• No results found

The role of abca1 in atherosclerosis: lessons from in vitro and in vivo models - Chapter 7 Efflux and Atherosclerosis: The clinical and biochemical impact of variations in the ABCA1 gene

N/A
N/A
Protected

Academic year: 2021

Share "The role of abca1 in atherosclerosis: lessons from in vitro and in vivo models - Chapter 7 Efflux and Atherosclerosis: The clinical and biochemical impact of variations in the ABCA1 gene"

Copied!
25
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

s

The role of abca1 in atherosclerosis: lessons from in vitro and in vivo models

Singaraja, R.R.

Publication date

2003

Link to publication

Citation for published version (APA):

Singaraja, R. R. (2003). The role of abca1 in atherosclerosis: lessons from in vitro and in vivo

models.

General rights

It is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s)

and/or copyright holder(s), other than for strictly personal, individual use, unless the work is under an open

content license (like Creative Commons).

Disclaimer/Complaints regulations

If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please

let the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the material

inaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letter

to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. You

will be contacted as soon as possible.

(2)

Chapter r

Effluxx and Atherosclerosis: The clinical and

biochemicall impact of variations in the ABCA1 gene

Roshnii R. Singaraja

1

*, Liam R. Brunham

1

*, Henk Visscher

1

, John J.P.

Kastelein

22

and Michael R. Hayden

1

"Thee Centre for Molecular Medicine and Therapeutics, University of British Columbia and Children'ss and Women's Hospital, Vancouver, B.C., V5Z4H4, Canada

:

Thee Department of Vascular Medicine, Academic Medical Centre, Amsterdam,, the Netherlands 'Thesee authors contributed equally to this work.

(3)

Abstract t

Approximatelyy 50 mutations and many SNPs have been described in the ABCA1 gene, with mutationss leading to Tangier Disease and Familial Hypoalphalipoprotememia- Homozygotes andd heterozygotes for mutations in ABCA1 display a wide range of phenotypes. Identification off ABCA1 as the molecular defect in these diseases has allowed for ascertainment based on geneticc status, determination of genotype-phenotype correlations, and has permitted us to identifyy mutations conferring a range of severity of cellular, biochemical and clinical phenotypes Heree we review how genetic variation at the ABCA1 locus affects its role in the maintenance off lipid homeostasis and the natural progression of atherosclerosis.

Abbreviations: :

(HDL)) high density lipoprotein, (SNP) single nucleotide polymorphism, (CAD) coronary artery disease,, (TD) tangier disease, (FHA) familial hypoalphalipoproteinemia, (NBF) nucleotide binding fold d

(4)

Partt I: The ABCA1 gene and its biological role

Atheroscleroticc coronary artery disease (CAD) constitutes the major public health burden in developedd countries, and by 2020 is predicted to be the single greatest cause of death worldwide ff I 2). Dc-ctc-aseJ HDLC is the must common iipiü abnormality in patients with premature CAD (4,5).. ABCA1 encodes the key protein regulating the efflux of lipids from peripheral cells to HDL,, following which these lipids are transported back to the liver and excreted as bile in a processs termed reverse cholesterol transport (6). At least 50 mutations have been identified in thee ABCA1 gene, leading t o the allelic disorders Tangier Disease (TD) and Familial Hypoalphalipoprotememiaa (FHA), which are associated with a wide range of phenotypic consequencess and putative biochemical defects. Here we review how genetic alterations of thee ABCA1 gene highlight its role in lipid homeostasis and atherosclerosis. Although TD is exceedinglyy rare, and ABCA1 mutations appear to be an infrequent cause of FHA, the study of m u t a t i o n ss in this gene has shed new light on a key pathway in the pathogenesis of atherosclerosis,, and opened up new approaches for its prevention and treatment.

1.11 Super-family: The ATP-binding cassette (ABC) transporters

Thee transport of specific molecules across membranes is critical for survival, and the ABC proteinss transport a wide variety of substances including lipids and sterols, metabolic products andd drugs across both intra and extracellular membranes (7). The first ABC transporter was clonedd in 1982 (8). ABC transporters are the largest membrane transporter family, consisting off 48 members in humans (9), 52 members in the mouse (10), 56 in Drosophila (9), 58 in C

eleganselegans (10), 31 in yeast (11) and 129 in Arabidopsis (10). The genome of E. coli contains 80

ABCC transporters, corresponding to 2 % of its genome (12). Despite their large numbers and substratee diversity, all ABC proteins bind and hydroiyze ATP and use the derived energy for transportt of the various molecules (13-15).

