Purification of human liver cytochrome P-450 and comparison to the enzyme isolated from rat liver

Purification of human liver cytochrome P-450 and comparison to the enzyme isolated from rat liver

ARCHIVES OF .BIOCHEMISTRY Vol. 199, No. 1, January, Purification PHILIP AND BIOPHYSICS pp. 206-219, 1980 of Human Liver Cytochrome P-450 and Compar...

1MB Sizes 0 Downloads 67 Views

ARCHIVES OF .BIOCHEMISTRY Vol. 199, No. 1, January,

Purification PHILIP

AND BIOPHYSICS pp. 206-219, 1980

of Human Liver Cytochrome P-450 and Comparison Enzyme Isolated from Rat Liver1 WANG, Department Vanderbilt

PATRICIA

S. MASON,

AND

F. PETER

to the

GUENGERICH

of Biochemistry and Center in Environmental Toxicology, School of Medicine, Nashville, Tennessee 37232

University

Received

July

9, 1979; revised

August

22, 1979

Human liver cytochrome P-450 was isolated from autopsy samples using cholate extraction and chromatography on n-octylamino-Sepharose 4B, hydroxylapatite, and DEAE-cellulose gels. Purified preparations contained as much as 14 nmol cytochrome P-450 rng-’ protein, were free of other hemoproteins, and were active in the mixedfunction oxidation of d-benzphetamine and 7-ethoxycoumarin when coupled with either rat or human liver NADPH-cytochrome P-450 reductase. Some of the preparations were apparently homogeneous as judged by sodium dodecyl sulfate-polyacrylamide gel electrophoresis; apparent subunit M,s estimated for several preparations were 53,000 or 55,500. The amino acid composition of one preparation was determined and found to resemble those of rat liver cytochromes P-450, although some variations were noted. Rabbit antibodies raised to phenobarbital-treated rat liver cytochrome P-450 were more effective in inhibiting d-benzphetamine N-demethylase activity in human liver microsomes than were antibodies raised to 3-methylcholanthrene-treated rat liver cytochrome P-450. These antibodies also inhibited benzo(a)pyrene hydroxylation in human liver microsomes, although the inhibition patterns did not follow a general pattern as in the case of benzphetamine demethylase activity. Microsomes prepared from three different human liver samples were more effective in eliciting complement fixation with antibodies raised to phenobarbitalthan to 3-methylcholanthrene-treated rat liver cytochrome P-450. Complement fixation in such systems appears to result from similarity of certain rat and human liver cytochrome P-450 antigenic determinants, as fixation could be inhibited by removal of cytochrome P-450-directed antibodies from the total immunoglobulin population and purified human cytochrome P-450 was more effective (on a protein basis) than liver microsomes in producing fixation. Human liver microsomes prepared from five different individuals all produced 290% complement fixation, but variations were observed in the fixation curves plotted either Ljers*bs microsomal protein or versus spectrally detectable microsomal cytochrome P-450. These results indicate that human liver microsomal cytochromes P-450 can be isolated using modifications of techniques developed for laboratory animals and that human and rat liver cytochromes P-450 share certain features of structural, functional, and immunological similarity. The available data suggest the existence of multiple forms of human liver microsomal cytochrome P-450, but possible artifacts associated with the use of autopsy samples suggest caution in advancing such a conclusion.

function oxidase system that functions in the bioactivation and detoxification of a wide variety of potentially toxic xenobiotics

P-4502 plays an important role as the terminal oxidase of the microsomal mixedI This research was supported by United States Public Health Service Grants ES 01590 and ES 00267 and Contract NO 1 CP 85672 (National Cancer Institute). F.P.G. is the recipient of a United States Public Health Service Research Career Development Award (ES 00041). 2 Abbreviations used: P-450, liver microsomal cytochrome P-450; SDS, sodium dodecyl sulfate; C’, com0003.9861/80/010206-14$02.00/O Copyright All rights

0 1980 by Academic Press, of reproduction in any form

plement; PB, phenobarbital; 3MC, 3-methylcholanthrene; di-12 GPC, L-cY-dilauroylglyceryl3-phosphorylcholine; IgG, immunoglobulin G; PB-rat IgG, IgG raised to P-450 isolated from PB-treated rats; 3MCIgG, IgG raised to P-450 isolated from 3MCtreated rats. 206

Inc. reserved.

COMPARISON

OF

HUMAN

AND

(l-4). A large body of evidence now indicates that the enzyme exists in multiple forms in several tissues of different experimental animals (1, 5- 10). P-450s have been purified to apparent homogeneity from rabbit liver (1, ll-14), rat liver (5, 7, 10, 11, 15, 16), mouse liver (8), and rabbit lung (17, 18). At this time, much less is known about every aspect of P-450 in humans than in laboratory animals. Most studies with human P-450 have focused upon spectra and activity of microsomal preparations (19-26). Kaschnitz and Coon solubilized human liver microsomes and established the roles of P450, NADPH-cytochrome P-450 reductase, and phospholipid fractions in the reconstitution of the mixed-function oxidase system (27). More recently, Kitada and Kamataki have purified fetal human liver microsomal P-450 to a specific content of as high as 6.7 nmol mg-’ protein and reported some of the spectral properties; these preparations also exhibit activity toward aniline and N-ethylmorphine in the presence of rat liver NADPH-cytochrome P-450 reductase and phosphatidylcholine (28). Antibodies have been raised to rabbit and rat liver P-450s and used to establish the multiplicity of P-450 (1, 5-7, 16, 29-32), to quantitate individual forms of P-450 (32), to explore the topical arrangement of P-450 in microsomal membranes (30, 33), to establish the role of P-450 in microsomal reactions (34, 35), to compare the P-450s of different tissues and subcellular organelles (35-38), to determine that different P-450s catalyze different reactions involving a single substrate (33, 39), to localize P-450s in tissues (40), and to examine the biosynthesis of P-450 (41, 42).” Such immunological studies have been important in aiding the understanding of P-450 and utilization of such techniques could be quite useful in studying various aspects of P-450 in human populations. This project was undertaken in order to better describe P-450 in humans by purification and using antibodies raised to rat liver enzymes. Methods are presented for the reproducible isolation of human liver P-450 ” Ohlsson, R. I., Lane, submitted for publication.

