Anda di halaman 1dari 14

Attenuation of plasma low density lipoprotein cholesterol

by select 3-hydroxy-3-methylglutaryl coenzyme A


reductase inhibitors in mice devoid of low density
lipoprotein receptors
Charles L. Bisgaier,'?*Arnold D. h e n b u r g , * Bruce J. Auerbach," Michael E. Pape,t
Catherine S. Sekerke," Andrew Gee,* Sabine Wolle,* and Roger S . Newton"
Department of Vascular and Cardiac Diseases" and Molecular Biolob7,t Parke-Davis Pharmaceutical
Research, Division ofb'arner-Lambert Colnpany, 2800 Plymouth Road, Ann Arbor, MI 48105

Downloaded from www.jlr.org by guest, on September 17, 2018


Abstract Low density lipoprotein (LDL) reduction indepetl- lipidemias caused by LDL-receptor disorders.-Bisgaier, C. L.,
dent of LDL receptor regulation was investigated using HMG- A. D. Essenburg, B. J. Auerbach, M. E. Pape, C. S. Sekerke, A.
CoA reductase inhibitors in LDL, receptor-deficient mice. In Gee, S. Wolle, and R. S. Newton. Attenuation of plasma l o w
males, LDL cholesteroldose-dependentlydecreased with density lipoprotein cholesterol by select 9-hydroxy-3-~nethylglu-
atorvastatin treatment after 1 week. As untreated mice grew tar$ coenzyme A reductase inhibitors in mice devoid o f ' l o w
older, their LDL cholesterol progressively rose above basal density lipoproteinreceptors. ,/. I,ipid Krc. 1997. 38: 2502-
levels, but was quelled with atorvastatin treatment. I n females, 2515.
atorvastatin treatment time-dependently decreased LDI. cho-
lesterol levels and induced hepatic HMCXoA reductase activ- Supplementary key words acorvastatin lovastatin pi-avastatin
ity. Unlike males, cholesterol-lowering effects of thedrug sirnvastatin
were sustained in females. Lovastatin, simvastatin, and prav-
astatin also reduced total and LDL, cholesterol; however, addi-
tional studies i n females demonstrated that atorvastatin
caused the greatest dose-dependent and sustained efyect after
2 weeks. I n females, hepatic HMGCoA reductase IIIRNA i n - Homozygous familial hypercholesterolemia (FH) is ;I
versely correlated with LDL cholesterol lowering, with ator- rare vascular disease caused by defective or deficient
vastatin showing the greatest increase in mRNA levels (17.2- L,DL receptors and is characterized by marked hyper-
fold), followed by lovastatin (10.7-folti),simvastatin (4.1-fold), cholesterolemia, tendon xanthomas, arcus comeae, and
and pravastatin (2.5-fold). Atorvastatin effects on lipoprotein life expectancy of less than 30 years d u e to myocardial
production were determined after acute ( 1 day) or chronic (2
infarction (1-3). Treatmentsincludeplasmapheresis,
week) treatment prior to intraperitoneal injection of Triton
WR1339. Acute treatment reduced cholesterol (-29%) ant1 LDL-apheresis, combined liver and heart transplants,
apoB ( - 16%) secretion, with no change in triglyceride secre- and experimental trials of LDL receptor replacement
tion. I n rontrast,chronictreatment elevated cholesterol by gene therapy (4-8). As up-regulation of LDL, recep-
(+20%), apoB (+310/0), and triglyceride (+57%) secretion. tors is thought to be the major mechanismof action of'
Despite increased cholesterol and apoB secretion, plasma le^-
els were reduced by 51% and 46%, respectively. Ovcrall,
HMG-ChA reductase inhibitors, their effectiveness in
under acute or chronic conditions, apoB paralleled choles- LDL receptor-deficientsubjectswouldnot he antici-
terol secretion rates, and triglyceride to cholcstc~-olsecrction pated (1, 9, 10). I n contrast, however, we recently a n d
ratios were elevated by 88% and 32%, respectively. we. pro- unexpectedlydiscovered that
atorvastatin
lowered
pose that atorvastatin limits cholesterol for lipoprotein assem- plasma LDL cholesterol in dietary casein-induced pre-
bly, which is compensated for by triglyceride enrichment. In
addition, with either acute or chronic atorvaslatin treatment,
apoB-100 secretion was blockcd, and compensated for by a n
increased secretion of apoB-48. The apoB-48 particles pro-
Ahbreviatiorls: ANOVA, analysis 01. variance; HDL., high dcnsit?
ducedarc cleared b y LDL, receptor-indeperldent mecha- lipoprotein; H M ( X o . 4 , ~ h ~ ~ l r o x y - Y n l c t h y l gc-ocwyme
i u ~ ~ ~ l A; H P ( X
tlisms, with an overall effect of reducing LDI, production i l l high performancc gel-filtration chromatography; I , I ~ t , ,low clcnsity
these 1nice.m These studies suppor~ theidea that HM(;-<:oA lipoprotein; TRS, Tri+huttt.rcrl salinc: VI.DI,. \.VI-? l o w tlcnsitv
reductase inhibitors modulate lipoprotein levcls indepentie~rt lipopl-otci~~.
of I,DI, receptors, and suggest thry tnay havc utility i n 11ypc1.- "Io whom (.orresporrdcrl(.c slloulti Ilc a t i t l r c s w d .

2502 Journal of LipidResearch Volume 88, 1997


established hypercholesterolemia in normal rabbits Vastatins
through the direct inhibition of LDL production and Atorvastain was prepared at Parke-Davis (21) . Lova-
not enhanced clearance (11). In addition, studies of statin (Merck, Sharp & Dohme) was prepared by ex-
Grundy and Vega (12), Vega, East, and Grundy (13,14) tracting formulated capsules of Mevacor (22). Prava-
and Arad, Ramakrishnan and Ginsburg (15, 16) have statin was provided by Bristol-Meyers Squibb and
suggested that lovastatin can act, in part, by limiting simvastatin was provided by Merck, Sharpe & Dohme.
LDL production in specific human hyperlidemias.
More recently, Shiomi et al. (17) have shown that flu- Blood sampling
vastatin sodium modestly reduced VLDL production in
the Watanabe heritable hyperlipidemic (WHHL) rab- Blood samples (100-200 ~ 1 prior
) to and after drug
bit. However, the WHHL rabbit expresses about 2% of treatment were collected at 8 AM in 0.04% EDTA and
the normal amount of LDL receptors, and therefore an 0.01% sodium azide from mice while under Metofane
effect on LDL receptor up-regulation cannot be totally (Pitman-Moore, Mundelein, IL) anesthesia after an 8-
ruled out as partially responsible for cholesterol lower- 10-h fast. For production studies, non-fasting blood
ing. Similarly, of the three FH subjects studied by Arad samples were collected before (at 9 PM) and after (9 AM)
et al. (15) one was heterozygous, another LDL receptor Triton WR 1339 treatment as described below.
defective, while the third subject was not classified. To
Determination of plasma lipids
directly test the hypothesis that HMGCoA reductase in-
hibitors can lower LDL cholesterol independent of Plasma total cholesterol was determined enzymati-
functional LDL receptors, we treated LDL receptor-de- cally by the method of Allain et al. (23) and plasma tri-

Downloaded from www.jlr.org by guest, on September 17, 2018


ficient mice with various concentrations of vastatins ad- glycerides were determined with a commercially avail-
ministered as diet admixtures and then measured able kit (Triglyceride G Kit #997-69801 Wako Pure
changes in their lipoprotein profiles. In addition, the Chemical Industries, Ltd., Richmond, VA) using a mi-
acute and chronic effects of atorvastatin administra- crotiter plate method (24). The reagent kit measures
tion on cholesterol, triglyceride, and apoB secretion plasma free glycerol after hydrolysis of triglycerides.
rates and apoB forms in these mice were also deter- Values were not corrected for the presence of plasma
mined. Although this mouse model has allowed us to free glycerol. Distribution of cholesterol among plasma
study effects of the vastatins on LDL cholesterol levels lipoproteins was determined by on-line continuous
and production independent of the LDL receptor, in- monitoring of postcolumn eluant after Superose 6
herent differences in hepatic apolipoprotein B mRNA high performance gel-filtration chromatography (HPGC)
editing (18), lipoprotein production as well as differ- of 5 ~1 individual or 5 pl pooled plasma on a high pres-
ences in lipoprotein metabolism between mice and hu- sure liquid chromatography system (Rainin Instrument
mans may limit the interpretation of how these com- Go., Inc., Woburn, MA) as previously described (25,
pounds act to lower LDL cholesterol in this mouse 26). Peaks were collected and areas were integrated
model and humans. and averaged using Dynamax and Compare Module
Software (Rainin Instrument Co., Inc., Woburn, MA) de-
veloped for Macintosh computers. Lipoprotein choles-
terol was determined from independent total choles-
terol determinations and percent area distribution of
METHODS cholesterol determined by HPGC. In studies where
samples were pooled for HPGC, plasma total choles-
Mice terol was determined on both the pools as well as the
Breeding pairs of low density lipoprotein receptor- individual samples. In these studies, the pooled plasma
deficient mice originally created by Ishibashi et al. (19, total cholesterol determinations (which were essen-
20) were obtained from Jackson Laboratories and a col- tially identical to those obtained from the average of
ony was established and maintained at Parke-Davis. the individual determinations) were used to determine
Mice were maintained with up to 5 mice per cage on cholesterol in the lipoprotein fractions. To verify that
chow and water ad libitum in temperature-controlled the two major plasma cholesterol peaks separated by
rooms on a 12 h light/l2 h dark cycle (lights on at 6 the HPGC procedure corresponded to LDL and HDL,
A M ) prior to study. Mice were assigned to be either in a pooled plasma sample from untreated female LDL
control groups fed ground rodent chow alone or to receptor-deficient mice was gel-filtered, and individ-
groups fed chow such that approximately 10, 30, 100, ual fractions were collected and analyzed for apoH,
or 300 mg/kg vastatins were consumed daily as a diet apoA-I (27), and total cholesterol. Essentially all the
admixture. apoB eluted in the first major cholesterol peak, while

