scholarly journals Bortezomib inhibits STAT5-dependent degradation of LEF-1, inducing granulocytic differentiation in congenital neutropenia CD34+ cells

Blood ◽  
2014 ◽  
Vol 123 (16) ◽  
pp. 2550-2561 ◽  
Author(s):  
Kshama Gupta ◽  
Inna Kuznetsova ◽  
Olga Klimenkova ◽  
Maksim Klimiankou ◽  
Johann Meyer ◽  
...  

Key Points Hyperactivated STAT5a binds LEF-1 protein leading to NLK/NARF/ubiquitin-dependent degradation of LEF-1 followed by defective granulopoiesis. In CN patients, elevated levels of phospho-STAT5a resulted in diminished LEF-1 expression, which could be restored by bortezomib treatment.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 316-316
Author(s):  
Kshama Gupta ◽  
Julia Skokowa ◽  
Karl Welte

Abstract Congenital hematological disorders are excellent models for investigating the regulation of hematopoiesis in humans. For instance, Severe Congenital Neutropenia (CN) is a heterogeneous syndrome characterized by a maturation arrest of granulopoiesis at the level of promyelocytes with no mature neutrophils in the peripheral blood. Even though G-CSF treatment results in increased neutrophil numbers in more than 90 % of CN patients, G-CSF-dependent granulocytic differentiation is severely affected in these patients. CN patients are found to be at increased risk of developing acute myeloid leukemia or myelodysplastic syndrome (AML/MDS) (cumulative incidence ~ 20 %). Since AML/MDS are not observed in cyclic or idiopathic neutropenia patients treated with G-CSF, an underlying defect down stream of G-CSF signaling rather than G-CSF therapy per se predisposes to malignant transformation in CN. STAT5 is activated by G-CSF signaling pathway and has also been found to be activated in AML. Recently we found that downregulation of LEF-1 transcription factor and its target gene C/EBPa are a common pathologic mechanism for CN. Therefore, we investigated the effects of G-CSF on the phosphorylation status of STAT5a in CN and whether it leads to the down modulation of LEF-1 expression and functions. Indeed, we detected elevated phosphoSTAT5 expression in CD34+ cells from CN patients before G-CSF stimulation in vitro, as compared to these cells from healthy individuals. Moreover, treatment with G-CSF resulted in a significantly higher phosphorylation of STAT5a in CN. Intriguingly, levels of phosphoSTAT5 in myeloid blast cells from one CN patient who developed AML was even higher and was in line with undetectable LEF-1 protein expression. Transduction of constitutive active STAT5a (STAT5a 1*6) in CD34+ cells from healthy individuals resulted in significant downregulation of LEF1 levels in a dose dependent manner. A screen of 10 kb upstream region of LEF1 gene revealed two putative STAT5 binding sites (−3891bp to −3909bp and −3714bp to −3732bp) and the specificity of this binding was confirmed in the nuclear extracts of CD34+ cells by chromatin immunoprecipitation assay. We found enhanced and prolonged STAT5a binding to the LEF-1 promoter in G-CSF treated CD34+ cells from CN patients, as compared to healthy individuals. Additionally, transfection of CD34+ cells with LEF-1 cDNA resulted in elevation of LEF-1 promoter activity, which suggests a strong LEF-1 autoregulation. Co-transfection with STAT5a 1*6 significantly disrupted LEF-1- dependent activation of LEF-1 promoter. Moreover STAT5a 1*6 severely abrogated the LEF-1 dependent regulation of C/EBPα gene promoter. Taken together phosphorylation of STAT5 is upregulated in hematopoietic progenitors from CN patients which lead to subsequent down regulation of LEF-1. These downstream effects of activated STAT5a may contribute to the malignant transformation of myelopoiesis in CN.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. SCI-45-SCI-45
Author(s):  
Julia Skokowa

