Endoplasmic Reticulum Stress Is a Target for Therapy in Paroxysmal Nocturnal Hemoglobinuria.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 160-160
Author(s):  
Regis Peffault de Latour ◽  
Valeria Visconte ◽  
Keyvan Keyvanfar ◽  
Olga Nunez ◽  
Marie Desierto ◽  
...  

Abstract Abstract 160 Paroxysmal nocturnal hemoglobinuria (PNH) is characterized by hemolytic anemia, bone marrow failure and venous thromboembolism. The disease is caused by a somatic mutation of the X-linked gene PIG-A, encoding a key enzyme responsible for the biosynthesis of the glycosylphosphatidylinositol anchored proteins (GPI-APs). Unfolded GPI-APs are translated and degraded intracellularly in the endoplasmic reticulum (ER) via the proteasome machinery. We hypothesized that the accumulation of misfolded proteins in mutant PNH cells activates the unfolded protein response (UPR) signaling cascade, which physiologically maintains the quality of newly synthesized proteins. UPR could thus represent a new target for therapy in PNH. We first assessed the sensitivity of GPI-AP- deficient K562 cells to the proteasome inhibitor PS-341 (Velcade‘) by flow cytometry after staining for annexin V and propidium iodide (PI). After 24 hour exposure to PS-341, cytotoxicity was significantly higher in the K562 GPI-AP-deficient cell line compared to their wild type counterpart at all dose tested (0 to 200nM). At 50nM, the early stage apoptosis was 21±2% in the K562 GPI-AP-deficient cell line versus 9.3±2% for their wild type counterpart (p=0.013) while the late stage apoptosis was 38±2.3% versus 22±2.3% (p<10−3), respectively. Similar results were obtained in immortalized lymphoblastoid cells derived from a PNH patient after PS-341 exposure. We reproduced those results with 2 other proteasome inhibitors (MG132 and Lactacystine) confirming the fundamental role of the proteasome in PNH. By immunoblot, K562 GPI-AP-deficient cell line exhibited more expression of chaperone proteins (CHOP and BiP/GRP78) compared to wild type cells after PS-341 exposure; evidence of the UPR is initiated by accumulation of unfolded proteins. UPR activation was confirmed by the detection of XBP1 splicing form in K562 GPI-AP-deficient cells by polymerase chain reaction (PCR). Moreover, PS-341 promoted pro-apoptotic/terminal UPR gene expression in K562 GPI-APs deficient cells as illustrated by the increase expression of an UPR specific pro-apoptotic protein, NOXA. We observed a concomitant arrest in the G2 phase of the cell cycle, as detected by flow cytometry after staining with PI. We employed a mouse model bearing a conditional Pig-a gene deletion in hematopoietic cells. When the animals were treated with PS-341 (15 ug/day once a week, intraperitoneally), there was a significantly higher proportion of apoptotic GPI-AP-deficient B cells compared to normal B cells in early stage apoptosis (p=0.01) and a slight increase of in the late stage apoptosis (p=0.13). PS-341 induced apoptosis also in bone marrow samples from two patients with classical and aplastic PNH as assessed by flow cytometry, based on CD59 and CD55 expression in the CD34+ population (Figure 1). GPI-AP-CD34+ deficient cells were more sensitive to PS-341 at the early stage of apoptosis (at 50 nM, 14% vs 4.6%), as well as late stage (at 200nM: 22% vs 7.2%). In conclusion, we demonstrate that GPI-AP-deficient cells are more susceptible to proteasome inhibition through the activation of the proapoptotic/terminal UPR, which leads to specific apoptosis in these cells. The sensitivity of PNH cells in vivo and in vitro to proteasome inhibitors might allow for the development of therapeutic strategies specifically directed to target one or several components of the UPR in PNH.Figure 1:Representative apoptosis assay in CD34+ cells according to GPI expression after 24 hours exposition to PS-341Figure 1:. Representative apoptosis assay in CD34+ cells according to GPI expression after 24 hours exposition to PS-341 Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2904-2904
Author(s):  
Robyn M. Dennis ◽  
Brandon Ballard ◽  
David John Tweardy ◽  
Karen Rabin

