10
(CANCER RESEARCH 50. 677-685. Februar} l, 1990) Inhibition of Rodent Protein Kinase C by the Anticarcinoma Agent Dequalinium1 Susan A. Rotenberg,2 Stephen Smiley, Marius Ueffing, Robert S. Krauss, Lan Bo Chen, and I. Bernard Weinstein Institute of Cancer Research, Columbia University, New York, New York 10032 fS. A. R., M. £/., R. S. A'., /. B. W.], and Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115 [S. S., L. B. C.¡ ABSTRACT Dequalinium has previously been shown to be an anticarcinoma agent (M. J. Weiss et al., Proc. Nati. Acad. Sci. USA, 84: 5444-5448, 1987). The present study demonstrates that it can inhibit protein kinase C-ß, isolated from an overproducing cell line with a 50% inhibitory concentra tion of8-15 MM-Further examination of the inhibition by using structural analogues of dequalinium reveals that the length of the méthylène bridge between the two quinaldinium moieties, the presence of the ring substit- uents, and the bipartite character of the compound each contributes to the inhibitory potency. Related studies show that the analogues display the same rank order of inhibitory potency when tested with the trypsin- generated catalytic fragment of the enzyme, indicating that dequalinium inhibits kinase activity through an interaction with the catalytic subunit. Further studies argue that the ability of a given analogue to inhibit phosphotransferase activity correlates with its ability to compete with |JH|phorbol-12,13-dibutyrate binding on the intact enzyme (50% inhibi tory concentration of 2-5 MM)-This suggests that the inhibitor is either binding directly to the regulatory subunit as well, or that due to its interaction with the catalytic subunit, dequalinium produces an indirect effect on sites defined by phorbol ester binding. Kinetic analysis revealed that inhibition is noncompetitive with respect to ATP or phosphatidyl- serine. Studies conducted with types I, II, and III rat brain isozymes, resolved by hydroxylapatite chromatography, demonstrate that dequa linium inhibits each of them with similar potency (50% inhibitory con centration of 11 MM)and imply that the site of contact on the enzyme is a highly conserved region. Morphology studies with dequalinium in intact cells demonstrate that the inhibitor can protect control cells against phorbol ester-induced morphology changes but cannot protect protein kinase C-overproducing cells, suggesting that an elevation in protein kinase C levels alone is sufficient to overturn the protection conferred by dequalinium. On the basis of these results, we propose that protein kinase C could be a critical in vivo target of dequalinium. INTRODUCTION Over the last five years the fields of biochemistry and molec ular biology have been harnessed to study the structure and function of PKC,1 a calcium- and phospholipid-dependent pro tein kinase that participates in many cell regulatory processes. This enzyme serves as both the receptor for phorbol ester tumor promoters (1) and as a key enzyme in growth factor-mediated signal transduction (2). It is activated by diacylglycerol, a prod- Received5/19/89;revised9/27/89;accepted10/17/89. The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. 1This work was supported by the following grants: NIH CA 02656 (S. A. R., I. B. W.): National Research Service Award CA 08346 (R. S. K.); CA 19589-11 and CA 22427-10 (S. S.. L. B. C): M. U. was a recipient of a DAAD fellowship (Federal Republic of Germany) in the Gene Technology Program. 2To whom requests for reprints should be addressed, at Room 1020, Institute of Cancer Research, Columbia University, 701 W. 168th St., New York, NY 10032. ' The abbreviations used are: PKC. protein kinase C; PS, phosphatidylserine; PDBu, phorbol-12.13-dibutyrate: TPA, 12-O-tetradecanoylphorbol-13-acetate; EGTA, ethyleneglycol bis(/j-aminoethyl ether)-A',/V,A'',A''-tetraacetic acid; PMSF, phenylmethylsulfonyl fluoride; IC50, 50% inhibitory concentration; SD, standard deviation: TLC, thin layer chromatography; DECA, l.l'-decamethy- lene-bis-4-aminoquinaldinium diiodide (dequalinium); OCTA, l.l'-octamethy- lene-bis-4-aminoquinaldinium diiodide; HEXA, 1,1'-hexamethylene-bis-4-amin- oquinaldinium diiodide; MAQ, l-methyl-4-aminoquinaldinium iodide; HAQ, I- hexyl-4-aminoquinaldinium iodide; BQ-10. 1,1'-decamethylene-bis-quino- Iininin diiodide; cDNA, complementary DNA; TPCK. L-l-tosylamido-2-phenyl- ethyl chloromethyl ketone; EGFR. epidermal growth factor receptor: DMSO, dimethyl sulfoxide. uct of phosphatidylinositol turnover, which, in turn, is stimu lated by a variety of hormones and agonists (2). The enzymatic activity termed "PKC" is expressed by a family of at least seven known structurally and functionally related isozymes (3-9) whose distribution varies widely among somatic and neural tissues (10, 11). Rat brain expresses at least four isozymes, o, j3i, 02, and 7, each of which is distinct immunogenically and is found in characteristic anatomical locations of this tissue (11, 12). Whether the diversity of enzyme structure and tissue distribution reflect differences in the response to stimuli or in the selection of substrates is not yet known, although recent studies provide evidence for subtle functional differences in cofactor requirements between individual PKC isozymes (11, 13). One approach to studying the PKC isozymes individually has been to utilize the cDNA clone for a given isozyme in order to overproduce the protein product in a suitable cell type (14, 15). This laboratory recently cloned a cDNA encoding the rat brain 0i isoenzyme. The cDNA was inserted into the retroviral vector pMV7 (16) which was then packaged into defective retrovirus particles by ^ 2 cells (17). Rat 6 fibroblasts were virally infected with the vector such that the 0, enzyme was constitutively expressed at high levels. The overexpression of PKC caused abnormalities in morphology and growth control (14). More recently, we used the same strategy to develop derivatives of the murine embryo fibroblast cell line C3H10T'/j that stably express high levels of rat PKC-0, (18). This approach provided a novel cell culture model for functional studies of PKC and also permitted the convenient isolation of a single isozyme in large quantity. Biochemical characterization of this PKC activ ity with regard to its Chromatographie behavior on hydroxyl apatite, response to tumor promoters, reactivity with PKC- specific antibodies, and sensitivity to known PKC inhibitors, indicated that the overproduced PKC-0, enzyme exhibits prop erties consistent with those of native brain PKC (19). Because PKC is thought to have a key role in regulating growth in many cell types, it may be a candidate target enzyme for the design of novel antineoplastic agents (20). The present work describes the investigation of dequalinium as a potential inhibitor of PKC, a study prompted by the published report (21) that this lipophilic, antimicrobial agent exhibits anticarci noma activity. In that study, dequalinium was shown to be 125- fold more toxic to human carcinoma cells such as MCF-7 than to normal epithelial cells such as CV-1. Furthermore, dequa linium potently inhibited the growth of human colon carcinoma (MB49 or CXI), or rat colon carcinoma (W163) that had been s.c. implanted in nude mice. Dequalinium also inhibited growth and led to the regression of mammary tumors chemically in duced in situ by the carcinogen 7,12-dimethylbenz(a)anthracene (22). The selective toxicity of this drug apparently arises from the higher membrane potential maintained across membranes of mitochondria in carcinoma cells (23). Thus, mitochondria concentrate lipophilic cations such as dequalinium to a much higher degree than normal cells leading, in response to the large concentration gradient, to a slow release of the drug into the cytoplasm. A previous study has shown that one of the cyto- 677 Research. on January 24, 2021. © 1990 American Association for Cancer cancerres.aacrjournals.org Downloaded from

Inhibition of Rodent Protein Kinase C by the Anticarcinoma ... · (CANCER RESEARCH 50. 677-685. Februar} l, 1990) Inhibition of Rodent Protein Kinase C by the Anticarcinoma Agent

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Inhibition of Rodent Protein Kinase C by the Anticarcinoma ... · (CANCER RESEARCH 50. 677-685. Februar} l, 1990) Inhibition of Rodent Protein Kinase C by the Anticarcinoma Agent

(CANCER RESEARCH 50. 677-685. Februar} l, 1990)

Inhibition of Rodent Protein Kinase C by the Anticarcinoma Agent Dequalinium1

Susan A. Rotenberg,2 Stephen Smiley, Marius Ueffing, Robert S. Krauss, Lan Bo Chen, and I. Bernard Weinstein

Institute of Cancer Research, Columbia University, New York, New York 10032 fS. A. R., M. £/.,R. S. A'., /. B. W.], and Dana-Farber Cancer Institute, Harvard Medical