1.22 Classification and topology of ABC proteins and ABCA1

Knowledgee of the normal topology and organization of ABC proteins and ABCA1 is important ass it provides insight into potential functional domains. ABC transporters are defined based on thee presence of ATP binding domains, also known as nucleotide binding folds (NBFs) which conlamm three characteristic conserved regions, the Walker A and B domains, which are separated byy approximately 90-110 amino acids, and a signature (C) motif, located just upstream of the Walkerr B site (16,17) (Figure 1). Using flag tags and antibodies, and also by deciphering the glycosylationn status of ABCA1, the most current topological analysis reveals that ABCA1 consists off t w o large extracellular loops, one between the first and second transmembrane domains, andd the other following the intracellular NBF1 domain (18,19) (Figure 1),

(5)

Figuree 1. Schematic diagram of ABCA1 with each octagon representing 2 amino acids. The location of the

transmembranee domains (blue), extracellular loops (purple), Walker A motifs (green), Walker B motifs (royal) and Walkerr C signatures (red) and their residues are indicated.

domainss (MSD), each usually comprised of six membrane spanning a-helices which provide substratee specificity (20)

Thee mammalian ABC genes are divided into seven subfamilies, ABCA to ABCG, based on similarityy in gene structure, order of the domains, and sequence homology in the NBF and TM domains.. Figure 2 shows the phylogenetic relationship of selected ABC family members, and thee percent identity and similarity of these proteins.

1.33 Mutations in ABC genes cause many human genetic diseases:

Geneticc variation in ABC genes have been shown to be the cause of at least 12 genetic diseasess (Table 1). The ABCA1 gene is highly conserved between species (Figure 3). Human ABCA11 is 95.2% identical to mouse, 85.3% to chicken, 25.5% to drosophila, 21.6% to C.

eleganselegans and 10 2 % identical to fugu ABCA1. We have performed identity and similarity

searches,, and generated a phylogenetic tree (Figure 3). Although the C elegans gene CED7 wass proposed as the orthologue of ABCA1 based on similarities of function (36), the C.

(6)

eleganselegans t r a n s p o r t e r CE2 ( Q 9 T X V 8 ) is t h e closest m e m b e r t o A B C A 1 , as s u p p o r t e d by similar

analysiss by Peelman a n d colleagues (37). Thus A B C A 1 a n d CED7 are likely t o be paralogs a n d n o tt o r t h o l o g s .

similarity y

ABCA11 ABCA4 ABCA7 ABCA2 A B C B 1 ABCC1 ABCC7 ABCG5

A B C A 11 ABCA1 A B C A 4 4 6 0 0 0 A B C A 2 2 ABCA7 7 ABCG5 5 ABCA4 4 ABCA2 2 ABCC11 ABCB1 ABCC77 . „ „ „ . . ABCC1 1 ABCC7 7 4 8 2 2 544 5 4 5 9 177 3 1788 15 1 1 7 22 141 1800 15.1 9.4 4 244 7 23 0 134 300 9 10 6 identity y

Figuree 2. (A) The phylogenetic relationship of selected ABC Family Proteins. Sequences from ABCA1 (Genbank

accessionn number NM_00502), ABCA2 (NM_001606), ABCA4 (NM_000350): ABCA7 (AF_328787), ABCB1

(NM_000927),, ABCC1 (NM_004996), ABCC7 (NM_000492) and ABCG5 (NM_022436) were aligned using Clustall X. 1.8. A Neighbourhood-Joining tree was generated in ClustalX and viewed using phylodendron webservice att http://iubio.bio.indiana.edu/treeapp/treeprint-form,html. (B) Identity/similarity table of selected ABC family memberss generated using Clustal X 1.8. The same Genbank sequences as above were used.

Tablee 1 . Mutations in ABC genes are associated with several genetic diseases.

Genee Disease ABCA1 1 ABCA4 4 ABCB4 4 ABCB7 7 ABCB11 1 ABCC6 6 ABCC8 8 ABCD1 1 ABCC2 2 ABCC7 7 ABCG5andABCG8 8

Tangierr disease, familial Hypoalphalipoproteinemia (21)

Stargardtt disease, Retinitis pigmentosum 19, cone-rod dystrophy, age-related macularr degeneration (22, 23)

Progressivee familial intrahepatic cholestasis type 3 (PFIC) (24) X-linkedd Sideroblastic anemia and cerebellar ataxia (25) Progressivee familial intrahepatic cholestasis type 2 (26, 27, 28) Pseudoxanthomaa elasticum (29)

Persistentt hypermsulinemic hypoglycemia of Infancy (PHHI) (30) X-lmkedd adrenoleukodystrophy (31)

Dubin-Johnsonn syndrome (32) Cysticc fibrosis (33, 34) Sitosterolemiaa (35;

(7)

AA B s i m i l a r i t y y ii D. M e l a n o g a s t e r H.. M. G. D. C F. S a p i e n ss M u s c u l u s G a l l u s M e l a n o g a s t e r E l e g a n s R u b r i p e s r-- H. S a p i e n s H. S a p i e n s — 96 6 89 9 36.9 28 0 19 8 II | i-j M. M u s c u l u s 95 2 — 89.5 37.0 33 9 19 6 —— M. M u s c u l u s :: a G a l l u s 85 3 85 1 — 37 4 33 7 19 7

i i

G.. G a l l u s D. M e l a n o g a s t e r 25 5 25 6 25 8 — 4u 7 1 5 ' C.. E l e g a n s 2 1 6 jj C. E l e g a n s F.. R u b r i p e s 10 2 11 F R u b r i p e s i d e n t i t y y

Figuree 3. (A) A phylogenetictreeof ABCA1 orthologuesfromH. sapiens (NM_005502), M. musculus (NMJ313454),

GG gallus (AF 362377), C. elegans (AF_ 101313), D melanogaster (NM_1 34601) and F lubripes (Hayden laboratory, unpublishedd data) were aligned using Clustal X 1.8, Neighbourhood-Joining tree was generated in ClustalX and viewedd using phylodendron webservice at httpy/iubio. bio.indiana.edu/treeapp/treeprint-form html as above. (B) Ann identity/similarity matrix consisting of the orthologues of ABCA1 was generated as described above.

