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: Cancer Cell, Elsevier BV, Vol. 22, No. 1 ( 2012-07), p. 51-65
    Type of Medium: Online Resource
    ISSN: 1535-6108
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2012
    detail.hit.zdb_id: 2074034-7
    detail.hit.zdb_id: 2078448-X
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Leukemia & Lymphoma, Informa UK Limited, Vol. 56, No. 3 ( 2015-03-04), p. 577-586
    Type of Medium: Online Resource
    ISSN: 1042-8194 , 1029-2403
    Language: English
    Publisher: Informa UK Limited
    Publication Date: 2015
    detail.hit.zdb_id: 2030637-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Genetics, Elsevier BV, Vol. 204, No. 4 ( 2011-4), p. 187-194
    Type of Medium: Online Resource
    ISSN: 2210-7762
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2011
    detail.hit.zdb_id: 2594323-6
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    SAGE Publications ; 2016
    In:  Therapeutic Advances in Hematology Vol. 7, No. 1 ( 2016-02), p. 52-64
    In: Therapeutic Advances in Hematology, SAGE Publications, Vol. 7, No. 1 ( 2016-02), p. 52-64
    Abstract: High-grade B-cell lymphomas with recurrent chromosomal break points have been termed ‘double hit lymphoma’ (DHL). The most commonly seen DHL is diffuse large B-cell lymphoma (DLBCL) with t(14;18) and t(8;14) or t(8;22) resulting in overexpression of BCL2 and MYC, respectively. The increased proliferation due to MYC overexpression, without the ability for an apoptotic brake as a result of BCL2 overexpression, results in ‘the perfect storm of oncogenesis’. Thus this disease presents a number of diagnostic and therapeutic challenges for the hematologist. The first and foremost challenge is to recognize the DHL. As different morphological entities can be affected it is incumbent on pathologists and clinicians to maintain a high index of suspicion especially in disease that appears unusually aggressive or refractory to therapy. Diagnosis by fluorescence in situ hybridization (FISH) is a sensitive and specific method for detection of the disease but is time-consuming and expensive. While detection by immunohistochemistry (IHC) is sensitive and correlates with survival, standardized methods for this are not widely agreed upon. The second and equally important challenge in DHL is optimizing clinical outcome in a group of patients for whom the prognosis is widely regarded as poor. While improvements have been achieved by dose escalating standard chemotherapeutic regimens, many patients continue to do badly. Furthermore as a disease of aging many patients are unsuitable for dose-intensive chemotherapy regimens. There are now multiple novel targeted agents in various stages of clinical development that offer hope for better outcomes without undue toxicity. Among the most exciting of these developments include specific inhibitors of both BCL2 and MYC.
    Type of Medium: Online Resource
    ISSN: 2040-6207 , 2040-6215
    Language: English
    Publisher: SAGE Publications
    Publication Date: 2016
    detail.hit.zdb_id: 2585183-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Science Signaling, American Association for the Advancement of Science (AAAS), Vol. 4, No. 188 ( 2011-08-30)
    Abstract: Precise regulation of ribosome biogenesis is fundamental to maintain normal cell growth and proliferation, and accelerated ribosome biogenesis is associated with malignant transformation. Here, we show that the kinase AKT regulates ribosome biogenesis at multiple levels to promote ribosomal RNA (rRNA) synthesis. Transcription elongation by RNA polymerase I, which synthesizes rRNA, required continuous AKT-dependent signaling, an effect independent of AKT’s role in activating the translation-promoting complex mTORC1 (mammalian target of rapamycin complex 1). Sustained inhibition of AKT and mTORC1 cooperated to reduce rRNA synthesis and ribosome biogenesis by additionally limiting RNA polymerase I loading and pre-rRNA processing. In the absence of growth factors, constitutively active AKT increased synthesis of rRNA, ribosome biogenesis, and cell growth. Furthermore, AKT cooperated with the transcription factor c-MYC to synergistically activate rRNA synthesis and ribosome biogenesis, defining a network involving AKT, mTORC1, and c-MYC as a master controller of cell growth. Maximal activation of c-MYC–dependent rRNA synthesis in lymphoma cells required AKT activity. Moreover, inhibition of AKT-dependent rRNA transcription was associated with increased lymphoma cell death by apoptosis. These data indicate that decreased ribosome biogenesis is likely to be a fundamental component of the therapeutic response to AKT inhibitors in cancer.