School, Boston, Massachusetts 02115 [S. S., L. B. C.¡

ABSTRACT

Dequalinium has previously been shown to be an anticarcinoma agent(M. J. Weiss et al., Proc. Nati. Acad. Sci. USA, 84: 5444-5448, 1987).The present study demonstrates that it can inhibit protein kinase C-ß,isolated from an overproducing cell line with a 50% inhibitory concentration of 8-15 MM-Further examination of the inhibition by using structuralanalogues of dequalinium reveals that the length of the méthylènebridgebetween the two quinaldinium moieties, the presence of the ring substit-uents, and the bipartite character of the compound each contributes tothe inhibitory potency. Related studies show that the analogues displaythe same rank order of inhibitory potency when tested with the trypsin-generated catalytic fragment of the enzyme, indicating that dequaliniuminhibits kinase activity through an interaction with the catalytic subunit.Further studies argue that the ability of a given analogue to inhibitphosphotransferase activity correlates with its ability to compete with|JH|phorbol-12,13-dibutyrate binding on the intact enzyme (50% inhibitory concentration of 2-5 MM)-This suggests that the inhibitor is eitherbinding directly to the regulatory subunit as well, or that due to itsinteraction with the catalytic subunit, dequalinium produces an indirecteffect on sites defined by phorbol ester binding. Kinetic analysis revealedthat inhibition is noncompetitive with respect to ATP or phosphatidyl-serine. Studies conducted with types I, II, and III rat brain isozymes,resolved by hydroxylapatite chromatography, demonstrate that dequalinium inhibits each of them with similar potency (50% inhibitory concentration of 11 MM)and imply that the site of contact on the enzyme isa highly conserved region. Morphology studies with dequalinium in intactcells demonstrate that the inhibitor can protect control cells againstphorbol ester-induced morphology changes but cannot protect proteinkinase C-overproducing cells, suggesting that an elevation in proteinkinase C levels alone is sufficient to overturn the protection conferred bydequalinium. On the basis of these results, we propose that protein kinaseC could be a critical in vivo target of dequalinium.

INTRODUCTION

Over the last five years the fields of biochemistry and molecular biology have been harnessed to study the structure andfunction of PKC,1 a calcium- and phospholipid-dependent pro

tein kinase that participates in many cell regulatory processes.This enzyme serves as both the receptor for phorbol ester tumorpromoters (1) and as a key enzyme in growth factor-mediatedsignal transduction (2). It is activated by diacylglycerol, a prod-

Received5/19/89;revised9/27/89;accepted10/17/89.The costs of publication of this article were defrayed in part by the payment

of page charges. This article must therefore be hereby marked advertisement inaccordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1This work was supported by the following grants: NIH CA 02656 (S. A. R.,I. B. W.): National Research Service Award CA 08346 (R. S. K.); CA 19589-11and CA 22427-10 (S. S.. L. B. C): M. U. was a recipient of a DAAD fellowship(Federal Republic of Germany) in the Gene Technology Program.

2To whom requests for reprints should be addressed, at Room 1020, Institute

of Cancer Research, Columbia University, 701 W. 168th St., New York, NY10032.

' The abbreviations used are: PKC. protein kinase C; PS, phosphatidylserine;PDBu, phorbol-12.13-dibutyrate: TPA, 12-O-tetradecanoylphorbol-13-acetate;EGTA, ethyleneglycol bis(/j-aminoethyl ether)-A',/V,A'',A''-tetraacetic acid;

PMSF, phenylmethylsulfonyl fluoride; IC50, 50% inhibitory concentration; SD,standard deviation: TLC, thin layer chromatography; DECA, l.l'-decamethy-lene-bis-4-aminoquinaldinium diiodide (dequalinium); OCTA, l.l'-octamethy-lene-bis-4-aminoquinaldinium diiodide; HEXA, 1,1'-hexamethylene-bis-4-amin-oquinaldinium diiodide; MAQ, l-methyl-4-aminoquinaldinium iodide; HAQ, I-hexyl-4-aminoquinaldinium iodide; BQ-10. 1,1'-decamethylene-bis-quino-Iininin diiodide; cDNA, complementary DNA; TPCK. L-l-tosylamido-2-phenyl-ethyl chloromethyl ketone; EGFR. epidermal growth factor receptor: DMSO,dimethyl sulfoxide.

uct of phosphatidylinositol turnover, which, in turn, is stimulated by a variety of hormones and agonists (2). The enzymaticactivity termed "PKC" is expressed by a family of at least seven

known structurally and functionally related isozymes (3-9)whose distribution varies widely among somatic and neuraltissues (10, 11). Rat brain expresses at least four isozymes, o,j3i, 02, and 7, each of which is distinct immunogenically and isfound in characteristic anatomical locations of this tissue (11,12). Whether the diversity of enzyme structure and tissuedistribution reflect differences in the response to stimuli or inthe selection of substrates is not yet known, although recentstudies provide evidence for subtle functional differences incofactor requirements between individual PKC isozymes (11,13).

One approach to studying the PKC isozymes individually hasbeen to utilize the cDNA clone for a given isozyme in order tooverproduce the protein product in a suitable cell type (14, 15).This laboratory recently cloned a cDNA encoding the rat brain0i isoenzyme. The cDNA was inserted into the retroviral vectorpMV7 (16) which was then packaged into defective retrovirusparticles by ^ 2 cells (17). Rat 6 fibroblasts were virally infectedwith the vector such that the 0, enzyme was constitutivelyexpressed at high levels. The overexpression of PKC causedabnormalities in morphology and growth control (14). Morerecently, we used the same strategy to develop derivatives ofthe murine embryo fibroblast cell line C3H10T'/j that stablyexpress high levels of rat PKC-0, (18). This approach provideda novel cell culture model for functional studies of PKC andalso permitted the convenient isolation of a single isozyme inlarge quantity. Biochemical characterization of this PKC activity with regard to its Chromatographie behavior on hydroxylapatite, response to tumor promoters, reactivity with PKC-specific antibodies, and sensitivity to known PKC inhibitors,indicated that the overproduced PKC-0, enzyme exhibits properties consistent with those of native brain PKC (19).

Because PKC is thought to have a key role in regulatinggrowth in many cell types, it may be a candidate target enzymefor the design of novel antineoplastic agents (20). The presentwork describes the investigation of dequalinium as a potentialinhibitor of PKC, a study prompted by the published report(21) that this lipophilic, antimicrobial agent exhibits anticarcinoma activity. In that study, dequalinium was shown to be 125-fold more toxic to human carcinoma cells such as MCF-7 thanto normal epithelial cells such as CV-1. Furthermore, dequalinium potently inhibited the growth of human colon carcinoma(MB49 or CXI), or rat colon carcinoma (W163) that had beens.c. implanted in nude mice. Dequalinium also inhibited growthand led to the regression of mammary tumors chemically induced in situ by the carcinogen 7,12-dimethylbenz(a)anthracene(22). The selective toxicity of this drug apparently arises fromthe higher membrane potential maintained across membranesof mitochondria in carcinoma cells (23). Thus, mitochondriaconcentrate lipophilic cations such as dequalinium to a muchhigher degree than normal cells leading, in response to the largeconcentration gradient, to a slow release of the drug into thecytoplasm. A previous study has shown that one of the cyto-

677

Research. on January 24, 2021. © 1990 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 2: Inhibition of Rodent Protein Kinase C by the Anticarcinoma ... · (CANCER RESEARCH 50. 677-685. Februar} l, 1990) Inhibition of Rodent Protein Kinase C by the Anticarcinoma Agent

INHIBITION OF PROTEIN KINASE C BY DEQUALINIUM

plasmic effects of dequalinium is to act as an antagonist ofcalmodulin (24).

Using PKC-/Õ,isolated from the C3H10T'/2 cell system de

scribed above, we show that dequalinium is also an inhibitor ofprotein kinase C-ßtactivity in vitro and present structure-activity studies that examine the structural determinants of theinhibition. We determine the sites on the enzyme with whichthe inhibitor interacts by testing for its interference with phos-photransferase activity of the isolated catalytic fragment, andfor competition with phorbol ester binding on the regulatorysubunit of the holoenzyme and in intact cells. We also testwhether the individual rat brain isozymes types I, II, and IIIresolved by hydroxylapatite chromatography, exhibit a difference in sensitivity to dequalinium. Preliminary studies withintact C3H10'/2T cells which overproduce PKC-/Õ,, and the

vector control cell line, provide evidence that dequalinium caninterfere directly with a PKC-mediated event in vivo.

MATERIALS AND METHODS

Materials

Tissue culture medium and calf serum were purchased from FlowLaboratories, Inc., 150-mm plates were from Nunc, and G418 wasfrom Gibco. Protamine sulfate, histone III-S, DEAE-Sephacel, bovineserum albumin, bovine -y-globulin, polyethylene glycol (M, 6000-8000),TPCK-trypsin-agarose, and phosphatidylserine were from Sigma.Phenyl-Sepharose was purchased from Pharmacia, hydroxylapatite(Bio-Gel HT) was a product of Bio-Rad, and [7-32P]ATP (0.5-3 Ci/mmol) was bought from Amersham. The EGF receptor synthetic pep-tide substrate (R-K-R-T-L-R-R-L) was synthesized by J. Wideman(Columbia University), and leupeptin was a gift from W. Troll (NewYork University). ['HJPDBu was purchased from Du Pont New Eng

land Nuclear, TPA and nonradioactive PDBu were products of LCServices (Woburn, MA).