Abstract Clinical observations revealed that congenital neutropenia (CN) patients harboring either ELANE or HAX mutations have similar bone marrow morphology, responses to G-CSF therapy, requirements of G-CSF dosages, and the risk of developing leukemia. Therefore, we suggested a common pathomechanism of defective G-CSFR-triggered granulopoiesis downstream of both mutated genes in these patients. We identified severely diminished expression and functions of the transcription factors lymphoid enhancer binding factor-1 (LEF-1) and C/EBPa in myeloid cells of CN patients, in comparison to healthy individuals and patients with cyclic neutropenia (CyN). LEF-1 expression was abrogated in patients harboring either ELANE or HAX1 mutations, which suggested LEF-1 as a possible common candidate factor for defective G-CSFR signaling. We further identified a mechanism of the diminished LEF-1 expression downstream of HAX1 or ELANE mutations. HAX1 is HCLS1-Associated protein X1. HCLS1 is Hematopoietic Cell-Specific Lyn Substrate 1. We found that HCLS1 protein is expressed at high levels in human myeloid cells and is phosphorylated upon stimulation with G-CSF. HCLS1 interacted with LEF-1 protein, inducing nuclear translocation of LEF-1, LEF-1 autoregulation, C/EBPa activation, and granulocytic differentiation. In CN patients with HAX1 mutations, we found profound defects in the G-CSF-triggered phosphorylation of HCLS1, subsequently leading to abrogated nuclear transport and autoregulation of LEF-1. In CN patients with ELANE mutations we detected severely reduced levels of the natural inhibitor of neutrophil elastase (NE), and secretory leukocyte protease inhibitor (SLPI). We demonstrated the important role of SLPI in myeloid differentiation by activation of Erk1/2 phosphorylation and subsequent phErk1/2-triggered tyrosine phosphorylation and activation of the LEF-1 protein. Therefore, the direct link between ELANE mutations and diminished LEF-1 expression was established: in these patients LEF-1 protein expression is diminished due to the reduced levels of SLPI. We further evaluated how G-CSF treatment overcomes maturation arrest of granulopoiesis in CN patients despite the absence of LEF-1 and C/EBPa in myeloid cells. We identified nicotinamide phosphoribosyltransferase (NAMPT) as an essential enzyme mediating G-CSF-triggered granulopoiesis in healthy individuals and in CN patients. Treatment of healthy individuals with G-CSF resulted in upregulation of NAMPT levels in myeloid cells and in plasma. NAMPT and NAD+ amounts were even more dramatically elevated by G-CSF treatment of CN individuals. The molecular events triggered by NAMPT included elevation of NAD+, NAD+-dependent activation of protein deacetylase sirtuin-1 (SIRT1), binding of SIRT1 to the myeloid specific transcription factors C/EBPα and C/EBPβ, and activation of these transcription factors. In CN patients, C/EBPα expression is severely diminished; therefore “steady-state” granulopoiesis could not be activated. G-CSF treatment induces expression of C/EBPβ in these patients via NAMPT and SIRT1 and operated via the “emergency” pathway. We also investigated the patterns of acquisition of leukemia-associated-mutations in 31 CN patients developing leukemia using next-generation DNA deep sequencing. Intriguingly, 20 of the 31 patients (64.5%) demonstrated mutations within RUNX1. The majority of patients with RUNX1 mutations (85%) had acquired CSF3R mutations. Other leukemia-associated mutations in the patients with RUNX1 mutations were found infrequently. Cytogenetics of the leukemic cells revealed that 10 patients with RUNX1 mutations developed monosomy 7, and six patients had trisomy 21. Single cell analysis in two patients revealed that RUNX1 and CSF3R mutations were segregated in the same malignant clone. Functional studies demonstrated proliferative advantage of CD34+ cells transduced with mutated RUNX1 and CSF3R. By analysis of the leukemogenic role of the defective G-CSFR signaling in CN patients we identified a significant and sustained elevation in the levels of phospho-STAT5 in hematopoietic CD34+ cells of CN patients which were even higher in CN/ acute myeloid leukemia patients. The other possible reason for the leukemogenic transformation could be elevated NAMPT/SIRT-triggered deacetylation of tumor supressor protein p53, proto-oncogene FOXO3a and Akt proteins. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 123 (14) ◽  
pp. 2229-2237 ◽  
Author(s):  
Julia Skokowa ◽  
Doris Steinemann ◽  
Jenny E. Katsman-Kuipers ◽  
Cornelia Zeidler ◽  
Olga Klimenkova ◽  
...  

