Ck2 How Long Till Suicide Again

J Prison cell Biochem. Writer manuscript; available in PMC 2015 Dec 1.

Published in final edited form every bit:

PMCID: PMC4199905

NIHMSID: NIHMS619091

Protein kinase CK2 inhibition induces cell death via early impact on mitochondrial function*

Fatima Qaiser

ˆCellular and Molecular Biochemistry Enquiry Laboratory (151), Minneapolis Veterans Diplomacy Wellness Care System, Minneapolis, MN 55417

Department of Laboratory Medicine and Pathology, Army Medical Higher, National University of Sciences and Technology, Islamabad, Pakistan

Janeen H. Trembley

ˆCellular and Molecular Biochemistry Inquiry Laboratory (151), Minneapolis Veterans Affairs Health Care Organisation, Minneapolis, MN 55417

Department of Laboratory Medicine and Pathology, Regular army Medical Higher, National University of Sciences and Technology, Islamabad, Pakistan

§Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455

Betsy T. Kren

ˆCellular and Molecular Biochemistry Research Laboratory (151), Minneapolis Veterans Affairs Health Care Arrangement, Minneapolis, MN 55417

Department of Medicine, University of Minnesota, Minneapolis, MN 55455

Jing-Jiang Wu

ˆCellular and Molecular Biochemistry Research Laboratory (151), Minneapolis Veterans Diplomacy Wellness Care System, Minneapolis, MN 55417

Department of Laboratory Medicine and Pathology, Army Medical College, National University of Sciences and Technology, Islamabad, Pakistan

A. Khaliq Naveed

Department of Biochemistry & Molecular Biology, Army Medical College, National University of Sciences and Technology, Islamabad, Islamic republic of pakistan

Khalil Ahmed

ˆCellular and Molecular Biochemistry Research Laboratory (151), Minneapolis Veterans Affairs Health Intendance Organization, Minneapolis, MN 55417

Section of Laboratory Medicine and Pathology, Army Medical Higher, National University of Sciences and Technology, Islamabad, Pakistan

Abstract

CK2 (official acronym for casein kinase two or Ii) is a potent suppressor of apoptosis in response to diverse apoptotic stimuli —thus its molecular downregulation or action inhibition results in stiff induction of jail cell death. CK2 downregulation is known to impact mitochondrial apoptotic circuitry just the underlying mechanism(s) remain unclear. Utilizing prostate cancer prison cell lines subjected to CK2-specific inhibitors which cause loss of cell viability, we take institute that CK2 inhibition in cells causes rapid early on decrease in mitochondrial membrane potential (Δψthousand). Cells treated with the CK2 inhibitors TBB (4,5,6,vii-tetrabromobenzotriazole) or TBCA (tetrabromocinnamic acid) demonstrate changes in Δψthousand which become apparent within 2 h, i.e., significantly prior to evidence of activation of other mitochondrial apoptotic signals whose temporal expression ensues subsequent to loss of Δψm. Further, nosotros have demonstrated the presence of CK2 in purified mitochondria and it appears that the event on Δψthou evoked past inhibition of CK2 may involve mitochondrial localized CK2. Results also suggest that alterations in Ca2+ signaling may be involved in the CK2 mediated regulation of Δψm and mitochondrial permeability. Thus, nosotros advise that a key mechanism of CK2 touch on mitochondrial apoptotic circuitry and jail cell expiry involves early loss of Δψm which may be a main trigger for apoptotic signaling and cell decease resulting from CK2 inhibition.

Keywords: Apoptosis, mitochondrial membrane potential, mitochondrial permeability transition, prostate cancer, signaling

Poly peptide kinase CK2 is a highly conserved and ubiquitous protein Ser/Thr kinase consisting of ii catalytic subunits α and α′ and two regulatory β subunits with the catalytic subunits linked through the β subunits. CK2 has been found to play a function in a vast number of normal and abnormal cell functions and has emerged as a primal cellular regulator with a big number of substrates [Ahmed, 1999; Guerra and Issinger, 1999; Meggio and Pinna, 2003; Pinna, 2002; St-Denis and Litchfield, 2009; Tawfic et al., 2001]. In item, much piece of work has been undertaken regarding its function in cancer pathobiology and information technology is remarkable that CK2 has been found to be uniformly dysregulated in all cancers examined [Ahmed et al., 2000; Guerra and Issinger, 2008; Tawfic et al., 2001; Trembley et al., 2009]. While it was known for a long time that CK2 was elevated in rapidly growing normal and cancer cells, its role in cancer cells was unclear. Nosotros demonstrated that CK2, in add-on to its office in prison cell growth and proliferation, is also a stiff suppressor of apoptosis; further, the CK2α catalytic subunit is responsible for the majority of the apoptosis suppression [Ahmad et al., 2008; Ahmed et al., 2002; Guo et al., 2001; Tawfic et al., 2001]. Thus, the latter characteristic of CK2 provided an important link of this kinase to the cancer cell phenotype equally continued cell proliferation and resistance to cell expiry are two consequent features of cancer cell biological science [Hanahan and Weinberg, 2011].

Much evidence has suggested that elevated CK2 levels have a broad role in cell death suppression mediated through diverse signals [Ahmad et al., 2008]. In this context, we demonstrated that one fashion of CK2 mediated suppression of apoptosis involves the mitochondrial apoptotic circuitry. In these studies, we observed that downregulation of CK2 resulted in upregulation of Bax and downregulation of Bcl-2 and Bcl-xL accompanied with release of cytochrome c. These events were completely blocked when forced overexpression of CK2α was instituted [Wang et al., 2005a; Wang et al., 2006]. In subsequent studies information technology was observed that downregulation of CK2 activeness or expression in prostate cancer cells for six–24 h either by chemical inhibition or use of antisense to CK2α resulted in production of H2O2 hinting that this may initiate apoptotic signaling under these weather [Ahmad et al., 2006]. However, the mechanism underlying the consecration of apoptosis post-obit downregulation of CK2 is non fully understood, especially with respect to the earliest events that occur upon inhibition of CK2.

Here we have examined the event of CK2 inhibition on mitochondrial function and observed that, following the handling of cells with a chemical inhibitor of CK2, a change in the mitochondrial membrane potential (Δψm) is detected as early every bit 2 h, thus occurring significantly prior to activation of other apoptotic signals. While CK2 is known to be localized in the nuclear and cytoplasmic fractions, nosotros accept besides identified its presence in prostate jail cell mitochondria. Farther, our results provide show for the first time suggesting that inhibition of mitochondrial CK2 may be involved in causing rapid loss of Δψm as a primary event that triggers the mitochondrial apoptotic circuitry nether these conditions.

Materials and Methods

Materials

Sources of various chemicals and reagents used in these studies are as follows: BAPTA (Calbiochem); BSA (Thermo Fisher); Dulbecco's PBS (Gibco Life Technologies); bovine liver catalase (Sigma Aldrich); CCCP (Calbiochem); CellTiter 96® Aqueous One solution (Promega); FBS (Atlanta Biologicals); oligomycin (Sigma Aldrich); Matrigel® (BD Biosciences); TBB, TBCA and thapsigargin (EMD Millipore); trypsin + EDTA (Gibco Invitrogen); and JC-1 (Life Technologies). The following antibodies were used for western absorb assay: AKT-one phospho-Ser129 (ane:1000, Epitomics 5508-i); AKT-i (1:thousand, Cell Signaling 9272); actin (one:1000, Santa Cruz sc-1616); cleaved caspase-3 (1:g, Jail cell Signaling 9661); caspase-ix (1:g, Jail cell Signaling 9508); lamin A/C (ane:1000, Cell Signaling 2032); CKII α (one:3000, Bethyl Laboratories A300-197); CKII α′ (1:3000, Bethyl Laboratories A300-199); cytochrome c (1:10,000, Epitomics 2119-ane); Bax (1:1000, Cell Signaling 2772); Bid (ane:1000, Cell Signaling 2002); and Cox 4 (1:1000, Cell Signaling 4850).

Cell culture

The cell lines employed were PC3-LN4, LNCaP and C4-two (homo prostate cancer cell lines) and BPH-1 (human benign prostate epithelial cell line), as described previously [Slaton et al., 2004]. PC3-LN4 cells were maintained in RPMI 1640 media with 5% FBS, 2 mM glutamine, and one% penicillin-streptomycin (P-S), whereas LNCaP, C4-ii and BPH-one cells were maintained in RPMI 1640 with 10% FBS, 2 mM glutamine, and one% P-S [Trembley et al., 2012].

