Targeting protein palmitoylation: selective inhibitors and implications in disease

15
1. Introduction 2. PATs and thioesterases 3. PATs, substrate specificity and human disease 4. Palmitoyl-cysteine prediction 5. Methods for palmitoyl-cysteine enrichment and quantitation 6. Pharmacological modulation and inhibitors of palmitoylation 7. Conclusion 8. Expert opinion Review Targeting protein palmitoylation: selective inhibitors and implications in disease Burzin Chavda, John A Arnott & Sonia Lobo Planey The Commonwealth Medical College, Department of Basic Sciences, Scranton, PA, USA Introduction: Palmitoylation describes the enzymatic attachment of the 16-carbon fatty acid, palmitate, to specific cysteines of proteins via a labile thioester bond. This post-translational modification increases the lipophilicity of the modified protein, thus regulating its subcellular distribution and func- tion. The transfer of palmitate to a substrate is mediated by palmitoyl acyltransferases (PATs), while depalmitoylation is catalyzed by acyl protein thioesterases (APTs). Nearly one-third of the 23 genes that encode PATs are linked to human diseases, representing important targets for drug development. Areas covered: In this review, the authors summarize the recent technical advances in the field of palmitoylation and how they will affect our ability to understand palmitoylation and its relevance to human disease. They also review the current literature describing existing palmitoylation inhibitors. The aim of this article is to increase the awareness of the importance of palmi- toylation in disease by reviewing the recent progress made in identifying pharmacological modulators of PATs/APTs. It alsoaims to provide suggestions for general considerations in the development of selective and potent PAT inhibitors. Expert opinion: Developing therapeutically useful pharmacological modula- tors of palmitoylation will require that they be developed within the context of well-characterized PAT/APT-related signaling systems. The successful development of potent, specific drugs in similarly complex systems suggests that development of useful drugs targeting PATs is feasible. Keywords: acyl protein thioesterases, Asp-His-His-Cys, Asp-His-His-Cys 2, cancer, cysteine, palmitoyl acyltransferases, palmitoylation Expert Opin. Drug Discov. [Early Online] 1. Introduction Protein palmitoylation describes the enzymatic, covalent attachment of the 16-carbon fatty acid palmitate to specific cysteine residues of a protein via a labile thioester bond. This essential post-translational modification increases the lipophilicity of the modified protein, thereby dynamically regulating its subcellular distribution, trafficking and function in dramatic and subtle ways. Recent proteo- mic studies have revealed that the number of palmitoylated proteins in mammals is both abundant (hundreds) and diverse; some being initially synthesized on cytosolic ribosomes, whereas others are integral membrane proteins synthesized on endoplasmic reticulum (ER)-bound ribosomes [1,2]. In the former case, palmi- toylation serves as a hydrophobic membrane anchor that permits controlled associ- ation of the cytosolic protein with a membrane, often resulting in its redistribution to a specific membrane compartment as is seen for Ras proteins [3,4]. Similarly, the palmitoylation of integral membrane proteins at cysteines adjacent to or within the transmembrane domain (TMD) can regulate their intracellular trafficking from one 10.1517/17460441.2014.933802 © 2014 Informa UK, Ltd. ISSN 1746-0441, e-ISSN 1746-045X 1 All rights reserved: reproduction in whole or in part not permitted Expert Opin. Drug Discov. Downloaded from informahealthcare.com by Universitaet Zuerich on 06/30/14 For personal use only.

Transcript of Targeting protein palmitoylation: selective inhibitors and implications in disease

Page 1: Targeting protein palmitoylation: selective inhibitors and implications in disease

1. Introduction

2. PATs and thioesterases

3. PATs, substrate specificity and

human disease

4. Palmitoyl-cysteine prediction

5. Methods for palmitoyl-cysteine

enrichment and quantitation

6. Pharmacological modulation

and inhibitors of

palmitoylation

7. Conclusion

8. Expert opinion

Review

Targeting protein palmitoylation:selective inhibitors andimplications in diseaseBurzin Chavda, John A Arnott & Sonia Lobo Planey†

The Commonwealth Medical College, Department of Basic Sciences, Scranton, PA, USA

Introduction: Palmitoylation describes the enzymatic attachment of the

16-carbon fatty acid, palmitate, to specific cysteines of proteins via a labile

thioester bond. This post-translational modification increases the lipophilicity

of the modified protein, thus regulating its subcellular distribution and func-

tion. The transfer of palmitate to a substrate is mediated by palmitoyl

acyltransferases (PATs), while depalmitoylation is catalyzed by acyl protein

thioesterases (APTs). Nearly one-third of the 23 genes that encode PATs

are linked to human diseases, representing important targets for drug

development.

Areas covered: In this review, the authors summarize the recent technical

advances in the field of palmitoylation and how they will affect our ability

to understand palmitoylation and its relevance to human disease. They also

review the current literature describing existing palmitoylation inhibitors.

The aim of this article is to increase the awareness of the importance of palmi-

toylation in disease by reviewing the recent progress made in identifying

pharmacological modulators of PATs/APTs. It also aims to provide suggestions

for general considerations in the development of selective and potent PAT

inhibitors.

Expert opinion: Developing therapeutically useful pharmacological modula-

tors of palmitoylation will require that they be developed within the context

of well-characterized PAT/APT-related signaling systems. The successful

development of potent, specific drugs in similarly complex systems suggests

that development of useful drugs targeting PATs is feasible.

Keywords: acyl protein thioesterases, Asp-His-His-Cys, Asp-His-His-Cys 2, cancer, cysteine,

palmitoyl acyltransferases, palmitoylation

Expert Opin. Drug Discov. [Early Online]

1. Introduction

Protein palmitoylation describes the enzymatic, covalent attachment of the16-carbon fatty acid palmitate to specific cysteine residues of a protein via a labilethioester bond. This essential post-translational modification increases thelipophilicity of the modified protein, thereby dynamically regulating its subcellulardistribution, trafficking and function in dramatic and subtle ways. Recent proteo-mic studies have revealed that the number of palmitoylated proteins in mammalsis both abundant (hundreds) and diverse; some being initially synthesized oncytosolic ribosomes, whereas others are integral membrane proteins synthesizedon endoplasmic reticulum (ER)-bound ribosomes [1,2]. In the former case, palmi-toylation serves as a hydrophobic membrane anchor that permits controlled associ-ation of the cytosolic protein with a membrane, often resulting in its redistributionto a specific membrane compartment as is seen for Ras proteins [3,4]. Similarly, thepalmitoylation of integral membrane proteins at cysteines adjacent to or within thetransmembrane domain (TMD) can regulate their intracellular trafficking from one

10.1517/17460441.2014.933802 © 2014 Informa UK, Ltd. ISSN 1746-0441, e-ISSN 1746-045X 1All rights reserved: reproduction in whole or in part not permitted

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsita

et Z

ueri

ch o

n 06

/30/

14Fo

r pe

rson

al u

se o

nly.

Page 2: Targeting protein palmitoylation: selective inhibitors and implications in disease

membrane system to another, such as from the ER to theplasma membrane (PM) [5]. More subtle changes (in termsof distance) can also occur at the nanoscale level within amembrane as a result of palmitoylation. For instance, theincrease in lipophilicity upon palmitoylation often results inan altered affinity of a protein for a particular lipid microen-vironment within that membrane [6]. For example, lipid raftsare small yet dynamic islands in membranes with distinct lipidcompositions that selectively attract or exclude both periph-eral (often exclusively by virtue of palmitoylation) and inte-gral membrane proteins [7]. Palmitoylated proteins haveaffinity for lipid rafts that are rich in cholesterol, whereas pre-nylated proteins have little or no affinity for these rafts [6].Such lipophilicity-driven changes in protein distributionmay alter the access of a palmitoylated protein to extracellularligands, protein--protein interactions or the engagement of thepalmitoyl-protein in multimolecular signaling complexes.Although palmitate may be the most common lipid species

to occupy cysteine residues, it is not the only one. MarilynResh and colleagues identified the lipid moieties resident onthe cysteine residue of the N-terminal tail of Src familykinases [8-10]. For these proteins, although the cysteine residuenear the N-terminus is most frequently palmitoylated, it isalso modified by palmitoleate, stearate or oleate with afrequency that is apparently related to the abundance ofpalmitate in cells [11]. The physiological differences that resultfrom proteins being modified by these other lipids have not

been explored extensively; however, given their differentphysical properties, it seems reasonable that their impact ona protein should be subtly different than palmitate.

2. PATs and thioesterases

The cellular concentrations of palmitoyl-CoA are generallyinsufficient for spontaneous palmitoylation to occur as wasonce believed, as the concentration of free unbound acyl-CoA esters is in the low nanomolar range under normalphysiological conditions and buffered by specific acyl-CoAbinding proteins [12]. Furthermore, it is now well establishedthat palmitoylation is mostly enzymatic and mediated by afamily of Asp-His-His-Cys (DHHC) motif containing palmi-toyl acyltransferases (PATs), although exception to thiscommon view exists [4]. The highly conserved DHHCcysteine-rich domain (DHHC-CRD) not only defines PATsbut is also directly involved in the transfer of palmitate to asubstrate [13,14]. Most PATs also have a conserved aspartate-proline-glycine motif and Thr-Thr-Xxx-Glu motif, but theirrole in PAT function is not yet known. The DHHC proteinsencoded by these genes all contain four or more TMDs andhave the greatest diversity at the amino acid level in the N-and C-terminal, cytoplasmic tails.

Mammalian genomes contain at least 23 members ofthe ZDHHC PAT gene family. The genomic structure ofZDHHC genes varies widely, including the number anddifferential use of exons that are knit together to generatethe mRNA. ECgene analyses (http://genome.ewha.ac.kr/ECgene/) of the mRNAs that encode PATs suggest that allof the genes are alternatively spliced at various sites through-out the protein coding sequence as well as within untranslatedregions. Many of the putative, alternatively spliced exons arepredicted to encode small peptides that change the structureof the protein in a way that may alter substrate specificity.Likewise, splicing may alter sites for other post-translationalmodifications, such as phosphorylation or glycosylation allof which may regulate activity, substrate specificity, subcellu-lar distribution or interactions with nonsubstrate proteins. Forexample, ZDHHC7 alters the use of a 111 bp exon that isdifferentially and specifically expressed in tissues such as pla-centa, lung, liver, thymus and small intestine [15]. This exonencodes a 37-residue peptide within the cytoplasmic loopbetween TM2 and TM3 that contains a protein kinase Cphosphorylation site. As it is known that multiple PATs canpalmitoylate a single substrate, one might speculate that alter-native splicing of a PAT like ZDHHC7 (or its substrate forthat matter) might shift the balance of responsibility forpalmitoylation of a substrate between multiple PATs.