Partt II: Variation in ABCA1:

Insightss into protein function and its contribution to atherosclerosis

2.11 Mutations in ABCA1

Att least 50 mutations in the ABCA1 gene have been identified (21,38-54,Hayden group unpublishedd data). These include 23 missense, 6 nonsense and 21 insertions or deletions. Forty-ninee of the reported mutations occur in exons. One mutation in intron 2 leads to abnormally splicedd transcripts lacking exon 2 or exon 4, or both (48). All mutations by definition result in decreasedd lipid efflux. The extremely high correlation between phospholipid and cholesterol effluxx (r = 0.86, p <0 0001 ) in more than 15 mutations tested (Singaraja and Hayden, unpublishedd data) indicates that ABCA1 influences efflux of both lipid types

2.22 Non-random distribution of mutations in t h e ABCA1 gene

Whilee mutations do occur throughout the gene, mutations in ABCA1 are not in random distribution.. Four mutations cluster between amino acids 230 and 282, 6 between residues 5877 and 635, 8 occur between amino acids 909 and 1099, 5 between residues 1145 and 12899 and 5 mutations occur between 2144 and 2215 (Figure 4). Conversely, with the exception off a large deletion, only one mutation occurs in the transmembrane regions between residues 6366 and 908, and no mutations occur in the second set of transmembrane domains. Many of thee residues harboring mutations are highly conserved with C elegans, a nematode that is

21.77 2 1 6 28 0 — 16 6

(8)

IVS22 +5 G>C —-R282X X

Figuree 4. Location of mutations in the ABCA1 protein. At least 50 mutations have been reported thus far in the

ABCA11 gene, many of which show a non-random distribution and cluster in specific regions of the protein (yelloww boxes).

estimatedd to have diverged from other metazoans 600-1,200 million years ago (55,56), thus indicatingg their functional importance (Table 2).

2.33 Functional effects of mutations in the extracellular loops of ABCA1

Approximatelyy half of the missense mutations in the ABCA1 gene associated with TD and FHA falll within the t w o extracellular loops. Mutations in the first and second extracellular loops mightt be expected to result in a lack of lipid efflux caused by dysfunctional interaction of ABCA11 with ApoA-l, since lipid poor pre-B HDL particles would require either direct interaction orr close proximity to ABCA1 in order for the lipid transfer to occur. Thus the extracellular loops off ABCA1 might provide a potential binding target for ApoA-l. Impaired transport of ABCA1 to thee plasma membrane could prevent interaction with ApoA-l. It Is also possible that mutations inn the extracellular domains will result in a disruption of the tertiary structure of ABCA1, therebyy preventing its function at the plasma membrane where it is normally localized. This conceptt is further supported by studies of ABCR, which shows very similar topology to ABCA1 (57).. The t w o halves of ABCR each contain a large extracellular loop, a transmembrane domain regionn and a nucleotide binding domain that interact together through one or more disulfide bonds,, putatively involving cysteines located in the extracellular loops in ABCR (57). Interestingly, mostt of the cysteines are conserved in these regions between ABCA1 and ABCR (57) suggesting

(9)

Tablee 2. Conservation of amino acid residues mutated in the human ABCA1 gene. MUTATION N P8BL L R230C C A255T T R587W W W590S S Q597R R \ L 6 9 3 3 T929I I N935S/H H A937V V A1046D D M1091T T D1099Y Y D1289L/N N C1477R R S1506L L N161ID D R1680W W N1800H H F20095 5 R2081W W P2150L L AE1893 3 ADD 1894 H,Sapiens s P P R R A A R R W W Q Q L L T T N N A A A A M M D D D D C C S S N N R R N N F F R R P P E E D D M.Musculus s P P R R A A R R W W Q Q L L T T N N A A A A M M D D D D C C

s s

N N R R N N F F R R P P E E D D G.Gallus s P P R R S S R R W W 0 0 L L T T N N A A A A M M D D D D C C

s s

N N R R N N F F R R P P E E D D D.Meianogaster r P P R R Q Q L L T T N N A A A A M M D D D D N N R R A A 1 1 R R R R D D D D C.EIeg g P P G G 0 0 Q Q L L T T N N A A A A M M D D D D S S R R W W M M R R N N S S D D

23/244 (95.83%) mutations are conserved down to the chicken

thatt these residues are essential for folding and interactions between the different domains. Of note,, ABCA1 contains a cysteine at position 1477 that is mutated to an argmme residue (21) andd could thereby disrupt proper three dimensional folding of the ABCA1 protein necessary forr its ability to efflux lipids.

