scholarly journals CRISPR/Cas9 Mediated ELANE Knock-out Restores Survival and Granulocytic Differentiation of HL60 Cells Expressing Mutant Neutrophil Elastase: Is Neutrophil Elastase a Dispensible Granulocyte Protease?

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 435-435
Author(s):  
Vahagn Makaryan ◽  
Isabella N. Archibald ◽  
Merideth L. Kelley ◽  
Breanna Fletcher ◽  
David C. Dale

Background : Mutations in ELANE are the most common cause of both cyclic and severe congenital neutropenia. ELANE encodes neutrophil elastase (NE), a tissue specific serine protease expressed primarily in neutrophils. Expression of the mutant protein impairs survival and maturation of myeloid precursors in bone marrow. More than 130 different ELANE mutations have been found in patients with cyclic and congenital neutropenia, and genotype-phenotype studies suggest that specific mutations cause more severe disease. (Curr Op Hematol. 2015;22:3-11) Mutant NE is also implicated as the primary cause of ELANE associated neutropenia by studies showing that cell permeable inhibitors of NE correct the defect in cell survival and maturation in cellular models. (Makaryan et al. J Leukoc Biol. 2017;102:1143). Hypothesis: If inhibition of NE can correct the cellular defect, deletion of ELANE will have similar effects and permit studies to see if NE is an essential neutrophil protease. Methods: We used CRISPR/Cas9 mediated gene editing to create a cellular model of congenital neutropenia in the commercially available human promyelocytic cell line, HL60. We used CRISPR/Cas9 editing technology to create engineered HL60 cell lines with knock-in (KI) ELANE P139L, C151Y and G214R single point heterozygous mutations. Using the same technology, we then performed ELANE gene complete knock out (KO) of ELANE in each mutant cell line. All engineered cell lines as well as wild-type HL60 cells were cultured for 5-7 days in complete RPMI supplemented with 2uM all-trans retinoic acid (ATRA) to trigger myeloid differentiation. Survival of these cell lines was investigated using Annexin V-PE staining and flow cytometric analysis. Granulocytic differentiation was evaluated using CD11b surface marker staining and flow cytometry and by performing manual differential cell counts. We measured the unfolded protein response (UPR) by western blotting using UPR specific antibodies, phagocytosis with E. coli particles using fluorescence detection, IL-8 stimulated chemotaxis in trans-well system and PMA activated respiratory burst by flow cytometry. Results: Cells expressing the P139L and C151Y mutant ELANE had increased Annexin V staining more than a 2-fold increase in apoptotic cells at 7 days in culture. Granulocytic differentiation, measured by surface CD11b expression, was significantly impaired. (p<0.0001). Cytospins stained with Diff-Quik showed a typical block of myeloid differentiation and a significant deficiency of mature neutrophils in 7-day cultures. Western blot analysis using antibodies to GRP78/BiP and ATF6 showed a typical UPR signature in both ELANE mutant cell lines compared to wild type. Chemotaxis to the IL8 chemo-attractant was severely impaired in cells expressing mutant NE. Respiratory burst and phagocytosis was also altered in cells expressing mutant NE. ELANE KO corrected all these cellular and functional abnormalities, reverting these functions toward the wild phenotype. Studies are in progress for the third cell line expressing the G214R mutation. Conclusions: CRISPR/Cas9 engineered HL60 cell lines expressing mutant NE are a highly reproducible and reliable cellular model for investigating ELANE associated neutropenia. The presence of mutant NE severely impairs neutrophil functions, and its deletion through KO of ELANE corrects the cellular abnormalities and reverses the induced abnormalities. These results suggest that CRISPR/Cas9 mediated genetic knock down of NE is a novel therapeutic approach for treatment of this disorder. This data also suggests that total absence of NE does not alter basic functions of granulocytes. Disclosures Dale: Sanofi Aventis: Consultancy, Honoraria; Athelas: Equity Ownership; Amgen: Consultancy, Research Funding; Cellerant: Other: Scientific Advisory Board; Hospira: Consultancy; Prolong: Consultancy; Beheringer/Ingelheim: Consultancy; Coherus: Consultancy; x4pharma: Consultancy, Honoraria, Research Funding.