As predicted by hydropathy analyses, the DHHC proteinsencoded by these genes all contain four or more TMDs. Pre-dictions using TopPred II 1.1 [16] as presented by Ohno andcolleagues show that most PATs have an even number of TMdomains with the DHHC-CRD motif in the cytoplasm andusually located between the second and third TM domain [15].

Article highlights.

. Protein palmitoylation describes the enzymaticattachment of the 16-carbon fatty acid, palmitate, tospecific cysteine residues of a protein via a labilethioester bond.

. In humans, there are at least 23 members of the ZDHHCpalmitoyl acyltransferases (PAT) gene family withpossible overlapping or at least partially overlappingmechanisms/motifs for substrate recognition, specificityand activity.

. Approximately one-third of the genes encoding PATs areassociated with human diseases, including neurological/neuropsychiatric disorders, cancer, dermatologicalpathologies and, most recently, osteoporosis.

. Recent methodological advances in higher throughputproteomic quantification and site-specific labeling haveimproved our ability to annotate palmitoylated proteinson a larger scale and to identify PAT/substrate pairs.

. The field of protein palmitoylation has suffered from alack of specific, potent inhibitors, previously relying ongeneric lipid-based compounds such as2-bromopalmitate, tunicamycin and cerulenin withmultiple, undesired off-target effects.

. The development of specific PAT/APT (acyl proteinthioesterase) inhibitors would provide vital reagents withwhich to study the pathophysiological importance ofmany palmitoylated proteins and may offer potential fortherapeutic development.

This box summarizes key points contained in the article.

B. Chavda et al.

2 Expert Opin. Drug Discov. (2014) 9 (9)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsita

et Z

ueri

ch o

n 06

/30/

14Fo

r pe

rson

al u

se o

nly.

Page 3: Targeting protein palmitoylation: selective inhibitors and implications in disease

In addition to the importance of PAT membrane topology,their membrane system of residence is likely to be an importantaspect of their function. Most mammalian DHHC proteinshave been shown to localize to the ER and Golgi [15]; however,some have also been localized to the PM or to endocyticvesicles [15,17,18]. Yet, little is known about how these proteinsachieve their respective localizations. Some interesting excep-tions include DHHC4/6 and DHHC2. Lysine-based sortingsignals were mapped in DHHC4 (KXX) and DHHC6(KKXX) that determine their restricted localization to ERmembranes [19]. DHHC2 has recently been shown to trafficbetween the PM and intracellular membranes via recyclingendosomes [20]. PM targeting of DHHC2 has also been shownto recruit postsynaptic density-95 to the PM and is essential forpostsynaptic nanodomain formation [21]. Importantly, the C-terminal 68 amino acids of DHHC2 were shown to play animportant role in defining its intracellular localization;however, a defined targeting signal present within this regionof DHHC2 and in other DHHC proteins has yet to bedefined.

Palmitoylation is reversed by a small and not well-characterized family of enzymes called acyl protein thioesterases(APTs), which includes APT1, APT2, APTL1 and PPT1(palmitoyl-protein thioesterase-1). APT1 and APT2, whichare encoded by genes LYPLA1 and LYPLA2, respectively, wereoriginally isolated as lysophospholipases and later demonstratedto be effective as cytosolic protein thioesterases [22-25]. Com-pared with the molecular diversity of PATs, only one APT(i.e., APT1) has been shown to deacylate intracellular proteinsunder conditions where it could play a role in physiologicalregulation of reversible palmitoylation. APT1 appears to beengaged in regulation of the acylation-deacylation cycle ofseveral cytoplasmic proteins including Ga subunits, H-Ras,endothelial nitric oxide synthase and synaptosomal-associatedprotein (SNAP)-23 [2,23,25,26]. APT2 is 68% homologous toAPT1 [24] but catalyzes the depalmitoylation of semisyntheticNRas more efficiently than APT1 [27], and specifically depami-toylates the axonal, peripheral membrane-associated GAP-43[23]. APTL1 (LYPLAL1) is a distant homolog of APT1 thatwas shown to have limited function as a palmitoyl thioesterasedue to its narrow substrate-binding pocket and preference forshort-chain lipid substrates in vitro [28]; however, genetic studiesshow that it is upregulated in obesity [29]. PPT1 is a lysosomalenzyme involved in the degradation of palmitoylated pro-teins [30] and inactivating mutations in the PPT1 gene are acause of infantile neuronal ceroid lipofuscinosis [25]. The dispro-portionately small number of APTs relative to PATs is reminis-cent of the disparity between phosphatases (< 100) and proteinkinases (> 500) in human cells, suggesting that APTs may func-tion as broad specificity enzymes that depalmitoylate a widerange of substrates, whereas PATs harbor the specificity fordynamic palmitoylation. Alternatively, this disparitymay reflectthe fact that only a limited subset of proteins undergo palmitoy-lation turnover. Yet, it is also possible that manymore thioester-ases exist but have yet to be identified.

3. PATs, substrate specificity and humandisease

Discovering the molecular identity of PATs was a pivotal eventthat dramatically accelerated the pace of discovery in the field.Likewise, there has been increased interest in palmitoylationpartly because many of the genes encoding PATs have beenlinked to human diseases. Over the last several years, an increas-ing body of evidence from genetic studies and animal modelsystems has demonstrated the involvement of palmitoylationin numerous and diverse pathological conditions including neu-rological/neuropsychiatric disorders, cancer, dermatologicalpathologies and, most recently, osteoporosis. To date, sevenPAT genes have been shown to be associated with humandisease [31-38], and others have been suggested to be associatedwith disease based on phenotypes seen in animal models [39-41].

The majority of these diseases or phenotypes occur as a resultof the disruption of the homeostatic balance of protein palmi-toylation in the nervous system, highlighting the importanceof palmitoylation for normal neuronal function. To date, eightPATs including ZDHHC7, ZDHHC21, ZDHHC17/huntington interacting protein (HIP)14, HIP14L, ZDHHC8,ZDHHC9, ZD HHC12 and ZDHHC15 and the thioesterasePPT1 have been implicated in neurological/neuropsychiatricdisorders, including Huntington’s disease (HD) [32,42],Alzheimer’s disease [43-45], schizophrenia [31], bipolar disorder[31,46], X-linked mental retardation [33,34,47] and forms of neuro-nal ceroid lipofuscinosis [48,49]. In general, PATs appear tomanifest their effects in neuronal tissue through regulation ofclustering and activity of postsynaptic scaffolds, receptors, ionchannels and other vesicle-associated proteins whose palmitoy-lation status is critical for proper localization and function.

The best studied of these associations is involvement of

DHHC17/HIP14 in HD. HD is a fatal, autosomal-dominant

neuropsychiatric disorder characterized by motor control defi-

cits, depression, anxiety, irritability and cognitive decline [50].

HD is caused bymutation of the huntingtin gene (HTT) result-

ing in abnormal expansion of polyglutamine-encoding CAG

trinucleotide greater than 35 repeats [51]. Genetic and molecular

studies have suggested that mutated HTT with abnormal

expansion of glutamine leads to neuronal damage by gain of

toxic function; however, strong evidence also suggests that

loss of function of HTT also plays a role in pathological

mechanisms of HD [50]. More recently, aberrant HTT palmi-

toylation has been implicated in HD pathogenesis. HTT

palmitoylation is reduced in the brain of the yeast artificial chro-

mosome (YAC)128 mouse model for HD, and this can be

reversed with DHHC17/HIP14 overexpression in YAC128

cortical neurons [32]. Additionally, mutation of HTT, rendering

it palmitoylation resistant, resulted in toxicity and other neuro-

nal cell changes associated with HTT-mutant mouse models of

HD [32,52].HTT has also been shown to interact with PATs.

DHHC17/HIP14 is highly expressed in the brain and has

Targeting protein palmitoylation

Expert Opin. Drug Discov. (2014) 9(9) 3

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsita

et Z

ueri

ch o

n 06

/30/

14Fo

r pe

rson

al u

se o

nly.

Page 4: Targeting protein palmitoylation: selective inhibitors and implications in disease

several other known synaptic substrates [53-55]. DHHC17/HIP14 was recently discovered to be a PAT for HTT [32],and its interaction/binding with HTT is inversely correlatedwith GAC length, suggesting importance in HD pathogene-sis [56]. These observations have recently been explored inmore detail in animal models. In Drosophila, DHHC17/HIP14 loss of function impairs neurotransmitter release [57]

and DHHC17/HIP14 knockout mice share similarities toother late-stage HD mouse models in behavioral, biochemicaland neuropathological features [53]. For example, DHHC17/HIP14 knockout mice generated from gene-trapped ES celllines revealed that loss of DHHC17/HIP14 resulted in striatalnerve count and volume reduction, together with reductionin neurochemical signaling and behavioral deficits seen inHD patients [53]. Recently, a secondary analysis on theseDHHC17/HIP14 knockout mice also revealed marked alter-ations in synaptic function in varied brain regions and signif-icantly impaired hippocampal memory and synaptic plasticity[58]. Interestingly, HTT palmitoylation was not altered inthese mice; however, other DHHC17/HIP14 substrateswere found to be hypo-palmitoylated [53]. This suggests thatother PATs may compensate for DHHC17/HIP14 loss, butthat this compensation is incomplete as not all DHHC17/HIP14 substrates are palmitoylated [48]. Significantly though,these results demonstrated that even though the substrate pooloverlaps for the 23 known PATs, loss of function of a singlePAT could have dramatic pathological effects.DHHC13/HIP14L is a DHHC17/HIP14 homolog [56].