Furtherr insights into how the mutations R587W, W590S and Q597R that occur in the extracellularr loops affect ABCA1 function have recently been described (58-60). Two studies havee reported that ABCA1 containing the point mutation Q597R, which occurs in the first extracellularr loop, does not localize to the plasma membrane (59,60). However, other studies havee reported that this mutant is expressed at the plasma membrane, but at reduced levels relativee to wildtype ABCA1 (58,44). R587W, another missense mutation in the first extracellular loop,, also prevents the trafficking of ABCA1 to the plasma membrane, although results with thiss mutant have been variable (58-60). Both the R587W and Q597R mutants are resistant to PNGasee digestion indicating that they are not glycosylated, suggesting that ABCA1 harboring thesee mutations does not traverse the medial and trans Golgi network. However, not all mutations inn the extracellular loops prevent the export of ABCA1 to the plasma membrane. ABCA1

(10)

harboringg the W590S mutation does reach the cell surface and cross-linking studies reported normall interaction of the W590S mutant with ApoA-l despite defective efflux, suggesting that interactionn with ApoA-l may not be sufficient for lipid efflux (58),

ABCA11 that dues nol leach the plasma membrane cannot induce the binding of ApoA-l. This iss indeed the case with mutant Q597R (58,60) which shows no ApoA-l binding. However, failuree of binding may also occur due to disruption of residues crucial for this function. Indeed, thee variants C1477R and S1506L which are both localized in the second large extracellular loopp are normally translocated to the plasma membrane but show no ApoA-l binding, indicating thatt specific amino acids in the large extracellular loops are also necessary for ApoA-l binding. A l t h o u g hh thus far only a small subset of the naturally occurring mutations have been biochemicallyy characterized, these in vitro studies of ABCA1 mutations have begun to provide valuablee information on structure-function relationships of the protein.

2.44 Mutations in t h e transmembrane domain and impact on ABCA1 function

Onlyy one mutation has been described in the transmembrane domain of ABCA1. five mutations havee been described in the transmembrane domains of the ABCR gene. Small deletions and mutationss that introduce charged amino acids into transmembrane regions of ABCR result in greatlyy reduced amounts of ABCR protein (22,61). Mutations in the transmembrane domain regionn of ABCA1 might also disrupt ABCAI's integration into membranes and therefore prevent itt from exiting the endoplasmic reticulum and the golgi, or prevent its integration into the plasmaa membrane. This could result in the rapid turnover of the mutant ABCA1 protein. The onlyy described mutation in ABCA1 that occurs in the transmembrane domain, AL693, results inn the deletion of one amino acid. ABCA1 with this mutation does not exit from the endoplasmic reticulumm and therefore it also shows no ApoA-i binding (60).

2.55 The integrity of the nucleotide binding folds is essential for ABCA1 function

Severall mutations have been described in the NBF region of the ABCR gene, and these mutants aree defective in ATP binding (22,61). Thus ABCA1 harboring mutations in the NBFs may not generatee the ATP necessary for active transport of substrates. Although ABCA1 harboring thesee mutations would be expected to show defects in lipid efflux which may be energy requiring,, no detect in localization to the plasma membrane is expected in these mutants. A totall of 6 missense mutations have been described in the NBF region between the Walker A andd B motifs. None of these mutations have yet been characterized biochemically for their abilityy to bind ATP, their localization, their ability to bind ApoA-l, or their ability to induce lipid efflux.. Interestingly, of the six mutations in the NBFs, five occur in the first NBF but only one occurss in the second NBF. The t w o NBFs in ABCR show significantly different ATP binding and hydrolysiss properties, with the NBF1 being active as an ATPase and binding ATP, CTP, GTP and UTPP (62). The NBD1 of the CFTR protein also shows greater affinity for ATP than does NBD2

(11)

(63,64).. Studies of the MRP1 molecule have shown that ATP binding at NBD1 induces conformationall changes in the protein and enhances ADP trapping at NBD2 (65). These data suggestt that the two NBD domains in ABCA1 have differential function, with NBD1 being rate-limitingg for proper function.

2.66 Critical role of the C-terminus of ABCA1

Thee CFTR protein, another homolog of ABCA1, is usually targeted to the apical surface of cells Whenn the C-terminal portions of CFTR are disrupted, there is a redistribution of the protein to bothh the apical and basolateral surfaces of cells, as well as a reduction in its half-life (66). In additionn the C-terminus of the CFTR protein contains a PDZ binding domain that when mutated causess redistribution of the protein (67).

ABCA11 also contains a PDZ binding domain in its C-terminus (43,68) which when mutated couldd lead to protein mislocalization. The functional significance of the PDZ domain remains to bee determined, though binding of PDZ proteins has been shown to occur (68). Mutations in thee C-terminus of ABCA1 might similarly impact the normal targeting of ABCA1 to the basolateral surfacee in polarized cells and influence its stability. The five naturally occurring mutations in thee C-terminal region of ABCA1 have yet to be functionally characterized.