Prison cell fractionation

Cell pellets were suspended gently in 9 packed cell volumes of homogenization buffer A1 (ten mM Tris-HCl (pH 7.4), 5 mM MgCltwo, 25 mM KCl, 0.25 Grand sucrose) with phosphatase and protease inhibitors added at ane:200 only earlier use (Sigma Aldrich: P5726, P8340). The suspension was incubated for 10 min on ice to promote cell swelling after which the cells were ruptured using a Dounce homogenizer using 9 strokes with an "A" pestle. The intermission was centrifuged at 12,000 × grand for 30 min at 4 °C to remove the mitochondria. The supernatant (cytosolic fraction) was subjected to a second centrifugation at 12,000 × g for 30 min at iv °C. The last supernatant was filtered through a 0.2 μm Ultrafree MC filter (Millipore) by centrifuging at 12,000 × g for two min at four °C. Aliquots were flash frozen in liquid nitrogen.

Isolation of purified mitochondria and analysis of mitochondrial membrane permeability

Training of mitochondria from cultured prostate cells was carried out according the manufacturer's instructions (Pierce 89874). Training and purification of rat liver mitochondria was performed co-ordinate to a previously described procedure [Schnaitman and Greenawalt, 1968]. Analysis of mitochondrial permeability changes was carried out as described [Roughshod et al., 1991] utilizing the purified mitochondrial preparation resuspended in a medium consisting of 213 mM D-mannitol, 71 mM sucrose, and 3 mM HEPES buffer (pH 7.4). Details of weather used for analysis of mitochondrial swelling are outlined in the fable for Fig. 5.

An external file that holds a picture, illustration, etc.  Object name is nihms619091f5.jpg

Consequence of CK2 inhibitors on membrane permeability transition in isolated mitochondria

(A) Effect of TBB (left console) or TBCA (right panel) treatment on mitochondrial permeability transition. Purified mitochondria were subjected to various treatments as shown in the graph legends. Mitochondrial respiration was maintained using 10 mM Na-succinate as the substrate. When used, EGTA or cyclosporin A was added at fourth dimension zero; "inducers" were added at the start of the 5th min afterwards mitochondria equilibration in the incubation medium and baseline absorbance (540 nm) had been captured for iv min. Absorbance was measured for an additional 10 min following the addition of the various chemicals as indicated. The concentrations of various agents were: 80 μM TBB; lxxx μM TBCA; 70 μM Ca2+, three mM Pi; 0.5 μM cyclosporin A; 150 μM EGTA. A volume of DMSO equivalent to the volume of TBB/TBCA was used. All other details are every bit described under Materials and Methods. (B) Left panel, rat CK2α, CK2α′, CK2β′ and COX Four were detected in all preparations of rat liver mitochondria by western absorb analysis; 3 representative preparations are shown. Right panel, cytochrome c that was retained in the mitochondria or released in the medium corresponding to the various treatments nether A (left and right panels) was detected past western blot analysis.

Western absorb analysis

Whole jail cell and mitochondrial lysates prepared using RIPA buffer [Trembley et al., 2012] and cytosolic fractions in buffer A1 (50 μg) were subjected to SDS polyacrylamide gel electrophoresis using Tris-Glycine Laemmli gels. Proteins were transferred onto nitrocellulose membrane and v% non-fatty dairy milk in TBS/0.1% Tween 20 was used for blocking and antibody incubations.

Cell viability analysis

CellTiter 96® Aqueous One Analysis was used to assess cell viability following diverse treatments. Cells were plated in 96-well plates (4000 cells/well) and allowed to adhere overnight. Time course experiments were performed with incubation of cells in complete media with 8 or 80 μM TBB for 2, 4, half-dozen, 24 or 48 h. For experiments with TBCA, concentrations of 1, 10, 20, 40, and fourscore μM were applied for 24 and 48 h. Controls included untreated and DMSO treated cells. Aqueous One assay solution was combined with complete media at a ratio of 100 μ of media plus 20 μl of the assay solution per well, and cells were incubated for 3 h at 37 °C. Absorbance was measured at 490 and 700 nm using a Molecular Devices v plate reader with absorbance values for media alone subtracted from the experimental values.

Crystal violet clonal survival assays

Cells were treated with eight or fourscore μM TBB or equivalent volume of DMSO for four, half dozen, 8, and 24 h. Cells were trypsinized at the end of treatment catamenia and replated (in triplicate) using complete fresh media at a concentration of k cells per 35 mm plate for 7 d (media was replaced at 4 days). On twenty-four hour period seven cells were stained with crystal violet (i× PBS containing 1% (v/5) methanol, one% (5/v) formaldehyde and 0.05% (w/v) crystal violet) for 20 min, the stain removed, and plates washed by immersion in water with continuous h2o flow. Plates were air-dried, colonies (containing at least 50 cells/colony) were counted, and plates were scanned. The data shown represent the results of three replicates per experiment and a minimum of 3 experiments.

Coating of cover slips with Matrigel

Individual sterile encompass slips (22 × 22 mm) were placed in 6-well tissue civilisation plates and covered with 750 μl of sterile filtered Matrigel® (333 μg/ml; BD Biosciences 354234) in i× PBS. Open plates were placed overnight to dry in a laminar flow biosafety chiffonier. Plates were and so sealed with alkane series flick.

JC-1 fluorescence imaging

JC-one was employed every bit a marker for studying Δψm changes. Cells were plated on cover slips coated with Matrigel® in 6-well plates to reach 60–70% confluence the adjacent solar day. Each well was treated for 2, 4, six, or 24 h with TBB or TBCA at varying concentrations as indicated in the figure legends. All experiments included cells treated with equivalent volumes of DMSO or untreated as controls. When used, CCCP (10 μM) and oligomycin (2.five μg/ml) were added for 30 min prior to termination of the experiment. JC-1 was added at a concentration of 5 μg/ml in consummate media and cells were incubated at 37 °C in a 5% COii atmosphere for the final i or ii h depending on the experiment. Later, the cover slips were inverted onto slides containing Slow-Fade Golden (Invitrogen) solution. Images were taken immediately at twenty× and 40× magnification employing an Olympus BX60 conventional chemical compound fluorescence microscope. Ruby-red (emission = 519 nm) and green (emission = 565,615 nm) fluorescence were captured using appropriate filters. The information presented in the tables correspond counting cells in 4 fields (60 – 100 cells per field) per condition from 3 different experiments for the PC3-LN4/TBB, 2 different experiments for the PC3-LN4/TBCA, two different experiments for BPH-one/TBB, and 1 experiment for LNCaP/TBB.

JC-1 FACS analysis

JC-1 was added at a final concentration of 2 μM in complete media containing 8 or 80 μM TBB or equivalent volume of DMSO and the cells incubated at 37 °C in five% CO2 for 1 h for the 2 h time-point or 2 h for the 24 h time-point. CCCP at a final concentration of ten μM was added to untreated cells 30 min prior to termination of JC-ane labeling as a positive control. At the termination of labeling, the cells were detached using Tryple Express (Invitrogen), collected by centrifugation (5 min at 500 × g) at 25 °C, resuspended in 0.5 ml of 37 °C live prison cell imaging solution (Invitrogen) and immediately analyzed using a FACS ARIA 3 (Becton Dickinson) excitation at 488 nm, collecting data using the 530/30 nm bandpass filter (greenish) and 582/15 nm bandpass filter (red) with a minimum of twenty,000 gated events. The information were analyzed using the FACS ARIA III software.

Intracellular calcium assay

Measurement of intracellular Ca2+ concentration was performed using the FluoForte® calcium assay kit (Enzo Life Sciences). PC3-LN4 cells were plated in complete media at 1 × x5 cells/well using 96-well black wall plates. The following twenty-four hours, the cells were loaded with 0.1 ml dye/HBSS solution per well for 45 min at 37 °C/5 % CO2 according to kit instructions; BAPTA was included in the dye-loading solution for the appropriate wells. The plate was removed from the incubator and placed at room temperature for 15 min. Drug or diluent was first diluted from stock solutions into Catwo+ -free PBS and so added to every well in a volume of 25 μl to final concentration every bit indicated in Fig. 3B. The plate was immediately placed in a microplate reader (Molecular Devices 5, excitation 490 nm, emission 525 nm, cutoff 515 nm) and data collected every 1 min for 5 min. This experiment was performed three times.