Abstract Abstract 2904 Survival has improved dramatically in acute lymphoblastic leukemia (ALL), but further gains are unlikely using conventional chemotherapy alone. Several recently discovered, novel cytogenetic lesions with adverse prognostic impact, JAK2 activating mutations and CRLF2 rearrangements, occur in up to 15% of adult and pediatric ALL. These lesions are associated with activation of Jak2 and Stat5, and hold promise as targets for novel therapies affecting these signaling pathways. We performed in vitro testing of a novel small molecule Stat inhibitor, C188-9, in B-lineage ALL cell lines and patient samples with and without JAK2/CRLF2 alterations. C188-9 treatment for one hour at 10 μM inhibited Stat3 and Stat5 phosphorylation in ALL cell lines with JAK2 and CRLF2 alterations, but not in cell lines with wild-type JAK2 and CRLF2, as measured by phospho-flow cytometry (Fig. 1A). Only the cell lines with JAK2 and CRLF2 alterations demonstrated basal Stat5 phosphorylation on Western blot analysis, and this was inhibited by C188-9 treatment (Fig. 1B). C188-9 demonstrated cytotoxicity in ALL cell lines regardless of JAK2/CRLF2 status, with IC50s in the low micromolar concentration range (Fig. 1C). While C188-9 is undergoing investigation currently as a potent inhibitor of Stat3 in acute myeloid leukemia (AML), it also merits further investigation as an agent with Stat5 inhibitory activity and cytotoxicity in ALL. Figure 1. Effects of C188-9 in ALL cell lines. A. Stat3 and Stat5 phosphorylation were determined by flow cytometry in the ALL cell lines MHH-CALL-4 (JAK2/CRLF2 mutated) and Reh (JAK2/CRLF2 wild-type). In each condition, cells were incubated in serum-free media for one hour, followed by incubation with C188-9 or vehicle for one hour, stimulation with vehicle or pervanadate 125 mM for 15 minutes, fixation, permeabilization, phospho-antibody staining for phospho-Stat3 and phospho-Stat5, and flow cytometric analysis. B. Western blot for phospho-Stat5 in K562 cell line (positive control); MHHCALL-4 treated for one hour with C188-9 at 0, 5, or 10 uM; and RS4;11 (JAK2/CRLF2 wild-type ALL cell line). C. IC50 determination by ATP assay for C188-9 in the ALL cell lines MHH-CALL-4 and RS4;11. Each experiment was performed in triplicate. Figure 1. Effects of C188-9 in ALL cell lines. A. Stat3 and Stat5 phosphorylation were determined by flow cytometry in the ALL cell lines MHH-CALL-4 (JAK2/CRLF2 mutated) and Reh (JAK2/CRLF2 wild-type). In each condition, cells were incubated in serum-free media for one hour, followed by incubation with C188-9 or vehicle for one hour, stimulation with vehicle or pervanadate 125 mM for 15 minutes, fixation, permeabilization, phospho-antibody staining for phospho-Stat3 and phospho-Stat5, and flow cytometric analysis. B. Western blot for phospho-Stat5 in K562 cell line (positive control); MHHCALL-4 treated for one hour with C188-9 at 0, 5, or 10 uM; and RS4;11 (JAK2/CRLF2 wild-type ALL cell line). C. IC50 determination by ATP assay for C188-9 in the ALL cell lines MHH-CALL-4 and RS4;11. Each experiment was performed in triplicate. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2717-2717
Author(s):  
Céline Bourgne ◽  
Alexandre Janel ◽  
Juliette Berger ◽  
Agnès Guerci ◽  
Caroline Jamot ◽  
...  

