The Neuronal Protein GAP-43 Associates with Microtubules and Lipid Rafts in Platelets and Megakaryocytes and Is Involved in Platelet Genesis

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2201-2201
Author(s):  
Antonija Jurak Begonja ◽  
Lilly Nguyen ◽  
Joseph E. Italiano ◽  
Herve Falet ◽  
Karina F. Meiri ◽  
...  

Abstract Abstract 2201 GAP-43 (neuromodulin) is a membrane-associated protein involved in neurite outgrowth, pathfinding and branching during development where it regulates neurotransmitter release and endocytosis, and plays a role in cytoskeletal signal transduction at the nerve ending. We have found GAP-43 to be expressed in platelets and megakaryocytes (MK) as evidenced by immunoblot and flow cytometry analysis. Immunofluorescence shows that GAP-43 localizes intracellularly as discrete punctuate structures mostly near the marginal microtubule (MT) coil of platelets and along the MT bundles in MKs and proplatelets. In contrast to neuronal cells where GAP-43 is associated with their detergent insoluble F-actin cytoskeleton, it is mostly soluble in platelets unless the platelets are first treated with taxol, a drug that stabilizes MTs. MT disruption with nocodazole or by chilling decreases the amount of GAP-43 in the detergent-insoluble cytoskeleton and disturbs its localization in proplatelets. GAP-43 colocalizes in platelets and MKs with the glycosylphosphatidylinositol-linked lipid raft marker Thy-1 and Cherry-PH PLC-delta1 (pleckstrin homology domain of phospholipase C-delta1) that stains membrane rafts. Since GAP-43 overexpression in MKs diminishes proplatelet formation, it appears to play a negative role in thrombopoiesis. Together, the data shows that GAP-43 is associated with MT and lipid rafts and suggests a role in signaling reactions between these two components to modulate proplatelet elaboration. Disclosures: No relevant conflicts of interest to declare.

2006 ◽  
Vol 34 (3) ◽  
pp. 374-376 ◽  
Author(s):  
E.B. Babiychuk ◽  
A. Draeger

The spatial segregation of the plasma membrane plays a prominent role in distinguishing and sorting a large number of signals a cell receives simultaneously. The plasma membrane comprises regions known as lipid rafts, which serve as signal-transduction hubs and platforms for sorting membrane-associated proteins. Ca2+-binding proteins of the annexin family have been ascribed a role in the regulation of raft dynamics. Glycosylphosphatidylinositol-anchored 5′-nucleotidase is an extracellular, raft-associated enzyme responsible for conversion of extracellular ATP into adenosine. Our results point to a regulation of ecto-5′-nucleotidase activity by Ca2+-dependent, annexin-mediated stabilization of membrane rafts.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 7-7
Author(s):  
Lin Yuehui ◽  
Qinglong Zheng ◽  
Tong Wu ◽  
DAN LIU