DHHC13/HIP14L-deficient mice develop adult-onset, wide-spread and progressive neuropathology accompanied by earlymotor deficits in climbing, impaired motor learning andreduced palmitoylation of the novel HIP14L substrate,SNAP25 [59]. Although the phenotype in these mice is similarto DHHC17/HIP14 mutant mice, a more progressive pheno-type, similar to that of the YAC128 transgenic mouse modelof HD, was observed. Interestingly, in a different DHHC13/HIP14L mutant mouse model (Hip14LR425X), a severe pheno-type with profound and diverse pathology involving multior-gan/systems was found. These mice developed cachexia,alopecia, osteoporosis, systemic amyloidosis, failure to thriveand early death [41]. This was the first study that implicatedPATs in regulation of skeletal physiology. More detailed exam-ination of the skeletal phenotype in these mice revealedZDHHC13 appears to be a novel regulator of postnatal skeletaldevelopment and bone mass acquisition as mutants displayeddelayed ossification, disorganized growth and early osteoporo-sis [60]. The bone developmental regulator membrane type 1-metalloprotease (MT1-MMP) was identified as a target ofZDHHC13/HIP14L palmitoylation [60], suggesting thatZDHHC13-mediated MT1-MMP palmitoylation is a keymodulator of bone homeostasis. However, this report did notexamine any brain or neurological phenotypes and thus wasunable to corroborate the neuropathology seen in the otherDHHC13/HIP14L-deficient mouse model. Thus, althoughboth DHHC17/HIP14 and DHHC13/HIP14L may be

dysfunctional inHD [59], DHHC13/HIP14Lmutation appearsto be more severe and may also be involved in skeletal and skindiseases/pathologies.

Studies have implicated the loss of ZDHHC8 function toschizophrenia [61]; however, there is still some debate concern-ing the consistency of this finding among various populationsof patients tested [62-66]. The ZDHHC8 gene is located onchromosome 22 and is part of large subset of genes on this chro-mosome responsible for 22q11.2 deletion syndrome [31,67-69]. Inchildren, microdeletions in chromosome 22q11 can result in arange of symptoms including birth defects and various neurode-velopmental and neuropsychiatric symptoms. It is estimatedthat ~ 30%of these patients will develop schizophrenia or schiz-oaffective disorder in adolescence or early adulthood [70,71].Recent work in DHHC8 null mice confirmed that these micemanifest a sexually dimorphic phenotype where only femalemice displayed a decrease in exploratory activity and a defi-ciency in prepulse inhibition, deficits consistent in individualswith schizophrenia [31]. These DHHC8 null mice abnormalitiesare also similar to affects seen in the mouse model for22q11.2 deletion syndrome [72]. Additional genetic associationevidence in the form a single-nucleotide polymorphism inDHHC8 that links it to schizophrenia has also beenfound [31,46], but a link between this small nucleotide polymor-phism and schizophrenia is not a consistent finding [63,65,73].

Other PATs have also been implicated in other neurologi-cal conditions. For example, genetic studies have linkedimpaired ZDHHC15 and ZDHHC9 expression/function tobe a potential cause in X-linked mental retardation [33,34].Additionally, although no direct genetic link has yet to beestablished between palmitoylation and Alzheimer’s disease,ZDHHC12 is thought to contribute to the trafficking andmetabolism of amyloid precursor protein (APP), the precur-sor protein for the generation of neurotoxic b-amyloid seenin Alzheimer’s disease [74]. Palmitoylation may also be impor-tant for localization of the enzymes involved in the APPcleavage process [75,76].

To date, ZDHHC2, ZDHHC7, ZDHHC8, ZDHHC9,ZDHHC11, ZDHHC14, ZDHHC17 and ZDHHC20 haveall been associated with cancer [35-38,77-80]. These PATs canfunction in an antimetastasis or in an oncogenic manner. Forexample, deletion or epigenomic silencing of ZDHHC2, previ-ously known as REAM, for reduced expression associated withmetastasis [35], has been detected in various types of metastaticcancers including breast [81,82], lung [83,84], urinary bladder [85],prostate [86], colorectal [87], hepatocellular [83] and gastriccancers [88], suggesting that palmitoylation of DHHC2 sub-strates impairs metastatic potential. Conversely, ZDHHC17(HIP14) is oncogenic and ZDHHC9 and ZDHHC11 displaycharacteristics that strongly suggest they are oncogenic whenoverexpressed [36-38]. Overexpression of ZDHHC17 has theability to induce colony formation and anchorage-independentgrowth in cell culture and tumors in mice and it palmitoylatesoncogenic RAS proteins [38]. ZDHHC9 is strongly upregulatedin some adenocarcinomas of the gastrointestinal tract at the

B. Chavda et al.

4 Expert Opin. Drug Discov. (2014) 9 (9)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsita

et Z

ueri

ch o

n 06

/30/

14Fo

r pe

rson

al u

se o

nly.

Page 5: Targeting protein palmitoylation: selective inhibitors and implications in disease

transcript and protein levels [37] and it too palmitoylates onco-genic RAS proteins in vitro. Additionally, ZDHHC11 is pres-ent on chromosome 5 that has a high incidence of additionalgenomic copies in cases of bladder cancers with high malignantpotential and in nonsmall cell lung cancer [36].

A single amino acid deletion in DHHC21, resulting in lossof PAT activity, has recently been mapped to the depilatedmutant mouse model [40]. The depilated mutant mouse wasdescribed over three decades ago and results in a recessive phe-notype characterized by variable hair loss, with thinner andshorter hairs remaining in a greasy coat [89]. A more detailedstudy of the phenotype demonstrates that lack of ZDHHC21palmitoylation results in hyperplasia of the interfollicular epi-dermis and sebaceous glands and delayed differentiation ofthe hair shaft [40]. Although the exact DHHC21 substrate(s)responsible for the phenotype have not been identified, Fyn,a member of the Src family of protein tyrosine kinases, wasidentified as a target and was mislocalized in the mutantmice hair follicles [40].

From these examples, it is clear that disrupting the homeo-static balance of protein palmitoylation causes significant anddeleterious effects on normal physiology and further identifi-cation of the substrates of each of these and other DHHCproteins will provide important information concerning themolecular mechanisms underlying human disease as well asreveal novel targets for pharmacological intervention. Thedevelopment of specific DHHC protein inhibitors wouldprovide vital reagents with which to study the pathophysio-logical importance of many palmitoylated proteins and mayoffer potential for therapeutic development.

4. Palmitoyl-cysteine prediction

Due to the recent widespread interest in the effects of proteinpalmitoylation and in the possible outcomes of pharmacolog-ical modulation of palmitoylation dynamics, more and morestudies have focused on identifying and annotating palmitoy-lated proteins [1,2,90-93]. This increasing dataset has fosteredthe development of improved and more accurate algorithmsfor predicting palmitoyl-cysteines.

Although PAT specificity for palmitoylating a particularcysteine residue is highly selective, no unique consensus sitesfor palmitoylation have been definitively established. However,sequence and structural alignment studies have highlightedsome common patterns. For example, palmitoylation iscommonly observed close to the cytosolic side of TMDs.Cysteine-rich motifs as well as cysteines proximal to the N-and C-termini are also commonly palmitoylated [94]. The firstmodel for predicting palmitoylation sites was developed byZhou et al. by employing a clustering and scoring strategy(CSS) (known as CSS-Palm 1.0) using a training set of210 experimentally determined palmitoylation sites from83 distinct proteins [95]. Since 2006, updated versions of CSS-Palm have been released with considerable improvements inprediction capacity and efficiency [96,97]. The current version,

CSS-Palm 4.0, utilizes the latest training set containing 583palmitoylation sites from 277 distinct proteins.

Other popular in silico platforms for palmitoyl-cysteineprediction include incremental feature selection (IFS)-Palm [98], weight, amino acid composition and position spe-cific scoring (WAP)-Palm [99] and PalmPred [100]. PalmPred’sapproach for identification of palmitoylation sites relies on fea-tures extracted only from the primary amino acid sequence,viz., sequence conservation, secondary structure and disor-dered regions, and is claimed to be more efficient than IFS-Palm and WAP-Palm. However, experimental determinationof palmitoyl-cysteines should be emphasized, which, with theapplication of emerging chemo-proteomic approaches, isbecoming more routine and straightforward. Nevertheless,such prediction algorithms offer a much needed support toolfor rapid preliminary characterization and/or validation ofpalmitoylation sites.

5. Methods for palmitoyl-cysteine enrichmentand quantitation

Initial attempts to identify palmitoylated proteins and assign-ing PAT/substrate specificity relied on metabolic labelingwith 3H-palmitate and subsequent candidate detection bySDS-PAGE and fluorography and/or immunoblotting [101,102].Due to recent advances in higher throughput proteomic quan-tification technologies, newer site-specific labeling methodolo-gies have emerged that permit nonradioactive and lesstime-consuming enrichment and annotation of palmitoylatedproteins on a much larger scale.

The first systematic approach for palmitoyl-cysteine enrich-ment, called acyl-biotin exchange (ABE), is based on a muchearlier technique called thiol-disulfide interchange, which wasfirst introduced as a method for isolating cysteine-containingenzymes like cysteine endopeptidases [103]. ABE utilizes selec-tive thioester cleavage by hydroxylamine, which follows ablocking step to cap free (nonpalmitoylated) cysteines. Thefreshly exposed (formerly palmitoylated) cysteines are thenlabeled with a thiol-reactive biotinylated probe via stable(but reversible) disulfide interaction for subsequent biotinylenrichment and detection by mass spectrometry. The firstdemonstration of ABE for large-scale analysis of palmitoy-lated proteins was accomplished by Roth et al. in yeast [92]

and has since been widely used to identify hundreds of palmi-toylated proteins in a variety of proteomes [2,90,104-106].

In a variation of ABE, Forrester et al. demonstrated the useof a thiol affinity resin for direct palmitoyl-cysteine enrich-ment that bypasses biotin-streptavidin capture, thus eliminat-ing nonspecific enrichment of endogenous biotinylatedproteins [107]. Similar in concept to ABE, we developedanother method for identifying substrates of specific PATsin mammalian cells, called palmitoyl-cysteine isolation cap-ture and analysis (PICA), by incorporating isotope-codedaffinity tags (ICAT) [108]. In PICA, heavy (H) and light (L)thiol-reactive, biotinylated ICAT reagents are used to

Targeting protein palmitoylation

Expert Opin. Drug Discov. (2014) 9(9) 5

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsita

et Z

ueri

ch o

n 06

/30/

14Fo

r pe

rson

al u

se o

nly.

