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
  • 1
    In: Nature Medicine, Springer Science and Business Media LLC, Vol. 18, No. 10 ( 2012-10), p. 1550-1559
    Type of Medium: Online Resource
    ISSN: 1078-8956 , 1546-170X
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2012
    detail.hit.zdb_id: 1484517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 223-223
    Abstract: Severe congenital neutropenia (CN) is a preleukemic bone marrow failure syndrome with a high risk of evolving into leukemia or myelodysplastic syndrome (MDS). Recently we demonstrated a very high frequency of cooperating RUNX1 and CSF3R mutations in CN patients who developed leukemia or MDS (Skokowa, et al. Blood 2014). We proposed a novel molecular pathway of leukemogenesis: mutations in the cytokine receptor (G-CSFR) in combination with the second mutations in the hematopoietic transcription fator (RUNX1). In the majority of CN patients, CSF3R mutations were acquired prior to RUNX1 mutations. CSF3R mutations alone are unable to induce leukemia in CN patients or in mice expressing a transgenic d715 G-CSFR. Co-acquisition of RUNX1 mutations is an essential step in the leukemogenic transformation in CN. To characterize the expression signature of hematopoietic cells of CN/AML patients carrying CSF3R mutations prior to and after acquisition of RUNX1 mutations, we analyzed expression profiles of CD34+ hematopoietic cells of CN patient who developed AML. This patient acquired CSF3R mutation (p. Q718*) five years and RUNX1 mutation (p. R139G) 16 months prior to leukemia. We compared expression profiles of CD34+ cells harbouring CSF3R mutation only, or both CSF3R and RUNX1 mutations. Co-acquisition of RUNX1 and CSF3R mutations led to marked reduction of the expression of hematopoietic growth factors such as IL6 and NAMPT, inhibitors of cytokine signaling SOCS3, as well as of components of neutrophil granules OLFM4, DEFA4, MMP8, SLPI, CRISP3 and CTSG. At the same time expression levels of pro-proliferative downstream effectors of G-CSF such as STAT5A, STAT5B, SMAD1 and cyclin A1 (CCNA1) were dramatically elevated. Moreover, genes overexpressed in early hematopoietic stem/progenitor cells (HSPCs) as compared to more mature progenitors, such as DNTT, BAALC, CD109, HPGDS, PDLIM1, MLLT11 and FLT3 were strongly upregulated in CN/AML blasts harbouring both RUNX1 and CSF3R mutations. Intriguingly, elevated expression of DNTT, BAALC, CD109 and FLT3 was described previously in RUNX1-mutated de novo AML blasts (Mendler et al., JCO 2012). This genetic signature suggests rapid transformation of hematopoietic progenitors carrying mutated CSF3R into more primitive hematopoietic progenitors after acquisition of RUNX1mutation. To elucidate the role of cooperative CSF3R and RUNX1 mutations on the clonogenic capacity and myeloid differentiation of hematopoietic progenitors, we performed functional studies in mice. We transduced lineage negative (lin-) bone marrow hematopoietic progenitor cells of WT or transgenic d715 G-CSFR mice with lentiviral expression constructs containing either WT or mutated forms of RUNX1 cDNA. We used two different mutants of RUNX1 by introduction of mutations at amino acid positions 139 and 174. Acquired RUNX1 mutations in these amino acids were presented with high frequency in our cohort of CN/AML patients and in most of the cases were associated with acquired CSF3R mutations. We found that similar to the effect of CSF3R mutations, lin-hematopoietic cells of WT mice transduced with mutated RUNX1 alone did not show elevated clonogenic capacity in replating experiments. Interestingly, transduction of WT cells with RUNX1 mutants resulted in severely reduced numbers of CFU-G colonies but unaffected CFU-M and BFU-E colonies. Intriguingly, transduction of lin- hematopoietic cells from transgenic d715 G-CSFR mice with RUNX1 mutants resulted in a markedly elevated clonogenic capacity in replating experiments, as compared to cells transduced with WT RUNX1 or control vector: numbers of colonies after second replating were 7 and 8 times higher in RUNX1-R139G and RUNX1-R174X mutants, respectively, in comparison to RUNX1 WT transduced cells. Moreover, granulocytic differentiation of lin- cells from d715 G-CSFR mice transduced with RUNX1-R139G mutant was severely diminished, in comparison to cells transduced with WT RUNX1, as revealed by 5-fold reduction of CFU-G colonies. Taken together, co-acquisition of RUNX1 and CSF3R mutations shifted the hematopoietic differentiation program towards more primitive hematopoietic progenitors with elevated proliferative capacity and reduced myeloid differentiation, which ultimately lead to leukemia. 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: 2014