    Type of Medium: Online Resource
    ISSN: 1945-0877 , 1937-9145
    Language: English
    Publisher: American Association for the Advancement of Science (AAAS)
    Publication Date: 2011
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Leukemia, Springer Science and Business Media LLC, Vol. 36, No. 12 ( 2022-12), p. 2883-2893
    Type of Medium: Online Resource
    ISSN: 0887-6924 , 1476-5551
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2022
    detail.hit.zdb_id: 2008023-2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Cell Reports, Elsevier BV, Vol. 14, No. 6 ( 2016-02), p. 1488-1499
    Type of Medium: Online Resource
    ISSN: 2211-1247
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2016
    detail.hit.zdb_id: 2649101-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Scientific Reports, Springer Science and Business Media LLC, Vol. 9, No. 1 ( 2019-05-07)
    Abstract: Deletion of long arm of chromosome 20 [del(20q)] is the second most frequent recurrent chromosomal abnormality in hematological malignancies. It is detected in 10% of myeloproliferative neoplasms, 4–5% of myelodysplastic syndromes, and 1–2% of acute myeloid leukaemia. Recurrent, non-random occurrence of del(20q) indicates that it is a pathogenic driver in myeloid malignancies. Genetic mapping of patient samples has identified two regions of interest on 20q – the “Common Deleted Region” (CDR) and “Common Retained Region” (CRR), which was often amplified. We proposed that the CDR contained tumor suppressor gene(s) (TSG) and the CRR harbored oncogene(s); loss of a TSG together with over-expression of an oncogene favored development of myeloid malignancies. Protein Tyrosine Phosphatase Receptor T (PTPRT) and Hemopoietic cell kinase (HCK) were identified to be the likely candidate TSG and oncogene respectively. Retroviral transduction of HCK into PTPRT-null murine LKS+ stem and progenitor cells resulted in hyperproliferation in colony forming assays and hyperphosphorylation of intracellular STAT3. Furthermore, over half of the murine recipients of these transduced cells developed erythroid hyperplasia, polycythemia and splenomegaly at 12 months, although no leukemic phenotype was observed. The findings suggested that HCK amplification coupled with PTPRT loss in del(20q) leads to development of a myeloproliferative phenotype.
    Type of Medium: Online Resource
    ISSN: 2045-2322
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2019
    detail.hit.zdb_id: 2615211-3
    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. 591-591
    Abstract: Rothmund-Thomson Syndrome (RTS) is a rare autosomal recessive disorder that presents with congenital skeletal malformations, premature ageing and an increased incidence of malignant disease including osteosarcoma and hematologic malignancy. The majority of RTS patients have deleterious germ-line mutations in the RECQL4 helicase. RECQL4 is a member of a family of DNA helicases including Bloom (BLM) and Werner (WRN) syndrome helicases and is thought to play an important role in maintaining chromosome stability. Recql4 deficiency is associated with karyotypic abnormalities and increased rates of aneuploidy. To enable lineage-restricted deletion of Recql4 we generated a mouse Recql4fl/fl line where exon 9 and 10 are flanked by loxP sites. This region encodes the start of the ATP dependent helicase domain and corresponds to human exons 8 and 9, mutations of which are commonly associated with cancers in RTS patients. Germ-line deletion of Recql4 led to embryonic lethality before E10.5. To understand the role of Recql4 in adult hematopoiesis, Rosa26-CreERT2 Recql4+/+, Recql4fl/+ and Recql4fl/fl mice were fed a tamoxifen containing diet (which activates Cre) for up to 4 weeks from 7 weeks of age to somatically delete Recql4. The most striking phenotype observed in these mice was that all R26-CreERT2 Recql4fl/fl animals became anaemic and developed white extremities, which in nearly all cases necessitated euthanasia by 30 days after starting tamoxifen. No differences were observed between R26-CreERT2Recql4+/+ and R26-CreERT2Recql4fl/+. Hematopoietic analyses revealed a severe multi-lineage cytopenia including profound red cell aplasia in the peripheral blood, spleen and bone marrow of Recql4 deficient animals. Effects were observed across all lineages with myeloid, B-lymphoid and T-lymphoid cell numbers severely reduced with a phenotype consistent with acute bone marrow failure. Loss of Recql4 led to a block in B-lymphoid development in the BM at the PrePro-B to Pro-B cell stage. T-lymphoid development was severely interrupted and most populations in the thymus were impacted. Within the stem and progenitor fractions, phenotypic HSCs were preserved in the absence of Recql4 but we found that the erythroid progenitors were nearly completely lost and only CMP/GMP remained. Colony formation was reduced by ∼70% in the deleted bone marrow. Bone marrow transplantation was used to assess the functionality of the Recql4 deleted HSCs. Strikingly, and consistently observed using both R26-CreERT2 and an additional hScl-CreER model, there was a strong selection against deficient cells and a recovery of hematopoiesis by cells with incomplete deletion of Recql4. This result demonstrates an essential role for Recql4 in the maintenance of hematopoiesis. In vitro B and T cell cultures recapitulated the developmental defects observed in the R26-CreERT2 Recql4fl/fl demonstrating a cell intrinsic requirement for Recql4. Loss of Recql4 lead to an accumulation of cells in S-phase and increased levels of DNA damage as measured by gH2Ax staining. Interestingly, the failure in B cell, T cell development and LKS+ CFC formation could be rescued by overexpression of a either WT or a helicase-dead Recql4-K508A mutant. Collectively these data demonstrate that Recql4 is essential for the maintenance of hematopoiesis and acts to maintain the committed progenitor pool. Loss of Recql4 leads to bone marrow failure, demonstrating a unique requirement for Recql4 in the regulation of hematopoiesis. 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