C., and Guengerich,

F. P..

RAT

CYTOCHROMES

P-450

207

in high purity and some chemical, physical, and immunological comparisons to the rat liver enzymes are made. EXPERIMENTAL

PROCEDURES

Preparation of human liver microsomes and enzymes. Human liver autopsy samples were obtained 2-8 h after death from the Department of Pathology, Vanderbilt University. Seven preparations were used in this work: patient 2, 54-year-old male, death due to cardiac arrest; patient 3,70-year-old male, death due to myocardial infarction; patient 4, S-day-old female, death due to cardiac arrest; patient 5. 69.year-old female, death due to primary breast and ovarian carcinoma; patient 6, 32.year-old female, death due to gunshot wound; patient 8, Yl-year-old female, death due to cerebral edema; and patient 9, 14-year-old male, death due to cardiac arrest. All liver samples were sliced into I-cm sections, washed with a cold solution of 1.15% (w/v) KC1 containing 0.1 mM phenylmethylsulfonyl fluoride and 0.1 mM dithiothreitol, blotted free of excess buffer, and stored at -20°C. Microsomes were prepared as described elsewhere (36). Specific contents of P-450 for the various microsomal preparations were, respectively (in nmol P-450 rng-’ protein): patient 2, 0.069; patient 3, 0.13; patient 4. 0.36; patient 5, 0.067; patient 6, 0.18. patient 8, 0.35; and patient 9, 0.17. All purification steps were carried out at 0-4°C‘ and used potassium phosphate buffers. Microsomes were suspended at a concentration of 2 mg protein ml- ’ in 0.1 M phosphate buffer (pH 7.25) containing 20%’ (v/v) glycerol, 1 mM EDTA, 20 pM butylated hydroxytoluene, 0.1 mM phenylmethylsulfonyl fluoride, and 0.1 mM dithiothreitol. A 2Oyc (w/v) solution of recrystallized sodium cholate (7) was added to a final concentration of 0.6% (w/v) over 30 min. and the microsomal suspension was centrifuged for 60 min at 105,OOOg. The resulting pellets were pooled and suspended in the original buffer (one-third the original volume), and cholate was added to a final roncentration of 1.54 (w/v) as before. After continued stirring and centrifugation as before, the pellets were pooled and suspended in the original buffer (onesixth original volume). Cholate was added to 0.5% (w/v) and Lubrol PX (Sigma Chemical Co., St. Louis, MO.) was added to 1% (w/v). After stirring for 30 min, the solution was centrifuged as before; an additional 14% of the P-450 was recovered in the supernatant. The three supernatant fractions were assayed for protein and P-450; the second fraction (1.5% cholate) was applied to a 2.5 x 40-cm octylamino-Sepharose 4B column which was washed and eluted as described elsewhere (7, 11, 17). The fractions eluted with the buffer containing 0.33% cholate and 0.06% Emulgen 913 (7, 11. 17) were diluted threefold with a 20% solution of glycerol and applied to a 1 x lo-cm hydroxylapatite (43) column.

208

WANG,

MASON,

AND

The hydroxylapatite column was eluted with a stepwise gradient of 20, 40, 80, 150, 300, and 500 mM potassium phosphate buffers (pH 7.25; 50 ml each buffer); all buffers contained 20% glycerol, 0.1 mM EDTA, and 0.2% (w/v) Emulgen 913. The 300 mM fractions were consistently highest in purity and were used for all of the experiments described here. Excess detergent was removed with Bio-Beads SM-2 (11) unless otherwise noted; less than 20 /*g of Emulgen 913 remained per nanomole P-450. In some cases the fractions eluted from hydroxylapatite columns with 150 or 300 mM phosphate could be further purified by subsequent dialysis against 5 mM potassium phosphate buffer (pH 7.25) containing 20% glycerol, 0.1 mM EDTA, and 0.2% Emulgen 913 and passage through DEAE-Sephacel (5 ml column volume, Pharmacia Fine Chemicals, Uppsala) equilibrated with the dialysis buffer-the yield for this step was routinely about 50%. Human liver NADPH-cytochrome P-450 reductase, assayed as NADPH-cytochrome c reductase, was eluted from the octylamino-Sepharose 4B column after P-450 using a buffer containing 0.35% cholate and 0.15% deoxycholate (44). The preparation was dialyzed and chromatographed on DEAE-cellulose (45) and 2’,5’-ADP-agarose (46) gels essentially as described elsewhere. Rat liver enzymes and antibodies. Rat liver P450s (“B” fractions) were purified and used to raise antibodies in female rabbits as previously described (7). IgG fractions were used in all experiments; preparation was as described elsewhere (7. 30, 36) except that antisera were heated for 20 min at 56°C and then centrifuged for 10 min at 104g prior to ammonium sulfate fractionation in order to inhibit C’. Specific contents (nmol P-450 mg -’ protein) were 2.0 and 1.4 for PB-treated and 3MCtreated rat livei microsomes, respectively. Assays. Protein concentrations were estimated according to Lowry et al. (47). In the case of purified human P-450 preparations. samples were dialyzed versus H,O, concentrated to dryness under N,, and washed twice with CH,OH to remove detergent prior to analysis. Rat P-450 and purified human P-450s were quantitated according to Omura and Sato (48). The extinction coefficient used was Aerao-r!lo = 91 mM-l cm-’ (48); subsequent determinations with P-450 purified from patient 6 gave At = 90 mM-’ cm-‘, based upon quantitation of the heme using the pyridine hemochrome assay (Atai7m375 = 32.4 mM-’ cm-’ for reduced versus oxidized pyridine hemochrome) (48, 49). P-450 was assayed in human liver microsomes according to Matsubara et al. (50) because of hemoglobin contamination. All spectral measurements were made at 20°C using a Cary 219 spectrophotometer in the automatic baseline correction mode. NADPH-cytochrome c reductase activity was assayed (at 30°C) as described elsewhere (45).