    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. 118, No. 21 ( 2011-11-18), p. 2165-2165
    Abstract: Abstract 2165 Secretory Leukocyte Protease Inhibitor (SLPI) is a cationic serine protease inhibitor with antiprotease, primarily anti-Neutrophil ELastase (NE), activities. Moreover, SLPI modulates intracellular signal transduction pathways such as NF-kB and Erk. The molecular interaction and the balance between NE and SLPI is tightly regulated. On the one side, NE upregulates the SLPI expression and at the other hand SLPI inhibits the NE-induced degradation of proteins. We identified severe diminished levels of SLPI mRNA in CD33+ myeloid cells and in PMNs of patients with severe congenital neutropenia (CN) harbouring either ELANE or HAX1 mutations, as compared to patients with cyclic neutropenia (CyN) and to healthy individuals. SLPI protein levels in plasma of CN patients were also significantly reduced. We further analysed whether diminished levels of SLPI are associated with the „maturation arrest“ of myeloid cells seen in CN patients. We inhibited SLPI using lentivirus-based transduction of the myeloid cell line NB4 with SLPI-specific shRNA and analysed ATRA-triggered myeloid differentiation. Indeed, myeloid differentiation was severely affected in NB4 cells transduced with SLPI-specific shRNA, as compared to control shRNA transduced cells. Further, we analysed the mechanisms leading to SLPI downregulation. Previously, we identified severely reduced mRNA and protein levels of NE in myeloid cells and in plasma of CN patients with either ELANE or HAX1 mutations, as compared to healthy individuals. Knowing that NE induces SLPI expression, we assumed that diminished NE levels may be responsible for the low SLPI expression in CN patients. Indeed, inhibition of NE in the myeloid cell line NB4 using NE-specific shRNAs led to diminished expression of SLPI mRNA, as compared to ctrl shRNA transduced cells. At the same time, we also found that transduction of the myeloid cell line NB4 with wild type (WT) NE resulted in the increased expression of SLPI mRNA but mutated (MUT) forms of NE as found in CN patients were not able to induce SLPI mRNA, as compared to ctrl transduced cells. Taken together, both diminished NE levels and mutations in ELANE gene may cause downregulation of SLPI. In summary, SLPI is severely downregulated in CN patients due to defective NE protein levels and ELANE mutations. As a consequence, the anti-microbial and antiinflammatory activities of SLPI are diminished in CN patients. 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: 2011
    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. 122, No. 21 ( 2013-11-15), p. 444-444
    Abstract: Congenital neutropenia (CN) is a rare inherited disorder of hematopoiesis with a 20% risk of evolving into acute myeloid leukemia (AML) or myelodysplastic syndrome (MDS). Using next-generation sequencing in 31 CN patients who developed leukemia we found that 20 of the 31 patients (64.5%) had mutations in RUNX1 (runt-related transcription factor 1). Of these 20 patients, 19 had inherited mutations associated with CN. Intriguingly, the majority of patients with RUNX1 mutations (80.5%) also had acquired CSF3R (colony stimulating factor 3 receptor) mutations. Other leukemia-associated mutations (EP300, FLT3-ITD, CBL, and SUZ12) were less frequent. In eight patients, we detected two distinct heterozygous RUNX1 mutations. These mutations were localized to the splice-acceptor site of intron 4, affecting splicing of exons 3 and 4, which encode the Runt homology/DNA binding domain (RHD) of RUNX1, or solely in the RHD or were present in both RHD and trans-activation domain (TAD). In two patients, we were able to perform allele-specific analysis of RUNX1 mutations. Patient #10 had an Phe13TrpfsX14 deletion on one allele of RUNX1 and an Arg139ProfsX47 deletion on the other allele. In Patient #14, two RUNX1 mutations were on the same allele; one of the mutations (Met240Ile) was inherited from the mother and was localized two amino acids before the TAD, and the second acquired mutation (Arg139Gly) was in the RHD of RUNX1. Ten patients with RUNX1 mutations developed monosomy 7 and six patients developed trisomy 21 at diagnosis of leukemia. In contrast to their high frequency