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. 12, No. 10 ( 2006-10-01), p. 1191-1197
    Type of Medium: Online Resource
    ISSN: 1078-8956 , 1546-170X
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2006
    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. 112, No. 11 ( 2008-11-16), p. 5-5
    Abstract: We here describe a previously unrecognized nosological entity in 12 patients from 8 unrelated pedigrees. All patients presented with severe congenital neutropenia and severe invasive bacterial infections. In addition, patients had a variety of additional syndromic features such as congenital heart disease (8/12), urogenital malformations (5/12), inner ear hearing loss (2/12), and myopathy (1/12). Furthermore, most patients (10/12) showed increased visibility/angiectasia of subcutaneous veins. The bone marrow smear was characterized by a typical “maturation arrest” due to premature apoptosis of mature neutrophils. Similar to Kostmann’s disease secondary to mutations in HAX1, myeloid cells from patients with this novel syndrome showed increased susceptibility to apoptosis. Myeloid progenitor cells revealed an abnormally enlarged rough endoplasmic reticulum and increased endoplasmic reticulum stress evidenced by increased expression of BiP. A genome-wide linkage study, performed in two consanguineous pedigrees, gave statistical evidene of a linkage interval on chromosome 17q21 (LOD score 5.74). We identified homozygous missense mutations in G6PC3, a ubiquitously expressed paralog of glucose-6-phosphatase. Biochemical studies confirmed deficient enzymatic activity. Using retroviral G6PC3-gene transfer into primary hematopoietic stem cells and in vitro differentiation into myeloid cells, the phenotype of increased susceptibility to apoptosis could be reverted. Eight distinct biallelic mutations were found, including missense and nonsense mutations. G6PC3-deficient myeloid cells showed a predominance of the unphosphorylated form of GSK3beta, a key molecule controlling cellular differentiation and apoptosis. As a consequence of increased GSK3beta activity, increased phosphorylation of the antiapoptotic molecule Mcl1 was detected, explaining increased susceptibility to apoptosis in neutrophils. In summary, our study describes a novel disease, determines its molecular etiology, and sheds light on the role of glucose-dependent pathways in controlling the homeostasis of the endoplasmic reticulum and control of apoptosis.
    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 ...
  • 3
    In: Blood, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. 2532-2532
    Abstract: An increased risk for malignant transformation (myelodysplastic syndrome (MDS) or leukemia) is well documented in patients with congenital neutropenia (CN). However, the risk of defined genetic subgroups of CN remains unknown. We therefore assessed the incidence of leukemic transformation and potential risk factors for leukemic transformation in CN patients with ELA2 and HAX1 mutations, respectively. The overall cumulative incidence of secondary leukemias in CN, as documented by long term follow up data from the European Severe Chronic Neutropenia Registry (SCNER) and the French Neutropenia Registry (FR) is 39 out of 343 patients (11.4 %). Up to now genetic testing has been performed in 177 of the 343 CN patients revealing ELA2 mutations in 99 (56%) and HAX1 mutations in 19 (11%) of tested CN patients. In 107patients no ELA2 mutation was detectable. Out of these 107 ELA2 negative patients 59 were also negative for HAX1 mutations (33% ELA2/HAX1 negative patients). In both, patients with ELA2-CN and HAX1-CN, the clinical phenotype is characterized by a maturation arrest of myelopoiesis and severe neutropenia and cannot be discriminated morphologically. There is also no difference in the gender distribution. Both subgroups benefit from G-CSF treatment and respond well to similar G-CSF doses (median G-CSF dose in ELA2-CN is 8,5 μg/kg/day versus 6 μg/kg/day in HAX1-CN. Secondary malignancies occurred in 17 (5 MDS, .11. AML, 1 bi-phenotypic leukemia) out of 99 (17%) ELA2-CN patients, 4 (2 MDS, 1 AML, 1 bi-phenotypic leukemia) out of 19 (21%) HAX1-CN patients, and 5 (1 MDS, 3 AML, 1 ALL) out of 59 (8 %) double negative patients. Mutations in the G-CSF receptor gene have been detectable in both groups of CN patients. The frequency of mutations increases over time with a significant higher frequency of G-CSFR mutations in the CN patients who developed leukemia indicative for an involvement of G-CSFR mutations in the process of leukemic transformation. Interestingly, G-CSFR mutations are detectable in 5 of 8 ELA2-CN leukemic patients tested and 1 of 2 HAX1-CN leukemic patients tested, but also in 1 out of 4 ELA2/HAX1 negative leukemic patients. Independent of the underlying genetic defect the dose response to G-CSF treatment seems to indicate the risk for leukemia. Patients being treated with less than 5 μg/kg/day had an leukemia incidence of 14% versus 26% in patients who received 5 μg/kg/day or more (p=0.027). Within the group of patients being treated with less than 5 μg/kg/day, 14% developed leukemia, whereas in the group of patients receiving = 5 μg/kg/day 26% developed leukemia (p=0.027), suggesting that those patients requiring higher doses of GCSF may be at higher risk for malignant transformation. Leukemia outcome is dependent on the success of hematopoietic stem cell transplantation. The outcome could be improved dramatically since SCT was initiated immediately after confirmation of the leukemia diagnosis. It would be interesting to further discuss why different genetic backgrounds (ELA2 and HAX1) reveal the same clinical phenotype in terms of morphology, response to G-CSF treatment, acquisition of G-CSFR mutations and the development of leukemia.
