the journal of biological chemistry © 2003 by the … 2… · navdar sever‡, bao-liang song‡,...

12
Insig-dependent Ubiquitination and Degradation of Mammalian 3-Hydroxy-3-methylglutaryl-CoA Reductase Stimulated by Sterols and Geranylgeraniol* Received for publication, September 10, 2003, and in revised form, October 9, 2003 Published, JBC Papers in Press, October 16, 2003, DOI 10.1074/jbc.M310053200 Navdar Sever‡, Bao-Liang Song‡, Daisuke Yabe‡, Joseph L. Goldstein§, Michael S. Brown, and Russell A. DeBose-Boyd From the Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9046 The endoplasmic reticulum enzyme 3-hydroxy-3-me- thylglutaryl-CoA reductase produces mevalonate, which is converted to sterols and to other products, including geranylgeraniol groups attached to proteins. The enzyme is known to be ubiquitinated and rapidly degraded when sterols and nonsterol end products of mevalonate metabolism accumulate in cells. Here, we use RNA interference to show that sterol-accelerated ubiquitination of reductase requires Insig-1 and Insig-2, membrane-bound proteins of the endoplasmic reticu- lum that were shown previously to accelerate degrada- tion of reductase when overexpressed by transfection. Alanine substitution experiments reveal that binding of reductase to Insigs and subsequent ubiquitination re- quire the tetrapeptide sequence YIYF in the second membrane-spanning helix of reductase. The YIYF pep- tide is also found in the sterol-sensing domain of SCAP, another protein that binds to Insigs in a sterol- stimulated fashion. When lysine 248 of reductase is sub- stituted with arginine, Insig binding persists, but the reductase is no longer ubiquitinated and degradation is markedly slowed. Lysine 248 is predicted to lie immedi- ately adjacent to a membrane-spanning helix, suggest- ing that a membrane-bound ubiquitin transferase is re- sponsible. Finally, we show that Insig-dependent, sterol- stimulated degradation of reductase is further accelerated when cells are also supplied with the 20- carbon isoprenoid geranylgeraniol, but not the 15- carbon farnesol, raising the possibility that the non- sterol potentiator of reductase regulation is a geranylgeranylated protein. The enzyme 3-hydroxy-3-methylglutaryl-CoA reductase (HMG-CoA reductase) 1 catalyzes the two-step reduction of HMG-CoA to mevalonate, a crucial intermediate in the synthe- sis of cholesterol and nonsterol isoprenoids (1). Mevalonate- derived products include ubiquinone, heme, and the farnesyl and geranylgeranyl groups that become attached to many pro- teins, directing them to membranes. HMG-CoA reductase is subject to tight regulation by a multivalent feedback mecha- nism mediated by nonsterol and sterol end products of meval- onate metabolism (2). These end products decrease reductase activity by inhibiting transcription of the reductase gene, block- ing translation of the reductase mRNA, and accelerating deg- radation of the reductase protein. The transcriptional effects are mediated through the action of sterol regulatory element- binding proteins (SREBPs), membrane-bound transcription factors that enhance transcription of genes encoding choles- terol biosynthetic enzymes and the low density lipoprotein receptor (3). The translational effects are mediated by non- sterol mevalonate-derived isoprenoids, which act by an unde- fined mechanism (4). The accelerated degradation of reductase is mediated both by sterols and nonsterol end products of mevalonate metabolism (4, 5) and appears to be carried out by the 26 S proteasome (6, 7). Hamster HMG-CoA reductase is an endoplasmic reticulum (ER) resident glycoprotein of 887 amino acids and consists of two distinct domains (8). The NH 2 -terminal domain of 339 amino acids contains eight membrane spanning regions sepa- rated by short loops (9); it anchors the protein to ER mem- branes. The COOH-terminal domain of 548 amino acids projects into the cytoplasm and exerts all of the catalytic activ- ity (8). Two lines of evidence indicate that the membrane do- main is crucial for sterol-accelerated degradation of the en- zyme: 1) the truncated cytoplasmic COOH-terminal domain, expressed by transfection, retains full catalytic activity and exhibits a long half-life that is not shortened by sterols (10); and 2) the transmembrane domain of reductase fused to soluble -galactosidase exhibits sterol-regulated degradation in a man- ner similar to wild-type reductase (11). Sterol-accelerated deg- radation of reductase occurs in the ER and is blocked by inhib- itors of the 26 S proteasome, which lead to the accumulation of ubiquitinated forms of the enzyme (6, 7). A series of studies in the yeast Saccharomyces cerevisiae support a role for the ubiquitin-proteasome pathway in the control of HMG-CoA reductase degradation (12). HMG2p, one of two reductase isozymes expressed in yeast, undergoes accel- erated degradation when flux through the mevalonate pathway is high (12, 13). Degradation of HMG2p is dependent upon a membrane-bound ubiquitin ligase complex that interacts with * This work was supported in part by National Institutes of Health Grant HL20948 and grants from the Perot Family Foundation and W. M. Keck Foundation. The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. ‡ These authors contributed equally to this work. § To whom correspondence may be addressed. E-mail: joe.goldstein@ utsouthwestern.edu. To whom correspondence may be addressed. E-mail: mike.brown@ utsouthwestern.edu. Recipient of National Institutes of Health Mentored Minority Fac- ulty Development Award HL70441. 1 The abbreviations used are: HMG-CoA, 3-hydroxy-3-methylglutaryl coenzyme A; CHO, Chinese hamster ovary; ER, endoplasmic reticulum; GG-OH, geranylgeraniol; LPDS, lipoprotein-deficient serum; PP, pyro- phosphate; RNAi, RNA interference; siRNA, small-interfering RNA; SREBP, sterol regulatory element-binding proteins; SCAP, SREBP cleavage-activating protein; VSV-G, vesicular stomatitis virus glycopro- tein; CMV, cytomegalovirus; TM, transmembrane; HA, hemagglutinin; FCS, fetal calf serum. THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 278, No. 52, Issue of December 26, pp. 52479 –52490, 2003 © 2003 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in U.S.A. This paper is available on line at http://www.jbc.org 52479

Upload: others

Post on 15-Nov-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: THE JOURNAL OF BIOLOGICAL CHEMISTRY © 2003 by The … 2… · Navdar Sever‡, Bao-Liang Song‡, Daisuke Yabe‡, Joseph L. Goldstein§, Michael S. Brown¶, and Russell A. DeBose-Boyd

Insig-dependent Ubiquitination and Degradation of Mammalian3-Hydroxy-3-methylglutaryl-CoA Reductase Stimulated bySterols and Geranylgeraniol*

Received for publication, September 10, 2003, and in revised form, October 9, 2003Published, JBC Papers in Press, October 16, 2003, DOI 10.1074/jbc.M310053200

Navdar Sever‡, Bao-Liang Song‡, Daisuke Yabe‡, Joseph L. Goldstein§, Michael S. Brown¶,and Russell A. DeBose-Boyd�

From the Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9046

The endoplasmic reticulum enzyme 3-hydroxy-3-me-thylglutaryl-CoA reductase produces mevalonate,which is converted to sterols and to other products,including geranylgeraniol groups attached to proteins.The enzyme is known to be ubiquitinated and rapidlydegraded when sterols and nonsterol end products ofmevalonate metabolism accumulate in cells. Here, weuse RNA interference to show that sterol-acceleratedubiquitination of reductase requires Insig-1 and Insig-2,membrane-bound proteins of the endoplasmic reticu-lum that were shown previously to accelerate degrada-tion of reductase when overexpressed by transfection.Alanine substitution experiments reveal that binding ofreductase to Insigs and subsequent ubiquitination re-quire the tetrapeptide sequence YIYF in the secondmembrane-spanning helix of reductase. The YIYF pep-tide is also found in the sterol-sensing domain of SCAP,another protein that binds to Insigs in a sterol-stimulated fashion. When lysine 248 of reductase is sub-stituted with arginine, Insig binding persists, but thereductase is no longer ubiquitinated and degradation ismarkedly slowed. Lysine 248 is predicted to lie immedi-ately adjacent to a membrane-spanning helix, suggest-ing that a membrane-bound ubiquitin transferase is re-sponsible. Finally, we show that Insig-dependent, sterol-stimulated degradation of reductase is furtheraccelerated when cells are also supplied with the 20-carbon isoprenoid geranylgeraniol, but not the 15-carbon farnesol, raising the possibility that the non-sterol potentiator of reductase regulation is ageranylgeranylated protein.

The enzyme 3-hydroxy-3-methylglutaryl-CoA reductase(HMG-CoA reductase)1 catalyzes the two-step reduction of

HMG-CoA to mevalonate, a crucial intermediate in the synthe-sis of cholesterol and nonsterol isoprenoids (1). Mevalonate-derived products include ubiquinone, heme, and the farnesyland geranylgeranyl groups that become attached to many pro-teins, directing them to membranes. HMG-CoA reductase issubject to tight regulation by a multivalent feedback mecha-nism mediated by nonsterol and sterol end products of meval-onate metabolism (2). These end products decrease reductaseactivity by inhibiting transcription of the reductase gene, block-ing translation of the reductase mRNA, and accelerating deg-radation of the reductase protein. The transcriptional effectsare mediated through the action of sterol regulatory element-binding proteins (SREBPs), membrane-bound transcriptionfactors that enhance transcription of genes encoding choles-terol biosynthetic enzymes and the low density lipoproteinreceptor (3). The translational effects are mediated by non-sterol mevalonate-derived isoprenoids, which act by an unde-fined mechanism (4). The accelerated degradation of reductaseis mediated both by sterols and nonsterol end products ofmevalonate metabolism (4, 5) and appears to be carried out bythe 26 S proteasome (6, 7).