in CN evolving into AML, RUNX1 mutations were found in only 9 of 307 (2.9%) patients with de novo pediatric AML. RUNX1 mutations were mainly found in pediatric AML patients with an adverse prognosis. A sequential analysis at stages prior to overt leukemia in ten CN/AML patients showed that RUNX1 mutation is a late event in leukemogenic transformation. In 6 of 10 patients, a CSF3R mutation occurred prior to RUNX1 mutations (24-192 months prior to CN/AML for CSF3R mutations vs. 1-36 months prior to CN/AML for RUNX1 mutations). Interestingly, monosomy 7 or trisomy 21 appeared after acquisition of RUNX1 mutations and no additional chromosomal aberrations were detected by array-CGH. Single-cell analyses in two patients revealed that RUNX1 and CSF3R mutations were segregated in the same malignant clone. Moreover, functional studies demonstrated elevated G-CSF-induced proliferation with diminished myeloid differentiation of hematopoietic CD34+ cells after co-transduction with mutated RUNX1 and CSF3R, in comparison to cells transduced with mutated RUNX1 or mutated CSF3R only. The importance of RUNX1 mutations in leukemogenic transformation was substantially strengthened by the analysis of a unique family with two siblings suffering from CN that subsequently transformed to AML. In both children, cooperating RUNX1 and CSF3Rmutations were detected that were not present in healthy family members. Taken together, the high frequency and the time course of cooperating RUNX1 and CSF3R mutations in CN patients who developed leukemia suggests a unique molecular pathway of leukemogenesis similar as has been reported in the Gilliland-Griffin two-hit hypothesis for AML development. The concomitant detection of RUNX1 and CSF3Rmutations represents a useful biomarker for identifying CN patients with a high risk of progressing to leukemia or MDS. Disclosures: Schnittger: MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Kohlmann:MLL Munich Leukemia Laboratory: Employment.
    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 ...
  • 5
    In: Blood, American Society of Hematology, Vol. 126, No. 15 ( 2015-10-08), p. 1865-1867
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    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. 127, No. 21 ( 2016-05-26), p. 2638-2641
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    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. 123, No. 14 ( 2014-04-03), p. 2229-2237
    Abstract: CN/AML patients have a high frequency of CSF3R and RUNX1 mutations. CSF3R and RUNX1 mutations induce elevated proliferation of CD34+ cells.
    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 ...
  • 8
    Online Resource
    Online Resource
    American Society of Hematology ; 2012
    In:  Blood Vol. 120, No. 21 ( 2012-11-16), p. 10-10
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 10-10
    Abstract: Abstract 10 Previously, we demonstrated severe diminished mRNA and protein levels of secretory leukocyte protease inhibitor (SLPI) in CD33+ myeloid cells in PMNs and in plasma of patients with severe congenital neutropenia (CN), as compared to G-CSF treated healthy individuals. We further analysed whether diminished levels of SLPI are associated with the “maturation arrest“ of myeloid cells seen in CN patients. We inhibited SLPI in the CD34+ bone marrow hematopoietic progenitor cells and in the acute myelid leukemia cell line NB4 using lentivirus-based transduction with SLPI-specific shRNA and analysed G-CSF- or ATRA-triggered myeloid differentiation, respectively. We found that G-CSF-triggered myeloid differentiation of CD34+ cells transduced with SLPI-specific shRNA was significantly diminished, in comparison to ctrl shRNA transduced cells (on day 14 of differentiation 53.8 % of CD11b+ cells in SLPI shRNA group vs 86.3 % in the ctrl shRNA group (ttest: p = 0.005); 13.1 % of CD16+ cells in SLPI shRNA group vs 48.8 % in the ctrl shRNA group (ttest: p = 0.0006)). This was accompanied by G0/G1 cell cycle arrest and elevated apoptosis of SLPI shRNA transduced cells: on day 7 of differentiation 83.9 % of SLPI shRNA transduced cells and 66.6 % of the ctrl shRNA transduced cells were in G0/G1 phase (ttest: p = 0.03); 11 % of annexin V positive cells in SLPI shRNA transduced group vs 6.5 % in ctrl shRNA group (ttest: p = 0.004). Similar effects on the ATRA-induced myeloid differentiation were observed after inhibition of SLPI in the NB4 cell line. We next analysed the mechanisms of diminished myeloid differentiation due to diminished