Background:The progression of Philadelphia chromosome positive chronic myeloid leukemia (CML) is frequently accompanied by cytogenetic evolution, commonly unbalanced chromosomal changes. but balanced chromosomal translocations are very rare in CML, especially translocations involving the 11q23. The few reported cases with blast phase (BP) of CML carrying a 11q23 rearrangement results in insufficient responses to tyrosine kinase inhibitors (TKIs) and possess a poor prognosis. Methods:Cytogenetic analysis , fluorescence in situ hybridization (FISH), RNA sequencing (RNA-seq), targeted genomic sequencing and multi-parametric flow cytometry analysis were performed to identify the chromosome translocations and pathogenic gene alterations in a 36-year-old female with myeloid BP of CML. Results: In BP, the bone marrow (BM) aspiration showed 61% myeloid blasts; Multi-parametric flow cytometry analysis revealed the abnormal myeloid blasts expression of the following antigens: CD117, CD13, CD33, CD38, partially expressed CD15, CD64. Chromosome analysis revealed a t(11;22)(q23;q11) translocation in addition to the t(9;22)(q34;q11). Fluorescence in situ hybridization (FISH) test confirmed that the t(11;22)(q23;q11) involved the mixed lineage leukemia(MLL) gene on 11q23 and RNA sequence revealed MLL-SEPT5 and BCR/ABL1(p210) fusion transcripts positive. Mutations on 339 commonly mutated genes in hematologic malignancies were analyzed by targeted next-generation sequencing showed ASXL1 p.G949Vfs*2 mutation. The patient failed to respond to both imatinib and dasatinib despite the absence of resistance-associated mutations in the BCR/ABL1 gene and she had a myeloid blast crisis at 19 months after initiation of first- and second-generation TKI treatment. After BP, she received ponatinb, a third-generation TKI with chemotherapy. Regretly she didn't achieve a complete remission(CR) and was in the process of salvage transplantation at present. Conclusions:The presence of 11q23 rearrangements in BC of CML is rare and most likely accounts for the adverse clinical outcome. We first report a patient who diagnosed CML with t(11;22)(q23;q11) and MLL-SEPT5 fusion gene positive in BP of CML. The clinical course was aggressive, and therapy was poorly tolerated. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1319-1319
Author(s):  
Massimiliano Bonifacio ◽  
Antonella Rigo ◽  
Angela Bonalumi ◽  
Emanuele Guardalben ◽  
Ilaria Nichele ◽  
...  

Abstract Abstract 1319 We have recently demonstrated that the sesquiterpene oil α-bisabolol is cytotoxic against primary acute leukemia cells ex vivo and in chronic myeloid leukemia cell lines. It enters cells via lipid rafts and activates the mitochondrial-dependent intrinsic pathway of apoptosis, exerting a preferential toxicity against malignant vs normal cells probably due to their higher content in lipid rafts. Here we investigated the in vitro activity of α-bisabolol in primary cells from patients with B-Chronic Lymphocytic Leukemia (B-CLL). Twenty-six patients with newly diagnosed B-CLL gave their informed consent to the study. Cells were collected before any treatment, purified and cultured for 24 hours with serial dilutions of α-bisabolol. Citotoxicity was quantified in flow cytometry by the BD Trucount™ technology to allow comparison between neoplastic and normal residual lymphocytes. B-CLL cells (IC50 42±15 μM) were significantly more sensitive towards α-bisabolol than normal B- (IC50 82±34 μM, p=.005) and T-cells (IC50 120±35 μM, p<.001). Citotoxicity was similar between the IgVH mutated (n=11) and the IgVH unmutated samples (n=7), as well as between the Binet stage A (n=20) and B-C (n=6) patients. To investigate the mechanisms of α-bisabolol-induced toxicity we treated B-CLL cells with 40 μM α-bisabolol for up to 3 hours. We observed a time-dependent increase in fluorescence of cells treated with the membrane-impermeant nucleic acid stain TO-PRO-3, already detactable after 30 minutes. When cells were loaded with the Ca2+ indicator Fluo-4 AM, an increase of Ca2+ influx was revealed already after 15 minutes. These early events indicate that α-bisabolol induces the loss of cellular membrane integrity, so triggering the apoptotic cascade. Then we assessed the mitochondrial transmembrane potential (ΔΨm) with the fluorochrome JC-1 to confirm that a mitochondrial damage is a concurrent mechanism in the apoptotic process induced by α-bisabolol. By flow cytometry we demonstrated that, after 3-hour incubation with 40 μM α-bisabolol, ΔΨm dissipation was already detectable in leukemic cells, while T-lymphocytes, evaluated as internal control in the same samples, stayed vital. To investigate the mitochondrial target of α-bisabolol we examined the function of the mitochondrial permeability transition pore (mPTP). After 5-hour incubation with 40 μM α-bisabolol we loaded cells with the calcein AM dye and added CoCl2 to distinguish between intact and damaged mitochondria, confirming that the function of mPTP was compromised in B-CLL cells but not in normal controls. Finally, to determine whether α-bisabolol affects the oxydative state of treated cells, we evaluated the intracellular concentration of reactive oxygen species (ROS) by measuring the fluorescent signal of CM-H2DCFDA loaded cells. When B-CLL cells were exposed to 40 μM a-bisabolol for 3 hours, they exhibited a clear fluorescence increase, indicating the striking generation of ROS: this was completely abrogated by the addition of N-acetylcysteine, a scavenger of intracellular ROS. Clues about the molecular mechanistics of α-bisabolol have also emerged from in vitro models based on treating cells previously transfected with BH3-only molecules. In this setting, α-bisabolol exposed cells seem to undergo detrimental, non-selective autophagy-like phenomena. Our data indicate that α-bisabolol exerts a level of cytotoxicity against B-CLL cells at concentrations that only partially affect normal B- and T-cells. Moreover, a brief exposure (3–5 hours) to α-bisabolol is sufficient to elicit multiple pro-apoptotic signals independently of the patients' mutational status. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 737-737
Author(s):  
Guilherme A dos Santos ◽  
Carolina H Thomé ◽  
Germano Aguiar Ferreira ◽  
Priscila S Scheucher ◽  
Antonio R Lucena-Araujo ◽  
...  