GUENGERICH Benzphetamine demethylase (51), benzo(a)pyrene hydroxylase (52), 7-ethoxycoumarin 0-deethylase (7), and epoxide (7,8-styrene oxide) hydratase (53) activities were assayed as described. Emulgen 913 was estimated as described by Garewal (54). C’$xatcntion techniques. The basic procedure was used as described by Levine and his associates (55, 56). Guinea pig C’ was purchased from Cappel Laboratories, Cochranville, Pennsylvania and sheep erythrocytes were obtained fresh weekly from Grand Island Biological Company Diagnostics, Madison, Wisconsin. IgGs and test antigens were diluted in 10 mM Tris-HCl buffer (pH 7.4) containing 0.14 M NaCl, 0.5 mM MgCl,, 0.15 mM CaCl,, and 1 mg ml-’ bovine serum albumin, and the appropriate volumes of each were mixed on ice. C’ (0.2 ml of a ca. 11800 dilution) was added along with sufficient diluent to bring the total volume in each tube to 0.8 ml. After 16 h at 4”C, 0.2 ml of diluent was added (to each tube) containing enough washed erythrocytes to give a final A,,, reading of ca. 1.0. Tubes were incubated for 30 min at 37°C with gentle shaking, chilled on ice, and centrifuged at IOOOg for 5 min. The supernatants were transferred to clean tubes and absorbances were measured at 413 nm. Percentage C’ fixation (anticomplementarity) was calculated using minus antigen blanks for complete lysis and minus C’ blanks for complete inhibition of lysis: minus minus

antigen antigen

A,,:, A,,,

~ observed A,,, - minus C’ A,,:, = percentage

Individual curves presented from experiments carried out single sets of reagents.

C’ fixation.

in each figure on single days

were with

RESULTS

Stability of Huma,n Enzymes

Liver Microsomal

The amounts of liver needed for the purification of substantial amounts of human enzymes required that autopsy material be used, as samples from different patients were not pooled. Preliminary studies were carried out with portions of the liver of a single patient (No. 9) to determine the stability of the enzymes under consideration (Fig. 1). Epoxide hydratase activity was rather stable, even when liver was kept at 23°C (Fig. 1A); 7,8styrene oxide hydratase activity had a t1,2 of about 43 h. NADPH-cytochrome c reductase and P-450 were less stable, with respective t ,,2 values of 14 and 8 h, respec-

COMPARISON

OF HUMAN

AND

RAT

CYTOCHROMES

P-450

209

FIG. 1. Stability of human liver microsomal enzymes under varying conditions. An autopsy sample from patient 9, obtained 3.5 h after death, was divided into several portions (lobes were distributed randomly) and microsomes were prepared. Assays were carried out as described under Experimental Procedures, except in the case of benzphetamine demethylase activity in part A, where a calorimetric assay was used as previously described (11) with minus benzphetamine blank corrections; results are expressed relative to activities determined immediately after preparation of microsomes from freshly procured liver-O.17 nmol P-450 rng-’ protein, 150 t 5 nmol cytochrome c reduced mini rng-’ protein, 19.7 -+ 0.3 nmol 7,8-styrene oxide hydrated min’ mg-’ protein, 0.36 2 0.05 nmol %hydroxybenzo(a)pyrene formed min’ rng-’ protein, and 0.29 t 0.03 nmol HCHO formed (from benzphetamine) min’ rng-’ protein. When SD is indicated by bars, triplicate experiments were carried out. (A) Portions of intact liver were maintained at 23°C; microsomes were prepared at the indicated time points and assays were carried out immediately. (B) Portions of intact liver were maintained at -20°C; microsomes were prepared at the indicated time points and assays were carried out immediately. (C) Microsomes were prepared (at 3.5 h after death of patient), frozen at -2o”C, and assayed at the indicated time points.

tively. Benzo( a)pyrene hydrodroxylase activity was very unstable, having a t1,2 of 1.5 h for the rapid phase of inactivation. However, d-benzphetamine N-demethylase activity was more stable under these conditions (t,,* ea. 30 h). Parallel results were obtained when liver was stored at -20°C (Fig. la), although losses of activity were significantly decreased and were apparently abolished in the cases of epoxide hydratase and benzphetamine demethylase. The stability of the various enzymes was also examined in microsomes (maintained at -20°C in 10 mM Tris-acetate buffer (pH 7.4) containing

1 InM EDTA and 20% glycerol) as a function of time after preparation (Fig. 1C). Benzo(a)pyrene hydroxylase activity was actually less stable than in intact liver. Purification

of Human

Liver P-450

The procedures chosen were based upon previous success in the purification of P-450s from rabbit liver (12), rat liver (7, II), and rabbit lung (17). The first attempt resulted in extensive purification of P-450 from patient 3; the nominal specific content was 9.4 nmol P-450 mg-’ protein and the overall yield was 0.7%.

210

WANG,

MASON,

AND TABLE

GUENGERICH I

PURIFICATION OF HUMAN LIVER MICROSOMALP-450"

(mg)

P-450 (nmol)

Apparent yield (%)

4200 3200 785 41 2.6

767 278 217 121 35

(100) 36 28 16 4.5

Protein Fraction Microsomes 0.6% Cholate extract 1.5% Cholate extract Octylamino-Sepharose Hydroxylapatite

4B

Specific content (nmol mg-‘) 0.18 0.017 0.28 2.97 13.1

a The procedure was carried out using a portion of the liver sample obtained cholate extract was used for subsequent steps. Data are shown only for the 300 hydroxylapatite column.