Abstract Introduction Chronic Phase - Chronic Myeloid Leukemia (CP-CML) is a myeloproliferative disorder characterized by malignant proliferation of the granulocytic lineage without the arrest of cell differentiation. Tyrosine Kinase Inhibitors (TKI) have revolutionized CML treatment but several studies showed that a combination of TKI and Interferon alpha (IFNα) provides better clinical response. Myeloid Nuclear Differentiation Antigen (MNDA), which belongs to the hematopoietic interferon-inducible nuclear proteins with the 200-amino-acid repeat (HIN200) gene family, encodes a protein expressed in myeloid cells but whose function remains poorly understood. Because of its high expression in polymorphonuclear cells, its involvement in cell differentiation and apoptosis, and its induction by IFNα, we evaluated MNDA expression in CML cells and its modulation after incubation with IFNα. Material and methods We tested MNDA expression in several cell lines (K562, KCL22, LAMA84, TF1 and U937 (positive control)), in polymorphonuclear cells from healthy donors (HD-PMN, n=13) and in primary cells from patients with CP-CML at diagnosis (CP-CML; n=17). The relative expression of the MNDA transcript was analyzed using the 2-ΔΔCt method and was normalized to the endogenous reference gene GAPDH. HD-PMN were used as calibrator. We developed a multiparametric flow cytometry assay (CD45-V500/CD14-APC-H7/CD15-PerCpCy5.5/CD34-PC7/CD38-V450/MNDA-FITC) to detect MNDA protein in the different cell subsets, particularly in CD34+cells. Results As previously described, MNDA was poorly expressed in the K562 cell line. Similarly, mRNA was detected at low levels in two other CML cell lines (KCL22, LAMA84) and in TF1 cells, but at a high level in the U937 cell line, used as a positive control. In each cell line, the transcript expression was correlated to the protein level, as evaluated by flow cytometry (MFI ratio: 2.04±0.21, 2.36±0.24, 1.59±0.14, 1.88±0.11 and 8.77±0.54 for K562, KCL22, LAMA84, TF1 and U937, respectively (n=3)). In CP-CML primary cells, MNDA expression was greatly diminished as compared with HD-PMN in both mRNA (0.20±0.08 (n=17) vs. 1.32±0.21 (n=10); p=1.52x10-6) and protein (MFI ratio: 6.9±0.98 vs. 16.31±1.25, p=0.001). After having verified that IFNα (2000 U/ml, 16 hours) induced MNDA expression in HD mononuclear cells but not in PMN, we observed that induction of MNDA was moderate in CML cell lines K562 and LAMA84 (2-fold increase, n=3) whereas the level of MNDA mRNA was significantly increased in TF1 cells (28-fold increase, n=4). Induction in primary CML cells was variable (3/5 patients). Aiming to evaluate the expression of MNDA in leukemic stem cells (LSC), we first analyzed MNDA expression in CD34+ and CD34+/CD38- cells from HD. We observed that MNDA is down-regulated in healthy CD34+ and CD34+/CD38- cells compared to mature cells (mRNA: about 4 logs, protein: 8-10 fold lower, n=4), but we always detected a significant signal in CD34+cells (MFI ratio: 2.76±0.46, n=3). However, MNDA was not expressed by CML cells from the LSC compartment (n=4). This inhibition does not seem to be antagonized by nilotinib or IFNα (n=2). Discussion/Conclusion MNDA expression appears to be clearly down-regulated in CP-CML cells and dramatically so in the LSC compartment. In some patients, we observed sustained sensitivity to IFNα, but only in the compartment of more mature cells. This suggests early deregulation of MNDA expression which seems to be only partially dependant on differentiation. The mechanisms involved in this down-regulation remain to be elucidated but could be independent to TK activity of BCR-ABL protein and resistant to IFNα in the LSC compartment. This marked deregulation of MNDA in the LSC compartment is an additional argument in favor of intrinsic changes specific to primitive cells. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1688-1688 ◽  
Author(s):  
Christopher O. Eden ◽  
David K. Edwards ◽  
Christopher A. Eide ◽  
Elie Traer ◽  
Jeffrey W. Tyner ◽  
...  