Bz~gazpret nl. Vastatins reduce plasma LDL in LDL receptordeficient mice 2503
all the apoA-I eluted in the second major peak (data an internal standard/RNAse protection assay essentially
not shown). as described previously in detail (31, 32). The protec-
tion assay contained 30 Fg of total liver RNA and 10 pg
Determination of plasma apoB of internal standard RNA. Autoradiographic images
were analyzed using the Molecular Dynamics 400E Phos-
Plasma apoB was determined by an immunoturbido-
phorImager (Molecular Dynamics, Sunnyvale, (A).
metric assay as previously described (27). The differ-
ence in plasma turbidity (A340) measured immediately Lipoprotein production studies in female LDL
after treatment and after overnight incubation with receptor-deficientmice
sheep anti-mouse apoB antiserum is directly linear to
For lipoprotein production studies, 'Triton WR 1339
the amount of mouse plasma added to the assay. For
was used to block plasma lipoprotein clearance,
each sample, turbidity change was assessed at multiple
thereby allowing measurement of cholesterol, triglycer-
plasma volumes (1-4 ~ 1 to) assure detection in the lin-
ide, arid apoB accumulation (Le., secretion rate) in
ear portion of the response curve. ApoB data are arbi-
plasma (33,34). Pilot studies demonstrated linear accu-
trarily expressed as change in OD340.
mulation of triglycerides in these mice after intraperi-
toneal administration of 0.9 mg Triton WR 1339/g
Determination of hepatic HMG-CoA reductase activity body wt up to 12 h (not shown). Female LDL, receptor-
Hepatic HMGCoA reductase activity was determined deficient mice were acclimated to eating ground ro-
in microsomes prepared from livers of control and dent chow prior to the study. Studies were carried out

Downloaded from www.jlr.org by guest, on September 17, 2018


atorvastatin-treated mice (28). Briefly, to prepare mi- to test the acute (1 day) and chronic (2 week) effects ol'
crosomes, mice were killed and livers were excised and atorvastatin. Prior to the initiation of the studies, a
then homogenized in 5 m L of cold 0.3 M sucrose, 5 nihi baseline blood sample (under Metofane) was obtained
EGTA, 5 inM dithiothreitol, 53 mM leupeptin, pH 7.4, at 9 P M in the fed state. To determine the acute effects
with 10 strokes of a tight-fitting Potter-Elvehjem ho- of atorvastatin, ground chow alone or chow containing
mogenizer. The homogenates were centrifuged for the equivalent of 300 mg/kg per day atorvastatin was
12,000 gfor 15 min; decanted supernatants were respun placed in cages in the morning (9 A V ) . At 9 I'M that
at 12,000 g f o r 15 min, and supernatants were centri- same day (e.g., 3 h after dark cycle start), a blood sam-
fuged at 100,000gfor 1 h to prepare microsomal-rich pel- ple (under Metofane) was obtained, and mice were
lets. The microsomal pellets were resuspended in 200 mhf then administered intraperitoneal Triton WR 1339 (0.9
phosphate-buffered saline, pH 7.4, and frozen immedi- nig/g body wt) under Metofane. Mice were returned t o
ately in liquid nitrogen, and assayed within 1 month. their cages without removal of food. Additional blood
HMGCoA reductase activities were determined as preti- samples were obtained at 9 AM the following day (itndt~r
ously described by Kim, Brown, and Goldstein (28). Metofme). For the chronic effects of atorvastatin, niicr.
had access to ground chow alone or chow containing
the equivalent of 500 mg/kg per day atorvas~atirifbr 2
Determination of hepatic HMG-CoA reductase weeks, when blood samples were obtained beforc (at 9
mRNA levels I Y ) or 12 h post-Triton WR 1339 administration (at 9 AM).
Hepatic RNA was isolated with RNAzol (Cinna/Bio- In addition, iinmunoblot analysis ( 3 5 , 36) of apoB-
tecx, Inc., Houston, TX) and assessed for any degrada- 100 arid apoB-48 in acute atorvastatin, chronic atorva-
tion by agarose electrophoresis (29). A partial cDNA statin, or control treatment groups wits carried o u t l x -
clone for mouse HMGCoA reductase (30) was isolated forc arid after Triton WR-1539 injections. Briefly,
by PCR. The amplified product of approximately 250 pooled plasma (0.7 ~ 1 proteins
) were separatcd on a 3-
base pairs was obtained by performing PCR with mouse 12% sodium dodecyl sulfate polyacrylamide gel (Novcx,
liver cDNA (Clontech, Palo Alto, CA) as template and San Diego, <A) and transferred to a nitrocellulose
conserved primers for HMCXoA reductase which were membrane. Af'ter- blocking with 3%) gelatin in Tris-
described previously (31). The reaction buffer con- buffered saline, pH 7.4 (TBS), niernbraries werc incu-
tained 60 i n k t Tris-HC1, 15 mivi ammonium sulfate, 2 mM bated with a 1:5000 dilution of polyclonal sheep anti-
MgCl,, pH 8.5, and the 35 cycles were run at 94°C for 15 mouse apoB serum in 10% fecal calf serum in TBS,
sec and 60°C for 30 sec. The amplified product was washed with TBS, and then incubated with a 1:5000 di-
cloned into pCRII (Invitrogen, San Diego, ('A) and se- lution donkey anti-sheep Ig(; conjugated t o alkalinc
quenced. The cDNA was found to be 100% identical to phosphatase (Sigma Chemical Co., St. I,ouis, MO) in
mouse HMGCoA. A specific antisense riboprobc and an 10% fetal calf serum in TBS. After TBS washing, Lhc
internal standard RNA for mouse HMGCoA reductase membrane was developed using a bromochloi-oindolyl
were synthesized and mRNA levels were determined by phosphatc/nitro blur tetrazoliuni substrate.

2504 Journal of Lipid Research Volume 38,1997


Statistical analysis progressively rose over 3 weeks compared to the basal
Statview (Abacus Concepts, Inc., Berkeley, CA) devel- bleed (Table 1). A similar effect on plasma lipoprotein
oped for Macintosh computers was used to statistically cholesterol was observed at the lowest dose of atorvastat-
analyze data. Data were analyzed by t-tests, where P in (10 mg/kg per day) tested. At 30 mg/kg per day,
values <0.05 were considered statistically significant. atorvastatin neutralized the time dependent rise in
For multiple group comparisons, analysis of variance plasma cholesterol. At the two highest atorvastatin
(ANOVA) and post-hoc Fisher's protected least signifi- doses used (100 and 300 mg/kg per day), both total
cance difference analysis was used to analyze data. and LDL cholesterol were reduced. A reduction in
HDL cholesterol was also observed at these higher
doses. At 300 mg/kg per day, both total and LDL cho-
lesterol were markedly reduced by 36% at 1 week com-
RESULTS pared to the level observed at 2 (13 and 9% reduction
for total and LDL cholesterol, respectively) and 3 weeks
Baseline cholesterol values from age-matched, fasted (10% reduction for total and a 1% increase for LDL
chow-fed female (89 .f 1 days old, n = 50) and male cholesterol, respectively). Thus, the mice were able to
(90 2 2 days old, n = 50) LDLr-/- mice are shown in compensate for the drug effect, possibly by increased
Fig. 1. Female mice had significantly higher total drug inactivation or induction of HMGCoA reductase.
plasma cholesterol (219 +- 7 mg/dl) than males (192 +- Studies were initially performed in male mice (Table
6 mg/dl) , which was largely reflected by significantly el- 1); however, because of potential compensatory mecha-
evated LDL cholesterol (149 ? 6 mg/dl versus 91 ? 4 nisms to inactivate HMGCoA reductase inhibitors, as

Downloaded from www.jlr.org by guest, on September 17, 2018


mg/dl) and reduced HDL cholesterol (59 2 2 mg/dl demonstrated for simvastatin in male rodents ( 3 7 ) ,we
versus 91 2 2 mg/dl) . This marked difference between sought to determine atorvastatin's effectiveness in fe-
females and males under basal conditions can also be males. We chose the 300 mg/kg dose as this appeared
appreciated as the significantly reduced ratio of HDL to be initially most effective in the males despite its de-
cholesterol to VLDL plus LDL cholesterol in the fe- creased effectiveness with time. At the time of killing,
males (0.404 +- 0.016) compared to the males (0.995 2 liver biopsies were removed for isolation of microsomes
0.039). No differencs in VLDL cholesterol was ob- for determination of HMGCoA reductase activity. In
served between the genders; however, triglycerides addition, single male mice were also treated for 1, 2, or
were significantly reduced in females (67 2 2 mg/dl) 3 weeks with 300 mg/kg per day atorvastatin for deter-
compared to males (99 2 4 mg/dl). mination of hepatic microsomal HMGCoA activity.
To investigate the role atorvastatin might have on Prior to atorvastatin treatment, the plasma lipoprotein
plasma lipoprotein cholesterol levels in these mice, cholesterol levels (Table 2) were typical of female mice
males were treated for 3 weeks with various doses of (Fig. 1) having more LDL than HDL. Similarly, as was
atorvastatin and bled weekly for determination of previously observed in the males at 1 week, atorvastatin
plasma lipoprotein cholesterol levels and distribution treatment at 300 mg/kg per day reduced total and LDL
(Table 1 ) . In control mice fed chow without drug, total cholesterol. However, unlike males, this reduction was
plasma cholesterol and specifically LDL cholesterol more pronounced and sustained at 2 and 3 weeks in