    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 ...
  • 4
    Online Resource
    Online Resource
    American Society of Hematology ; 2007
    In:  Blood Vol. 110, No. 11 ( 2007-11-16), p. 3296-3296
    In: Blood, American Society of Hematology, Vol. 110, No. 11 ( 2007-11-16), p. 3296-3296
    Abstract: Congenital neutropenia (CN) is a heterogeneous group of disorders, characterized by an absolute neutrophil count (ANC) of less than 500 cells per mm3 from birth on and a predisposition of the patients to recurrent and partial life threatening bacterial and fungal infections as a consequence thereof. CN is associated with an exclusive deficiency in neutrophil development. It usually presents early in infancy during the first year of life, and its infectious complications include cellulitis, perirectal abscesses, pneumonitis, peritonitis, stomatitis, and meningitis due to the extremely low ANC ( 〈 200 cells mm 3). The numbers of circulating monocytes and eosinophils are often increased. However, there is a cumulative incidence of 21% for a malignant transformation in CN after 10 years on G-CSF. Point mutations in the gene for the G-CSF receptor CSF3R have been implicated in the progression of CN to leukemia: In 42% out of 219 CN patients tested so far we could demonstrate acquired nonsense mutations in the CSF3R gene. However, in the subpopulation of patients who have developed leukemia 78% are revealing CSF3R mutations. To clarify the involvement of CSF3R mutations in the process of malignant transformation it is important to know at which stage of hematopoietic development these mutations emerge. Therefore we analyzed single cells of flow cytometrically sorted subpopulations of hematopoietic progenitors and mature hematopoietic cells from bone marrow and peripheral blood of 4 CN patients for the existence of CSF3R mutations. The different cell populations reflect the different lineages of hematopoietic development (CD33+, CD34+, CD14+, CD3+, CD19+, CD16+). Two patients characterized by the occurrence of multiple mutations were screened before any signs of malignant transformation and two patients were analyzed at time of secondary AML. Mutated cells were found in all analyzed populations including lymphocytes. However, in the CD34+ cells, eosinphil granulocytes and in the lymphocytic cells the frequency was much lower than in the G-CSFR expressing monocytes and neutrophil granulocytes. These results suggest an in vivo growth advantage of cells expressing a truncated G-CSFR confirming in vitro results with transfected cell lines and underlines the significance of CSF3R mutations in leukemogenesis in CN patients.
    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 ...
  • 5
    In: Blood, American Society of Hematology, Vol. 111, No. 10 ( 2008-05-15), p. 4954-4957
    Abstract: Homozygous mutations in HAX1 cause an autosomal recessive form of severe congenital neutropenia (CN). By screening 88 patients with CN, we identified 6 additional patients with HAX1 mutations carrying 4 novel mutations. Of these, 2 affect both published transcript variants of HAX1; the other 2 mutations affect only transcript variant 1. Analysis of the patients' genotypes and phenotypes revealed a striking correlation: Mutations affecting transcript variant 1 only were associated with CN (23 of 23 patients), whereas mutations affecting both transcript variants caused CN and neurologic symptoms, including epilepsy and neurodevelopmental delay (6 of 6 patients). In contrast to peripheral blood, transcript variant 2 was markedly expressed in human brain tissue. The clinical phenotype of HAX1 deficiency appears to depend on the localization of the mutation and their influence on the transcript variants. Therefore, our findings suggest that HAX1 isoforms may play a distinctive role in the neuronal system.
    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 ...
