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: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 4402-4402
    Abstract: Abstract 4402 It is well established that patients with congenital neutropenia due to mutations of ELANE or HAX1 have a strongly increased risk of developing secondary leukemia. In contrast, the leukemia risk is not strongly increased in patients with specific other neutropenia subtypes (e. g. cyclic neutropenia with ELANE mutations). Here we report the first glycogen storage disease type Ib (GSD1b) patient in our registry who developed acute myelogenous leukemia (AML). GSD1b is an autosomal recessive disorder of glycogen metabolism with intracellular accumulation of glycogen caused by mutations in the glucose-6-phosphate translocase gene. Patients present with hepatomegaly, growth retardation, hypoglycemia, lactic acidosis as well as neutropenia and neutrophil dysfunction predisposing to bacterial infections. G-CSF has been shown to be an effective long-term treatment for GSD1b patients leading to an increase in neutrophil counts and enhancement of neutrophil function resulting in fewer infections and improved quality of life. The European Branch of the Severe Chronic Neutropenia International Registry (SCNIR) has collected long-term data from 23 patients with GSD1b. Median age was 17 years, 3 patients expired from infections at an age of 8; 9 and 26 years, respectively. All 23 patients received G-CSF treatment for a median duration of 9.5 years, 22 patients received continuous G-CSF with a median dose of 3.4 mcg/kg/day (range 0.8 to 5 mcg/kg/day), one patient received G-CSF interventionally only. Our patient was diagnosed with AML FAB classification M2 linked to cytogenetic aberrations involving trisomy 21. Leukemia cells also harboured a missense RUNX 1 mutation in exon 3. G-CSF treatment was initiated at age 5 months at a dose of 2.3 mcg/kg/d and was continued until AML diagnosis at age 10 years. The G-CSF dose was increased due to low ANCs and infectious episodes with otitis media and pneumonitis up to 4.05 mcg/kg/day. After initial AML treatment with a hypometylating chemotherapy consisting of azacytidine and cytarabine she developed partially reversible renal insufficiency and oxygen dependent pneumonitis which led to a reduction of chemotherapy despite persistence of AML blasts. Remission was achieved with the use of mylotarg. Subsequently, she received a conditioning regimen with melphalan, fludarabine and treosulfan plus ATG-Fresenius followed by infusion of 10/10 HLA matched peripheral blood stem cells from an unrelated donor. Leukocyte engraftment occurred on day +18 followed by neutrophil engraftment on day +21. Post-SCT she developed grade III skin GvHD on day +8 which was responsible to steroid treatment. Discussion: Up to date 3 GSD1b patients with development of AML have been reported in the literature. Two patients were on long-term G-CSF treatment (Pinsk et al. J Pediatr. 2002; Schroeder et al. J Medical Case Reports 2008), one patient transformed predating the availability of G-CSF (Simmons et al. J Pediatr. 1984). Conclusion: According to the literature, leukemic transformation in patients with GSD1b is a rare event. However, with a total of four patients reported with secondary AML with or without G-CSF treatment, GSD1b patients may have a higher leukemia risk than previously anticipated. Independent from G-CSF treatment, regular bone marrow examinations (morphology, cytogenetics, mutation analysis of candidate genes such as RUNX1) may help to detect malignant transformation early and therefore avoid chemotherapy prior to SCT. SCT is a treatment option for AML in GSD1b, but further data is required. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2012
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 3347-3347