    Online Resource
    Online Resource
    American Society of Hematology ; 2011
    In:  Blood Vol. 118, No. 21 ( 2011-11-18), p. 2423-2423
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 2423-2423
    Abstract: Abstract 2423 Homeobox (Hox) genes have been shown to play critical roles in normal and aberrant hematopoiesis, however, little is known about the role of Hoxa1. Unlike most Hox genes, Hoxa1 is expressed as two different isoforms: full length Hoxa1 (FL-Hoxa1) and a truncated form, Hoxa1-T, resulting from alternative splicing within exon 1 of the gene. The two isoforms are expressed differentially in immature hematopoietic cells. Overexpression of FL-Hoxa1 and Hoxa1-T in mouse bone marrow (BM) cells significantly increased, and significantly decreased, respectively, cell proliferation in vitro compared to control (MXIE) transduced BM cells. These findings suggested that Hoxa1-T may negatively regulate FL-Hoxa1. We therefore generated a mutant Hoxa1 (muHoxa1) that expresses FL-Hoxa1 but was no longer capable of generating Hoxa1-T by changing the AGC serine to a TCT serine at the splice site of Hoxa1-T. Hoxa1 levels in muHoxa1-overexpressing BM were significantly higher than those in FL-Hoxa1 BM. Furthermore, BM cells overexpressing muHoxa1 had higher proliferative potential in vitro than FL-Hoxa1-overexpressing BM. Their in vivo potential was therefore assessed. At 12 weeks post-transplant, all primary transplant recipients had similar %GFP expression in the peripheral blood (PB), being approximately 10%. At this time point all recipients of muHoxa1-overexpressing BM cells displayed thrombocytopenia (mean ± SEM PB platelets (x 106/ml): MXIE= 1062 ± 75; FL-Hoxa1= 1053 ± 146; muHoxa1= 681 ± 71*, *P 〈 0.05 vs MXIE). Secondary transplants were performed into irradiated and non-irradiated recipients. Strikingly, the %GFP+ve cells were markedly increased in the PB of recipients of muHoxa1 BM (%GFP: MXIE: 0.29 ± 0.06; FL-Hoxa1: 3.33 ± 1.38; muHoxa1: 27.46 ± 8.35*; *P 〈 0.05 vs MXIE and FL-Hoxa1). All secondary recipients of muHoxa1 BM developed myeloid neoplasias, resembling myelodysplastic syndrome (MDS). The thrombocytopenia persisted (PB platelets (x 106/ml): MXIE: 923 ± 42; FL-Hoxa1: 812 ± 38; muHoxa1: 388 ± 108*; *P 〈 0.001 vs MXIE and FL-Hoxa1) and secondary recipients of muHoxa1 BM developed anemia (PB Hb (g/L): MXIE: 143 ± 2.6; FL-Hoxa1: 146 ± 2.6; muHoxa1: 117 ± 5.2*, *P 〈 .0005 vs MXIE and FL-Hoxa1). The PB leukocyte counts in the majority of muHoxa1 recipients were unchanged compared to MXIE and FL-Hoxa1 recipients, however, muHoxa1 PB cells were predominantly granulocytes. These neoplasms also occurred in non-irradiated recipients of muHoxa1 BM, although they had a much longer latency. Interestingly, 40% of the non-irradiated recipients developed acute myeloid leukemia between 6 and 12 months post-transplant. Importantly, integration site and cytogenetic analysis demonstrated that the malignant phenotype was not due to co-operating insertional mutagenesis or chromosomal instability in the transduced cells. The PB anemia was accompanied by a significant two-fold reduction in Ter119+ cells in BM of muHoxa1-overexpressing cells (GFP+ve= 16.3± 5.3%, GFP-ve= 34.8 ± 5.1%, P 〈 0.05). This was due to an accumulation of the cells in early stages of erythroid differentiation, with increased proportions of proerythroblasts (GFP+ve: 36.6 ± 7.6%; GFP-ve: 8.6 ± 1.3% P 〈 0.002) and basophilic erythroblasts (GFP+ve: 34.3 ± 3.4%; GFP-ve: 13.2 ± 3.5%, P 〈 0.001) in muHoxa1 Ter119+ cells. The block in erythroid differentiation was also accompanied by significant alterations in the proportions of immature progenitors in recipients of muHoxa1 BM. GFP+ve cells were detectable in HSC and myeloid progenitor cell subsets, however, there was a significant increase in the proportion of megakaryocyte erythroid progenitors (MEPs) within the lineage negative, c-kit+, Sca-1 negative progenitor cell fraction (GFP+ve: 42.9 ± 2.1%; GFP-ve: 8.2 ± 1.3%, P 〈 0.01). Significantly reduced GATA-1 expression was observed in both the GFP+ve proerythroblasts (400-fold) and MEPs (100-fold) compared to GFP-ve populations sorted from the same mice (P 〈 0.001). Taken together, these results suggest that overexpression of FL-Hoxa1 in the absence of Hoxa1-T results in the development of MDS. This is partly due to an accumulation of MEPs and impaired differentiation of erythrocytes, both of which have significantly downregulated expression of GATA-1. Given the striking similarities in hematological phenotype to human patients with MDS, this novel mouse model will be invaluable in identifying the mechanisms contributing to this disease. 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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...