Preparative Biochemistry

ISSN: 0032-7484 (Print) (Online) Journal homepage: http://www.tandfonline.com/loi/lpbb19

Stabilization and Purification of Tyrosine Aminotransferase from Rat Liver James L. Hargrove To cite this article: James L. Hargrove (1990) Stabilization and Purification of Tyrosine Aminotransferase from Rat Liver, Preparative Biochemistry, 20:1, 11-22, DOI: 10.1080/00327489008050174 To link to this article: http://dx.doi.org/10.1080/00327489008050174

Published online: 23 Oct 2006.

Submit your article to this journal

Article views: 9

View related articles

Citing articles: 2 View citing articles

Full Terms & Conditions of access and use can be found at http://www.tandfonline.com/action/journalInformation?journalCode=lpbb20 Download by: [NUS National University of Singapore]

Date: 08 November 2015, At: 20:54

PREPARATIVE BIOCHEMISTRY, 20(1), 11-22 (1990)

Downloaded by [NUS National University of Singapore] at 20:54 08 November 2015

STABILIZATION AND PURIFICATION OF TYROSINE AMINOTRANSFERASE FROM RAT LIVER James L. Hargrove Department of Foods and Nutrition Dawson Hall The University of Georgia Athens, GA 30602 Abbreviations: CM, carboxymethyl; DEAE, diethylaminoethyl; DTT, dithiothreitol; EDTA, Ethylenediamine tetraacetic acid; HEPPS, Hydroxyethylpiperazinepropanesulfonic acid; PLP, pyridoxal 5'-phosphate; SDS, sodium dodecyl sulfate. Purification of unmodified tyrosine aminotransferase from rat liver requires that the activity of cathepsin T be minimized, and that losses of enzyme due to dilution or oxidation be prevented. The enzyme was stabilized by pyridoxal 5'-phosphate, dithiothreitol, and potassium phosphate, but was destabilized by L-tyrosine or L-plutamate. A rapid, efficient method for purification of this enzyme included the following steps: twenty-fold induction with a high-casein diet plus dexamethasone phosphate administered in the drinking water; a heat step (65OC) followed by precipitation from 0.20 M_ sucrose at pH 5.0; and small-scale chromatography on DEAE-cellulose, hydroxyapatite and CM-Sephadex C50 at pH 6 . 0 . These steps yielded more than 10 mg of native enzyme from 35 rats, with a recovery of 68% of the initial activity

.

INTRODUCTION Recent studies suggest that tyrosine aminotransferase (E.C. 2.6.1.5)

contains structural features that are involved in the

rapid rate of degradation which characterizes this enzyme in vivo, and other features which underlie its ability to transaminate tyrosine (1-3). Thus, the enzyme contains PEST elements that are thought to participate in proteolytic degradation of soluble proteins (21, and features that may permit it to be taken up into lysosomes in a serum-dependent manner ( 3 ) .

11 Copyright 0 1990 by Marcel Dekker, Inc.

While it should be

12

HARGROVE

useful

to

characterize

the

features

causing

tyrosine

aminotransferase to interact with these degradative systems, the low abundance and instability of the enzyme in rat 1l.ver impede progress.

Downloaded by [NUS National University of Singapore] at 20:54 08 November 2015

In order to overcome these obstacles, the present work describes conditions permitting induction of the enzyme to high levels in rat liver, and rapid purification in high yield. method

involves

acidification

of

a

sucrose

homogenate

The to

precipitate the enzyme, which is stabilized in the presence of phosphate buffer, pyridoxal 5'-phosphate (PLP), and dithiothreitol (DTT). using

The resulting preparation can then be purified in one day small

columns

of

DEAE-cellulose, hydroxyapatite, and

CM-Sephadex C50 to avoid dilution and loss of activity. MATERIALS AND METHODS Sprague-Dawley rats were bred in the departmental vivarium, where they were given free access to Purina rat chow and water. To survey treatments that provide a high concentration of enzyme, the rats were either fed a high-protein diet (50% of calories as casein; ICN Biomedical Products) for three days, or were fasted overnight before being killed. Injections of hormones and other agents were given between 9 and 10 p.m., and animals were killed between 8 and 9 a.m. the next morning. Hydroxyapatite (Biogel HTP) was obtained from BioRad, and other media for chromatography were from Sigma.