An external file that holds a picture, illustration, etc.  Object name is nihms619091f3.jpg

Mitochondrial membrane potential change mediated past CK2 inhibition is blocked by pretreatment of cells with BAPTA but not catalase and is associated with transient release of intracellular Ca2+

(A) PC3-LN4 cells were treated with catalase (3000 units) or BAPTA (10 μM) for 1 h followed by treatment with lxxx μM TBB for 4 h. JC-1 loading was performed for the final one h of incubation. Controls included pretreatment for 1 h with drug solvent (DMSO for BAPTA; 50 mM potassium phosphate buffer pH 7.0 for catalase) followed by DMSO. Untreated cells were analyzed as a further control. Panels are accordingly labeled. Mitochondrial membrane potential was measured as described under materials and methods. (B) PC3-LN4 cells were loaded with FluoForte calcium assay dye, and with 10 μM BAPTA as indicated, for 1 h as described nether Materials and Methods. Drug or diluent was added to each well equally indicated in the legend (untreated cells received PBS diluent), and the plate immediately read once per min for v min. DMSO was used at the same concentration as 80 μM TBB. * p < 0.05 to untreated.

Statistical assay

The master cell line for written report of the various parameters was PC3-LN4, and most experiments using these cells were performed at least 3 times. The data are expressed as mean ± standard error (SE). To make up one's mind the generality of the results obtained for Δψm changes in PC3-LN4 cell line, they were confirmed in two other prison cell lines (LNCaP and BPH-1); in this example the experiments were performed 2 and 1 times, respectively. Statistical significance was analyzed for the Aqueous One prison cell viability and the intracellular Ca2+ assays using assay of variance with Bonferroni correction, and for clonal survival using t-tests.

Results

Effect of TBB on prostate cancer PC3-LN4 cell survival

Experiments were undertaken to determine the time class of the effect of inhibition of CK2 on cell viability in the presence of the relatively specific CK2 inhibitor TBB at varying concentrations. In these experiments we determined the effects of TBB handling for much shorter fourth dimension periods than previously examined. We employed the CellTiter 96® Aqueous I assay which measures the bioreduction (thought to occur via NADPH/NADH) of an MTS tetrazolium compound to a colored formazan product whose quantity as measured by absorbance at 490 nm is directly proportional to the number of metabolically active cells [Berridge and Tan, 1993]. This assay is a snapshot of the bioreduction activeness of the cells at the specific time-point during the incubation. The results in Fig. 1A demonstrate that PC3-LN4 cells treated with TBB at 80 μM begin to show a change in viability as early as 2 h following the addition of TBB. Past 24 h and 48 h the corresponding prison cell viability was 40% and 22%, equally indicated graphically in Fig. 1A. Similar observations were made when cells were treated with another CK2 inhibitor TBCA, although a less dramatic loss of viability at 24 and 48 h using 80 μM TBCA was observed (Fig. 1A).

An external file that holds a picture, illustration, etc.  Object name is nihms619091f1.jpg

Cell viability and clonal survival in cells treated with inhibitors of CK2

(A) Prostate cancer PC3-LN4 cells were treated with varying concentrations of TBB or TBCA (two inhibitors of CK2) over different periods of time (varying from ii to 48 h) every bit indicated. Cell viability was determined relative to the controls in which no additions (untreated) were made. Statistical analysis of the data is equally follows: * p < 0.001 to untreated, p < 0.0001 to DMSO; & p < 0.001 to untreated, p < 0.0001 to DMSO; # p = 0.04 to untreated. (B) Clonal survival of cells treated with eighty μM of TBB for varying periods of fourth dimension (4 to 24 h). Representative colony formation results are shown on the right side while their quantitation is presented graphically on the left side of the panel. All other details are equally described under Materials and Methods. Statistical assay of the information is equally follows: * p = 0.004 to DMSO; # p < 0.0001 to DMSO.

In order to assess the survival and proliferative capacity of the cells over a period of vii d following TBB handling, crystal violet clonal survival assays were carried out post-obit iv, vi, 8, and 24 h treatment with TBB. In this analysis, TBB was removed from the cells later the indicated period of treatment, and the cells were replated in complete media. The results demonstrated that 6 h of handling with 80 μM TBB was sufficient to cause meaning loss of cell proliferation and survival (Fig. 1B). Thus, the initiation of loss of cell viability occurring every bit early on every bit 6 h following inhibition of CK2 suggested the presence of preceding events that triggered these changes. Concordant with this information technology may be noted that exposure of prostate cancer cells in culture to fourscore μM TBB for 6 h shows an inhibition of cellular CK2 activity by 20–35% and this inhibition progresses with time [Wang et al., 2008].

Effect of CK2 inhibition on mitochondrial membrane potential

Nosotros have previously demonstrated that modulations in CK2 activeness bear on mitochondrial apoptotic circuitry observed at 24 h following changes in CK2 [Wang et al., 2005a; Wang et al., 2006]. Since Δψm is regarded equally a fundamental determinant of cell viability [Mayer and Oberbauer, 2003], we investigated the effect of CK2 inhibition on mitochondrial role by examining the effect of TBB treatment at varying concentrations over fourth dimension. The results in Fig. 2A prove testify of a rapid loss of Δψm as measured by microscopic detection of JC-1 uptake and fluorescence in cells treated with TBB. Crimson JC-1 fluorescence capture indicates intact Δψ1000, whereas greenish JC-i fluorescence capture indicates that membrane potential has been lost. It is particularly noteworthy that the loss of membrane potential (Δψm) is credible as early on as 2 h post-obit treatment with viii or 80 μM TBB, occurring in a concentration dependent style. These experiments were also performed using TBCA. Like to the data presented in Fig. 1A, the effects of TBCA-mediated CK2 inhibition were slightly less dramatic but substantially the aforementioned as TBB. The quantitation of these data is presented in Tables ane and ii. Nosotros also investigated the effect of TBB on Δψm in two other prostate prison cell lines (LNCaP and BPH-1); representative results of these experiments are shown in Fig. 2, B and C. The response of LNCaP cancer cells was like to that observed for PC3-LN4 cells (Fig. 2B). However, the Δψthou modify in the benign prostate epithelial cell line (BPH-1) was notably lower in response to TBB under similar conditions (Fig. 2C). The quantitation of these data is presented in Tables 3 and 4. These results suggest that cancer cell lines are more than susceptible to inhibition of CK2 action and respond by a dramatic modify in the Δψm and loss of viability. This observation accords with our previous findings on the relative resistance of normal versus beneficial cells to inhibition of CK2 activity [Slaton et al., 2004].

An external file that holds a picture, illustration, etc.  Object name is nihms619091f2.jpg

Changes in mitochondrial membrane potential (Δψm) caused by treatment of cells with the CK2 inhibitor TBB

(A) PC3-LN4 cells were treated with 8 or 80 μM TBB as shown. DMSO command cells were treated with a book of DMSO equivalent to that for 80 μM TBB. The menses of treatment varied from 2 to 24 h. (B) LNCaP cells were treated with TBB at 8 or 80 μM for the periods of time varying from 4 to 24 h. (C) BPH-i cells were treated with eight or 80 μM TBB for the periods of fourth dimension varying from two to 24 h. Mitochondrial membrane potential was measured using JC-1 every bit described under Materials and Methods.

Table 1

Issue of TBB on mitochondrial membrane potential in PC3-LN4 cells over time

Time of handling (h) Amanuensis Mitochondrial membrane potential (Δψm) in PC3-LN4 cellsa
High Δψyard
(red)
Low Δψm
(yellow)
No Δψ1000
(dark-green)
2 None 80.seven ± 6.ix 18.nine ± 2.3 0
DMSO 64.ii ± four.ii 35.iv ± 6.1 0
8 μM TBB 55.1 ± 5.nine 39.viii ± iii.6 11.8 ± 1.2
lxxx μM TBB 16.8 ± ane.3 0.6 ± 0 82.vii ± 2.2
iv None 93.4 ± 3.v v.one ± iii.iv 1.3 ±0
DMSO 93.9 ± iv.8 six.seven ± 2.7 0
8 μM TBB 66.ane ± iv.6 29.one ± 4.0 3.7 ± 1.4
80 μM TBB 0.3 ± 0 0.1 ± 0 99.iv ± 8.2
vi None xc.5 ± 4.5 nine.2 ±4.5 0.iii ± 0
DMSO 85.5 ± 4.6 14.5 ± 4.6 0 ± 0
8 μM TBB 41.0 ± 4.7 41.two ± 4.one 17.viii ± ii.6
eighty μM TBB 0.9 ± 0 0 ± 0 99.i ± i.5
24 None 97.0 ± 0.two 2.ii ± 0 0 ± 0
DMSO 98.ix ± 0.4 0.v ± 0 0 ± 0
8 μM TBB 58.2 ± 3.ane b 13.7 ± ii.5 28.i ± ii.2
80 μM TBB 0.seven ± 0 0 ± 0 99.2 ± 1.9

