Role of Casein Kinase 2 in Platelets Release from Megakaryocytes.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1534-1534
Author(s):  
Alieta Ciocea ◽  
Tatiana V. Byzova ◽  
Michael Kalafatis

Abstract Megakaryoblasts are precursors of platelets. Megakaryoblasts first differentiate to the stage of megakaryocytes. Megakaryocytes mature by increasing their size and by undergoing nuclear endoreplication and cytoplasmic maturation. Mature megakaryocytes form pseudopodia and reach pro-platelet bearing stage. Proplatelets bearing megakaryocytes fragment to give rise to platelets, through the process of thrombocytopoiesis. Both the thrombocytopoiesis and the megakaryocytopoiesis processes are linked to the constitutive apoptosis of megakaryocytic cells. The BCR/ABL oncoprotein is the result of a chromosomal translocation known as Philadelphia positive chromosome (Ph+). This is the main cause of several myeloproliferative disorders, like chronic myelogenous leukemia (CML). In CML the rate of platelet production is abnormal due to malignant megakaryoblast overproliferation. Casein kinase 2 (CK2) plays a crucial role in CML. CK2 was found to interact with BCR/ABL and to modulate its function. CK2 is also involved in many pathways downstream of BCR/ABL. CK2 acts on apoptotic pathways and also in the blood coagulation cascade. We studied the effect of casein kinase 2 alpha subunit (CK2α) inhibition in a megakaryoblastic cell line from a CML patient in blast crisis (MEG-01). We found that these inhibitors induce proliferation arrest while maintaining a steady cell number for a period of one week. Treated cells grew at a lower and constant rate than the non-treated ones, which are extremely prolific. Apoptosis of MEG-01 was induced by CK2 inhibitors, and this phenomenon was dose and time dependent. No necrosis was detected in the presence of the inhibitors, demonstrating that such compounds are not cytotoxic. Treatment of MEG-01 cells with CK2 inhibitors resulted in cells with apoptotic features like blebbing and specific apoptotic DNA fragmentation. Interestingly, these treatments lead to stimulation of megakaryocytopoiesis and thrombocytopoiesis processes in MEG-01 megakaryoblasts. In the presence of CK2 inhibitors megakaryocytes matured to the pro-platelets bearing stage. Platelets were released through rupture, following cytoplasmic fragmentation and nuclear extrusion. Thrombocytopoiesis due to the use of CK2 inhibitors occurred both in suspension as well as with MEG-01 cells grown on a fibronectin matrix. Platelets obtained following these treatments were harvested and analyzed for function. Such platelets were identified as anucleated cells by flow cytometry and microscopy techniques using DAPI and PI staining. These platelets were found to undergo shape change in response to various agonists (human thrombin, TRAP, ADP, PMA, fibronectin). MEG-01-derived platelets were also found to stain positive with CD41a antibody (αIIbβ3) as determined by flow cytometry using a monoclonal antibody specific to the receptor. Following agonist activation, the platelets expose phosphatidylserine and P-Selectin. The specific use of PAC-1, an antibody that only recognizes the activated form of αIIbβ3, together with fibrinogen binding studies, demonstrated that the MEG-01-derived platelets can be activated. Fibronectin, RGDS and EDTA inhibited activation of MEG-01-derived platelets. Addition of human thrombin (5 U/ml) and fibrinogen to these platelets resulted in a stable clot. These findings suggest that CK2 is involved in MEG-01 differentiation, platelets production, and activation. Thus, by using a CK2 inhibitor we have successfully stopped the abnormal proliferation of a transformed cell line and reversed the path towards its normal function.