2.77 The nature of mutations in ABCA1 contributes t o t h e biochemical, cellular and clinicall phenotype

Itt is likely that the phenotypic heterogeneity in TD patients or in those heterozygous for ABCA11 deficiency might at least in part be accounted for by the nature of the mutation and its effectt on the protein. Until the genetic basis for TD and FHA were discovered, these patients weree diagnosed based on their phenotype. Since the genetic defect underlying both diseases hass been discovered, the assignment of disease has been mainly based on genotype with heterozygotess for ABCA1 mutations being classified as having FHA, and those homozygous forr ABCA1 mutations being categorized as having TD. Thus, in the past there was a potential forr underascertainment, with TD patients carrying mutations conferring milder phenotypes beingg classified as having FHA, and those heterozygous for very mild mutations not being recognizedd at all. The converse is also true, with those carrying severe heterozygous mutations beingg designated as having TD. Indeed, phenotypic variability of TD is now readily apparent withh some TD patients having very low HDL levels (<1%), and others having >10% compared too age and sex matched controls.

Basedd on the knowledge of the mutations in affected individuals, it is now possible to ascertain functionall deficits for a spectrum of phenotypes associated w i t h either heterozygosity or homozygosityy for mutations in ABCA1 (Figure 5). For TD, individuals with severe clinical phenotypes mayy show no ABCA1 protein at the plasma membrane or have ABCA1 at the plasma membrane thatt is completely lacking in function. This could result from two null alleles for ABCA1 preventing

(12)

normal l C A DD Incidence A B C A 11 Activity A B C A 11 Allele 1 A B C A 11 Allele 2 normal l normal l FHA A normal l >50% % HDL-C C normal l -50% % HDL-C C normal l <50% % HDL-C C TD D 10-15% % HDL-C C 10-15% % HDL-C C 1-4% % HDL-C C 1-4% % HDL-C C A255T T W590S S T929I I A937V V R1680W W P85L L R587W W \L693 3 R909X X N935S S A1046D D D1099Y Y C1477R R \E,DD 1893/94 R2081W W 2145X X 2203X X M1091T T A255T T Q597R R R1680W W 635X X N935S S N1800H H 1851X X 2203X X C-termm deletion

Figuree 5. A model showing genotype/phenotype correlations for heterozygous and homozygous mutations in

ABCA1.. HDL-C levels reported in the literature for patients harboring specific mutations in ABCA1 were compared too age and sex matched control HDL-C levels from the LRC database. Increasing CAD incidence and decreasing HDL-CC levels correlating to decreasing ABCA1 activity is modeled.

exportt of the protein to the plasma membrane, or from ABCA1 at the plasma membrane harboring mutationss in residues crucial for its function. Indeed patients harboring the mutations 635X, N935S,, N1800H, 1851X, 2203X and the large C-terminal deletion all have below 1 % of HDL-C levelss of age and sex matched controis from the LRC population.

Patientss homozygous for the mutations A255T and R1680W show HDL-C levels that are greater thann 10% of age and sex matched population controls. These patients with a less severe clinicall phenotype imply that the ABCA1 protein shows some residual activity.

Inn general, of those patients heterozygous for ABCA1 mutations, those with a mutation resulting inn completely dysfunctional protein w o u l d be expected to show HDL-C levels that are approximatelyy 50% of those for age and sex matched controls. The protein generated from thiss allele either would not be expected to reach the plasma membrane or would affect a residuee essential for its function. Most patients harboring heterozygous mutations do show HDL-CC levels that are close to 50% of those of normal age and sex matched controls. Somee patients heterozygous for ABCA1 mutations show >50% HDL-C levels compared to controlss when environmental effects such as age are controlled for. These patients would be hypothesizedd to synthesize a mutant ABCA1 protein that shows some residual activity. In this case,, mutant ABCA1 would be localized at the plasma membrane, and show a mild defect.

(13)

Thiss is indeed the case in heterozygous patients harboring mutations A255T, W590S, T929I, R1680WW and A937V who all show HDL-C levels >75% of normal age and sex matched controls. Anotherr possibility is that heterozygotes for mutations in ABCA1 show <50% of normal levels off HDL-C. This could be caused by dominant negative effects of ABCA1, as previously shown forr truncation mutations (69). Patients harboring the mutation M1091T show HDL-C levels thatt are 30% of age and sex matched controls (70). This finding suggests that ABCA1 acts as aa dimer or as part of a complex in the exertion of its function.

2.88 ABCA1 heterozygotes have increased atherosclerosis

Priorr to the cloning of the ABCA1 gene, studies of obligate heterozygotes had reported conflicting findingss on whether individuals heterozygous for mutations in ABCA1 are at an increased risk off developing CAD (71,72). This is not surprising considering that patients harboring mutations inn ABCA1 show a wide range of phenotypes, and thus misclassification of patients was likely. Cloningg of the gene and descriptions of the mutations allowed for the direct assessment of atherosclerosiss in heterozygotes. In one large study of 13 different mutations in 11 families, bothh w i t h TD and FHA, phenotypic analysis in a cohort of heterozygous individuals was undertakenn (70). The control cohort consisted of unaffected family members. A greater than three-foldd increase in CAD in adult heterozygotes compared to controls, with earlier age of onsett (by 10 years) was evident. Intriguingly, the relative cholesterol efflux levels were directly relatedd to CAD, with families with the clearest evidence for premature CAD having individuals withh the lowest cholesterol efflux.

