8
A Disease-causing Mutation in the Active Site of Serine Palmitoyltransferase Causes Catalytic Promiscuity * Received for publication, March 10, 2010, and in revised form, May 7, 2010 Published, JBC Papers in Press, May 26, 2010, DOI 10.1074/jbc.M110.122259 Kenneth Gable ‡1 , Sita D. Gupta ‡1 , Gongshe Han , Somashekarappa Niranjanakumari , Jeffrey M. Harmon § , and Teresa M. Dunn ‡2 From the Departments of Biochemistry and Molecular Biology and § Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20184-4799 The autosomal dominant peripheral sensory neuropathy HSAN1 results from mutations in the LCB1 subunit of serine palmitoyltransferase (SPT). Serum from patients and transgenic mice expressing a disease-causing mutation (C133W) contain elevated levels of 1-deoxysphinganine (1-deoxySa), which pre- sumably arise from inappropriate condensation of alanine with palmitoyl-CoA. Mutant heterodimeric SPT is catalytically inac- tive. However, mutant heterotrimeric SPT has 10 –20% of wild-type activity and supports growth of yeast cells lacking endogenous SPT. In addition, long chain base profiling revealed the synthesis of significantly more 1-deoxySa in yeast and mam- malian cells expressing the heterotrimeric mutant enzyme than in cells expressing wild-type enzyme. Wild-type and mutant enzymes had similar affinities for serine. Surprisingly, the enzymes also had similar affinities for alanine, indicating that the major affect of the C133W mutation is to enhance activation of alanine for condensation with the acyl-CoA substrate. In vivo synthesis of 1-deoxySa by the mutant enzyme was proportional to the ratio of alanine to serine in the growth media, suggesting that this ratio can be used to modulate the relative synthesis of sphinganine and 1-deoxySa. By expressing SPT as a single-chain fusion protein to ensure stoichiometric expression of all three subunits, we showed that GADD153, a marker for endoplasmic reticulum stress, was significantly elevated in cells expressing mutant heterotrimers. GADD153 was also elevated in cells treated with 1-deoxySa. Taken together, these data indicate that the HSAN1 mutations perturb the active site of SPT resulting in a gain of function that is responsible for the HSAN1 phenotype. Serine palmitoyltransferase (SPT) 3 catalyzes the committed and rate-limiting step in sphingolipid biosynthesis. Until recently, it was believed that there was a single isozyme of this enzyme containing two subunits, LCB1 and LCB2, with the catalytic domain located at the interface and composed of res- idues from each subunit. However, Hornemann et al. showed that there was a second isoform of LCB2 (1). More recently, we identified two highly related isoforms of a third subunit, ssSPTa and ssSPTb, which are essential for maximal enzymatic activity (2). Thus, there are at least four potential SPT isozymes, each containing a common hLCB1 subunit and one of each of the two hLCB2 and ssSPT subunits. Examination of the acyl-CoA preferences of the hLCB1/LCB2a and hLCB1/LCB2b het- erodimers and the hLCB1/LCB2/ssSPT heterotrimers showed that both the LCB2 isoform and the ssSPT isoform conferred different acyl-CoA preferences (2, 3), revealing a previously unappreciated complexity in SPT substrate selectivity, and the corresponding long chain base (LCB) products. Mutations in the hLCB1 subunit of SPT have been demon- strated to cause the autosomal dominant late-onset hereditary sensory neuropathy, type I (HSAN1) (4 – 6). In addition, an HSAN1-like phenotype is seen in transgenic mice overexpress- ing the LCB1 C133W mutation (7). Because modeling studies placed the Cys-133 residue near the active site (8 –10), reduced SPT activity was seen in transgenic mice (7), and in vitro char- acterization indicated that the mutant heterodimers were cat- alytically inactive (7, 8), it appeared that the HSAN1 phenotype might be the result of haploinsufficiency. However, several lines of evidence suggest that this is not the case. First, measurement of SPT activity in lymphoblastoid cell lines established from normal and affected members of HSAN1 families showed a wide range of SPT activities with some unaffected individuals having less SPT activity than some of the affected relatives (11). Second, despite the fact that heterozygous mLCB1 knock-out mice have a greater reduction in SPT activity than the trans- genic mice overexpressing the LCB1 C133W allele, they show no neurologic impairment (12). Third, the presence of deoxy- LCBs has been reported in both the serum of affected patients (13) and the transgenic mice expressing the mutant allele (12) suggesting that the mutant enzyme is active but has acquired the ability to condense alanine and glycine with acyl-CoAs. Thus, the disease-causing HSAN mutations may result in a gain-of-function. This hypothesis is entirely consistent with the fact that, although many mutations in hLCB1 would inactivate the subunit, only a few mutations have been identified in vari- ous HSAN1 kindreds (14). In addition, because transgenic mice co-overexpressing both mutant and wild-type LCB1 have no obvious phenotype (12), it does not appear that disease-causing mutations lead to misfolding of LCB1 resulting in a toxic pro- tein analogous to those responsible for other neurodegenera- tive diseases (15, 16). The most common HSAN1 mutation resides in a highly con- served domain. Previous studies of yeast SPT demonstrated * This work was supported, in whole or in part, by National Institutes of Health Grant NS47717 (to T. M. D.) from the USPHS, National Institute of Neuro- logical Disorders and Stroke. 1 Both authors contributed equally to this work. 2 To whom correspondence should be addressed: 4301 Jones Bridge Road, Bethesda, MD 20184-4799. Tel.: 301-295-3592; Fax: 301-295-3512; E-mail: [email protected]. 3 The abbreviations used are: SPT, serine palmitoyltransferase; 1-deoxySa, 1-deoxysphinganine; HSAN1, human sensory neuropathy type 1; LCB, long-chain base; HA, hemagglutinin; CHO, Chinese hamster ovary. THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 285, NO. 30, pp. 22846 –22852, July 23, 2010 Printed in the U.S.A. 22846 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 285 • NUMBER 30 • JULY 23, 2010 by guest on January 30, 2018 http://www.jbc.org/ Downloaded from