Tabular array two

Effect of TBCA on mitochondrial membrane potential in PC3-LN4 cells over time

Time of treatment (h) Agent Mitochondrial membrane potential (Δψm) in PC3-LN4 cellsa
Loftier Δψm
(red)
Low Δψm
(yellowish)
No Δψthou
(green)
2 None 86.iii ± 1.iii xiii.8 ± 1.3 0
DMSO 90.5 ± 0.9 vii.ix ± 0.viii one.six ± 1.0
20 μM TBCA 35.3 ± three.4 29.three ± 1.4 34.3 ± i.ix
80 μM TBCA 20.three ± two.4 26.6 ± 1.0 53.1 ± ii.1
iv None 90.viii ± ane.2 4.four ± 0.8 3.0 ± 1.1
DMSO 89.ii ± i.0 10.three ± 0.7 0.7 ± 0.i
20 μM TBCA 40.9 ± v.eight 36.0 ± 3.7 23.0 ± three.6
fourscore μM TBCA fourteen.0 ± 2.1 31.3 ± 2.0 54.vi ± 5.8
6 None 88.8 ± two.half dozen 10.1 ± 2.0 1.0 ± 0
DMSO 91.four ± 1.1 7.v ± ane.2 one.iii ± 0.five
twenty μM TBCA 88.five.0 ± 2.7 9.six ± 2.1 1.9 ± 0.five
80 μM TBCA 13.five ± iii.1 37.1 ± i.6 49.3 ± 2.v
24 None xc.iv ± 1.0 9.vi ± one.0 0 ± 0
DMSO 94.8 ± 0.3 v.iii ± 0.3 0 ± 0
20 μM TBCA 17.vi ± 3.6 33.5 ± ii.5 48.nine ± one.9
lxxx μM TBCA 0.six ± 0.5 0 ± 0 99.iv ± i.seven

Tabular array three

Event of TBB on mitochondrial membrane potential in LNCaP cells over fourth dimension

Time of treatment (h) Agent Mitochondrial membrane potential (Δψthousand) in LNCaP cellsa
High Δψm
(blood-red)
Low Δψm
(yellow)
No Δψyard
(green)
4 None 81.five ± 2.five eighteen.v ± 1.5 0
8 μM TBB 45.seven ± 1.five x.1 ± 3.5 44.5 ± 1.5
80 μM TBB xv.6 ± three.3 0 80.ane ± 5.0
six None xc.8 ± ane.0 9.ane ±1.2 0
8 μM TBB 55.1 ± 1.8 9.iv ± 0.7 35.half-dozen ± ane.ii
80 μM TBB 19.nine ± 1.5 0 ± 0 eighty.9 ± five.0
24 None 87.one ± one.iii 12.nine ± 0.9 0 ± 0
DMSO 93.6 ± 2.four half-dozen.4 ± 1.2 0 ± 0
8 μM TBB 43.9 ± v.9 16.2 ± 1.8 twoscore.0 ± 3.0
80 μM TBB 23.ii ± one.9 2.8 ± 0.nine 74.1 ± 4.vi

Table 4

Effect of TBB on mitochondrial membrane potential in BPH-1 cells over time

Time of treatment (h) Agent Mitochondrial membrane potential (Δψm) in BPH-1 cellsa
High Δψm
(red)
Depression Δψchiliad
(yellow)
No Δψg
(green)
two None 86.three ± 5.6 12.5 ± 3.1 0
DMSO 85.1± 4.five 14.eight ± 4.2 0.12
8 μM TBB 81.three ± 3.6 xiv.9 ± 1.nine 3.viii ±2.i
lxxx μM TBB 71.7 ± 6.iii 13.8 ± two.8 12.6 ± 5.5
iv None 89.8 ± 6.6 iv.four ± i.3 5.9 ±0
DMSO 94.one ± 3.3 3.ix ± 0.9 one.viii ± 1.1
8 μM TBB 54.i ± ane.8 13.7 ± two.0 35.4 ± 3.9
80 μM TBB 13.4 ± 9.half-dozen sixteen.0 ± two.7 seventy.6 ± 4.9
6 None 88.vi ± 1.nine 11.four ± 2.2 0
DMSO 85.2 ± 12.six 14.0 ± 1.5 0
8 μM TBB 63.1 ± 21.v ± 2.8 15.four ± 1.ane
80 μM TBB 30.3 ± 0 20.7 ± 2.vi 49.five ± 3.6
24 None 95.5 ±2.0 3.4 ± 2.2 0
DMSO 91.6 ± 12.6 eight.4 ± 1.5 0
viii μM TBB 66.9 ± one.8 20.5 ± 1.six 11.4 ± 1.0
fourscore μM TBB 52.six ± 7.2 15.8 ± 2.6 31.6 ± 3.vi

We used a 2d technique to verify loss of Δψm following CK2 inhibition with TBB. PC3-LN4 cells were treated with 1 or lxxx μM TBB using equivalent volumes of DMSO as controls, and JC-1 fluorescence was analyzed by FACS after 2 or 24 h of TBB treatment. The results again demonstrate that CK2 activity inhibition by TBB results in a dramatic loss of Δψm afterward 2 h of handling (Tabular array 5). Continued incubation with 80 μM TBB for 24 h resulted in virtually all cells demonstrating a loss of Δψm, which is in agreement with the JC-1 microscopic assay. We as well used siRNA transfection to downregulate CK2αα′ protein expression in PC3-LN4 cells. At 48 h post-transfection, we observed a 21.iii ± i.03% loss in Δψm relative to 4.ii ± 1.42% loss in the command siRNA transfected cells. Loss of well-nigh seventy% of CK2αα′ protein expression was indicated past immunoblot (data not shown). This information is supportive of a CK2-specific function in maintaining Δψm; however, the extended nature of siRNA-mediated downregulation of protein expression over time compared to the kinetically precise inhibition of CK2 action using a small-scale molecule inhibitor is a probable explanation for the less dramatic change in Δψm at a given timepoint post-transfection (please see Give-and-take section likewise).

Table five

FACS analysis of the effect of TBB on mitochondrial membrane potential in PC3-LN4 cells over fourth dimensiona

Agent Time of treatment
(h)
Cells with intact Δψg
(%)
Cells with loss of Δψ1000
(%)
None 97.3 two.seven
DMSOb 2 96.half-dozen 3.4
1μM TBB 2 97.7 ii.3
DMSOb 2 65.4 34.6
fourscore μM TBB ii 21.0 79.0
DMSOb 24 55.3 44.7
80 μM TBB 24 0.3 99.vii

Effect of catalase on CK2 inhibition mediated mitochondrial membrane potential

Information technology was previously observed that inhibition of CK2 activity by TBB or downregulation of CK2 expression levels by antisense CK2α in prostate cancer cells promoted product of ROS such as H2O2 which became apparent between iv to viii h after treatment [Ahmad et al., 2006]. Later on, similar results were observed in the homo leukemia Cem cell line treated with TBB. Compared with prostate cancer prison cell lines employed in the nowadays work, leukemia Cem cells are significantly more sensitive to inhibition of CK2 such that in the presence of fifty μM TBB less than 40% jail cell survival was observed at 24 h post-treatment, and accordingly under these experimental weather the product of ROS was shown to occur at 3 h [Hanif et al., 2009]. In these experiments, treatment of cells with the HiiO2 scavenger catalase for i h prior to handling with TBB resulted in a l% reduction in the product of HiiO2 confirming the nature of the ROS produced on inhibition of CK2 in Cem cells. Nosotros had originally suggested that a possible mechanism of induction of apoptosis upon inhibition or downregulation of CK2 was via the upstream production of HiiOii [Ahmad et al., 2006]. Nonetheless, as described above, we have now observed that an event on the Δψm is credible at an even earlier fourth dimension (2 h in the case of prostate cancer cells employed here). If production of ROS preceded changes in Δψm in these cells, and then treatment of cells with a ROS scavenger should block the changes in Δψone thousand. Thus, we investigated if Δψm was effected when PC3-LN4 cells were treated with 3000 and 6000 units of catalase for 1 h prior to inhibition of CK2 with 80 μM TBB for 4 h. The results indicated that there was no effect of catalase handling on Δψgrand under these conditions, suggesting that the Δψm change in prostate cancer cells occurred prior to the production of H2O2 (Fig. three, left panels).