However,, several caveats were evident in this study. Firstly, the collection of the kindreds may havee been biased by clinical sampling, since only families with the most severe phenotypes mayy have presented at clinics. Secondly, a very low number of events were seen. Thirdly, using ann endpoint of CAD as an outcome measure might have underestimated the effects. CAD is an insensitivee marker for atherosclerosis and does not address the effects of mutations on the naturall history of presymptomatic atherogenesis.

Too address these issues, a second study elucidating the association between mutations in ABCA11 and surrogate markers, namely, increased arterial wall thickness and ABCA1 mediated cholesteroll efflux was performed (73). The study group consisted of 30 individuals heterozygous forr four different missense mutations in the ABCA1 gene, C1477R, M1091T, P2150L and T929I.. Importantly, the mean intima-media thickness in carriers was higher than in controls andd carriers for mutations in ABCA1 also showed increased progression of arterial thickening thatt reached the upper limit of normal (0.8mm) much earlier (55 years) when compared to controlss (80 years, p<0.0001). Similar to the previous study, regression analysis of the data fromm this study indicated that a 50% increase in ABCA1 mediated cholesterol efflux would resultt in a 3 0 % increase in HDL-C concentrations, and that this could translate into a 35-50% reductionn in the risk of CAD.

(14)

Interestingly,, mutations in ABCA1 do affect susceptibility to atherosclerosis not only by influencingg lipids but by direct effects on the vessel wall (74). Nitric oxide is derived from the vascularr endothelium, and is a crucial antiatherogenic agent that maintains vascular homeostasis. Diminishedd NO availability represents an e-aily step in atheiuscleiusis. ABCA1 hetetozygotes showw significantly impaired endothelial function with impaired basal and stimulated nitric oxidee activity when compared to controls, indicating that ABCA1 affects vessel wall function.

3.11 Single Nucleotide Polymorphisms (SNPs) in ABCA1

Tenn coding single nucleotide polymorphisms (cSNPs) along w i t h hundreds of non-coding SNPss have been described in the ABCA1 gene (see Table 3) (41,75-80). Among the non-coding SNPss are 9 promoter and 5' UTR variants that have been analyzed for functional significance (76,78,79,81).. Most cSNPS are found distal to known functional domains (Figure 6). In addition, thee amino acid residues affected by cSNPs are less conserved compared to those affected by mutationss (50% compared to >95%, Table 4).

Tablee 3. Regulatory and coding single nucleotide polymorphisms described thus far in the ABCA1 gene, and

theirr relative frequencies in the general population. Nucleotide e -1095A/G G -477C/T T -419A/C C -320G/C C -191G/C C C69T T C117G G lnsG319 9 G378C C G1051A A T1591C C G2706A A A2715C C G2723C C G2826A A A3044G G G3911C C G5255A A C5587G G ndd = not determined Aminoo Acid promoter r promoter r promoter r promoter r promoter r 5'UTR R 5'UTR R 5'UTR R 5'UTR R R219K K V399A A V771M M T774P P K776N N V825! ! I883M M E1172D D R1587K K S1731C C Exon n

— —

--1 --1 1 1 2 2 2 2 7 7 11 1 16 6 16 6 16 6 17 7 18 8 24 4 35 5 38 8 Frequency y nd d 0.42 2 0.01 1 0.42 2 0.42 2 0.138 8 0.065 5 0.085 5 nd d 0.254 4 0.008 8 0.029 9 0 0 0 3 3 0 0 0 3 3 0.0811 - 0.541 0.0799 - 0 737 0.026 6 0.259 9 nd d

3.22 Association of ABCA1 cSNPs and regulatory SNPs w i t h HDL and atherosclerosis

Soonn after mutations in ABCA1 were found to be causative of TD and FHA, our laboratory and otherss investigated whether common variation in ABCA1 could contribute to variation in HDL-CC levels and atherosclerosis in the general population (41,75,77).

(15)

I883M M

E1172D D

Figuree 6. Distribution of cSNPs in the ABCA1 gene. Interestingly, no cSNPs have been found thus far in the

c-terminuss of the ABCA1 gene.

Tablee 4. Conservation of amino acid residues polymorphic in humans

cSNP P R219K K V399A A V771M M T774P P K776N N V825I I I883M M E1172D D R1587K K S1731C C apiens s R R V V V V T T

< <

V V I I E E R R 5 5 M.Musculus s R R V V V V S S K K V V V V E E K K S S G.Gallus s K K V V V V

s s

K K A A P P E E K K S S D.M M elanogaster r A A L L 5 5 K K M M R R E E T T C.EIegans s L L Y Y G G R R

_ _

A A

--V --V H H

5/100 (50%) of cSNPs are conserved to the chicken

Remarkably,, of the 10 cSNPs described, 6 are associated with potential functional effects, includingg alterations of lipid levels or measures of atherosclerosis (see Table 5). However, it is importantt to note that few of these findings have been replicated, and some results are inconsistent.. Still, it is remarkable that so many cSNPs have been associated with functional effects,, suggesting that ABCA1 may be a major atherosclerosis susceptibility iocus in the generall population. The R219K, V771M and I883M variants have been recognized as putative

(16)