A Disease-causing Mutation in the Active Site of Serine

  • Upload
    lykhue

  • View
    217

  • Download
    2

Embed Size (px)

Citation preview

A Disease-causing Mutation in the Active Site of SerinePalmitoyltransferase Causes Catalytic Promiscuity*

Received for publication, March 10, 2010, and in revised form, May 7, 2010 Published, JBC Papers in Press, May 26, 2010, DOI 10.1074/jbc.M110.122259

Kenneth Gable‡1, Sita D. Gupta‡1, Gongshe Han‡, Somashekarappa Niranjanakumari‡, Jeffrey M. Harmon§,and Teresa M. Dunn‡2

From the Departments of ‡Biochemistry and Molecular Biology and §Pharmacology, Uniformed Services University ofthe Health Sciences, Bethesda, Maryland 20184-4799

The autosomal dominant peripheral sensory neuropathyHSAN1 results from mutations in the LCB1 subunit of serinepalmitoyltransferase (SPT). Serum frompatients and transgenicmice expressing a disease-causing mutation (C133W) containelevated levels of 1-deoxysphinganine (1-deoxySa), which pre-sumably arise from inappropriate condensation of alanine withpalmitoyl-CoA.Mutant heterodimeric SPT is catalytically inac-tive. However, mutant heterotrimeric SPT has �10–20% ofwild-type activity and supports growth of yeast cells lackingendogenous SPT. In addition, long chain base profiling revealedthe synthesis of significantlymore 1-deoxySa in yeast andmam-malian cells expressing the heterotrimeric mutant enzyme thanin cells expressing wild-type enzyme. Wild-type and mutantenzymes had similar affinities for serine. Surprisingly, theenzymes also had similar affinities for alanine, indicating thatthemajor affect of theC133Wmutation is to enhance activationof alanine for condensation with the acyl-CoA substrate. In vivosynthesis of 1-deoxySa by the mutant enzyme was proportionalto the ratio of alanine to serine in the growth media, suggestingthat this ratio can be used to modulate the relative synthesis ofsphinganine and 1-deoxySa. By expressing SPT as a single-chainfusion protein to ensure stoichiometric expression of all threesubunits, we showed that GADD153, a marker for endoplasmicreticulum stress, was significantly elevated in cells expressingmutant heterotrimers. GADD153 was also elevated in cellstreatedwith 1-deoxySa. Taken together, these data indicate thattheHSAN1mutations perturb the active site of SPT resulting ina gain of function that is responsible for the HSAN1 phenotype.

Serine palmitoyltransferase (SPT)3 catalyzes the committedand rate-limiting step in sphingolipid biosynthesis. Untilrecently, it was believed that there was a single isozyme of thisenzyme containing two subunits, LCB1 and LCB2, with thecatalytic domain located at the interface and composed of res-idues from each subunit. However, Hornemann et al. showedthat there was a second isoform of LCB2 (1). More recently, we

identified two highly related isoforms of a third subunit, ssSPTaand ssSPTb, which are essential for maximal enzymatic activity(2). Thus, there are at least four potential SPT isozymes, eachcontaining a common hLCB1 subunit and one of each of thetwo hLCB2 and ssSPT subunits. Examination of the acyl-CoApreferences of the hLCB1/LCB2a and hLCB1/LCB2b het-erodimers and the hLCB1/LCB2/ssSPT heterotrimers showedthat both the LCB2 isoform and the ssSPT isoform conferreddifferent acyl-CoA preferences (2, 3), revealing a previouslyunappreciated complexity in SPT substrate selectivity, and thecorresponding long chain base (LCB) products.Mutations in the hLCB1 subunit of SPT have been demon-

strated to cause the autosomal dominant late-onset hereditarysensory neuropathy, type I (HSAN1) (4–6). In addition, anHSAN1-like phenotype is seen in transgenic mice overexpress-ing the LCB1C133W mutation (7). Because modeling studiesplaced the Cys-133 residue near the active site (8–10), reducedSPT activity was seen in transgenic mice (7), and in vitro char-acterization indicated that the mutant heterodimers were cat-alytically inactive (7, 8), it appeared that the HSAN1 phenotypemight be the result of haploinsufficiency.However, several linesof evidence suggest that this is not the case. First, measurementof SPT activity in lymphoblastoid cell lines established fromnormal and affected members of HSAN1 families showed awide range of SPT activities with some unaffected individualshaving less SPT activity than some of the affected relatives (11).Second, despite the fact that heterozygous mLCB1 knock-outmice have a greater reduction in SPT activity than the trans-genic mice overexpressing the LCB1C133W allele, they show noneurologic impairment (12). Third, the presence of deoxy-LCBs has been reported in both the serum of affected patients(13) and the transgenic mice expressing the mutant allele (12)suggesting that the mutant enzyme is active but has acquiredthe ability to condense alanine and glycine with acyl-CoAs.Thus, the disease-causing HSAN mutations may result in again-of-function. This hypothesis is entirely consistentwith thefact that, although many mutations in hLCB1 would inactivatethe subunit, only a few mutations have been identified in vari-ousHSAN1 kindreds (14). In addition, because transgenicmiceco-overexpressing both mutant and wild-type LCB1 have noobvious phenotype (12), it does not appear that disease-causingmutations lead to misfolding of LCB1 resulting in a toxic pro-tein analogous to those responsible for other neurodegenera-tive diseases (15, 16).Themost commonHSAN1mutation resides in a highly con-

served domain. Previous studies of yeast SPT demonstrated

* This work was supported, in whole or in part, by National Institutes of HealthGrant NS47717 (to T. M. D.) from the USPHS, National Institute of Neuro-logical Disorders and Stroke.

1 Both authors contributed equally to this work.2 To whom correspondence should be addressed: 4301 Jones Bridge Road,

Bethesda, MD 20184-4799. Tel.: 301-295-3592; Fax: 301-295-3512; E-mail:[email protected].

3 The abbreviations used are: SPT, serine palmitoyltransferase; 1-deoxySa,1-deoxysphinganine; HSAN1, human sensory neuropathy type 1; LCB,long-chain base; HA, hemagglutinin; CHO, Chinese hamster ovary.

THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 285, NO. 30, pp. 22846 –22852, July 23, 2010Printed in the U.S.A.

22846 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 285 • NUMBER 30 • JULY 23, 2010

by guest on January 30, 2018http://w

ww

.jbc.org/D

ownloaded from

that this mutation rendered it catalytically inactive (8). Thesame result was obtained when Chinese hamster LCB1C133Wwas cotransfected with LCB2a in CHO cells (7). However, thelack of activity of the mammalian LCB1133W/LCB2a het-erodimers might simply reflect an extremely reduced basalactivity of the mutant enzymes that lack an ssSPT subunit. Ourrecent discovery of the ssSPT subunits and our ability toexpress the human isozymes in yeast cells lacking endogenousSPT (2) has allowed us to directly test the possibility that thetrimeric human SPT isozymes containing the most commonHSAN1 mutant, hLCB1C133W, are in fact enzymatically activeand capable of utilizing alanine to synthesize the deoxy-long-chain base, 1-deoxySa. Our results demonstrate that this isindeed the case and provide additional evidence that theseunusual products may be causally related to the loss of neuronsin the dorsal root ganglia of patients with HSAN1.

EXPERIMENTAL PROCEDURES

Yeast Strains—The yeast strains used were TDY4116,Mat�lcb1� tsc3� ura3 leu2 lys2 his 3 met15, and TDY8055, Matalcb1� lcb2� leu2 ura3 his3 met 15 lys2 trp1::His3 (2). Themutants were cultured in medium containing 15 �M phyto-sphingosine and 0.2% tergitol.Plasmids—Construction of plasmids used to express the

human SPT subunits in yeast and mammalian cells has beenpreviously described (2). The C133Wmutation was introducedinto the yeast hLCB1 expression plasmid using QuikChangemutagenesis (Stratagene, La Jolla, CA), and theAvrII restrictionfragment from this plasmidwas substituted for the correspond-ing fragment in themammalian expression plasmid. Yeast plas-mids expressing HA-ssSPTa and HA-ssSPTb were constructedby PCR amplifying the ssSPTa or ssSPTb open reading frame togenerate a fragment flanked by NheI-XbaI sites, which wasligated into pGH316 (2), followed by insertion of an SpeI-endedHA epitope. The plasmid expressing the hLCB2a-ssSPTa-LCB1 triple fusion protein was constructed by ligating an XbaI-NheI-ended PCR fragment containing hLCB2a into the NheIsite of pGH316, followed by insertion of an NheI-XbaI-endedPCR fragment containing hLCB1 into the NheI site to create anhLCB2a-hLCB1 fusion with an in-frame NheI site at the junc-tion. Finally, an XbaI-Nhel-ended ssSPTa PCR fragment wasligated into the Nhe1 between hLCB2a-LCB1 to generate theyeast expression plasmid. To create themammalian expressionplasmid, a BglII/SalI-ended PCR fragment containing hLCB2a-ssSPTa-LCB1 was ligated between the BglII and XhoI sites ofpDual GFP-CCM (Vector Biolabs, Philadelphia, PA).CHO Cell Culture and Transfection—The CHO cell line

CHO-LyB (RIKEN Bioresource Center, Japan) deficient inLCB1 expression (17) was maintained in 95:5 Ham’s F-12/Dul-becco’s modified Eagle’s medium containing 4.5 g/liter glucose(Invitrogen) supplemented with 10% fetal bovine serum (JRHBiosciences, Lenexa, KS) and penicillin and streptomycin. Cellswere transfected using Lipofectamine 2000 (Invitrogen) as pre-viously described (18).Microsome Preparation and Immunoblotting—Microsomes

were prepared from yeast and mammalian cells as previouslydescribed (18). hLCB1, hLCB2a, HA-ssSPTa, and HA-ssSPTbwere resolved by SDS-PAGE and visualized by immunoblotting

using antibodies directed against LCB1, LCB2a, and the HAepitope as previously described (2). GADD153 was detectedusing the SC-7351 mouse monoclonal IgG (Santa Cruz Bio-technology, Santa Cruz, CA) at a dilution of 1:250 and glycer-aldehyde-3-phosphate dehydrogenase was detected usingmouse monoclonal antibody MA B374 (Millipore, Billerica,MA) at a dilution of 1:10,000.SPT Assays—Microsomal SPT activity was assayed by mea-

suring acyl-CoA-dependent incorporation of [3H]serine intolong-chain bases as previously described (2). For determinationof kinetic constants, microsomes were incubated for 10 min atthe indicated serine or alanine concentrations, with 18 �Ci/ml[3H]serine or [3H]alanine. For determination of the alanine Ki,the apparent Km values for serine were determined using 0.1mM serine with 18 �Ci/ml [3H]serine at varying alanineconcentrations.LCB Analysis—LCBs were extracted, derivatized, and ana-

lyzed by reversed-phase high-performance liquid chromatog-raphy on a C18 column (4 �m, Grace Vydac, Hesperia, CA)using an HP Series II 1090 liquid chromatograph with HPChemstation coupled to an Agilent 1100 series fluorescencedetector as previously described (19). The 1-deoxySa standardswere either purchased (C18) from Avanti Polar Lipids (Alabas-ter, Al) or were synthesized (C20) using purified Sphingomonaspaucimobilis SPT enzyme (20) with alanine and stearoyl-CoAas substrates to produce C20-3-keto-deoxysphinganine fol-lowed by sodium borohydride reduction.