Most chemicals

and reagents were obtained from standard suppliers.

Triamcinolone

acetonide was a gift from Lederle Laboratories (Pearl River, NY). Tyrosine aminotransferase was assayed by the procedure of Granner and Tomkins (41, with one unit of activity equal to formation of one micromole of product per minute at 37'C.

Protein

was measured by a dye binding assay using pre-mixed reagent from Pierce Chemical Company. Purity of the enzyme was assessed by SDS-polyacrylamide gel electrophoresis (5) using minigel apparatus from BioRad. RESULTS AND DISCUSSION Stabilization

of

Tyrosine

Aminotransferase.

Since

tyrosine

aminotransferase gradually loses activity during the time required

PURIFICATION OF TYROSINE AMINOTRANSFERASE

13

TABLE I: Stability of Tyrosine Aminotransferase in Vitro

Downloaded by [NUS National University of Singapore] at 20:54 08 November 2015

37", 4h

Addition

Minus

Plus

Glutamate

Glutamate

None

60", 15 min

84

18

63

106

84

91

a-Ketoglutarate, 2 mM

99

25

93

L-Tyrosine, 1 mM

36

N.D.~

18

Pyridoxal 5'-phosphate, 0.2 mM

E-Hydroxyphenylpyruvate, 1 mM

106

N.D.

N.D.

Dithiothreitol, 2 mM

113

23

110

108

100

N.D.

Potassium phosphate, pH 7 , 25 mM

aN.D., value not determined.

for its purification, means to optimize yield by stabilizing the enzyme were tested. A preparation of enzyme was incubated at 37°C in buffers from pH 6 to 8, in the presence of potassium phosphate, PLP,

dithiothreitol, and a-ketoglutarate.

The enzyme was most

stable in phosphate buffer at pH 6-7, and least stable at pH 8 in synthetic buffers. For the experiments shown in Table I, one hundred milliunits of partially purified tyrosine aminotransferase (30 unitslmg protein) was incubated for 4 h at 37" in 0.25 ml of

14

HARGROVE

0.1 M - Hepes, pH 7.5, with 1 mM EDTA and 0.5 mg of fatty acid-free

bovine serum albumin.

One set of samples included 1 mM glutamate,

which removes PLP from the enzyme.

Other agents were included at

the concentrations shown, and samples were taken for assay of

Downloaded by [NUS National University of Singapore] at 20:54 08 November 2015

enzyme activity before and after the incubation.

In a second

experiment, samples were heated to 60" for 15 min in 0 . 2 5 ml of 25 mM potassium phosphate, pH

7, containing 0 . 5 mg albumin.

Values

are expressed as percentages of the initial activity remaining after the incubations, and are means of duplicate determinations. In the presence of 1 mM L-glutamic acid (which resolves the coenzyme

[unpublished

observation]),

maximum

stabilization

occurred with 0 . 2 mM PLP or 25 mM potassium phosphate. During the heat step, the enzyme was also stablized by 1 mM a-ketoglutarate or 2 mM dithiothreitol.

Combinations of these agents totally

stabilized the enzyme; therefore, most of this work employed potassium phosphate buffer at pH 6 , containing 0 . 1 mM PLP, 2 mM a-ketoglutarate, and 2 mM dithiothreitol. The reducing agent was added to buffers immediately before use. However, the initial extract must be buffered

to pH 8 and heated to inactivate

lysosomal proteases that generate multiple

forms by

limited

proteolysis (6,7), for which reason hydroxyethylpiperazinepropanesulfonic acid (HEPPS) was chosen for that step. Induction of Tyrosine different

procedures

aminotransferase purification.

to

Aminotransferase have

high

been levels

used in

in Rat

Liver.