Effect of BAPTA on CK2 inhibition mediated mitochondrial membrane potential

Since it is well known that Δψ1000 and mitochondria permeability are both regulated by cellular Ca2+ (for a review encounter, east.g., [Perry et al., 2011]), we investigated the effect of treatment of cells with the Catwo+ chelator BAPTA (10 μM) for 1 h prior to treatment of cells with eighty μM TBB for iv h. As shown in Fig. 3, center panels, pre-handling of cells with BAPTA prevented Δψm loss suggesting a role of Catwo+ in promoting these changes in response to CK2 inhibition. These results predict the interest of Caii+ signaling equally an early on response to inhibition of CK2 activity in promoting loss of Δψm.

Release of intracellular Ca2+ stores upon CK2 inhibition

Because we observed that BAPTA handling protected cells from Δψm loss subsequently inhibition of CK2, we tested whether there is a detectable release of intracellular Ca2+ upon addition of TBB to PC3-LN4 cells. Intracellular Caii+ was measured at 2 h, 1 h, 30 min, fifteen min and immediately after addition of TBB. A meaning alter in detectable Ca2+ was observed upon treatment with 80 μM TBB immediately afterwards its addition (Fig. 3B). Thapsigargin is known to cause a rapid increase in intracellular costless Ca2+ concentration by release of intracellular Caii+ stores and was used as positive command. A bottom amount of Caii+ release was detected following 8 μM TBB treatment (Fig. 3B). No difference between untreated and TBB treated cells was detected at the other fourth dimension points, suggesting the transient nature of the Ca2+ signal response to CK2 inhibition (data non shown).

Sit-in of presence of CK2 in mitochondria

CK2 is distributed in various compartments within the nucleus and cytoplasm of the cell. Previous reports had indicated CK2-like activity in the mitochondria of mammalian organs, including rat liver and bovine kidney [Damuni and Reed, 1988; Sarrouilhe and Baudry, 1996]. Here nosotros provide evidence on the presence of CK2 in human prostate mitochondria using diverse jail cell lines. We prepared highly purified mitochondria from several prostate cell lines including LNCaP, C4-2, BPH-1, and PC3-LN4. All of them showed the presence of CK2 past immunoblot assay. Fig. 4 shows the representative results on iii different prostate cell lines (LNCaP, C4-2, and BPH-1) which indicate the distribution of the various CK2 subunits in dissimilar cell fractions, including the presence of all three subunits of CK2 in the mitochondrial fraction. The purity of these mitochondrial preparations was established by incubating the blots with antibodies that notice specific markers for mitochondrial (Cox IV) and non-mitochondrial fractions (lamin A/C and β-tubulin). Note the presence of Cox IV in mitochondria and its absence in the cytosol, the absence of lamin A/C in both the mitochondrial and cytosolic fractions, and the absence of β-tubulin in mitochondria. As shown subsequently, we also studied if the mitochondrial localized CK2 might be involved in the rapid response of cells to inhibition of CK2.

An external file that holds a picture, illustration, etc.  Object name is nihms619091f4.jpg

Presence of CK2 in mitochondria

The results demonstrate the presence of CK2 in the purified mitochondrial fractions from prostate cells C4-two, LNCaP, and BPH-1. Markers for nuclear, cytosolic, or mitochondrial fractions were analyzed in whole cell lysate (W), cytosolic fraction (C), and mitochondria (M). Markers include Cox Four for mitochondria; β-tubulin for cytosol; and lamin A/C for nuclei.

Effect of CK2 inhibitors TBB and TBCA on isolated mitochondria

The results on the effect of CK2 inhibition on Δψgrand changes that were blocked by prior treatment of cells with the Ca2+ chelator BAPTA suggested that rapid mitochondrial permeability changes may exist involved in the Δψthou changes. Mitochondrial permeability transition (MPT) tin can occur associated with dissipation of the difference in voltage across the inner mitochondrial membrane, i.eastward., Δψthousand. Further, it is more often than not recognized that a complex interaction between Ca2+ shuttling, mitochondrial membrane permeability transition, and mitochondrial membrane potential exists in relation to consecration of cell death, although this relation can vary with the cell type [Isenberg and Klaunig, 2000; Lemasters et al., 1998; Lemasters et al., 2009; Stavrovskaya and Kristal, 2005]. In view of these various considerations, our observation on the presence of CK2 in the mitochondrial fraction raised the question of the possible involvement of intrinsic CK2 in influencing mitochondrial function pertinent to membrane permeability transition.

To address this question, nosotros employed rat liver mitochondria using procedures that yield highly pure preparations and evaluated the effects of endogenous CK2 inhibition on MPT every bit measured by mitochondrial swelling [Schnaitman and Greenawalt, 1968]. Cyclosporin A was added every bit a control to block the permeability transition, whereas Ca2+ and Pi combined were added as positive control inducers to cause permeability. The results in Fig. 5A prove the effect of two CK2 inhibitors, TBB and TBCA, tested at eighty μM concentrations. When added to purified mitochondria these inhibitors produced a rapid (within v min) significant change in permeability transition resulting in mitochondrial swelling. This was likewise accompanied by cytochrome c release (Fig. 5B, right panel). Cyclosporin A partially inhibited the mitochondrial swelling in response to CK2 inhibition with either TBB or TBCA, indicating interest of the mitochondrial permeability transition pore (PTP), whereas information technology did non significantly inhibit the cytochrome c release (Fig. 5B, right panel). The swelling of the mitochondria subsequently CK2 inhibition was rescued more than finer by the addition of excess EGTA (chelator of Ca2+), implying that Catwo+ plays a pivotal role in the CK2-dependent process. Farther, in the presence of EGTA the release of cytochrome c was besides significantly mitigated (Fig. 5B, right console). Of item note is the observation that comparison of the effect of TBB or TBCA with that of EGTA+TBB or TBCA demonstrates a articulate reduction in the consequence of TBB or TBCA when EGTA is included. However, an additive effect was observed when EGTA and cyclosporin A were combined, resulting in almost complete inhibition of mitochondrial swelling and prevention of cytochrome c release. The presence of endogenous CK2 in the mitochondria used for these assays was verified by immunoblot assay (Fig. 5B, left panel). These observations agree with the information presented in Fig. two and Fig. 3, reinforcing that early changes in mitochondrial function relate to involvement of the mitochondrial transition pore and Catwo+ mediated mitochondrial membrane permeability coupled with loss of Δψm. Taken together, it appears that these early events are disquisitional for initiation of the apoptotic machinery in response to inhibition of CK2. Farther, our results hint that mitochondrial associated CK2 may exist the primary site of initiation of these events.

Temporal relation of CK2 inhibition and mitochondrial membrane potential changes compared with other markers of apoptosis

The presence of the CK2 inhibitor TBB promoted loss of prostate cancer cell viability that becomes apparent around 4–6 h equally shown in Fig. 1. To further identify the temporal response of various mitochondrial apoptotic signals to inhibition of CK2, nosotros examined several markers of apoptosis over time following handling of cells with TBB. The results in Fig. 6 prove the issue of treating PC3-LN4 cells with 80 μM TBB for varying periods of time. In cytosolic lysates in which the mitochondria have been removed past centrifugation, loss of full length Bid compared to DMSO controls was apparent from 4 h through 24 h. In the same cytosolic lysates, release of cytochrome c and Cox IV from the mitochondria into the cytosol was evident at 24 h (Fig. 6A). Assay of other signals associated with apoptosis such as lamin A/C, caspase-nine, and caspase-3 cleavage was performed in whole cell lysates and indicated their appearance at 24 h (Fig. 6B). The effectiveness of TBB as inhibitor of CK2 was also analyzed by examining the phosphorylation condition of AKT-1. The inhibition of phosphorylation at Ser129 which is a specific target of CK2 was indicated at 24 h and further increased at 48 h post-obit treatment with TBB (Fig. 6B). Concurrent loss of the full general AKT-1 betoken was also observed and was specific to TBB and non etoposide treatment. These results farther suggest that the activation of these various cellular effects and apoptotic signals are downstream of the to a higher place described changes in Δψm most likely triggered by contradistinct mitochondrial Ca2+ flux in response to inhibition of CK2 activity, as discussed after.

An external file that holds a picture, illustration, etc.  Object name is nihms619091f6.jpg

Temporal response of CK2 substrate and apoptotic signals following TBB mediated inhibition of CK2 in PC3-LN4 cells

(A) Effect of 80 μM TBB handling of cells for varying times on apoptotic signals measured in the cytosol. Cells were treated with TBB for 4, 6, 8 and 24 h. Analysis of Bid, cytochrome c, and Cox IV was carried out in the purified mitochondria-free cytosolic fractions. (B) Consequence of 80 μM TBB handling of cells for varying times in whole prison cell lysates. Cells were treated with TBB for 6, 24, and 48 h as shown, with DMSO as the corresponding control. Full length and cleaved lamin A/C, full length and cleaved caspase 9, cleaved caspase three, full and phospho-Ser129 AKT-i were analyzed. Etoposide (100 μM for 24 h) was employed equally the positive command for induction of apoptosis. All other details were as described under Materials and Methods.