Methods

Cell Culture. C3H10T'/2-PKC-4 cells (18) were cultured in Dulbec-co's modified Eagle's medium containing 10% calf serum for a maxi

mum of 2 months. With the exception of the final passage prior toharvesting, the cells were maintained under selection with 50 Mg/mlG418 (14). Cells were grown in 150-mm plates for 4 days or untilconfluent. These cells have an 11-fold increase in PKC activity whencompared to control C3H10T'/2 (18).

Isolation of PKC. PKC-/3,, derived from C3H10T'/2-PKC-4 cells was

isolated by using previously described methods (14). Briefly,C3H10T'/2-PKC-4 cells were harvested at confluence, typically fromtwenty-five 150-mm plates (2.7 x IO7cells/plate), and lysed in 10 m\i

Tris, pH 7.5, 5 mivi EGTA, 5 mivi EDTA, 0.25 mivi PMSF, 10 Mg/mlleupeptin, 10 Mg/ml soybean trypsin inhibitor, 15 mivi 2-mercaptoeth-anol, and 0.1% Triton X-100 (v/v) (extraction buffer).

Following 25 strokes with a Dounce homogenizer and centrifugationat 9000 rpm for 20 min at 4°C,the extract was applied to a disposable

Bio-Rad column containing 1 ml DEAE-Sephacel that had been equilibrated with extraction buffer containing no Triton X-100. After theftowthrough activity (typically <1 unit/ml) was collected, the columnwas washed with 10 column volumes of Buffer A (20 mM Tris, pH 7.5,2 mM EGTA, 2 mM EDTA, 1 mM dithiothreitol, 1 mM PMSF, 10 n%/ml leupeptin, 10 Mg/ml soybean trypsin inhibitor), and PKC was theneluted with 3-5 column volumes of Buffer A plus 100 mM NaCl. Whenstored at -70°C in 10% glycerol and 0.05% Triton X-100, this crude

preparation of PKC-ßi(0.3 Mg/ml) showed significant loss of activity,sometimes as high as 50%, probably due to the freeze-thaw process.The length of storage at -70"C, however, did not further affect the

recovered activity. The specific activity of the enzyme used for theexperiments described was typically 6-17 units/mg, where 1 unit is 1nmol "Pi transferred to substrate per min (see below). Partially purifiedPKC-01 preparations were judged adequate for these studies since, with

respect to total PKC activity, such preparations consisted of approximately 93% ßiisoform. This homogeneity was shown by hydroxylapatite chromatography (19), and represents the highest enrichment inPKC-ß,reported for this isoform.

Isolation of the mixture of isozymes (3.4 units/mg) from rat brainwas carried out similarly, except that the extraction buffer contained20 mM Tris, pH 7.5, 10 mM EGTA, 2 mM EDTA, 5 mM dithiothreitol.1 mM PMSF, 10 Mg/ml aprotinin, 10 Mg/ml leupeptin, and 10 Mg/mlsoybean trypsin inhibitor. Further resolution of the rat brain enzymepreparation into individual isozymes was accomplished by using a stepelution procedure with phenyl-Sepharose, in order to exclude a largelyhydrophilic population of proteins, followed by elution from hydroxylapatite with a potassium phosphate gradient (20-200 mM). Thehydroxylapatite chromatography is a method previously shown to resolve the individual isozymes of the brain mixture (19, 25, 26). Following storage at -70°C, the specific activities of types I, II, and III

isoforms on the day of the experiment were 71, 29, and 40 units/mg,respectively. Purification of the /3, isoform from C3H10T'/2-PKC-4

cells was carried out in an identical manner.Protein content of each enzyme sample was determined by the Bio-

Rad protein assay with lysozyme as standard.Protein Kinase C Assay. PKC activity was taken as the difference in

the amount of 12P¡transferred from |7-'2P]ATP to a synthetic peptide

sequence of the EGF receptor (R-K-R-T-L-R-R-L) in the presenceand absence of PS. The reaction medium (0.12 ml), placed in 13- x100-mm disposable glass test tubes, consisted of 20 mM Tris, pH 7.5,10 mM MgCl2, 1 mM CaCl2, and the following components were addedin order: 4.0 Mgsynthetic peptide, 10 MgPS or 10 M'H^O, 10 p\ enzyme(3.8 fig), and 10 M!of test compound. All dequalinium solutions weremade up fresh on the day of the experiment. They were solubilized inmethanol and serially diluted into water for preparation of workingsolutions. At no time did the final percentage of methanol exceed 8%;control experiments demonstrated that the presence of methanol up to10% had no effect on PKC phosphotransferase activity. For eachinhibitor concentration, duplicate sets of tubes were prepared with andwithout phospholipid. PS working solutions were prepared by dryingthe stock PS (dissolved in chloroform), using N2, adding 1 ml sterilized,deionized H2O. and sonicating with three 10-s bursts with a sonifiercell disrupter. Model W185 (Heat Systems, Ultrasonics, Inc.). Following the addition of test compound, the tubes were incubated on ice for10 min. The kinase reaction was initiated by the addition of 73 MM[7-32P]ATP (approximately 200 cpm/pmol) to each tube, which was immediately transferred to a 30°Cwaterbath for 10 min. The reaction was

quenched by transferring 40 M'of the reaction medium to a 3- x 3-cmsquare of phosphocellulose paper and immersing it in a 1.0-liter beakerof tap water. After terminating all reactions, the squares were washed5 times with 1 liter of water. Finally, each square was placed in ascintillation vial containing 7 ml Hydrofluor and was measured for 32P

content. The results for each duplicate set of tubes were averaged. Thedifference between the (+) PS and the (-) PS tubes was judged to beCa2+/PS-dependent activity. The presence of PS typically stimulatedthe kinase reaction 6- to 10-fold with the synthetic EGFR peptide assubstrate.

For assays measuring phosphorylation of histone III-S (average M,16,000), the conditions were identical to those described above exceptthat histone III-S (15, 21, or 42 MM)replaced the EGF receptor peptide.For assays in which protamine sulfate phosphorylation was measured,the reaction medium (0.12 ml) consisted of 20 mM Tris, pH 7.5, 10mM MgCl2, protamine sulfate (0.083 - 0.67 mg/ml), 10 ¡Aenzyme (0.3mg/ml), and test compound which was added last; no phospholipid orCa2* was present. The kinase reaction was carried out in triplicate

under the conditions described above. The reported results were givenas the means of these triplicates which differed by less than 15%. Allexperiments were repeated at least twice.

Isolation of Catalytic Fragment. Using PKC prepared by DEAE-Sephacel chromatography, as described above, the catalytic fragmentof the enzyme was released by limited proteolysis with trypsin. For thepurpose of controlling the time of exposure to protease, TPCK-trypsinimmobilized on agarose was used since centrifugation rapidly removesit from the reaction medium. The reaction was initiated by adding the

678

Research. on January 24, 2021. © 1990 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 3: Inhibition of Rodent Protein Kinase C by the Anticarcinoma ... · (CANCER RESEARCH 50. 677-685. Februar} l, 1990) Inhibition of Rodent Protein Kinase C by the Anticarcinoma Agent

INHIBITION OF PROTEIN KINASE C BY DEQUALINIUM

TPCK-trypsin-agarose to the enzyme suspension (l mg/ml) in anEppendorf tube such that there was 100 ^1 TPCK-trypsin-agarose/500p\ enzyme. The tube was rotated for 2 min at 8°C,and immediately

centrifugad for 1 min in a Fisher microcentrifuge (Model 235C). Thesupernatant, containing the catalytic fragment, was quickly transferredto a fresh tube to which PMSF (1 mM) and soybean trypsin inhibitor( 10 Mg/"1') were further added. The extent of proteolysis was confirmedby demonstrating the loss of PS and Ca"+ dependence of the kinase

reaction and the 6- to 8-fold higher level of activity in the absence ofPS. In order to resolve the catalytic fragment from any remaining intactenzyme, the product mixture was first diluted 3-fold with Buffer A,followed by chromatography on 0.5 ml DEAE-Sephacel. Typically, anunproteolyzed preparation of the PKC-ft isozyme yields a single peakwhen eluted from DEAE-Sephacel with a gradient of 0-0.3 M NaCl inBuffer A.4 Using a 10-ml gradient of 0-0.4 M NaCl in Buffer A, thetrypsin-proteolyzed PKC preparation eluted as two distinct activitypeaks (27). The first peak, representing residual intact PKC. displayeda 6-fold enhancement when PS was added. The second peak, representing the catalytic fragment, exhibited less than 2-fold stimulation byPS. Fractions (0.7 ml) representing the second peak were pooled to atotal volume of 2.1 ml. The PKC catalytic fragment in this pool (77units/mg) was used for triplicate assays with the dequalinium analogues,as described, but without added phospholipid. The results are given asthe means of these triplicates which differed by less than 10%.