Many

to

induce

tyrosine

liver

before

starting

Since it i s essential to chose an efficent method

for induction, Table I1 summarizes results obtained in this laboratory

using

various

procedures.

The

concentration of

tyrosine aminotransferase is greater in male rats than in females both in the basal and induced states (8), so it is preferable to use males. However, as shown here, the enzyme can be increased to levels exceeding 250 units per liver in females.

This compares

well to maximum values observed in males, so that animals of either gender may be used. The procedure involves inducing the enzyme overnight

so

and purification.

that a full day may be used for the extraction Results of induction with combinations of

PURIFICATION OF TYROSINE AMINOTRANSFERASE

15

TABLE I1

Downloaded by [NUS National University of Singapore] at 20:54 08 November 2015

Induction of Tyrosine Aminotransferase in Rats

Sex

Treatment

Average

Tyrosine

Liver

Aminotransferase

Weight

Units/g Units /Rat

8.1

2.0

16.3

0 . 1 mg/100 g

8.2

17.4

142

10 mg/100 g

8.6

38.2

329

12.3

14.7

182

14.3

23.2

333

10.4

36.1

375

15.8

23.6

375

Males,

Fasted 20 h

fasted

Triamcinolone,

Males,

50X casein diet,

fed

3 days

Triamcinolone, 0.1 mg/100 g (fed) Triamcinolone, 0 . 1 mg/100 g (fed),

Cycloheximide, 10 d 1 0 0 g

Dexamethasone phosphate in drinking water, 1 mg/100 ml (fed)

Females, Triamcinolone, fasted

0 . 1 mg/100g, plus

Cycloheximide, 25 pg/100 g 12.4 23.9 296 Animals ( 5 per group) were injected with Triamcinolone and/or

cycloheximide at 9 p.m.

and killed between 8 and 9 a.m.

Enzyme

activity was determined in duplicate for 9,OOOxg supernatants of individual livers.

16

HARGROVE

Triamcinolone acetonide or dexamethasone phosphate, cycloheximide, and high-protein diets are shown in Table 11.

Lower levels result

from use of hydrocortisone, insulin, and theophylline (not shown). Although

10 ug of synthetic

steroid per

100 g body weight

Downloaded by [NUS National University of Singapore] at 20:54 08 November 2015

provides maximum induction in 5 h (9), overnight induction with steroid suspensions yields highest levels of enzyme at 10 mg per 100 g (10). Comparable effects can be achieved at 0.1-1 mg per 100 g

body

weight

by

dissolving

Triamcinolone

acetonide

in

dimethylsulfoxide and including 10-50 pg of cycloheximide/lOOg

(11).

Cycloheximide functions by elevating the mRNA for this

enzyme, which leads to increased enzyme synthesis when translation begins again (12). Induction to similar levels can be achieved without injecting the rats by adapting them to a diet containing 50% protein by weight for at least three days, and adding dexamethasone phosphate to the drinking water at a concentration of 10 ug/ml on the day prior to sacrifice. Preparation of Liver Homogenates. ether and decapitated between steroid was administered.

Rats were anesthetized with

8-9 a.m.

on the day after the

Rats were exsanguinated and the excised

livers were placed in a beaker of 0.154 M NaCl at

4OC.

The livers

were rinsed several times with the cold saline solution, blotted on paper towels, and weighed.

Samples weighing one gram were used

to obtain the values shown in Table 11, and the rest of the livers were pooled and used to purify the enzyme as shown in Table 111. They were minced with scissors and homogenized in four volumes of 0.2 M sucrose containg 0.05 M HEPPS, pH 8.2, dithiothreitol, and 0.2 mM PLP.