Tablee 5. Clinical and biochemical consequences of ABCA1 cSNPs

Nucleotidee Ammo Acid/position Lipids s CAD/Atherosclerosis s Anti-atherogpnirr SNPs C-17G G IN5G319 9 G1051A A A3044G G promoterr no change j 5'UTRR no change J, R219KK JTG, THDL, fApoAl, TApoB, | LDL j I883MM JTG, f HDL f Pro-atherogenicc SNPs -191C/-320 0 G-19K K A-1095G G C117G G G2706A A G2868A A G3911C C G5155A A C/-477TT haplotype promoter r promoter r promoter r 5'UTR R V771M M V825I I E1172D D R1587K K noo change noo change noo change | T G G noo change noo change JJ HDL, JApoB JJ HDL

t t

t t

I I

noo change

t t

t t

T T

noo change

Associationss with lipid levels and coronary artery disease (CAD) are shown by arrows to represent the direction of association. .

anti-atherogenicc polymorphisms, associated with increased HDL-C and decreased TG levels (K2199 and M883), and increased HDL-C and ApoA-l (M771) (41,75,82). The E1172D, R1587K cSNPss have been reported to be associated with decreased HDL-C (75).

Fivee promoter SNPs are associated with increased severity of atherosclerosis, including the -191C/-320C/-477TT haplotype (76,78) as well as the G-191C and A-1096G SNPs. In contrast thee C-17G variant was associated with less atherosclerosis (78). Interestingly, these SNPs are nott associated with changes in lipid levels, suggesting that changes in ABCA1 activity can occurr without changes in steady state plasma lipid levels. The V825I, I883M and E1172D SNPs havee also been associated with increased clinical events and severity of atherosclerosis (75,77).

3.33 The R219K Variant

Thee R219K SNP has been most studied, and highlights many of the difficulties associated with thee study of SNPs in general. At least 8 studies have examined the role of this SNP in lipid homeostasiss or atherogenesis (75,77,79,81-84) (Table 6). Of these, 5 have reported positive associationn with either increased HDL-C or reduced severity of atherosclerosis. This number of positivee replications in independent studies consistently in the same direction indicates that thiss is truly an important variant with a significant athero-protective effect.

Inn 3 studies, the athero-protective effect of the K219 allele was observed only in certain circumstances,, for example in w o m e n (83), in individuals with elevated Lp(a) or with the ApoE3/E33 genotype (82), or in smokers (84). These findings indicate the functional effect of K2199 may be particularly significant in certain genetic and environmental backgrounds.

(17)

Tablee 6. Functional associations reported for the R219K variant. HDL-C C Atherosclerosis s 3rousseauu et al '.,85; Cleeett al. (83) Takagii et al t.87) Lutucutaa et al. (89) Yamakawa-Kobayashii eta (personall communication] Evanss et al. (90) nc c T T Kakkoetal.(91)) t Cenarroo et al (92) nc

ncc - no change - - not assesed

Tablee 7. Ethnic variation of SNP frequency in the ABCA1 gene.

TG G

nc c nc c

comments s

Differencess in changes of HDL-C withh age observed

Onlyy in ApoE3/E3(TG)and elevatedd Lpiaj (atherosclerosis)

ndividuals s Onlyy in women

FHH patients, K allele more protectivee in smokers R219K K V825I I I883M M E1172D D R1587K K Dutch h 0.254 4 0.081 1 0.136 6 0.026 6 0.259 9 Japanese e 11 51 0.54 4 1.60 0 nd d 0.71 1 German n 0.26 6 nd d nd d nd d nd d Finn n 0.22 2 nd d 0.112 2 0.026 6 nd d Inuit t nd d 0.200 0 0.294 4 nd d 0.561 1 Oji-Cree e nd d 0.250 0 0.690 0 nd d 0.378 8 South-Asian n nd d 0.053 3 0.145 5 nd d 0.303 3 ndd - not determined

Thee 3 reports that found no association with HDL-C or severity of atherosclerosis may have beenn confounded by population admixture, as t w o of these studies examined ethnically mixed urbann U.S. populations (77,81). The frequencies of ABCA1 cSNPs are highly divergent across populationss (Table 7), and interestingly, the 'minor' K219 allele is actually the wildtype allele in aa cohort of 327 Japanese school-aged children (Yamakawa-Koboyashi, personal communication). Linkagee disequilibrium among ABCA1 cSNPs is a further confounding variable, which to date hass not been adequately addressed. Clee et al. reported significant LD between the R219K and thee V771M, K776N, I883M, and R1587K cSNPs, but found that after carriers of these variants weree excluded, R219K remained significantly associated with degree of atherosclerosis and TG levelss (75). Further study of population-specific patterns of LD among these SNPs and haplotype analysiss should clarify these results. In addition, biochemical and functional assessment of thesee SNPs is needed for definitive clarification of their effects.