2019 ◽  
Vol 18 (11) ◽  
pp. 1551-1562 ◽  
Author(s):  
Abbas Kabir ◽  
Kalpana Tilekar ◽  
Neha Upadhyay ◽  
C.S. Ramaa

Background: Cancer being a complex disease, single targeting agents remain unsuccessful. This calls for “multiple targeting”, wherein a single drug is so designed that it will modulate the activity of multiple protein targets. Topoisomerase 2 (Top2) helps in removing DNA tangles and super-coiling during cellular replication, Casein Kinase 2 (CK2) is involved in the phosphorylation of a multitude of protein targets. Thus, in the present work, we have tried to develop dual inhibitors of Top2 and CK2. Objective: With this view, in the present work, 2 human proteins, Top2 and CK2 have been targeted to achieve the anti-proliferative effects. Methods: Novel 1-acetylamidoanthraquinone (3a-3y) derivatives were designed, synthesized and their structures were elucidated by analytical and spectral characterization techniques (FTIR, 1H NMR, 13C NMR and Mass Spectroscopy). The synthesized compounds were then subjected to evaluation of cytotoxic potential by the Sulforhodamine B (SRB) protein assay, using HL60 and K562 cell lines. Ten compounds were analyzed for Top2, CK2 enzyme inhibitory potential. Further, top three compounds were subjected to cell cycle analysis. Results: The compounds 3a to 3c, 3e, 3f, 3i to 3p, 3t and 3x showed excellent cytotoxic activity to HL-60 cell line indicating their high anti-proliferative potential in AML. The compounds 3a to 3c, 3e, 3f, 3i to 3p and 3y have shown good to moderate activity on K-562 cell line. Compounds 3e, 3f, 3i, 3x and 3y were found more cytotoxic than standard doxorubicin. In cell cycle analysis, the cells (79-85%) were found to arrest in the G0/G1 phase. Conclusion: We have successfully designed, synthesized, purified and structurally characterized 1- acetylamidoanthraquinone derivatives. Even though our compounds need design optimization to further increase enzyme inhibition, their overall anti-proliferative effects were found to be encouraging.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 4328-4328
Author(s):  
Alieta Ciocea ◽  
Natalya V. Narizhneva ◽  
Evrim Erdogan ◽  
Tatiana V. Byzova ◽  
Michael Kalafatis

Abstract Megakaryocytes are polyploid cells, originating from hematopoietic stem cells in the bone marrow. Megakaryocytopoiesis is the process of production of anucleated cells, the platelets, from megakaryocytes. Megakaryocytes undergo endomitosis and maturation to the stage of proplatelets bearing megakaryocytes, which fragment to give rise to platelets. Platelets are vital for both maintaining normal hemostasis and for the response of the human body to trauma. Apoptosis is a genetically programmed and evolutionary conserved mechanism through which the normal development and tissue homeostasis are maintained. The platelets production is the result of constitutive apoptosis of megakaryocytes. Casein kinase II (CKII) is a pleiotropic, ubiquitous ectokinase that phosphorylates Serine, Threonine and Tyrosine amino acid residues. CKII has two catalytic subunits, α and α’ and two regulatory β subunits. The upregulation and hyperactivity of CKII has a general anti-apoptotic, pro-survival function in different overproliferative and carcinogenic processes. In chronic myelogenous leukemia (CML), specifically, CKII upregulation and hyperactivity induces stem cells increased proliferation. This results in bone marrow and circulating high blast levels and abnormal blood cells counts (trombocytosis, leukocytosis). We used the megakaryoblastic CML cell line, MEG-01, to study the effect of the inhibition of CKII, by CKII α subunit specific inhibitors, on platelet formation and cell proliferation. MEG-01 cells, which are defined as cytokine independent, are unresponsive to thrombopoietin and interferon. Incubation of MEG-01 cells with two different CKII inhibitors results in proliferation arrest, megakaryopoiesis, apoptosis and megakaryocytopoiesis. Platelet release from MEG-01 cells strongly depends on inhibitors concentration and treatment length. The platelets obtained from MEG-01 cells, following incubation with CKII inhibitors, get activated when stimulated by agonists, like phorbol-12-mystrate-13-acetate (PMA) and the peptide TRAP. These agonist-activated platelets undergo shape change and expose P-Selectin as a consequence of their activation. The platelet population stains positive for GpIIb/IIIa complex. Small size and negative staining for propidium iodide clearly distinguish platelets from megakaryocytes. In conclusion, we found a novel activator of platelet release from megakaryocytes. PMA, used as a megakaryocytopoiesis inducer, is known to stimulate release of functional platelets from MEG-01 cells, with normal blood platelet morphology. However, PMA, a very potent tumor promoter, induces cutaneous squamous cell carcinoma in mice. Our findings are important and demonstrate that platelets release from MEG-01 cells is a form of apoptosis that can be initiated following specific inhibition of CKII α subunit. Therefore, the processes of megakaryocytopoiesis and megakaryopoiesis are controllable through the fine-tuning of apoptosis. These results are original and of high physiological relevance since the high blast cell percentage can be decreased by proliferation arrest and differentiation. So the malignant processes in this type of leukemia can be stopped by controlling the stem cell number and their proliferation rate following inhibition of CKII α subunit. Thus, regulation of CKII α catalytic subunit may play a very important role in the future treatment of patients with CML that are resistant to the currently available treatments.