Abstract Background: Inflammatory cytokines secreted in the bone marrow microenvironment play important roles in modulating cell survival, proliferation, differentiation, and immune responses in cancer. Perhaps not surprisingly, there is also an association between chronic inflammation and tumor progression. We recently used an ex vivo functional screen of 94 cytokines to show that the pro-inflammatory cytokines IL1α and IL1β promoted the expansion of AML progenitors in 70% (40/60) of primary samples. We therefore hypothesized that inflammatory cytokines are crucial to clonal expansion and disease progression in AML and that therapeutic targeting of these pathways may circumvent disease heterogeneity. Here we provide in vitro and in vivo evidence that IL1-mediated signaling elicits profound expansion of leukemia progenitors in AML patients harboring various genetic mutations and promotes in vivo clonal expansion and disease progression in a murine AML model. Further, these effects are reversed by targeting IL1 signaling. Methods: We validated the role of IL1 signaling using a shRNA approach and in murine competitive repopulation and bone marrow transplantation models. We evaluated the influence of these cytokines on inflammatory markers using immunoblotting, flow cytometry, and Luminex assays, and assessed strategies to target these pathways using small-molecule inhibitors. Results: IL1 stimulation promoted a 3- to 20-fold increase in growth, survival, and clonogenic potential of AML CD34+ cells, while paradoxically suppressing growth of healthy CD34+ cells. To identify the influence of IL1 on in vivo clonal expansion of healthy and leukemic progenitors simultaneously, we established an in vivo murine competitive repopulation study utilizing TET2-null mice. IL1 treatment promoted clonal expansion of TET2-null myeloid cells over wild-type cells during 6 weeks of IL1β treatment. Consistent with this, both flow cytometry analysis and blood differential counts showed an increased percentage of granulocytes and reduced percentage of lymphocytes in IL1-treated mice. In this model, TET2-null monocytes have greater expression of IL1 than wild-type cells, suggesting IL1 promotes clonal growth of TET2-mutated early leukemic progenitors. Similarly, IL1β and IL1 receptors (IL1R1 and IL1RAP) were overexpressed in IL1-sensitive AML bone marrow and peripheral blood samples compared to nonsensitive AML samples and normal samples. Intracellular FACS showed that the majority of IL1β was secreted by monocytes and to some extent by myeloid progenitors. Accordingly, IL1-sensitive AML samples exhibited trends towards monocytic and myelomonocytic clinical features. Reduced survival of AML cells after monocyte depletion was rescued by IL1 treatment, suggesting that IL1 mediates paracrine regulation of AML cell growth. Silencing of the IL1 receptor, IL1R1, reduced the clonogenic potential of AML primary samples and oncogene (AML1-ETO9a, NRASG12D, and MLL-ENL)-transduced mouse bone marrow. In a murine bone marrow transplantation model, recipients of IL1R1-/- marrow transduced with AML1-ETO9a/NRASG12D survived significantly longer than did recipients of wild-type marrow. We also found that IL1β increased phosphorylation of p38MAPK and MK2, as well as secretion of multiple downstream inflammatory cytokines (IL6, IL8, MCP1, MIP1α, and MIP1β) from CD34+ progenitors, in IL1-sensitive AML samples compared to IL1-nonsensitive progenitors. Conversely, treating AML cells with p38MAPK inhibitors such as doramapimod andralimetinib reduced the growth of AML cells by decreasing p38MAPK and MK2 phosphorylation and reducing secretion of inflammatory cytokines from AML progenitors. Clinical and demographic analyses suggest that AML patients are dependent on IL1 signaling irrespective of mutation status and clinical features. Targeting this unifying mechanism of IL1-mediated clonal expansion may thus have application across heterogeneous AML subtypes. Conclusion: We demonstrate that IL1 promotes in vitro and in vivo clonal expansion of leukemic cells and promotes disease progression in AML. As IL1 signaling is active across heterogeneous disease subtypes, AML patients may therefore benefit from drugs targeting IL1/p38MAPK signaling because of their potential to inhibit AML while enhancing normal hematopoiesis, a significant clinical advantage over traditional chemotherapy. Disclosures Agarwal: CTI BioPharma Corp: Research Funding.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 660-660 ◽  
Author(s):  
Susann Szmania ◽  
Balaji Balasa ◽  
Priyangi Malaviarachchi ◽  
Fenguhuang Zhan ◽  
Yongsheng Huang ◽  
...  

