ASXL1), found in 15-20% of MDS patients, have been identified as an independent poor prognostic factor. We previously demonstrated that C-terminal–truncating ASXL1 mutations (ASXL1-MT) inhibited myeloid differentiation and induced an MDS-like disease in mice after 1~2 years by inhibiting polycomb repressive complex 2–mediated methylation of histone H3K27 (Inoue et al. J Clin Invest. 2013). Given that ASXL1 mutations have been shown to be related to high-risk MDS or leukemic transformation, it is not clear how ASXL1-mutated MDS clones can transform into advanced MDS or AML. First, we examined genetic alterations in 368 WHO-defined MDS patients; ASXL1 mutations were detected in 64 of them (17.39%). Intriguingly, the patients with ASXL1 mutations had a significantly higher incidence of the concurrent SET binding protein 1 (SETBP1) mutation than those with the wild-type ASXL1 (6 out of 64, 9.38% vs. 2 out of 304, 0.66%, P=0.0005). Moreover, among ASXL1-mutated MDS patients, those harboring SETBP1 mutations had a higher incidence of leukemic transformation than those without (P=0.042), and MDS patients with both mutations had a significantly shorter overall survival compared to those without SETBP1 mutations (median, 10.5 vs. 22.5 months, P=0.046). In addition, we demonstrated that most SETBP1 mutations, such as D868N, occur in the PEST domain of the SKI homology region, preventing ubiquitination and subsequent proteasomal degradation. These results prompted us to investigate whether SETBP1 mutations play a critical role in the leukemic transformation of ASXL1-mutated MDS cells. In in vitro experiments, the expression of SETBP1-D868N enhanced myeloid colony formation of ASXL1-MT-transduced LSK cells, augmenting ASXL1-MT-induced differentiation blocking of 32Dcl3 cells. Of note, SETBP1-D868N collaborated with ASXL1-MT to induce AML after a short latency (median survival, 73 days) in a murine BMT model, while all mice expressing either ASXL1-MT or SETBP1-D868N survived for 6 months after transplantation (P 〈 0.0001). Mice with leukemia induced by the combination of ASXL1-MT and SETBP1-D868N exhibited remarkable leukocytosis, anemia, thrombocytopenia, macrocytosis, hematosplenomegaly and hypercellular BM when compared to control mice. To clarify the molecular mechanism leading to leukemic transformation, we first investigated the Pp2a-Akt pathway because SETBP1 protein has been shown to interact with SET oncoprotein, resulting in Pp2a phosphorylation and subsequent inhibition. Consistent with previous reports using overexpression systems of SETBP1 wild type protein (SETBP1-WT), BM cells of leukemic mice displayed phosphorylated Pp2a and Akt compared to those of the control mice. Administration of FTY720, a Pp2a activator, efficiently repressed the growth rate in vitro and slightly improved the survival of serially transplanted mice. Next, using RNA-seq and GSEA, we demonstrated that SETBP1-D868N enriched hematopoietic stem cell-related genes and posterior Hoxa genes. Chromatin immnoprecipitation assay showed that both SETBP1-WT and SETBP1-D868N interacted with the promoter regions of Hoxa9 and Hoxa10, raising the possibility that a gain-of-function mutant of SETBP1 enhances transcription of these genes, directly or indirectly. Moreover, GSEA indicated global repression of the TGF-β signaling pathway and reciprocal upregulation of the Myc pathway in leukemic mice. In conclusion, our data provide evidence for the role of SETBP1 mutations in leukemic transformation and suggest the resulting deregulated pathways as potential therapeutic targets to prevent disease progression in MDS. Disclosures Harada: Kyowa Hakko Kirin Co., Ltd.: Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    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. 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 ...
  • 10
    In: Blood, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. 197-197
    Abstract: In attempts to isolate myeloid tumor-suppressor genes responsible for 7q deletion, we identified a common microdeletion cluster in chromosome subband 7q21.2 by microarraybased CGH analyses of JMML (ASH Annual Meeting, 2006). This region was also deleted in nearly 30% of unselected adult MDS/AML patients, mostly as a part of monosomy 7 or larger 7q deletions. In this region, there are three poorly-characterized genes (Miki = LOC253012, Kasumi = Samd9, and Titan = Samd9L). Miki encoding a centrosomal protein is likely involved in myelodysplasia and chromosomal instability, which are characteristic of -7/7q- MDS/AML, as is presented in this meeting elsewhere. Kasumi (Samd9) and Titan (Samd9L) are related genes that encode 60% homologous proteins. Neither Kasumi nor Titan has homology with any other proteins or contain known functional motifs. Kasumi and Titan were ubiquitously expressed at a relatively constant level. However, in six cell lines derived from MDS/AML patients harboring monosomy 7, Kasumi protein was barely detectable, whereas Titan expression levels were roughly half of those in other AML cells. The mouse genome contains only Titan and lacks Kasumi gene, suggesting that the function of these two gene products are overlapping. We started to characterize these genes by generating mice deficient in Titan (titan−/−). titan−/− mice appear normal and no hematological abnormalities have been observed, suggesting that additional gene alterations are required for leukemia development. To address this issue, retroviral insertional mutagenesis was applied to the mice. Virus infection induced acute leukemia in homozygous (titan−/−) and heterozygous (titan+/−) mice with higher morbidity and mortality than in wild-type (titan+/+) littermates. Leukemias developed in titan+/+ mice were mainly of T-cell lineage. By contrast, those developed in titan−/− and titan+/− mice were negative for lymphoid markers but expressed various combination of cell surface markers for myeloid (Gr1), monocytic (Mac1), erythyroid (Ter119) and megakaryocytic (CD61) progenitors. Histopathology demonstrated that leukemia cells infiltrated the liver, lung, kidneys and spleen, and a portion of the infiltrated cells were maturated. These data suggests that leukemias that developed in titan-deficient mice represent stem cell malignancy rather than AML. Inverse PCR detected two common integration sites (CIS) specific for titan−/− and titan+/− mice, which induced deregulated expression of a zinc finger transcription factor, Evi1, and a histone demethylase, Fbxl10. In addition, although it was not a CIS, TGFβ was isolated as a major viral integration site in one tumor. These results demonstrated that haploinsufficiency and deficiency of Titan predispose leukemia development through inhibition of TGFβ-mediated signaling or an epigenetic change. Recently, deleterious mutations in the Titan gene were reported to be involved in Normophosphatemic Familial Tumoral Carcinosis, a rare autosomal recessive disease in five families of Jewish-Yemenite origin. Impairment of cell migration is suspected to be a cause of this disease and, indeed, wound healing test revealed that fibroblasts established from titan−/− and titan+/− mice migrate slower than those established from wild-type mice. Relevance of the impairment of cell migration to development of leukemia in titan-deficient mice is currently under investigation.
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...