Hamster HMG-CoA reductase is an endoplasmic reticulum(ER) resident glycoprotein of 887 amino acids and consists oftwo distinct domains (8). The NH2-terminal domain of 339amino acids contains eight membrane spanning regions sepa-rated by short loops (9); it anchors the protein to ER mem-branes. The COOH-terminal domain of 548 amino acidsprojects into the cytoplasm and exerts all of the catalytic activ-ity (8). Two lines of evidence indicate that the membrane do-main is crucial for sterol-accelerated degradation of the en-zyme: 1) the truncated cytoplasmic COOH-terminal domain,expressed by transfection, retains full catalytic activity andexhibits a long half-life that is not shortened by sterols (10);and 2) the transmembrane domain of reductase fused to soluble�-galactosidase exhibits sterol-regulated degradation in a man-ner similar to wild-type reductase (11). Sterol-accelerated deg-radation of reductase occurs in the ER and is blocked by inhib-itors of the 26 S proteasome, which lead to the accumulation ofubiquitinated forms of the enzyme (6, 7).

A series of studies in the yeast Saccharomyces cerevisiaesupport a role for the ubiquitin-proteasome pathway in thecontrol of HMG-CoA reductase degradation (12). HMG2p, oneof two reductase isozymes expressed in yeast, undergoes accel-erated degradation when flux through the mevalonate pathwayis high (12, 13). Degradation of HMG2p is dependent upon amembrane-bound ubiquitin ligase complex that interacts with

* This work was supported in part by National Institutes of HealthGrant HL20948 and grants from the Perot Family Foundation andW. M. Keck Foundation. The costs of publication of this article weredefrayed in part by the payment of page charges. This article musttherefore be hereby marked “advertisement” in accordance with 18U.S.C. Section 1734 solely to indicate this fact.

‡ These authors contributed equally to this work.§ To whom correspondence may be addressed. E-mail: joe.goldstein@

utsouthwestern.edu.¶ To whom correspondence may be addressed. E-mail: mike.brown@

utsouthwestern.edu.� Recipient of National Institutes of Health Mentored Minority Fac-

ulty Development Award HL70441.1 The abbreviations used are: HMG-CoA, 3-hydroxy-3-methylglutaryl

coenzyme A; CHO, Chinese hamster ovary; ER, endoplasmic reticulum;GG-OH, geranylgeraniol; LPDS, lipoprotein-deficient serum; PP, pyro-phosphate; RNAi, RNA interference; siRNA, small-interfering RNA;SREBP, sterol regulatory element-binding proteins; SCAP, SREBP

cleavage-activating protein; VSV-G, vesicular stomatitis virus glycopro-tein; CMV, cytomegalovirus; TM, transmembrane; HA, hemagglutinin;FCS, fetal calf serum.

THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 278, No. 52, Issue of December 26, pp. 52479–52490, 2003© 2003 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in U.S.A.

This paper is available on line at http://www.jbc.org 52479

Page 2: THE JOURNAL OF BIOLOGICAL CHEMISTRY © 2003 by The … 2… · Navdar Sever‡, Bao-Liang Song‡, Daisuke Yabe‡, Joseph L. Goldstein§, Michael S. Brown¶, and Russell A. DeBose-Boyd

HMG2p and mediates its ubiquitination in response to degra-dative signals (14).

Although accelerated degradation of mammalian reductaseand that of yeast HMG2p seem similar, major differences exist.Although the membrane domain of yeast HMG2p includes mul-tiple membrane spanning segments (13), it bears no primarysequence resemblance to that of mammalian reductase (8, 15).Moreover, accelerated degradation of HMG2p in yeast is trig-gered maximally by a nonsterol isoprenoid (16), although max-imal degradation of reductase in mammalian cells requiressterols plus nonsterol isoprenoids (4, 5).

Recent clues to the mechanism of degradation of mammalianHMG-CoA reductase have emerged from the study of the tran-scriptional axis of sterol regulation as mediated by SREBPs.The key discovery was SREBP cleavage-activating protein(SCAP), a polytopic membrane protein that resembles the re-ductase by virtue of eight membrane spanning segments.SCAP is a sterol-responsive escort protein that forms a complexwith SREBPs and escorts them from the ER to the Golgi whereSREBPs are proteolytically released from membranes, therebyallowing transcriptionally active fragments to enter the nu-cleus where they activate the reductase gene (3, 17, 18). Sterolsblock SREBP activation by preventing the exit of SCAP-SREBP complexes from the ER, leading to decreased synthesisof cholesterol and other lipids. The block in SREBP export isachieved through sterol-induced binding of SCAP to one of apair of ER retention proteins called Insig-1 and Insig-2 (19, 20).The binding site for Insig in SCAP has been localized to a�170-amino acid segment that comprises transmembrane seg-ments 2–6. These segments show significant sequence similar-ities to helices 2–6 of the membrane domain of HMG-CoAreductase, and this region has been termed the sterol-sensingdomain (21–23). Point mutations within the sterol-sensing do-main of SCAP prevent it from binding to Insigs (19, 20), thuspreventing the sterol-mediated ER retention of SCAP/SREBP(18, 24, 25).

The resemblance between the sterol-sensing domains ofSCAP and HMG-CoA reductase led us to test the possible roleof Insig-1 in the sterol-regulated degradation of the reductase(26). When reductase is overexpressed in Chinese hamsterovary (CHO) cells, the enzyme is no longer degraded rapidly inresponse to sterol treatment. Overexpression of Insig-1 re-stores regulated degradation of overexpressed reductase, andthis degradation can be inhibited by overexpressing the sterol-sensing domain of SCAP, suggesting that reductase and SCAPcompete for limiting amounts of Insig when sterol levels arehigh. Although both proteins bind to Insig-1, the binding leadsto markedly different consequences: binding leads to ER reten-tion of SCAP and to accelerated degradation of reductase. HowInsigs orchestrate seemingly discordant modes of cellular reg-ulation requires further knowledge of the mechanisms under-lying the sterol-mediated effects on reductase and SCAP. Inthis paper, we explore the role of Insigs, sterols, and nonsterolisoprenoids in regulating degradation of reductase.

EXPERIMENTAL PROCEDURES

Materials—We obtained MG-132 and digitonin from Calbiochem andhorseradish peroxidase-conjugated, donkey anti-mouse IgG (affinitypurified) from Jackson ImmunoResearch Laboratories; farnesol andgeranylgeraniol from Sigma; NB-598 from Banyu Pharmaceutical Co.,Ltd; and hydroxypropyl-�-cyclodextrin from Cyclodextrin Technologies,Inc. (Gainesville, FL). Other reagents were obtained from describedsources (17). Lipoprotein-deficient serum (LPDS, d � 1.215 g/ml) wasprepared from newborn calf serum by ultracentrifugation (27).

Expression Plasmids—The following plasmids were described in theindicated reference: pCMV-Insig-1-Myc, which encodes amino acids1–277 of human Insig-1 followed by six tandem copies of a c-Myc epitopetag under control of the cytomegalovirus promoter (19); pRc/CMV7S-Mev, which encodes amino acids 1–494 of the hamster Mev protein

under control of the CMV promoter (28, 29); and pCMV-HMG-Red-T7(TM1–8), which encodes amino acids 1–346 of hamster HMG-CoAreductase followed by three tandem copies of the T7 epitope tag undercontrol of the CMV promoter (26). pEF1a-HA-ubiquitin (provided by Dr.Zhijian Chen, University of Texas Southwestern Medical Center) en-codes amino acids 1–76 of human ubiquitin preceded by an epitope tagderived from the influenza hemagglutinin (HA) protein (YPYDVPDY)under the control of the EF1a promoter.

pCMV-HMG-Red-T7, which encodes full-length hamster reductase(amino acids 1–887) followed by three tandem copies of the T7-epitopetag (MASMTGGQQMG), was constructed by standard methods. Lysinemutations (K89R, K248R, and K89R/K248R) and sterol sensing-domainmutations (Y75A, Y77A, Y75A/Y77A, I76A, F78A, I76A/F78A, andYIYF-(75–78) to AAAA) in pCMV-HMG-Red-T7 and pCMV-HMG-Red-T7(TM1–8) were generated with the QuikChange XL site-directed mu-tagenesis kit (Stratagene). The integrity of all plasmids was confirmedby DNA sequencing of their open reading frames and ligation joints.

Cell Culture—Monolayers of Chinese hamster ovary cells (CHO-K1)were maintained in tissue culture at 37 °C in 8–9% CO2. Stock culturesof CHO-K1 cells were maintained in medium A (1:1 mixture of Ham’sF-12 medium and Dulbecco’s modified Eagle’s medium containing 100units/ml penicillin and 100 �g/ml streptomycin sulfate) supplementedwith 5% (v/v) fetal calf serum (FCS).

Monolayers of SV-589 cells, an immortalized line of human fibro-blasts expressing the SV40 large T antigen (30), were grown at 37 °C in5% CO2. Stock cultures of SV-589 cells were maintained in medium B(Dulbecco’s modified Eagle’s medium containing 100 units/ml penicillinand 100 �g/ml streptomycin sulfate) supplemented with 10% FCS.SV589/pMev cells are derivatives of SV-589 cells stably expressingpMev, a cDNA encoding a mutated monocarboxylate transporter thatconfers efficient uptake of mevalonate from tissue culture medium (28).SV-589/pMev cells were generated as follows. On day 0, SV-589 cellswere set up at 2 � 105 cells per 60-mm dish in medium B supplementedwith 10% FCS. On day 1, the cells were transfected with 2 �g ofpRc/CMV7S-Mev using the FuGENE 6 transfection reagent as de-scribed below. Twenty-four hours after transfection, cells were switchedto selection medium that consisted of medium B supplemented with10% LPDS, 50 �M sodium compactin, 200 �M sodium mevalonate, and700 �g/ml G418. Fresh medium was added every 2–3 days until coloniesformed after about 10 days. Individual colonies were isolated withcloning cylinders, and pMev expression was assessed by the ability ofexogenous mevalonate to induce reductase degradation. Once stablytransfected cell lines were established, they were each maintained at37 °C, 5% CO2 in medium B containing 10% LPDS, 50 �M sodiumcompactin, 200 �M sodium mevalonate, and 500 �g/ml G418. Sterolswere added to the culture medium at a final concentration of 0.1 or 0.2%(v/v) ethanol.

Transient Transfection, Cell Fractionation, and Immunoblot Analy-sis—Transfections were performed as described (31) with minor modi-fications. CHO-K1 cells were transfected with 3 �g of DNA per 60-mmdish. For each transfection, FuGENE 6 DNA transfection reagent(Roche Diagnostics) was added to 0.2 ml of medium A at a ratio of 3 �lof FuGENE-6 per 1 �g of DNA. Conditions of incubation are describedin the figure legends. At the end of the incubation, dishes of cells wereharvested and pooled for analysis.