RESULTS

Expression and Activities of Wild-type and Mutant SPTHeterodimers—It was previously shown that the yeastLcb1pC180W mutant, which is analogous to the hLCB1C133W

mutant, heterodimerizes with yeast Lcb2p, but that the mutantheterodimers are catalytically inactive (8). To determinewhether the hLCB1C133W mutation abolishes the activity ofhLCB1/hLCB2 heterodimers, hLCB1 or hLCB1C133W wascoexpressed with hLCB2a in CHO-LyB cells, and the activitiesof the human mutant and wild-type heterodimers were com-pared. Immunoblotting revealed that the hLCB1C133W mutantand wild-type hLCB1 proteins were expressed at similar levels,and both stabilized cotransfected hLCB2a (Fig. 1A). BecausehLCB2a stability is dependent upon its ability to form a het-erodimer with hLCB1 (21–22) (Fig. 1A), this result indicatesthat hLCB1C133W forms stable heterodimers with hLCB2a.Measurement of SPT activity in microsomes prepared fromtransfected cells showed that, unlike wild-type hLCB1, whichrestored some SPT activity that was further increased by theco-expression of hLCB2a, the hLCB1C133W subunit did notrestore SPT activity with endogenous CHO LCB2 or cotrans-fected hLCB2a (Fig. 1B). Indeed, cotransfection of increasingratios of the hLCB1C133W to wild-type hLCB1 showed that SPTactivity was inversely proportional to the ratio of mutant/wild-type hLCB1 (Fig. 1C). Thus, although wild-type hLCB1 andhLCB1C133W have similar affinities for the hLCB2a subunit, themutant hLCB1 subunit does not form a catalytically active het-erodimer with hLCB2a, and behaves as a classic dominant neg-ative mutant.

HSAN1 Mutation Alters Substrate Utilization

JULY 23, 2010 • VOLUME 285 • NUMBER 30 JOURNAL OF BIOLOGICAL CHEMISTRY 22847

by guest on January 30, 2018http://w

ww

.jbc.org/D

ownloaded from

Expression and Activity of Mutant SPT Heterotrimers—Be-cause evenwild-type human SPT heterodimers have onlymod-est activity in yeast and transfected CHO-LyB cells (2), the pos-sibility that the disease-causing mutation simply reduces SPTactivity below the level of detection could not be excluded. Wetherefore investigated the effect of the highly homologous smallproteins (ssSPTa and ssSPTb) recently shown to increase SPTactivity 50- to 100-fold (2) on the activity of the hLCB1C133W/LCB2 mutant heterodimers. For these experiments, we tookadvantage of the ability to express active human SPT isozymesin yeast cells lacking any endogenous SPT activity. As previ-ously reported (2), coexpression of ssSPTa or ssSPTb withhLCB1 and hLCB2a resulted in sufficient increase in activity ofthe wild-type heterodimer to support growth of the lcb1�lcb2�mutant at 37 °C (Fig. 2A). More significantly, coexpression ofeither ssSPTa or ssSPTb with hLCB1C133W/LCB2a also pro-vided sufficient SPT activity to support growth at both 26 °Cand 37 °C (Fig. 2A). Measurement of SPT activity in micro-somes prepared from yeast cells expressing mutant isozymescontaining hLCB2a with either ssSPTa or ssSPTb showed thatthemutant enzymes had 10–20% of wild-type activity (Fig. 2C).This is not a result of differential expression of wild-type andmutant hLCB1 or differential stabilization of hLCB2a, because

bothmutant andwild-type proteinswere expressed to the samelevel, and equivalent amounts of hLCB2a were seen in the pres-ence of either hLCB1 or hLCB1C133W (Fig. 2B). A similar acti-vation of hLCB1C133W/LCB2a by ssSPTa or ssSPTb was alsoobserved inCHO-LyB cells (Fig. 2E). Thus, the C133Wdisease-causingmutant forms a catalytically active enzymewhen eitherof the small subunits is present.Altered Substrate Specificity of Mutant SPT Isozyme—We

have recently shown that SPT isozymes containing ssSPTbhave an increased ability to condense stearoyl-CoA with serine(2). In addition, although serine is its preferred amino acid sub-strate, SPT can also condense alanine or glycine with palmitoyl-CoA to generate 1-deoxysphinganine (1-deoxySa) and 1-amino-2-deoxy-n-heptadecane (23–24). Furthermore, the presence ofthese LCB species has been reported in HEK 293 cells express-ing hLCB1C133W in Epstein-Barr virus-transformed lympho-blastoid cell lines and plasma derived fromHSANpatients (13),and in transgenic mice expressing CHO LCB1C133W (12). To

FIGURE 1. Activity of SPT containing the hLCB1C133W mutant subunit.hLCB1 or hLCB1C133W were expressed with or without hLCB2a in CHO-LyBcells, and microsomes were prepared for immunoblotting to ascertain theexpression of transfected genes (A) or used to assay SPT activity (B). Alterna-tively, a constant amount of hLCB1 (0.25 �g) was expressed with increasingamounts of hLCB1C133W and microsomal SPT activity was determined (C).

FIGURE 2. Coexpression of the ssSPT subunits with the hLCB1C133W/hLCB2a heterodimer results in significant catalytic activity. A, yeastlcb1�lcb2� mutants were transformed with plasmids expressing hLCB2a,wild-type, or mutant hLCB1, and empty vector (top row), HA-tagged ssSPTa(middle row), or HA-ssSPTb (bottom row). Transformants were tested forgrowth on YPD plates at 26° or 37 °C or with phytosphingosine. B, microsomalproteins obtained from the lcb1�lcb2� yeast cells expressing hLCB2a withhLCB1 or hLCB1C133W without (lanes 1 and 4) or with HA-ssSPTa (lanes 2 and 5)or HA-ssSPTb (lanes 3 and 6) were fractionated by SDS-PAGE, and subunitexpression was visualized by immunoblotting using anti-hLCB1, anti-hLCB2a,and anti-HA antibodies. The lower of the two bands seen in immunoblots ofyeast microsomal proteins using the anti-hLCB2a antibody is a nonspecificprotein. C, microsomal SPT activities from yeast cells expressing various com-binations of human subunits as in panel B were determined according to“Experimental Procedures.” D, microsomal protein obtained from CHO-LyBcells expressing hLCB2a, wild-type, or C133W mutant hLCB1, and HA-ssSPTa(lanes 1 and 3) or HA-ssSPTb (lanes 2 and 4) were fractionated by SDS-PAGE,and subunit expression was visualized by immunoblotting using anti-hLCB1,anti-hLCB2a, and anti-HA. E, microsomal SPT activities from CHO-LyB cellsexpressing various combinations of human subunits as in panel D weredetermined.