  • 6
    In: Blood, American Society of Hematology, Vol. 97, No. 3 ( 2001-02-01), p. 829-830
    Type of Medium: Online Resource
    ISSN: 1528-0020 , 0006-4971
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2001
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    Wiley ; 2002
    In:  British Journal of Haematology Vol. 118, No. 3 ( 2002-09), p. 923-924
    In: British Journal of Haematology, Wiley, Vol. 118, No. 3 ( 2002-09), p. 923-924
    Type of Medium: Online Resource
    ISSN: 0007-1048 , 1365-2141
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2002
    detail.hit.zdb_id: 1475751-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Nature Medicine, Springer Science and Business Media LLC, Vol. 12, No. 11 ( 2006-11), p. 1329-1329
    Type of Medium: Online Resource
    ISSN: 1078-8956 , 1546-170X
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2006
    detail.hit.zdb_id: 1484517-9
    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. 3299-3299
    Abstract: Severe congenital neutropenia (CN) is a heterogeneous disorder of myelopoiesis with two major types of inheritance: autosomal dominant CN defined by mutations in ELA2 gene encoding neutrophil elastase (NE) (Horwitz M., et al., Nat Genet.1999;23:433) and autosomal recessive CN (including Kostmann syndrome) carrying HAX-1 mutations (Klein C., et al., Nat Genet.2007;39:86), both characterized by a maturation arrest of granulopoiesis at the level of promyelocytes. In the present study we aimed to evaluate the expression profile of genes specifically expressed in the CD33+ bone marrow promyelocytes of both patient groups harbouring ELA2 or HAX1 mutations. In healthy individuals mRNA expression levels of neutrophil serine proteases (neutrophil elastase (ELA2), cathepsin G, cathepsin D, proteinase 3 and azurocidin) as well as of myeloperoxidase (MPO) and defensins reached highest levels in the azurophil granules at the promyelocytic stage of neutrophil differentiation (Borregaard N., et al., Curr Opin Hematol.2001;8:23). We found downregulation of mRNA expression levels of ELA2 (8.9 fold), cathepsin G (7.6 fold), cathepsin D (11.2 fold), proteinase 3 (9.2 fold) and defensin B1 (6.5 fold) in both groups of CN patients (with ELA2 or HAX1 mutations), in comparison to G-CSF-treated patients with idiopathic neutropenia (IN) and G-CSF-treated healthy controls. In contrast, there were no difference in mRNA expression levels of azurocidin and only slight decrease in the expression of MPO mRNA in CN patients. Additionally, we found significantly reduced protein levels of neutrophil elastase (NE) in plasma of CN patients irrespective of “ELA2 or HAX1” inheritance, in comparison to cyclic neutropenia (CyN) patients, IN patients and G-CSF-treated healthy controls. Taken together, both ELA2 and HAX1 mutations are associated with defective expression of neutrophil serine proteases such as NE, cathepsin G, cathepsin D, proteinase 3 as well as of defensin B1 in CD33+ myeloid progenitor cells of CN patients, suggesting a common pathway for both patient groups. Intriguingly, ELA2 expression is directly regulated by LEF-1, suggesting that abrogated LEF-1 expression in CN promyelocytes (Skokowa J., et al., Nat. Med.2006;12:1191) may be responsible for defective serine proteases expression in both groups, since all are regulated by a similar mechanism.
    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, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 3597-3597
    Abstract: Abstract 3597 Poster Board III-534 An increased risk for malignant transformation (MDS or leukemia) is well documented in patients with congenital neutropenia (CN). In this study we assessed the incidence of leukemic transformation and potential risk factors for leukemic transformation in CN patients with known gene mutations, e.g. ELA2, HAX1, G6PC3, p14, WAS or no identified mutation, respectively, by combining all available data from the European and US Branches of the Severe Chronic Neutropenia Registry (SCNIR) and the French Neutropenia Registry (FR). Data from mutational analysis were available for 407 patients. Mutations were identified in 259 CN patients, of whom 209 patients revealed ELA2 mutations, 20 HAX1 mutations, 18 WAS, 8 G6PC3 and 4 p14 mutations. In addition, in 57 patients neither ELA2 nor HAX 1 mutation were detectable and in another 91 patients ELA2 mutations could be excluded, but further genetic evaluation is not yet completed. Secondary malignancies occurred in 50 of the 407 CN patients. The distribution by genetic subtype is shown in the table below: CN subtype by gene mutation Total patient number (n) MDS/Leukemia (n/%) ELA2-CN 209 31 (14. 8%) HAX1-CN 20 4 (20%) ELA2 neg + HAX1 neg 57 6 (10.5%) ELA2 neg/ HAX1 not tested 91 5 (5.5%) WAS 18 4 (22.2%) G6PC3 8 0 p14 4 0 Total 407 50 (12.3%) All subgroups benefit from G-CSF treatment. Median G-CSF maintenance doses required during the years prior to leukemic transformation compared by genetic subtype is shown in the following table 2: CN subtype by gene mutation Without leukemia (n) Median G-CSF dose (μg/kg/d) With leukemia (n) Median G-CSF dose (μg/kg/d) ELA2-CN 75 5.0 15 15 HAX1-CN 15 5.0 4 13.4 ELA2-/HAX1- 36 7.2 4 10.5 WAS 15 2.6 4 3.0 G6PC3 8 3.9 0 na p14 4 5.1 0 na Conclusion Patients with severe congenital neutropenia who have mutations in ELA2, HAX1, or WAS and also those with no recognized mutation are at risk of secondary leukemia. So far, progression to MDS leukemia has not yet been described in the small number G6PC3 or p14 CN cases in our database. ELA2-CN or HAX1-CN patients requiring higher doses of G-CSF are at greater risk. Mutational analysis is helpful to identify the genetic cause of severe congenital neutropenia but does not serve to identify patients at risk of leukemic transformation. 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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...