Discussion

Much work has established CK2 to be involved in the regulation of a wide range of cellular activities both in normal cells and in many disease processes [Guerra and Issinger, 2008]. In particular, CK2 has emerged equally a particularly important indicate in cancer biology [Ahmed et al., 2000; Ahmed et al., 2002; Trembley et al., 2010; Trembley et al., 2009]. Over fourth dimension, it has been observed that CK2 is markedly elevated at the protein level in all cancers that have been examined [Tawfic et al., 2001]. Several hallmarks of cancer accept been described [Hanahan and Weinberg, 2011], and there is considerable evidence of functional association of CK2 in several of these hallmarks [Trembley et al., 2010; Trembley et al., 2009; Trembley et al., 2013]. Amid the hallmarks of cancer two features are consistently noted, namely deregulated growth and proliferation and altered resistance to cell death. The involvement of CK2 in both normal and abnormal jail cell growth was recognized for a long time; still, its relation to the cancer cell phenotype became credible with recognition that CK2 not only promotes cell growth and proliferation but as well suppresses apoptosis [Ahmad et al., 2008; Ahmed et al., 2002; Guo et al., 2001]. A general concept that emerged from our studies was that nuclear CK2 dynamics were intimately related to cell growth and cell expiry so that removal of growth stimuli resulted in the loss of CK2 from the nuclear structures (chromatin and nuclear matrix) which preceded the consecration of apoptosis [Ahmed et al., 1993; Guo et al., 1999; Yu et al., 2001]. Further, our demonstration that prior forced overexpression of CK2 in cells protected them from undergoing apoptosis mediated by etoposide and diethylstilbestrol provided direct evidence of the ability of CK2 to suppress apoptosis [Ahmed et al., 2002; Guo et al., 2001]. The now accepted role of CK2 as a suppressor of apoptosis is underscored past its impact on apoptosis mediated by diverse stimuli such as loss of survival factors (hormones, growth factors), chemical agents (drugs such as etoposide, diethylstilbestrol, inhibitors of CK2), death receptor mediated signals (such as TNFα, TRAIL, FasL), physical agents (such as heat, radiation), and molecular downregulation of CK2 [Ahmad et al., 2008; Ahmed et al., 2002; Davis et al., 2002; Guo et al., 1999; Guo et al., 2001; Trembley et al., 2012; Trembley et al., 2011; Trembley et al., 2013; Wang et al., 2005a; Wang et al., 2006; Wang et al., 2008; Wang et al., 2001; Yamane and Kinsella, 2005]. These various observations prompted the states to advise that CK2 could serve every bit a cancer therapy target [Ahmad et al., 2005; Slaton et al., 2004; Unger et al., 2004; Wang et al., 2005b; Wang et al., 2001]; the potential of CK2 as a target continues to proceeds wide acceptance with mounting evidence of its druggability for cancer therapy [Pinna and Allende, 2009; Sarno and Pinna, 2008].

Several factors and pathways that may play a role in the apoptotic procedure triggered by the inhibition of CK2 have been examined, including IAPs, Bcl family proteins, ROS, and caspases [Ahmad et al., 2006; Duncan et al., 2011; McDonnell et al., 2008; Ponce et al., 2011; Tapia et al., 2006; Turowec et al., 2011; Wang et al., 2006; Wang et al., 2008; Wang et al., 2001]. These activities point to the temporal progression of apoptotic signaling that occurs in response to inhibition or downregulation of CK2. All the same, the nature of the chief trigger(due south) for the initiation of apoptotic signaling has been unclear. To address this issue, we have presented novel observations that suggest a potential mechanism for the initiation and progression of apoptosis after CK2 inhibition. We have shown that when cells are treated with the relatively specific CK2 inhibitors TBB or TBCA the initiation of cell survival loss occurs between 4 to six h post-obit inhibitor treatment, implying the existence of events preceding this time frame. Appropriately, we have found that under the present experimental conditions, loss of Δψthou is apparent equally early every bit 2 h following treatment with CK2 inhibitors. Further, our results hint that this alteration in mitochondrial physiology involves changes in the permeability transition pore besides equally Caii+ dynamics since prior treatment with the specific Catwo+ chelator BAPTA blocked the change in membrane permeability and Δψm.

Information technology is well known that CK2 is localized in the nuclear and cytoplasmic fractions and that its level in dissimilar loci is of a dynamic nature depending on the cell country. We accept documented here that highly purified mitochondria from prostate cells as well harbor some CK2, and our results hint that the mitochondrial CK2 may accept a role in the regulation of mitochondrial permeability and hence the membrane potential. In addition, a previous documentation of the localization of CK2 activity in the isolated rat liver mitochondria inner membrane and intermembrane space is consequent with inhibition of its action being able to directly affect the PTP also every bit the mitochondrial apoptosis channel (MAC). Previously, information technology was as well suggested that production of HiiOtwo may be the proximal trigger for initiation of apoptosis on downregulation or inhibition of CK2 in prostate cancer cells; even so, this event occurred at around 4 to 6 h following CK2 inhibition [Ahmad et al., 2006]. Farther, nosotros have observed that prior treatment of cells with the ROS scavenger catalase did non influence the rapid changes in Δψk caused by CK2 inhibition. This argues against H2O2 being the initiator of apoptotic signaling in response to CK2 inhibition in prostate cancer cells. Additional analysis of the temporal response for several of the apoptotic signals on inhibition of CK2 suggested changes in Bid to occur at iv to 6 h while other signals of apoptosis such as the cytosolic appearance of cytochrome c, activation of caspases, and production of lamin A/C cleavage products to exist amongst the afterwards events. Finally, the JC-one microscopy and FACS data together with the results using isolated mitochondria also imply a direct office for CK2 activeness in maintaining the mitochondrial membrane potential. The effect of TBB and TBCA on isolated mitochondria suggests that these drugs are able to cross the mitochondrial membranes.

In previous piece of work, nosotros have shown that manipulation of CK2 expression levels in cells influences the mitochondrial apoptotic circuitry [Wang et al., 2006]. However, in the present piece of work we observed that mitochondrial membrane potential loss was less dramatic following siRNA treatment for 48 h to downregulate CK2 poly peptide levels than following TBB mediated inhibition of CK2 activity at early time points. We suggest that this may be due to the almost immediate effect that TBB or TBCA may take on mitochondrial located CK2, whereas loss of CK2 protein due to cytosolic siRNA downregulation of expression would likely take a much longer fourth dimension to take impact on the mitochondrial CK2 population. Essentially, use of siRNA to CK2 produces an effect on CK2 level that does not become pregnant until about 48 h and even then a notable amount of CK2 poly peptide would be expected to be available since the half-life of CK2 protein is relatively long. Thus, the consecration of apoptosis past molecular downregulation of CK2 becomes apparent at later time points and in an asynchronous manner making it difficult to undertake a precise assay of the initiating events. It is also plausible that the method of modulating CK2 (i.e., rapid action inhibition versus tedious loss of protein expression) may co-opt different jail cell expiry pathways. These aspects of CK2 regulated cell death are also focus of our future work. To reiterate, we emphasize that the goal of the present work was to place the earliest preceding events that lead to the orchestration of the mitochondrial apoptotic pathway and induction of prison cell death on reduction of CK2 activity.

Based on the various aforementioned observations, we nowadays the following scheme of progression of events subsequent to downregulation of CK2 activity in the cell (Fig. seven). We propose that i of the earliest events in response to CK2 inhibition in the cells is rapid intracellular release also equally contradistinct Caii+ flux into or within mitochondria which promotes a change in mitochondrial permeability and loss of mitochondrial membrane potential. It is conceivable that in response to CK2 inhibition, at that place is also ER stress and consequent release of Ca2+ from the ER, equally is suggested past our detection of transiently increased intracellular Ca2+ after CK2 inhibition. While these studies on the regulation of various cellular Ca2+ pumps are currently existence pursued in our laboratory, it is noteworthy that our data using isolated rat liver mitochondria employing buffers containing Caii+ chelating agents suggest that only a pocket-size Ca2+ flux alter may be needed in response to CK2 inhibition to cause cell decease signaling. Thus, the in a higher place-described early changes in Catwo+, mitochondrial membrane permeability, and Δψm upon inhibition of CK2 are likely to stand for the "point of no return" for cell fate decision so that loss of survival begins at four to half dozen h. Associated with this time bespeak are changes in Bid translocation as evidenced by reduction of cytosolic total length Bid (every bit early on every bit 4 to 6 h) and production of H2O2 (around iv to 6 h) followed past the previously described changes in the Bcl family unit proteins and activation of caspases which stand for terminal phase events leading to cell death. The machinery of change(south) in the Caii+ dynamics in mitochondria in response to loss of CK2 signaling, the relative role of CK2 inhibition in the cell cytoplasm compared with that in the mitochondria, and the nature of conditions that make up one's mind the level of CK2 in mitochondria are currently under investigation in our laboratory.