250
225
p=OOO32
I-+

I,
Female (n=50)
Male (n=50)
x1.2
p < 0.0001

Fig. 1. Fasting plasma lipid levels in female and


male LDL receptordeficient mice maintained on
150 chow diets. Fifty male (90 ? 2 day old) and 50 fe-
125 male (89 t 1 day old) LDL receptor-deficient
100 mice maintained on chow were bled after an 8-h
fast to determine baseline lipid values. Lipid val-
ues representing mean ? standard error of the
50 mean were determined as described in Materials
and Methods. Data were analyzed by a two-tail un-
25
paired t-test.
. . . .
Total VLDL ' LDL' 'HDL' ,Triglycerides Ratio

Cholesterol

Bisgaier et al. Vastatins reduce plasma LDL in LDL receptordeficientmice 2505


TABLE I . Effect ol.atorvastatin on lipopi-otein cholesrcrol and triglyccritles in nialc
LDI>receptor-deficient mice

0 142 2 I 1 15s I x (-3) l ~ 5 9 i -I9 ( + I ? ) 201 i- I 1 (+"')'


IO 197 + 13 202 i- 8 ( + 3 ) 242 t 16 ( + 2 3 ) " 2.55 t 13 (+29jt
:30 I64 5 1 J 159 t 6 ( - 3 ) 1x1 I 9 ( + I O ) I66 i- 9 ( + 1 )
100 15s 1 4 1:w t 0 ( - 1 X) I29 2 9 (-18) 124 + 13 (-22)"
300 164 I .5 105 + 1 1 (-?l6)( 1.13 I x (-- 1 X) 147 18 (-IO)+

VLDI. cholesterol
0 :3.9 t 0.8 4.0 t 0.5 ( + I ) 6.1 + 1.7 (+57) 8.4 I 2 3 ( I 15)' +
IO (i.2 + 0.2 6.4 i 0 . 3 (+1) 6.2 f 0.6 ( + I ) 10.4 2 0.6 (+(i7j"
30 ?)
!
I i- 0.3 4.1 t 0.4( + l 3 ) 4.6 -t 0.4 (+?PO) .5.6 2 0.7 (+60)
100 4.0 i- 0.:3 5.0 1 0 . 5 (+23) 5.1I 0.9 (+?d) .5.7i 0.5 ( + 4 l )
300 4.1 + 0.5 !).ti i I , < ) (+12X)" 5.2t 1.3 ( + 2 7 ) 14.1 2 1.x ( t 2 4 2 ) '
L,DI>cholesterol
0 68 I !I 6'7 i !) ( - 1 ) 79 f I I ( + I ( ? ) 121 % 1 1 ( + 7 X ) '
IO SH x * 94 2 4 (+7) I18 + 9 (+?J4)" 142 I x ( + W ) '
30 82 + 1 78 + 7 (-5j 00 2 7 ( + 10) X6 2 4 ( + 5 )
IO0 ti7 5 3 i t i t 7 (-17) 58 t 6 (-14) 55 I x ( - I S )
*-
300 t : > i- 5 18 -i 7 (-?di)[' ti8 f 6 ( - 9 ) 76 I 9 ( t I j
HDI. cholesterol

Downloaded from www.jlr.org by guest, on September 17, 2018


0 (iX t 7 t i l -+ 2 ( - 6 ) 72 + 8 (+6) 65 t 1 ( - I )
IO <I!) +
9 99 I IO ( 0 ) L15 t I 1 ( i - I [ ? ) 09 t I I ( 0 )
50 7s i 9 75 2 5 (--4) 84 + 5 (+7) +
71 (i( - 1 0 )
100 x4 i- 5 6 i 17 (-20)" ti4 t 7 (-24)" 01 I 8 (-2s)'
300 84 t L' 46 I 5 (-4.5)' (i9 t 5 ( - I S ) 5b t 8 ( - 3 3 ) '
Hl)l~/(\'l.l)I. + 1,DI.)
0 O.!)S i 0.14 0.94 t 0. 19 ( - 4 ) 0 3 7 2 0.0s (- 11 ) 0..5:3 t 0.06 (-45)'
IO 1.07 I 0.1 1 1 . 0 1 i- 0.13 ( - 6 ) 0.94 t 0.10 (-12) 0.65 1 0 . 0 6 (-?&))'
50 0.91 i 0. IO 0.96 t 0.1.5 ( + 3 ) 0.91 t 0.10 (-1) 0.77 f 0.0.5 ( - 16)
IO0 I . I I)t 0. 10 1.17 t 0.16 ( - 2 ) 1.07 2 0.16 (-11) +
1.03 0.15 (-14)
300 1.07 i 0.08 O S ? i- 0.08(-24)(' 0.97 + 0.12 (-10) +
0.62 0.03 (-42)"
I'lasma triglycerides
0
..-
/ 3 i '7 Oi%i(-II) I20 I 1'7 (+bo)' fi5 + -1 ( -I X)
-.
IO 04 t 7 70 2 i( - 19) 83 t IO (-12) lh t I ( - I!))
30 (iJ + (i 57 + 4 (-10) +
65 r, ( + 9 ) ! + 4 (-6)
54
IO0 72 2 :I 58 t 2 ( - 1 9 ) 73 t h ( + I ) 75 i (i( + . I )
300 74 + 5 X?I')(+ll) 02 i- 9 ( - 1 t i ) 7X i- 2 ( + . 5 )
~~

Dosedependent effccts of atorvastatin in rnalr 1.111. rereptor-tleficicnrmice. Mice were fed chow alone (11 =
4) 01-chow containing atonastatin such that approximately 10 ( n = 5 ) , 30 (n = 5), 100 (11 = ~ 5 )o. r 300 (11= 4)
m g drug/kg body weight was consumed daily. Values represent mg/dl. -t SEM. Numbers in parenthesis repre-
sent percent change from basal Irvt.1. I)ara wci-c aiialy/cd by ANOVA of the perccllr changes Iroiii pretreatillelit
(hasal) ~ A I L I Y S
I ' i 0.01; ' l J <0.005; "/'< 0.0005: ' P i 0.0001, c o n l p ~ l r ~ Yt ol I,aaal \.;tIIIc.

the females (Table 2) compared to males (Table 1).De- CoA reductase inhibitor through a mechanism unre-
termination of hepatic microsomal HMGCoA reduc- lated to LDL receptor elevation is not unique. Indeed,
tase activity in the females demonstrated a marked in- kinetic studies, albeit in LDL receptorcompetent ani-
duction of the enzyme as early as 1 week, reaching levels mals, have suggested a potential block in LDL produc-
greater than 10 times that of untreated females by 3 tion by HMGCoA reductase inhibitors (11-16). To de-
weeks (Fig. 2). Hepatic microsomes prepared from single termine whether LDL reduction is a class specific
male mice after 1, 2, and 3 weeks of 300 mg/kg per day effect, LDL receptor-deficient mice were treated for 1
atorvastatin treatment also suggested a timedependent week with 300 mg/kg per day with various HMGCoA
rise in HMGCoA reductase activity (not shown). reductase inhibitors, and their plasma lipoprotein cho-
The observation that atorvastatin reduced total and lesterol distributions were cornpared to pretreatment
LDL cholesterol in LDL receptor-deficient mice im- levels (Fig. 3).All treatment groups consisted of thrcc
plies that LDL production might be impeded by H M G males and three females, except the pravastatin group
CoA reductase inhibitors. LDL lowering by an HMC; in which there were two males and three females. At

2506 Journal of Lipid Research Volume 38, 1997


,!,09Z asnu $napppSo$daaar?(I? u! ?<I? lrurslrtd asnpa q m ~ .p
e $2~uzv2xzg
dnoJB s!ql u! q u r U ~ A 30 ~ SJnoj 'asop sly1 1~ .pamasqo W L W W O
0 0 0
: 3 0 0 1
aJaM 1o~a1saloy3 7 a H pue 7 a T pampa1 pue U O ! ~ E A ~ :
" - 1; 5
-la 1o~alsaloq3 Tam pay~eurXqaJayM pa1nqpls!paJ WM GZ$ $ -
A, n ,9 $.
~
I O J a W 3 l O y 3 'U!ll?lSEAOl 8y/BLU 006 1V 3SOp By/8W 001 gg G: I S
aq1 1e pauasqo SEM 1o~alsa1oy37a7 pue le101 u! uop c cg
2
-3npaJr 1q8qs e 'upqsenol 103 .(ap e d 'p '816)sasop 11e GG 8 WN-LW
e ?
8 .;1
le 1uaJedde s! 1o~a1sa1oy3 T a T pue le101 u! uoy3npaJ A8 3 ~S
g7iy
luapuadap-asop F: 'u!l~ls~mole JOJ .salgOJd ~ U ~ U I J E ~ J ~ c" g e Ips.**
-aJd aq1 uopasodurpadns a x (salyoJdpapeqs) sa19
- y"2
%x M N N N
qrncccc
-old 1uauru3ea~1 )sod paIood aq1 (a pued 'p %A) sdnod3 ;gg 0 - N N I3 j
I t I t It I t
lUaUIleaJ1 U!lI?lSEA 91 aql J O j 'XIJel!W!S '(p!EIJaAO) by5
"e5 2.
g 4 - - N
.I<&