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3683-3683
Author(s):  
Vahagn Makaryan ◽  
Breanna Fletcher ◽  
Merideth L. Kelley ◽  
Masoud Nasri ◽  
Julia Skokowa ◽  
...  

Abstract Background: Mutations in ELANE are the most common cause of both cyclic and severe congenital neutropenia. Congenital neutropenia is characterized by low neutrophil counts in peripheral blood and impaired survival and maturation of myeloid precursors in bone marrow. G-CSF and HSCT are currently the only effective treatment options. To date, more than 100 different ELANE mutations have been reported. We previously described that some mutations (e.g. G214R and C151Y) are associated with more severe outcomes, while others (e.g. P139L and R220Q) result in relatively mild clinical phenotypes. (Curr Op Hematol. 2015;22:3-11) To explain the biological effects of various ELANE mutants we have created CRISPR/Cas9 edited ELANE mutants using the commercially available human promyelocytic cell line HL60. Methods: We used CRISPR/Cas9 editing technology to create engineered HL60 cell lines with ELANE P139L and C151Y single point heterozygous mutations. Both ELANE mutant cell lines as well as wild-type HL60 cells were cultured for 7 days in complete RPMI supplemented with 2uM all-trans retinoic acid (ATRA) to trigger myeloid differentiation. Survival of these cell lines was investigated using Annexin V-PE staining and flow cytometric analysis. Granulocytic differentiation was evaluated using CD11b surface marker staining and flow cytometry and by performing manual differential cell counts. Unfolded protein response (UPR) was measured by western blotting using UPR specific antibodies. MK-0339 is a potent, cell permeable, orally absorbed inhibitor of neutrophil elastase (NE), previously investigated in preclinical and clinical studies by Merck/DuPont as a potential anti-inflammatory drug. We have recently reported that MK-0339 increases cell survival and myeloid differentiation in cellular models of ELANE associated neutropenia. (Makaryan, et al, J Leukoc Biol. 2017;102(4):1143-1151). We examined the effects of MK-0339 on these cell lines. Results: Annexin V staining showed more than 2-fold increase in apoptotic cells in both mutant cell lines compared to wild-type. Granulocytic differentiation measured by surface CD11b expression was significantly impaired in both mutant cell lines (p-values <0.0001). Cytospins stained with Diff-Quik showed a typical block of myeloid differentiation and a significant deficiency of mature myelocytes. Western blot analysis using antibodies to GRP78/BiP and ATF6 showed a typical UPR signature in both ELANE mutant cell lines compared to wild type. It is important to note that the C151Y mutant, the mutant clinically associated with more severe disease, shows more severe impairment compared to P139L. Addition of 1uM MK-0339 to the culture completely restored normal survival and myeloid differentiation of both mutant cell lines. Conclusions: We believe CRISPR/Cas9 engineered HL60 cell lines expressing mutant NE are a highly reproducible and reliable cellular model for investigating genetic neutropenias. These results suggest that a panel of different mutant ELANE HL60 cell lines will help to elucidate the molecular and biochemical origin of phenotypic variability in ELANE associated neutropenia. Disclosures Dale: Athelas, Inc.: Equity Ownership; Amgen: Consultancy, Research Funding; Sanofi-Aventi: Consultancy, Honoraria; Cellerant: Other: Scientific Advisory Board; Hospira: Consultancy; Prolong: Consultancy; Beheringer-Ingelheim: Consultancy; Coherus: Consultancy.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 406-406
Author(s):  
Vahagn Makaryan ◽  
Merideth L. Kelley ◽  
Breanna Fletcher ◽  
David C. Dale