An external file that holds a picture, illustration, etc.  Object name is nihms619091f7.jpg

A cartoon depicting the proposed sequence of events in cells treated with CK2 inhibitors that pb to induction of apoptosis

It is proposed that among the earliest changes in response to treatment of cells with CK2 inhibitors is the effect on mitochondrial membrane permeability transition associated with Ca2+ flux (before ii h) and loss of mitochondrial membrane potential which occur as early as ii h post-obit the handling of cells with TBB (earliest practical fourth dimension measurements in this experimental model). The subsequent serial of events that become apparent between half dozen to 24 h following inhibition of CK2 appear to exist production of ROS, loss of jail cell viability, and loss of proliferative capacity (by four–6 h) followed by other apoptotic signals such as activation of caspases, cleavage of lamin A/C, loss of phospho-Ser129 and total AKT-i, release of cytochrome c and Cox IV from mitochondria, and loss of IAPs expression observed by 24 h [Tapia et al., 2006; Wang et al., 2008].

In summary, while it has been well documented that inhibition or downregulation of CK2 promotes apoptosis in cells through various mechanisms depending on the nature of the cell decease signal, the underlying triggering mechanism that initiates the mitochondrial apoptotic signaling pathway was not well understood. In this written report we take documented, for the starting time time, that downregulation of CK2 activity evokes an early alter in mitochondrial Catwo+ dynamics associated with altered mitochondrial permeability and loss of Δψyard every bit a novel mechanism of how CK2 may be involved in regulation of cell apoptosis.

Acknowledgments

Nosotros give thanks Omar Cespedes-Gomez and Alexandra Rex for their aid in the laboratory.

Contract grant sponsor: U.S. Department of Veterans Diplomacy Merit Review Program; Contract grant number: I01BX001731.

Contract grant sponsor: National Cancer Plant; Contract grant numbers: R01CA150182 (KA), R21CA158730 (BTK).

Abbreviations

CK2 official acronym for the inappropriate former name casein kinase 2 or II
TBB 4,5,half-dozen,vii-tetrabromobenzotriazole
TBCA tetrabromocinnamic acrid
BAPTA 1,ii-bis(o-aminophenoxy)ethane- N,N,N′,N′-tetraacetic acrid
BSA bovine serum albumin
P-S penicillin-streptomycin
PBS phosphate buffered saline
CCCP Carbonyl cyanide m-chlorophenyl hydrazone
FBS fetal bovine serum
TRAIL tumor necrosis factor–related apoptosis inducing ligand
ROS reactive oxygen species
TNFα Tumor necrosis factor α
FasL Fas ligand
IAPs inhibitor of apoptosis proteins
FACS fluorescent activated cell sorting
RIPA radioimmunoprecipitation assay buffer
TBS Tris buffered saline, pH 7.4
MPT mitochondrial permeability transition
PTP permeability transition pore
MAC mitochondrial apoptosis-channel

Footnotes

Conflict of Involvement: No conflicts to declare.

Publisher's Disclaimer: Disclaimer: The views expressed in this article are those of the authors and do not necessarily reflect the position or policy of the U.Southward. Section of Veterans Affairs or the U.S. regime.