Page 6: Targeting protein palmitoylation: selective inhibitors and implications in disease

differentially label newly exposed cysteines (after selectivethioester cleavage, as in ABE) in control cells versus cells inwhich levels of a single PAT are reduced by si-RNA-mediatedknockdown (Figure 1). This allows rapid identification ofpotential PAT substrates following tandem mass spectrometry(MS/MS) and analyzing H/L ratios to quantify the robustnessof palmitoylation. Using this method, we identified and con-firmed by incorporation of radiolabeled palmitate thatcytoskeleton-associated protein 4 (CKAP4) is a key physiolog-ical substrate of DHHC2 in HeLa cells and demonstratedthat loss of DHHC2 expression impairs trafficking ofCKAP4 to the cell surface where it acts as a receptor for anti-proliferative factor (APF) [108]. Without surface expression ofCKAP4, APF is unable to initiate a wide range of downstreameffects, including halting cellular proliferation and altering theexpression of genes related to cancer progression [18]. The dis-covery that CKAP4 is a substrate of DHHC2 provides animportant link between the proliferative properties of manytypes of cancer cells and the reduced expression or mutationof ZDHHC2 that has been associated with metastatic cellularbehavior [35,88,109]. Thus, PICA provides the advantage ofspecifically identifying the palmitoyl-cysteine(s) within apalmitoyl protein, allowing a rapid transition to follow-upexperiments to evaluate the specific role of the palmitoyl-cysteine(s) (versus the many non-palmitoyl-cysteines existingin the protein) to the function of the protein and downstreamsignaling pathways.Hemsley and coworkers described yet another ABE enhance-

ment by employing a biotin switch isobaric tagging for relativeand absolute quantification-based approach to identifyS-acylated proteins from Arabidopsis [93,110]. They identified~ 600 putative S-acylated proteins involved in diverse cellularprocesses and also reported experimental validation on a subsetof those proteins via various in vitro S-acylation assays.Although ABE has proved to be a largely successful methodfor identifying palmitoylated proteins, there are some inherentdrawbacks. For example, there is the requirement of highfidelity protocols for blocking free cysteines as well as for selec-tive thioester hydrolysis, disulfide--interchange reactions andbiotin--streptavidin-based enrichment. Challenges associatedwith such techniques can result in substantial false-positives [92].Another method that has recently gained prominence in

palmitoylation-related studies is target protein labeling withw-azido-fatty acids [111-113]. Building on these initial reports,Martin and Cravatt first demonstrated the use of such azido-fatty acid probes as a method for large-scale identification ofpalmitoylated proteins using the commercially available alkynylfatty acid analogue 17-octadecynoic acid (17-ODYA) [1].17-ODYA is readily incorporated into endogenous palmitoyla-tion sites by the cellular palmitoylation machinery and is thenconjugated to rhodamine-azide or biotin-azide reporter tagsby Cu(I)-catalyzed click chemistry for subsequent gel-baseddetection or biotinyl enrichment. Approximately 125 putativepalmitoylated proteins were identified in human Jurkat T-cellsusing this approach. To improve sensitivity and quantification

of dynamic palmitoylation events, Martin et al. reported anenhancement of their 17-ODYA labeling technique by combin-ing it with stable isotope labeling of cells (SILAC) [91]. Usingthis modified approach, they identified over 400 palmitoylatedproteins in mouse T-cell hybridoma cells. Furthermore, theydemonstrated the applicability of this technique in measuringdynamic palmitoylation turnover by pulse chase experimentswith a serine lipase-specific inhibitor. The subset of these palmi-toylated proteins susceptible to presumably APT-mediatedturnover was identified to contain oncogenes and other proteinslinked to aberrant cell growth, migration and cancer [91], thusalso implying the importance of APTs as targets forpharmacological modulation.

In contrast to ABE, target labeling with bioorthogonalprobes such as 17-ODYA offers some distinct advantages.Chiefly, this approach minimizes nonspecific labeling andthus circumvents generation of false-positives associated withABE protocols, as discussed earlier. It offers flexibility withdetection methods for rapid validation, and temporal controlof probe incorporation to accommodate pulse chaseanalysis [114]. Additionally, in comparison to the traditionallyused spectral counting method for quantitation of palmitoy-lated proteins, SILAC significantly improves on reproducibil-ity, accuracy and sensitivity toward quantifying subtle changesassociated with palmitoylation turnover. However, it shouldalso be noted that chemical probes require metabolic labelingof cells or organisms, unlike in vitro methods such as ABEthat can be performed directly with tissue or cell lysates.

6. Pharmacological modulation and inhibitorsof palmitoylation

As discussed earlier, palmitoylation is catalyzed by PATs,whereas deacylation by APTs. In recent years, the develop-ment of pharmacological modulators of these enzymes hasgarnered increasing interest due to the prospect of alteringthe localization and activity of palmitoylated proteins, severalof which are involved in pathological processes.

6.1 PAT inhibitorsThe use of lipid-based PAT inhibitors such as cerulenin,tunicamycin and the palmitate analog, 2-bromopalmitate(2-BP), has been featured extensively in the literature, with2-BP being most common (Figure 2). Over the years 2-BPhas been widely utilized as an inhibitor of PAT-mediatedpalmitoylation [105,115-118], and sometimes even referred to asa ‘specific’ inhibitor of palmitoylation. Ironically, however,2-BP has long since been known to nonselectively inactivateseveral other types of membrane-bound enzymes [119], includ-ing several enzymes involved in lipid metabolism [120], andeven the deacylating enzymes, APT1 and APT2 [121]. Somerecent studies have explicitly highlighted the promiscuity of2-BP, thus further discrediting its use, at least as an exclusiveinhibitor of enzymatic palmitoylation [122].

B. Chavda et al.

6 Expert Opin. Drug Discov. (2014) 9 (9)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsita

et Z

ueri

ch o

n 06

/30/

14Fo

r pe

rson

al u

se o

nly.

Page 7: Targeting protein palmitoylation: selective inhibitors and implications in disease

P P P

P1

P2

P3

P1

P2

P3

P1

P2

P3

P10

5

10

15

P2 P3

Protein extraction

MMTS

Hydroxylamine

• Protects free thiols (X)

• Palmitates are removed

Differential labelingwith biotinylated

thiol-reactive heavyand light ICAT

reagents

ICAT labeled proteinscombined 1:1,

digested with trypsin,purified with avidin affinity

column

MS/MS peptideidentification; focus on

proteins with reduced H/Lratio

Peptides

Rel

ativ

e ab

unda

nce

• No effect on palmitoylatedcysteines (P)

• Creates reactive thiols (SH)

Heavy

Light

HeavyLight

Ste

p 1

Ste

p 2

Ste

p 3

Ste

p 4

Ste

p 5

Ste

p 6

PATknockdown

Controlcells

Promitoyl-cysteine identification capture and analysis

X

X X XX

PP X

X

PP P P

PP

XX

SHSHSHSHSHSH

SH SH SHSH

SHSH SH

SH

X

X

XX

XXX

1:1

XXXX

X

X

XX

X

X

X

X

X

P P PX

Figure 1. Palmitoyl-cysteine identification capture and analysis: determining palmitoyl acyltransferase-substrate specificity

by differential labeling of palmitoylated proteins with isotope-coded affinity tags.H: Heavy; ICAT: Isotope-coded affinity tags; L: Light; MS: Mass spectrometry; PAT: Palmitoyl acyltransferase.

Targeting protein palmitoylation

Expert Opin. Drug Discov. (2014) 9(9) 7

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsita

et Z

ueri

ch o

n 06

/30/

14Fo

r pe

rson

al u

se o

nly.

Page 8: Targeting protein palmitoylation: selective inhibitors and implications in disease

Due to the nonspecificity of 2-BP and other lipid-baseinhibitors, it is essential to screen for newer selective inhibitorsto accurately establish the role of individual PATs in regulat-ing protein palmitoylation and the resulting downstreameffects. Such efforts have been partly hindered due to a lackof suitable assays for high-throughput screening, as well asinsufficient knowledge of the precise mechanisms of PAT-mediated palmitoylation. Although recent studies haveoffered some mechanistic and kinetic evaluation of PATfunction [14,123,124], without a detailed atomic structure themechanism of palmitoylation remains largely unsubstanti-ated. Another complication arises due to the molecular diver-sity of PATs. There are at least 23 members of the ZDHHCPAT gene family in the mammalian genome with possibleoverlapping or at least partially overlapping mechanisms/motifs for substrate recognition, specificity and activity. Onthe assay front, novel methodologies for palmitoylation assaysthat are amenable to higher-throughput compound screeninghave been emerging (see review by Draper and Smith [125]).Smith and coworkers used a cell-based assay approach to

screen a compound library to identify more selective inhibi-tors of palmitoylation [126]. The screen utilized, among otherassays, cell-permeable green fluorescent protein-linked sub-strate peptides to observe the effects of each compound ininhibiting the localization of such substrates to the PM.They identified five lead compounds, designated compoundsI -- V (Figure 2), that inhibited cellular processes associatedwith palmitoylation. However, follow-up studies revealedthat just one of the compounds, compound V (or2-(2-hydroxy-5-nitro-benzylidene)-benzo[b]thiophen-3-one),inhibited the activity of all four DHHC proteins that weretested [127]. It was also determined that compound V inhibits

PAT autoacylation, a common property of all knownPATs [13], in the same way that 2-BP does; thus, it wouldnot be selective for different PATs. However, unlike 2-BP,inhibition by compound V was largely reversible. Conse-quently, given that 2-BP is also known to inhibit otherenzyme families, compound V is a good candidate for futurestructure-activity tuning studies to increase selectivityand potency.

6.2 APT inhibitorsCertain palmitoylated proteins are dynamically regulated bythe palmitoylation machinery and undergo enzymatic deacy-lation catalyzed by APTs. The cyclic regulation of acylationand deacylation constitutes an important trigger for governingthe intracellular trafficking of such proteins and distinct sig-naling pathways, as was demonstrated with Ras isoforms [128].As mentioned previously, APTs have not been characterizedas extensively as PATs, and only two cytosolic APTs (APT1and APT2) have been reported to have depalmitoylatingactivity against diverse palmitoylated substrate proteinsin vitro [23,129-131], making them attractive targets for pharma-cological intervention. The availability of the APT1 crystalstructure [132] has further enhanced the discovery of APTinhibitors.

One of the first efforts to identify novel APT inhibitors wasbased on the commercially marketed weight loss drug tetrahy-drolipstatin (THL or Orlistat), a prototypical serine hydrolaseinhibitor, following its observed structural homology withgastric lipase [26]. THL possesses an electrophilic b-lactonemoiety that covalently inactivates certain serine hydro-lases [133]. Derivatization of the b-lactone scaffold and screen-ing for inhibition against APT1 led to the discovery of

2-bromopalmitate

Compound V [100][2-(2-hydroxy-5-nitro-benzylidene)-benzo[b]thiophen-3-one]

Cerulenin

Tunicamycin

Figure 2. Palmitoyl acyltransferase inhibitors.