Abstract Background : Mutations in ELANE, the gene for neutrophil elastase (NE), are the most frequent cause of both cyclic neutropenia (CyN) and severe congenital neutropenia (SCN). G-CSF and HSCT are currently the only effective treatment options. We have examined the possibility that inhibitors of NE might also be treatment options. Methods: Cells: HL60 human promyelocytic cells, a commercially available cell line, and induced pluripotent stem cells (iPSc) derived from patient dermal fibroblasts or bone marrow stromal cells, reprogrammed using episomal vectors. NE inhibitors: Two cell permeable inhibitors were used: MK0339 provided by Merck and sivelestat from Sigma. Results: Expression of mutant NE in HL60 Cells Transient Expression:Transient transfection of HL60 cells with 4 different ELANE mutants (P139L, C151Y, V174_C181del, G214R) cloned in pcDNA 3.1 vector showed increased apoptosis after 48h of culture in 10% FBS RPMI and expression of mutant constructs. Addition of Merck NE inhibitor MK0339 with 1uM final concentration in culture significantly improved survival of cells expressing each of the mutations tested detected by Annexin V staining using flow cytometry. (p<0.0001) The effect of the sivelestat (100nM) was significantly effective on only G214R mutant. (p= 0.035) Regulated Expression:We tested the effects of Merck inhibitor MK0339 on tet-inducible HL60 cells with regulated expression of mutant NE (V174_C181del), a mutation found in a patient with severe congenital neutropenia who died of AML. We examined 3 characteristics of these cells: cell survival, myeloid differentiation, and proliferation. The cell lines, expressing wild type or mutant NE under a tetracycline regulated promoter, were treated with MK0339 and cultured for 72 hours in the presence or absence of doxycycline. MK0339 significantly reduced mutant NE triggered apoptosis and increased granulocytic differentiation induced by addition of all-trans retinoic acid (ATRA) in 5-day cultures. Expression of mutant NE also significantly reduced the growth rate of the HL60 cells, but addition of MK0339 promoted growth of these HL60 cells. Induced Pluripotent Stem Cells (iPSC) We have established iPS cell lines from three neutropenic patients (two brothers with P139L and another patient with G214R) and two healthy volunteers. Cell lines from all 3 patients consistently grew much slower and formed much smaller colonies than the controls using both mTeSR1/matrigel and DEF-CS feeder free culture medium systems. We observed more than 3-fold growth reduction of patient derived lines compared to healthy volunteers. We therefore tested the effects of MK-0339 and sivelestat under the same culture conditions. Growth of patient cell lines was enhanced and the cells appeared more robust, especially after addition of 1uM MK0339. The latter normalized patient derived iPSc proliferation. We also investigated survival of the iPS cells during growth in feeder free settings and the effect of the NE inhibitors. After 4 days of culture in DEF-CS system, the patient derived cell lines showed increased apoptosis measured by Annexin V. We observed more than 9-fold difference in increased apoptosis of patient derived cells compared to healthy volunteers. Addition of MK-0339 and sivelestat reduced the apoptosis. MK0339 significantly reduced the apoptosis in both mutant cell lines, while sivelestat significantly reduced the apoptosis in only P139L mutant cell lines. Again, sivelestat appeared to be less effective. Myeloid differentiation of iPSc was also impaired and inefficient in patient derived cell lines detected with granulocytic differentiation markers using flow cytometry. Introduction of MK0339 in this system normalizes the impaired myeloid differentiation capacities of patient derived iPSc lines. Summary/Conclusion: These studies clearly show effects of these selected inhibitors to enhance neutrophil formation in reproducible cellular models that mimics features of SCN and CyN. We believe this work will lead to novel therapies for ELANE associated neutropenia and open the door to other new therapies for neutropenia. Disclosures Dale: Amgen: Consultancy, Honoraria, Research Funding.


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.


1991 ◽  
Vol 273 (3) ◽  
pp. 573-578 ◽  
Author(s):  
M C Garcia ◽  
C Garcia ◽  
M A Gijon ◽  
S Fernandez-Gallardo ◽  
F Mollinedo ◽  
...  