References

  • Ahmad KA, Wang Thousand, Ahmed Yard. Intracellular hydrogen peroxide production is an upstream result in apoptosis induced by down-regulation of casein kinase ii in prostate cancer cells. Mol Cancer Res: MCR. 2006;four:331–8. [PubMed] [Google Scholar]
  • Ahmad KA, Wang M, Slaton J, Unger G, Ahmed K. Targeting CK2 for cancer therapy. Anticancer Drugs. 2005;16:1037–43. [PubMed] [Google Scholar]
  • Ahmad KA, Wang Thousand, Unger Chiliad, Slaton J, Ahmed K. Protein kinase CK2–a key suppressor of apoptosis. Adv Enzyme Regul. 2008;48:179–87. [PMC free article] [PubMed] [Google Scholar]
  • Ahmed K. Nuclear matrix and protein kinase CK2 signaling. Crit Rev Eukaryot Cistron Expr. 1999;9:329–36. [PubMed] [Google Scholar]
  • Ahmed Yard, Davis AT, Wang H, Faust RA, Yu S, Tawfic S. Significance of protein kinase CK2 nuclear signaling in neoplasia. J Jail cell Biochem Suppl Suppl. 2000;35:130–5. [PubMed] [Google Scholar]
  • Ahmed K, Gerber DA, Cochet C. Joining the jail cell survival squad: an emerging role for protein kinase CK2. Trends in Jail cell Biology. 2002;12:226–30. [PubMed] [Google Scholar]
  • Ahmed Yard, Yenice Southward, Davis A, Goueli SA. Association of casein kinase 2 with nuclear chromatin in relation to androgenic regulation of rat prostate. Proc Natl Acad Sci U Southward A. 1993;xc:4426–thirty. [PMC gratuitous article] [PubMed] [Google Scholar]
  • Berridge MV, Tan AS. Characterization of the cellular reduction of 3-(4,5-dimethylthiazol-2-yl)-2,five-diphenyltetrazolium bromide (MTT): subcellular localization, substrate dependence, and interest of mitochondrial electron send in MTT reduction. Arch Biochem Biophys. 1993;303:474–82. [PubMed] [Google Scholar]
  • Damuni Z, Reed LJ. Purification and properties of a protamine kinase and a blazon II casein kinase from bovine kidney mitochondria. Arch Biochem Biophys. 1988;262:574–84. [PubMed] [Google Scholar]
  • Davis AT, Wang H, Zhang P, Ahmed 1000. Estrus shock mediated modulation of poly peptide kinase CK2 in the nuclear matrix. J Cell Biochem. 2002;85:583–91. [PubMed] [Google Scholar]
  • Duncan JS, Turowec JP, Duncan KE, Vilk K, Wu C, Luscher B, Li SS, Gloor GB, Litchfield DW. A peptide-based target screen implicates the protein kinase CK2 in the global regulation of caspase signaling. Science Signaling. 2011;4:ra30. [PubMed] [Google Scholar]
  • Guerra B, Issinger OG. Protein kinase CK2 and its part in cellular proliferation, development and pathology. Electrophoresis. 1999;20:391–408. [PubMed] [Google Scholar]
  • Guerra B, Issinger OG. Poly peptide kinase CK2 in man diseases. Curr Med Chem. 2008;xv:1870–86. [PubMed] [Google Scholar]
  • Guo C, Yu S, Davis AT, Ahmed K. Nuclear matrix targeting of the protein kinase CK2 signal every bit a common downstream response to androgen or growth cistron stimulation of prostate cancer cells. Cancer Res. 1999;59:1146–51. [PubMed] [Google Scholar]
  • Guo C, Yu Southward, Davis AT, Wang H, Dark-green JE, Ahmed Thousand. A potential function of nuclear matrix-associated poly peptide kinase CK2 in protection against drug-induced apoptosis in cancer cells. J Biol Chem. 2001;276:5992–9. [PubMed] [Google Scholar]
  • Hanahan D, Weinberg RA. Hallmarks of cancer: the side by side generation. Prison cell. 2011;144:646–74. [PubMed] [Google Scholar]
  • Hanif IM, Ahmad KA, Ahmed M, Pervaiz S. Involvement of reactive oxygen species in apoptosis induced by pharmacological inhibition of protein kinase CK2. Register New York Acad Sci. 2009;1171:591–9. [PMC free article] [PubMed] [Google Scholar]
  • Isenberg JS, Klaunig JE. Role of the mitochondrial membrane permeability transition (MPT) in rotenone-induced apoptosis in liver cells. Toxicol Sci : an official journal of the Society of Toxicology. 2000;53:340–51. [PubMed] [Google Scholar]
  • Lemasters JJ, Nieminen AL, Qian T, Trost LC, Elmore SP, Nishimura Y, Crowe RA, Cascio WE, Bradham CA, Brenner DA, Herman B. The mitochondrial permeability transition in cell expiry: a common mechanism in necrosis, apoptosis and autophagy. Biochim Biophys Acta. 1998;1366:177–96. [PubMed] [Google Scholar]
  • Lemasters JJ, Theruvath TP, Zhong Z, Nieminen AL. Mitochondrial calcium and the permeability transition in jail cell decease. Biochim Biophys Acta. 2009;1787:1395–401. [PMC free article] [PubMed] [Google Scholar]
  • Mayer B, Oberbauer R. Mitochondrial regulation of apoptosis. News in physiological sciences : an international journal of physiology produced jointly past the International Union of Physiological Sciences and the American Physiological Social club. 2003;18:89–94. [PubMed] [Google Scholar]
  • McDonnell MA, Abedin MJ, Melendez M, Platikanova TN, Ecklund JR, Ahmed K, Kelekar A. Phosphorylation of murine caspase-9 by the protein kinase casein kinase 2 regulates its cleavage by caspase-8. J Biol Chem. 2008;283:20149–58. [PMC free article] [PubMed] [Google Scholar]
  • Meggio F, Pinna LA. Ane-1000-and-1 substrates of protein kinase CK2? FASEB J. 2003;17:349–368. [PubMed] [Google Scholar]
  • Perry SW, Norman JP, Barbieri J, Brown EB, Gelbard HA. Mitochondrial membrane potential probes and the proton gradient: a applied usage guide. BioTechniques. 2011;50:98–115. [PMC free article] [PubMed] [Google Scholar]
  • Pinna LA. Protein kinase CK2: a challenge to canons. J Cell Sci. 2002;115:3873–8. [PubMed] [Google Scholar]
  • Pinna LA, Allende JE. Protein kinase CK2 in health and disease: Protein kinase CK2: an ugly duckling in the kinome swimming. Cell Mol Life Sci. 2009;66:1795–9. [PubMed] [Google Scholar]
  • Ponce DP, Yefi R, Cabello P, Maturana JL, Niechi I, Silva E, Galindo Yard, Antonelli M, Marcelain Yard, Armisen R, Tapia JC. CK2 functionally interacts with AKT/PKB to promote the beta-catenin-dependent expression of survivin and enhance cell survival. Mol Cell Biochem. 2011;356:127–32. [PubMed] [Google Scholar]
  • Sarno Southward, Pinna LA. Poly peptide kinase CK2 as a druggable target. Mol Biosyst. 2008;four:889–94. [PubMed] [Google Scholar]
  • Sarrouilhe D, Baudry M. Evidence of true protein kinase CKII activity in mitochondria and its spermine-mediated translocation to inner membrane. Jail cell Mol Biol. 1996;42:189–97. [PubMed] [Google Scholar]
  • Savage MK, Jones DP, Reed DJ. Calcium- and phosphate-dependent release and loading of glutathione past liver mitochondria. Arch Biochem Biophys. 1991;290:51–half dozen. [PubMed] [Google Scholar]
  • Schnaitman C, Greenawalt JW. Enzymatic properties of the inner and outer membranes of rat liver mitochondria. J Cell Biol. 1968;38:158–75. [PMC free commodity] [PubMed] [Google Scholar]
  • Slaton JW, Unger GM, Sloper DT, Davis AT, Ahmed 1000. Induction of apoptosis by antisense CK2 in homo prostate cancer xenograft model. Mol Cancer Res. 2004;2:712–21. [PubMed] [Google Scholar]
  • St-Denis NA, Litchfield DW. Protein kinase CK2 in health and illness: From birth to expiry: the role of protein kinase CK2 in the regulation of prison cell proliferation and survival. Cell Mol Life Sci. 2009;66:1817–29. [PubMed] [Google Scholar]
  • Stavrovskaya IG, Kristal BS. The powerhouse takes control of the prison cell: is the mitochondrial permeability transition a viable therapeutic target against neuronal dysfunction and death? Free Radical Biol & Med. 2005;38:687–97. [PubMed] [Google Scholar]
  • Tapia JC, Torres VA, Rodriguez DA, Leyton L, Quest AF. Casein kinase 2 (CK2) increases survivin expression via enhanced beta-catenin-T cell factor/lymphoid enhancer binding factor-dependent transcription. Proc Natl Acad Sci U S A. 2006;103:15079–84. [PMC gratis commodity] [PubMed] [Google Scholar]
  • Tawfic South, Yu Southward, Wang H, Faust R, Davis A, Ahmed 1000. Protein kinase CK2 betoken in neoplasia. Histol Histopathol. 2001;16:573–82. [PubMed] [Google Scholar]
  • Trembley JH, Chen Z, Unger G, Slaton J, Kren BT, Van Waes C, Ahmed Yard. Emergence of protein kinase CK2 as a fundamental target in cancer therapy. BioFactors. 2010;36:187–95. [PMC costless commodity] [PubMed] [Google Scholar]
  • Trembley JH, Unger GM, Korman VL, Tobolt DK, Kazimierczuk Z, Pinna LA, Kren BT, Ahmed One thousand. Nanoencapsulated anti-CK2 small molecule drug or siRNA specifically targets malignant cancer but not benign cells. Cancer Lett. 2012;315:48–58. [PMC free article] [PubMed] [Google Scholar]
  • Trembley JH, Unger GM, Tobolt DK, Korman VL, Wang Thou, Ahmad KA, Slaton JW, Kren BT, Ahmed Thousand. Systemic administration of antisense oligonucleotides simultaneously targeting CK2alpha and alpha′ subunits reduces orthotopic xenograft prostate tumors in mice. Mol Cell Biochem. 2011;356:21–35. [PMC complimentary commodity] [PubMed] [Google Scholar]
  • Trembley JH, Wang Thousand, Unger M, Slaton J, Ahmed K. Protein kinase CK2 in health and illness: CK2: a cardinal thespian in cancer biology. Cell Mol Life Sci. 2009;66:1858–67. [PMC free commodity] [PubMed] [Google Scholar]
  • Trembley JH, Wu J, Unger GM, Kren BT, Ahmed G. CK2 suppression of apoptosis and its implications in cancer biology and therapy. In: Pinna LA, editor. The Wiley-IUBMB Series on Biochemistry and Molecular Biology: Protein Kinase CK2. 2013. pp. 319–333. [Google Scholar]
  • Turowec JP, Duncan JS, Gloor GB, Litchfield DW. Regulation of caspase pathways past protein kinase CK2: identification of proteins with overlapping CK2 and caspase consensus motifs. Mol Cell Biochem. 2011;356:159–67. [PubMed] [Google Scholar]
  • Unger GM, Davis AT, Slaton JW, Ahmed K. Protein kinase CK2 every bit regulator of cell survival: implications for cancer therapy. Curr Cancer Drug Targets. 2004;four:77–84. [PubMed] [Google Scholar]
  • Wang Yard, Ahmad KA, Ahmed K. Modulation of death receptor-mediated apoptosis by CK2. Mol Prison cell Biochem. 2005a;274:201–v. [PubMed] [Google Scholar]
  • Wang G, Ahmad KA, Ahmed K. Function of protein kinase CK2 in the regulation of tumor necrosis gene-related apoptosis inducing ligand-induced apoptosis in prostate cancer cells. Cancer Res. 2006;66:2242–9. [PubMed] [Google Scholar]
  • Wang K, Ahmad KA, Harris NH, Ahmed K. Touch on of protein kinase CK2 on inhibitor of apoptosis proteins in prostate cancer cells. Mol Prison cell Biochem. 2008;316:91–7. [PubMed] [Google Scholar]
  • Wang Thousand, Unger G, Ahmad KA, Slaton JW, Ahmed K. Downregulation of CK2 induces apoptosis in cancer cells–a potential arroyo to cancer therapy. Mol Cell Biochem. 2005b;274:77–84. [PubMed] [Google Scholar]
  • Wang H, Davis A, Yu Southward, Ahmed K. Response of cancer cells to molecular interruption of the CK2 indicate. Mol Cell Biochem. 2001;227:167–74. [PubMed] [Google Scholar]
  • Yamane Thou, Kinsella TJ. CK2 inhibits apoptosis and changes its cellular localization following ionizing radiations. Cancer Res. 2005;65:4362–7. [PubMed] [Google Scholar]
  • Yu S, Wang H, Davis A, Ahmed K. Consequences of CK2 signaling to the nuclear matrix. Mol Cell Biochem. 2001;227:67–71. [PubMed] [Google Scholar]

wadsworthladest78.blogspot.com

Source: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4199905/

0 Response to "Ck2 How Long Till Suicide Again"

Post a Comment

Iklan Atas Artikel

Iklan Tengah Artikel 1

Iklan Tengah Artikel 2

Iklan Bawah Artikel