B. Chavda et al.

8 Expert Opin. Drug Discov. (2014) 9 (9)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsita

et Z

ueri

ch o

n 06

/30/

14Fo

r pe

rson

al u

se o

nly.

Page 9: Targeting protein palmitoylation: selective inhibitors and implications in disease

palmostatin B (APT1 IC50 = 5.4 nM) (Table 1), the first com-pound shown to inhibit Ras depalmitoylation in cells [26].Further optimization yielded a more potent and more solubleanalog, palmostatin M (APT1 IC50 = 2.5 nM) (Table 1) [134].Both palmostatin B and palmostatin M are mechanism-basedinhibitors of APTs that covalently modify and inactivate theactive site serine residue. Although both compounds displayhigh selectivity for APTs among other cellular lipid esterases,neither is selective between the two APT isoforms, APT1 andAPT2 [26,134].

In search of a newer APT inhibitor chemotype, Zimmer-man et al. screened a compound library of natural productsand inhibitors for APT binding, and identified boronic acidderivatives as a new class of potent APT inhibitors [135]. Theboron-based inhibitors were observed to have similar inhibi-tion characteristics as palmostatins. Although less potentthen palmostatins, boron-based inhibitors displayed somefavorable attributes, like robust APT binding (slower off-rates), low toxicity, as well as some selectivity between APTisoforms, thus establishing them as a new APT inhibitor che-motype and suitable candidates for future optimization [135].

Recently, Bachovchin et al. introduced an enhancedsubstrate-free methodology, called fluopol-activity-based pro-tein profiling (ABPP), to accommodate high-throughputscreening programs aimed at identifying inhibitor compoundsfor largely uncharacterized enzymes [136]. Fluopol-ABPP capi-talizes on existing ABPP technology by incorporating the useof broad spectrum fluorescent probes. The assay relies oncompetitive active site occupancy between putative inhibitorsand the fluorescent probe, enabling real-time measurementof deviations in enzymatic activity by monitoring the

fluorescence polarization signal [136]. With the aim of develop-ing APT isoform specific inhibitors, fluopol-ABPP was usedto screen a large compound library (315,004 compounds)against APT1 and APT2 in parallel, and the resulting leadcompounds were then selected for downstream gel-basedABPP assays [137]. The authors reported that a majority ofthe lead compounds contained a piperazine amide motif,establishing yet another APT inhibitor chemotype. Eventuallytwo compounds, designated inhibitor 21 and inhibitor 1,were identified as potent and selective inhibitors of APT1and APT2, respectively (Table 1); moreover, the compoundswere reported to maintain in vivo potency and selectivity inmice, thus representing valuable tools for future studiestoward profiling dynamic protein palmitoylation as well asindividually establishing the roles of APTs.

7. Conclusion

Protein palmitoylation increases the lipophilicity of proteins,thereby dynamically regulating their subcellular distribution,trafficking and function in dramatic and subtle ways. Suchlipophilicity-driven changes in protein distribution can alteraccess of a palmitoylated protein to extracellular ligands, canaffect protein--protein interactions or alter the engagementof the palmitoyl-protein in multimolecular signaling com-plexes that can have dramatic resultant physiological andpathological effects. Indeed, over the last several years, anincreasing body of evidence from genetic studies and animalmodel systems has demonstrated that disrupting the homeo-static balance of protein palmitoylation causes significantand deleterious effects on normal physiology. Identification

Table 1. Acyl protein thioesterase inhibitors.

Inhibitor Structure Inhibition-APT1 Inhibition-APT2

Palmostatin B OO

OCH3

OCH3

IC50 = 5.4 nM[16]

IC50 = 37.7 nM[16]

Palmostatin M OO

S

O ONMe2

IC50 = 2.5 nM[107]

IC50 = 19.6 nM[107]

Inhibitor 21

F

CI

NN

O

O

O

FF

NH

Ki = 300 nM[110]

Ki > 10 µM[110]

Inhibitor 1

N

H3CO

N

O

S SO

O

Ki > 10 µM[110]

Ki = 230 nM[110]

Targeting protein palmitoylation

Expert Opin. Drug Discov. (2014) 9(9) 9

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsita

et Z

ueri

ch o

n 06

/30/

14Fo

r pe

rson

al u

se o

nly.

Page 10: Targeting protein palmitoylation: selective inhibitors and implications in disease

of the full complement of substrates of each of these PATsand other DHHC proteins will provide important informa-tion concerning the molecular mechanisms and targets under-lying human diseases. In recent years, the development ofpharmacological modulators of PATs, and more recently,APTs, has garnered increasing interest due to the prospectof altering the localization and activity of palmitoylated pro-teins, several of which are involved in pathological processes.Developing therapeutically useful, pharmacological modula-tors of palmitoylation will require that they be developedwithin the context of well-characterized PAT/APT-relatedsignaling systems. Thus, the further development/refinementof screening approaches for palmitoylated proteins and sub-strates is necessary to identify targets and provide vitalreagents with which to study the pathophysiological impor-tance of many palmitoylated proteins. The successful develop-ment of potent, specific drugs in similarly complex systemssuggests that development of useful drugs targeting PATsis feasible.

8. Expert opinion

Our ability to understand palmitoylation and its importanceto human health and disease is only as good as the technolog-ical methods we use to make accurate and valid measure-ments. Recent methodological advances have improved ourability to annotate palmitoylated proteins and to identifyPAT/substrate pairs, yet a framework for predicting PAT/substrate selectivity is lacking and our understanding of howpalmitoylation affects disease-related signaling processes stillunclear. Selective and potent pharmacological inhibitors ofPATs, like those recently developed for APT, would signifi-cantly advance progress in this field.One major consideration regarding the design of PAT

inhibitors is the specificity of PAT/substrate recognition. TheDHHC motif in a PAT defines the active site and is highlyconserved in all known mammalian PATs [13]. Studies havedemonstrated that for all DHHC proteins studied so farmutation of just the cysteine in the DHHC motif preventsPAT autoacylation and subsequent substrate palmitoylation.This high degree of homology within the active site is not aunique feature to PATs and is seen in many kinases [138,139],where development of selective and potent active-site, ATP-competitive inhibitors has been successful [140]. Evidencederived from knockout animal models clearly demonstratesthat even though the substrate pool overlaps for all the knownPATs, loss of function of a single PAT can have dramaticpathological effects. Presumably, this is the direct result ofhypo-palmitoylation of a combination of multiple substratesconsisting of PAT-specific substrates and more common (i.e.,substrates palmitoylated by multiple PATs) target substrates.However, given the homology of the DHHC protein activesites, specificity of palmitoylation must be derived in partfrom other unique physical interactions of individual PATswith their substrates. It is known that the sequence of amino

acids surrounding a substrate cysteine partially defines thepotential for that cysteine to be palmitoylated. However, thephysical determinants for substrate recognition will likelyextend throughout the accessible portions of the PAT and sub-strate, as is the case for DHHC17 [55]. An important futurestep will be to identify and characterize substrates for sequencepatterns and other potential predictors of activation via specificPATs. Additionally, other factors that are likely to regulatepalmitoylation are the temporal and spatial aspects of PATand substrate expression.

Another complication of PAT-substrate recognition lies inthat each PAT can traverse the membrane multiple times.Thus, it is logical to assume that beyond sequence alone, thelocal membrane environment (i.e., exposed/accessible regionsof each PAT) is important for determining in vivo PAT struc-ture and substrate recognition. However, several PATs havebeen purified from membrane that can remain enzymaticallyactive [127], so it may be possible to use enzyme activity-basedand drug-binding screens for selective PAT inhibitors.

Until such selective PAT inhibitors are available, we shouldbe mindful that using compounds like 2BP, cerulenin andtunicamycin may continue to lead to erroneous conclusionsabout every aspect of palmitoylation. Further, we should notlimit our view of PAT function (or many other proteinsfor that matter) only to palmitoylation as both HIP14(DHHC17) and HIP14L (DHHC13) have been shown tomediate the transport of Mg2+ and fall into a category ofenzymes called ‘chanzymes’ or ion channels that also haveenzymatic activity [141]. Developing nonlipid, selective inhib-itors that target the PAT active site is feasible, and thechallenges that exist are similar to those faced during thedevelopment of selective, small-molecule inhibitors of kinasesthat do not resemble ATP. Targeting those PATs forwhich overexpression is oncogenic----DHHC9, DHHC17 orDHHC11----may be the most logical ones to target first.More conclusive data from in vivo experiments that linkPATs to oncogenesis are warranted to motivate drug discoveryprograms on a large scale toward this goal.

Acknowledgements

The opinions, interpretations, conclusions and recommenda-tions are those of the authors and are not necessarily endorsedby the US Army.

Declaration of interest

The authors declare support from the CommonwealthMedical College. B Chavda is also supported by the USArmy Medical Research and Material Command underAward No W81XWH-13-1-0454. The authors have no otherrelevant affiliations or financial involvement with any organi-zation or entity with a financial interest in or financial conflictwith the subject matter or materials discussed in themanuscript apart from those disclosed.

B. Chavda et al.

10 Expert Opin. Drug Discov. (2014) 9 (9)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsita

et Z

ueri

ch o

n 06

/30/

14Fo

r pe

rson

al u

se o

nly.

Page 11: Targeting protein palmitoylation: selective inhibitors and implications in disease

BibliographyPapers of special note have been highlighted as

either of interest (�) or of considerable interest(��) to readers.

1. Martin BR, Cravatt BF. Large-scale

profiling of protein palmitoylation in

mammalian cells. Nat Methods

2009;6(2):135-8

.. Important methodological advance for

large-scale identification of

palmitoylated proteins.

2. Yang W, Di Vizio D, Kirchner M, et al.

Proteome scale characterization of human

S-acylated proteins in lipid raft-enriched

and non-raft membranes.