Methods were refined as noted under Experimental Procedures to purify P-450 from patient 6 to apparent homogeneity in 4.5% yield (Table I). Three separate purifications with portions of the same liver (patient 6) yielded P-450 ranging in nominal specific content from 9.5-13.8 nmol P-450 mg-’ protein in 1.6-4.5% overall apparent yield; P-450 was also purified from patient 8 to a specific content of 14.1 nmol P-450 mg-’ (0.5% yield). The P-450s obtained from patients 6 and 8 migrated as single bands upon SDSpolyacrylamide gel electrophoresis as described by Laemmli (57) or as subsequently modified (7). The apparent M,.s were 53,000 d for those preparations (Fig. 2); the major

I 2

3

4567

Fold purification

from

patient

mM fraction

(1) 0.5 1.5 16.2 71.6 6. Only recovered

the 1.5% from the

band in the preparation derived from patient 3 had an apparent M, of 55,500. For comparison, the following apparent M,s were estimated for rat and rabbit liver P-450s (7, 11) in these electrophoretic experiments: PB-treated rat P-450, 53,000; 3MC-treated rat P-450, 55,000; PB-treated rabbit LM-2, 51,500; and P-naphthoflavonetreated rabbit LM-4, 53,500. Spectra indicated the absence of other hemoproteins (Fig. 3). The oxidized form of the isolated P-450 existed as a low-spin hemoprotein with absorption maxima at 416, 528, and 570 nm. The A39,,LA4,6 ratio was 0.55, as also found with several isolated rat and rabbit liver P-450 preparations (11). Other maxima were observed

8

9

IO

FIG. 2. SDS-polyacrylamide gel electrophoresis of human liver P-450 preparations. Samples were electrophoresed according to Laemmli (57) except that the concentrations of the components of the cathode buffer were doubled (7). The anode was at the bottom of each of the three gels; gels were stained and destained according to Fairbanks et al. (58). Samples were as follows: 1, 7, and 9, 3.75 pg each of standard bovine serum albumin (accepted M, 68,000), bovine liver catalase (M, 58,000), Escherichia coli L-glutamate dehydrogenase (M, 53,000), hen egg ovalbumin (M, 43,000), and rabbit muscle aldolase (M, 40,000); 2, octylamino-Sepharose 4B fraction from patient 3, 10 pg; 3, hydroxylapatite-DEAE-cellulose fraction from patient 3, 3 pg; 4 -6, hydroxylapatite fraction from patient 6, 5, 10, and 13 Fg, respectively; 8, hydroxylapatite fraction from patient 8, 4.5 pg; and 10, hydroxylapatite fraction from patient 6, 5 Fg.

COMPARISON

OF

HUMAN

AND

RAT

CYTOCHROMES

P-450

FIG. 3. (A) Difference spectrum of purified human liver P-450. A 0.25 pM solution of a final human liver P-450 preparation (patient 6) in 5 mM potassium phosphate buffer (pH 7.7) containing 20% glycerol. 0.1 mM EDTA, and 0.2% Emulgen 913 was divided into two 0.2ml cuvettes which were equilibrated with CO; a baseline (- - -) was established and Na,S,O? was added to the sample cuvette (---). (R) Absolute spectra of purified human liver P-450. The sample cuvette contained 0.25 @M purified P-450 (patient 6) in the same buffer as used in part A. The oxidized (---1. Na,S,O,-reduced (- - -), and reduced-CO (-.-) spectra are shown.

at 418 and 560 nm in the reduced state cated that the estimates of protein conand 450 (450.0 -+ 0.2) and 552 nm for the centration using the method of Lowry reduced-CO complex. Of all the microsomal et al. (47) were reasonably accurate (i.e., and purified preparations examined, none within 5%). had reduced-CO maxima that were out of the range of 449-451 nm. PuriIcation of Human Liver The amino acid composition of a human NADPH-cytochrome P-450 Reductase liver P-450 preparation showed some similarity to the major P-450s isolated from The reductase was purified from patient PB- and SMC-treated rats (Table II). The 6 to a specific activity of 53 pm01 most striking difference was in the com- cytochrome c reduced min-’ mg-’ protein position of basic residues; i.e., the human using techniques developed for the purificapreparation contained very little histidine tion of the enzyme from other species and much more lysine than either of the (44-46). The enzyme was not homogeneous two rat P-450s. Difference indices were as judged by SDS-polyacrylamide gel calculated according to Metzger et al. (60): electrophoresis (58), but >80% of the the two rat liver P-450s had an index of 3.1; Coomassie blue stain was associated with a the human liver P-450 had an index of 6.3 single band of apparent M, 70,000 (not when compared to the PB-treated rat P-450 shown). This M, was slightly less than and 5.7 when compared to the 3MC- that observed for the rat liver enzyme treated rat P-450. These values indicate under identical conditions (M, 74,000) (46). that the two rat P-450s are more closely The preparation was active in supporting related to each other than to the human P-450-associated hydroxylation activities enzyme. The amino acid analysis data indi(vide infra).