    Abstract: Severe chronic neutropenia is a group of rare disorders including inborn genetic defects (congenital neutropenia;CN and cyclic neutropenia; CyN) and acquired diseases associated with severe chronic neutropenia. CN includes a variety of genetic subgroups and is well known as one of the premalignant bone marrow failure syndromes with an overall incidence of secondary leukemia of more than 10 percent. Since the availability of G-CSF for the treatment of severe chronic neutropenia patients the life expectancy and quality of life has improved significantly. Therefore, stem cell transplantation is to date limited to few indications, e. g. G-CSF treatment non-response or secondary leukemia. Here we report on the outcome of stem cell transplantations in 71 patients (70 CN, 1 CyN) documented by the SCNIR since 1994. Results Reason for SCT was myelodysplastic syndrome or secondary leukemia in 34 CN and 1 CyN patients (LEUK+), G-CSF treatment non-response in 17 CN patients and other non-malignant events (e.g. G-CSF receptor mutation) in 19 CN patients (LEUK-). In 56 of 71 patients genetic testing was available revealing 28 ELANE (11 LEUK+; 17 LEUK-), 7 HAX1 (6 LEUK+; 1 LEUK-), 7 SBDS (5 LEUK+; 2 LEUK-), 1 G6PT (LEUK+), 1 G6PC3 (LEUK-) 1 p14 (LEUK-), digenic mutation (HAX1 plus G6PC3) in 1 (LEUK-) and no gene mutation in 10 patients (6 LEUK+; 4 LEUK-). In 13 CN (7 Leuk+/6 Leuk-)and the CyN (Leuk+) patient no genetic testing was performed. For further analysis patients were divided by SCT indication (35 LEUK+ versus 36 LEUK-) and both cohorts by year of SCT (before and since 2001) to acknowledge differences in SCT outcome over time due to improved HLA-typing and concomitant treatment (e. g. GvHD prophylaxis) and a change in SCT strategy in the LEUK+ cohort by introducing a new SCT protocol in 2001. Patients are characterized as follows: LEUK+ prior to 2001 (n=11):5/6 male/female. Median age at SCT 10 years. Median follow-up of 0.54 years (max. 13.2 years). Median time between leukemia diagnosis to SCT 6.5 months. Median G-CSF dose 4.5 mcg/kg/day. LEUK+ since 2001 (n=24):9/15 male/female. Median age at SCT 12 years. Median follow-up of 4.5 years (max. 10.42 years). Median time between leukemia diagnosis to SCT 2.5 months. Median G-CSF dose 15.11 mcg/kg/day. LEUK- prior to 2001 (n=9):4/5 male/female. Median age at SCT 8 years. Median follow-up of 10.9 years (max. 27.2 years). Median G-CSF dose 24.5 mcg/kg/day. LEUK- since 2001 (n=27):15/12 male/female. Median age at SCT 5 years. Median follow-up of 3.08 years (max. 9.2 years). Median G-CSF dose 9.9 mcg/kg/day. 22 different chemo-conditioning regimens were used. Busulfan based regimens were chosen in the majority of LEUK+ and LEUK- patients independent from HLA-donor and stem cell resource. In the LEUK+ cohort before 2001 SCT was performed mainly in 2ndremission following standard chemotherapy protocols, whereas since 2001 due to the new protocol SCT was performed immediately after leukemia diagnosis without standard anti-leukemic therapy. An increase of survival rate from 27.3% (8 deaths out of 11 patients) to 83.3% (4 deaths in 24 patients) in the LEUK+ cohort is highly significant (p 〈 0,001). In the LEUK- cohortsurvival has improved over time from 66.7% prior 2001 to 77.8% since 2001. Conclusion Secondary leukemia is the major reason for SCT in CN patients followed by G-CSF treatment non-response. Comparing SCT prior 2001 with SCT since 2001, SCT survival has improved in the LEUK- indications by 13 % independent from the conditioning regimens used for SCT and HLA match of SCT donors. These results may indicate improved HLA typing, GvHD prophylaxis and treatment for infectious episodes. In comparison, in the LEUK+ cohort a dramatic improvement was documented with an increase of survival rate from 27 % to 83% indicating the additional impact of a shortened time interval between leukemia diagnosis and SCT in combination with the avoidance of standard chemotherapy treatment prior to SCT by the new SCT protocol. 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 ...
  • 3
    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 ...