Mol Cell Proteomics 2010;9(1):54-70

3. Chamberlain LH, Lemonidis K,

Sanchez-Perez M, et al. Palmitoylation

and the trafficking of peripheral

membrane proteins. Biochem Soc Trans

2013;41(1):62-6

4. Rocks O, Gerauer M, Vartak N, et al.

The palmitoylation machinery is a

spatially organizing system for peripheral

membrane proteins. Cell

2010;141(3):458-71

5. Blaskovic S, Blanc M, van der Goot FG.

What does S-palmitoylation do to

membrane proteins? FEBS J

2013;280(12):2766-74

6. Zacharias DA, Violin JD, Newton AC,

et al. Partitioning of lipid-modified

monomeric GFPs into membrane

microdomains of live cells. Science

2002;296(5569):913-16

7. Levental I, Lingwood D, Grzybek M,

et al. Palmitoylation regulates raft affinity

for the majority of integral raft proteins.

Proc Natl Acad Sci USA

2010;107(51):22050-4

8. Liang X, Lu Y, Neubert TA, et al. Mass

spectrometric analysis of GAP-43/

neuromodulin reveals the presence of a

variety of fatty acylated species.

J Biol Chem 2002;277(36):33032-40

9. Liang X, Lu Y, Wilkes M, et al. The

N-terminal SH4 region of the Src family

kinase Fyn is modified by methylation

and heterogeneous fatty acylation: role in

membrane targeting, cell adhesion, and

spreading. J Biol Chem

2004;279(9):8133-9

10. Liang X, Nazarian A,

Erdjument-Bromage H, et al.

Heterogeneous fatty acylation of Src

family kinases with polyunsaturated fatty

acids regulates raft localization and signal

transduction. J Biol Chem

2001;276(33):30987-94

11. Jones TL, Degtyarev MY,

Backlund PS Jr. The stoichiometry of G

alpha(s) palmitoylation in its basal and

activated states. Biochemistry

1997;36(23):7185-91

12. Faergeman NJ, Knudsen J. Role of long-

chain fatty acyl-CoA esters in the

regulation of metabolism and in cell

signalling. Biochem J

1997;323(Pt 1):1-12

13. Mitchell DA, Vasudevan A, Linder ME,

et al. Protein palmitoylation by a family

of DHHC protein S-acyltransferases.

J Lipid Res 2006;47(6):1118-27

14. Jennings BC, Linder ME. DHHC

protein S-acyltransferases use similar

ping-pong kinetic mechanisms but

display different acyl-CoA specificities.

J Biol Chem 2012;287(10):7236-45

15. Ohno Y, Kihara A, Sano T, et al.

Intracellular localization and tissue-

specific distribution of human and yeast

DHHC cysteine-rich domain-containing

proteins. Biochim Biophys Acta

2006;1761(4):474-83

16. Claros MG, von Heijne G. TopPred II:

an improved software for membrane

protein structure predictions.

Comput Appl Biosci 1994;10(6):685-6

17. Greaves J, Chamberlain LH. Differential

palmitoylation regulates intracellular

patterning of SNAP25. J Cell Sci

2011;124(Pt 8):1351-60

18. Planey SL, Keay SK, Zhang CO, et al.

Palmitoylation of cytoskeleton associated

protein 4 by DHHC2 regulates

antiproliferative factor-mediated

signaling. Mol Biol Cell

2009;20(5):1454-63

19. Gorleku OA, Barns AM, Prescott GR,

et al. Endoplasmic reticulum localization

of DHHC palmitoyltransferases mediated

by lysine-based sorting signals.

J Biol Chem 2011;286(45):39573-84

20. Greaves J, Carmichael JA,

Chamberlain LH. The palmitoyl

transferase DHHC2 targets a dynamic

membrane cycling pathway: regulation by

a C-terminal domain. Mol Biol Cell

2011;22(11):1887-95

21. Fukata Y, Dimitrov A, Boncompain G,

et al. Local palmitoylation cycles define

activity-regulated postsynaptic

subdomains. J Cell Biol

2013;202(1):145-61

22. Sugimoto H, Hayashi H, Yamashita S.

Purification, cDNA cloning, and

regulation of lysophospholipase from rat

liver. J Biol Chem

1996;271(13):7705-11

23. Tomatis VM, Trenchi A, Gomez GA,

et al. Acyl-protein thioesterase 2 catalyzes

the deacylation of peripheral membrane-

associated GAP-43. PLoS One

2010;5(11):e15045

24. Toyoda T, Sugimoto H, Yamashita S.

Sequence, expression in Escherichia coli,

and characterization of lysophospholipase

II. Biochim Biophys Acta

1999;1437(2):182-93

25. Zeidman R, Jackson CS, Magee AI.

Protein acyl thioesterases (Review).

Mol Membr Biol 2009;26(1):32-41

26. Dekker FJ, Rocks O, Vartak N, et al.

Small-molecule inhibition of

APT1 affects Ras localization and

signaling. Nat Chem Biol

2010;6(6):449-56

27. Rusch M, Zimmermann TJ, Burger M,

et al. Identification of acyl protein

thioesterases 1 and 2 as the cellular

targets of the Ras-signaling modulators

palmostatin B and M. Angew Chem Int

Ed Engl 2011;50(42):9838-42

28. Burger M, Zimmermann TJ, Kondoh Y,

et al. Crystal structure of the predicted

phospholipase LYPLAL1 reveals

unexpected functional plasticity despite

close relationship to acyl protein

thioesterases. J Lipid Res

2012;53(1):43-50

29. Fox CS, Liu Y, White CC, et al.

Genome-wide association for abdominal

subcutaneous and visceral adipose reveals

a novel locus for visceral fat in women.

PLoS Genet 2012;8(5):e1002695

30. Verkruyse LA, Hofmann SL. Lysosomal

targeting of palmitoyl-protein

thioesterase. J Biol Chem

1996;271(26):15831-6

31. Mukai J, Liu H, Burt RA, et al.

Evidence that the gene encoding

ZDHHC8 contributes to the risk of

schizophrenia. Nat Genet

2004;36(7):725-31

32. Yanai A, Huang K, Kang R, et al.

Palmitoylation of huntingtin by HIP14 is

Targeting protein palmitoylation

Expert Opin. Drug Discov. (2014) 9(9) 11

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsita

et Z

ueri

ch o

n 06

/30/

14Fo

r pe

rson

al u

se o

nly.

Page 12: Targeting protein palmitoylation: selective inhibitors and implications in disease

essential for its trafficking and function.

Nat Neurosci 2006;9(6):824-31

33. Mansouri MR, Marklund L,

Gustavsson P, et al. Loss of

ZDHHC15 expression in a woman with

a balanced translocation t(X;15)(q13.3;

cen) and severe mental retardation. Eur J

Hum Genet 2005;13(8):970-7

34. Raymond FL, Tarpey PS, Edkins S, et al.

Mutations in ZDHHC9, which encodes

a palmitoyltransferase of NRAS and

HRAS, cause X-linked mental retardation

associated with a Marfanoid habitus.

Am J Hum Genet 2007;80(5):982-7

35. Oyama T, Miyoshi Y, Koyama K, et al.

Isolation of a novel gene on 8p21.3-

22 whose expression is reduced

significantly in human colorectal cancers

with liver metastasis.

Genes Chromosomes Cancer

2000;29(1):9-15

36. Yamamoto Y, Chochi Y, Matsuyama H,

et al. Gain of 5p15.33 is associated with

progression of bladder cancer. Oncology

2007;72(1-2):132-8

37. Mansilla F, Birkenkamp-Demtroder K,

Kruhoffer M, et al. Differential

expression of DHHC9 in microsatellite

stable and instable human colorectal

cancer subgroups. Br J Cancer

2007;96(12):1896-903

38. Ducker CE, Stettler EM, French KJ,

et al. Huntingtin interacting protein

14 is an oncogenic human protein:

palmitoyl acyltransferase. Oncogene

2004;23(57):9230-7

39. Li Y, Hu J, Hofer K, et al.

DHHC5 interacts with PDZ domain

3 of post-synaptic density-95 (PSD-95)

protein and plays a role in learning and

memory. J Biol Chem

2010;285(17):13022-31

40. Mill P, Lee AW, Fukata Y, et al.

Palmitoylation regulates epidermal

homeostasis and hair follicle

differentiation. PLoS Genet

2009;5(11):e1000748

41. Saleem AN, Chen YH, Baek HJ, et al.

Mice with alopecia, osteoporosis, and

systemic amyloidosis due to mutation in

Zdhhc13, a gene coding for palmitoyl

acyltransferase. PLoS Genet

2010;6(6):e1000985

42. Huang K, Sanders SS, Kang R, et al.

Wild-type HTT modulates the enzymatic

activity of the neuronal palmitoyl

transferase HIP14. Hum Mol Genet

2011;20(17):3356-65

43. Bhattacharyya R, Barren C, Kovacs DM.

Palmitoylation of amyloid precursor

protein regulates amyloidogenic

processing in lipid rafts. J Neurosci

2013;33(27):11169-83

44. Benjannet S, Elagoz A, Wickham L,

et al. Post-translational processing of

beta-secretase (beta-amyloid-converting

enzyme) and its ectodomain shedding.

The pro- and transmembrane/cytosolic

domains affect its cellular activity and

amyloid-beta production. J Biol Chem

2001;276(14):10879-87

45. Sidera C, Parsons R, Austen B.

Post-translational processing of beta-

secretase in Alzheimer’s disease.

Proteomics 2005;5(6):1533-43

46. Chen WY, Shi YY, Zheng YL, et al.

Case-control study and transmission

disequilibrium test provide consistent

evidence for association between

schizophrenia and genetic variation in the

22q11 gene ZDHHC8. Hum Mol Genet

2004;13(23):2991-5

47. Ropers HH. X-linked mental retardation:

many genes for a complex disorder.

Curr Opin Genet Dev 2006;16(3):260-9

48. Young FB, Butland SL, Sanders SS, et al.

Putting proteins in their place:

palmitoylation in Huntington disease and

other neuropsychiatric diseases.

Prog Neurobiol 2012;97(2):220-38

49. Vesa J, Hellsten E, Verkruyse LA, et al.

Mutations in the palmitoyl protein

thioesterase gene causing infantile

neuronal ceroid lipofuscinosis. Nature

1995;376(6541):584-7

50. Pla P, Orvoen S, Saudou F, et al. Mood

disorders in Huntington’s disease: from

behavior to cellular and molecular

mechanisms. Front Behav Neurosci

2014;8:135

51. A novel gene containing a trinucleotide

repeat that is expanded and unstable on

Huntington’s disease chromosomes.