Abstract Introduction: One-third of multiple myeloma (MM) patients exhibit high-risk features such as abnormal cytogenetics, high LDH, amplification of CKS1-B or spiked expression of MAF, MAF-B or FGFR3. While not affecting complete response rates, median durations of event-free and overall survival, even with high-dose melphalan-based tandem autotransplants of such patients, do not exceed 24 mo and 36 mo, compared to 60 mo and ≥ 90 mo for the remainder. Monoclonal antibody (mAb)-mediated therapy may target a chemotherapy-resistant myeloma cell pool. CS1 (CD2 subset 1, CRACC, SLAMF7), a member of the CD2 family of cell surface glycoproteins, exhibits high-level expression on primary myeloma cells, indicating that CS1 is a potential target for treatment in MM. Methods: Gene expression was assessed using an Affymetrix GeneChip array. Protein expression was measured by flow cytometry, and immunohistochemistry (IHC), using HuLuc63, a novel humanized anti-CS1 mAb. HuLuc63-mediated lysis of myeloma cells via antibody dependent cellular cytotoxicity (ADCC) was measured by 51Cr-release. Results: CS1 mRNA was detected in &gt;95% of CD138+ purified plasma cells from &gt;95% of healthy donors, newly diagnosed myeloma patients, and those with relapsed myeloma (Fig. 1a). CS1 remained highly expressed in patients following VDTPACE treatment, albeit at a reduced level. CS1 expression was also high following bortezomib (Velcade®) treatment, with a subset of patients showing increased expression post-treatment. CS1 protein expression on primary myeloma cells was confirmed by flow cytometry, while IHC analysis of normal tissues revealed anti-CS1 staining primarily on CD138+ tissue plasma cells. Finally, we determined that HuLuc63 could induce killing of myeloma cells using purified allogeneic NK cells (Fig. 1b). Blocking the Fc receptor greatly reduced this activity indicating an ADCC mechanism. Killing of myeloma targets was also observed in autologous systems suggesting that HuLuc63 can overcome KIR-mediated NK cell inhibition of autologous NK cells. In summary, we observed high mRNA and protein expression of CS1 in myeloma from early stage, late stage, and treated patients, and showed enhanced lysis of myeloma cells in vitro with HuLuc63. Our data support the potential clinical utility of CS1-targeted therapy. HuLuc63 will be entering a phase I clinical trial for advanced myeloma patients. Figure 1a. CS1 mRNA is highly expressed in CD138+ purified plasa cells. Figure 1a. CS1 mRNA is highly expressed in CD138+ purified plasa cells. Figure 1b. CS1 antibody enhances killing of myeloma cells by allo - NK cells. Figure 1b. CS1 antibody enhances killing of myeloma cells by allo - NK cells.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 5034-5034
Author(s):  
Karel Svojgr ◽  
Tomas Kalina ◽  
Tomas Brdicka ◽  
Tereza Kacerova ◽  
Jana Volejnikova ◽  
...  

Abstract Abstract 5034 Transmembrane adaptor proteins NTAL, LAT, PAG and LIME have an important role in proximal signaling of T-lymphocytes. They lack enzymatic or kinase function, but when tyrosine-phosphorylated they bind other signaling molecules and mediate signaling from T-cell receptor (TCR) to nucleus. In our previous studies, we showed that in childhood T-cell acute lymphoblastic leukemia (T-ALL) the level of NTAL significantly correlates with early response to treatment. Patients with high NTAL levels responded favorably (prednisone good-responders) whereas patients with low NTAL were prednisone poor responders (p=0.05). We confirmed this clinical data also in in-vitro experiment - derivative Jurkat cell line (human T-ALL) transfected with NTAL expression construct (Jurkat/NTAL+) was more sensitive to corticosteroid treatment compared to wild-type Jurkat cell line (Jurkat/wt, NTAL negative). In the present study, we performed a more detailed analysis of a relationship between adaptor proteins, TCR signaling and apoptosis in T-ALL. Monoclonal IgM antibody C305 binding the Jurkat TCR with high affinity was used to stimulate TCR signalization in Jurkat/wt and Jurkat/NTAL+ cell lines. To assess the influence of TCR signaling on corticosteroid-driven apoptosis, we treated both the Jurkat cell lines with the C305 antibody and/or with methylprednisolone. At 24 hours we detected higher percentage of cells with the stimulation marker CD69 in Jurkat/NTAL+ cells compared to the Jurkat/wt (median 70% vs. 60%, p<0.05). More cells treated with C305 and methylprednisolone together expressed higher CD69 than cells treated by C305 alone (median 75% vs. 65%, p<0.004). At 24 and 48 hours we determined the level of apoptosis. At each time point the number of living cells in the untreated control was set to 100%. Jurkat/NTAL+ cell line was more prone to apoptosis than the wild-type cell line in all settings. At 24 hours, the percentage of surviving cells Jurkat/NTAL+ vs. Jurkat/wt was 44% vs. 50% when treated with methylprednisolone alone (p<0.1), 24% vs. 42% (C305 alone, p<0.1) and 15% vs. 26% (methylprednisolone + C305, p<0.1). Similar differences were detected after 48hours of treatment. Using flow cytometry, we further determined phosphorylation status of key downstream kinases ERK, p38 and JNK after TCR stimulation by the C305 antibody. We detected hyperphosphorylation of ERK higher in Jurkat/NTAL+ compared to Jurkat/wt (1.57, 1.53 and 1.46 fold at 5, 15 and 30 minutes after stimulation, respectively). Levels of phosphorylated P38 and JNK did not differ from the unstimulated controls or between the two cell lines. Our data show that the differences in apoptosis are not driven by an upregulation of FAS or FAS ligand as any significant increase in the levels of FAS/FASL was not detected by flow cytometry throughout the above described experiments. It was shown previously that aberrant TCR signaling affects the incidence of dominant-negative isoforms of IKAROS. Thus, to further elucidate the role of the signaling in T-ALL we analyzed presence and expression profile of IKAROS splicing variants (using on-chip electrophoresis) in a cohort of 29 pediatric T-ALL patients. While we did not observe any link between NTAL or LAT levels and expression of different IKAROS isoforms, we found a close correlation between IKAROS isoforms IK1 and IK4a and another adaptors, PAG (major inhibitor of Src kinases in lymphocytes) and LIME (adaptor involved in signalization via CD4 and CD8). The two patients expressing IK1 and IK4A as dominant transcripts had >2 logs lower PAG (p=0.02) and LIME (p<0.1) expression levels compared to the rest of the cohort (n=27) showing IK2 as a dominant transcript. Based on this data, we propose that NTAL acts as a tumor suppressor enhancing proximal signaling of lymphocytes. Via phosphorylation of ERK, the NTAL sensitizes T-leukemic cells to corticosteroid induced apoptosis that is not mediated by FAS or FASL. The role of IKAROS isoforms and its connection with adaptor proteins is under investigation. The work was supported by grants MSM0021620813 and GAUK35607. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4977-4977
Author(s):  
Denise Carvalho Rezende ◽  
Lorena Pacheco ◽  
Gustavo Loureiro ◽  
Elisa Kimura ◽  
Laila Silva ◽  
...  