Pooled cell pellets were used to isolate either nuclear extracts, 2 �104-g membrane fractions (26), or whole cell lysate fractions (18). Allfractions were subjected to 8% SDS-PAGE and immunoblot analysis asdescribed (26). Primary antibodies used for immunoblotting were asfollows: mouse monoclonal anti-T7-Tag (IgG2b) (Novagen); mouse mono-clonal anti-Myc (IgG fraction) from the culture medium of hybridomaclone 9E10 (American Type Culture Collection); IgG-A9, a mouse mono-clonal antibody against the catalytic domain of hamster reductase(amino acids 450–887) (32); IgG-1D2, a mouse monoclonal antibodyagainst the NH2 terminus of human SREBP-2 (amino acids 48–403)(20); IgG-P4D1, a mouse monoclonal antibody against bovine ubiquitin(Santa Cruz Biotechnology); IgG-CD71, a mouse monoclonal antibodyagainst the human transferrin receptor (Zymed Laboratories); and IgG-HA-7, a mouse monoclonal antibody against the HA epitope tag(Sigma).

Blue Native-PAGE for Detection of HMG-CoA Reductase-Insig-1Complex—Monolayers of SRD-13A cells, a line of mutant CHO cellsthat lacks SCAP (31), were set up on day 0 (8 � 105 cells/100-mm dish)and cultured in 8–9% CO2 at 37 °C in medium A supplemented with 5%FCS, 5 �g/ml cholesterol, 1 mM sodium mevalonate, and 20 �M sodiumoleate. On day 2, the cells were transfected as described in the legend toFig. 5, after which they were harvested for preparation of a 1.6 � 104-gmembrane pellet, which was solubilized with 1% (w/v) digitonin and

Insig-dependent Ubiquitination of HMG-CoA Reductase52480

Page 3: THE JOURNAL OF BIOLOGICAL CHEMISTRY © 2003 by The … 2… · Navdar Sever‡, Bao-Liang Song‡, Daisuke Yabe‡, Joseph L. Goldstein§, Michael S. Brown¶, and Russell A. DeBose-Boyd

centrifuged as described (19). One aliquot of the resulting 20,000 � gsupernatant was mixed with a 6-amino-n-hexanoic acid containingbuffer and subjected to electrophoresis on a 4–16% blue native gel asdescribed (19). A second aliquot of the 20,000 � g supernatant wasmixed with SDS-containing buffer and subjected to 10% SDS-PAGE.

RNA Interference—The protocol was adapted from Elbashir et al.(33). Duplexes of small-interfering RNA (siRNA) were synthesized byDharmacon Research (Lafayette, CO). siRNA sequences targeting hu-man Insig-1 and human Insig-2 (GenBankTM accession numbersAY112745 and AF527632, respectively) corresponded to the followingnucleotide positions relative to the first nucleotides of the start codons,respectively: Insig-1, 691–711; and Insig-2, 324–344. The siRNA se-quence targeting an irrelevant control gene, vesicular stomatitis virusglycoprotein (VSV-G), was reported (34).

On day 0, SV-589 cells were set up at a density of 5 � 104 cells per60-mm dish in medium B supplemented with 10% FCS. On day 1, thecells were transfected with siRNA using a ratio of 6 �l of OligofectAMI-NETM reagent (Invitrogen) to 400 pmol of siRNA duplexes per dish asdescribed by the manufacturer. Cells were washed once with 2 ml ofmedium B (without antibiotics) and refed with 1.6 ml of medium B(without antibiotics). The siRNA/OligofectAMINE mixture (in a totalvolume of 0.4 ml) was added to each dish and incubated at 37 °C. After6 h, the cells received a direct addition of 1 ml of medium B containing(final concentrations) 10% FCS, 5 �g/ml cholesterol, 1 mM sodiummevalonate, and 20 �M sodium oleate, and were then incubated at37 °C. On day 3, the cells were transfected with siRNA duplexes asdescribed above. Six hours after the final transfection, the cells receiveda direct addition of 1 ml of medium B containing (final concentration)10% LPDS, 50 �M sodium compactin, and 50 �M sodium mevalonate. Onday 4, the cells were treated as described in the figure legends, andharvested for analysis.

Real-time PCR—The protocol was identical to that described byLiang et al (35). Triplicate samples of first-strand cDNA were subjectedto real-time PCR quantification using forward and reverse primers forhuman Insig-1, human Insig-2, and human 36B4 (invariant control)(Table I). Relative amounts of mRNAs were calculated using the com-parative CT method.

Pulse-chase Analysis of HMG-CoA Reductase—Cells were pulse-la-beled in medium C (methionine/cysteine-free Dulbecco’s modified Ea-gle’s medium containing 100 units/ml penicillin, 100 �g/ml streptomy-cin sulfate, 10% LPDS, and 50 �M sodium compactin), and pulse-chaseanalysis was carried out as described (26). Immunoprecipitation oflabeled reductase was carried out with 30 �g of a polyclonal antibodydirected against the 60-kDa COOH-terminal domain of human reduc-tase (36). Immunoprecipitates were subjected to SDS-PAGE and trans-ferred to Hybond C-extra nitrocellulose filters. Dried filters were ex-posed to an imaging plate at room temperature and scanned in a Fuji XBas 1000 Phosphorimager.

Ubiquitination of HMG-CoA Reductase—Conditions of incubationsare described in the figure legends. At the end of the incubation, cellswere harvested, and immunoprecipitations with either polyclonal anti-body against endogenous human reductase (see above) or monoclonalanti-T7 IgG-coupled agarose beads (Novagen) against transfected re-ductase were carried out as described (26) with two modifications: thelysis buffer used to harvest the cells was supplemented with 10 mM

N-ethylmaleimide and the pelleted beads were washed three times (30min each). Immunoprecipitates were subjected to SDS-PAGE on 5–12%gradient gels and transferred to nitrocellulose filters.

RESULTS

The experiment in Fig. 1 was designed to examine the rela-tionship between ubiquitination and sterol-accelerated degra-dation of HMG-CoA reductase. SV589 cells, a line of SV-40-transformed human fibroblasts, were depleted of sterols byincubation for 16 h in LPDS containing the reductase inhibitor,

compactin, and a low level of mevalonate (50 �M), which is thelowest level that assures viability. Cells were then treated foreither 2 or 5 h with a mixture of sterols (1 �g/ml 25-hydroxy-cholesterol and 10 �g/ml cholesterol) and a high concentrationof mevalonate (10 mM) in various combinations. Additionally,some of the cells received the proteasome inhibitor MG-132 toblock the degradation of any ubiquitinated reductase mole-cules. Detergent-solubilized extracts were subjected to immu-noprecipitation with polyclonal antibodies against reductase,and the resulting immunoprecipitates were subjected to SDS-PAGE and blotted with anti-ubiquitin (Fig. 1A, top panel) oranti-reductase (Fig. 1A, lower panel) monoclonal antibodies. Incells treated for 2 h, sterols caused a slight decrease in theamount of reductase. The addition of mevalonate had no effect(lower panel, lanes 1–4). The sterol-dependent decrease of re-

FIG. 1. Sterols promote ubiquitination of endogenous HMG-CoA reductase in SV589 cells. A and B, SV-589 cells were set up forexperiments on day 0 at 2 � 105 cells per 100-mm dish in medium Bwith 10% FCS. On day 2, cells were washed with phosphate-bufferedsaline and refed medium B supplemented with 10% LPDS, 50 �M

sodium compactin, and 50 �M sodium mevalonate. After 16 h at 37 °C,cells were switched to medium B containing 10% LPDS, 50 �M compac-tin, and the indicated additions. A, cells were treated in the absence (�)or presence (�) of sterols (1 �g/ml 25-hydroxycholesterol and 10 �g/mlcholesterol), 10 mM sodium mevalonate, and 10 �M MG-132 as indi-cated. After 2 (lanes 1–8) or 5 h (lanes 9–16) at 37 °C, cells wereharvested, lysed, and subjected to immunoprecipitation with polyclonalanti-reductase as described under “Experimental Procedures.” Aliquotsof immunoprecipitates were subjected to SDS-PAGE, transferred tonylon membranes, and immunoblotted with 5 �g/ml monoclonal IgG-A9(against reductase) or 0.2 �g/ml monoclonal IgG-P4D1 (against ubiq-uitin). Filters were exposed to film for 2 (top gel) and 30 s (bottom gel).B, cells were incubated at 37 °C for the indicated times in the absence(�) or presence (�) of sterols and 10 �M MG132 as indicated, harvested,and subjected to immunoprecipitation and immunoblot analysis as in A.Filters were exposed to film for 30 (top gel) and 10 s (bottom gel).

TABLE INucleotide sequences of gene-specific primers used for quantitative real-time PCR

mRNA Sequences of forward and reverse primers (5� to 3�) GenBank™ accession No.

Insig-1 CCCAGATTTCCTCTATATTCGTTCTT AY112745CACCCATAGCTAACTGTCGTCCTA

Insig-2 TGTCTCTCACACTGGCTGCACTA AF527632CTCCAAGGCCAAAACCACTTC

36B4 TGCATCAGTACCCCATTCTATCA XM_065715AAGGTGTAATCCGTCTCCACAGA

Insig-dependent Ubiquitination of HMG-CoA Reductase 52481

Page 4: THE JOURNAL OF BIOLOGICAL CHEMISTRY © 2003 by The … 2… · Navdar Sever‡, Bao-Liang Song‡, Daisuke Yabe‡, Joseph L. Goldstein§, Michael S. Brown¶, and Russell A. DeBose-Boyd

ductase was blocked by MG-132, (lower panel, lanes 5–8), in-dicating that this decrease reflected accelerated degradationmediated by proteasomes. After 2 h of sterol treatment in thepresence of MG-132, we observed the appearance of ubiquiti-nated reductase as indicated by the high molecular weightsmear in the anti-ubiquitin immunoblots of the reductase im-munoprecipitates (lanes 6 and 8, upper panel), but not in cellstreated with mevalonate alone (lane 7). The addition of meva-lonate did not enhance the sterol effect (compare lanes 6 and 8).After 5 h in the presence of sterols alone, reductase was stilldetectable, although diminished (lower panel, lane 10). In thepresence of sterols plus mevalonate, the reductase was furtherreduced (lower panel, lane 12). This result is consistent withother studies demonstrating that sterols alone accelerate thedegradation of reductase and that nonsterol mevalonate-de-rived products further accelerate degradation (4, 5). Acceler-ated degradation of reductase was completely blocked by MG-132 (lower panel, lanes 13–16). In the presence of MG-132,sterols caused a marked increase in the amount of ubiquiti-nated reductase, and there was no further effect with meval-onate addition (compare lanes 14 and 16 in upper panel of Fig.1A). Mevalonate alone had only a slight effect (upper panel,lane 15), which is likely attributable to the incorporation ofmevalonate into sterols after 5 h.