Abstract Abstract 737 Lipid rafts are highly ordered membrane domains that are enriched in cholesterol and sphingolipids and function in providing a scaffold to signal transduction. Altered raft assembly has been implicated in breast and colorectal cancer progression; rafts are essential to sustained p38K activation by GM-CSF and VEGFa in acute myeloid leukemia (Saulle et al. BJH, 2009). In a previous study, we showed that 10-(Octyloxy) decyl-2-(trimethylammonium) ethyl phosphate (ODPC), an alkylphospholipid, targets raft like domains in model membranes and induces apoptosis in leukemia cells, (ED-50 25mM - NB4), but spares normal hematopoietic and epithelial cells (dos Santos et al. BBACTA 2010). Here, we performed an unbiased quantitative proteomic screen of ODPC targets in lipid rafts. We used stable isotope labeling in cell culture (SILAC) to reveal proteins that are modulated by ODPC before the installation of apoptosis, to gain access to the primary mechanisms of drug action (Fig 1A, B and C). We used successive detergent extraction (Fig 1D) to isolate detergent resistant membranes (DRM) enriched in lipid rafts; blots with specific markers prove the quality of fractionation (Fig 1E). Proteins were considered modulated if: fold change of 2/0.5 or greater plus two or more quantification events and a P value <0.1. A small fraction of proteins was modulated by ODPC (Fig 1F). Of the 1426 proteins identified in the total extract, only one protein (0.07%) was upregulated. Of the 1314 proteins identified in the DRM, 6 proteins (0.45%) were donwregulated. Of these, 4 have predicted or validated palmitoylation sites (Fig 1F). We validated that ODPC downregulates LAT-2 (Fig 2A), an adaptor protein that is target to rafts by palmitoylation and is expressed in B cells and myeloid cells. Interesting, LAT-2 was not expressed in K562 (Fig 2A), a cell resistant to ODPC-induced apoptosis (Fig 1A and B). Moreover, ODPC induced a loss of LAT-2 in the lipid rafts of NB4 cells after 3h of treatment (Fig 2B). Since LAT-2 may function as an adaptor protein in AKT signalization (Roget et al. J.Immunol. 2008) we evaluate the effect of ODPC on the ATK pathway. Fig 2C shows that ODPC induces a downregulation of all components from AKT signaling, compatible with a membrane target drug. However, in functional experiments, ODPC inhibited AKT activation in NB4 cells by myeloid growth factors (MGF), after few minutes of incubation, in a similar way of the specific PI3K inhibitor Wortmannin (Fig 2D). Moreover, in these experiments, MGF induced a rapid and translation-dependent up regulation in LAT-2 expression in control treated cells (Fig 2E). This effect was partially suppressed by ODPC treatment and was suppressed by Wortmannin (Fig 2D). This suggests that LAT-2 translation is highly regulated and dependent on a functional AKT pathway. Indeed, phosphorylation of 4EBP-1 and ribossomal S6 kinase (S6K) with consequent upregulation of translation in the ribosome is downstream of PI3K/AKT/mTOR (Mamane et al. Oncogene, 2006). Moreover, we confirmed a hypophosphorilation state of S6K in the presence of ODPC or Wortmannin (Fig 2D). Interesting, we have identified two potential-palmitoylated ribosomal proteins (RP) to be downregulated in the lipid raft fraction of ODPC treated cells (Fig 1F). This finding confirms previous reports that a raft disruption treatment (methyl-β-cyclodextrin) causes downregulation of several RP (Foster, De Hoog and Mann. PNAS, 2003) and that palmitoylation target ribosome proteins to lipid rafts (Wei et al. MCP, 2010). This indicates that ODPC may present also a direct or indirect inhibitory effect on translational apparatus. Our results strongly indicate that ODPC acts on LAT-2 by interference with its protein-lipid interactions, since experimental-induced disturbances in lipid raft physiology recapitulate or increase ODPC effects on LAT-2 expression (Fig 3A and B). To further clarify the mechanism of LAT-2 downregulation we evaluate if it is degraded by the proteasome after ODPC or 2BrPA treatments. Fig 3C and D reveals that this is the case. This confirms the findings of Orr and McVicar (J.Leuk.Biol. 2011) of ubiquitin-proteasomal regulation of LAT-2 stability. Taken together, our results demonstrate that a lipid raft targeting drug may present several effects on signal transduction, causing a severe toxicity to leukemia cells. Moreover, adaptor proteins like LAT-2 emerge as possible new therapeutic targets in leukemia. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2133-2133
Author(s):  
Gheath Alatrash ◽  
Pariya Sukhumalchandra ◽  
Mao Zhang ◽  
Celine Kerros ◽  
Anna Sergeeva ◽  
...  