1986 ◽  
Vol 164 (1) ◽  
pp. 251-262 ◽  
Author(s):  
L S Park ◽  
D Friend ◽  
S Gillis ◽  
D L Urdal

125I-labeled recombinant human GM-CSF was used to identify and characterize receptors specific for this lymphokine on both a mature primary cell, human neutrophils, and on the undifferentiated promyelomonocytic leukemia cell line, HL-60. Human GM-CSF also bound to primary human monocytes and to the myelogenous leukemia cell line, KG-1, but not to any of the murine cells known to express the murine GM-CSF receptor. In addition, although some murine T lymphomas can express the GM-CSF receptor, none of the human cell lines of T cell lineage that we examined bound iodinated human GM-CSF. Binding to all cell types was specific and saturable. Equilibrium binding studies revealed that on all cell types examined, GM-CSF bound to a single class of high affinity receptor (100-500 receptors per cell) with a Ka of 10(9)-10(10)/M. More extensive characterization with neutrophils and HL-60 cells showed that in both cases, binding of GM-CSF was rapid at 37 degrees C with a slow subsequent dissociation rate that exhibited marked biphasic kinetics. Among a panel of lymphokines and growth hormones, only human GM-CSF could compete for binding of human 125I-GM-CSF to these cells. GM-CSF can not only stimulate the proliferation and differentiation of granulocyte/macrophage precursor cells, but can modulate the functional activity of mature granulocytes and macrophages as well. No significant differences in the kinetic parameters of receptor binding were seen between mature neutrophils and the undifferentiated promyelocytic leukemia cell line HL-60, indicating that maturation-specific responses to GM-CSF are not mediated by overt changes in the binding characteristics of the hormone for its receptor.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4930-4930
Author(s):  
Joshua Acklin ◽  
Mitra Azadniv ◽  
Scott Portwood ◽  
Kimberly Lacagnina ◽  
Charlotte Beers ◽  
...  