The binding and metabolism of platelet-activating factor (PAF) was studied in human cell lines resembling myeloid cells (HL60 and U937) and B and T lymphocytes (Daudi and Jurkat). All of the cell lines were found to bind and catabolize exogenous [3H]PAF in a time- and temperature-dependent manner. PAF binding could also be demonstrated in isolated membrane fractions, which provides further evidence of the existence of true membrane receptors. Myeloid cell lines contained numbers of receptors at least 10-fold higher than in lymphoid cell lines. Biosynthesis of PAF upon challenge by ionophore A23187 could be demonstrated in HL60 and U937 cells. In contrast, lymphoid cell lines were unable to produce PAF. Incubation with [14C]acetate showed incorporation of the label into three main fractions: neutral lipids, phosphatidylcholine and PAF, but the distribution of the label varied depending on the cell line. Significant incorporation into phosphatidylcholine was observed in uninduced myeloid cell lines. A phospholipase A2 acting on 1-O-hexadecyl-2-arachidonoyl-sn-glycero-3-phosphocholine and an acetyl-CoA:lyso-PAF acetyltransferase were expressed in the HL60 cell line and showed variations in specific activity with granulocytic differentiation. In contrast, these enzyme activities were not expressed in Daudi and Jurkat cell lines. These data indicate (1) the occurrence of PAF binding and catabolism in both myeloid and lymphoid cell lines; (2) the restriction of PAF biosynthesis to myeloid cell lines, especially HL60 cells; (3) the occurrence of differentiation-elicited changes in the specific activities of the enzymes involved in PAF biosynthesis by the remodelling pathway; and (4) the central role played by the disposal of lyso-PAF, a product of the phospholipase A2 reaction, in PAF biosynthesis.


2000 ◽  
Vol 113 (8) ◽  
pp. 1405-1414 ◽  
Author(s):  
P.F. van der Ven ◽  
J.W. Bartsch ◽  
M. Gautel ◽  
H. Jockusch ◽  
D.O. Furst

Titin, also called connectin, is a giant muscle protein that spans the distance from the sarcomeric Z-disc to the M-band. Titin is thought to direct the assembly of sarcomeres and to maintain sarcomeric integrity by interacting with numerous sarcomeric proteins and providing a mechanical linkage. Since severe defects of such an important molecule are likely to result in embryonic lethality, a cell culture model should offer the best practicable tool to probe the cellular functions of titin. The myofibroblast cell line BHK-21/C13 was described to assemble myofibrils in culture. We have now characterized the sub-line BHK-21-Bi, which bears a small deletion within the titin gene. RNA analysis revealed that in this mutant cell line only a small internal portion of the titin mRNA is deleted. However, western blots, immunofluorescence microscopy and immunoprecipitation experiments showed that only the N-terminal, approx. 100 kDa central Z-disc portion of the 3 MDa titin protein is expressed, due to the homozygous deletion in the gene. Most importantly, in BHK-21-Bi cells the formation of thick myosin filaments and the assembly of myofibrils are impaired, although sarcomeric proteins are expressed. Lack of thick filament formation and of ordered actin-myosin arrays was confirmed by electron microscopy. Myogenisation induced by transfection with MyoD yielded myofibrils only in myotubes formed from wild type and not from mutant cells, ruling out that a principal failure in myogenic commitment of the BHK-21-Bi cells might cause the observed effects. These experiments provide the first direct evidence for the crucial role of titin in both thick filament formation as a molecular ruler and in the coordination of myofibrillogenesis.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 3550-3550
Author(s):  
Vadim Markovtsov ◽  
Diane Yu ◽  
Marina Gelman ◽  
Wayne Lang ◽  
Vanessa C. Taylor ◽  
...  