~I ; ,
~ ; ?
2
pasodurpadns osle pue '(qgoJd papeys) M O Y ~ uo 5
G o j. j --row .,
-
s y a a ~ age ' (alyoJd uado) a~o3aqsalgoJd aql sMoqs
z rnmwrp 3
mkbptnx
z
v laued '(v Iaued 'p .&J) syaaM .Ioj M O ~ Duo p a u ~ e ~ &PA ES 2 I t It I t I t ;?
N - N -
-u!eur a>!w lua!3gap-Jo1dam_r T a T ap2ura3 1 0 ~ 1 ~ 0 3 $;."K w-urrn $ 3
R S L
""
ayl U! paAJXq0 aJaM Sa8UEq3 OU Xl[E!lUaSS7J.paA!AJnS x;; I I I I F
" '4 r 4 '
a
0 . 'Q -4-
1eur!ue auo XIUO a ~ a dnolS q ~ U ~ E J S E A U I ! ~lsod Xep _rad $2 "V
- a

- 5
By/%ur 00%a q 3~0 uopda3xa aql q l ! ~dnoJS lad a3!ur n 0 2
ag - 2
U ~ A ~ 3
~ 1S0 1 curseid 30 saurnIoA Ienba urog p a ~ ! ~ aSEM p mm.000 Fp
I t It I t I t 2 %
Iood eursqd q x q '(p %d) 3 3 d H kqs!sX~eue a1goJd $6 M-NIC. ; ? $ 2
1o~alsaloq3u!a~o~dodq~ 0 3sluaur1ea~1U!WISEA ~a1je gg N 1 N r
c ; e?
r
!$ - C N M c:E
3
pue aJojaq sdnoJS luaurlea~lWOJJ palood SEM eursqd I t It I t I t 0 ;
3 2. 5, Cil F
s .3
'SyaaM 2 103 U!lElSeAUI!S JO 'LI!~E)SEAE.I~ 'U!l4SEAOl 'U! ""
- 0
7i? ++++ $ a
i -
3
-mse?~ole '~o~!q!qu! asm3npaJ~ 0 3 a q330 Xep ~ Jad
~ % e
ESm-uI 6'
s,< 0.TN.C
w 7 0- 0-
By/Bur 006 10 '001 '06 '01 30 1uapqnba ay1 Bu!u!e~uo~ "
25 '?
sa.~nlx!urpe 10 auole M O Y ~paj a!" l u a ! q a p - ~ o ~ d a ~ a ~ - 7 -"I
z
54 5,m.l-J E
2.
T a T a~eura3 u! pauno3~adSEM Xpms X3e3g3a upels W"N I
% $ 0
I t I t I t It j.
-EA aAgeJeduro3 e 'suopemasqo aAoqe aq1 uo pasea ?ro -00 5,N
3'0, r
3 2 I N o
.1o~aalsaloq3 7 a H p a m p a os@ e a
_I
J
=
E5
sugelseil ayl 1~ Ala~p3adsa~ 'u!~e~se~ur!s kq %op pue 16 3g .I"* 0
.%a 3 m 0 4 , % ' -.
pue 'upelseAeJd dq %sz pue 0z 'upqseao~Xq %rp pue G' r It It I t I t
I
2 It ",+I
5s 'u!lelsemo1e Xq %pp pue 86 Xq pampa1 a ~ lo~a1sal a ~ - ,a0 g if ;
Z K I I"
-0y3 7 a T pue le101 'uo!lmnp 1uau11eaq pue asop s!qs LnmLL
2F <&g
$5 ~
Downloaded from www.jlr.org by guest, on September 17, 2018

%$ - 1
7- F F
.sooo'o >d*.* 'gyo > d * :10auo3p a l e a ~ ~ u0 n1 uosuedwo3 0'9 0 x tC
;
S $ O O N N 0
'VAONV dq ~ ~ Z X I E U aE ~ meaa '(p
~ = u ) s g a a ~E JO '(5 = u) S X ~ ~ M I t It I t I t s
z 3
'(6 = u) g a a I~ ~ 0 Bmp Xep lad 8y/8w 006 Bu~~eur!xo~dde sa~n~
5 5' -"a
- m o
-X!UlpE )a!p LI!lElSI?AJOlr? + MOY3 J 0 (6 = U ) aUO[E MOY3 pa3 aJaM Ez ; e? %= j
a3!w lua!3yap~o1da~1a~ a Ta ~ a ~
7a7 ' a s p alewa3 ~ u a ! ~ y a p - ~ o ~T d 2;
0 3
.llrDwa n
I p ' Q W N Y .
a5 It I t I t I t
2 0
palk?an-ugQWOlt?U! k)!,i!pE am3npaJ V O ~ W H 3gedaH 'X *%Id 2 2
3 2 il"Uc
N W "
2-. 2 " h
'+
ci
% =2 l o
3.
SyaaM SyaaM yaaM 3 2 l + l r CD
SIOJ)UO~ N-lrDtC
aaJlll OM1 aU0 peleeltun 0
FT0 mo--
"
~
' I
1 I I
O G K
3 s
"
1 - 1 2T
wrc
X&&$
R;1 NWSQIffi g i
=I
- ooz-z(P a0 It It It It
-a 0% 3 2 zzgg I
i;' n
3 0-
0-0 5. ?a ooIpcc js
3
- eDc w ZYPP
-<e,
OOP 3 3 5 '
a m 5, 7
58.3 N'WLnC
yr
g I t It I t I t
e+
2:8 PPPP
g N N 0 0 $ 5
3 <
L'C W N
- - 009-$$ "" 2 c
2 ++++
--N
v
2 a=oarnip 0"
IY.Y.1 -
008 r
Pravastatin (300mgikg/d) Basal
N=5 (3F, 2M)
One Week Rx
Fig. 3. Comparative effects of HMGCoA I-educ-
tase inhibitors in LDL receptordeficient mice. Fe-
male and male mice (number of each indicated
on figure) were fed chow alone (Untreated Con-
trols) or the indicated vastatin for 1 week as a diet
Simvastatin (300 mgikgld) admixtures approximating 300 mg/kg per day
N=6 (3F. 3M)

3
drug.

Downloaded from www.jlr.org by guest, on September 17, 2018


One Week Rx

N=6 (3F. 3M)


Untreated Controls

One-week

0 Time (min) 35

were lethargic at the time of the 2 week post treatment (Table 3 ) . For atorvastatin, the dose-dependent reduc-
bleed. For pravastatin, essentially no reduction in total tion in total cholesterol is largely reflected by reduced
or LDL cholesterol was observed at any dose. For sim- LDL cholesterol (Table 3 ) and to a lesser extent HDL,
vastatin, essentially no reduction in total or LDL choles- and therefore an elevation of the ratio of HDL choles-
terol was observed at the 10 or 30 mg/kg dose. At 100 terol to VLDL plus LDL cholesterol becomes apparenl
mg/kg simvastatin, cholesterol began to redistribute (Table 3). The pooled total cholesterol determinations
from LDL and HDL to VLDL. In the 300 mg/kg simva- (Table 3) were essentially identical to those obtained
statin treatment group, the lipoprotein cholesterol pro- from the average of individual total cholesterol determi-
file of the single surviving morbid mouse showed re- nations. Statistical analysis to determine whether va-
duced total, LDL, and HDL cholesterol and elevation statin treatment results in reduction of the individually
of VLDL cholesterol. The independent total choles- determined total cholesterol (ANOVA of percent
terol determination of the plasma pools (Table 3) and change from pretreatment values) demonstrated signifi-
the percent distribution of total cholesterol from the cant lowering at the two highest atorvastatin doses used
profiles (Fig. 4) were used to determine the average (no treatment, +6%; 10 mgikg, -11%; 30 mgikg,
amount of cholesterol in each lipoprotein fraction for -12%; 100 mg/kg, -15%, P < 0.05; 300 mg/kg, -45%,
the various treatments and are shown in Table 3. In ad- P < 0.0001). No other vastatin tested significantly low-
dition, cholesterol of the combined VLDL plus LDL ered total plasma cholesterol in this experiment.
fraction, the ratio of HDL cholesterol to VLDL plus To determine whether differences in the degree of
LDL cholesterol, and plasma triglycerides are shown total and LDL, cholesterol lowering between the va-

2508 Journal of Lipid Research Volume 38,1997


Dose Atorvastatin Lovastatin Pravastatin Simvastatin
“/kg/d)
- ~-

0
10 (N=7) (N = 7) (N = 7) (N = 7) U
P
co
0
h

30 (N=7) (N = 7) (N = 7) W=7)
2ru
100 (N = 7) (N = 7) (N=7) 3
0

1 300 I ( N = 7 ) m I ( N = 7 ) k (N = 7)
Pre (N=7)
v
4e
2

Downloaded from www.jlr.org by guest, on September 17, 2018


Time (minutes)