212

WANG, TABLE

MASON,

AND

Reconstitution

II

COMPARISON OF AMINO ACID COMPOSITION OF HUMAN ANDRAT LIVER CYTOCHROMESP-450" Number of residues subunit P-450

Amino

acid

Human

Lysine Histidine Arginine Asx Threonine Serine Glx Proline Glycine Alanine Valine Methionine Isoleucine Leucine Tyrosine Phenylalanine Half-cystine Tryptophan Total

46 1 19 47 25 36 45 28 45 27 22 15 28 53 2: 5b 4 474

GUENGERICH

Phenobarbitaltreated rat

per

S-Methylcholanthrenetreated rat

27 13 25 42 29 30 52 27 33 29 27 11 27 59 14 32 7 2 486

31 13 25 43 29 38 43 37 32 26 28 10 24 54 14 28 6 6 487

of Human Liver Microsomal Mixed-Function Oxidase Activity

The P-450 preparation derived from patient 6 was active toward d-benzphetamine and ‘i-ethoxycoumarin when coupled with either rat or human liver NADPH-cytochrome P-450 recluctase (Table III). The dealkylation rates were higher than those observed with the microsomal preparation when expressed on the basis of P-450. However, benzo(a)pyrene hydroxylase activity was not detected with this same P-450. However, benzo(a)pyrene hydroxylase activity was not detected with this same P-450 preparation. Other experiments inclicatecl that the rates of benzphetamine and 7-ethoxycoumarin metabolism were lowered to less than 11 and 40%, respectively, when di-12 GPC was omitted from the respective systems, and that the addition of Emulgen 913 to 10 pg ml-’ did not affect the rates of metabolism. Effect of Antibodies upon Activity Human Liver P-450

0 Data for the rat liver proteins were reported elsewhere (7). The preparation derived from patient 6 (Table I) was hydrolyzed and analyses were done in triplicate essentially as described elsewhere (7). b Estimated by titration with 4,4’-pyridine disulfide (59). TABLE

of

PB rat-IgG was found to inhibit benzphetamine clemethylase activity in microsomes prepared from three different individuals (Fig. 4); the extent of maximum inhibition ranged from 25-65% and was statistically significant when compared to the effect of preimmune IgG. The effect of 3MC rat-IgG was rather small if significant at all. III

METABOLISMOF SUBSTRATES BYHUMAN LIVER MICROSOMESAND PURIFIED P-450" Turnover

number Purified

Substrate d-Benzphetamine ‘I-Ethoxycoumarin

Microsomes (min-‘) 2.7 ” 0.20 0.0137 5 0.0006

Plus rat reductase (min-‘) 27.0 k 10.9 0.023 2 0.004

human

P-450 Plus human reductase (min-I) 21.9 0.021

t 8.0 + 0.011

a Assays measuring benzphetamine (51) and 7-ethoxycoumarin (7) activities were carried out in triplicate as described. Incubations contained 0.5 mg microsomal protein or reconstituted systems composed of 5 pmol purified human P-450 (patient 6) plus an equimolar amount of rat (7,46) or human liver NADPH-cytochrome P-450 reductase and 50 PM di-12 GPC. Turnover numbers (means 2 SD) are expressed as nmol product formed min-’ nmol-’ P-450.