Abstract Limited options provided by the current standard of care for the patients suffering from myeloproliferative diseases (MPDs) prompted an extensive search for the underlying molecular mechanisms of these disorders. Recent discovery of a single activating mutation (V617F) in JAK2 kinase gene associated with the development of the polycythemia vera (PV), essential thrombocythemia (ET) and chronic idiopathic myelofibrosis (CIMF) opened up a possibility to develop highly targeted therapies against these debilitating ailments. To that end, we engineered cytokine-independent Ba/F3 cell line expressing the V617F mutant of JAK2 to screen a focused small molecule library for potential inhibitors of JAK2 V617F -dependent proliferation. We confirmed the ability of hit compounds to inhibit proliferation of JAK2-dependent tumor cell lines using UKE-1 and SET-2 cells carrying the V617F JAK2 mutation. A FACS-based phosphoSTAT5 assay was then used to demonstrate that the hits directly targeted mutant JAK2. JAK3 activity of each compound was evaluated in IL-2-dependent CTLL-2 cell line using phosphoSTAT5 FACS and proliferation assays. To avoid hits with nonspecific antiproliferative activity, the hits were tested in JAK2-independent MOLT4, A549 and H1299 cell lines. Compound hits with the desirable properties were further evaluated for their ability to inhibit JAK2, JAK3 and other kinases in the context of T cell, B cell, or mast cell activation using a variety of cell-based assays as well as in the in vitro biochemical assays. We identified a number of compounds that potently inhibit growth of the two V617F mutant cell lines with EC50s varying from 20 to 500 nM, but do not affect proliferation of control cell lines MOLT4, A549 and H1299 to the same degree. These compounds induce strong and highly specific suppression of STAT5 phosphorylation with IC50s of 10 to 200 nM in SET-2 and V617F JAK2 expressing Ba/F3 cells. One of the hits with the desirable biological and pharmacokinetic profiles was further evaluated in V617F JAK2 Ba/F3 engraftment mouse model where it demonstrated significant extension of survival at 150 and 200 mg/kg bid. Such potent JAK2 inhibitors could become the basis for the next generation of compounds targeting JAK2-dependent myeloproliferative diseases.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1589-1589
Author(s):  
Jenny E. Hernandez ◽  
Junling Li ◽  
Ru-Qi Wei ◽  
Paul Tapang ◽  
Steven K. Davidsen ◽  
...  

Abstract FLT3 is an receptor tyrosine kinase of the subclass III family that plays a vital role in the regulation of the differentiation, proliferation and survival of normal hematopoietic cells. FLT3 mutations are often found in patients with Acute myelogenous leukemia (AML) and confer poor prognosis. Of these mutations, 15–35% are FLT3 ITD (internal tandem duplication) mutations and 5–7% are point mutations on the FLT3 kinase activation loop (e.g. D835V). Our laboratory is studying the signaling pathways associated with a newly identified multi-targeted tyrosine kinase receptor small molecule inhibitor (RTKI), ABT-869. Recently published work in our laboratory showed that using ABT-869 to treat MV4-11, a human AML FLT-3 ITD mutant cell line, resulted in the inhibition of phosphorylation of FLT-3 with a downstream inhibitory effect on the activation of STAT5, ERK, and Pim-1. Cell viability assays determined that MV-411 cells responded to ABT-869 in a concentration dependent manner (IC50 = 10nM). Apoptosis studies also showed an induction of apoptosis in ABT-869 treated cells. In vivo studies involving xenograft injections of MV-411 cells into SCID mice and subsequent treatment with ABT-869 demonstrated regression of tumor formation. In this study, a Ba/F3 mouse pro-B lymphocytic cell line harboring the FLT-3 ITD or FLT-3 D835V mutation is used as an isolated Flt-3 mutant model system. In vitro, ABT-869 is effective in inhibiting the proliferation of Ba/F3 Flt-3 ITD mutant cells when compared to Ba/F3 Flt-3 D835V mutant and Ba/F3 Flt-3 WT cells. Trypan Blue Exclusion and Alamar Blue assays were used to demonstrate that there is 50% inhibition of growth and proliferation (IC50) of Ba/F3 FLT3 ITD mutant cells at a concentration of 1nM after 48 hours of treatment. Ba/F3 FLT3 D835V mutant cells show an IC50 between 1μM and 10μM after 48 hours of treatment. In contrast, Ba/F3 FLT3 WT cells demonstrate an IC50 of 10μM only after 72 hours of treatment. Annexin V and propidium iodide staining of cells revealed that an increase in apoptosis (41.2%) occurred in Ba/F3 Flt-3 ITD mutant cells treated with 10nM ABT-869 after 24 hours when compared to untreated (6.5%) or vehicle control (6.1%) cells. Staining of Ba/F3 Flt-3 WT treated cell lines revealed no difference in apoptosis when compared to untreated Ba/F3 Flt-3 WT cell only and DMSO controls. PARP cleavage was observed in Ba/F3 FLT-3 ITD mutant cells following treatment with ABT-869 whereas no cleavage was observed with Ba/F3 WT cells treated with ABT-869. In vivo, the activity of ABT-869 treatment of SCID mice injected with Baf3 Flt-3 ITD, Baf3 Flt-3 D835V, or Baf3 Flt-3 WT cells is also being evaluated. Using bioluminescence imaging, it was determined that Ba/F3 FLT-3 ITD mutant and Ba/F3 Flt-3 D835Vmutant cell lines result in metastases and subsequent death in SCID mice after 2 weeks for ITD and 5 weeks for D835V, whereas mice injected with Ba/F3 WT survive longer than 5 weeks. Preliminary data demonstrated that ABT-869 prolonged survival in mice injected with the Ba/F3 FLT3-ITD cells compared to controls. Our preclinical data demonstrate that ABT-869 is effective specifically with FLT-3 ITD mutant cell lines in an isolated system. These studies provide rationale for the treatment of AML patients and the prevention of relapse.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1609-1609
Author(s):  
Federica Servida ◽  
Francesco Onida ◽  
Domenico Delia ◽  
Cinzia Scavullo ◽  
Daniele Lecis ◽  
...  