1 mM EDTA, 2 mM

Disruption of tissue was achieved

with a Tekmar tissue homogenizer in 1 minute at 70% maximum power. The extract was immediately centrifuged at 9,OOOxg for 20 minutes in a preparative centrifuge, and the resulting supernatant solution was decanted through a double layer of cheesecloth. The supernatant was divided into two portions of about 600 ml each, and was heated to 65OC in a water bath maintained at 70°C

.

After five minutes, the sample was placed into an ice bath and cooled to 23°C.

The cooled extract was then centrifuged for ten

PURIFICATION OF TYROSINE AMINOTRANSFERASE

17

TABLE III Purification of Tyrosine Aminotransferase from 35 Rats (Liver wet weight, 370 g)

Downloaded by [NUS National University of Singapore] at 20:54 08 November 2015

Volume (ml)

Enzyme

Protein

(Units/ml)

(mg/ml)

Total

Specific

Units

Activity

(Units/mg protein)

12,000 xg 1290

5.4

21.6

6960

0.25

1090

5.6

5.7

6120

1.0

precipitation

180

34

6.0

6180

5.7

DEAE Cellulose

45

110

1.4

4980

78

Hydroxyapatite

9

492

3.1

4780

158

CM Sephadex C50

10.9

436

0.85

4760

510

Supernatant Heat to 65" for 5 m i n Isoelectric

Liver supernatants from the hormone-treated groups of male and female rats shown in Table XI were pooled and purified as described in the text.

minutes at

9,OOOxg

in a rotor at 4OC.

The volume of the

supernatant fluid was recorded, and the solution was acidified to pH 5.0 by dropwise addition of 10% acetic acid.

This causes

tyrosine aminotransferase to precipitate; after stirring the fluid for fifteen minutes at 4OC, the solution was centrifuged for five minutes at 2,700xg.

The supernatant fluid was decanted and

HARGROVE

18

discarded after verifying that the tyrosine aminotransferase had precipitated quantitatively.

The pellet was resuspended using a

Dounce-type tissue grinder in 50 mM potassium phosphate, pH 7.0, containing 0 . 1 M KC1, 2 mM dithiothreitol, 0.1 mM PLP, and 0.5 m M

Downloaded by [NUS National University of Singapore] at 20:54 08 November 2015

EDTA,

using

one-fifth

of

the

volume

recorded

prior

to

acidification. The suspended material was then centrifuged for 30 minutes

material

and

nucleoproteins that impede chromatography on DEAE cellulose.

at

100,OOOxg

to

remove

undissolved

The

precipitation yields a five-fold purification of the enzyme and a five-fold reduction in volume to permit more rapid chromatographic steps. Chromatography of Tyrosine Aminotransferase. next

series

of

steps

is

that

the

The basis for the

enzyme

DEAE-cellulose and CM-Sephadex C50 at pH 6.0,

binds

to

both

and that it can be

concentrated from solutions containing KC1 without dialysis using a small column of hydroxyapatite. Therefore, the redissolved enzyme was loaded onto a column of DEAE-cellulose equilibrated 50

with

mM

potassium

phosphate,

pH

7.0.

One

gram

of

DEAE-cellulose is used per rat, and the column should be 2 . 5 cm o r more

in diameter to permit application at a rate of about 100 ml

per hour without using a pump.

The column used for DEAE

-

cellulose chromatography was made from a plastic, 50 ml syringe, and

the ones used

for chromatography on hydroxyapatite and

CM-Sephadex C50 were made from 10 ml syringes.

After the enzyme

has been applied, the column is washed with 100 ml of column buffer (25 mM potassium phosphate, pH 6 . 0 , 2 mM dithiothreitol, 2 a-ketoglutaric acid, and 0.5 mM EDTA) containing 0.1 M KC1. PLP binds to DEAE cellulose and i s omitted during this step. A

mM

400 ml gradient of 0.1 to 0.5 M KC1 in column buffer is then

applied.

The enzyme elutes between 0.2-0.3 M KC1 (not sham).