Abstract Introduction : Mammalian target of rapamycin (mTOR) signaling has previously been identified as a possible therapeutic target in acute myelogenous leukemia (AML), as the PI3K/AKT/mTOR pathway has been shown to be upregulated in blasts of up to 90% of AML cases through the constitutive loss of the tumor suppressor PTEN. The activation of this pathway is implicated in synthesis of prosurvival transcription and translational factors responsible for cellular resistance to apoptosis, resistance to chemotherapy, and enhanced survival in the marrow microenvironment. Unfortunately, exploitation of this pathway has been largely ineffective in clinical studies. A second mTOR complex (mTORC2) will reestablish the activation of the first when the first mTOR complex (mTORC1) is shut down under treatment with traditional rapalogs, such as rapamycin and everolimus. Additionally, the cellular process of autophagy has been postulated to be a protective mechanism for leukemic blasts during treatment with mTOR inhibitors, therefore decreasing their efficacy. For these reasons, this work explores means to optimize mTOR pathway inhibition by examining effects of dual mTORC inhibition (OSI027), dual PI3K/mTOR inhibition (BEZ235/Dactilosib), combination of mTOR inhibitors with a histone deacetylase inhibitor with ability to inhibit AKT (LBH589/Panobinostat), and the combination of mTOR inhibitors with hypomethylating agents able to overcome the hypermethylation of mTOR pathway components such as TSC1, TSC2 and PTEN (decitabine/5-azacytidine). Materials and Methods : Primary AML leukapheresis samples obtained with informed consent were cultured for 48 hours in the presence of OSI027, LBH589 (Novartis), BEZ235 (Novartis), decitabine, or OSI027 in combination with each of these individual inhibitors and compared with control culture conditions. Cells were stained with annexinV and DAPI as previously described, and the percent of living, pre-apoptotic, necrotic and dead cells were determined via flow cytometry. Synergy calculations were completed using Calcusyn™ software as described by Chao and Talalay (1977). Autophagic flux was determined in the OSI027 and LBH589 combination exposure via flow cytometry using anti-LC3. Lastly, protein expression under treatment with various combinations was determined via standard western blotting techniques. Results: The MV411 AML cell line was utilized to establish efficacy of combination exposures. These cells expressed phosphorylated (p)Akt (ser 473), p4EBP1, and pmTOR. OSI027 10µM, LBH589 1µM, and BEZ235 10nM were able to inhibit pmTOR, pAKT, and p4EBP1 expression as determined by western blotting. In the MV411 cell line, IC50s were determined, and combination indices were determined for OSI027+BEZ235, panobinostat+BEZ235, and panobinostat+OSI027; all of which demonstrated synergy. In primary AML samples, the combination of OSI027 10uM and LBH589 10nM significantly increased the percentage of dead cells in comparison to OSI027 treatment alone. Additionally in primary AML blasts, pmTOR and pAKT expression were decreased in the combination of OSI027 and LBH589 when compared to single agent treatment via western blot. The combination of OSI027 and LBH589 demonstrated a significant reduction in the autophagic flux in comparison to OSI027 treatment alone, suggesting an anti-autophagic effect, which correlates with the increased rates of cellular death in the combination. It was also found in AML cell lines that the combination of 10 µM azacytidine and 100 nM rapamycin resulted in synergistic suppression of U937, MV411, and KG1a survival as measured by MTT. No synergy was noted in the HL60 cell line. This combination effectively suppressed CFU-L in primary AML cases and suppressed p70S6K and p4EBP1 expression more than either agent alone by western blotting. The combination of decitabine 500nM with OSI027 10uM did not demonstrate an increase in apoptotic cells in primary blasts compared to OSI027 alone. Conclusions : Based on these findings, we conclude that mTOR inhibition in AML cell lines and primary samples can be enhanced through dual mTORC inhibition, PI3K modulation, and histone deacetylase inhibition. This may occur through suppression of activated pathway mediators or through suppression of autophagic flux. These results suggest that there is merit in exploring these combinations for therapeutic potential in AML. Disclosures Portwood: ImmunoGen: Research Funding. Wang:ImmunoGen: Research Funding.


2020 ◽  
Author(s):  
Andreas Krämer ◽  
Christian Georg Kurz ◽  
Benedict-Tilman Berger ◽  
Ibrahim Ethem Celik ◽  
Stefan Knapp ◽  
...  