Key Points CN/AML patients have a high frequency of CSF3R and RUNX1 mutations. CSF3R and RUNX1 mutations induce elevated proliferation of CD34+ cells.


Blood ◽  
2014 ◽  
Vol 123 (8) ◽  
pp. 1239-1249 ◽  
Author(s):  
Olga Klimenkova ◽  
Wienke Ellerbeck ◽  
Maksim Klimiankou ◽  
Murat Ünalan ◽  
Siarhei Kandabarau ◽  
...  

Key Points The natural inhibitor of neutrophil elastase, SLPI, is severely reduced in severe congenital neutropenia patients. SLPI controls myeloid differentiation by regulation of NFκB, ERK1/2:LEF-1, and c-myc activation.


Blood ◽  
2009 ◽  
Vol 114 (14) ◽  
pp. 3044-3051 ◽  
Author(s):  
Julia Skokowa ◽  
John Paul Fobiwe ◽  
Lan Dan ◽  
Basant Kumar Thakur ◽  
Karl Welte

Abstract Severe congenital neutropenia (CN) is a heterogeneous disorder of myelopoiesis which follows an autosomal dominant or autosomal recessive pattern of inheritance. Genetic analyses indicate mutations in the ELA2 gene in most patients. We have identified LEF-1 as a decisive transcription factor in granulopoiesis controlling proliferation and granulocytic differentiation by direct activation of its target gene, C/EBPα. In patients with CN, the expression of LEF-1 and C/EBPα was abrogated in myeloid progenitors leading to maturation arrest of granulopoiesis. In the present study we demonstrated that ELA2 mRNA expression in myeloid progenitors and plasma protein levels of neutrophil elastase (NE) were markedly reduced in patients with CN harboring mutations in either ELA2 or HAX-1 genes. The ELA2 gene promoter is positively regulated by the direct binding of LEF-1 or C/EBPα, documenting the role of LEF1 in the diminished ELA2 expression. We found that transduction of hematopoietic cells with LEF-1 cDNA resulted in the up-regulation of ELA2/NE synthesis, whereas inhibition of LEF-1 by shRNA led to a marked reduction in the levels of ELA2/NE. LEF-1 rescue of CD34+ cells isolated from 2 patients with CN resulted in granulocytic differentiation of the cells which was in line with increased levels of functionally active ELA2/NE.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 433-433
Author(s):  
Julia T Warren ◽  
Peeradol Wattanasirakul ◽  
David H Spencer ◽  
Adam Locke ◽  
Vahagn Makaryan ◽  
...  