COMPARISON

OF HUMAN

AND

RAT

CYTOCHROMES

213

P-450

i 0 0

.i

.----L--L

2

6

mg

0 I --~ 0 C

3

I 1

~~~-

L--

IgG

--e---F

nmol

-’

,-1

-‘,----3,.--

--yf-L-

*

5

mg

IgG

nmol-’

20

P-450

i 20

P-450

FIG. 4. Effect of antibodies raised to rat liver P-450s on d-benzphetamine N-demethylase activity of human liver microsomes. Incubations were carried out essentially as described elsewhere (36) using an amount of microsomal protein equivalent to 100 pmol of P-450 in each assay. Either preimmune IgG, IgG raised to PB-treated rat P-450, or IgG raised to 3MC-treated rat P-450 was present at each indicated concentration. All assays were carried out in triplicate and results are expressed as means 2 SD. The basal levels of HCHO formation were 0.156 C 0.016, 0.60 k 0.06, and 1.67 + 0.12 nmol min-’ nmol-’ P-450 for patients 4 (A), 8 (B), and 9 (Cl, respectively.

214

WANG,

MASON,

AND

The inhibition of benzo(a)pyrene hydroxylation was also examined (Fig. 5). 3MC-rat IgG produced varying levels of inhibition in three different patients. PB rat IgG had no effect in one case (Fig. 4B), inhibited metabolism by 70% in another case (Fig. 4C), and gave some stimulation in the third ease (Fig. 4A). C’ Fixation

Experiments

Preliminary experiments indicated that none of the human liver microsomal preparations yielded immunodiffusion precipitin lines with either PB rat-IgG or 3MC rat IgG when examined under conditions used for rat P-450s (6, ‘7, 36). Complexation of human liver P-450 with PB rat-IgG was

GUENGERICH

detected using C’ fixation techniques, as shown in Fig. 6. Considerably less fixation, if any, was detected using 3MC ratIgG (Fig. 7). The C’ fixation activity of the PB rat-IgG was nearly abolished by precipitation of the rat P-450-directed antibodies from the IgG with PB-treated rat liver microsomes as shown in Fig. 8, suggesting that similar immunological determinants are present in rat and human liver microsomal P-450s. The P-450-directed antibodies in the PB rat-IgG preparation could also be partially removed by centrifugation of mixtures of PB rat-IgG and highly purified rat P-450; after such treatment, PB-rat IgG was less effective than untreated PB-rat IgG in C’ fixation using either PB-treated rat microsomes or

i

FIG. 5. Effect of antibodies raised to rat liver P-450s on benzo(a)pyrene hydroxylase activity of human liver microsomes. Incubations were carried out essentially as described elsewhere (36) using an amount of microsomal protein equivalent to 100 pmol of P-450 in each assay. All assays were carried out in triplicate and are expressed as means 2 SD. Either preimmune IgG, IgG raised to PB-treated rat P-450, or IgG raised to 3MC-treated rat P-450 was present at each indicated concentration. The basal levels of 3-hydroxybenzo(a)pyrene formation were 0.201 2 0.085, 1.79 _f 0.23, and 2.65 2 0.12 nmol min-’ nmol-’ P-450 for patients 4 (A), 8 (B), and 9 (C), respectively.

COMPARISON

OF HUMAN

AND

RAT

CYTOCHROMES

215

P-450

FIG. 6. C’ fixation by PB rat-IgG and PB-treated rat liver or human liver microsomes. Varying levels of PB rat-IgG were mixed with either 0.2 kg of PB-treated rat liver microsomes (A) or 2 pg of liver microsomes derived from each of the following human samples: patient 2 CO), patient 3 (W), or patient 6 (A). C’ fixation assays were carried out as described under Experimental Procedures.

human microsomes (Fig. 8). The lack of complete inhibition of C’ fixation was due to incomplete precipitation of P-450lIgG complexes at these concentrations, as 30% apparent C’ fixation was found for incubations devoid of microsomes. No greater inhibition of C’ fixation was found with a number of higher or lower P-450:IgG ratios for removal of P-450-directed antibodies. Treatment of the PB rat-IgGIP-450 supernatant with varying levels of a second antibody (swine IgG raised to rabbit IgG) did not specifically precipitate PB rat-IgG/ P-450 complexes. Highly purified human P-450 was more effective in C’ fixation on a protein basis

than were human microsomes (Fig. 9>, further implicating P-450 in the C’ fixation process. The difference in the curves is not in direct proportion to the purification factor; however, other forms of P-450 may be present in the microsomes that are more similar immunologically. Differences Different

in C’ Fixation Humans

by P-450s of

Varying amounts of the different human microsomal preparations were incubated with a fixed amount of PB-rat IgG in C’ fixation studies, as curves from such experiments are more sensitive in detecting

FIG. 7. C’ fixation by 3MC rat-IgG and 3MC-treated rat liver or human liver microsomes. Varying levels of 3MC rat-IgG were mixed with either 0.2 pg of 3MC-treated rat liver microsomes (A) or 2 kg of liver microsomes derived from each of the following human samples: patient 2 (O), patient 3 (m), or patient 6 (A). C’ fixation assays were carried out as described under Experimental Procedures.

216

WANG, MASON, AND GUENGERICH

FIG. 8. Inhibition of PB-treated rat and human microsomal C’ fixation by removal of P-450. directed antibodies. C’ fixation was carried out as described under Fig. 6 with PB-treated rat liver microsomes (A A) and human liver microsomes derived from patient 6 (A A). PB-rat IgG was mixed with either 2 nmol purified PB-treated rat liver P-450 or 1 mg PB-treated rat liver microsomes/mg PB rat-IgG, allowed to stand 1 h at 23°C and 24 h at 4”C, and centrifuged at ZO,OOOgfor 30 min. Aliquots of the supernatants were used in parallel experiments without correction for the amounts of IgG removed: 0.2 pg PB-treated rat microsomes plus PB-rat IgG treated with purified PB rat P-450 (A - - - A); 2 *g human microsomes (patient 6) plus PB-rat IgG treated with purified PB rat P-450 (A - - - a), 0.2 gg PB-treated rat microsomes plus PB-rat IgG treated with PB-treated rat microsomes (A -.A); and 2 pg human microsomes (patient 6) plus PB-rat IgG treated with PB-treated rat microsomes (a -‘A).