ABSTRACTCasein kinase 2 (CK2) is a constitutively expressed serine/threonine kinase that has a large diversity of cellular substrates. Thus, CK2 has been associated with a plethora of regulatory functions and dysregulation of CK2 has been linked to disease development in particular to cancer. The broad implications in disease pathology makes CK2 an attractive target. To date, the most advanced CK2 inhibitor is silmitasertib, which has been investigated in clinical trials for treatment of various cancers, albeit several off-targets for silmitasertib have been described. To ascertain the role of CK2 inhibition in cancer, other disease and normal physiology the development of a selective CK2 inhibitor would be highly desirable. In this study we explored the pyrazolo[1,5-a]pyrimidine hinge-binding moiety for the development of selective CK2 inhibitors. Optimization of this scaffold, which included macrocyclization, led to IC20 (31) a compound that displayed high in vitro potency for CK2 (KD = 12 nM) and exclusive selectivity for CK2. X-ray analysis revealed a canonical type-I binding mode for IC20. However, the polar carboxylic acid moiety that is shared by many CK2 inhibitors including silmitasertib was required for potency and reduced somewhat cellular activity. In summary, IC20 represents a highly selective and potent inhibitor of CK2, which can be used as a tool compound to study CK2 biology and potential new applications for the treatment of diseases.NotesThe authors declare no conflict of interest.TOC Figure / Graphical Abstract


Stroke ◽  
2017 ◽  
Vol 48 (suppl_1) ◽  
Author(s):  
Sylvain Brunet ◽  
Chinthasagar Bastian ◽  
Danielle Aquila ◽  
Selva Baltan

Axonal injury and dysfunction are responsible for much of the disability observed following a stroke. Human brain comprises equal proportions of gray matter and white matter and white matter is injured in most strokes. Casein kinase 2 (CK2) is a protein kinase expressed in brain, including white matter, and is regulated by ischemia. We therefore hypothesized that transient CK2 inhibition would protect white matter from ischemic injury. To assess the impact of CK2 inhibition on axonal electrical activity following oxygen glucose deprivation (OGD), mouse optic nerves (MONs), a pure white matter track, from C57BL/6J were subjected to OGD (1h) while eliciting compound action potentials (CAPs) and exposed to CX-4945, a selective CK2 inhibitor, or control artificial cerebrospinal fluid (ACSF). We observed that CX-4945 preserved CAPs when applied either before or after OGD. Then to determine the impact of CK2 inhibition on glial cell survival following OGD, MONs exposed to OGD that were treated with either CX-4945 or control ACSF were processed for immunohistochemistry. We observed that CX-4945 treatment protected oligodendrocytes from OGD. And finally, to determine if CK2 inhibition protected mitochondrial from OGD, MONs from Thy-1 mito-CFP mice were similarly subjected to OGD in the presence of either CX-4945 or control ACSF. We observed that CX-4945 maintained Thy-1 mito-CFP fluorescence following OGD. In conclusion, our results suggest that CK2 inhibition preserves axonal function by preserving oligodendrocytes and mitochondrial function following ischemic injury. We propose that CK2 inhibitors, which are currently in phase II-III clinical trials for cancer therapy could be repurposed and provide a novel therapeutic target to protect white matter against ischemic injury, reducing mortality and morbidity and improving recovery following stroke.


2020 ◽  
Vol 15 (3) ◽  
pp. 209-215
Author(s):  
Luka Horvat ◽  
Mariastefania Antica ◽  
Maja Matulić

Background:: Casein Kinase 2 (CK2) is a Ser/Thr protein kinase that coregulates a great number of signalling pathways in the cell. It is involved in cell cycle regulation and cell proliferation, apoptosis, DNA damage response and gene transcription. Its substrates are numerous kinases and transcription factors. It was found to be upregulated in different tumours, and certain types of leukaemia are very sensitive to its inhibition. Objective:: We analysed the effects of casein kinase 2 inhibition on three leukaemia cell lines of B and T cell origin: Jurkat, a T cell line, CLL, a chronic B lymphocytic leukaemia cell line and 697, a pre-B acute lymphocytic leukaemia cell line. Besides cell proliferation and cytotoxicity analysis, the aim was to investigate the influence of CK2 inhibition on elements of the Notch signalling pathway. Notch signalling has an important role in blood cell differentiation, and CK2 regulates Ikaros, a tumour suppressor interfering with Notch signalling Methods:: and T leukaemia cells were treated with different concentrations of the CK2 inhibitor, CX-4945, for 6 days, and cell viability and proliferation were determined by Trypan Blue Exclusion Method. Analysis of gene expression was performed by RT-qPCR. Results:: All three cell lines were sensitive to CK2 inhibition and among them, 697 cells had two times lower IC50. In Jurkat and CLL cells changes in c-Myc and Notch pathway gene expression were found. Conclusion:: As CK2 is involved in numerous signalling circuits, we concluded that each cell type could have a cell-specific response in gene expression.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 4039-4039
Author(s):  
John T Patton ◽  
Theodore Smith ◽  
Leonard D. Shultz ◽  
John L. Magnani