Severe congenital neutropenia (SCN) is an inborn disorder of granulopoiesis characterized by severe chronic neutropenia from birth, premature death secondary to infectious complications, and transformation to myeloid malignancy. Although many cases of SCN are associated with mutations in ELANE, encoding the neutrophil elastase, roughly one-third of cases do not have an identifiable genetic cause. In collaboration with the Severe Chronic Neutropenia International Registry (SCNIR), we performed exome sequencing on 90 cases of congenital neutropenia. Heterozygous missense mutations of CLPB were identified in six patients with SCN. None of these patients had mutations in other genes known to cause SCN. A total of 5 different mutations were identified that clustered within the ATPase domain. Of note, all of these mutations were predicted to be functionally deleterious and had a frequency of <0.002% in the ExAC and gnomAD databases. We subsequently identified heterozygous CLPB mutations in an additional 3 cases of SCN that were not part of our original cohort. Prior studies showed that biallelic mutations of CLPB are associated with a syndrome defined by 3-methylglutaconic aciduria (3-MGA), cataracts, neurologic disease, and variable neutropenia. In our original cohort of 6 SCN patients with heterozygous CLPB mutations, 3-MGA was not present in the three cases where urine samples were available and none had reported cataracts. In total, 3 had neurologic abnormalities (2 seizures and 1 developmental delay). Of note, the CLPB mutations present in syndromic cases were distinct from those seen in our SCN cohort, and none of the mutations in our series are biallelic. CLPB encodes for caseinolytic peptidase B, a protein implicated in protein folding in bacteria and yeast but with an unknown role in human granulopoiesis. Based on these observations, we hypothesize that CLPB is required for normal basal granulopoiesis and that the heterozygous CLPB mutations identified in our study act in a dominant-negative fashion to disrupt granulopoiesis. To test this hypothesis, two complementary genetic approaches were employed. First, we used CRISPR-Cas9 gene editing to generate null mutations in CLPB in human cord blood-derived CD34+ hematopoietic stem/progenitor cells (HSPCs). Using this approach, we are able to achieve greater than 90% editing efficiency as assessed by next generation sequencing. The genetically modified HSPCs were cultured for 14 days under conditions that promote granulocytic differentiation. Modified HSPCs were also seeded into methylcellulose cultures to measure CFU-G. The percentage and absolute number of mature granulocytes, but not early granulocytic precursors (promyelocytes and myelocytes), were significantly reduced in cultures of gene-edited cord blood CD34+ cells. Moreover, the frequency of edited cells decreases over time in our culture system indicating that CLPB-knockout cells have a competitive disadvantage. A significant decrease in CFU-G also was observed. Second, we generated lentivirus expressing all 5 of the neutropenia-associated heterozygous CLPB mutations identified in our SCN cohort (N496K, E557K, R561G, R603H, and R620C). Expression of all of these mutants (except R603H) in cord blood-derived CD34+ cells was associated with a significant decrease in mature neutrophils and corresponding increase in early granulocytic precursors. These four CLPB mutants also resulted in a decrease in CFU-G. Collectively, these data strongly suggest that heterozygous mutations of CLPB are a new cause of congenital neutropenia. Indeed, in the North American population, CLPB mutations appear to be the second most common cause of congenital neutropenia, behind ELANE mutations. Studies are underway to examine the molecular mechanisms by which mutant CLPB disrupts granulopoiesis. Disclosures Dale: Coherus: Consultancy; Beheringer/Ingelheim: Consultancy; Athelas: Equity Ownership; Amgen: Consultancy, Research Funding; Sanofi Aventis: Consultancy, Honoraria; Cellerant: Other: Scientific Advisory Board; Hospira: Consultancy; Prolong: Consultancy; x4pharma: Consultancy, Honoraria, Research Funding.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 500-500 ◽  
Author(s):  
Julia Skokowa ◽  
Murat Uenalan ◽  
Axel Schambach ◽  
Michaela Scherr ◽  
Matthias Eder ◽  
...  

Abstract Lymphoid enhancer-binding factor 1 (LEF-1) belongs to the canonical Wnt signaling pathway acting in transcriptional complexes with β-catenin. LEF-1 can also act independent of β-catenin (i.e. in the TGF-β or Notch pathway). Additionally, recent studies described LEF-1 dominant negative isoform (dnLEF-1), which lack the β-catenin binding domain and functions as either a transcriptional repressor or activator. To date, analysis of the role of LEF-1 in hematopoiesis has been restricted to the lymphoid compartment. Previously we described the crucial role of lymphoid enhancer-binding factor 1 (LEF-1) in granulopoiesis. We found that LEF-1 mediates proliferation, survival, and differentiation of granulocyte progenitor cells. Moreover, abrogated LEF-1 expression is one of the pathomechanism of severe congenital neutropenia CN (Skokowa et al., Nature Medicine, in press). Based on these findings, we aimed to characterize the molecular mechanisms of LEF-1 in the regulation of granulocytic differentiation. C/EBPα is well known as a key transcription factor in granulopoiesis and we found it to be a target gene directly regulated by LEF-1. A screen of the known 566 bp upstream promoter of C/EBPα gene revealed a putative LEF-1 binding site (− 559 bp to − 538 bp). We confirmed LEF-1 binding to C/EBPα promoter in nuclear extracts from CD34+ and CD33+ cells in the transcription factor binding NoShift and ChIP assays. Interestingly, LEF-1 binds to the C/EBPα promoter more efficiently in CD33+ myeloid progenitors than in CD34+ cells. The direct regulation of C/EBPα by LEF-1 was further confirmed in functional studies. We found that in line with down-regulation of LEF-1, expression of C/EBPα was also significantly reduced in CD33+ myeloid progenitors of CN patients. Moreover, LEF-1 rescue of these cells resulted in a marked up-regulation of C/EBPα mRNA expression and in vitro restoration of defective granulocytic differentiation. Remarkably, transduction of CN CD33+ cells with dnLEF-1 isoform, which lacks the ß-catenin-binding domain, resulted in up-regulation of C/EBPα to a similar degree as it was observed with full-length LEF-1. A direct regulatory link between LEF-1 and C/EBPα was additionally confirmed in LEF-1 inhibition experiments. C/EBPα expression was significantly down-regulated in CD34+ cells of healthy individuals, transduced with LEF-1 shRNA. Therefore, we propose that LEF-1 is a key regulator of myeloid differentiation acting in a β-catenin-independent manner, similar as it is known for LEF-1 regulation of T-lymphocyte development.