differences in proteins than the type used in Figs. 6 and 7 (55, 56). When the results were plotted as a function of protein used, the order of C’ fixation (most efficient to least) was patient 2 > patient 4 > patient 6 = patient 3 s patient 5 (Fig. 10A). The amount of protein required for 50% C’ fixation differed 12-fold between patients 2 and 5. Plotting the results as a function of spectrally detectable P-450 also showed a 1Zfold difference in the amount I

1

FIG. 9. Comparison of C’ fixation by human liver microsomes and highly purified human liver microsomal P-450. C’ fixation assays were carried out using 5 pg of PB rat-IgG and varying levels of microsomes derived from patient 6 (A) or P-450 (hydroxylapatite preparation, Table I in duplicate) derived from patient 6 (0).

of P-450 required for 50% C’ fixation (Fig. 10B); the order of C’ fixation was patient 2 + patient 3 > patient 6 = patient 4 > patient 5. In all cases, 290% C’ fixation was achieved at some concentration of microsomes. DISCUSSION

With some modification, techniques developed for the purification of P-450 and NADPH-cytochrome P-450 reductase from experimental animals have been used to isolate the enzymes from human liver in high states of purity. Although some of the final preparations were apparently homogeneous as judged by SDS-polyacrylamide gel electrophoresis, specific contents of P-450 mg-’ protein were somewhat less than expected on the basis of subunit M, (i.e., 17-19 nmol). The protein estimation was not in error, as judged by quantitative amino acid analysis. Hydrodynamic studies indicate that M, estimates for rat liver P-450s obtained with SDS-polyacrylamide gel electrophoresis are correct (7),4 and we assume that this is the case d Guengerich, F. P., and Holladay, mitted for publication.

I>. A., sub-

COMPARISONOFHUMANANDRATCYTOCHROMESP-450

B

HUMeN

CYTOCHROME

P-450,

217

pmoles

FIG. 10. C’ fixation as a function of human microsomal protein or P-450. C’ fixation assays were carried out as described under Experimental Procedures with 5 pg of PB rat-IgG and varying amounts of human liver microsomes derived from patient 2 (O), patient 3 (m), patient 4 (O), patient 5 (O), and patient 6 (a). (A) Data are plotted versus concentration of human liver microsomal protein. (B) Data are plotted versus concentration of human liver microsomal protein. (B) Data are plotted versus concentration of spectrally detectable human liver P-450.

for human liver P-450 as well. The difference was not due to the presence of cytochrome P-420 (i.e., a form(s) of denatured P-450) (Fig. 3). The discrepancy was also probably not due to error in the extinction coefficient; ~~~~~~~~~= 91 MM~’ cm-’ has been confirmed with a variety of rat and rabbit preparations (5, 48) and a value of 90 mM-' cm-’ was obtained for a human preparation. Thus, the discrepancy must arise from the presence of apoenzyme or some other protein not resolved by SDSpolyacrylamide gel electrophoresis. The stability profiles deserve some comment. Benzo(a)pyrene hydroxylase activity is not very stable in human liver, in concurrence with a recent report by Prough et al. (26). This is somewhat surprising since Robinson et al. reported that this activity was stable in mice for 7 h after

death (61). Benzphetamine demethylase activity was much more stable under all conditions examined, as is the case for another microsomal enzyme, epoxide hydratase. The data suggest caution in using the measurement of benzo(a)pyrene hydroxylase activity in autopsy samples as an index of metabolism. The results also suggest considerable variation in stabilities of different human liver P-450s and that minor amounts of certain individual forms or subpopulations of P-450 may be responsible for the bulk of either benzo(a)pyrene hydroxylase or benzphetamine demethylase activity. Although repeated attempts to establish immunochemical similarity of various rat and human P-450s using double-diffusion analysis were negative, similarity was detected using C’ fixation and inhibition of

218

WANG,

MASON,

activity. The general conclusion can be drawn that the P-450s in the human liver samples examined here were immunologically more similar to P-450 isolated from PB-treated than SMC-treated rats. The apparent trend among various patients was that benzphetamine metabolism was more readily inhibited by PB-rat IgG and benzo(a)pyrene metabolism was more readily inhibited by 3MC-rat IgG, which might be expected on the basis of some of the results obtained with rat liver microsomes (31, 33, 36). Other experiments indicate that benzo(a)pyrene hydroxylase activity in human lymphocytes and monocytes is more readily inhibited by 3MC-rat IgG than by PB-rat IgG.” In conclusion, the available data suggest that rat and human P-450s are somewhat similar as judged by certain physical and immunological techniques, although differences definitely exist. The results also suggest that humans contain multiple forms of P-450, as judged by isolation of P-450s with different apparent M,s (Fig. 2), C’ fixation studies (Fig. lo), individual differences in susceptibility to antibodies (Fig. 4), differences in the stabilities of P-450catalyzed activities (Fig. l), and the separation of P-450 into several fractions in the various chromatographic procedures. However, such a conclusion must be considered tentative because of possible artifacts that may be associated with the use of autopsy material.

We thank Drs. Frank Chytil, Robert Briggs, and Lubomir Hnilica for their advice in carrying out the complement fixation work, Dr. John Edland for his help in procuring autopsy samples, and Mr. W. Morgan Crawford, Jr. and Mrs. Margaret B. Mitchell for their excellent technical assistance. REFERENCES J., VERMILION, J. L., VATSIS, K. P., J. S., DEAN, W. L., AND HAUGEN, (1977) in Drug Metabolism Concepts D. M., ed.), Amer. Chem. Sot. SymSeries, No. 44, pp. 46-71, Amer. Sot., Washington, D. C.