[3H|Phorbol Ester Binding. [3H)PDBu binding was assayed in a reaction (0.25 ml) consisting of 80 mM Tris-Cl, pH 7.4, 0.1 mivicalciumchloride, 7.5 mM magnesium acetate, 1 mM dithiothreitol, phosphati-dylserine (25 Mg/ml), bovine serum albumin (5 mg/ml), enzyme preparation (3.8 fig), 10 nM [3H]PDBu, and test compound added to theindicated concentration. In order to assess nonspecific binding of [3H[-

PDBu, a duplicate set of tubes was prepared for each concentration oftest compound that included 30 /¿Mnonradioactive PDBu. The reactionwas conducted in 1.5-ml Eppendorf tubes which were incubated for 5min at 30"C and then cooled in an ice water bath. Estimation of bound[3H]PDBu was by the polyethylene glycol precipitation method (28),which entails the addition of bovine -y-globulin (0.5 mg) and polyeth

ylene glycol (12% final concentration) to each tube. After at least 30min on ice, the tubes were microfuged for 15 min at 8°C.Followingthe removal of the supernatant, the 3H content of the pellet was

measured by severing the tip of the Eppendorf tube and placing it in 7ml Hydrofluor. In order to allow solubilization of the pellet material,scintillation counting was conducted 1-2 h later. Specific binding wastaken as the difference between 3H content in the pellets resulting from

binding reactions conducted with and without nonradioactive PDBu.Typically, the nonspecific binding represented 30% of the total 3H in

the pellet. The results are reported as the means of triplicate valueswhich differed by less than 15%.

Studies with Intact Cells. Cultures of 10T'/2-MV7-5 and 10T'/2-PKC-4 cells grown as described previously (18), were seeded at one-third toone-half saturation density on 60-mm plates. Forty-eight h later thecultures were treated with either 10MMDECA in DMSO [0.1% DMSOfinal concentration (v/v)] or DMSO alone. Fifteen min after that thecells were treated with 100 ng/ml TPA in DMSO [0.01% solvent finalconcentration (v/v)) or DMSO alone. The cultures were viewed microscopically every 15-30 min for 6 h and were photographed 90 minfollowing TPA treatment.

Synthetic Methods

General. All chemicals and reagents used for synthesis of the dequalinium series of compounds were purchased from the Aldrich Chemical Co. (Milwaukee, WI). HPTLC-HLF chromatography plates werepurchased from Analtech (Newark, DE). The synthetic methods described below are well established and have appeared in earlier studies(29-31). With the exception of HAQ, they included elemental analysisand melting point data of the products. Characterization of the purityof the products used for the present studies was by melting pointdetermination with a Meltemp and by TLC. The most discriminatingsolvent system found for compounds of this type was chloroform/

acetone/methanol/acetic acid/water (50/20/10/10/5). All compoundswere stored protected from light at —¿�20°C.

MAQ. 4-Aminoquinaldine was refluxed with an equimolar amountof iodomethane in 2-butanone for 72 h. The precipitate was recovered

and recrystallized from methanol. Purity was assessed at greater than99% by TLC. The melting range was 269-270°C; reported meltingpoint, 270°C(32).

HAQ. Prepared in the same manner as MAQ except that iodomethane was replaced by 1-iodohexane. Purity was assessed at greaterthan 99% by TLC. The melting point range was 232-233°C.

HEXA. Three equivalents of 4-aminoquinaldine were refluxed with1equivalent of 1,6-diiodohexane for 72 h in 2-butanone. The precipitatewas collected, washed with acetone and diethyl ether, and recrystallized3 times from ethanol and methanol. TLC showed a major product(>95%) and a secondary contaminant which turned brown upon prolonged exposure to air. This contaminant is likely to be l-(6-iodohexa-methylene)-4-aminoquinaldinium iodide, a result of incomplete reaction. A trace amount of other impurities was present which is likely torepresent isomerie products in which the alkylation occurred at the 4-amino nitrogen rather than the heterocyclic nitrogen. The melting rangeof the product was 294-300°C; reported melting, 295-296°C (29, 30).

OCTA. Prepared in the same manner as HEXA except that 1,6-diiodohexane was replaced by 1,8-diiodooctane. TLC showed a singlemajor product (>98%). The melting range was 288-291°C; reportedmelting range, 294-295"C (29, 30).

DECA. Prepared in the same manner as HEXA except that 1,6-diiodohexane was replaced by 1,10-diiododecane. TLC showed a majorproduct (>95%). The impurities are likely to be similar to the isomersdiscussed by HEXA. The melting range was 302-304°C; reportedmelting range, 308-309°C (29, 30). The dichloride salt of DECA

(DEÇA-CI)is available commercially from Sigma. The major component of DEÇA-CImigrated at nearly the same rate as the iodide salt,but the number and quantities of impurities were noticeably greater.The melting range for DEÇA-CIfrom Sigma was 303-306°C;reportedmelting range, 317-318°C(29).

BQ-10. Prepared in the same manner as DECA except that 4-aminoquinaldine was replaced by quinoline. TLC showed a majorproduct (>90%) and a single other component. The melting range was230-232°C;reported melting range, 213-214°C(31).

RESULTS

Our initial studies focused on the DECA analogue of thedequalinium series (Fig. \A) since it is this compound that wasused in the anticarcinoma studies (21), and thus serves as theparent compound of the series. As shown in Fig. 2, we found

CH3 CH3

A. Dequalinium Series (x=6, 8, or 10)

N-(CH2),o-N

B. BQ-10

N—CH3

C. HAQ MAQ

' S. A. Rotenberg. Unpublished observations.Fig. 1. Structural formulae of DECA-related analogues. Synthesis of these

compounds are given in "Materials and Methods".

679

Research. on January 24, 2021. © 1990 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 4: Inhibition of Rodent Protein Kinase C by the Anticarcinoma ... · (CANCER RESEARCH 50. 677-685. Februar} l, 1990) Inhibition of Rodent Protein Kinase C by the Anticarcinoma Agent

INHIBITION OF PROTEIN KINASE C BY DEQUALINIUM

100

mo

10 20 30 40

DECA (¡J.M)

50

Fig. 2. Inhibition of PKC-/Ì,by dequalinium. PKC-/3, was assayed as describedin "Materials and Methods" in the presence of increasing concentrations of

DECA. An inhibition curve representative of several experiments is shown. PKCactivity (100% activity) in the absence of the drug was 12 units/mg. The concentration of DECA at which activity was reduced 50% was 14 /JM.

Analogue (uM)Fig. 3. Inhibition of PKC by dequalinium-related analogues. PKC-/3, was

assayed for inhibition of activity by the following compounds: DECA (A); OCTA(D); HEXA (•);HAQ (O); MAQ (A): BQ-10 (•).PKC activity in the absence ofdrug was 5-12 units/mg.

that the DECA analogue exerts a strong inhibitory effect onthe kinase activity of PKC-/3,, exhibiting a IC50 in the range of8-14 ¿¿M.This discovery prompted us to explore the structure-activity relationships of a series of analogues in which the lengthof the méthylènebridge was varied. The HEXA and OCTAanalogues were synthesized, representing lengths of the méthylènebridge as 6 and 8 carbons, respectively (Fig. \A ). Fig. 3presents the results of testing each compound on the ß,isozymein the manner described for the DECA compound in Fig. 2.The results indicate that inhibitory potency increases as theméthylènebridge is lengthened. In this series, the HEXA compound was found to be the least effective PKC-ß,inhibitor. Forfour determinations, the IC50 value for this compound was 71±11 fiM (SD), which is about 6 times higher than that ofDECA, signifying a diminished inhibitory effectiveness by thepresence of 6 méthylènecarbons instead of 10. Increasing thenumber of carbon units in the méthylènebridge to 8 and 10enhanced the inhibitory potency as shown by the progressivelylower ICso values obtained from three determinations for theOCTA (ICso = 25 ±3 MM),and DECA (IC50 = 11 ±3 MM)structures. We tested a related structure, BQ-10 (Fig. IB),which retains the 10 carbon méthylènebridge but lacks theamino and methyl substituents on both ring systems. Relativeto the parent compound, BQ-10 was >40 times less potent,exhibiting a IC50 >400 ¡¿M.Thus, inhibition of PKC depends

on the presence of the exocyclic ring substituents. The absenceof effect on kinase activity by the monoquinaldinium analogues(Fig. 1C) HAQ and MAQ (no inhibition up to 0.83 HIM),demonstrates that the bis structure of an analogue is also acritical determinant of its potency.