The eluate from the DEAE-cellulose column is applied to column

containing

10

ml

of

hydroxyapatite;

a

tyrosine

aminotransferase binds without removal of potassium chloride. The column is washed with 20 ml of column buffer minus EDTA (EDTA is omitted because it chelates calcium to some extent); a yellow band should now be visible on the column, corresponding to bound

PURIFICATION OF TYROSINE AMINOTRANSFERASE tyrosine aminotransferase.

19

The enzyme is then eluted by washing

with 10 ml each of 0.2 M and 0.3 M potassium phosphate, pH 6.0, containing the same concentrations of dithiothreitol, PLP, and a-ketoglutaric acid as used in the earlier steps.

Elution can be

Downloaded by [NUS National University of Singapore] at 20:54 08 November 2015

monitored by watching movement of the yellow band.

The enzyme

should now be concentrated in less than 10 ml of buffer,

so

that

loss of activity due to dilution can be avoided. The optimum conditions for binding aminotransferase to CM-Sephadex C50 are

of 25-50

native mM

tyrosine

sodium or

potassium phosphate at pH 6.0 or less (unpublished observation). Therefore, the eluate from the hydroxyapatite may either be dialyzed or simply diluted to equilibrate it with the appropriate buffer.

Here, the enzyme was diluted with five parts of deionized

water containing 2 mM dithiothreitol and 0.2 mM PLP.

The pH was

adjusted with a few drops of 10% acetic acid, and the conductivity was checked to be sure it was very close to that of 50 mM potassium phosphate. The diluted enzyme was then applied to a column containing ten ml of CM-Sephadex C50 equilibrated with 50 mM potassium phosphate, pH 6.0, containing 2 mM DTT, 2 mM a-ketoglutarate, and 0.2 mM PLP.

The column was washed with 20 ml of the same buffer,

and the enzyme was eluted

stepwise with 10 mls each of buffer

containing 0.1, 0.2 and 0.4 M KC1.

The enzyme was recovered in

nine ml of buffer, was dialyzed against four liters of 25

mM

ammonium bicarbonate and then lyophilized.

A summary of the procedure is shown in Table 111, and a photograph of an electrophoretic gel containing enzyme purified by this method is shown in Figure 1.

The undegraded enzyme migrates

with an apparent molecular weight of 52,000,

indicating that

cathepsin T activity has been suppressed. Otherwise, a band would be present at 48,000 Daltons (6,7). Comments on the Procedure.

Purified tyrosine aminotransferase is

extremely stable, so long as it is protected against oxidation and dilution (unpublished observations). than at pH 6-7,

It is less stable at pH 8

but must be extracted from liver under alkaline

conditions to minimize the activity of cathepsin T, the lysosomal

Downloaded by [NUS National University of Singapore] at 20:54 08 November 2015

20

HARGROV E

FIGURE 1 Analysis of Purified Tyrosine Aminotransferase by SDS-PAGE Tyrosine aminotransferase samples resulting from chromatography on DEAE cellulose , hydroxyapatite (HAP) and Carboxymethyl Sephadex C50 (CM) were analyzed on a polyacrylamide gel containing 12% total acrylamide monomer, in the following amounts:

(lanes 1 and 2),

and 16 pg of protein; CM (lanes 1 and 2), Molecular

DEAE

7 and 14 pg of protein; HAP (lanes 1 and 2 1 , 8

weight

standards

are

5 and 10 pg of protein.

bovine

serum

albumin,

immunoglobulin G, and ovalbumin.

protease that rapidly cleaves an N-terminal segment from tyrosine aminotransferase (6,7).

Since cathepsin T is inactivated by

heating, the chromatographic steps may be performed safely at pH 6 after the initial heat step, Conditions that effectively stabilize tyrosine aminotransferase in vitro are shown in Table I.

The cofactor, PLP, protects

against inactivation, as does phosphate

ion.

Dithiothreitol

protects against losses due to oxidation, mechanical agitation, freezing and thawing, or heating, with an optimum concentration of

PURIFICATION OF TYROSINE AMINOTRANSFERASE about 20 mM.