The Huntington’s Disease Collaborative

Research Group. Cell 1993;72(6):971-83

52. Slow EJ, van Raamsdonk J, Rogers D,

et al. Selective striatal neuronal loss in a

YAC128 mouse model of Huntington

disease. Hum Mol Genet

2003;12(13):1555-67

53. Singaraja RR, Huang K, Sanders SS,

et al. Altered palmitoylation and

neuropathological deficits in mice lacking

HIP14. Hum Mol Genet

2011;20(20):3899-909

54. Huang K, Yanai A, Kang R, et al.

Huntingtin-interacting protein HIP14 is

a palmitoyl transferase involved in

palmitoylation and trafficking of multiple

neuronal proteins. Neuron

2004;44(6):977-86

55. Huang K, Sanders S, Singaraja R, et al.

Neuronal palmitoyl acyl transferases

exhibit distinct substrate specificity.

FASEB J 2009;23(8):2605-15

56. Singaraja RR, Hadano S, Metzler M,

et al. HIP14, a novel ankyrin domain-

containing protein, links huntingtin to

intracellular trafficking and endocytosis.

Hum Mol Genet 2002;11(23):2815-28

57. Ohyama T, Verstreken P, Ly CV, et al.

Huntingtin-interacting protein 14, a

palmitoyl transferase required for

exocytosis and targeting of CSP to

synaptic vesicles. J Cell Biol

2007;179(7):1481-96

58. Milnerwood AJ, Parsons MP, Young FB,

et al. Memory and synaptic deficits in

Hip14/DHHC17 knockout mice.

Proc Natl Acad Sci USA

2013;110(50):20296-301

59. Sutton LM, Sanders SS, Butland SL,

et al. Hip14l-deficient mice develop

neuropathological and behavioural

features of Huntington disease.

Hum Mol Genet 2013;22(3):452-65

60. Song IW, Li WR, Chen LY, et al.

Palmitoyl acyltransferase, zdhhc13,

facilitates bone mass acquisition by

regulating postnatal epiphyseal

development and endochondral

ossification: a mouse model. PLoS One

2014;9(3):e92194

61. Liu H, Abecasis GR, Heath SC, et al.

Genetic variation in the 22q11 locus and

susceptibility to schizophrenia. Proc Natl

Acad Sci USA 2002;99(26):16859-64

62. Demily C, Legallic S, Bou J, et al.

ZDHHC8 single nucleotide

polymorphism rs175174 is not associated

with psychiatric features of the

22q11 deletion syndrome or

schizophrenia. Psychiatr Genet

2007;17(5):311-12

63. Glaser B, Schumacher J, Williams HJ,

et al. No association between the

putative functional ZDHHC8 single

nucleotide polymorphism rs175174 and

schizophrenia in large European samples.

Biol Psychiatry 2005;58(1):78-80

B. Chavda et al.

12 Expert Opin. Drug Discov. (2014) 9 (9)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsita

et Z

ueri

ch o

n 06

/30/

14Fo

r pe

rson

al u

se o

nly.

Page 13: Targeting protein palmitoylation: selective inhibitors and implications in disease

64. Glaser B, Moskvina V, Kirov G, et al.

Analysis of ProDH, COMT and

ZDHHC8 risk variants does not support

individual or interactive effects on

schizophrenia susceptibility.

Schizophr Res 2006;87(1-3):21-7

65. Otani K, Ujike H, Tanaka Y, et al. The

ZDHHC8 gene did not associate with

bipolar disorder or schizophrenia.

Neurosci Lett 2005;390(3):166-70

66. Saito S, Ikeda M, Iwata N, et al. No

association was found between a

functional SNP in ZDHHC8 and

schizophrenia in a Japanese case-control

population. Neurosci Lett

2005;374(1):21-4

67. Gothelf D, Eliez S, Thompson T, et al.

COMT genotype predicts longitudinal

cognitive decline and psychosis in

22q11.2 deletion syndrome.

Nat Neurosci 2005;8(11):1500-2

68. Paterlini M, Zakharenko SS, Lai WS,

et al. Transcriptional and behavioral

interaction between 22q11.2 orthologs

modulates schizophrenia-related

phenotypes in mice. Nat Neurosci

2005;8(11):1586-94

69. Raux G, Bumsel E, Hecketsweiler B,

et al. Involvement of hyperprolinemia in

cognitive and psychiatric features of the

22q11 deletion syndrome.

Hum Mol Genet 2007;16(1):83-91

70. Chow EW, Watson M, Young DA, et al.

Neurocognitive profile in 22q11 deletion

syndrome and schizophrenia.

Schizophr Res 2006;87(1-3):270-8

71. Pulver AE, Nestadt G, Goldberg R, et al.

Psychotic illness in patients diagnosed

with velo-cardio-facial syndrome and

their relatives. J Nerv Ment Dis

1994;182(8):476-8

72. Mukai J, Dhilla A, Drew LJ, et al.

Palmitoylation-dependent

neurodevelopmental deficits in a mouse

model of 22q11 microdeletion.

Nat Neurosci 2008;11(11):1302-10

73. Faul T, Gawlik M, Bauer M, et al.

ZDHHC8 as a candidate gene for

schizophrenia: analysis of a putative

functional intronic marker in case-control

and family-based association studies.

BMC Psychiatry 2005;5:35

74. Mizumaru C, Saito Y, Ishikawa T, et al.

Suppression of APP-containing vesicle

trafficking and production of beta-

amyloid by AID/DHHC-12 protein.

J Neurochem 2009;111(5):1213-24

75. Cordy JM, Hussain I, Dingwall C, et al.

Exclusively targeting beta-secretase to

lipid rafts by GPI-anchor addition up-

regulates beta-site processing of the

amyloid precursor protein. Proc Natl

Acad Sci USA 2003;100(20):11735-40

76. Riddell DR, Christie G, Hussain I, et al.

Compartmentalization of beta-secretase

(Asp2) into low-buoyant density,

noncaveolar lipid rafts. Curr Biol

2001;11(16):1288-93

77. Draper JM, Smith CD. DHHC20:

a human palmitoyl acyltransferase that

causes cellular transformation.

Mol Membr Biol 2010;27(2-3):123-36

78. Hungermann D, Schmidt H,

Natrajan R, et al. Influence of whole arm

loss of chromosome 16q on gene

expression patterns in oestrogen receptor-

positive, invasive breast cancer. J Pathol

2011;224(4):517-28

79. Yu L, Reader JC, Chen C, et al.

Activation of a novel palmitoyltransferase

ZDHHC14 in acute biphenotypic

leukemia and subsets of acute myeloid

leukemia. Leukemia 2011;25(2):367-71

80. Sudo H, Tsuji AB, Sugyo A, et al. A loss

of function screen identifies nine new

radiation susceptibility genes.

Biochem Biophys Res Commun

2007;364(3):695-701

81. Yaremko ML, Kutza C, Lyzak J, et al.

Loss of heterozygosity from the short

arm of chromosome 8 is associated with

invasive behavior in breast cancer.

Genes Chromosomes Cancer

1996;16(3):189-95

82. Anbazhagan R, Fujii H, Gabrielson E.

Allelic loss of chromosomal arm 8p in

breast cancer progression. Am J Pathol

1998;152(3):815-19

83. Fujiwara Y, Ohata H, Emi M, et al. A 3-

Mb physical map of the chromosome

region 8p21.3-p22, including a 600-kb

region commonly deleted in human

hepatocellular carcinoma, colorectal

cancer, and non-small cell lung cancer.

Genes Chromosomes Cancer

1994;10(1):7-14

84. Ohata H, Emi M, Fujiwara Y, et al.

Deletion mapping of the short arm of

chromosome 8 in non-small cell lung

carcinoma. Genes Chromosomes Cancer

1993;7(2):85-8

85. Knowles MA, Shaw ME, Proctor AJ.

Deletion mapping of chromosome 8 in

cancers of the urinary bladder using

restriction fragment length

polymorphisms and microsatellite

polymorphisms. Oncogene

1993;8(5):1357-64

86. Bova GS, Carter BS, Bussemakers MJ,

et al. Homozygous deletion and frequent

allelic loss of chromosome 8p22 loci in

human prostate cancer. Cancer Res

1993;53(17):3869-73

87. Emi M, Fujiwara Y, Ohata H, et al.

Allelic loss at chromosome band

8p21.3-p22 is associated with progression

of hepatocellular carcinoma.

Genes Chromosomes Cancer

1993;7(3):152-7

88. Yan SM, Tang JJ, Huang CY, et al.

Reduced expression of ZDHHC2 is

associated with lymph node metastasis

and poor prognosis in gastric

adenocarcinoma. PLoS One

2013;8(2):e56366

89. Mayer TC, Kleiman NJ, Green MC.

Depilated (dep), a mutant gene that

affects the coat of the mouse and acts in

the epidermis. Genetics

1976;84(1):59-65

90. Kang R, Wan J, Arstikaitis P, et al.

Neural palmitoyl-proteomics reveals

dynamic synaptic palmitoylation. Nature

2008;456(7224):904-9

91. Martin BR, Wang C, Adibekian A, et al.

Global profiling of dynamic protein

palmitoylation. Nat Methods

2012;9(1):84-9

. Report on improvement of palmitoyl-

proteomic quantification by

incorporating stable isotope labeling of

cells into previous click labeling

method; applicability in measuring

dynamic palmitoylation.

92. Roth AF, Wan J, Bailey AO, et al.

Global analysis of protein palmitoylation

in yeast. Cell 2006;125(5):1003-13

. First demonstration of large-scale

analysis of palmitoylated proteins.

93. Hemsley PA, Weimar T, Lilley KS, et al.

A proteomic approach identifies many

novel palmitoylated proteins in

Arabidopsis. New Phytol

2013;197(3):805-14

94. Tom CT, Martin BR. Fat chance!

Getting a grip on a slippery

modification. ACS Chem Biol

2013;8(1):46-57

95. Zhou F, Xue Y, Yao X, et al. CSS-Palm:

palmitoylation site prediction with a

Targeting protein palmitoylation

Expert Opin. Drug Discov. (2014) 9(9) 13

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsita

et Z

ueri

ch o

n 06

/30/

14Fo

r pe

rson

al u

se o

nly.

Page 14: Targeting protein palmitoylation: selective inhibitors and implications in disease

clustering and scoring strategy (CSS).

Bioinformatics 2006;22(7):894-6

96. Ren J, Wen L, Gao X, et al. CSS-Palm

2.0: an updated software for

palmitoylation sites prediction.