s Robie-Suh, K., Robinson, Guengerich, F. P., submitted

GUENGERICH

2. CONNEY, A. H. (1967) Pharmacol. Rev. 19, 317-366. 3. GELBOIN, H. V. (1967) Advan. Cancer Res. 10, 1-81. 4. GILLETTE, J. R. (1966) Advan. Pharmacol. 4, 219-261. 5. GUENCERJCH, F. P. (19’79) Pharmacol. Ther. A 6, 99-121. 6. THOMAS, P. E., Lu, A. Y. H., RYAN, D., WEST, S. B., KAWALEK, J., AND LEVIN, W. (1976) Mol. Pharmacol. 12, 746-758. 7. GUENGERICH, F. P. (1978) J. Bi01. Chem. 253, 7931-7939. 8. HUANG, M-T., WEST, S. B., AND Lu, A. Y. H. (1976) J. Biol. Chem. 251, 4659-4665. 9. ULLRICH, V., AND KREMERS, P. (1977) Arch. Toxicol. 39, 41-50. 10. LEVIN, W. (1977) in Microsomes and Drug Oxidations (Ullrich, V., Roots, I., Hildebrandt, A., Estabrook, R. W., and Conney, A. H., eds.), pp. 735-747, Pergamon Press, New York. 11. GUENGERICH, F. P. (1977) J. Biol. Chem. 3970-3979. 12. IMAI, Y., AND SATO, R. (1974) Biochem. phys. Res. Commun. 60, 8-14. 13. JOHNSON, E. F., AND MULLER-EBERHARD, (1977) Biochem. Biophys. Res. Commun. 652-659. 14. PHILPOT, R. M., AND ARINC, E. (1976) Pharmacol. 12, 483-493.

R., Gelboin, H. V., and for publication.

252,

BioU. 76, Mol.

15. RYAN, D., Lu, A. Y. H., KAWALEK, J., WEST, S. B., AND LEVIN, W. (1975) Biochem. Biophys. Res. Commun. 64, 1134-1141. 16. RYAN, D. E., THOMAS, P. E., AND LEVIN, W. (1977) Mol. Pharmacol. 13, 521-532. 17. GUENGERICH, F. P. (1977) Mol. Pharmacol. 13, 911-923. 18. WOLF, C. R., SZUTOWSKI, AND PHILPOT, R. M. Interac. 21, 29-43.

ACKNOWLEDGMENTS

I. COON, M. FRENCH, D. A. (Jerina, posium Chem.

AND

M. M., (1978)

BALL, L. M., Chem.-Biol.

19. KUNTZMAN, R., MARK, L. C., BRAND, L., JACOBSON, M., LEVIN, W., AND CONNEY, A. H. (1966) J. Phamacol. Ezp. Ther. 152, 151156. 20. ALVARES, A. P., SCHILLING, G., LEVIN, W., KUNTZMAN, R., BRAND, L., AND MARK, L. C. (1969) Clin. Pharmacol. They. 10, 655-659. 21. DARBY, F. J., NEWNES, W., AND PRICE EVANS, D. A. (1970) Biochem. Phawnacol. 19, 15141517. 22. NELSON, E. B., RAJ, P. P., BELFI, K. J., AND MASTERS, B. S. S. (1971) J. Pharmacol. Exp. They. 178, 580-588. 23. SCHOENE, B., FLEISCHMANN, R. A., REMMER, H., AND v. OLDERSHAUSEN, H. F. (1972) Eur. J. Clin. Pharmacol. 4, 65-73. 24. KAPITULNIK, J., POPPERS, P. J., BUENING, M. K.,

COMPARISON

25.

26.

27. 28. 29.

30. 31.

32.

OF HUMAN

AND RAT CYTOCHROMES

FORTNER, J. G., AND CONNEY, A. H. (1977) Clin. Pharmacol. Ther. 22, 475-484. KAPITULNIK, J., POPPERS, P. J., AND CONNEY, A. H. (1977) Cl&. Pharmacol. Thu. 21, 166176. PROUGH, R. A., PATRIZI, V. W., OKITA, R. T., MASTERS, B. S. S., AND JAKOBSSON, S. W. (1979) Cancer Res. 39, 1199-1206. KASCHNITZ, R. M., AND COON, M. J. (1975) Biochem. Pharmacol. 24, 295-297. KITADA, M., AND KAMATAKI, T. (1979) Biochem. Pharmacol. 28, 793-797. THOMAS, P. E., Lu, A. Y. H., RYAN, D., WEST. S. B., KAWALEK, J., AND LEVIN, W. (1976) J. Biol. Chem. 251, 1385-1391. DEAN, W. L., AND COON, M. J. (1977) J. Biol. Chem. 252, 3255-3261. WELTON, A. F., O’NEAL, F. O., CHANEY, L. C., AND AUST, S. D. (1975) J. Biol. Chem. 250, 5631-5639. THOMAS, P. E., KORZENIOWSKI, D., RYAN, D., AND LEVIN, W. (1979) Arch. Biochem. Biophys. 192, 524-532.

33. THOMAS, P. E., Lu, A. Y. H., WEST, S. B., RYAN, D., MIWA, G. T., AND LEVIN, W. (1977) Mol. Pharmacol. 13, 819-831. 34. KAMATAKI, T., BELCHER, D. H., AND NEAL, R. A. (1976) Mol. Pharmacol. 12, 921-932. 35. GCEPU‘GERICH, F. P. Biochern. Pharr)!acol., in press. 36. GUENGERICH, F. P., AND MASON, P. S, (1979) Mol. Pharmacol. 15, 154-164. 37. SAGARA, Y., HARANTO, T., AND OMURA, T. (1978) J. Biochem. (Tokyo) 83, 807-812. 38. THOMAS, P. E., KORZENIOWSKI, D., BRESNICK, E., BORNSTEIN, W. A., KASPER, C. B., FAHL, W. E., JEFCOATE, C. R., AND LEVIN, W. (1979) Arch. Biochem. Biophys. 192, 22-26. 39. KAMINSKY, L. S., FASCO, M. J., AND GUENGERICH, F. P. (1979) Fed. Proc. 38, 732. 40. BARON, J., REDICK, J. A., AND GUENGERICH, F. P. (1978) Life Sci. 23, 2627-2632. 41. CRAFT, J. A., COOPER, M. B., AND RABIN, B. R. (1978) FEBS Lett. 88, 62-66. 42. BHAT, K. S., AND PADMANABAN, G. (1978) FEBS Lett. 89, 337-340.

P-450

219

43. MAZIN, A. L., SULIMOVA, G. E., AND VANYUSHIN, B. F. (1974) Anal. Biochem. 61, 62-71. 44. IMAI, Y. (1976) J. Biochem. (Tokyo) 80,267-276. 45. STROBEL. H. W., AND DIGNAM, J. D. (1978) in Methods Enzymology (Fleischer, S., ed.), Vol. 52, pp. 89-96, Academic Press. New York. 46. GUENGERICH, F. P. (1978) Biochemistry 17, 3633-3639. 47. LOWRY, 0. H., ROSEBROUGH, N. .J. FARR, A. L.. AND RANDALL, R. J. (1951) J. Biol. Chem. 193, 265-275. 48. OMURA, T., AND SATO, R. (1964) J. Biol. Chem. 239, 2370-2378. 49. FALK, J. E. (1964) Porphyrins and Metalloporphyrins, pp. 181-182. American Elsevier, New York. 50. MATSUBARA, T., KOIKE, M., TOUCHI. A., ToCHINO, Y., AND SUGENO. K. (1976) Anal. Biochem. 75, 596-603. 51. GUENGERICH, F. P., BALLOU, D. P., AND COON. M. J. (1975) J. Biol. C’hem. 250, 7405-7414. 52. NEBERT. D. W.. AND GELBOIN, H. V. (1968) J. Biol. Chem. 243, 6242-6249. 53. JERINA, D. M., DANSETTE, P. M., Lu, A. Y. H., AND LEVIN. W. (1977) Mol. Pharmacol. 13, 342-351. 54. GAREWAL, H. S. (1973) Anal. Biochern. 54, 319-324. 55. WASSERMAN, E., AND LEVINE, L. (1961) J. Immunol. 87, 290-295. 56. LEVINE, L., AND VAN VUNAKIS, H. (1967) in Methods in Enzymology (Hirs, C. H. W., ed.), Vol. 11, pp. 928-936, Academic Press, New York. 57. LAEMMLI, U. K. (1970) Nature (London) 227, 680-685. 58. FAIRBANKS, G., STECK, T. L., AND WALLACH, D. F. H. (1971) Biochemistry 10, 2606-2617. 59. GRASSETTI. D. R.. AND MURRAY, J. F., JR. (1967) Arch. Biochem. Biophys. 119, 41-49. 60. METZGER, H., SHAPIRO, M. B., MOSIMANN, J. E., AND VINTON, J. E. (1968) Nature (London) 219, 1166-1168. 61. ROBINSON, J. R., FELTON. J. S., LEVITT, R. C., THORGEIRSSON, S. S., AND NEBERT, D. W. (1975) Mol. Pharmacol. 11, 850-865.