Abstract The apoptotic process and its dysfunctions have become the focus of extensive pharmaceutical research in solid and hematopoietic tumors as well as neurodegenerative diseases. The X-Inhibitor of Apoptosis Protein (XIAP) binds caspase 9, 3 and 7, preventing their activation and, consequently, apoptosis. The Smac/DIABLO protein, released from mitochondria, binds XIAP as a dimer on the same caspase 9 (BIR3 domain) binding site. Similarly, the Smac protein interferes with the XIAP binding site for caspases 3 and 7, thus promoting both the extrinsic and intrinsic apoptotic paths. The thin balance of this binding equilibrium is altered in various tumors, including leukemia, where XIAP is overexpressed and a caspase-dependent resistance to enter apoptosis is usually observed. Thus, XIAP inhibition via Smac mimetics’ binding is at the same time a characterized protein-protein interaction, and a validated mechanism for intervention in cancer therapy. We tested 56 Smac mimetic compounds (designed by CISI - Center for biomolecular Interdisciplinary Studies and Industrial applications of the Milan University) for their in vitro capacity to bind to the XIAP BIR3 domain. We also evaluated the ability of the Smac mimetic compounds to inhibit the growth of the human leukemia HL60, K562 and Jurkat cell lines (derived from patients with promyelocyitic leukemia, blastic phase-CML and T acute lymphoblastic leukemia, respectively). Nine compounds which were shown to be active, were further investigated for their effect on cell cycle (by DNA staining with propidium iodide and cytofluorimetric analysis) and for possible synergistic effect in combination with other chemotherapeutic drugs (Cytarabine, Etoposide and Idarubicine). The same compounds were also tested on normal CD34+ hematopoietic progenitor cells. The cytotoxicity was evaluated after 72 hours treatment with Smac mimetic compounds by a colorimetric assay for the quantification of cell proliferation and viability based on the cleavage of the WST-8 tetrazolium salt by mitochondrial dehydrogenases. The effect of Smac mimetic compounds on CD34+ cells enriched from mobilized peripheral blood was assessed as the capability of inhibiting the myeloid colony growth (CFU-GM). The data were expressed as mean percentage of 3 replicates normalized to the untreated control. Overall, a strong correlation between the binding affinity to the XIAP BIR3 domain and the cytotoxic effect on the leukemic cell lines was observed. The more promising compounds showed IC50 ranging from 0,3 to 1 microM on the HL60 cell line. The Jurkat and K562 cell lines were less sensitive, with IC50 ranging from 11,8 microM to more than 50 microM. However, in the K562 cell line, the combined treatment unveiled synergistic effect with Cytarabine and Etoposide (R Kern index = 1,4 and 1,5 respectively). No cytotoxic effect was observed on normal controls at doses up to 80 microM. A consistent sub G1 apoptotic peak (up to 53% of apoptotic cells) was observed in the HL60 cell line after 48 hrs treatment, thus suggesting a strong activation of the apoptotic process. All together, our data suggest that Smac mimetics may have a promising therapeutic potential as a new class of anticancer drugs in hematopoietic malignancies. Further experiments are currently ongoing to confirm the effectiveness of these compounds also on primary cells from leukemia patients, both as single agents and in combination with conventional drugs. In particular, due to their ability to enhance pro-apoptotic effect, Smac mimetic compounds may allow to overcome resistance of cancer cells to standard chemotherapy.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2474-2474
Author(s):  
Jesus Duque-Afonso ◽  
Aitomi Essig ◽  
Leticia M Solari ◽  
Tobias Berg ◽  
Heike L. Pahl ◽  
...  