Protein Eng Des Sel 2008;21(11):639-44

97. Xue Y, Liu Z, Cao J, et al. editors.

Computational prediction of

post-translational modification sites.

Intech China, Shanghai; 2011

98. Hu LL, Wan SB, Niu S, et al. Prediction

and analysis of protein palmitoylation

sites. Biochimie 2011;93(3):489-96

99. Shi SP, Sun XY, Qiu JD, et al. The

prediction of palmitoylation site locations

using a multiple feature extraction

method. J Mol Graph Model

2013;40:125-30

100. Kumari B, Kumar R, Kumar M.

PalmPred: an SVM based palmitoylation

prediction method using sequence profile

information. PLoS One

2014;9(2):e89246

101. Baker TL, Zheng H, Walker J, et al.

Distinct rates of palmitate turnover on

membrane-bound cellular and oncogenic

H-ras. J Biol Chem

2003;278(21):19292-300

102. Fukata Y, Bredt DS, Fukata M. Protein

Palmitoylation by DHHC Protein

Family. In: Kittler JT, Moss SJ, editors.

The dynamic synapse: molecular methods

in ionotropic receptor biology. 2011/01/

05 ed CRC Press, Boca Raton; 2006

103. Brocklehurst K, Carlsson J, Kierstan MP,

et al. Covalent chromatography by thiol-

disulfide interchange. Methods Enzymol

1974;34:531-44

104. Ivaldi C, Martin BR, Kieffer-Jaquinod S,

et al. Proteomic analysis of S-acylated

proteins in human B cells reveals

palmitoylation of the immune regulators

CD20 and CD23. PLoS One

2012;7(5):e37187

105. Jones ML, Collins MO, Goulding D,

et al. Analysis of protein palmitoylation

reveals a pervasive role in Plasmodium

development and pathogenesis.

Cell Host Microbe 2012;12(2):246-58

106. Marin EP, Derakhshan B, Lam TT,

et al. Endothelial cell palmitoylproteomic

identifies novel lipid-modified targets and

potential substrates for protein acyl

transferases. Circ Res

2012;110(10):1336-44

107. Forrester MT, Hess DT, Thompson JW,

et al. Site-specific analysis of protein

S-acylation by resin-assisted capture.

J Lipid Res 2011;52(2):393-8

108. Zhang J, Planey SL, Ceballos C, et al.

Identification of CKAP4/p63 as a major

substrate of the palmitoyl acyltransferase

DHHC2, a putative tumor suppressor,

using a novel proteomics method.

Mol Cell Proteomics 2008;7(7):1378-88

109. Li MH, Dong LW, Li SX, et al.

Expression of cytoskeleton-associated

protein 4 is related to lymphatic

metastasis and indicates prognosis of

intrahepatic cholangiocarcinoma patients

after surgery resection. Cancer Lett

2013;337(2):248-53

110. Hemsley PA, Grierson CS. Multiple roles

for protein palmitoylation in plants.

Trends Plant Sci 2008;13(6):295-302

111. Hang HC, Geutjes EJ, Grotenbreg G,

et al. Chemical probes for the rapid

detection of Fatty-acylated proteins in

Mammalian cells. J Am Chem Soc

2007;129(10):2744-5

112. Kostiuk MA, Corvi MM, Keller BO,

et al. Identification of palmitoylated

mitochondrial proteins using a bio-

orthogonal azido-palmitate analogue.

FASEB J 2008;22(3):721-32

113. Kostiuk MA, Keller BO, Berthiaume LG.

Palmitoylation of ketogenic enzyme

HMGCS2 enhances its interaction with

PPARalpha and transcription at the

Hmgcs2 PPRE. FASEB J

2010;24(6):1914-24

114. Martin BR. Nonradioactive analysis of

dynamic protein palmitoylation.

Curr Protoc Protein Sci

2013;73:Unit 14 5

115. Fukata M, Fukata Y, Adesnik H, et al.

Identification of PSD-95 palmitoylating

enzymes. Neuron 2004;44(6):987-96

116. Leong WF, Zhou T, Lim GL, et al.

Protein palmitoylation regulates

osteoblast differentiation through BMP-

induced osterix expression. PLoS One

2009;4(1):e4135

117. Resh MD. Use of analogs and inhibitors

to study the functional significance of

protein palmitoylation. Methods

2006;40(2):191-7

118. Webb Y, Hermida-Matsumoto L,

Resh MD. Inhibition of protein

palmitoylation, raft localization, and T

cell signaling by 2-bromopalmitate and

polyunsaturated fatty acids. J Biol Chem

2000;275(1):261-70

119. Coleman RA, Rao P, Fogelsong RJ, et al.

2-Bromopalmitoyl-CoA and 2-

bromopalmitate: promiscuous inhibitors

of membrane-bound enzymes.

Biochim Biophys Acta

1992;1125(2):203-9

120. Chenette EJ, Abo A, Der CJ. Critical

and distinct roles of amino- and

carboxyl-terminal sequences in regulation

of the biological activity of the Chp

atypical Rho GTPase. J Biol Chem

2005;280(14):13784-92

121. Pedro MP, Vilcaes AA, Tomatis VM,

et al. 2-Bromopalmitate reduces protein

deacylation by inhibition of acyl-protein

thioesterase enzymatic activities.

PLoS One 2013;8(10):e75232

122. Davda D, El Azzouny MA, Tom CT,

et al. Profiling targets of the irreversible

palmitoylation inhibitor

2-bromopalmitate. ACS Chem Biol

2013;8(9):1912-17

123. Mitchell DA, Mitchell G, Ling Y, et al.

Mutational analysis of Saccharomyces

cerevisiae Erf2 reveals a two-step reaction

mechanism for protein palmitoylation by

DHHC enzymes. J Biol Chem

2010;285(49):38104-14

124. Gonzalez Montoro A, Quiroga R,

Maccioni HJ, et al. A novel motif at the

C-terminus of palmitoyltransferases is

essential for Swf1 and Pfa3. Biochem J

2009;419(2):8

125. Draper JM, Smith CD. Palmitoyl

acyltransferase assays and inhibitors

(Review). Mol Membr Biol

2009;26(1):5-13

126. Ducker CE, Griffel LK, Smith RA, et al.

Discovery and characterization of

inhibitors of human palmitoyl

acyltransferases. Mol Cancer Ther

2006;5(7):1647-59

. Describes the most recent

improvement in the development

of PAT.

127. Jennings BC, Nadolski MJ, Ling Y, et al.

2-Bromopalmitate and 2-(2-hydroxy-5-

nitro-benzylidene)-benzo[b]thiophen-3-

one inhibit DHHC-mediated

palmitoylation in vitro. J Lipid Res

2009;50(2):233-42

128. Rocks O, Peyker A, Kahms M, et al. An

acylation cycle regulates localization and

activity of palmitoylated Ras isoforms.

Science 2005;307(5716):1746-52

129. Duncan JA, Gilman AG. A cytoplasmic

acyl-protein thioesterase that removes

B. Chavda et al.

14 Expert Opin. Drug Discov. (2014) 9 (9)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsita

et Z

ueri

ch o

n 06

/30/

14Fo

r pe

rson

al u

se o

nly.

Page 15: Targeting protein palmitoylation: selective inhibitors and implications in disease

palmitate from G protein alpha subunits

and p21(RAS). J Biol Chem

1998;273(25):15830-7

130. Hirano T, Kishi M, Sugimoto H, et al.

Thioesterase activity and subcellular

localization of acylprotein thioesterase

1/lysophospholipase 1.

Biochim Biophys Acta

2009;1791(8):797-805

131. Tian L, McClafferty H, Knaus HG,

et al. Distinct acyl protein transferases

and thioesterases control surface

expression of calcium-activated potassium

channels. J Biol Chem

2012;287(18):14718-25

132. Devedjiev Y, Dauter Z, Kuznetsov SR,

et al. Crystal structure of the human acyl

protein thioesterase I from a single X-ray

data set to 1.5 A. Structure

2000;8(11):1137-46

133. Hadvary P, Sidler W, Meister W, et al.

The lipase inhibitor tetrahydrolipstatin

binds covalently to the putative active

site serine of pancreatic lipase.

J Biol Chem 1991;266(4):2021-7

134. Hedberg C, Dekker FJ, Rusch M, et al.

Development of highly potent inhibitors

of the Ras-targeting human acyl protein

thioesterases based on substrate similarity

design. Angew Chem Int Ed Engl

2011;50(42):9832-7

135. Zimmermann TJ, Burger M, Tashiro E,

et al. Boron-based inhibitors of acyl

protein thioesterases 1 and 2.

ChemBioChem 2013;14(1):115-22

136. Bachovchin DA, Brown SJ, Rosen H,

et al. Identification of selective inhibitors

of uncharacterized enzymes by high-

throughput screening with fluorescent

activity-based probes. Nat Biotechnol

2009;27(4):387-94

137. Adibekian A, Martin BR, Chang JW,

et al. Confirming target engagement for

reversible inhibitors in vivo by kinetically

tuned activity-based probes. J Am

Chem Soc 2012;134(25):10345-8

. First report on development of isoform

specific inhibitors of acylprotein

thioesterases (APT1 and APT2).

138. Fabian MA, Biggs WH III, Treiber DK,

et al. A small molecule-kinase interaction

map for clinical kinase inhibitors.

Nat Biotechnol 2005;23(3):329-36

139. Karaman MW, Herrgard S, Treiber DK,

et al. A quantitative analysis of kinase

inhibitor selectivity. Nat Biotechnol

2008;26(1):127-32

140. Feng Y, Yin Y, Weiser A, et al.

Discovery of substituted 4-(pyrazol-4-yl)-

phenylbenzodioxane-2-carboxamides as

potent and highly selective Rho kinase

(ROCK-II) inhibitors. J Med Chem

2008;51(21):6642-5

141. Goytain A, Hines RM, Quamme GA.

Huntingtin-interacting proteins,

HIP14 and HIP14L, mediate dual

functions, palmitoyl acyltransferase and

Mg2+ transport. J Biol Chem

2008;283(48):33365-74

AffiliationBurzin Chavda, John A Arnott &

Sonia Lobo Planey†

†Author for correspondence

The Commonwealth Medical College,

Department of Basic Sciences, Scranton,

PA 18509, USA

E-mail: [email protected]

Targeting protein palmitoylation

Expert Opin. Drug Discov. (2014) 9(9) 15

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsita

et Z

ueri

ch o

n 06

/30/

14Fo

r pe

rson

al u

se o

nly.