HSAN1 Mutation Alters Substrate Utilization

22848 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 285 • NUMBER 30 • JULY 23, 2010

by guest on January 30, 2018http://w

ww

.jbc.org/D

ownloaded from

determinewhether SPTheterotrimers containing hLCB1C133Whave an increased ability to utilize alanine, wild-type andmutant heterotrimerswere expressed in yeast, and formation ofC18-1-deoxySa and C20-1-deoxySa was determined. Theresults showed that wild-type SPT isozymes synthesized nei-ther C18-1-deoxySa nor C20-1-deoxySa at detectable levels. Incontrast, yeast cells expressing the hLCB1C133W mutant alongwith hLCB2a and ssSPTa synthesized significant amounts ofC18-1-deoxySa, but no C20-1-deoxySa (Fig. 3A), whereas cellsexpressing the hLCB1C133W and hLCB2a along with ssSPTbproduced both C18-1-deoxySa and C20-1-deoxySa (Fig. 3B).As we have previously shown that ssSPTa confers a preferencefor C16-CoA, whereas ssSPTb confers a preference for C18-CoA (2), we conclude that the C133W mutation allows themutant SPT isozymes to react with alanine without alteringtheir acyl-CoA preferences. Cells expressing the SPT isozymescontaining hLCB2b instead of hLCB2a also accumulated 1-de-oxySa.4 It is also interesting to note that, although the productsof SPT are 3-ketosphinganine or 3-ketodeoxysphinganine, itappears that both are efficiently reduced by Tsc10p and FVT1in yeast and mammalian cells, respectively, because the 3-ketoproducts were not detected.

Similar resultswere obtainedwhen themutant SPT isozymeswere expressed in CHO-LyB cells. Long-chain base analysis ofCHO-LyB cells expressing SPT isozymes containing mutanthLCB1C133W revealed the presence of substantial amounts ofC18-1-deoxySa not found in cells expressing any wild-typeisozyme (Fig. 3C). In addition, in contrast to the hLCB1/LCB2a/ssSPTa heterotrimers, expression of mutant hLCB1C133W/LCB2a/ssSPTb heterotrimers also produced low but detectableamounts of C20-1-deoxySa (Fig. 3D). Taken together, theseresults indicate that the mutant heterotrimeric SPT enzymesare active in both yeast and mammalian cells and have anenhanced ability to condense alanine with the preferredacyl-CoA.Kinetic Properties of the Mutant SPT—To investigate the

effect of theHSAN1mutation on amino acid selectivity, micro-somes prepared from yeast expressing the wild-type hLCB1/hLCB2a/ssSPTa and mutant hLCB1C133W/hLCB2a/ssSPTaheterotrimeric SPT isozymes were used to measure theKm andVmax for serine. The results showed that the wild-type enzymehad a Km for serine of �0.75 mM and a Vmax of �1350 pmol/mg/min (Fig. 4A). By comparison the Km andVmax for serine ofthe mutant enzyme were �1.4 mM and �275 pmol/mg/min,respectively (Fig. 4B). Thus, the major affect of the HSAN1mutation is on Vmax. With alanine as substrate, the HSAN1mutant enzyme had a Km of �9.6 mM and a Vmax of �110pmol/mg/min (Fig. 4C). The wild-type SPT did not efficientlyutilize alanine, precluding the direct measurement of a Km.However, inhibition by alanine of serine utilization by the wild-type and mutant enzymes revealed a similar relative affinity foralanine with Ki values of 2 mM and 5mM, respectively (Fig. 4D).Combined with the similar affinities of the mutant and wild-type enzymes for serine and the significant rate of reaction ofthe mutant enzyme with alanine, these data suggest that themajor effect of theHSAN1mutation is to perturb the active sitein such a manner as to allow bound alanine to be activated toreact with the acyl-CoA substrate.Effects of Varying Extracellular Alanine and Serine on 1-

DeoxySa Accumulation—Because the normal extracellularconcentrations of serine and alanine are well below the Km val-ues of SPT, the possibility that altering the ratio of serine toalanine would affect the amount of 1-deoxySa synthesized wasinvestigated. It is clear from the data in Figs. 3 and 5B that yeastand mammalian cells expressing the mutant HSAN1 enzymeproduce a small but significant amount of 1-deoxySa evenwhenno additional alanine is added to the growthmedium. Based onthe serum concentration of alanine, �0.12 mM alanine waspresent in the growth medium of the CHO-LyB cells. Additionof as little as 1 mM alanine elicited a large increase in 1-deoxySaproduction (Fig. 5C). Perhapsmore importantly, increasing theserine concentration from 0.12 mM in normal medium to 1.12mM reversed the effect of added alanine (Fig. 5D), and additionof 10mM serine completely abolished 1-deoxySa synthesis (Fig.5E). Thus, relatively small increases in the ratio of serine toalanine have the potential to inhibit 1-deoxySa synthesis by theHSAN1 enzyme containing the C133Wmutation in hLCB1.1-DeoxySa Induces ER Stress Response—1-deoxySa has been

implicated as the potentially neurotoxic agent in HSAN1 (12–13). However, the basis for this toxicity has not been deter-4 K. Gable, S. D. Gupta, J. M. Harmon, and T. M. Dunn, unpublished results.

FIGURE 3. Mutant human SPT heterotrimers synthesize 1-deoxySa(doxSA). Long-chain bases were analyzed from yeast lcb1�lcb2� mutantsexpressing wild-type (broken line) or mutant (solid line) hLCB1/hLCB2a/ssSPTa(A) or hLCB1/hLCB2a/ssSPTb (B) heterotrimers or from CHO-LyB cells express-ing wild-type (broken line) or mutant (solid line) hLCB1/hLCB2a/ssSPTa (C) orhLCB1/hLCB2a/ssSPTb (D) heterotrimers. The C18-1-deoxySa (C18 DoxSa)eluted at �36 min and the C20 DoxSa at �73 min.