21

These three protective agents are very effective

when combined in the concentrations used in this protocol.

By

minimizing dilution and the time required for preparation, the yield observed in the present work was 6 8 % . as compared to 15-43%

Downloaded by [NUS National University of Singapore] at 20:54 08 November 2015

in previous reports.

Glycerol at concentrations of 25-50% (v:v>

protects against inactivation due to freezing and thawing, and may be added during storage.

Enzyme stored at -20" in 40% glycerol

retains its primary structure but loses activity with a half-time of about 6 months; an inert atmosphere such as nitrogen or argon is beneficial.

This rapid procedure for purifying tyrosine aminotransferase

relies upon precipitation with acetic acid to reduce the volume of the initial extract, which must be done after the heat step because cathepsin T has an acidic pH optimum. Precipitation works reliably only in the absence of KC1, which solubilizes the enzyme. Therefore, sucrose i s included to lessen release of cathepsin T from lysosomes due to osmotic lysis.

Using ammonium sulfate,

polyethylene glycol, or ethanol as precipitating agents either gave less purification, lower recovery, or introduced salts that must be removed prior to chromatography. Small volumes of chromatographic media are used to minimize dilution during

application

contaminating proteins.

of

gradients

and

adsorption of

To provide for moderate rates of flow,

columns may conveniently be made from 50-ml or 10-ml plastic syringes, with sample being applied by way of a second syringe (minus plunger) connected to the column with plastic tubing. Together, these steps minimize dilution and permit a high degree of purification to be achieved in one or two days.

Since several

milligrams of enzyme may be prepared quickly from a small number of rats, it should now be possible to characterize functional regions in tyrosine aminotransferase with comparative ease.

ACKNOWLEDGEMENT This work was supported by U . S . P . H . S .

grant number DK39329.

HARGROVE

22

REFERENCES 1.

J. L. Hargrove, H. A. Scoble, W. R. Mathews, B. R. Baumstark, and K. Biemann, J. Biol. Chem.

2.

S. Rogers, R.

3.

J. F. Dice and H.

4.

D. K. Granner and G. M.

Downloaded by [NUS National University of Singapore] at 20:54 08 November 2015

(1986)

.

(1989)

264,

45-53

(1989).

Wells, and M. Rechsteiner, Science

Chiang, Biochem.

L.

SOC.

234, 364-368

Symp.

. Tomkins, Meth. Enzymol.

55,

45-55

E , 633-637

(1970). 5.

U. K. Laemmli, Nature

6.

J. L. Hargrove, M.

227,

680-685

(1970).

D i e s t e r h a f t , T. Noguchi, and D.

Granner

J. Biol. Chem 255, 71-77 (1980). 7.

Godha and H.

E.

C.

Pitot,

J.

Biol.

Chem.

255,

7371-7379

235,

E374-E380

(1980). 8.

J. Voigt and C.

E. S e k e r i s , h e r . J. Physiol.

(1978). 9.

T. H i r o t a , K. H i r o t a , Y. Sanno, and T. Tanaka, Endocrinology

117, 10.

1788-1795 (1985).

F A. V a l e r i o t e , F. Auricchio, G . M. Tomkins, and D. R i l e y , J. Biol. Chem. 244, 3618-3624 (1969).

11.

F.

M.

Baccino,

T e s s i t o r e , and M.

G.

Cecchini,

F.

Palmucci,

V.

Z u r e t t i , Biochim. Biophys. Acta

Sverko,

L.

9. 91-97

(1977). 12.

M. J. Ernest, L. DeLap, and P. Feigelson, J. Biol. Chem. 2895-2897 (1978).

253,

Stabilization and purification of tyrosine aminotransferase from rat liver.

Purification of unmodified tyrosine aminotransferase from rat liver requires that the activity of cathepsin T be minimized, and that losses of enzyme ...
648KB Sizes 0 Downloads 0 Views