SLPI levels. SLPI is a cationic serine protease inhibitor with antiprotease, primarily anti-neutrophil elastase, activities. Previously we showed regulation of neutrophil elastase by SLPI in myeloid cells. However, SLPI is also known to modulate intracellular signal transduction pathways such as NF-kB and Erk. SLPI is involved in the phosphorylation of Erk1/2 protein. Erk1/2 protein plays a role in myeloid differentiation. We found that ATRA treatment of NB4 cells transduced with ctrl shRNA led to the transient time-dependent phosphorylation of Erk1/2 on day 2 of culture with subsequent decline on day 4 of treatment. Interestingly, Erk1/2 phosphorylation was almost completely abolished on day 2 of ATRA treatment of NB4 cells transduced with SLPI shRNA. Recently, we demonstrated an essential role of LEF-1 in the granulocytic differentiation and severe diminished LEF-1 expression levels in myeloid cells of CN patients. It has been shown that Erk1/2 phosphorylates LEF-1 transcription factor. In line with defective phosphorylation of Erk1/2 we measured severe diminished levels of phosphorylated LEF-1 in SLPI shRNA transduced cells, in comparison to ctrl shRNA samples. Therefore, we concluded that SLPI regulates myeloid differentiation by regulation of Erk1/2 dependent LEF-1 phosphorylation and autoregulation. To evaluate additional intracellular signaling pathways which are affected during G-CSF-triggered myeloid differentiation if SLPI is downregulated, we performed microarray analysis of RNA from CD34+ cells transduced with SLPI or ctrl shRNA and subsequently treated with G-CSF for 24 hours. Analysis of the microarray data using Ingenuity Systems Pathway Analysis (IPA) revealed severe significant defects in the signaling cascades involved in the differentiation (p-Value = 0.0093), proliferation (p-Value = 0.015), survival (p-Value = 0.012) and chemotaxis (p-Value = 0.0093) of myeloid progenitor cells and neutrophils which was correlated with significant downregulation of FPR2, MMP8, XCL1, S100A9, IL2 and OLFM4 mRNA expression. Moreover, signaling systems regulating DNA replication checkpoint and arrest in Go/G1 phase were also significantly affected (p-Value = 0.0009 and 0.0065, respectively) due to downregulation of mRNA expression of CHEK1, CCNE2, CDC6, MYBL1 and MYC. Taken together, SLPI is a new candidate factor playing an important role in myelopoiesis by controlling proliferation, differentiation and cell cycle of myeloid cells not only by inhibition of neutrophil elastase but also by regulation of intracellular signaling pathways. 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 ...
  • 9
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 11-11
    Abstract: Abstract 11 Cyclic neutropenia (CyN) is a hematologic disorder in which blood cell counts particularly granulocytic neutrophil numbers show cycles at 21 day intervals. The majority of CyN patients (ca 90 %) harbor inherited mutations in the ELANE gene. Intriguingly, same ELANE mutations are present in two different hematologic syndromes: congenital as well as in cyclic neutropenias. It is unclear how mutation in the same gene cause congenital or cyclic neutropenia. We aimed to identify genes which are exclusively mutated in cyclic or in congenital neutropenia additionaly to the ELANE gene mutations. Recently, we found in congenital neutropenia patients additional to ELANE mutations inherited mutations in for example the G6PC3 gene or the HAX1 gene (Germeshausen, M., et al, Haematologica 2010). This suggests cooperating effects of different defective intracellular signaling pathways and excludes that mutated ELANE alone is responsible for the pathogenesis of congenital or cyclic neutropenia. To identify gene mutations causing cyclic neutropenia in association with ELANE mutation, we performed whole genome sequencing using Complete Genomics technology (Complete Genomics. Inc, Mountain View, CA.) of a family with an affected CyN patient. The CyN patient harbors sporadic heterozygous ELANE mutation (c.761C 〉 G p.W241L) and her family consists of a healthy brother and healthy parents. We identified a novel heterozygous point mutation in the tumor necrosis factor receptor superfamily, member 1A (TNFRSF1A) gene (c.664C 〉 T; p.R121Q) in the affected patient and her mother. This mutation was confirmed by Sanger sequencing. The TNFRSF1A gene encodes p55 subunit of the TNFa receptor (TNFR1) and intriguingly this gene is known to be frequently mutated in patients with Tumour necrosis factor