Fig. 4. Dose<lependent lipoprotein cholesterol profiles i n female LDL receptor-deficient mice treated with various vastatins for 2 weeks
(group plasma pools). Blood samples from chow-fcd mice were obtained after an 8-11 fast before (open profiles) and after 2 weeks on
chow alone or chow + vastatin diet admixtures approximating 10, 30, 100, and 300 mg/kg per day clriig (shaded profiles). Panel A dis-
plays the hefore (open profile), after (shaded profile), and these profiles overlaid from mice fed chow alone for 2 weeks. Panel 8 displays
the before (open profiles) overlaid on the treatment profile (shaded profiles) for each treatment group. In some cases the baseline id-
lies were essentially identical to treatment values and therefore the overlay of the open profile on the shaded profile is not apparent.
Each profile was generated from a pool containing equivalent amoiints of plasma from each moiise in the group.

statins used in this model were related to the induction cholesterol, triglyceride, and apoB secretion rates were
level of HMGCoA reductase, hepatic mRNA levels assessed after acute (1 day) and chronic (2 weeks) dos-
were determined by a sensitive internal standard/ ing in females by utilizing Triton WR 1339 to block l i p
RNAse protection assay. Female LDL receptordeficient oprotein clearance. We observed a significant 29% re-
mice were bled prior to and after 2 weeks treatment duction in cholesterol secretion in plasma after 1 day of
with vastatins at the 100 mg/kg per day treatment. In 300 mg/kg atorvastatin treatment (Fig. 6A). No signifi-
this experiment, LDL and total plasma cholesterol cant difference in triglyceride o r apoB secretion rates
were significantly lowered with atorvastatin (-41 and was observed (Fig. 6B and 6C). After 2 w e e k treatment,
-27%), lovastatin (-27 and -21%) and simvastatin the total plasma cholesterol and apoB levels were re-
(-22and -15%), but notwith control ( + s a n d +11%) duced by51% (P< 0.0001) and 46% (P< 0.0001) in the
o r pravastatin (+8 and + 12%) treatment, respectively. atowastatin group, respectively, and not changed in the
After 2 weeks of treatment, hepatic HMGCoA reductase control group (Fig. 7). However, with 2 weeks atorvastatin
mRNA levels were elevated 17.2-fold with atorvastatin, treatment, cholesterol (+20%), triglyceride (+57%),
1O.7-fold with lovastatin, 2.5-fold with pravastatin, and and apoB (+31%) secretion rates were significantly ele-
4.1-fold with simvastatin treatment (Fig. 5A). These val- vated (Fig. 6A, 6R, and 6C). Therefore, the secretion ra-
ues were inversely related to the plasma LDL cholesterol tio of triglyceride to cholesterol was significantly elevated
levels (Fig. 5B). An almost identical relationship was o h whether the mice were treated acutely o r chronically
served with total plasma cholesterol (not shown). Inter- with atorvastatin(Fig. 6D).
estingly, a positive relation between the ratio of HDL to To determine whether atorvastatin treatment had ef-
VLDL plus LDL cholesterol with that of HMGCoA re- fects on the molecular forms of apoB secreted, plasma
ductase was also observed (Fig. 5C). samples before o r after Triton WR 1339 treatment were
To explore a possible mechanism of action responsi- analyzed by apoB immunoblot analysis (Fig. 8). In un-
ble for reduced total and LDL cholesterol, especially in treated mice, apoBlOO was predominantly secreted,
the atorvastatin-treated LDL receptordeficient mice, whereas during acute atorvastatin treatment only a p o B