  • 4
    Online Resource
    Online Resource
    Springer Science and Business Media LLC ; 2010
    In:  Clinical Reviews in Allergy & Immunology Vol. 38, No. 1 ( 2010-2), p. 68-74
    In: Clinical Reviews in Allergy & Immunology, Springer Science and Business Media LLC, Vol. 38, No. 1 ( 2010-2), p. 68-74
    Type of Medium: Online Resource
    ISSN: 1080-0549 , 1559-0267
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2010
    detail.hit.zdb_id: 2091859-8
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    Elsevier BV ; 2014
    In:  The Lancet Respiratory Medicine Vol. 2, No. 10 ( 2014-10), p. e19-
    In: The Lancet Respiratory Medicine, Elsevier BV, Vol. 2, No. 10 ( 2014-10), p. e19-
    Type of Medium: Online Resource
    ISSN: 2213-2600
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2014
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    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 ...
  • 7
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 439-439
    Abstract: Analysis of patients with severe congenital neutropenia (SCN) may shed light on the delicate balance of factors controlling differentiation, maintenance, and decay of neutrophil granulocytes. Mutations in ELANE, GFI1, HAX1, G6PC3, WAS, and VPS45 are known to cause SCN. We here describe a new monogenetic SCN variant with biallelic mutations in the gene encoding Jagunal homolog 1 (JAGN1). We studied an index family from Northern Africa with a total of 5 children suffering from SCN. An Affymetrix SNP array-based genetic linkage analysis was performed and identified a single interval of perfect segregation with highly significant multi-marker LOD scores of at least 4.5spanning approximately 1.5Mbp from 9.52Mb to 11.04Mb on chromosome 3 of NCBI’s human genome build 36.3. This interval contained a total of 30 genes, including JAGN1 which encodes an ER-resident protein. Sanger sequencing revealed a homozygous mutation c.3G 〉 A in exon 1 of the JAGN1 gene; this mutation leads to disruption of the defined start of translation. Systematic analysis of a cohort of 90 SCN patients identified 9 distinct homozygous mutations in the gene encoding Jagunal homolog 1 (JAGN1) in 14 SCN patients, thus accounting for approximately 10% of SCN patients. The clinical phenotype was variable and included failure to thrive, developmental delay and bone skeletal abnormalities. The only consistent finding in all JAGN1-deficient patients was SCN and partial or complete refractoriness to therapy with rh-GCSF. JAGN1 is the human ortholog of a gene originally identified in Drosophila melanogaster. Jagunal-deficient oocytes are characterized by defective ER reorganization and aberrant membrane trafficking during vitellogenesis. We found that JAGN1-mutant human granulocytes showed aberrantly enlarged ER structures and paucity of secretory vesicles. We hypothesized that that ER aberrations may be associated with defective N-glycosylation of multiple proteins in neutrophil granulocytes and found that JAGN1-mutant neutrophil granulocytes exhibited anomalous N-glycomic profiles characterized by a marked reduction in fucosylation of all their multi-antennary glycans. JAGN1-deficient neutrophil granulocytes showed increased apoptosis in response to TNFa and staurosporine, likely accounting for the lack of mature neutrophils in these patients. Additional studies in JAGN1-knockdown cells indicate that JAGN1 participates in the secretory pathway and is required for granulocyte-colony stimulating factor receptor-mediated signaling. Global proteomic analysis of the JAGN1-interactome identified a limited number of interaction partners including members of the Coat Protein I (COPI) complex (COPA, COPB2, and COPG2) which suggest a role for JAGN1 in vesicular trafficking from Golgi to ER. Taken together, JAGN1 emerges as a hitherto unrecognized factor necessary in differentiation and survival of neutrophil granulocytes and a novel gene implicated in SCN. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 117, No. 3 ( 2011-01-20), p. 1030-1041
    Abstract: Adoptive transfer (AT) of T cells forced to express tumor-reactive T-cell receptor (TCR) genes is an attractive strategy to direct autologous T-cell immunity against tumor-associated antigens. However, clinical effectiveness has been hampered by limited in vivo persistence. We investigated whether the use of major histocompatibility complex–mismatched T cells would prolong the in vivo persistence of tumor-reactive TCR gene expressing T cells by continuous antigen-driven proliferation via the endogenous potentially alloreactive receptor. Donor-derived CD8+ T cells engineered to express a TCR against a leukemia-associated antigen mediated strong graft-versus-leukemia (GVL) effects with reduced graft-versus-host disease (GVHD) severity when given early after transplantation. AT later after transplantation resulted in a complete loss of GVL. Loss of function was associated with reduced expansion of TCR-transduced T cells as assessed by CDR3 spectratyping analysis and PD-1 up-regulation on T cells in leukemia-bearing recipients. PD-L1 blockade in allogeneic transplant recipients largely restored the GVL efficacy without triggering GVHD, whereas no significant antileukemia effects of PD-L1 blockade were observed in syngeneic controls. These data suggest a clinical approach in which the AT of gene-modified allogeneic T cells early after transplantation can provide a potent GVL effect without GVHD, whereas later AT is effective only with concurrent PD-L1 blockade.