Under our standard assay conditions (see "Materials andMethods") the IC50 values described above were highly repro

ducible. Inhibition by DECA was found to be equally effectivewith PKC-01 preparations of different levels of purity. Thus,enzyme that had been isolated by DEAE-Sepharose, phenyl-Sepharose, and hydroxylapatite chromatographies, was as sensitive to DEÇAas enzyme prepared only by step elution fromDEAE-Sephacel, i.e., despite an 8-fold difference in proteincontent. Systematic variations in certain components of thereaction system, however, yielded pronounced alterations in theapparent IC50, indicating that the IC50 values reported here arenot absolute. One component found to be critical to inhibitorsensitivity was the PKC substrate chosen for the assay. Ourstandard assay contained as substrate a 30 ßMconcentration ofa synthetic octapeptide fragment of the EGFR, which containsthe threonine site phosphorylated by PKC in vivo (33). Whenhistone III-S was examined as a substrate of PKC-ßi,theenzyme was about 5-fold less sensitive to inhibition by theDECA analogue (IC50 = 50 ^M). In addition, we addressed theinhibitory effectiveness of the DECA analogue by using prota-mine sulfate as substrate. This artificial substrate of PKC canbe phosphorylated in the absence of phospholipid and calcium(34) and thereby reports solely on PKC catalytic function. Wefound that over a wide range of protamine sulfate concentration(0.083-0.67 mg/ml), PKC-ß,activity was consistently refractory to inhibition by DECA, even when tested at 100 MM(datanot shown here). These data indicate that the choice of substratecan dramatically determine the occurrence and extent of inhibition by dequalinium of PKC activity. In studies with stauro-sporine, a potent inhibitor of PKC (35), we have observed asimilar dependence of IC50on the substrate used in the assay.4

In addition to choice of substrate, variations in the concentrations of two other assay components, ATP or PS, affectedthe extent of inhibition by DECA. Shown in Fig. 4 are Line-weaver-Burk analyses of the dependence of inhibition on theconcentration of ATP and PS, respectively. For ATP concentrations in the range of 3-50 ßM,noncompetitive action byDECA was observed (Fig. 4A) such that K¡= 50 MM(i.e., theinhibitor concentration which doubled the slope of the reciprocal plot). At ATP concentrations higher than 50 MM(data notshown), no ATP-dependent effect of DECA was observed. Sincethe standard assay medium contained 73 MMATP, the observation of 50% inhibition by 8-14 UM DECA under standardconditions (Fig. 2) cannot be explained by interference withATP binding to the enzyme. Noncompetitive inhibition byDECA was also observed with respect to activation of PKC bythe phospholipid PS (Fig. 4B). Such findings indicated thatover the range of 60-240 UM PS, inhibition by DECA wasdecreased as the PS concentration was progressively elevated.The noncompetitive character of the plots for ATP and PSdependence implies that DECA is not interacting directly withthe sites occupied by these cofactors.

To gain further insight into the mechanism of dequalinium-mediated inhibition, we sought to identify the site or sites onthe enzyme with which DECA directly interacts to produceinhibition. We first considered that dequalinium might interferedirectly with the function of the catalytic subunit. To test this,we prepared the catalytic fragment by subjecting PKC-/3i tolimited proteolysis by trypsin, followed by chromatography on

680

Research. on January 24, 2021. © 1990 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 5: Inhibition of Rodent Protein Kinase C by the Anticarcinoma ... · (CANCER RESEARCH 50. 677-685. Februar} l, 1990) Inhibition of Rodent Protein Kinase C by the Anticarcinoma Agent

INHIBITION OF PROTEIN KINASE C BY DEQUALINIL'M

Fig. 4. Kinetic analysis of inhibition ofPKC-/3| by dequalinium. A, dependence onATP concentration. Lineweaver-Burk plot ofPKC-/J, activity (7 /jg enzyme/assay) wasmeasured in duplicate at various concentrations of [7-32P]ATP having a constant specific

radioactivity of 613 cpm/pmol (•).The/fm forATP was determined to be 15 JIM. The non-competitive behavior of inhibitor was measured at DECA concentrations of 50 MM(•)and 100 MM(A). B, dependence on PS concentration. Lineweaver-Burk plot of PKC-i), activity (3.5 pg enzyme/assay) was measured induplicate at various concentrations of PS(Avanti Polar Lipids) in the presence of DECAconcentrations of 26 MM(•).52 MM(•).and75 MM(A). In the absence of inhibitor, theenzyme activity was a constant value over therange of PS concentrations presented (60-240MM).

1/V(pmoles/min)"1 1/V(pmoles/min)'1

1/ATP(uM)-

0.005 0.010 0.015

1/PS (uM) -1

DEAE-Sephacel to resolve the catalytic fragment from residualintact enzyme ("Materials and Methods"). The results, shown

in Fig. 5, demonstrate the effects of the DECA, HEXA, andBQ-10 compounds on the activity of the catalytic fragment. Itwas found that the DECA analogue inhibited phosphotransfer-ase activity with a IC50 of 10 ^M; the HEXA analogue wassignificantly less inhibitory (IC50 = 120 MM),and the BQ-10compound was ineffective. Significantly, the rank order ofinhibitory potencies of these analogues obtained with the catalytic fragment was consistent with the rank order observed withthe intact enzyme (Fig. 3). Thus, the catalytic domain of PKC-ßiis a target for inhibition by DECA. When protamine sulfatewas used as a substrate of the catalytic fragment, there wasagain no interference by the DECA compound even when testedat 100 /^M(data not shown).

It is noteworthy that at DECA concentrations sufficient toinhibit >95% of the total activity of the holoenzyme (Fig. 1),there was only 75% inhibition of the activity obtained with theisolated catalytic fragment activity (Fig. 5). Unlike the experiments with the intact enzyme which measured the difference inphosphotransferase activity with and without phospholipid, theactivities reported for the catalytic fragment represented totalkinase activity in the absence of phospholipid. Thus, the residual activity evident in Fig. 5 may reflect either the presence ofother kinases which are able to phosphorylate the EGF receptor

100 Bj—O

>>A

—¿� 50

10

I10 20 50 100 150

Analogue (uM)

Fig. 5. Effect of DECA analogues on kinase activity of the catalytic fragmentof PKC-fi]. Assays of the phosphotransference activity of the catalytic fragmentin the presence of increasing concentrations of the indicated analogue were carriedout under the conditions described for the intact enzyme (see "Materials andMethods"), but without the addition of phosphatidylserine. All assays were

conducted in triplicate for each concentration of drug and data are expressed asthe average values. DECA (A), HEXA (•).and BQ-10 (HI).

peptide substrate, and/or a subpopulation of the PKC catalyticfragment molecules that is resistant to inhibition by DECA.

In light of the dual character of some other PKC inhibitorswhich can interact with both the catalytic and regulatory sub-units (36), the inhibitory potency of the dequalinium analogueswas assessed in terms of another function residing in the regulatory subunit, the region on PKC recognized by diacylglyceroland by phorbol esters such as PDBu. By using [3H]PDBu as a

probe, we examined the ability of dequalinium to displace thisradioactive marker from the intact enzyme. As shown in Fig.6, the specific binding of [3H]PDBu to PKC-/3, was dramaticallyreduced by the DECA analogue, with a IC50 of 2-5 MM,in thepresence of 25 MgPS/ml assay medium. HEXA was consistentlyless potent than DECA, while BQ-10 and HAQ had no inhibitory effect (data not shown). Thus, these studies demonstratethat the structural features which determine the potency forinhibiting the phosphotransferase activity of either the intactenzyme or the catalytic fragment are also reflected in the abilityto inhibit phorbol ester binding.

The identification of the regulatory subunit as another possible locus of action of dequalinium led us to consider that theinhibitor may discriminate among the a, ß,and y isozymesfrom rat brain since each isozyme has a characteristic aminoacid sequence at its NH2 terminus (11). PKC activity assayswere performed with DEÇAon a mixture of rat brain isozymesand also on the PKC isoforms resolved by hydroxylapatite,types I, II, and III. The results of these assays are shown inTable 1. In agreement with the potency of the DECA compound

Analogue (i/M)

Fig. 6. Effect of DECA analogues on [3H]PDBu binding. Assays for specificbinding of [3H]PDBu to PKC-/3, were carried out at the indicated concentrationsof the dequalinium analogues, as described in "Materials and Methods." PKC-/3,was prepared by DEAE-Sephacel chromatography. DECA (A), HEXA (•),andBQ-10 (•).