RiTgaiPr ~1 nl. Vastatins reduce plasma LDL in LDL receptordeficient mice 2509
TABLE 3. Dose-dependent plasma lipid changes in femalc IDI, receptor-deficient mice Lreated
with various vastatins for 2 weeks (group plasma pools)
~~~ - ... -~~
".~ ~~. .~

VLDL + LDI. HDI./


Total Cholestrrol VLDL Chnlrsterol LDL C;hnlrstrrol Cholesterol l l D L Cholesterol (VLDL + LDL) Triglycerides
~-~ .~

Group Dose Psc Post Prc PoTt Prc Poyt Pre Post Prr Po\t Pre Post PI-e Po7t
~"

Control 0 224 217 ( - 3 ) (-23)


79 131 118
(-10)
140
125 (-11) 84 92 (10)
0.60
0.74
(24)
(-4)
80
83
Atorvastatin 10 239 206 (-14)7 6 (-12)
149
117
(-21)
155
123
(-21)
84
83 (-1) 0.54 0.68 (26) 81 69 (-15)
30 205 172
(-16)
13
(64)
8 124
95
(-23)
132
108 (-18) 73 64 (-13)0.55 0.59 (7)
94
57
(-39)
100 204 159 (-22) 177 (144) 130 84 (-35) 137 I02 (-26) 67 57 (-15) 0.49 0.57 (15) 72 69 ( - 3 )
300 211 107 (-49) 6 9 (68) 138 51 (-63) 143 61 (-58) 68 46 (-32) 0.47 0.76 (62) 68 72 (6)
Lovastatin 10 218 219 ( 0 ) (-18)
56 121 126
128
(4) 131 (3) 90 88 (-3) 0.71 0.67
(-6) 8'2 (-20)
66
30 216 2'24 (4) 4 8 (76) 131 136 (4) 136 144 (6) 80 80 (-1) 0.59 0.55 (-7) 78 70 (-10)
100 236 197 (-17) 11 7 (64) 149 123 (-17) 156 134 (-14) 80 63 (-22) 0.52 0.47 (-9) 56
69 (-18)
300 214 220 ( 3 ) 8 71 (816) 143 113 (-21) 151 184 (22) 63 36 (-44) 0.42 0.19 (-54) 85 117 (38)
Pravastarin 10 212 219 (3) 8 (-41)
142
4 150 (5) 150 154 (3) (i2 (4)
65 0.41 0.42 (1) 78 (-34)
52
30 228 226 (-1) 7 8 (11) 141 139 (-2) 149 147 (-1) 79 79 (0) 0.53 0.54 (1) 80 63 (-22)
100 224 221 (-1) 9 6 (-33) 145 149 (3) 154 155 (I) 70 66 ( - 6 ) 0.46 0.43 (-7) 77 53 (-31)
300 202 196 ( - 3 ) 9 11 (20) 125 120 (-4) 134 131 (-2) 68 65 (-4) 0.51 0.50 (-2) 95 61 (-36)
Simvastatin 10 232 213 (-8) 7 6 (-6) 155
143 (-8) 162
150 (-8) 70 63 (-9) 0.43 0.42 (-2) 77 56 (-27)
30 227 215 (-5)7 7 (-10) 148 135 (-9) 156 142 (-9) 71 73 (2) 0.46 0.51 (12) 78 53 (-32)
100217 242 (12) 347 (411) 137 146 (7) 143 180 (26) 74 62 (-16) 0.51 0.34 (-33) 77 73 (-5)
300 234 147 (-37) 395 (700) 146 82 (-44) 151 120 (-20) 83 27 (-68) 0.55 0.22 (-60) 65 45 (-31)

Downloaded from www.jlr.org by guest, on September 17, 2018


~~ ~~ ~ ~~-
.. " ~ ~~~~ ."~

Dose-dependent plasma lipid changes in female LDL receptor-deficient mice t r a t c d with vastatin for 2 weeks (group plasma pc)ols). All
treatment groups consisted of sevcn female mice. Mice were fed chow alone or chow containing the indicated vastatin such that appt-oximately
10, 30, 100, or 300 mg drug/kg body wcight was consumed daily. Values represent mg/dI, of plasma pools fronl each group and arc dcrived
from the total cholesterol determination and the lipoproteindistribution o f t h e pools (ix., Fig. 5). Numbers in parentheses reprcscnt percent
change from basal level. For sinwastatin, only one animal survived at the 300 rng/kg per clay dose and the comparison for this single mouse is
made to its pretreatment group pool.

48 was detected. With chronic atorvastatin treatment, Indeed, when levels of atorvastatin were equal to or
both apoB-100 and apoB-48were secreted; however, greater than 30 mg/kg per day, the rise in LDL choles-
apoB-48 was the major form detected. terol observed with time in male mice was quelled, and
furthermore, reduced at the 100 and 300 mg/kg per
day doses. Thus, these studies demonstrated a direct ef-
DISCUSSION fect by an HMGCoA reductase inhibitor on lowering
L,DL cholesterol in the absence of LDL receptors. Even
In the current study, the effects of various vastatins though atorvastatin dose-dependently lowered plasma
on plasma lipoprotein cholesterol in the LDL receptor- total and LDL cholesterol in male LDL receptor-defi-
deficient mouse were determined. It is generally ac- cient mice, with time the effectivenessof thedrug
cepted that HMGCoA reductase inhibitors act prima- waned, suggestive of compensatory mechanisms to
rily to lower plasma LDL cholesterol by up-regulation maintain hepatic cholesterol homeostasis. Possibly,
ofLDL receptors (1, 9, 10). However, lipoprotein ki- P450 induction (37-40) or HMG-CoA reductase over-
netic studies, albeit in LDL receptor-competent mod- production (28) could reverse the effectiveness of the
els, have suggested that HMGCoA reductase inhibitors compound. Indeed, HMGCoA reductase activity and
could, in addition, actby blocking lipoprotein produc- mRNAlevelswere markedly induced in atorvastatin-
tion (11-16). Therefore, we chose to directly test the treated mice.
hypothesis that HMGCoA reductase inhibitors could Interestingly, we observed a marked gender-specific
lower plasma and LDL cholesterol in the LDL receptor- difference in both total and lipoprotein cholesterol in
deficient mouse. As prior studies in both mice and rats these mice. Females appearedto have moreathero-
have suggested marked inactivation of HMGCoA re- genic profiles, having markedly more LDL and less
ductase inhibitors caused by robust P450 enzyme in- HDL cholesterol than males. Therefore, we initially
duction (37-40) and elevation of HMGCoA reductase tested a single dose of atorvastatin (300 mg/kgper
levels (28), it was necessary to use a relatively high dose day) in females, and observed that their response to
(e.g., in comparison to doses used in humans) of the treatment was more profound and longer lasting than
HMGCoA reductase inhibitors to supersede compen- observed in males treated with the same dose. Females
satory mechanisms and to observe a biological effect. also had markedly elevated hepatic HMGCoA reduc-

2510 Journal of Lipid Research Volume 38,1997


Fig. 5. Hepatic HMGCoA reductase mRNA levels in female
LDL receptor-deficient mice treated with various vastatins for 2

10 1 I* weeks. Mice were fed diet admixtures approximating 100 mg/kg


per day of each statin for 2 weeks. Hepatic HMGCoA reductase
levels were determined by an internal standard/RNAse protection
assay as described in Materials and Methods. Blood samples were

* taken before and after treatment and assessed for total and lipo-
protein cholesterol as described in Materials and Methods. (A)
Hepatic HMGCoA reductase mRNA levels; (B) correlation be-
tween hepatic HMG-CoA reductase mRNA level5 and percent
change in LDL cholesterol separated by treatment; and (C) corre-
lation between hepatic HMGCoA reductase mRNA levels and the
percent change in the ratio of HDL cholesterol to VLDL plus
LDL cholesterol separated by treatment (indicated on panel B).
Data in panel A were analyzed by ANOVA. Comparison to control;
*P< 0.005, * * P < 0.0001.
Cont Atorva Lova Prava Simva
B
males. In this regard, studies of Ohtawa and Uchiyama
(37) have shown that hepatic microsomes isolated from
male rats were markedly more active than those iso-
lated from female rats for yielding simvastatin meta-

Downloaded from www.jlr.org by guest, on September 17, 2018


bolic products. Although not directly determined in
the current studies, perhaps the female mice are less
capable of atorvastatin inactivation than males.
We were curious as to whether the hypercholester-
olemic effect in these mice could be observed with
other HMGCoA reductase inhibitors, indicating a class
specific effect. In a 1-week study at the 300 mg/kg per
day dose, we observed a significant total and LDL cho-
lesterol reduction for atorvastatin, lovastatin, and sim-
vastatin, indicating a class specific effect. Therefore,
the relative effectiveness of select vastatins was com-

-6
0
240- Ra.587 pared. Atorvastatin treatment for 2 weeks resulted in
220- significant and dose-dependent lowering of total cho-
lesterol in female LDL receptordeficient mice. Total
i-g 200: 0
cholesterol lowering was largely due to reduction of
LDL cholesterol. Taken together with observations in
males, these reductions are more profound and sus-
z: tained in the females. In contrast to the 1-week experi-
ment at the highest dose, under these experimental
'f
3 0 120
1 4 A0 k
A
conditions (female LDL receptor-deficient mice vasta-
& loo A tin-treated for 2 weeks) and at similar doses, pravasta-
0 I tin, lovastatin, and simvastatin were less effective for
lowering total and non-HDL cholesterol than atorvasta-
60
0 2 4 6 8 10 12 14 16 18 tin and suggest induction of compensation mecha-
nisms or lower efficacies. Interestingly, the efficacy of
HMG CoA Reductase (pg mRNAIFgRNA) the vastatins for lowering total and LDL cholesterol was
inversely related to the level of induction of HMGCoA
reductase mRNA. In that the only major source of cho-
tase activity, as did a few males treated with the drug, lesterol in this model is de novo synthesis and mainte-
and this raises the possibility that females are less effi- nance of cholesterol homeostasis is tightly regulated,
cient than males in inactivating atorvastatin. Measure- the level of HMGCoA reductase mRNA may therefore
ment of plasma drug and metabolite levels, although reflect the relative effectiveness of these compounds.
not determined in the current study, may also shed Lipoprotein cholesterol production was identified as
light on the response differences between males and fe- the initial mechanism by which atorvastatin lowered

Bisgaier et al. Vastatins reduce plasma LDL in LDL receptor-deficient mice 251 1
6
'=
e-
1.2

1 .o

0.8
A
I
p = $0022
I p c o.Ooo1
("2)
I 10
l2 r p c 0.m1

-f 0.6
2,
9s
*E
2- 0.4

0.2

0.0
Control Acute Chronic Control Acute Chronic
o c 20
p = 0.0100
D p = 0.0020

I NS 1
(n=_'2)

Downloaded from www.jlr.org by guest, on September 17, 2018


0.01
Control Acute Chronic Control Acute Chronic

Fig. 6. (A) Cholesterol, (B) triglyceride, ((:) apoB secretion rates and the ratio of the triglyceride to cho-
lesterol secretion rates (D) in atorvastatin-treated female LDL receptor-deficient mice. Mice were fed ii
diet of admixture approximating 300 mg/kg per day atorvastatin acutely (1 day) or chronically (14 days).