The sterol stimulation of reductase ubiquitination was re-markably rapid (Fig. 1B). In the absence or presence of theproteasome inhibitor, sterol-dependent ubiquitination of reduc-tase was observed after as little as 5 min of sterol treatment(lanes 2 and 3) and reached a maximum after 10 min (lanes 5and 6). Thereafter, in the absence of MG-132 ubiquitinatedreductase declined, presumably because of degradation (lanes 8and 11). This decline was prevented by MG-132 (lanes 9 and12).

Although the data of Fig. 1 indicate that reductase is ubiq-uitinated and degraded in a sterol-dependent manner, the pro-portion of reductase that was detectably polyubiquitinated atany one time was very small, even when degradation wasblocked by MG-132. Thus, in the presence of MG-132 most ofthe reductase that accumulated was not polyubiquitinated, asindicated by its continued migration on SDS-PAGE as a 97-kDaband (lower panel in Fig. 1). We interpret these findings toindicate a balance between ubiquitination and de-ubiquitina-tion reactions. According to this hypothesis, sterols stimulatethe ubiquitination of reductase. When proteasomal function isintact, most of the ubiquitinated reductase is degraded. Whenproteasomal degradation is blocked, the amount of ubiquiti-nated reductase at any instant reflects an equilibrium betweenubiquitination and de-ubiquitination reactions. Similar conclu-sions have been reached by others in studies of the ubiquitina-tion of other proteins (37). The addition of mevalonate en-hances reductase degradation, but it is not clear whethermevalonate increases reductase ubiquitination or whether itacts to enhance degradation of reductase that has become ubiq-uitinated under the influence of sterols. It is clear, however,that nonsterol mevalonate products cannot act by themselves.They only act in the presence of sterols.

In previous studies of reductase that was overexpressed bytransfection, the sterol-mediated acceleration of degradationwas blunted, but it could be restored by simultaneous overex-pression of Insig-1 (26) or its close relative Insig-2.2 Thesefindings raised the possibility that accelerated reductase deg-radation requires Insig protein. To demonstrate this require-ment directly, we decreased the amount of the two Insigs

through the use of RNA interference (RNAi) in human SV589fibroblasts (Fig. 2). Cells were transfected twice with duplexesof siRNA targeting a control mRNA encoding vesicular stoma-titis virus glycoprotein (VSV-G, lanes 1–4), which is not presentin the cells, Insig-1 (lanes 5–8), Insig-2 (lanes 9–12), or thecombination of Insig-1 and Insig-2 (lanes 13–16). Cells werethen treated for 5 h with sterols and/or mevalonate, and deter-gent extracts were analyzed by immunoblotting with reduc-tase-specific antibody. In control cells receiving VSV-G siRNAduplexes, sterols alone caused a modest, but detectable de-crease in reductase (compare lanes 1 and 2), and this reductionwas complete upon addition of sterols plus mevalonate (lane 4).Similar responses were observed when Insig-1 and Insig-2siRNA duplexes were transfected alone (lanes 5–8 and 9–12,respectively). However, when both Insig-1 and Insig-2 siRNAduplexes were introduced into cells, no degradation of reduc-tase was observed under any conditions (lanes 13–16). As acontrol, we immunoblotted identical extracts for the trans-ferrin receptor, which remained unchanged regardless ofsiRNA transfection or treatment (lower panel, lanes 1–16). Inthe same experiment, we used quantitative real-time PCR toshow that Insig-1 and Insig-2 mRNAs were reduced by 90 and50%, respectively, by RNAi (Fig. 2B). It should be pointed outthat in SV-589 cells Insig-1 mRNA is normally expressed atlevels 10-fold higher than Insig-2 mRNA.

To demonstrate directly that the RNAi knockdown of Insig-1and Insig-2 abolishes sterol-accelerated degradation of reduc-tase, we performed a pulse-chase experiment. Cells transfectedwith siRNA duplexes were pulse-labeled for 1 h with 35S-labeled methionine plus cysteine, after which they werewashed and switched to medium containing an excess of unla-beled methionine and cysteine in the absence or presence ofsterols plus mevalonate. Radiolabeled reductase was moni-tored by immunoprecipitation with anti-reductase antibodiesfollowed by SDS-PAGE and visualization in a phosphorimager.Fig. 2C shows the time course of disappearance of 35S-labeledreductase in cells treated with siRNA targeting VSV-G (upperpanel) or Insig-1 plus Insig-2 (lower panel). In control cellstreated with VSV-G siRNA, sterols plus mevalonate caused arapid disappearance of 35S-labeled reductase that was nearlycomplete after 5 h (upper panel, lanes 5–7). However, in cellstransfected with Insig-1 plus Insig-2 siRNA, labeled reductasecontinued to be present throughout the chase (lower panel,lanes 5–7). These results were quantified by scanning of imagesin a phosphorimager, and the data are plotted in the lowerpanel of Fig. 2C. The half-life of labeled reductase in controlcells declined from 11.5 to 0.9 h upon treatment with sterolsplus mevalonate. In cells treated with siRNAs targeting thetwo Insigs, reductase half-life was 10.4 h in the absence orpresence of sterols plus mevalonate.

We next addressed the question of whether Insigs are nec-essary for the ubiquitination of reductase or whether they arerequired for a post-ubiquitination step. To this end, we trans-fected cells with siRNA duplexes targeting VSV-G (Fig. 2D,lanes 1–4) or Insig-1 plus Insig-2 (lanes 5–8). Cells weretreated for 1 h with sterols and/or mevalonate in the presenceof MG-132. Detergent extracts were immunoprecipitated withanti-reductase polyclonal antibodies and subjected to immuno-blotting with anti-ubiquitin (upper panel) or anti-reductase(lower panel). In control cells treated with VSV-G RNAi, sterolscaused the appearance of ubiquitinated reductase (upper panel,lanes 2, 4), and mevalonate alone had only a slight effect (upperpanel, lane 3). In cells transfected with siRNAs targeting In-sig-1 and Insig-2, sterol-mediated ubiquitination of reductasewas greatly diminished (upper panel, lanes 5–8). Together,

2 N. Sever, B.-L. Song, D. Yabe, J. L. Goldstein, M. S. Brown, andR. A. DeBose-Boyd, unpublished observations.

Insig-dependent Ubiquitination of HMG-CoA Reductase52482

Page 5: THE JOURNAL OF BIOLOGICAL CHEMISTRY © 2003 by The … 2… · Navdar Sever‡, Bao-Liang Song‡, Daisuke Yabe‡, Joseph L. Goldstein§, Michael S. Brown¶, and Russell A. DeBose-Boyd

these results indicate that Insig-1 and Insig-2 are necessary forsterol-induced ubiquitination of reductase.

Ubiquitination of proteins is a multistep process, culminat-ing in the covalent attachment of ubiquitin to the �-aminogroup of a lysine residue in the target protein. The topology ofthe ubiquitination machinery makes it likely that potentialsites of ubiquitination are accessible to the cytosol. The mem-brane domain of reductase is not only necessary, but sufficientfor regulated degradation (10, 11), and we have determined in

transfection experiments that the membrane domain (aminoacids 1–346 corresponding to transmembrane segments 1–8;see Fig. 3A) of reductase retains the ability to undergo Insig-dependent, sterol-regulated degradation (data not shown). Asshown in Fig. 3A, amino acids 1–346 of reductase contain onlytwo lysines that are exposed to the cytosol (positions 89 and248). To ascertain their participation in regulated ubiquitina-tion of reductase, we changed the lysine codons at positions 89and 248 to arginine in pCMV-HMG-Red-T7, an expression

FIG. 2. Insig-1 and -2 are required for regulated degradation and ubiquitination of HMG-CoA reductase as determined by RNAi.SV-589 cells were set up on day 0 at 5 � 104 cells per 60-mm dish in medium B with 10% FCS. On days 1 and 3, cells were transfected with theindicated siRNA as described under “Experimental Procedures.” The total amount of siRNA was adjusted to 400 pmol/dish by addition of VSV-GsiRNA. After the second transfection on day 3, cells were incubated for 16 h at 37 °C in medium B containing 10% LPDS, 50 �M compactin, and50 �M mevalonate. A and B, on day 4, cells were switched to medium B containing 10% LPDS and 50 �M compactin in the absence (�) or presence(�) of sterols (1 �g/ml 25-hydroxycholesterol and 10 �g/ml cholesterol) and/or 10 mM mevalonate as indicated. After 5 h, cells were harvested forpreparation of whole cell extracts (A) and total RNA (B). Aliquots of extracts (10 �g of protein) were subjected to SDS-PAGE and immunoblotanalysis with 5 �g/ml monoclonal IgG-A9 (against reductase) and 1 �g/ml monoclonal IgG-CD71 (against transferrin receptor). Filters wereexposed to film for 1 min (top) and 1 s (bottom). Total RNA was prepared from cells treated with sterols and mevalonate (lanes 4, 8, 12, and 16).Aliquots of reverse-transcribed total RNA (20 ng) were subjected to real-time PCR. Each value for cells transfected with the indicated siRNArepresents the amount of Insig-1 mRNA or Insig-2 mRNA relative to that in control cells transfected with siRNA targeting VSV-G. C, on day 4,cells were preincubated for 1 h in medium C, after which they were pulse-labeled for 1 h with 100 �Ci/ml [35S]methionine in medium C. Cells werethen chased in medium C supplemented with 0.5 mM unlabeled methionine and 1 mM unlabeled cysteine in the absence (�) or presence (�) ofsterols (1 �g/ml 25-hydroxycholesterol and 10 �g/ml cholesterol) plus 10 mM mevalonate as indicated. After the indicated chase period, cells wereharvested, lysed, and subjected to immunoprecipitation with polyclonal anti-reductase. Aliquots of immunoprecipitates (normalized for equal 35Sradioactivity incorporated into total cell protein) were subjected to SDS-PAGE and transferred to nylon membranes. Filters were exposed for 48 hto an imaging plate at room temperature, scanned, and quantified in a Fuji X Bas 1000 Phosphorimager, and the image was photographed. Theamount of 35S-labeled reductase during the pulse (lane 1 of both gels) was arbitrarily set at 100%. D, on day 4, cells were switched to medium Bcontaining 10% LPDS, 50 �M compactin, and 10 �M MG-132 in the absence (�) or presence (�) of sterols (1 �g/ml 25-hydroxycholesterol and 10�g/ml cholesterol) and/or 10 mM mevalonate as indicated. After 1 h cells were harvested, lysed, and subjected sequentially to immunoprecipitation,SDS-PAGE, and immunoblot analysis as described in the legend to Fig. 1. Filters were exposed to film for 3 (top gel) and 30 s (bottom gel).