HSAN1 Mutation Alters Substrate Utilization

JULY 23, 2010 • VOLUME 285 • NUMBER 30 JOURNAL OF BIOLOGICAL CHEMISTRY 22849

by guest on January 30, 2018http://w

ww

.jbc.org/D

ownloaded from

mined. Because 1-deoxySa is synthesized in the ER, we exam-ined the possibility that it induces ER stress. Initial experimentsexpressing the three SPT subunits from three cotransfectedexpression plasmids provided some indication that the HSAN1mutant heterotrimer increased GADD153 immunoreactivity, amarker for ER stress and the unfolded protein response (25).However, even the wild-type enzyme, though to a lesser extent,also increased GADD153 expression (data not shown). This islikely due to non-stoichiometric expression of the three sub-units and the consequent misfolding of monomeric subunits.To obtain stoichiometric expression, we extended our previousobservation that a functional SPT enzyme could be synthesizedas an LCB2-LCB1 fusion protein (26) and created a triple fusioncontaining all three subunits (Fig. 6A). Their arrangement wasdictated by the observations that : 1) the C terminus of LCB2 iscytoplasmic5; 2) split ubiquitin two-hybrid experiments placethe N termini of the ssSPTs in the cytoplasm and the ssSPTscontain a single predicted transmembrane domain; and 3) the

N terminus of LCB1 is luminal and the C terminus is cytoplas-mic (18). This arrangement is analogous to the known organi-zation of the coccolithoviral single-chain SPT, in which theLcb2-like domain precedes the Lcb1-like domain with the twoseparated by a short spacer (26). Thus, we reasoned that anhLCB2-ssSPT-LCB1 triple fusion protein would adopt a topol-ogy that recapitulates that of the native SPT heterotrimer.Indeed, the triple fusion protein had activity equal or greater

to, and biochemical properties indistinguishable from, the nor-mal heterotrimer.5 In addition, introduction of the HSAN1mutation into the triple fusion results in 1-deoxySa synthesiscomparable to that obtained by coexpression of mutanthLCB1C133W, hLCB2a, and ssSPTa (compare Figs. 5B and 6B).Using the triple fusion protein, it is clear that the wild-type

enzyme has virtually no affect on GADD153 expression,whereas the mutant fusion protein significantly elevatesGADD153 immunoreactivity (Fig. 6C). This increase is greaterwhen cells are grown in 10 mM alanine and is blocked by inclu-sion of 10 mM serine (Fig. 6C), suggesting that 1-deoxySa, or ametabolite, induces ER stress. Indeed, the level of GADD153was substantially elevated in cells grown in 1 �M deoxySa (Fig.6D). Thus, there is a clear correlation between 1-deoxySa pro-duction and the ER stress response.

DISCUSSION

Until recently, SPT was thought to be a simple heterodimercomposed of two similarly sized subunits, hLCB1 and hLCB2,with each subunit contributing residues that form a catalyticsite at their interface for the condensation of serine and palmi-toyl-CoA. The identification a second isoform (hLCB2b) of thepyridoxal phosphate-binding subunit, LCB2, added some com-plexity, because this isoform appears to confer a broader acyl-CoA preference facilitating the incorporation of shorter acyl-5 G. Han, K. Gable, S. D. Gupta, J. M. Harmon, and T. M. Dunn, in preparation.

FIGURE 4. Properties of SPT enzymes containing wild-type and mutantLCB1 subunits. The serine Km and the Vmax of the wild-type (A) and HSAN1C133W mutant (B) SPT enzymes were determined using [3H]serine asdescribed under “Experimental Procedures.” The alanine Km and Vmax of themutant enzyme was determined using [3H]alanine (C). The Ki values for ala-nine inhibition of serine utilization by the mutant and wild-type enzymeswere determined by measuring the apparent Km for serine in the presence ofvarying concentrations of unlabeled alanine (D).

FIGURE 5. Synthesis of 1-deoxySa is modulated by the extracellular con-centrations of the amino acid substrates. A, the indicated LCB standards(stds) were analyzed by high-performance liquid chromatography asdescribed under “Experimental Procedures.” CHO-LyB cells expressinghLCB1C133W/hLCB2a/ssSPTa were grown in Ham’s F-12 with 5% Dulbecco’smodified Eagle’s medium (B), or in the same media supplemented with 1 mM

alanine (C), 1 mM alanine and 1 mM serine (D), or 1 mM alanine and 10 mM

serine (E), and long-chain bases were extracted and analyzed as describedunder “Experimental Procedures.”

HSAN1 Mutation Alters Substrate Utilization

22850 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 285 • NUMBER 30 • JULY 23, 2010

by guest on January 30, 2018http://w

ww

.jbc.org/D

ownloaded from

CoAs (2, 3). This is similar to what was observed for the single-chain SPT we previously characterized from Emiliania huxleyi(26). However, the activity of either heterodimer is negligible inthe absence of a third subunit (ssSPT) that increases SPT activ-

ity 50- to l00-fold (2). There are two isoforms of this subunit.Both increase SPT activity of hLCB1/hLCB2a and hLCB1/hLCB2b heterodimers. The hLCB1/hLCB2a/ssSPTa isozymehas a clear preference for C-16 acyl-CoA, and the hLCB1/hLCB2b/ssSPTa prefers C-14 acyl-CoAs. In contrast, thehLCB1/hLCB2a/ssSPTb and hLCB1/hLCB2b/ssSPTb iso-zymes have a clear preference for longer rather than shorteracyl-CoAs (2). Indeed, the hLCB1/hLCB2b/ssSPTb isozymeprefers C-18 and C-20 acyl-CoAs. Thus, the acyl-CoA prefer-ences ascribed to the hLCB1/hLCB2 heterodimers are likely aconsequence of the relative amounts of ssSPTa and ssSPTb intransfected cells and the complexities introduced by the tran-sient overexpression of individual subunits of a multimericenzyme.The activation of the hLCB1/LCB2a heterodimer by the