681

Research. on January 24, 2021. © 1990 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 6: Inhibition of Rodent Protein Kinase C by the Anticarcinoma ... · (CANCER RESEARCH 50. 677-685. Februar} l, 1990) Inhibition of Rodent Protein Kinase C by the Anticarcinoma Agent

INHIBITION OF PROTEIN KINASE C BY DEQUALINIUM

Table 1 Effect of DECA on PKC isozymes from rat brainThe mixture of PKC isozymes that eluted from DEAE-Sephacel (see "Mate

rials and Methods") and the three individual peaks resolved by hydroxylapatite

chromatography were assayed for inhibition of kinase activity by the DECAanalogue. For additional details see "Materials and Methods." Earlier studies(37) have established that types I, II, and III correspond genetically and ¡mmu-nochemically to y, ¡i.and a isoforms. respectively.

Rat brain isozyme IC,„(UM)

MixtureType IType IIType III

11IIII9

measured with PKC-0, isolated from the C3H10T'/2-PKC-4

cells, the mixture of rat brain isozymes exhibited a IC50 of 11¿iM.When tested with the DECA analogue, the rat brain peakactivities I, II, and III, which correspond to isozymes 7, 0, anda, respectively (37), displayed a similar IC50 value. Thus, de-qualinium effectively inhibits the native PKC activities frombrain but apparently does not recognize differences among theindividual isozymes. Because there may be other isozymes inrat brain that were not resolved by hydroxylapatite, we shouldstress that the a, ß,and 7 preparations from rat brain testedhere are probably not homogeneous.

Inhibition of PKC by DECA in Intact Cells. We next determined whether DECA could inhibit PKC in intact cells. Toaccomplish this, we used two derivations of the murine embryofibroblast line C3H10T'A developed in our laboratory (18).Cells designated 10T'/2-PKC-4 contain a retroviral expressionvector into which has been inserted a rat brain PKC-/3] cDNA.In these cells there is an 11-fold greater PKC enzyme activitythan in a control cell line, 10T'/2-MV7-5, which harbors theretroviral vector without a cDNA insert (18). 10T'/2-MV7-5cells, like parental lOT'/z cells, exhibit a characteristic morphological response when exposed to TPA; 10T'/2-PKC-4 cells

show an exaggerated response ( 18). We therefore tested whetherDECA could inhibit the morphological response of these celllines to the phorbol ester TPA. Cultures of 10T'/2-MV7-5 and10T'/2-PKC-4 were pretreated with DMSO (i.e., solvent con

trol) or 10 Õ/MDECA for 15 min and then challenged with 100ng/ml TPA. Ninety min after TPA treatment, 10T'/2-MV7-5

cells pretreated with DMSO showed a typical morphologicalresponse relative to non-TPA-treated control cultures (compareFig. 7, A and B). In contrast, cultures of 10T'/2-MV7-5 cells

pretreated with DECA show a much less profound alterationin morphology following TPA treatment (Fig. 1C). Treatmentwith DECA alone had no effect on the morphology of thesecells (data not shown). Thus, DECA can inhibit a primaryresponse of intact cells to TPA. 10T'/2-PKC-4 cells, which are

somewhat morphologically altered without TPA treatment ( 18),showed an exaggerated morphological response to TPA, relative to 10T'/2-MV7-5 cells (in Fig. 7, compare A and B with D

and £).However, DECA was unable to inhibit the response toTPA in these cells (Fig. IF). [DECA had a slight inhibitoryeffect in 10T'/2-PKC-4 at very early time points (~15 min),

before the full morphological response was achieved (data notshown)]. Thus, overproduction of PKC can overcome the inhibition by DECA of the effects of TPA. These data stronglysuggest that the ability of DECA to inhibit the morphologicalresponse to TPA in 10T'/2-MV7-5 cells occurs by its ability to

inhibit PKC directly.It should be noted that the inhibitory effect of DECA in

10T'/2-MV7-5 cells was transient. Five h after TPA treatment,the cultures receiving DECA and TPA showed a strong morphological response and the cultures were indistinguishablefrom cultures that received only TPA. Whether this is due to

metabolism of DECA to an inactive form, its uptake by mitochondria, or the gradual accumulation of TPA-induced, PKC-mediated phosphorylation events, is unclear.

DISCUSSION

The results of these studies indicate that micromolar concentrations of dequalinium markedly inhibit PKC activity. A seriesof analogues was synthesized to assess the contribution of theméthylènebridge, the ring substituents, and the bis structure tothe inhibition. The inhibitory potencies of these analogues weredetermined by using the partially purified /3¡isoform of PKCderived from C3H10T'/2 cells genetically engineered to over

produce this isoform (14, 18). Of those compounds tested, wefound that a bis structure containing a méthylènebridge of 10carbons and that retains the ring substituents is the most potentinhibitor of the /3i isoform. Analogues having a shorter méthylènebridge (6 or 8 carbons) attenuated the inhibitory potencyand analogues lacking either the ring substituents or the bisstructure were noninhibitory, implying that these features arecritical to inhibition. A future direction of the present workshould consider analogues having a méthylènebridge of 12 ormore carbons in order to establish an optimal bridge length.

Our studies also revealed that the sensitivity of PKC toinhibition by dequalinium is dependent on the substrate usedin the assay. Inhibition was highest with the EGFR syntheticpeptide (R-K-R-T-L-R-R-L) which contains the threoninesite phosphorylated by PKC in vivo (33). A 5-fold higher concentration of the drug was required to produce comparableinhibition when the substrate was histone, and no inhibitionwas seen when protamine was the substrate, even when theDECA concentration was 100 UM. Given that in all cases bothsubstrate and inhibitor were cationic, it is unlikely that thissubstrate-related effect is due to an interaction of substrate andinhibitor alone, although more extensive kinetic analysis wouldbe required to exclude this possibility. An alternative explanation of this phenomenon is suggested by previous studies ofPKC with synthetic peptides and histone substrates (38, 39).Such systematic surveys have indicated that the substrate affinity is enhanced significantly by increasing the number of cationic amino acids adjacent to the phosphorylation site. Moreover, relative to the peptide substrates tested, histone proteins,in general, were observed to have much lower Km values, suggesting that tertiary structure also has an important role indetermining this kinetic parameter. It is possible, therefore,that protein substrates like histone and protamine, which haveconsiderably more cationic character and tertiary structure thanthe EGFR peptide, can compete more effectively with DECAfor critical anionic residues in the substrate binding site of PKC.Further studies are required, however, to determine the exactmechanism. Nevertheless, our studies indicate that the choiceof substrate should be considered carefully when screeningpotential PKC inhibitors.

Another consideration emerging from these studies is ourfinding that increasing concentrations of PS decreased theinhibitory effect of DECA on PKC activity (Fig. 4B). In kineticstudies, phospholipid and DECA exhibited noncompetitive behavior implying that DECA does not bind directly to the PS-binding site. In view of the lipophilic nature of dequalinium,the drug may simply partition into the phospholipid micellarphase such that its availability to the enzyme is diminished.Consistent with this possibility, we found that high PS concentrations can attenuate the inhibitory potency of DECA evenwith the catalytic fragment of PKC (data not shown), i.e., under

682

Research. on January 24, 2021. © 1990 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 7: Inhibition of Rodent Protein Kinase C by the Anticarcinoma ... · (CANCER RESEARCH 50. 677-685. Februar} l, 1990) Inhibition of Rodent Protein Kinase C by the Anticarcinoma Agent

CONTROL

INHIBITION OF PROTEIN KINASE C BY DEQUALINIUM

+TPA +TPA + DECA

Fig. 7. Effect of DECA on phorbol ester action in intact cells. Photomicrographs of 10T'/¡-MV7-5and 10T'/2-PKC-4 cells exposed to DMSO (control), TPA, orTPA plus DECA. Cultures were exposed to 0.11% DMSO (A and D), 100 ng/ml TPA (B and £),or 100 ng/ml TPA and 10 JIMDECA (Cand F), x 200. See textfor details.

conditions in which the kinase activity is independent of phos- could bind in a substrate-independent manner consistent withpholipid. This result implies that PS can physically sequesterthe inhibitor. Thus, the IC50values we obtained for DECA withthe holoenzyme, and that have been determined for certainother lipophilic cations whose inhibition of PKC is a functionof the phospholipid concentration (40), are not absolute values.Consequently, it is difficult to predict from these subcellularstudies the inhibitory potency of this compound for PKC invivo.

The present studies suggest that DECA inhibits PKC bycausing perturbations in the function of both the catalytic andregulatory domains of this enzyme. Since we found that DECAand related analogues inhibited the phosphotransferase activityof the catalytic fragment with potencies similar to those seenwith the holoenzyme, it would appear that the primary targetof the drug is the catalytic domain. The fact that the inhibitoryefficacy of DECA is a function of the phosphotransferasesubstrate and the ATP concentration is also consistent withthis interpretation. At the same time, where previous studieshave established that the PDBu-binding site resides exclusivelyon the regulatory domain (41), we found that DECA inhibited[3H]PDBu binding to the PKC holoenzyme. Competition of

DECA with phorbol esters is compatible with the mechanismobserved for other PKC inhibitors that are lipophilic amines,i.e., the antineoplastic lipoidal amine CP-46,665-1 (42), andsphingosine (43).