Abstract Abstract 4977 Introduction PI3K/AKT signalling pathway is involved in cell growth, proliferation and apoptosis. PI3K/AKT constitutive activation is observed in several solid tumors and leukemic cells. Inhibition of PI3K/AKT activity using specific inhibitor, LY294002, results in apoptosis. Deguelin is a natural product isolated from the leguminous, Mundulea sericea, with antitumorigenic effect in vitro and in vivo. The inhibition effect of Deguelin in PI3K/AKT signalling pathway in leukaemia cells was also observed in vitro. Aims We evaluated PI3K/AKT activation using p-AKT expression by flow cytometry in P39 cell line and CD34+ hematopoietic progenitors. PI3K/AKT activity inhibition mediated by deguelin and its proapoptotic effect was tested in P39 cell line. Material and Methods The high-risk MDS cell line P39 and leukaemia cell lines, Jurkat and HL60, were maintained in RPMI1640 and FBS 10%. Jurkat and HL60 were used as positive and negative control for p-AKT expression, respectively. Cell lines and CD34+ cells from bone marrow healthy donors (n=5) were assessed for p-AKT expression. Flow cytometry detection of intracellular Ser 473 p-AKT was performed after permeabilization and fixation with a saponin based solution with Tween20 and FACSlysing solution (Becton Dickinson-BD). An alexa-fluor 488-conjugated rabbit antibody to Ser 473 p-AKT (Cell Signalling Technology) was employed. A double immunostaining procedure using CD45-PerCP and CD34+PE was performed to analyse p-AKT expression in CD34+ cells from bone marrow healthy donors. After incubation, cells were analysed on a FACSCalibur (BD). The p-AKT activity was determined using Kolmogorov-Smirnov test (D). Difference between groups was analyzed using the Mann Whitney test. For treatment and determination of apoptosis, cells were exposed to deguelin (at concentrations of 10-300nM) and LY294002 (15-50uM) for 24-48h. To determine apoptotic changes, cells were stained withy Annexin-V/FITC and propidium iodide and examined on a FACScalibur (BD). Results Jurkat cells showed constitutive PI3K/AKT activation with a mean D value for p-AKT expression of D=0,84 ± 0,02. P39 cells also showed a constitutive PI3K/AKT activation with p-AKT expression as high as in Jurkat cells (mean D= 0,76±0,07). P-AKT expression was significant lower in CD34+cells from health donors (D=0,38±0,06) than in Jurkat and P39, respectively (p=0,0043 and p=0,015). Inhibition of p-AKT was observed using different concentrations of LY294002 (15-50uM) in Jurkat cells and apoptosis was observed after 24 and 48h when cells were treated with 50uM. For P39, although apoptosis was observed, no p-AKT inhibition was detected after LY294002 treatment. Treatment with deguelin induced apoptosis in Jurkat via p-AKT inhibition. No effect on apoptosis or p-AKT expression was observed in P39 cell line after deguelin treatment. Conclusions PI3/AKT constitutive activation was observed in P39 cell line and suggests that PI3/AKT can play a role in MDS progression. Deguelin effect in PI3K/AKT pathway can be cell specific, as observed in Jurkat but not in P39 cell line. Thus, deguelin in combination with other agents should be tested as a potential therapeutic option for MDS via PI3K/AKT inhibition. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 4399-4399
Author(s):  
Kohei Hosokawa ◽  
Takamasa Katagiri ◽  
Chizuru Saito ◽  
Luis J. Espinoza ◽  
Yumi Sasaki ◽  
...  