Insig-dependent Ubiquitination of HMG-CoA Reductase 52483

Page 6: THE JOURNAL OF BIOLOGICAL CHEMISTRY © 2003 by The … 2… · Navdar Sever‡, Bao-Liang Song‡, Daisuke Yabe‡, Joseph L. Goldstein§, Michael S. Brown¶, and Russell A. DeBose-Boyd

FIG. 3. Lysines 89 and 248 are required for regulated ubiquitination and degradation of HMG-CoA reductase. A, amino acid sequence andtopology of the membrane domain of hamster reductase. Lysine residues are shown in black with those located in the cytosol denoted by arrows. B and C,CHO-K1 cells were set up on day 0 at 5 � 105 cells/60-mm dish in medium A with 5% FCS. On day 1, cells were transfected in 2 ml of medium A containing

Insig-dependent Ubiquitination of HMG-CoA Reductase52484

Page 7: THE JOURNAL OF BIOLOGICAL CHEMISTRY © 2003 by The … 2… · Navdar Sever‡, Bao-Liang Song‡, Daisuke Yabe‡, Joseph L. Goldstein§, Michael S. Brown¶, and Russell A. DeBose-Boyd

plasmid encoding full-length reductase with three tandem cop-ies of the T7 epitope tag at the COOH terminus of the protein.To enhance sensitivity of ubiquitin detection, cells were alsotransfected with pEF1a-HA ubiquitin, an expression plasmidencoding human ubiquitin with an NH2-terminal tag consist-ing of two copies of the HA epitope. In the experiment shown inFig. 3B, CHO cells were transfected with pEF1a-HA-ubiquitinand wild-type (lanes 1–4) or mutant versions (lanes 5–16) ofpCMV-HMG-Red-T7 in the absence or presence of pCMV-Insig-1-Myc. After treatment for 2 h with MG-132, cells were har-vested, transfected reductase was immunoisolated from deter-gent lysates with anti-T7 agarose beads, and samples weresubjected to SDS-PAGE. For simplicity, the cells were incu-bated with a mixture of sterols plus mevalonate instead ofadding each one separately. Immunoblot analysis with anti-HArevealed that ubiquitination of wild-type reductase requiredInsig-1 plus sterols and mevalonate (upper panel, lanes 1–4).Mutating lysine 89 to arginine had no effect on ubiquitination(upper panel, lanes 5–8). However, mutating lysine 248 toarginine individually or in combination with the K89R muta-tion eliminated the regulated ubiquitination of reductase (up-per panel, lanes 11–12 and 15–16). The introduced mutationsdid not affect the total amount of reductase that was produced(lower panel, lanes 1–16).

To test the effects of the lysine mutations on regulated deg-radation, wild-type or mutant versions of pCMV-HMG-Red-T7were transfected into cells with or without pCMV-Insig-1-Myc,and cells were incubated with or without sterols plus meval-onate for 5 h in the absence of the proteasome inhibitor. Asexpected, in the presence of Insig-1 sterols plus mevalonate ledto the disappearance of reductase (Fig. 3C, lane 4), and theK89R mutation had little effect (lane 8). Mutating lysine 248 toarginine partially blocked regulated degradation of reductase(lane 12), and the block was more pronounced with the K89R/K248R double mutant (lanes 13–16). Together, the results ofFig. 3, B and C, suggests that the cytosolically oriented lysine248 is the major site of Insig-dependent ubiquitination of re-ductase. Lysine 89 may play some role when lysine 248 isabsent.

The overexpressed sterol-sensing domain of SCAP blockssterol-mediated degradation of reductase, suggesting that thetwo proteins compete for the same site on Insig (26). Binding ofSCAP to Insig is abolished upon the substitution of a cysteinefor tyrosine 298 (Y298C mutant), which lies within one of thetransmembrane helices in the sterol-sensing domain of SCAP(18, 20, 25). The Y298C mutant of SCAP fails to competitivelyinhibit sterol-mediated degradation of reductase presumablybecause it fails to bind to Insigs (19). Fig. 4A shows an aminoacid alignment between the second transmembrane helices ofreductase and SCAP. Reductase and SCAP share a tetrapep-tide (YIYF) beginning at residue 75 of reductase, which corre-sponds to the crucial Tyr-298 residue in SCAP. To determinewhether this tetrapeptide sequence is essential for reductaseinteraction with Insigs, we generated a series of mutations inpCMV-HMG-Red-T7 and tested their effects on sterol-acceler-

ated degradation (Fig. 4B). CHO cells were transfected withpCMV-Insig-1-Myc and wild-type or mutant versions of pCMV-HMG-Red-T7 containing various combinations of alanine sub-stitutions in the YIYF motif. The level of Insig expression issufficient to permit nearly complete degradation of reductase in5 h in the presence of 25-hydroxycholesterol without addedmevalonate. After incubation with various concentrations of25-hydroxycholesterol for 5 h, cells were harvested, fraction-ated, and subjected to SDS-PAGE followed by immunoblotanalysis with anti-T7 antibody. Under the conditions of thisexperiment, 0.1 �g/ml 25-hydroxycholesterol was sufficient toaccelerate the degradation of wild-type reductase, and com-plete disappearance was observed at 0.3 �g/ml of the oxysterol(Fig. 4B, lanes 1–4 and 17–20). The Y75A mutant (correspond-ing to the Y298C mutant in SCAP) reduced only slightly theeffect of sterols on degradation of reductase (Fig. 4B, lanes5–8). However, substitution of the other tyrosine in this tet-rapeptide (Y77A) rendered reductase significantly resistant tothe sterol effect (lanes 9–12). The combined effect of the twotyrosine mutations (Y75A/Y77A) was more potent than eitherof the two substitutions alone. The isoleucine at the secondposition of the tetrapeptide was also important for sterol-accel-erated degradation (lanes 21–24), but the phenylalanine at thefourth position was not (F78A, lanes 25–28). Substitution of allfour residues of the tetrapeptide (YIYF to AAAA) preventedsterols from exerting any effect on the degradation of reductase(lanes 33–36).

We next tested whether ubiquitination of the various mu-tants of reductase correlates with their resistance to sterol-accelerated degradation (Fig. 4C). Cells were transfected withpCMV-Insig-1-Myc, pEF1a-HA-ubiquitin, and wild-type (lanes1 and 2) or mutant versions of pCMV-HMG-Red-T7 (lanes3–12). The cells were treated for 2 h with MG-132 in theabsence or presence of sterols plus mevalonate and harvested,and detergent extracts were prepared. Transfected reductasewas immunoprecipitated with anti-T7-coupled agarose beads.Immunoblotting the precipitates with anti-HA monoclonal an-tibody revealed that the Y75A mutant of reductase was ubiq-uitinated in a regulated manner, similar to that of the wild-type protein (lanes 1–4). In contrast, ubiquitination of all of thedegradation-resistant mutants was markedly reduced (lanes5–12). These results strengthen the view that the conservedYIYF sequence in reductase is essential for the ability of In-sig-1 to mediate the regulated ubiquitination and the subse-quent proteasomal degradation of reductase.

The results in Figs. 3 and 4 show that lysine 248 and theYIYF sequence are necessary for sterol-mediated reductaseubiquitination. We utilized blue native-polyacrylamide gelelectrophoresis to test the hypothesis that mutations in theYIYF sequence of reductase decrease the binding of protein toInsig-1, whereas substitution of lysine 248 preserves bindingbut prevents ubiquitination. This technique was recently em-ployed to identify sterol-induced complexes between Insig-1and SCAP (19). To eliminate the potential competition betweenSCAP and reductase for Insig, we utilized SRD-13A cells, a

5% LPDS and 1 �g of wild-type (lanes 1–4), K89R (lanes 5–8), K248R (lanes 9–12), or K89R/K248R (lanes 13–16) versions of pCMV-HMG-Red-T7in the absence or presence of 30 ng of pCMV-Insig-1-Myc as indicated. In B, cells in each lane were also transfected with 0.1 �g of pEF1a-HA-ubiquitin. The total DNA in each lane was adjusted to 3 �g/dish by the addition of pcDNA3 mock vector. Six hours after transfection, cells receiveda direct addition of 2 ml of medium A containing 5% LPDS, 10 �M compactin, and 50 �M mevalonate (final concentrations). After 16 h at 37 °C,cells were switched to medium A containing 5% LPDS and 50 �M compactin in the absence (�) or presence (�) of sterols (1 �g/ml 25-hydroxycholesterol and 10 �g/ml cholesterol) plus 10 mM mevalonate as indicated. In B, the medium also contained 10 �M MG-132. B, followingincubation at 37 °C for 2 h, cells were harvested, lysed, and subjected to immunoprecipitation with monoclonal anti-T7-coupled agarose beads.Aliquots of immunoprecipitates were subjected to SDS-PAGE and transferred to nylon membranes. Immunoblot analysis was carried out with1:1000 dilution of monoclonal anti-HA-IgG (against ubiquitin) or 1 �g/ml monoclonal anti-T7 IgG (against reductase). Filters were exposed to filmfor 5 s. C, after incubation for 5 h, cells were harvested, membrane fractions were prepared, and aliquots (10 �g) were subjected to SDS-PAGE.Immunoblot analysis was carried out with 1 �g/ml monoclonal anti-T7 IgG (against reductase) and 1 �g/ml monoclonal anti-Myc IgG (againstInsig-1). Filters were exposed to film for 7–10 s.