ssSPT subunits allowed us to investigate whether the unusualLCB, 1-deoxySa, suggested to underlie the pathophysiology ofHSAN1, is generated by the mutant SPT enzymes containingthe C133W mutation in hLCB1. Our results show that,although the mutant heterotrimeric SPTs have the same acyl-CoA preferences as their wild-type counterparts, the mutantenzymes have acquired the ability to activate and condense ala-nine with acyl-CoAs to produce the 1-deoxySas. Although ithas been suggested that the HSAN1 mutations cause a higheraffinity for alanine (13), two lines of evidence suggest that theC133W mutation does not significantly alter the amino acidbinding site of the mutant SPT enzyme. First, the mutantenzyme has only a 2-fold greater Km for serine than the wild-type enzyme. Second, the Ki for alanine inhibition of serineutilization by the mutant enzyme is similar to that of the wild-type SPT. The more likely effect of the mutation is to perturbthe catalytic site in such a way as to facilitate the formation ofthe external aldimine with alanine and/or allow the externalaldimine to be more efficiently deprotonated to form and sta-bilize the reactive quininoid intermediate. This is consistentwith the crystal structures of the wild-type and mutant solublebacterial enzymes, which show that the HSAN1-like mutationalters the relative positions of multiple hydrophobic residuescontributed by both subunits at the dimer interface (9). Furtherstudies of the HSAN1-like mutations of the bacterial SPTenzyme are likely to reveal the precise effect of these amino acidsubstitutions on catalysis.Interestingly, Hanada et al. (27) reported that alanine did not

inhibit SPT purified from CHO cells. In preliminary studies wehave observed that the ssSPT subunits dissociate during puri-fication. This raises the possibility that the ssSPT subunit isnecessary for alanine binding. Resolution of this question willrequire the purification of sufficient heterodimeric and hetero-trimeric SPT for kinetic and structural studies. The SPT fusionproteins should facilitate these studies.It has been suggested that 1-deoxySa contributes to the

pathophysiology of HSAN1 (12, 14). The observation thatGADD153, an ER stress response protein, is induced by growthin 1-deoxySa as well as the fact that GADD153 levels areincreased in cells expressing the disease-causing SPT isozymesupport this hypothesis. The ability of physiological extracellu-lar concentrations of alanine to increase both 1-deoxySa pro-duction and the ER stress response, combined with the finding

FIGURE 6. 1-deoxySa induces ER stress. A, model of the hLCB2-ssSPT-LCB1triple fusion SPT in which ssSPT (stippled) is inserted between hLCB2 andhLCB1. Topologies are based on hydropathy analyses as well as preliminaryexperimental data as described in the text. Cys-133, which when mutated totryptophan causes HSAN, is indicated (C). The pyridoxal phosphate-bindinglysine in hLCB2, which in the crystal structure of S. paucimobilis lies directlyacross the dimer interface from Cys-133 (9), is also indicated (K). B, CHO-LyBcells expressing mutant human LCB2a-ssSPTa-LCB1C133W (bottom panel), butnot wild-type LCB2-ssSPTa-LCB1 (top panel), fusion SPT accumulate 1-de-oxySa (DoxSA). C, cells transfected with vector (lanes 1–3), or plasmidsexpressing hLCB2a-ssSPTa-LCB1 (WT; lanes 4 – 6), or hLCB2a-ssSPTa-LCB1C133W (C133W; lanes 7–9) were grown in Ham’s F-12 with or without 10mM serine or 10 mM alanine for 48 h. As controls, untransfected cells weretreated with vehicle (lane 10) or 20 �g/ml tunicamycin for 5 h (lane 11). LCB2a-ssSPTa-LCB1, GADD153, and glyceraldehyde-3-phosphate dehydrogenase(GAPDH) were visualized by immunoblotting following fractionation of totalcellular proteins by SDS-PAGE. D, untransfected CHO-LyB cells were treatedwith vehicle (0.4% bovine serum albumin/phosphate-buffered saline/etha-nol) (lane 1), 1 �M 1-deoxySa (lane 2), 1 �M dihydrosphinganine (lane 3), 1 �M

sphingosine (lane 4), 0.25% DMSO (lane 5), or 20 �g/ml tunicamycin in DMSO(lane 6). GADD153 and glyceraldehyde-3-phosphate dehydrogenase(GAPDH) were visualized by immunoblotting as above. Results are represen-tative of data obtained in three independent experiments.

HSAN1 Mutation Alters Substrate Utilization

JULY 23, 2010 • VOLUME 285 • NUMBER 30 JOURNAL OF BIOLOGICAL CHEMISTRY 22851

by guest on January 30, 2018http://w

ww

.jbc.org/D

ownloaded from

that serine blocks both of these effects, suggests that it may bepossible to directly test this hypothesis bymodulating the ratiosof serine to alanine in the diet of HSAN1 transgenic mice. Apositive result would suggest a potentially simple therapeuticstrategy for treating affected individuals. A negative resultwould be harder to interpret. It could indicate thatmodificationof dietary serine does not alter the serine concentration in theDRG neurons that are most affected in HSAN1 patients. Alter-natively, it could indicate that the catalytic promiscuity reflectsa more subtle alteration in the enzyme, which over time elicitsthe late onset peripheral neuropathy. It will therefore be impor-tant to examine the enzymatic properties of the other HSAN1disease-causing mutations.

Acknowledgments—We are grateful to Dominic Campopiano,Marine Raman, and Jonathan Lowther for helpful discussions.

REFERENCES1. Hornemann, T., Richard, S., Rutti, M. F., Wei, Y., and von Eckardstein, A.