Abstract Abstract 4399 Background: An increase in glycosylphosphatidylinositol-anchored protein-deficient (GPI-AP−) blood cells is a significant predictor of a good response to immunosuppressive therapy in patients with aplastic anemia (AA) and low-risk myelodysplastic syndrome (MDS). Immune mechanisms are therefore thought to be involved in the increase of GPI-AP− cells in such bone marrow (BM) failure, though the exact mechanisms are unknown. The lack of GPI-APs due to PIGA mutations may render hematopoietic stem cells (HSCs) resistant to myelosuppressive cytokines such as TGF-b. This may lead to preferential activation of PIGA mutant HSCs in immune-mediated BM failure, although little is known about the GPI-APs involved. Objectives/Methods: We assessed the roles of GPI-APs in TGF-b-mediated inhibition of cell proliferation, cell cycle, and signal transduction in the myeloid leukemia cell line TF-1, TF-1 with a PIGA mutation (PNH-TF-1), and TF-1 cells transfected with siRNA against CD109, a GPI-AP that serves as a TGF-b co-receptor in human keratinocytes. We used flow cytometry to study expression of CD109 in BM CD34 cells of healthy individuals and other hematopoietic cell lines. Results: TGF-b inhibited PNH-TF-1 proliferation to a lesser degree (percentage of inhibition, 19±13%) than wild-type TF-1 cells (67±3%) in an MTT-based proliferation assay. Transfection of PIGA into PNH-TF-1 cells restored GPI-AP expression as well as sensitivity to TGF-b (53±10% versus 19±13% in PNH-TF-1 cells). CD109 coimmunoprecipitated with TGF-b in TF-1 cells, and its expression was confirmed on BM CD34+ cells of healthy individuals, particularly on CD34+CD38+CD123−CD45RA− megakaryocyte-erythroid progenitor cells, and also on TF-1 and KG1a cells. CD109 knockdown rendered TF-1 cells less sensitive to TGF-b-induced inhibition of proliferation (26±10% versus 67±3%), TGF-b induced cell cycle arrest (a relative increase in the G0/G1 phase of the cell cycle, 6.9% versus 20.7%, Figure) and TGF-b-induced SMAD2 phosphorylation (a relative increase in pSMAD2 MFI, 1±0.3% versus 16%±3%) compared to non-transfected TF-1 cells. Conclusions: The lower sensitivity of PIGA mutant TF-1 cells to TGF-b compared to wild-type TF-1 cells is partially due to CD109 deficiency. This deficiency may also account for the preferential activation of PIGA mutant HSCs in immune-mediated BM failure, in which TGF-b suppresses activation of wild-type HSCs. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 159-159
Author(s):  
Hongxia Yan ◽  
Yaomei Wang ◽  
Jie Li ◽  
Xiaoli Qu ◽  
Yumin Huang ◽  
...  