Abstract PR1 is an HLA-A2-retricted, nonameric peptide that is derived from the azurophil granule proteases neutrophil elastase (NE) and proteinase 3 (P3). PR1 has been targeted successfully in acute (AML) and chronic (CML) myeloid leukemia using anti-PR1/HLA-A2 antibody (8F4), PR1-peptide vaccine and PR1-specific cytotoxic T lymphocytes (PR1-CTL). We have previously reported that NE and P3 are cross-presented by normal B cells and dendritic cells (DC), leading to PR1 expression by HLA-A2. Since multiple myeloma (MM) is a B cell malignancy, we investigated whether MM cells can cross-present PR1 as a possible target for immunotherapy. To study whether PR1 is presented by MM cells, patient bone marrow (BM) was stained with 8F4 antibody and then imaged using confocal microscopy. PR1/HLA-A2 was detected on the surface of CD138+ MM cells in BM samples from 3 of 6 HLA-A2+ MM patients (Fig. 1). We then investigated whether cellular immunity to PR1 is detected in peripheral blood (PB) from patients with MM. PB samples from MM patients who had undergone allogeneic (allo) (n=9) and autologous (auto) (n=2) stem cell transplantation (SCT) were stained with PR1/HLA-A2 dextramer in addition to standard lineage markers. PR1-specific CD8+CTL were detected in PB of 10 of 11 patients (range=0.02%-2.9%). Because P3 and NE expression is limited to myeloid cells, we sought to determine the mechanism of PR1 presentation in MM. We performed RT-PCR and western blotting on seven MM cell lines, including U266, ARK, ARP-1, OPM-2, LP-1, IM-9 and RPMI 8226. Neither NE nor P3 were detected in the MM cell lines studied at either the mRNA or proteins levels. We then investigated whether MM cells took up NE and P3. We cultured MM cells for 30 hours with 10 ug/mL of soluble NE and P3 or irradiated HLA-A2 negative PMN, the latter as a source for cell-associated NE and P3. Cells were then stained intracellularly for NE and P3 at different time points. Flow cytometry analysis showed that all the cell lines analyzed took up NE and P3. Uptake was seen as early as 1 hour after co-culture with soluble NE and P3 and was higher for soluble P3. Additionally, more uptake was seen in the cells that were co-cultured with irradiated PMN in comparison with soluble NE and P3. We then investigated whether PR1 expression in MM was through NE and P3 cross-presentation. We focused our studies on the HLA-A2+ U266 MM cell line. U266 cells were co-cultured with soluble NE, P3 or irradiated PMN, as described in the previous section, and then surface stained with 8F4. We detected PR1/HLA-A2 on the surface of U266 cells by flow cytometry as early as 6 and 24 hours after co-culture with soluble NE and P3, respectively. Cross-presentation was also seen in the cells that were co-cultured with irradiated PMN to a similar extent in comparison with the cells that were cultured with soluble NE and P3, however, cross-presentation occurred at an earlier time point (1 hour) in the cells that were cultured with PMNs. Cross-presentation was abrogated by lactacystin, a proteasome inhibitor, and brefeldin A, an ER/Golgi transport inhibitor, indicating that NE and P3 cross-presentation occurs through conventional cross-presentation mechanisms. Furthermore, because immune modulatory drugs and proteasome inhibitors are part of the standard of care therapy for MM, and since both have been shown to affect cross-presentation by DC, we tested whether lenalidomide and bortezomib altered PR1 cross-presentation by U266 and normal DC. U266 and DC were cultured with irradiated PMN in the presence of increasing doses of bortezomib or lenalidomide and then stained with PR1/HLA-A2. Flow-cytometry analysis showed a significant inhibition of PR1-cross-presentation by U266 after addition of bortezomib, but not lenalidomide. PR1 cross-presentation by DC was not affected by either drug. Finally, we tested whether PR1 cross-presentation caused MM cells to become susceptible to PR1-targeting immunotherapies. U266 cells were cultured with NE or P3 for 24 hours, the time point that showed the maximal cross-presentation, followed by addition of 8F4 antibody or co-culture with PR1-CTL. Using calcein AM cytotoxicity assays, we showed dose dependent killing of U266 by 8F4 and PR1-CTL following PR1 cross-presentation (Fig. 2). Together our data show that PR1 is cross-presented by primary MM cells and cell lines. These findings lay the foundation for the future applications of PR1-targeting immunotherapies in MM. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4922-4922
Author(s):  
Bing Xu ◽  
Jiahong Tang ◽  
Feili Chen ◽  
Yanyan Li ◽  
Huijuan Dong ◽  
...  