Abstract Abstract 2474 Background: The leukemia-specific oncofusion protein AML1/ETO regulates different target genes, including the LAT2 gene (encoding the adaptor molecule LAT2/NTAL/LAB), which is epigenetically repressed by AML1/ETO as we have previously described. LAT2 is phosphorylated by c-kit and has a role in mast cell and B cell activation. To address the functional role of LAT2 during myeloid differentiation, expression studies were performed in myeloid cell lines, and LAT2 was overexpressed by retroviral gene transfer in AML1/ETO-positive Kasumi-1 cells and AML1/ETO-negative U-937 cells. Methods: To induce monocytic and granulocytic differentiation, the myeloid cell lines U-937, HL-60 and NB4 were treated with PMA and ATRA, respectively, and LAT2 expression measured by both Northern and Western blot. LAT2 was overexpressed in Kasumi-1 and U-937 cells by use of the retroviral vector pMYSiG-IRES-GFP. Virus was produced in 293T cells and titrated in TE671 cells. Following transduction, GFP-positive cells were sorted by fluorescence-activated cell sorting (FACS). Transduced cells were treated with PMA (2 and 10 nM for 24 and 48 hours) and ATRA (0.1 μM and 0.5 μM for 48 and 96 hours), respectively. Results: The AML1/ETO-negative myeloid cell lines HL-60, NB4 and U-937 readily expressed LAT2, which was further upregulated by PMA, and transiently downregulated with ATRA. In the AML1/ETO-positive Kasumi-1 and SKNO-1 cells, LAT2 expression was absent. To address the functional role of this repression, forced expression of LAT2 was achieved in Kasumi-1 and U-937 cells and resulted in effective processing of LAT2 protein (confirmed by Western blot), and a decrease in the expression of the differentiation markers CD11b and CD11c (FACS analysis) in Kasumi-1 but not U-937, with only minor effects of LAT2 overexpression upon apoptosis and cell growth arrest. Notably, during both PMA- and ATRA-induced differentiation, a striking maturation block occurred in Kasumi-1 (measured by CD11b/CD11c expression, observed at different doses and time points of these treatments), while differentiation of U-937 cells was unimpaired by overexpression of LAT2. Conclusions: In AML1/ETO-negative leukemia cells, LAT2 expression is differentially regulated during monocytic and granulocytic differentiation. In AML1/ETO-positive leukemia cells, in which LAT2 is repressed, LAT2 re-expression imposes a striking maturation block. Graded expression of this novel AML1/ETO target gene may therefore play an important role in maintaining the phenotypic characteristics of this leukemia subtype. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2165-2165
Author(s):  
Wienke Ellerbeck ◽  
Olga Klimenkova ◽  
Julia Skokowa ◽  
Karl Welte

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.


Sign in / Sign up

Export Citation Format

Share Document