receptor-associated periodic fever syndrome (TRAPS), a disease clinically similar to cyclic neutropenia. TRAPS is an autosomal dominant disorder characterized by episodes of fever, inflammation and periodical changes in the neutrophil counts. Functional studies in patients with TRAPS described heterogenous effects of different TNFRSF1A mutations on the surface expression and PMA-induced clevage of TNFR1 on the neutrophilic granulocytes and monocytes. In our CyN patient we measured elevated mRNA levels of TNFR1 on neutrophils, in comparison to her healthy family members and unrelated healthy individuals. However, we detected diminished surface expression of the TNFR1 protein, but elevated PMA-induced receptor shedding in the affected CyN patient, in comparison to healthy individuals, as assessed by estimation of soluble TNFR1 in supernatants of PMA-stimulated neutrophils. We also identified in the same patient and her father a second novel heterozygous point mutation in the CEBPE gene (c.636C 〉 A; p.L155M), which was also confirmed by Sanger sequencing. This C 〉 A substitution changes CTG to ATG creating a new start site for translation of a novel isoform of C/EBPε protein. C/EBPε is a myeloid-specific transcription factor playing an important role in granulopoiesis. Mutations in the CEBPE gene have been described in patients with neutrophil-specific granule deficiency (SGD). In summary, we identified additional to the ELANE mutation two novel mutations in a CyN patient, one in the TNFRSF1A gene inherited from the mother, another in the CEBPE gene inherited from the father. Mutations in both genes are already described in patients with TRAPS (periodic fever syndrome) and granulocyte abnormalities, respectively. These mutations in association with the ELANE gene mutation may contribute to the pathogenesis of cyclic neutropenia in this patient. Whether the combination of these three mutations might be responsible for a subgroup of CyN patients remains to be investigated. 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 ...
  • 10
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 2199-2199
    Abstract: Previously, we described new mechanism of G-CSF-triggered granulocytic differentiation of hematopoietic stem cells (HSCs) via activation of the enzyme Nicotinamide Phosphorybosyltransferase (NAMPT) leading to NAD+ production and activation of NAD+ -dependent protein deacetylase sirtuin 1 (SIRT1). We found, that upon stimulation of HSCs with NAMPT, SIRT1 bound to the key myeloid transcription factor C/EBPα followed by transcriptional induction of C/EBPα target genes G-CSFR and G-CSF and granulocytic differentiation. In the present work we investigated the mechanism of NAMPT/SIRT1-triggered deacetylation of C/EBPα. We found that C/EBPα is acetylated at the position Lys 161, which is evolutionarily conserved. Lys 161 is localized in the transactivation element III (TE-III) of the transactivation domain (TAD) of C/EBPα protein, which is responsible for recruitment of SWI/SNF and CDK2/CDK4. Western blot and DUOLINK analysis using rabbit polyclonal antibody specifically recognizing acetyl-Lys 161 of C/EBPα revealed predominantly nuclear localization of acetylated C/EBPα protein in acute myeloid leukemia cell lines NB4 and HL60 as well as in primary HSCs. Induction of myeloid differentiation of HSCs by treatment with G-CSF as well as ATRA-induced differentiation of NB4 cells resulted in the deacetylation of C/EBPα. NAMPT inhibition in NB4 and HL60 cell lines using specific inhibitor FK866 led to the dramatically elevated levels of acetylated C/EBPα and reduced amounts of total C/EBPα protein, which was in line with diminished mRNA expression of C/EBPα target genes (G-CSF, G-CSFR and ELANE). Interestingly, treatment of acute myeloid leukemia cell line HL60 with NAMPT or transduction of HL-60 cells with NAMPT-expressing lentiviral construct induced myeloid differentiation of these cells even without addition of ATRA. This was in line with time- and dose-dependent increase of total C/EBPα protein levels upon NAMPT treatment. Therefore, NAMPT overcomes transcriptional repression of C/EBPα in HL-60 cells by activation of positive CEBPA autoregulation. Taken together, we described a new mechanism of regulation of C/EBPα activities in hematopoiesis and leukemogenesis by its post-translational modification via NAMPT/SIRT1-triggered de-/acetylation. 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: 2015
    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...