Abstract Background Triptolide (TPL), a diterpenoid triepoxide derived from Tripterygium wilfordii, has been used in Traditional Chinese Medicine (TCM) for centuries. Recently, TPL has also been shown to have strong anti-cancer ability both in vitro and in vivo. It could induce apoptosis in a variety of cancer cell lines. However, the clinical applications of TPL are limited by its narrow therapeutic window and severe toxicity. Thus, the focus of study about TPL has been switched to the ability of low dose of TPL in enhancing the drug-induced apoptosis of cancer cells. KG1a cell line is a kind of cell line which has been demonstrated to have a certain proportion of leukemia stem cells which are responsible for the relapse of leukemia. Usage of this kind of cell line could resemble the relapse situation of patients. MDR1 and NF-ΚB are two important factors closely related to drug-resistance. Dow-regulation of these two genes could lead to the sensitization of cells to drugs. Aims This study aims to explore whether low-dose TPL could enhance the Idarubicin-induced apoptosis in KG1a cells through down-regulation of MDR1 and NF-κB. Methods Hoechst 33342 staining and flow cytometry analysis were used to determine the ability of TPL in enhancing the Idarubicin-induced apoptosis in KG1a cells. RT-PCR as well as western blotting analysis were used to determine whether TPL combined with Idarubicin could down-regulate MDR1 and NF-κB. Results Hoechst 33342 staining and flow cytometry analysis showed that low-dose of TPL((IC20F5.0nM)) could significantly enhance the Idarubicin-induced apoptosis in KG1a cells(Idarubicin alone vs Idarubicn combined with TPL: 15.13%: 60.17%; P<0.05). TPL combined with Idarubicin could down-regulate expression of MDR1 both at protein and mRNA level with the down-regulation of NF-κB at protein level. Conclusion Low-dose Triptolide could enhance the Idarubicin-induced apoptosis IN KG1a cells through down-regulation of MDR1 and NF-κB Disclosures: No relevant conflicts of interest to declare.