It is noteworthy that the potency with which DECA displacedphorbol ester (IC50 = 2-5 ßM)was similar to the potency withwhich it inhibited phosphotransferase activity (IC50 = 8-14MM).In addition, the DECA, HEX A, and BQ-10 analogues hadthe same rank order of potency in inhibiting [-'H]PDBu binding

as they did in inhibiting the kinase activity of the intact enzyme.These binding studies were carried out in the absence of thepeptide substrate, demonstrating that DECA and its analogues

their inhibitory potencies. Although the IC50values observed inthe binding studies were consistently lower than those determined for kinase inhibition, we attribute this higher sensitivityto the fact that the PS concentration in the binding studies (25Mg/ml) was lower than the PS concentration in the kinaseinhibition assay (83 /ug/ml).

Thus, it is possible that DECA interacts with specific bindingsites on both the catalytic and regulatory domains of PKC.Alternatively, the inhibitor might occupy a single major bindingsite (on the catalytic domain) which blocks or induces a confor-mational change such that phorbol ester-binding sites on theregulatory domain become inaccessible. This would be consistent with current models of PKC activation (20, 44) which invokean unfolding of the protein in the presence of activating cofac-tors. A complex or mixed mode of inhibition has also beenproposed for several other lipophilic cationic PKC inhibitors,including the aminoacridines (45), tamoxifen (36, 46), trifluo-perazine (36), Adriamycin (36), and gossypol (36). Gossypol,in particular, has been shown to inhibit both [3H]PDBu binding

to the holoenzyme and phosphotransferase activity of the catalytic fragment (with histone III-S as substrate) with similarpotency, a phenomenon observed with DECA in the presentstudy. The site(s) of action of DEÇAon PKC is (are) apparentlyhighly conserved, since our studies with the overexpressed ratß,form and the individual rat brain isozymes revealed nosignificant differences in sensitivity to inhibition by this drug.

Using cell morphology as an indicator of phorbol ester action,we have shown that dequalinium does indeed interfere with theresponse of intact cells to phorbol ester. The transient protection from a saturating dose of phorbol ester was conferred onvector control cells by an external concentration of 10 ¿ÕM,consistent with its IC50in vitro. This experiment indicated that,relative to TPA, when DECA is present at 60-fold excess, it

683

Research. on January 24, 2021. © 1990 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 8: Inhibition of Rodent Protein Kinase C by the Anticarcinoma ... · (CANCER RESEARCH 50. 677-685. Februar} l, 1990) Inhibition of Rodent Protein Kinase C by the Anticarcinoma Agent

INHIBITION OF PROTEIN KINASE C BY DEQUALINIUM

can cause effective inhibition of phorbol ester binding in vivoand that an elevation of PKC-0, levels alone is sufficient tooverturn that inhibition. It is also noteworthy that despite theability of DECA to inhibit PKC with only certain substrates invitro, our studies with intact cells demonstrate the inhibition ofPKC action with native substrates.

Although this experiment showed that PKC can serve as anin vivo target of dequalinium, it did not examine other cellularcomponents that could serve as targets in the drug concentration range studied. Dequalinium has been shown previously toact as an antagonist of calmodulin (IC50 = 1 MM)in the in vitroactivation of a cyclic AMP phosphodiesterase (24, 47), apparently by binding directly to calmodulin. This effect of DECAcorrelated with its potency as an antiproliferative agent in a C6astrocytoma cell line (47). That tamoxifen, another antitumorlipophilic cation, is both a PKC inhibitor and a calmodulinantagonist in the activation of a cyclic AMP phosphodiesterase(48) suggests that the antitumor effects of these and otherlipophilic cationic drugs are due to interactions with more thanone cellular target.

In the present study, the analysis of DECA-mediated inhibition of PKC was greatly faciliated by the use of preparations ofthe overproduced PKC-ß,isoform which were virtually homogenous (93%) with respect to total PKC activity (19). Consequently, the possible ambiguity imposed by the presence ofother PKC isozymes was avoided. We suggest that the use ofcell systems that have been genetically engineered to overproduce a specific PKC isoform should provide valuable tools forexamining the potency and specificity of any potential PKCinhibitor in both enzyme preparations and intact cells. Furtherstudies are in progress regarding the possible in vivo role ofPKC as a critical cellular target for the anticarcinoma activityof DECA and other potential PKC inhibitors, using thesegenetically engineered cell systems.

ACKNOWLEDGMENTS

We thank Sheila M. Washington for her expert skills in the preparation of this manuscript. We also acknowledge Dr. Darryl Rideout(Scripps Institute) for stimulating discussions.

REFERENCES

1. Nishizuka, Y. The role of protein kinase C in cell surface signal transductionand tumor promotion. Nature (Lond.). 308: 693-698. 1984.

2. Nishizuka, Y. Studies and perspectives of protein kinase C. Science (Wash.DC), 223: 305-312. 1986.

3. Cousscns, L.. Parker. P.. Rhee, L., Yang-Feng, T. L.. Chen. E.. Waterfield.M. D.. Francke. U., and Ullrich. A. Multiple distinct forms of bovine andhuman protein kinase C suggest diversity in cellular signaling pathways.Science (Wash. DC), 233: 859-866. 1986."

4. Housey, G. M., O'Brian. C. A.. Johnson, M. D.. Kirschmeier, P., and

Weinstein. I. B. Isolation of cDNA clones encoding protein kinase C:evidence for a protein kinase C-related gene family. Proc. Nati. Acad. Sci.USA, A4: 1065-1069, 1987.

5. Knopf, J. L., Lee, M-H.. Sultzman, L. A.. Kriz, R. W., Loomis, C. R..Hewick, R. M., and Bell. R. M. Cloning and expression of multiple proteinkinase C cDNAs. Cell, 46: 491-502. 1986.

6. Ono. Y., Kurokawa, T., Fujii, T.. Kawahara, K., Igarashi. K.. Kikkawa, U.,Ogita, K.. and Nishizuka, Y. Two types of complementary DNAs of rat brainprotein kinase C. FEBS Lett.. 206: 347-352. 1986.

7. Parker, P. J.. Coussens, L., Totty, N., Rhee. L.. Young. S.. Chen. E., Stabel,S., Waterfield, M. D.. and Ullrich, A. The complete primary structure ofprotein kinase C—the major phorbol ester receptor. Science (Wash. DC).233: 853-859, 1986.

8. Kikkawa, U., Ogita. K., Ono. Y., Asaoka, Y.. Shearman. M. S.. Fujii. T.,Ase, K., Sekiguchi, K., Igarashi, K., and Nishizuka, Y'.The common structure

and activities of four subspecies of rat brain protein kinase C family. FEBSLett., 2^:212-216, 1987.

9. Ono. Y., Fujii, T.. Ogita, K.. Kikkawa. U.. Igarashi. K.. and Nishizuka, Y.Identification of three additional members of rat protein kinase C family:6-, t-, and f-subspecies. FEBS Lett., 226: 125-128. 1987.

10.

11.

12.

13.

14.

15.

16.

17.

18.

19.

20.

21.

23.

24.

25.

26.

27.

28.

29.

30.

31.

32.

33.

34.

35.

36.

37.

38.

Kosaka. N'.. Ogita, K.. Äse.K.. Nomura. H.. Kikkawa, U.. and Nishi/uka, Y.

The heterogeneity of protein kinase C in various rat tissues. Biochem.Biophys. Res. Commun.. 151: 973-981. 1988.Nishizuka, Y. The molecular heterogeneity of protein kinase C" and itsimplications for cellular regulation. Nature (Lond.). 334: 661-665. 1988.Saito, N.. Kikkawa. U.. Nishizuka. Y.. and Tanaka. C'. Distribution of proteinkinase C-like ¡nimunoreactiveneurons in rat brain. J. Neurosci., 8: 369-382.1988.Huang. K-P., Huang. F. L., Nakabayashi. H.. and Yoshida, Y. Biochemicalcharacterization of rat brain protein kinase C isozymes. J. Biol. Chem., 263:14839-14845. 1988.Housey. G. M., Johnson, M. D.. Hsiao. W-L., O'Brian. C. A. Murphy, J.