Insig-dependent Ubiquitination of HMG-CoA Reductase 52485

Page 8: THE JOURNAL OF BIOLOGICAL CHEMISTRY © 2003 by The … 2… · Navdar Sever‡, Bao-Liang Song‡, Daisuke Yabe‡, Joseph L. Goldstein§, Michael S. Brown¶, and Russell A. DeBose-Boyd

CHO-derived mutant cell line that lacks SCAP (31). In theexperiment shown in Fig. 5, we transfected SRD-13A cells withpCMV-Insig-1-Myc in the absence (lanes 1 and 2) or presence ofwild-type or mutant versions of pCMV-HMG-Red-T7(TM1–8)(lanes 3–8). pCMV-HMG-Red-T7(TM1–8) is an expressionplasmid encoding the membrane domain of hamster reductase(amino acids 1–346) with a COOH-terminal T7 epitope tag (26).Following sterol depletion with hydroxypropyl-�-cyclodextrin,the cells were incubated in medium with or without sterols.The medium also contained MG-132 to prevent proteasomaldegradation of the ubiquitinated reductase membrane domain.After treatments, the cells were harvested, membranes wereisolated and solubilized with digitonin. Soluble material wasmixed with Coomassie Blue and 6-amino-n-hexanoic acid (toimpart a negative charge and prevent precipitation of proteins)and subjected to PAGE. The fractionated proteins were trans-ferred to membranes and blotted with various antibodies. Inthe absence of the reductase membrane domain, Insig-1 ap-peared as a single band as revealed by immunoblotting withanti-Myc. Its migration was not altered upon sterol treatment

(compare lanes 1 and 2 in the upper panel of Fig. 5). This bandwas designated as unbound Insig-1. When the cells expressedthe wild-type reductase membrane domain, some of the Insig-1migrated more slowly (lane 3). This slower migrating bandcontained Insig-1 bound to the membrane domain of reductase,as demonstrated by the fact that it was supershifted when thedigitonin-solubilized material was incubated prior to PAGEwith anti-T7 monoclonal antibody, but not with a control anti-body (data not shown). Thus, we designated the slower migrat-ing band as the reductase-Insig complex. In the presence ofsterols, unbound Insig-1 disappeared almost completely (lane4). The reductase-Insig-1 complex was also diminished, but to alesser extent (lane 4). The decrease in the reductase-Insig-1complex is likely caused by incomplete inhibition of proteaso-mal activity, as revealed by immunoblotting of another aliquotof the same extract that was subjected to SDS-PAGE. This blotrevealed a decrease in the total amount of the reductase mem-brane fragment in the presence of sterols (SDS-PAGE, lowerpanel, lanes 3 and 4).

In cells that expressed the K248R mutant of reductase, about

FIG. 4. Mutations in the sterol-sensing domain of HMG-CoA reductase confer resistance to sterol-mediated degradation. A,comparison of the amino acid sequence of the second transmembrane domain of reductase with that of the comparable sterol-sensing region inSCAP. Sequences are from Chinese hamster reductase (amino acids 57–84) and Chinese hamster SCAP (amino acids 280–307). GenBankTM

accession numbers are L00165 and U67060, respectively. Identical amino acids are highlighted in black. Asterisk denotes the site of sterol-sensingmutation (Y298C) in SCAP. B and C, on day 0, CHO-K1 cells were set up at 5 � 105 cells/60-mm dish in medium A with 5% FCS. On day 1, cellswere transfected with 1 �g of the indicated wild-type or mutant version of pCMV-HMG-Red-T7 together with 30 ng of pCMV-Insig-1-Myc. Cellsin C were also transfected with 0.1 �g of pEF1a-HA-ubiquitin. Six hours after transfection, cells received a direct addition of 2 ml of medium Ato produce final concentrations of 5% LPDS, 10 �M compactin, and 50 �M mevalonate. B, after 16 h, cells were switched to 3 ml of medium Acontaining 5% LPDS, 50 �M compactin, and the indicated concentration of 25-hydroxycholesterol. After incubation for 5 h, cells were harvested,membrane fractions were prepared, and aliquots (15–20 �g) were subjected sequentially to SDS-PAGE and immunoblot analysis as described inthe legend to Fig. 3. Filters were exposed to film for 5–20 s. C, following incubation for 16 h, cells were switched to 3 ml of medium A containing5% LPDS, 50 �M compactin, 10 �M MG-132 in the absence (�) or presence (�) of sterols (1 �g/ml 25-hydroxycholesterol and 10 �g/ml cholesterol)plus 10 mM mevalonate. After 2 h at 37 °C, cells were harvested, lysed, and subjected sequentially to immunoprecipitation, SDS-PAGE, andimmunoblot analysis as described in the legend to Fig. 3. Filters were exposed to film for 1–5 s.

Insig-dependent Ubiquitination of HMG-CoA Reductase52486

Page 9: THE JOURNAL OF BIOLOGICAL CHEMISTRY © 2003 by The … 2… · Navdar Sever‡, Bao-Liang Song‡, Daisuke Yabe‡, Joseph L. Goldstein§, Michael S. Brown¶, and Russell A. DeBose-Boyd

half of the Insig-1 was found in the reductase-Insig-1 complexin the absence of sterols (lane 5). Addition of sterols caused allof the Insig-1 to migrate as a complex with reductase (lane 6),indicating that this mutation does not block the ability ofsterols to enhance binding of the reductase membrane frag-ment to Insig-1. Sterol addition caused only a slight decline inthe total amount of the K248R reductase membrane fragment(SDS-PAGE, lane 6), presumably because this mutant is resist-ant to ubiquitination and degradation. A very different resultwas obtained with the tetrapeptide mutant (YIYF to AAAA). Incells that expressed this mutant protein, the majority of Insig-1was in the unbound state, and there was no increase in thecomplex when sterols were added (blue native-PAGE, lanes 7and 8), nor was there a detectable decrease in the total amountof the reductase membrane domain as revealed by SDS-PAGE(lanes 7 and 8).

We next conducted a series of studies designed to determinewhich product of mevalonate metabolism, in addition to sterols,is required to accelerate the degradation of reductase. To facil-itate cellular uptake of mevalonate, we used a permanentlytransfected line of human SV-589 cells that expresses a cellsurface mevalonate transporter (SV589-pMev cells). Thistransporter is a mutant version of a monocarboxylate trans-porter (MCT-1) with a substitution of phenylalanine for cys-teine at position 360, which lies in a putative transmembrane

helix (28). The mutant protein, but not the native protein,transports mevalonate into cells with high efficiency and thusfacilitates studies of the effects of mevalonate and its productson reductase.

Fig. 6A show three of the branches of the mevalonate path-way in animal cells. Mevalonate, produced by reductase, isconverted to the five-carbon intermediate isopentenyl pyro-phosphate (isopentyl-PP), which is polymerized to form gera-nyl-PP (10 carbons) and farnesyl-PP (15 carbons). Two farne-syl-PP molecules can undergo head-to-head condensation toproduce squalene (30 carbons), which is the first committedintermediate in the cholesterol synthetic pathway. Conversionof squalene to cholesterol is blocked by NB-598, which inhibitssqualene epoxidase (38). Farnesyl-PP can donate a farnesylgroup to certain proteins, and it can condense with anotherisopentenyl-PP to form geranylgeranyl-PP (20 carbons), whichis added to other proteins by two distinct geranylgeranyltransferases (39).

When SV589-pMev cells were incubated with a higher con-centration of mevalonate (10 mM) in the presence of the pro-teasome inhibitor MG-132, reductase was ubiquitinated as de-termined by immunoprecipitation and blotting with the anti-ubiquitin antibody (Fig. 6B, lane 2). Ubiquitination wasblocked by NB-598 (lane 4), indicating that it required theincorporation of mevalonate into cholesterol. Exogenous sterols

FIG. 5. Sterols promote interaction of Insig-1 with wild-type HMG-CoA reductase, but not sterol-sensing domain mutant. On day0, SRD-13A cells were set up for experiments as described under “Experimental Procedures.” On day 2, the cells were transfected in medium Acontaining 5% FCS and 60 ng of pCMV-Insig-1-Myc and/or 4 �g of the wild-type or mutant version of pCMV-HMG-Red-T7(TM1–8) as indicated.The total amount of DNA in each experiment was adjusted to 5 �g/dish by addition of pcDNA3 mock vector. On day 3, cells were switched tomedium A containing 5% LPDS, 1% hydroxypropyl-�-cyclodextrin, and 50 �M compactin. After incubation for 1 h at 37 °C, the cells were washedand switched to the same medium (without hydroxypropyl-�-cyclodextrin) containing 10 �M MG-132 in the absence or presence of 1 �g/ml25-hydroxycholesterol plus 10 �g/ml cholesterol (sterols) as indicated. After incubation for 2 h at 37 °C, cells from 4 dishes were pooled, and theresulting digitonin-solubilized extracts were subjected to blue native-PAGE and immunoblot analysis with 5 �g/ml monoclonal anti-Myc IgG formeasurement of reductase-Insig-1 complex. Filter was exposed to film for 30 s at room temperature. Digitonin-solubilized extracts were alsosubjected to SDS-PAGE and immunoblot analysis with either 5 �g/ml monoclonal anti-Myc IgG (against Insig-1) or 1:1000 dilution of monoclonalanti-T7 IgG (against reductase). Filters were exposed to films for 30 s.