Blood samples were obtained before (9 IW) and 12 h post-Triton WR1339 administration. In the chronic
study, blood samples were also obtained before initiation of atorvastatin treatment. Data wet-e derived
from two separate experiments in which the cholesterol and triglyceride secretion rates were measured in
both experiments, and in which the apoB secretion rate was determined in the second experiment. The
secretion rates of cholesterol, triglycerides, and apoB were determined by the difference in accumulated
lipid over 12 h as described in Materials and Methods. Data were analyzed by ANOVA.

total plasma cholesterol in female L,DL receptor- ever, with chronic treatment, mechanisms to compen-
deficient mice. Acute atorvastatin treatment resulted in sate for inhibition of cholesterol de novo synthesis, and
decreased cholesterol production and perhaps contrib- possibly drug inactivation, subsequently led to in-
uted to the reduced levels of LDL cholesterol. How- creased cholesterol production by 2 weeks. Surpris-

300 100 0.10


1A NS p c 0.m1
+-I
B
IC NS

i I ,

-.--
Pre Post Pre Post Pre Post Pre Post Pre post Pre Post
Control Atorvastatin Control Atowastatin Control Atorvastatin
(n=23) (n=22) (n=23) (n=22) (n=l2) (n=12)

Fig. 7. Total plasma cholesterol ( A ) , triglycerides (B), and apoB (C) in the chronic study before and 2 weeks after atorvastatin treat-
ment (i.e., prior to Triton WR1339 administration), Data were analyzed by one-tailed paired I-tests.

2512 Journal of Lipid Research Volume 38,1997


Fig. 8. ApoB immiinoblot analysis of pooled
Control Chronic Acute plasma from control (n = 12), chronic atonmtatin-
Pre- Post- Pre- Post- Pre- Post-
treated (n = 12), and acute atonmstatin-treated (n
Triton Triton Triton Triton Triton Triton = IO) mice before and after Triton M'R 1339 treat-
ment. Plasma proteins (0.7 p,I plasma/lanc) were
separated on 4 1 2 % sodium dodccyl sulfate poly-
acrylamide gel. High molecrrlar weight apoB-100
+ apoB-lOO and low molecuIar weiglit a p o w 8 were detectecl
with a polyclonal sheep and anti-mouse apoR se-
+ apoB-48 rum as described in Materials and Methods.

ingly, the reestablished increased cholesterol synthetic males. With treatment, males, unlike females, appeared
rate appeared to maintain the newly derived plasma to compensate for the drug effects for lowering total
cholesterol level rather than induce an elevation. and LDL cholesterol with time. Therefore, the greater
Thus, it appears that a new steady state plasma choles- response in females may also reflect an inability to inac-
terol level was achieved. In both the acute and chronic tivate the drug as efficiently as males. In a human ho-
studies, the ratio of triglyceride to cholesterol secre- mozygous familial hypercholesterolemia subject with
tion was elevated with atorvastatin treatment, and true receptor negative phenotype, Feher et al. (50) dem-
raises the possibility that production of triglyceride- onstrated that simvastatin monotherapy could sustain
rich cholesterol-poor VLDL may also contribute to the and reduce plasma cholesterol by 30% over approxi-

Downloaded from www.jlr.org by guest, on September 17, 2018


LDL cholesterol-lowering effects observed. mately 1 month of treatment. Although the cholesterol-
The observation that both acute and chronic atorva- lowering effect in this subject was sustained over the fol-
statin treatment resulted in a greater proportion of lowing 20 months, combination therapies were added
apoR48 secreted relative to apoB-100 may also contrib- during this time. In a preliminary report, Naoumova et
ute to the reduced production of LDL. ApoR48 parti- al. (51) showed that atorvastatin augmentation to usual
cles produced in the mice are cleared efficiently therapy in homozygous familial hypercholesterolemic
through a non-LDL receptor-mediated mechanism humans (bi-weekly LDL apheresis in seven subjects, no
(41). Similarly, clearance of apoR48containing lipo- other treatment in three subjects) over 2 months re-
proteins occurred normally in familial hypercholester- duced LDL cholesterol by 31 %. Furthermore, these in-
olemic humans (42) and WHHL rabbits (43). The re- vestigators (51) reported that using a combined ther-
duced secretion of apol3-100 may result from increased apy consisting of LDL apheresis, bile acid sequestration,
apoB mRNA editing (44), an enhanced intracellular and atorvastatin achieved a 70% reduction in LDL cho-
degradation (45), o r decreased synthesis when choles- lesterol. Despite the limited data regarding vastatin
terol becomes limiting for lipoprotein production. Al- treatment of humans with homozygous familial hyper-
ternatively, the apoR48containing lipoproteins that cholesterolemia, the above observations, taken together
accumulate with Triton WR 1339 treatment may r e p with the data presented in the current studies, suggest
resent a disproportionate contribution of particles that pharmacological inhibition of cholesterol produc-
from the intestine. During chronic atorvastatin treat- tion can lead to a reduction of plasma cholesterol in the
ment, when cholesterol secretion increased above con- absence of LDL recept0rs.l
trol levels, the apoB-48 still represented a significant
portion of the secreted apoR. Interestingly, in studies in
rabbit5 (46,47) and mice (48), when apoB mRNA edit- T h e authors acknowledge P a d Schultz and Steven Gray of
ing was increased by adenovirus gene therapy, a re- Parke-Davis for their help in the cloning of HMGCoA redric-
duced LDL production was observed. Under condi- tase and J a n e t Riley and Sandra Holmes, also o f Parke-Davis,
tions where only apoJ3-100 is made, such as in mice for veterinary assistance in maintaining the mouse colony. The
deficient in apoR editing, LDL levels increase (49). It authors gratefully acknowledge the generosity of Drs. S. Ishiba-
should be noted that in humans, unlike mice, apoB-100 shi, M. S.Brown, J. L. Goldstein, R. D. Gerard, R. E. Hammer,
is the only apoR produced in the liver (18), and per- D. K. Burns, and J. H e n of the University of Texas Southwest
Medical Research Foundation for allowing the distribution of
haps a direct effect of a vastatin on apoE-100 produc-
LDL receptor-deficient mice created in their laboratory to the
tion odd also lead to reduced LDL levels. scientific community for study. We also gratefully acknowledge
Overall these data suggest that vastatins lower total Bristol-Meyers Squibb for providing pravastatin and Merck,
and LDL cholesterol in LDL receptordeficient mice. Sharpe & Dohme for providing simvastatin.
For the vastatins tested, atorvastatin caused a sustained
and greatest plasma cholesterol-lowering effect in fe- Mnntrsrripl wwivpd 12 Mrq I997 rind in mii.rPdJm I9 A q ~ s 1997.
l

Bi.vgnim d nl. Vastatins reduce plasma LDL in LDL receptordeficient mice 2513
REFERENCES plications for the pathophysiology of apoB production. ,/.
lipid. RES.31: 567-582.
16. Arad,Y., R. Ramakrishnan, and H. N. Ginsburg. 1992. 1
1. Brown, M. S., and J. L. Goldstein. 1986. A receptor- fects of lovastatin therapy on very low density lipoprotein
mediated pathway for cholesterol homeostasis. Sciw/ce. triglyceride metabolism in subjects with combined hypcr-
232: 34-37. lipidemia: evidence for reduced assembly and secretion
2. Goldstein, J. L., and M. S. Brown. 1989. Familial hyper- of triglyceride-rich lipoproteins. Metabolism. 41: 487-493.
cholesterolemia. In The Metabolic Basis of Inherited Dis- 17. Shiomi, M., M. Shiraishi, T. Yata, and T. Ito. 1994. Effect
eases. c:. R. Scriver, A. L. Beaudet, W. S. Sly, arid D. Valle, of fluvastatin sodium on secretion of very low density lipo-
editors. McCraw-Hill, New York. 1215-1250. protein and serum cholesterol levels. ,4rznri7ti.-I:orscl,/
3. Patsch, W.,J. R. Patsch, and A. M. < h t t o , J r .1989. The hy- Drugfix 44(11),10: 1154-1156.
perlipoproteinemias. Mud. Clin. iVor//i Am. 73: 859-893. 18. Davidson, N. O., S. Anant, arid A. .J. MacGinnitie. 1995.
4. Davidson, B. L., E. D. Allen, K. F. Kwarsky,,J. M. Wilson, Apolipoprotein B messenger RNA editing: insights i n to
and B. J. Roessler. 1993. A model system for in vivo gene the niolecular regulation of post-transcriptiotial cytidine
transfer to the central nervous system using an adenoviral deamination. Cur%@in. Lipidol. 6: 70-74.
vector. Nalure Gunel. 3: 2 19-223. 19. Ishibashi, S., M. S. Brown,J. 1,. Goldstein, K. D. Gerard,
!I. Herz, J.. and R. D. Gerard. 1993. Adetiovinis-mediated K.E. Hammer, and J. Herz. 1993. Hypercholesterolemia
transfer of low density lipoprotein receptor gene acutely in low density lipoprotein receptor knockout mice and its
accelerates cholesterol clearance in norrnal micc. I’ror. reversal by adenovirus-mediated gene delivery.,j . Cliti. In-
N d l . Arnd. Sri. USA. 90: 2812-2816. UP,!/. 92: 883-893.
ti. Kozarsky, K., M. Grossman, and J. M. Wilson. 1993. 20. Ishibashi, S., .J. I>. Goldstein, M. S. Brown, J. Herr, and
Adenovirus-mediated correction of the genetic defect in D. K. Burns. 1994. Massive xanthomatosis and atheroscle-
hepatocytes from patients with Familial hypercholester- rosis in cholesterol-fed low density lipoprotein receptoi.-

Downloaded from www.jlr.org by guest, on September 17, 2018


olemia. Somat. Cull Mol. Gen. 19: 449-4.58. negative mice.J: Clin. hursi. 93: 1885-1893.
7. Kozarsky, K. F., D. R. McKinley, I,. I.. Austin, S. E. Raper, 21. Roth, B. D., (:. J. Blankley, A. W. Chucholowski, E.
I.. I). Stratford-Rerricaudet, atidJ. M. Wilson. 1994. I n vivo Fergusin, M. I,. Hoefle, D. F. Ortwine, R. S. Newton, C. S.
correction of low density lipoprotein receptor deficiency Sekerke, D. R. Sliskovic, C. D. Stratton, and M. W. M’ilson.
i n the Wataiiabe heritable hyperlipidemic rabbit with re- 1991. Inhibitors of cholesterol biosynthesis. 3. Tctrahydixr