P., Kirschmeier, P., and Weinstein. I. B. Overproduction of protein kinaseC causes disordered growth control in rat fibroblasts. Cell, 52: 343-354,1988.Persons, D. A.. Wilkison, W. O., Bell. R. M.. and Finn. O. J. Altered growthregulation and enhanced tumorigenicity of NIH 3T3 fibroblasts transfectedwith protein kinase C-I cDNA. Cell. 52: 447-458, 1988.Kirschmeier, P. T.. Housey, G. M.. Johnson, M. D.. Perkins. A. S., andWeinstein. I. B. Construction and characterization of a retroviral vectordemonstrating efficient expression of cloned cDNA sequences. DNA (NY),7:219-225. 1988.Mann. R., Mulligan. R. C.. and Baltimore. D. Construction of a retroviruspackaging mutant and its use to produce helper-free defective retrovirus.Cell. 33: 153-159. 1983.Krauss, R. S., Housey. G. M., Johnson, M. D.. and Weinstein, I. B. Disturbances in growth control and gene expression in a C3H10T1/; cell line thatstably overproduces protein kinase C. Oncogene. 4: 991-998, 1989.Rotenberg, S. A., Krauss. R. S.. Borner. C. M. B., and Weinstein, l. B.Characterization of a specific form of protein kinase C overproduced by aC3H10T'/z cell line. Biochem. J., in press, 1990.Weinstein. I. B. The origins of human cancer. Cancer Res., 48: 4135-4143.1988.Weiss, M. J., Wong, J. R.. Ha, C. S.. Bleday, R., Salem. R. R.. Steele, G.D., Jr., and Chen, L. B. Dequalinium. a topical antimicrobial agent, displaysanticarcinoma activity based on selective mitochondria] accumulation. Proc.Nati. Acad. Sci. USA 84: 5444-5448. 1987.Chen, L. B.. Weiss, M. J., Davis, S.. Bleday. R. S., Wong, J. R., Song, J.,Tcrasaki. M.. Shepherd, E. L., Walker. E. S.. and Steele, G. D., Jr. Mitochondria in living cells: effects of growth factors and tumor promoters,alterations in carcinoma cells, and targets for therapy. Cancer Cells (ColdSpring Harbor), 3: 433-443, 1985.Chen. L. B. Mitochondrial membrane potential in living cells. Annu. Rev.Cell Biol.. 4: 155-181. 1988.Hait. W. N. Targeting calmodulin for the development of novel cancerchcmotherapeutic agents. Anti-Cancer Drug Design. 2: 139-149. 1987.Huang. K-P.. Nakabayashi, H.. and Huang. F. L. Isozymic forms of rat brainCa2*-activated and phospholipid-dependent protein kinase. Proc. Nati. Acad.Sci. USA S3: 8535-8539. 1986.Pelosin. J-M.. Vilgrain. I., and Chambaz, E. M. A single form of proteinkinuse C is expressed in bovine adrenocortical tissue, as compared to fourchromatographically resolved isozymes in rat brain. Biochem. Biophys. Res.Commun., 147: 382-391, 1987.O'Brian. C. A., and Ward, N. E. Binding of protein kinase C to N-(6-aminohexyl)5-chloro-l-naphthalenesulfonamide through its ATP bindingsite. Biochem. Pharmacol.. 38: 1737-1742. 1989.Sharkey, N. A.. Leach, K. L., and Blumberg. P. M. Competitive inhibitionby diacylglycerol of specific phorbol ester binding. Proc. Nati. Acad. Sci.USA«/: 607-610. 1984.Taylor, E. P.. Austin, W. C.. Collier. H. O. J.. and Potter. M. D. BritishPatent 745.956. 1956.Austin. W. C., Potter. M. D., and Taylor. E. P. Potential trypanocides. Theaction of polymethylene dihalides on 4-aminoquinaldine. J. Chem. Soc.(Lond.). pp. 1489-1498. 1958.Taylor, E. P. Synthetic neuromuscular blocking agents. Part I. Heterocyclicdecamethylene bis (quaternary ammonium salts). J. Chem. Soc. (Lond.). pp.1150-1157, 1951.Ashley. J. N.. Browning, C. H.. Cohen. J. B.. and Gulbransen. R. Theantiseptic and trypanocidal properties of some anil and styryl derivatives of4-amino quinaldine. Proc. R. Soc. Lond. B Biol. Sci., 113: 293-299, 1933.Hunter. T.. Ling. N.. and Copper, J. A. Protein kinaseCof the EGF receptorat a threonine residue close to the cytoplasmic face of the plasma membrane.Nature (Lond.). 311: 480-483. 1984.Takai, Y.. Kishimoto. A.. Iwasa. Y., Kaw'ahara, Y.. Mori. T., and Nishizuka,Y. Calcium-dependent activation of a multifunctional protein kinase bymembrane phospholipids. J. Biol. Chem., 254: 3692-3695, 1979.Tamaoki, T., Nomoto. H., Takahashi. !.. Kalo, Y.. Morimoto, M., andTornita, F. Staurosporine, a potent inhibitor of phospholipid/Ca2* dependentprotein kinase. Biochem. Biophys. Res. Commun.. 135: 397-402. 1986.Nakadate. T.. Jeng. A. Y., and Blumberg, P. M. Comparison of proteinkinase C functional assays to clarify mechanisms of inhibitor action.Biochem. Pharmacol.. 3 7: 1541-1545. 1988.Kikkawa, U., Ono. Y., Ogita. K.. Fujii. T.. Asaoka. Y., Sekiguchi, K., Kosaka,Y'., Igarashi. K.. and Nishizuka. Y. Identification of the structures of multiplesubspecies of protein kinase C expressed in rat brain. FEBS Lett.. 2/7: 227-231, 1987.Woodgett. J. R.. Gould. K. L.. and Hunter. T. Substrate specificity of proteinkinase C. Eur. J. Biochem.. 161: 177-184, 1986.

684

Research. on January 24, 2021. © 1990 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 9: Inhibition of Rodent Protein Kinase C by the Anticarcinoma ... · (CANCER RESEARCH 50. 677-685. Februar} l, 1990) Inhibition of Rodent Protein Kinase C by the Anticarcinoma Agent

INHIBITION OF PROTEIN KINASE C BY DEQUALINIUM

39. Ferrari, S., Marchiori. F., Borin, G., and Pinna. L. A. Distinct structural binding in vitro and in human platelets. J. Biol. C'hem., 2dl: 12604-12609,requirements of Ca!*/phospholipid-dependent protein kinase (protein kinase 1986.C) and cAMP-dependent protein kinase as evidenced by synthetic peptide 44. House. C., and Kemp. B. E. Protein kinase C contains a pseudosubstratesubstrates. FEBS Lett., 184:12-11, 1985. prototype in its regulatory domain. Science (Wash. DC), 238: 1726-1728.

40. Mori, T., Takai, Y.. Minakuchi. R., Yu. B.. and Nishizuka. Y. Inhibitory 1987.action of chlorpromazine, dibucaine, and other phospholipid-interacting 45. Hannun. Y. A., and Bell, R. M. Aminoacridines, potent inhibitors of proteindrugs on calcium-activated, phospholipid-dependent kinase. J. Biol. C'hem.. kinase C. J. Biol. Chem.. 263: 5124-5131. 1988.255:8378-8380, 1980. 46. O'Brian. C. A.. Liskamp, R. M.. Solomon. D. H.. and Weinstein. I. B.

41. Lee, M-H.. and Bell. R. M. The lipid binding, regulatory domain of protein Inhibition of protein kinase C by tamoxifen. Cancer Res.. 45: 2462-2465.kinase C, J. Biol. Chem.. 261: 14867-14870. 1986. 1985.

42. Shoji. M.. Vogler. W. R., and Kuo, J. F. Inhibition of phospholipid/Ca!*- 47. Bodden. W. L., Palayoor, S. T.. and Hait. W. N. Selective antimitochondrialdependent protein kinase and phosphorylation of leukemic cell proteins by agents inhibit calmodulin. Biochem. Biophys. Res. Commun.. 135:574-582,CP-46,665-1. a novel antineoplastic lipoidal amine. Biochem. Biophys. Res. 1986.Commun., 127: 590-595. 1985. 48. Lam. H-Y. P. Tamoxifen is a calmodulin antagonist in the activation of a

43. Hannun. Y. A.. Loomis, C. R., Merrill. A. H.. Jr.. and Bell. R. M. Sphin- cAMP phosphodiesterase. Biochem. Biophys. Res. Commun.. 118: 27-32,gosine inhibition of protein kinase C activity and of phorbol dibutyrate 1984.

685

Research. on January 24, 2021. © 1990 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 10: Inhibition of Rodent Protein Kinase C by the Anticarcinoma ... · (CANCER RESEARCH 50. 677-685. Februar} l, 1990) Inhibition of Rodent Protein Kinase C by the Anticarcinoma Agent

1990;50:677-685. Cancer Res   Susan A. Rotenberg, Stephen Smiley, Marius Ueffing, et al.   Agent DequaliniumInhibition of Rodent Protein Kinase C by the Anticarcinoma

  Updated version

  http://cancerres.aacrjournals.org/content/50/3/677

Access the most recent version of this article at:

   

   

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected] at

To order reprints of this article or to subscribe to the journal, contact the AACR Publications

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://cancerres.aacrjournals.org/content/50/3/677To request permission to re-use all or part of this article, use this link

Research. on January 24, 2021. © 1990 American Association for Cancercancerres.aacrjournals.org Downloaded from