Insig-dependent Ubiquitination of HMG-CoA Reductase 52487

Page 10: THE JOURNAL OF BIOLOGICAL CHEMISTRY © 2003 by The … 2… · Navdar Sever‡, Bao-Liang Song‡, Daisuke Yabe‡, Joseph L. Goldstein§, Michael S. Brown¶, and Russell A. DeBose-Boyd

also provoked ubiquitination of reductase (lane 5), but thiseffect was not blocked by NB-598 (lane 7). In the presence ofmaximal levels of sterols, mevalonate had no additional effecton ubiquitination (lanes 6 and 8).

To determine the relation between ubiquitination and deg-radation of reductase, we made the same additions to SV589-pMev cells in the absence of MG-132 and measured the totalamount of reductase by SDS-PAGE (Fig. 7C). Mevalonate at 10mM caused the complete disappearance of reductase (Fig. 7C,lane 2) and this was blocked by NB-598 (lane 4). Exogenoussterols reduced the amount of reductase only partially (lane 5).Complete disappearance required the addition of mevalonate(lane 6), and this mevalonate effect was not blocked by NB-598(lane 8), indicating that it did not require conversion of meva-lonate to sterols.

To determine which of the nonsterol mevalonate-derivedproducts is required for accelerated degradation of reductase,we tested the ability of externally added farnesol or geranylge-raniol (GG-OH) to reproduce the effects of mevalonate (Fig. 7).For these experiments, we used native SV-589 cells that lackthe mevalonate transporter. Sterols added to these SV-589cells led to a partial decrease in reductase protein after 5 h (Fig.7A, lane 2). Addition of 10 mM mevalonate alone had no effect(lane 3), presumably because of the relatively slow uptake ofmevalonate in these cells. However, mevalonate synergizedwith sterols to give a complete disappearance of reductase (lane4). Farnesol at 10 or 100 �M had no effect on reductase, eitherin the absence or presence of sterols (lanes 5–8). In contrast,GG-OH at 10 �M synergized with sterols (lane 10), and theeffect was complete at 100 �M (lane 12).

As discussed above, Insig proteins are required for the reg-ulated degradation of reductase and for the ability of sterols toblock SCAP-dependent processing of SREBPs. To determine

whether mevalonate and GG-OH contribute to SCAP regula-tion as they do to reductase regulation, we added these com-pounds to cells in the absence or presence of sterols and thenmeasured the amount of membrane-bound and nuclearSREBP-2 as well as membrane-bound reductase (Fig. 7B). Asdescribed previously, both mevalonate and GG-OH synergizedwith sterols in eliminating reductase protein from membranes(Fig. 7B, lanes 4 and 6). On the other hand, sterols aloneblocked SREBP processing completely (lane 8). Mevalonate andGG-OH had no additional effect (lanes 10 and 12). To confirmthat GG-OH was acting by enhancing reductase degradation,we performed a pulse-chase experiment (Fig. 7C). Sterols aloneaccelerated the degradation of reductase (compare lanes e–gwith lanes b–d in Fig. 7C), but some 35S-labeled reductaseremained detectable at 5 h (lane g). GG-OH had little effect ondegradation by itself (lanes i–k), but it synergized with sterolsso that the reductase was markedly reduced at 1.5 h (lane l)and barely detectable at 3 h (lane m).

DISCUSSION

The data in this paper confirm and extend previous observa-tions by others that eukaryotic HMG-CoA reductase is de-graded by a ubiquitin-proteasome mechanism that is enhancedby sterols and nonsterol mevalonate-derived products (5, 7, 13).The new findings in the current paper are as follows: 1) ubiq-uitination and degradation of mammalian reductase is depend-ent upon Insigs, as revealed by the elimination of both pro-cesses when the two Insig mRNAs are removed through RNAinterference (Fig. 2). 2) Ubiquitination occurs primarily onlysine 248 of reductase (Fig. 3). 3) Replacement of lysine 248with arginine does not interfere with sterol-stimulated bindingof reductase to Insigs (Fig. 5), but it does block the subsequentubiquitination and degradation (Fig. 3). 4) Sterol-stimulated

FIG. 6. Sterol and nonsterol requirements for degradation and ubiquitination of endogenous HMG-CoA reductase in SV-589 cellsstably expressing mevalonate transporter, pMev. A, schematic representation of the mevalonate pathway. B and C, SV-589/pMev cells wereset up on day 0 at 4 � 105 cells/100-mm dish in medium B with 10% LDPS, 50 �M compactin, 200 �M mevalonate, and 500 �g/ml G418. On day2, the cells were switched to medium B with 10% LPDS, 50 �M compactin, and 50 �M mevalonate. After 16 h at 37 °C, the cells were switched tomedium B containing 10% LPDS and 50 �M compactin in the absence (�) or presence (�) of 10 mM mevalonate, 30 �M NB-598, and sterols (1 �g/ml25-hydroxycholesterol and 10 �g/ml cholesterol), as indicated. In B, the medium also contained 10 �M MG-132. B, after incubation for 1 h at 37 °C,cells were harvested, lysed, and sequentially subjected to immunoprecipitation, SDS-PAGE, and immunoblot analysis as described in the legendto Fig. 1. Filters were exposed to film for 2 min (top gel) and 30 s (bottom gel). C, following incubation at 37 °C for 5 h, the cells were harvestedand subjected to cell fractionation as described under “Experimental Procedures.” Aliquots of membranes (3 �g of protein) were subjected toSDS-PAGE, transferred to nylon membranes, and immunoblotted with 5 �g/ml monoclonal IgG-A9 (against reductase). The filter was exposed tofilm for 1 min.

Insig-dependent Ubiquitination of HMG-CoA Reductase52488

Page 11: THE JOURNAL OF BIOLOGICAL CHEMISTRY © 2003 by The … 2… · Navdar Sever‡, Bao-Liang Song‡, Daisuke Yabe‡, Joseph L. Goldstein§, Michael S. Brown¶, and Russell A. DeBose-Boyd

binding of reductase to Insigs requires the tetrapeptide YIYFthat is also present in the sterol-sensing domain of SCAP (Fig.5). 5) Mutations within the YIYF tetrapeptide prevent sterol-mediated ubiquitination and degradation (Fig. 4). 6) GG-OH,but not farnesol, can synergize with sterols in acceleratingdegradation of reductase, suggesting that a geranylgerany-lated protein may be involved (Fig. 7).

The data of Fig. 1B show that the kinetics of sterol-stimu-lated reductase ubiquitination in human fibroblasts are com-plex. The initial response is unexpectedly rapid, with ubiquiti-nation detectable within 5 min of sterol addition, suggestingthat the added sterols quickly reach the ER membrane. In theabsence of a proteasome inhibitor, the amount of ubiquitinatedreductase reaches a maximum at 10–30 min, then declinesdramatically by 60 min even though substantial reductaseremains within the cell. The fall in ubiquitinated reductase isblocked by MG-132, implicating proteasomal degradation. Butwhy is the remaining reductase not ubiquitinated? One expla-nation is that the rate of ubiquitination slows after 30 min;another is that the rate of de-ubiquitination increases. It is alsopossible that ubiquitination continues, but the partial declinein total reductase lowers the absolute amount of ubiquitinatedreductase below the threshold of detection by the immunoblot-ting assay. Part of the difficulty in interpretation arises be-cause only a small amount of reductase appears to be ubiquiti-nated at any instant in time, perhaps because of de-ubiquitinating enzymes operating in the cell.

Nonsterol, mevalonate-derived products are not capable ofstimulating reductase degradation when cells are sterol-defi-cient, but they accelerate degradation when sterols are abun-dant (4, 5, 40). Two lines of evidence in the current studyindicate that the major nonsterol product is geranylgeranylpyrophosphate. First, the nonsterol acceleration is not blockedby the squalene epoxidase inhibitor NB-598 (Fig. 6), suggestingthat it occurs at a branch prior to sterol synthesis. Second, theeffect can be reproduced by addition to the culture medium ofGG-OH, but not farnesol. Previous studies have shown thatcells have a salvage pathway by which they can activateGG-OH to the metabolically active pyrophosphate (41). Al-though there is some suggestion that GG-OH can be convertedto cholesterol, there is no known enzymatic pathway for thisconversion. Moreover, the failure of NB-598 to inhibit the me-valonate effect suggests that sterol synthesis is not required. Itseems more likely that GG-OH, after conversion to the pyro-phosphate, is being incorporated into a geranylgeranylatedprotein that is required for maximally rapid reductase degra-dation. Possible candidates include geranylgeranylated Rabproteins, which play roles in vesicular transport (42). We can-not exclude the possibility that GG-OH itself, or its pyrophos-phate derivative, is the actual regulator.

Our finding that GG-OH, but not farnesol, synergizes withsterols in accelerating degradation of mammalian reductasediffers from previous studies. In one study farnesol, but notGG-OH, promoted degradation of reductase in permeabilized

FIG. 7. Geranylgeraniol acceleratessterol-induced degradation of endog-enous HMG-CoA reductase in SV-589cells. A and B, SV-589 cells were set upon day 0 at 2 � 105 cells/100-mm dish inmedium B with 10% FCS. On day 2, thecells were refed medium B with 10%LPDS, 50 �M compactin, and 50 �M me-valonate. Following incubation for 16 h at37 °C, the cells were switched to mediumB containing 10% LPDS and 50 �M com-pactin in the absence (�) or presence (�)of sterols (1 �g/ml 25-hydroxycholesteroland 10 �g/ml cholesterol), 10 mM meval-onate, and the indicated concentration offarnesol or GG-OH. After incubation for5 h at 37 °C, the cells were harvested andsubjected to cell fractionation. A, aliquotsof membrane fractions (5 �g) were sub-jected to SDS-PAGE and immunoblotanalysis as described in the legend to Fig.6. Filters were exposed to film for 30 s. B,aliquots of membrane and nuclear extractfractions (3–12 and 20 �g, respectively)were subjected to SDS-PAGE and immu-noblot analysis as above. Filters were ex-posed to film for 30 s (top gel) and 1 min(bottom gel). P and N denote precursorand nuclear cleaved forms of SREBP-2,respectively. C, SV-589 cells were set upon day 0 at 2 � 105 cells/60-mm dish inmedium B with 10% FCS. On day 1, thecells were refed medium B with 10%LPDS, 50 �M compactin, and 50 �M me-valonate. Following incubation for 16 h at37 °C, cells were preincubated and pulse-labeled with [35S]methionine as describedin the legend to Fig. 2. Cells were thenchased in medium C with 0.5 mM methi-onine and 1 mM unlabeled cysteine in theabsence (�) or presence (�) of sterols (1�g/ml 25-hydroxycholesterol and 10�g/ml cholesterol) and 30 �M GG-OH asindicated. Following the indicated chaseperiod, cells were harvested, lysed, andsubjected to immunoprecipitation withpolyclonal anti-reductase and analyzed asdescribed in the legend to Fig. 2.