Blood ◽  
2021 ◽  
Author(s):  
Julia T Warren ◽  
Ryan R Cupo ◽  
Peeradol Wattanasirakul ◽  
David Spencer ◽  
Adam E Locke ◽  
...  

Severe congenital neutropenia (SCN) is an inborn disorder of granulopoiesis. Approximately one-third of cases do not have a known genetic cause. Exome sequencing of 104 persons with congenital neutropenia identified heterozygous missense variants of CLPB (caseinolytic peptidase B) in 5 SCN cases, with 5 more cases identified through additional sequencing efforts or clinical sequencing. CLPB encodes an adenosine triphosphatase (ATPase) implicated in protein folding and mitochondrial function. Prior studies showed that biallelic mutations of CLPB are associated with a syndrome of 3-methylglutaconic aciduria, cataracts, neurologic disease, and variable neutropenia. However, 3-methylglutaconic aciduria was not observed and, other than neutropenia, these clinical features were uncommon in our series. Moreover, the CLPB variants are distinct, consisting of heterozygous variants that cluster near the ATP-binding pocket. Both genetic loss of CLPB and expression of CLPB variants results in impaired granulocytic differentiation of human hematopoietic progenitors and increased apoptosis. These CLPB variants associate with wildtype CLPB and inhibit its ATPase and disaggregase activity in a dominant-negative fashion. Finally, expression of CLPB variants is associated with impaired mitochondrial function but does not render cells more sensitive to endoplasmic reticulum stress. Together, these data show that heterozygous CLPB variants are a new and relatively common cause of congenital neutropenia and should be considered in the evaluation of patients with congenital neutropenia.


Blood ◽  
2014 ◽  
Vol 124 (13) ◽  
pp. e21-e32 ◽  
Author(s):  
Ruggiero Norfo ◽  
Roberta Zini ◽  
Valentina Pennucci ◽  
Elisa Bianchi ◽  
Simona Salati ◽  
...  

Key Points Differential gene and miRNA expression analysis in PMF granulocytes identifies new biomarkers and putative therapeutic targets. Activation of the miR-155/JARID2 axis in PMF CD34+ cells results in overproduction of MK precursors.


Blood ◽  
2018 ◽  
Vol 132 (12) ◽  
pp. 1225-1240 ◽  
Author(s):  
Andrea Pellagatti ◽  
Richard N. Armstrong ◽  
Violetta Steeples ◽  
Eshita Sharma ◽  
Emmanouela Repapi ◽  
...  

Key Points RNA-seq analysis of CD34+ cells identifies novel aberrantly spliced genes and dysregulated pathways in splicing factor mutant MDS. Aberrantly spliced isoforms predict MDS survival and implicate dysregulation of focal adhesion and exosomes as drivers of poor survival.


Sign in / Sign up

Export Citation Format

Share Document