    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 ...
  • 9
    Online Resource
    Online Resource
    American Society of Hematology ; 2011
    In:  Blood Vol. 118, No. 21 ( 2011-11-18), p. 14-14
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 14-14
    Abstract: Abstract 14 Several studies found that in patients with severe congenital neutropenia (CN) harboring mutations in the ELANE gene mutated NE protein induced unfolded protein response (UPR) leading to elevated apoptosis and diminished differentiation of myeloid cells. However, it is unclear, why UPR was not detected in patients with cyclic neutropenia (CyN) carrying the same ELANE mutations, which have been found in CN patients. Several UPR components have been identified in mammalian cells, which include three transducers (IRE1, PERK, and activating transcription factor 6 (ATF-6) as well as one master regulator (BiP/GRP78). BiP is known to be regulated by ATF6. The activation of ATF6 and its target genes (GADD34, CHOP and BiP) in CN patients has not been studied yet. We were able to detect significantly elevated levels of ATF6 and BiP in myeloid cells of CN patients with ELANE mutations, in comparison to CyN patients and to healthy individuals. Therefore, we investigated the mechanism of UPR and activation of ATF6 and ATF6 target genes in CN patients in comparison to CyN patients. We transduced the myeloid cell lines HL60 and NB4 with lentiviral constructs contained either wild type (WT) ELANE cDNA, or mutated (MUT) ELANE cDNA and measured mRNA and protein expression of ATF6 as well as mRNA expression of ATF6 target genes. We compared the effects of three ELANE mutations: C42R, V145-C152del (both mutations presented in CN patients, but not in CyN patients) and S97L (typical for CN and CyN patients) with WT ELANE. We found that in both cell lines only C42R ELANE MUT, but not V145-C152del ELANE MUT or S97L ELANE MUT induced expression of ATF6, GADD34, CHOP and BiP, as compared to control transduced cells. Furthermore, we hypothesize that degradation of mutated NE protein by Secretory Leukocyte Protease Inhibitor (SLPI) might be involved in UPR induction. However, we detected only very low levels of SLPI mRNA in CD33+ myeloid cells and in PMNs of patients with severe congenital neutropenia (CN), as compared to patients with cyclic neutropenia (CyN) and to healthy individuals. The lack of the NE inhibitor, SLPI in CN patients may further contribute to elevated UPR triggered by ELANE MUT and normal levels of SLPI in CyN patients might protect from ELANE MUT-induced UPR. Indeed, inhibition of SLPI using SLPI-specific shRNA led to a significantly elevated expression levels of ATF6, GADD34 and BIP, as compared to ctrl shRNA transduced cells. More importantly, co-transduction of NB4 cells with SLPI shRNA in combination with ELANE S97L MUT (which is common for both CN and CyN patients), but not with WT ELANE led to elevated levels of ATF6, GADD34 and BIP. In summary, different ELANE mutations have different effects on UPR as judged by ATF6 activation and the level of ELANE-triggered UPR is regulated by SLPI. 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 ...