Abstract Most patients with acute myelogenous leukemia (AML) initially respond to chemotherapy but later relapse and die from the disease (50–70%). One mechanism by which AML cells resist treatment with anti-proliferative drugs is by cell adhesion in protective microenvironments in areas such as the bone marrow. Here, we investigate the effects of GMI-1070 on the mechanism of adhesion of an AML cell line (MV-4-11) derived from biphenotypic myelomonocytic cells. GMI-1070 is a small molecule rationally designed pan-selectin antagonist with particularly strong activity for E-selectin (IC50 = 3.4 μM). Over 95% of these AML cells express ligands for E and P-selectins as determined by fluorescence-activated cell sorter analysis using E or P-selectin/hIg chimeras. Virtually all of these cells strongly express the specific E-selectin ligands CD65 and FH-6 using flow cytometry. The ability of these AML cells to roll and adhere to E or P-selectin expressed on monolayers of human endothelial cells (HUVECs) under the shear forces of normal blood flow was determined in vitro using flow chambers. The data was captured by videomicroscopy and processed using digital imaging. AML cells rolled and adhered to endothelium expressing E or P-selectin and both forms of adhesion are inhibited by GMI-1070. In contrast, much less rolling and adhesion was observed by multiple myeloma cell line U-266 only on endothelial cells expressing P-selectin although these specific interactions are also inhibited by GMI-1070. To analyze the adhesion, homing and infiltration of this AML cell line in vivo, recently developed NOD-scid IL2rgnull mice were used as hosts for in vivo experimentation. These mice lack adaptive immune function, are completely deficient in host natural killer cells and support long term growth of primary human leukemias (AML, CML, and ALL). Mice were injected i.v. with MV-4-11 AML cells (1 × 106) and blood samples were taken from the retro-orbital plexus at 1 and 2 weeks post injection. Blood counts were performed using an ADVIA Hematology Analyzer. At intervals post injection, the presence of AML cells in the bone marrow (BM), spleen, and peripheral blood was determined by flow cytometry of cells co-expressing human CD45 and human CD33. At 3 weeks, AML cells represented 26% of cells in the bone marrow and 2.3% of cells in the spleen. At 6 weeks, high percentages of AML cells were found in both tissues as well as in the blood (bone marrow, 83%; spleen, 69%; blood, 36%). Histological analyses showed accumulations of AML cells in the BM, spleen, lungs, liver, kidneys, and ovaries. At these later stages, hematopoietic cells in the BM cavity were largely replaced by sheets of leukemic cells. This model proved to be effective in studying the homing of an AML cell line to protective environments in the bone marrow and dissemination of disease to other organs and is being used to determine the effects on cell adhesion-mediated drug resistance (CAM-DR) by selectin-mediated inhibition of adhesion using small molecule antagonist, GMI-1070. GMI-1070 is currently in Phase I clinical trials.


Diabetes ◽  
1985 ◽  
Vol 34 (4) ◽  
pp. 347-352 ◽  
Author(s):  
T. Yamanouchi ◽  
T. Tsushima ◽  
Y. Akanuma ◽  
M. Kasuga ◽  
H. Mizoguchi ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document