(2006) J. Biol. Chem. 281, 37275–372812. Han, G., Gupta, S. D., Gable, K., Niranjanakumari, S., Moitra, P., Eichler,

F., Brown, R. H., Jr., Harmon, J. M., and Dunn, T. M. (2009) Proc. Natl.Acad. Sci. U.S.A. 106, 8186–8191

3. Hornemann, T., Penno, A., Rutti, M. F., Ernst, D., Kivrak-Pfiffner, F., Ro-hrer, L., and von Eckardstein, A. (2009) J. Biol. Chem. 284, 26322–26330

4. Bejaoui, K., Wu, C., Scheffler, M. D., Haan, G., Ashby, P., Wu, L., de Jong,P., and Brown, R. H., Jr. (2001) Nat. Genet. 27, 261–262

5. Dawkins, J. L., Hulme, D. J., Brahmbhatt, S. B., Auer-Grumbach, M., andNicholson, G. A. (2001) Nat. Genet. 27, 309–312

6. Nicholson, G. A., Dawkins, J. L., Blair, I. P., Auer-Grumbach, M., Brahm-bhatt, S. B., and Hulme, D. J. (2001) Am. J. Hum. Genet. 69, 655–659

7. McCampbell, A., Truong, D., Broom, D. C., Allchorne, A., Gable, K., Cut-ler, R. G.,Mattson,M. P.,Woolf, C. J., Frosch,M. P., Harmon, J. M., Dunn,T. M., and Brown, R. H., Jr. (2005) Hum. Mol. Genet. 14, 3507–3521

8. Gable, K., Han, G., Monaghan, E., Bacikova, D., Natarajan, M., Williams,R., and Dunn, T. M. (2002) J. Biol. Chem. 277, 10194–10200

9. Raman, M. C., Johnson, K. A., Yard, B. A., Lowther, J., Carter, L. G., Nais-mith, J. H., and Campopiano, D. J. (2009) J. Biol. Chem. 284, 17328–17339

10. Yard, B. A., Carter, L. G., Johnson, K. A., Overton, I. M., Dorward,M., Liu,H., McMahon, S. A., Oke, M., Puech, D., Barton, G. J., Naismith, J. H., andCampopiano, D. J. (2007) J. Mol. Biol. 370, 870–886

11. Dedov, V. N., Dedova, I. V., Merrill, A. H., Jr., and Nicholson, G. A. (2004)Biochim. Biophys. Acta 1688, 168–175

12. Eichler, F. S., Hornemann, T., McCampbell, A., Kuljis, D., Penno, A., Var-deh, D., Tamrazian, E., Garofalo, K., Lee, H. J., Kini, L., Selig, M., Frosch,M., Gable, K., von Eckardstein, A., Woolf, C. J., Guan, G., Harmon, J. M.,Dunn, T. M., and Brown, R. H., Jr. (2009) J. Neurosci. 29, 14646–14651

13. Penno,A., Reilly,M.M.,Houlden,H., Laura,M., Rentsch, K.,Niederkofler,V., Stoeckli, E. T., Nicholson, G., Eichler, F., Brown, R. H., Jr., von Eckard-stein, A., and Hornemann, T. (2010) J. Biol. Chem., 285, 11178–11187

14. Hornemann, T., Penno, A., Richard, S., Nicholson, G., van Dijk, F. S.,Rotthier, A., Timmerman, V., and von Eckardstein, A. (2009)Neurogenet-ics 10, 135–143

15. Nakamura, T., and Lipton, S. A. (2009) Apoptosis 14, 455–46816. Luheshi, L. M., and Dobson, C. M. (2009) FEBS Lett. 583, 2581–258617. Hanada, K., Hara, T., Fukasawa,M., Yamaji, A., Umeda,M., andNishijima,

M. (1998) J. Biol. Chem. 273, 33787–3379418. Han, G., Gable, K., Yan, L., Natarajan, M., Krishnamurthy, J., Gupta, S. D.,

Borovitskaya, A., Harmon, J. M., and Dunn, T. M. (2004) J. Biol. Chem.279, 53707–53716

19. Tsegaye, Y., Richardson, C. G., Bravo, J. E., Mulcahy, B. J., Lynch, D. V.,Markham, J. E., Jaworski, J. G., Chen, M., Cahoon, E. B., and Dunn, T. M.(2007) J. Biol. Chem. 282, 28195–28206

20. Gupta, S. D., Gable, K., Han, G., Borovitskaya, A., Selby, L., Dunn, T. M.,and Harmon, J. M. (2009) J. Lipid Res. 50, 1630–1640

21. Gable, K., Slife, H., Bacikova, D., Monaghan, E., and Dunn, T. M. (2000)J. Biol. Chem. 275, 7597–7603

22. Hanada, K., Hara, T., and Nishijima, M. (2000) J. Biol. Chem. 275,8409–8415

23. Pruett, S. T., Bushnev, A., Hagedorn, K., Adiga,M., Haynes, C. A., Sullards,M.C., Liotta, D.C., andMerrill, A.H., Jr. (2008) J. Lipid Res.49, 1621–1639

24. Zitomer, N. C., Mitchell, T., Voss, K. A., Bondy, G. S., Pruett, S. T., Gar-nier-Amblard, E. C., Liebeskind, L. S., Park, H., Wang, E., Sullards, M. C.,Merrill, A. H., Jr., and Riley, R. T. (2009) J. Biol. Chem. 284, 4786–4795

25. Oyadomari, S., and Mori, M. (2004) Cell Death Differ. 11, 381–38926. Han,G., Gable, K., Yan, L., Allen,M. J.,Wilson,W.H.,Moitra, P., Harmon,

J. M., and Dunn, T. M. (2006) J. Biol. Chem. 281, 39935–3994227. Hanada, K., and Nishijima, M. (2003)Methods Mol. Biol. 228, 163–174

HSAN1 Mutation Alters Substrate Utilization

22852 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 285 • NUMBER 30 • JULY 23, 2010

by guest on January 30, 2018http://w

ww

.jbc.org/D

ownloaded from

Jeffrey M. Harmon and Teresa M. DunnKenneth Gable, Sita D. Gupta, Gongshe Han, Somashekarappa Niranjanakumari,

Causes Catalytic PromiscuityA Disease-causing Mutation in the Active Site of Serine Palmitoyltransferase

doi: 10.1074/jbc.M110.122259 originally published online May 26, 20102010, 285:22846-22852.J. Biol. Chem. 

  10.1074/jbc.M110.122259Access the most updated version of this article at doi:

 Alerts:

  When a correction for this article is posted• 

When this article is cited• 

to choose from all of JBC's e-mail alertsClick here

  http://www.jbc.org/content/285/30/22846.full.html#ref-list-1

This article cites 27 references, 16 of which can be accessed free at

by guest on January 30, 2018http://w

ww

.jbc.org/D

ownloaded from