Abstract TET family proteins (TET1, TET2 and TET3) have recently emerged as important epigenetic modifiers by catalyzing the conversion of 5-methylcytosine (5mc) to 5-hydroxymethylcytosine (5hmc). Although they have been documented to play important roles in a variety of biological processes, their function in erythroid differentiation has yet to be defined. In the present study, we show that of the three TET family members, TET2 and TET3 but not TET1 are expressed in erythroid cells and that TET3 is more abundantly expressed than TET2. Using shRNA-mediated knockdown approach we explored the role of TET proteins in erythroid differentiation of CD34+ human cells. We first showed that consistent with their role in the production of 5hmc, knockdown of either TET2 or TET3 led to a decrease in global 5hmc levels as assessed by mass spectrometric analysis. However, knockdown of TET2 or TET3 resulted in distinctly different phenotypic changes during erythropoiesis. Knockdown of TET3 in human CD34+ cells resulted in impaired cell growth which is accompanied by increased apoptosis of late stage erythroblasts. Knockdown of TET3 also led to generation of bi/multinucleated polychromatic/orthochromatic erythroblasts which is accompanied by impaired enucleation. To explore the molecular mechanisms for the observed phenotypic changes, we performed RNA-seq analysis on control and TET3 knockdown erythroblasts at same stages of development. Bioinformatics analysis revealed that the expression levels of several apoptosis-promoting genes such as FOXO1, TNFRSF10B, TGFB1 and BTG1 are increased and that of a mitosis/cytokinesis associated gene KLHDC8B is decreased in polychromatic and orthochromatic erythroblasts following TET3 knockdown. Measurement of 5hmc and 5mc at promoter region of KLHDC8B locus revealed decreased 5hmc level concurrent with increased 5mc level. Importantly, knockdown of KLHDC8B in CD34+ cells as with knockdown on TET3 led to generation of increased numbers of bi/multinucleated polychromatic/orthochromatic erythroblasts and impaired enucleation implying a role for this protein in cytokinesis of late stage but not early stage erythroblasts. These findings demonstrate that TET3 regulates erythropoiesis in a stage-specific manner by targeting different set of genes. Importantly, knockdown of TET2 led to phenotypic changes that were very different from that seen following knockdown of TET3 but the observed changes are similar to the erythroid development defects noted in myelodysplastic syndromes (MDS). These include hyper-proliferation of early stage erythroid cells; delayed terminal erythroid differentiation and increased apoptosis of late stage erythroblasts. Together with the fact that TET2 gene mutation is one of the most common mutations in MDS and dyserythropoiesis is a hallmark of this disorder, our findings suggest that TET2 gene mutations can directly account for dyserythropoiesis of MDS. Our findings demonstrate distinct and important roles for TET2 and TET3 in regulating erythropoiesis and provide significant new and novel insights into epigenetic regulation of erythropoiesis at distinct development stages. The findings are likely to be very useful for furthering our understanding of epigenetic regulation of normal and disordered human erythropoiesis. Disclosures No relevant conflicts of interest to declare.


PLoS Genetics ◽  
2021 ◽  
Vol 17 (4) ◽  
pp. e1009499
Author(s):  
Jordan J. Tyrrell ◽  
Jackson T. Wilbourne ◽  
Alisa A. Omelchenko ◽  
Jin Yoon ◽  
Lina Ni

Temperature sensation guides animals to avoid temperature extremes and to seek their optimal temperatures. The larval stage of Drosophila development has a dramatic effect on temperature preference. While early-stage Drosophila larvae pursue a warm temperature, late-stage larvae seek a significantly lower temperature. Previous studies suggest that this transition depends on multiple rhodopsins at the late larval stage. Here, we show that early-stage larvae, in which dorsal organ cool cells (DOCCs) are functionally blocked, exhibit similar cool preference to that of wild type late-stage larvae. The molecular thermoreceptors in DOCCs are formed by three members of the Ionotropic Receptor (IR) family, IR21a, IR93a, and IR25a. Early-stage larvae of each Ir mutant pursue a cool temperature, similar to that of wild type late-stage larvae. At the late larval stage, DOCCs express decreased IR proteins and exhibit reduced cool responses. Importantly, late-stage larvae that overexpress IR21a, IR93a, and IR25a in DOCCs exhibit similar warm preference to that of wild type early-stage larvae. These data suggest that IR21a, IR93a, and IR25a in DOCCs navigate early-stage larvae to avoid cool temperatures and the reduction of these IR proteins in DOCCs results in animals remaining in cool regions during the late larval stage. Together with previous studies, we conclude that multiple temperature-sensing systems are regulated for the transition of temperature preference in fruit fly larvae.


1997 ◽  
Vol 35 (11-12) ◽  
pp. 451-453
Author(s):  
F. X. Abad ◽  
A. Bosch ◽  
J. Comas ◽  
D. Villalba ◽  
R. M. Pintó

A method has been developed for the detection of infectious human rotavirus (HRV), based on infection of MA104 and CaCo-2 cell monolayers and ulterior flow cytometry. The sensitivity of the flow cytometry procedure for the cell-adapted HRV enabled the detection of 200 and 2 MPNCU in MA104 and CaCo-2 cells, respectively. Flow cytometry performed five days after infection of CaCo-2 enabled the detection of naturally occurring wild-type HRV in faecal samples and concentrated water samples.


Sign in / Sign up

Export Citation Format

Share Document