  • 10
    Online Resource
    Online Resource
    American Society of Hematology ; 2010
    In:  Blood Vol. 116, No. 21 ( 2010-11-19), p. 1480-1480
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 1480-1480
    Abstract: Abstract 1480 Severe congenital neutropenia (CN) is a heterogeneous disorder of hematopoiesis characterized by a maturation arrest of granulopoiesis at the level of promyelocytes with peripheral blood absolute neutrophil counts below 0.5 × 109/L. G-CSF treatment increases blood neutrophil counts in more than 90% of individuals with CN. CN is also considered as a pre-leukemic syndrome, since ca. 20% of CN patients develop Acute Myelocytic Leukemia (AML) or Myelodysplastic Syndrome (MDS). Surprisingly no mutations in genes, which typically occur in primary AML/MDS patients, were detected in CN patients who developed leukemia. But studies in CN patients reveal a high association of G-CSF receptor mutation and the incidence of leukemia, indicating the dysregulation of certain factors downstream of G-CSF receptor signalling. Recently, we reported that Nicotinamide Phosphoribosyltransferase (NAMPT), a protein involved in biosynthesis of NAD+, was significantly increased in CN patients treated with G-CSF as compared to healthy individuals. Elevated NAMPT/NAD+ levels correlated with increased levels of SIRT1, a NAD+-dependent deacetylase which is involved in the deacetylation of histone and non-histone proteins e.g. p53. The acetylation of tumor-suppressor p53 is considered necessary for its transcriptional activation, while SIRT1-mediated deacetylation of p53 has been shown to attenuate the transcriptional activity of p53. Therefore, we asked if deacetylation-dependent inactivation of p53 might play a role in leukemic transformation in CN patients. In this study we demonstrate that the presence of NAMPT or NAD+ enhances the SIRT1-mediated deacetylation of p53 in both the 293T cell line and the promyelocytic leukemia NB4 cell line. Treatment with exogenous recombinant NAMPT also leads to a decrease in the acetylation levels of endogenous p53 in CD34+ cells. The cyclin-dependent kinase inhibitor 1A (p21, Cip1) protein is a well-known target of p53 and is involved in cell cycle arrest. We have shown that over-expression of NAMPT leads to down-regulation of p21 mRNA, and specific knockdown of SIRT1 leads to up-regulation of p21 mRNA. The presence of NAMPT also decreases the mRNA levels of p21 in both NB4 and CD34+ cells. The compound FK866 specifically inhibits NAMPT and has recently entered clinical trials as a potential chemotherapeutic agent. In a recent preclinical in vitro study FK866 has been shown to elicit massive cell death in numerous leukemia/lymphoma cell lines, but the underlying molecular mechanism remains unknown. We tested if inhibition of NAMPT using FK866 enhances the tumor-supressing role of p53 by increasing its acetylation levels. We have demonstrated that the treatment of NB4 cells with FK866 increases the acetylation of endogenous p53, and this increased acetylation is in part due to decreased interaction of p53 with SIRT1. In addition, the mRNA levels of p21 down-regulated in CD34+ and NB4 cells on treatment with NAMPT were up-regulated on use of FK866. Knockdown of p53 using specific shRNA against p53 inhibits the expression of p21 and treatment with FK866 under p53 knockdown does not induce the expression of p21 when compared to control cells. In functional studies we show that over-expression of NAMPT leads to increased proliferation of both 293T and NB4 cells. Treatment with FK866 leads to increased death of NB4 cells compared to the cells in which p53 has been silenced, due to the lack of p53 available to be acetylated. Taken together, our conclusion is that NAMPT/NAD+ activated SIRT1 mediates deacetylation of p53 leading to down-regulation of the downstream target gene p21. This inhibition of the tumor-suppressor functions of p53 might be involved in the leukemic transformation seen in CN. 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: 2010
    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...