combinant aden0viruses.J. Biol. Churn. 269:13695-13702. 4-hydroxy-6-[2-(1H-pyrrol-l-yl)etliyl]-2H-pyran-2~~11e in-
8. Thompson, C;. R. 1989. History and evolution of extracor- hibitors of HMCXoA reductase. 2 . Effects o f introducing
poreal LDL elimination in severe hvpercholesteroleniia. substituents at positions three and four of‘the pyrrole 1111-
Treatment of severe hypercholesterolemia i n the preven- cleus. ,J. M e t . Clirm. 34: 357-366.
tion of coronary heart disease. Proceedings of the Second 22. Bocan, T. M. A,, E. Ferguson, W. McNally, 1.’ 1).
Symposium. 164-169. Uhlendorf, S. B. Mueller, P. Dehart, I). K. Sliskovic, B. D.
9. Bilheirncr, D. W., S. M. Grundy, M. S. Brown, and J. I,. lioth, B. R. Krause, and K.S.Newton. 1992. Hepatic and
Goldstein. 1983. Mevinoliri arid colcstipol stimulatc nonhepatic sterol synthesis arid tissue distribution follow-
t.cceptor-mediated clearance of low density lipoprotein ing administration of a liver specific HMGCoA reductasc
troni plasma in familial hypercholesterolemia heterory- inhibitor, CI-981: comparison with selected HMGCoA re-
gotes. I’rot.. Null. Arnd. . USA. 80: 4124-4128. ductase inhibitors. Uiochim. Biqidiys. Acta. 1123: 133-144.
IO. ReihnCr, E., M. Rudlirig, D. Stahlberg, L. Berlund, S. 23. Allain, C. C., I.. S. Poon, (:. S. C;. Chan, W. Richrnond,
Ewerth, I. Bjorkhem, K. Einarsson, and B. Angelin. 1990. and P. C. Fit. 1974. Enzymatic determination or total sc-
Influence of pravastatin, a specific inhibitor of HMGCoA r-um cholesterol. (Xn.O’hem. 20: 470-475.
I-eductase, on hepatic metabolism of cholesterol. N. E??$ 24. Auerbach, B. J., J. S. Parks, and I). Applebauni-Bowderi.
1. Mrd. 323: 224-228. 1990. A rapid and sensitive micro-assay for the e n q ” c
11. Aiierbach, B.,J.,B. K. Krause, C . L. Bisgaier, and R. S. N e w detemiin;ition of plasma and lipoprotein cholesterol. ,/.
1011. 1995. (:ornparativc effects of HMCX:oA reductase in- 1.ipit. Res. 31: 738-742.
hihitors on apoB production in the caseiri-fed rabbit: ator- 25. Kieft, K. A., T. M. A. Bocan, and B. K. Krause. 1991. Rapid
~ ~ I S t ~Ver‘SUS ~ d l / l ~ ( l c t l ~ O , S i115:
~ l i l ~OvdstdtiIl. , j . 173-1 80. oti-line deterrnination of cholesterol distribution ainong
12. Gntndy, S. M., and C.. L. Vega. 1985. Influerice of inrvirio- plasma lipoproteins after high-performance gel-filtratioii
lin on metdbohni of low density lipoproteins in primary chromatography. J. lipid. Res. 32: 859-866.
moderate hypercholesteroleniia. ,/. 1,ifid. RKS.26: 1464- 26. Aalto-Sedla, K., (1. I,. Bisgaier, A. Ho, K. A. heft, M. G.
1475. ‘G-atjer, H. J. Kayden, R. Rarnakrishnan, A. \iValsli, A. I).
13. Vega, C;. L., C. East, and S . M. Grundy. 1988. Lovastatiii Essenburg, and J . L,. Breslow. 1994. Intestinal expressiott
therapy in familial dysbetalipoproteinemia: effects on ki- of human apolipoprotein A-n’in transgenic mice fails t o
netics of apolipoprotein B. AthomrlPrr,.ris. 70: 131-143. influencr dietary lipid absorption of feeding behavior. ,/.
14. Vega, G. L., C . East, and s. M. GruI1dy. 1989. Effects of f,’lin. /mi,?jl. 93: 1776-1 786.
combined therapy with lovastatin and colestipol in het- 27. Haubenwallner, S., A. D. Essenburg, B. (;. Rarnctt, M . l:,.
erozygous Familial hypercholesterolemia. Effects o n ki- Papc, K. B. DeMattos, B. R. Krause, L,. I,. Minton, 13. .J.
netics~of apolipoprotein B. Art~riosrlrr.osis.SupplementI 9: iluerbach, R. S. Newton, ‘I:Leff, and <:.L. Bisgaier. 1995.
1-1 35-1-144, Hypolipidemic activity of select fibrates corre1att.s t o
15. Arad, Y, R. Ramakrishnan, and H. N. (;insburg. 1990. changes in hepatic apolipoprotein (XI1 expression: a po-
1.ovast;itin therapy reduces low densitv lipoprotein apoB trntial physiologic basis Tor their mode of action. ,j. I.i/>itf.
levels i n subjects with combined hyperlipidemia by reduc- 1Cr.c.36: 2541-25.51.
ing the production of apoB-containing lipoproteiris: im- 28. Kita, T., M. S. Brown, and]. L. Goldstein. 1980. Feedhack

2514 Journal of Lipid Research Volume 38, 1997


regulation of %hydroxy-3methylglutaryl-coenzyme A re- ing the low density lipoprotein receptor, apolipoprotein E
ductase in livers of mice treated with mevinolin, a competi- or both proteins. Proc. Nall. Acad. Sci. USA. 91: 4431-4435.
tive inhibitor of the reductase.J Clzn. Invest. 6 6 1094-1100. 42. Rubinsztein, D. C., J. C. Cohen, G. M. Berger, D. R. van
29. Chomczyski, P., and N. Sacchi. 1987. Single-step method der Westhuyzen, G. A. Coetzee, and W. Gevers. 1990. Chy-
of RNA isolation by acid guanidinium thiocyanate- lomicron remnant clearance from the plasma in familial
phenol-chlorofotm extraction. Anal. Bwchem. 162: 156-159. hypercholesterolemic homozygotes with defined recep-
30. Helmberg, A., R. Fassler, S. Geley, IL Johrer, G. Kroemer, tor defects.]. Clin. Inve.rt. 86: 1306-1312.
G. Bock, and R. Kofler. 1990. Glucocorticoid-regulated 43. Kita, T., J. L. Goldstein, M. S. Brown, Y. Watanabe, C. A.
gene expression in the immune system: analysis of gluco- Hornick, and R. J. Havel. 1982. Hepatic uptake of chylo-
corticoid-regulated transcripts from the mouse macro- micron remnants in WHHL rabbits: a mechanism geneti-
phage like cell line P388Dl.J. Immunol. 145: 4332-4337. cally distinct from the lowdensity lipoprotein receptor.
31. Rea, T. J., R. B. Demattos, and M. E. Pape. 1993. Hepatic ?’roc. Natl. Acad. Sci. USA. 79: 3623-3627.
expression of genes regulating lipid metabolism in rab- 44. Teng, B., C. E Burant, and N. 0. Davidson. 1993. Molecu-
bits.,!. Lipid Rps. 34: 1901-1910. lar cloning of an apolipoprotein B messenger RNA edit-
32. Pape, M. E., G. W’. Melchior, and K. R. Marotti. 1991. ing protein. Science. 260: 1816-1819.
mRNA quantitation by a simple and sensitive RNAse pro- 45. Ginsberg, H. N. 1997. Role of lipid synthesis, chaperone
tection assay. Genetic. Anal. 8: 206-213. proteins and proteasomes in the assembly and secretion
33. Otway, S., and D. S. Robinson. 1967. The use of a non- of apoprotein B-containing lipoproteins from cultured
ionic detergent (Triton WR 1339) to determine rates of liver cells. Clin. Exp. Pharmacal. Physiol. 24: A29-32.
triglyceride entry into the circulation of the rat under dif- 46. Kozarsky, K E, D. K. Bonen, F. Giannoni, T. Funahashi,
ferent physiological conditions.]. Physiol. 190: 321-332. J. M. Wilson, and N. 0. Davidson. 1996. Hepatic expres-
34. Sirtori, C. R., P. Camarasca, G. D’atri, S. Cerutti, G. sion of the catalytic subunit of the apolipoprotein B
Tronconi, and C. Scolastico. 1978. Pharmacological pro- mRNA editing enzyme (apobec-1) ameliorates hypercho-

Downloaded from www.jlr.org by guest, on September 17, 2018


file of BR-931, a new hypolipidemic agent that increases lesterolemia in LDL receptordeficient rabbits. Hem. &ne
high-density lipoproteins. Artherosclerosis. 30: 45-56. Thm 7: 943-957.
35. Towbin, H., T. Staehelin, and J. Gordon. 1979. Electro- 47. Greeve, J., V. K. Jona, N. R. Chowdhury, M. S . Honvitz,
phoretic transfer of proteins from polyacrylamide gels to and J. R. Chowdhury. 1996. Hepatic gene transfer of the
nitrocellulose sheets: procedure and some applications. catalytic subunit of the apolipoprotein B mRNA editing
Prw. Natt. Acad. Sci. USA. 7 6 4350-4354. enzyme results in a reduction of plasma LDL levels in
36. Harlow, E., and D. Lane. 1988. Immunoblotting. In Anti- normal and Watanabe heritable hyperlipidemic rabbits. J.
bodies: A Laboratory Manual. Cold Spring Harbor Labo- Lifiid. Res. 37: 2001-2017.
ratory, NewYork. 471-510. 48. Teng, B., S. Blumenthal, T. Forte, N. Navaratnam,J. Scott,
37. Ohtawa, M., and N. Uchiyama. 1992. Sex difference in A. M. Gotto,Jr., and L. Chan. 1994. Adenovirus-mediated
metabolism of simvastatin by rat hepatic microsomes. EUK gene transfer of rat apolipoprotein B mRNA-editing pro-
J. [>rugMetah. Pharmacokinet. 17: 175-181. tein in mice virtually eliminates apolipoprotein B-100 and
38. Greenspan, M. D., J. B. Yudkovitz, A. W. Alberts, L. S. Ar- normal low density lipoprotein production. ,I. Bid. (;hem.
genhright, B. H. Arison, and J. L. Smith. 1988. Metabo- 269 29395-29404.
lism of lovastatin by rat and human liver microsomes in 49. Morrison, J. R., C. Paszty, M. E. Stevens, S. D. Hughes, T.
vitro. I h u g Metoh. Dispo.s. 16: 678-682. Forte, J. Scott, and E. M. Rubin. 1996. Apolipoprotein B
Vyas, K. P., P. H. Kari, S. M. Pitzenberger, R. A. Halpin, RNA editing enzyme-deficient mice are viable despite al-
H. G. Ramjit, B. Arison,J. S. Murphy, W. F. Hoffman, M. S. terations in lipoprotein metabolism. Prof. N d . Amd. Sci.
Schwartz, E. H. Ulm, and D. E. Duggan. 1990. Biotrans- USA. 93: 7154-7159.
formation of lovastatin. 1. Structure elucidation of in vitro 50. Feher, M. D., J. C. Webb, D. D. Patel, A. F. Lant, P. D.
and in vivo metabolites in the rat and moiisc. h g Metab. Mayne, B. L. Knight, and A. K. Soutar. 1993. Cholesterol-
Di.spo.r. 18: 203-2 1 1. lowering drug therapy in a patient with receptor-negative
40. Vyas, K. P., P. H. Kari, S. R. Prakash, and D. E. Duggan. homozygous familial hypercholesterolemia. Athmoschosis.
1990. Biotransformation of lovastatin. 11. In vitro metabo- 103: 171-180.
lism by rat and mouse liver microsomes and involvement 51. Naoumova, R. P., D. Marais,J. C. Firth, C. K. Y. Neuwirth,
of cytochrome P-450 in dehydrogenation of lovastatin. G. W. Taylor, and G. R. Thompson. 1996. Atorvastatin
Drug Metal). Dispos. 18: 218-222. augments therapy of homozygous familial hypercholes-
41. Ishibashi, S.,J. Herz, N. Maeda, J. L. Goldstein, and M. S. terolemia by inhibiting upregulation of cholesterol syn-
Brown. 1994. The twc-receptor model of lipoprotein thesis after apheresis and bile acid sequestrants. Cirrulcl-
clearance: tests of the hypothesis in “knockout”mice lack- tion (Suppl.). 94: 1-583 (Abstract).

Bisgaier et al. Vastatins reduce plasma LDL in LDL receptor-deficient mice 2515

Anda mungkin juga menyukai