Insig-dependent Ubiquitination of HMG-CoA Reductase 52489

Page 12: THE JOURNAL OF BIOLOGICAL CHEMISTRY © 2003 by The … 2… · Navdar Sever‡, Bao-Liang Song‡, Daisuke Yabe‡, Joseph L. Goldstein§, Michael S. Brown¶, and Russell A. DeBose-Boyd

cells (43). However, subsequent studies showed that farnesolcaused the reductase to become detergent-insoluble and thusresistant to immunoprecipitation, which masqueraded as anapparent increase in degradation (44). In the same study, far-nesol synergized with sterols to accelerate reductase degrada-tion in intact cells, but GG-OH was not tested and kinetic datawere not reported.

The data of Figs. 4 and 5 implicate the YIYF sequence inreductase as a binding site for Insigs. We were attracted to theYIYF sequence because this sequence is conserved in the ste-rol-sensing domain of SCAP (25) and because the first tyrosineof this sequence is required for SCAP interaction with Insigs(19, 20). Surprisingly, the first tyrosine in the reductase YIYFsequence is not required for sterol-stimulated ubiquitinationand degradation, but the isoleucine and the second tyrosine arerequired (IY sequence, Fig. 4). Replacement of the entire YIYFsequence with AAAA blocks reductase binding to Insigs (Fig.5), which explains the lack of ubiquitination and degradation.Whether the IY sequence in SCAP is required for its interactionwith Insigs is currently being explored.

The conservative substitution of arginine for lysine 248 inreductase does not affect Insig binding (Fig. 5), but it abolishesdetectable reductase ubiquitination and delays degradation(Fig. 3). A similar replacement of lysine 89, the only otherlysine in the cytoplasmic loops of reductase, has little effect onits own, but it further delays degradation when lysine 248 isalso replaced (Figs. 3 and 5). These findings indicate that lysine248 is the primary site of reductase ubiquitination and thatlysine 89 can become ubiquitinated to a small degree whenlysine 248 is absent. It is striking that both of these lysines arepredicted to lie at the COOH-terminal ends of cytoplasmicloops where they join membrane-spanning helices (see diagramof Fig. 3). This juxtamembrane location raises the possibilitythat ubiquitin is transferred to reductase by a membrane-bound ubiquitin transferase. This would resemble the situationin S. cerevisiae where, Hrd1p, a membrane-bound ubiquitintransferase, is believed to attach ubiquitins to reductase, ear-marking it for degradation (12). In contrast to the mammaliansystem, the yeast ubiquitination reaction appears to be stimu-lated primarily by a nonsterol mevalonate metabolite, and nota sterol (12). Moreover, the yeast genome does not contain aneasily recognizable counterpart of Insigs.

A major remaining puzzle is how Insigs can interact withsimilar YIYF sequences in the membrane domains of bothSCAP and HMG-CoA reductase while producing dramaticallydifferent consequences. When SCAP binds to Insigs, it is notdegraded, but rather it is retained in the ER (19). Furthermore,we have been unable to find evidence that SCAP is ubiquiti-nated in response to sterols.3 In contrast, the Insig-reductaseinteraction leads to ubiquitination and degradation. Furtherprogress in understanding these important differences willcome through the identification of other proteins that are re-cruited to the Insig-reductase and Insig-SCAP complexes thatform in response to sterols.

REFERENCES

1. Goldstein, J. L., and Brown, M. S. (1990) Nature 343, 425–4302. Brown, M. S., and Goldstein, J. L. (1980) J. Lipid Res. 2l, 505–5l73. Brown, M. S., and Goldstein, J. L. (1997) Cell 89, 331–3404. Nakanishi, M., Goldstein, J. L., and Brown, M. S. (1988) J. Biol. Chem. 263,

8929–89375. Roitelman, J., and Simoni, R. D. (1992) J. Biol. Chem. 267, 25264–252736. Inoue, S., and Simoni, R. D. (1992) J. Biol. Chem. 267, 9080–90867. Ravid, T., Doolman, R., Avner, R., Harats, D., and Roitelman, J. (2000) J. Biol.

Chem. 275, 35840–358478. Liscum, L., Finer-Moore, J., Stroud, R. M., Luskey, K. L., Brown, M. S., and

Goldstein, J. L. (1985) J. Biol. Chem. 260, 522–5309. Roitelman, J., Olender, E. H., Bar-Nun, S., Dunn, W. A., Jr., and Simoni, R. D.

(1992) J. Cell Biol. 117, 959–97310. Gil, G., Faust, J. R., Chin, D. J., Goldstein, J. L., and Brown, M. S. (1985) Cell

41, 249–25811. Skalnik, D. G., Narita, H., Kent, C., and Simoni, R. D. (1988) J. Biol. Chem.

263, 6836–684112. Hampton, R. Y. (2002) Curr. Opin. Cell Biol. 14, 476–48213. Hampton, R. Y., Gardner, R. G., and Rine, J. (1996) Mol. Biol. Cell 7,

2029–204414. Gardner, R. G., Shearer, A. G., and Hampton, R. Y. (2001) Mol. Cell. Biol. 21,

4276–429115. Luskey, K. L., and Stevens, B. (1985) J. Biol. Chem. 260, 10271–1027716. Gardner, R. G., and Hampton, R. Y. (1999) J. Biol. Chem. 274, 31671–3167817. DeBose-Boyd, R. A., Brown, M. S., Li, W.-P., Nohturfft, A., Goldstein, J. L., and

Espenshade, P. J. (1999) Cell 99, 703–71218. Nohturfft, A., Yabe, D., Goldstein, J. L., Brown, M. S., and Espenshade, P. J.

(2000) Cell 102, 315–32319. Yang, T., Espenshade, P. J., Wright, M. E., Yabe, D., Gong, Y., Aebersold, R.,

Goldstein, J. L., and Brown, M. S. (2002) Cell 110, 489–50020. Yabe, D., Brown, M. S., and Goldstein, J. L. (2002) Proc. Natl. Acad. Sci.

U. S. A. 99, 12753–1275821. Hua, X., Nohturfft, A., Goldstein, J. L., and Brown, M. S. (1996) Cell 87,

415–42622. Brown, M. S., and Goldstein, J. L. (1999) Proc. Natl. Acad. Sci. U. S. A. 96,

11041–1104823. Kuwabara, P. E., and Labouesse, M. (2002) Trends Genet. 18, 193–20124. Nohturfft, A., Hua, X., Brown, M. S., and Goldstein, J. L. (1996) Proc. Natl.

Acad. Sci. U. S. A. 93, 13709–1371425. Nohturfft, A., Brown, M. S., and Goldstein, J. L. (1998) Proc. Natl. Acad. Sci.

U. S. A. 95, 12848–1285326. Sever, N., Yang, T., Brown, M. S., Goldstein, J. L., and DeBose-Boyd, R. A.

(2003) Mol. Cell 11, 25–3327. Goldstein, J. L., Basu, S. K., and Brown, M. S. (1983) Methods Enzymol. 98,

241–26028. Kim, C. M., Goldstein, J. L., and Brown, M. S. (1992) J. Biol. Chem. 267,

23113–2312129. Garcia, C. K., Goldstein, J. L., Pathak, R. K., Anderson, R. G. W., and Brown,

M. S. (1994) Cell 76, 865–87330. Yamamoto, T., Davis, C. G., Brown, M. S., Schneider, W. J., Casey, M. L.,

Goldstein, J. L., and Russell, D. W. (1984) Cell 39, 27–3831. Rawson, R. B., DeBose-Boyd, R. A., Goldstein, J. L., and Brown, M. S. (1999)

J. Biol. Chem. 274, 28549–2855632. Liscum, L., Luskey, K. L., Chin, D. J., Ho, Y. K., Goldstein, J. L., and Brown,

M. S. (1983) J. Biol. Chem. 258, 8450–845533. Elbashir, S. M., Harborth, J., Weber, K., and Tuschl, T. (2002) Methods 26,

199–21334. Moon, Y.-A., and Horton, J. D. (2003) J. Biol. Chem. 278, 7335–734335. Liang, G., Yang, J., Horton, J. D., Hammer, R. E., Goldstein, J. L., and Brown,

M. S. (2002) J. Biol. Chem. 277, 9520–952836. Sato, R., Goldstein, J. L., and Brown, M. S. (1993) Proc. Natl. Acad. Sci.

U. S. A. 90, 9261–926537. Mimnaugh, E. G., Bonvini, P., and Neckers, L. (1999) Electrophoresis 20,

418–42838. Horie, M., Tsuchiya, Y., Hayashi, M., Iida, Y., Iwasawa, Y., Nagata, Y.,

Swasaki, Y., Fukuzumi, H., Kitani, K., and Kamei, T. (1990) J. Biol. Chem.265, 18075–18078

39. Zhang, F. L., and Casey, P. J. (1996) Annu. Rev. Biochem. 65, 241–26940. Correll, C. C., and Edwards, P. A. (1994) J. Biol. Chem. 269, 633–63841. Crick, D. C., Andres, D. A., and Waechter, C. J. (1997) Biochem. Biophys. Res.

Commun. 237, 483–48742. Seabra, M. C., Mules, E. H., and Hume, A. N. (2002) Trends Mol. Med. 8, 23–3043. Correll, C. C., Ng, L., and Edwards, P. A. (1994) J. Biol. Chem. 269,

17390–1739344. Meigs, T. E., Roseman, D. S., and Simoni, R. D. (1996) J. Biol. Chem. 271,

7916–7922

3 J. D. Feramisco, M. S. Brown, and J. L. Goldstein, unpublishedobservations.

Insig-dependent Ubiquitination of HMG-CoA Reductase52490