Endocrinology ◽  
2004 ◽  
Vol 145 (2) ◽  
pp. 613-619 ◽  
Author(s):  
Jayoung Kim ◽  
Rosalyn M. Adam ◽  
Keith R. Solomon ◽  
Michael R. Freeman

Abstract IL-6 is an inflammatory cytokine that has been linked to aggressive prostate cancer (PCa). Previous studies have demonstrated that IL-6 can enhance the differentiation of PCa cells toward a neuroendocrine (NE) phenotype, a possible indicator of hormone-refractory disease. In this report, we present evidence that the mechanism of IL-6-stimulated NE differentiation employs a detergent-resistant (lipid raft) membrane compartment for signal transduction in LNCaP PCa cells. Signal transducer and activator of transcription (STAT)3, a mediator of IL-6 signaling, was rapidly phosphorylated and translocated to the nucleus in LNCaP cells treated with IL-6. Both processes were inhibited by filipin, a cholesterol-binding compound that disrupts plasma membrane lipid rafts. Isolation of Triton X-100-insoluble raft fractions from LNCaP cells by discontinuous sucrose gradient centrifugation demonstrated that the 80-kDa IL-6 receptor localized almost exclusively to the raft compartment. Although STAT3 was located predominantly in the Triton X-100-soluble subcellular fraction in exponentially growing cells, abundant phosphorylated STAT3 was detected in the raft fraction after stimulation with IL-6. Increases in expression of the NE marker, neuron-specific enolase, and neuron-specific enolase promoter activity after IL-6 treatment were reduced after membrane rafts were disrupted by filipin treatment. LNCaP cells expressed the raft-resident proteins flotillin-2 and Giα2, but notably not caveolins, the predominant structural protein present in caveolar membrane rafts in many tissues and tumor cells. These results are the first to define a role for lipid raft membrane microdomains in signal transduction mechanisms capable of promoting the NE phenotype in PCa cells, and they demonstrate that the raft compartment is capable of mediating such signals in the absence of caveolins. Our results also suggest a mechanistic role for membrane cholesterol in cell signaling events relevant to PCa progression.


Molecules ◽  
2021 ◽  
Vol 26 (5) ◽  
pp. 1461
Author(s):  
Nuno Mariz-Ponte ◽  
Laura Regalado ◽  
Emil Gimranov ◽  
Natália Tassi ◽  
Luísa Moura ◽  
...  

Pseudomonas syringae pv. actinidiae (Psa) is the pathogenic agent responsible for the bacterial canker of kiwifruit (BCK) leading to major losses in kiwifruit productions. No effective treatments and measures have yet been found to control this disease. Despite antimicrobial peptides (AMPs) having been successfully used for the control of several pathogenic bacteria, few studies have focused on the use of AMPs against Psa. In this study, the potential of six AMPs (BP100, RW-BP100, CA-M, 3.1, D4E1, and Dhvar-5) to control Psa was investigated. The minimal inhibitory and bactericidal concentrations (MIC and MBC) were determined and membrane damaging capacity was evaluated by flow cytometry analysis. Among the tested AMPs, the higher inhibitory and bactericidal capacity was observed for BP100 and CA-M with MIC of 3.4 and 3.4–6.2 µM, respectively and MBC 3.4–10 µM for both. Flow cytometry assays suggested a faster membrane permeation for peptide 3.1, in comparison with the other AMPs studied. Peptide mixtures were also tested, disclosing the high efficiency of BP100:3.1 at low concentration to reduce Psa viability. These results highlight the potential interest of AMP mixtures against Psa, and 3.1 as an antimicrobial molecule that can improve other treatments in synergic action.


Sign in / Sign up

Export Citation Format

Share Document