(18)

3.44 ABCA1 SNPs may be associated w i t h changes in atherosclerosis independent of changess in HDL-C levels

Off the 12 cSNPs and regulatory SNPs associated with alterations in plasma lipid levels or atherosclerosis,, 7 display altered severity of atherosclerosis without detectable changes in lipid levels.. This suggests that while ABCA1 may be an important atherosclerosis susceptibility locus,, the mechanisms by which it exerts this effect is not necessarily by altering steady state HDL-CC levels. The non-coding SNPs G-191C, C-69T, C-17G and lnsG319 and the cSNPs R219K, V 7 7 1 M ,, and V825I have all been found to be associated w i t h differences in severity of atherosclerosiss but not with changes in HDL-C levels in at least one study. The implication is that HDLL quality and composition, as determined by ABCA1-mediated efflux, may be a determinant of thee efficiency of reverse cholesterol transport, without actually affecting the levels of circulating HDL-C.. These results are consistent with efflux influencing atherogenesis without necessarily changingg lipid levels. Several studies have recently provided evidence into how this may occur. Bonee marrow transplant experiments between ABCA1 null and wildtype mice have demonstrated thatt deficiency of macrophage ABCA1 is associated with small changes in lipid levels, but significant increasess in atherosclerosis (85,86). This concept has been recapitulated by the study of ABCA1 BACC transgenic mice, in which a significant protection f r o m atherosclerosis is evident with minimall changes in HDL-C levels (87). Taken together, these studies indicate that ABCA1 can influencee atherogenesis independent of steady state HDL-C levels.

Conclusion n

Thee study of TD, a rare disorder of lipoprotein metabolism with less than 60 reported cases wondwiue,, ied to the identhication Oi the lunctionai impact oi the ABCA1 gene and protein. Variationn of this protein has now been shown to confer a risk for atherosclerosis in the general population,, and has provided an answer to a question asked for many years, namely, how lipidss are effluxed from cells in the first step of reverse cholesterol transport. The study of this raree disorder in a few has led to the identification of a validated drug target that offers hope forr raising HDL and prevention of atherosclerosis in many.

Acknowledgements s

Thiss work was supported by grants from the Canadian Institutes of Health Research and the Heartt and Stroke Foundation of British Columbia and Yukon. M.R.H. is an established investigator off the University of British Columbia and the Children and Women's Hospital and is a holder off a Canada Research Chair in human genetics. Prof. John J. Kastelein is an established investigatorr of the Netherlands Heart Foundation (2000D039)

(19)

References s

1,, Yusuf 5, Reddy S, Ounpuu S, and Anand S. Global burden of cardiovascular diseases, part I: general considerations, thee epidemiologic transition, risk factors, and impact of urbanization. Circulation 2001;104:2746-2753 2.. Murray CJL and Lopez AD. Alternative projections of mortality and disability by cause 1990-2020: Global

Burdenn of Disease Study. Lancet 1997;349:1498-1504.

33 Murray CJL and Lopez AD. Global mortality, disability, and the contribution of risk factors: Global Burden of Diseasee Study. Lancet 1997;349:1436-1442

4.. Genest JJr., Bard J-M, Fruchart J-C, Ordovas JM, and Schaefer EJ Familial hypoalphaiipoproteinemia in prematuree coronary artery disease Artenoscler Thromb 1993; 13:1728-1737.

5.. Genest JJr, Martin-Munley SS, McNamara JR, Ordovas JM, Jenner J, Myers RH, Silberman SR, Wilson PWF, Salemm DN.and Schaefer EJ. Familial lipoprotein disorders in patients with premature coronary artery disease. Circulationn 1992;85:2025-2033.

6.. Glomset JA. The plasma lecithimcholesterol acyltransferase reaction J Lipid Res 1968;9.1 55-167.

7.. Holland IB, Cole SPC, Kuchler K, and Higgins CF. ABC Proteins: From Bacteria to Man. San Diego: Academic Press;; 2003.

8.. Higgins CF, Haag PD, Nikatdo K, Ardeshir F, Garcia G, and Ames GF. Complete nucleotide sequence and identificationn of membrane components of the histidine transport operon of S. typhimurium. Nature 1982;298:723-727. .

9.. Dean M, RzhetskyA, and Allikmets R. The human ATP-binding cassette (ABC) transporter superfamily. Genome Ress 2001 ;11:1156-1166.

10.. Dean M. The Human ATP-Binding Cassette (ABC) Transporter Superfamily. Bethesda: National Library of Medicine,, 2002.

11.. Bauer BE, Wolfger H, and Kuchler K. Inventory and function of yeast ABC proteins: about sex, stress, pleiotropicdrugg and heavy metal resistance. Biochim Biophys Acta 1999; 1461:217-236.

12.. Linton KJ and Higgins CF. The Escherichia coli ATP-binding cassette (ABC) proteins. Mol Microbiol 1998,28:5-13. 13.. Higgins CF. ABC transporters: From microorganisms to man. Annu Rev Cell Biol 1992;8:67-1 1 3.

14.. Childs S and Ling V. The MDR superfamily of genes and its biological implications Important Adv Oncol 1994,21-36. .

11 5. Dean M a n d Allikmets R. Evolution of ATP-binding cassette transporter genes. CurrOpin Genet Dev 1995;5:779-785. .

16.. Hyde SC, Emsley P, Hartshorn MJ, Mimmack M M , Gileadi U, Pearce SR, Gallagher MP, Gill DR, Hubbard RE, and Higginss CF Structural model of ATP-binding proteins associated with cystic fibrosis, multidrug resistance and bacteriall transport. Nature 1990;346:362-365.

11 7. Gottesman M M , and Ambudkar SV. Overview:: ABC transporters and human disease. J Bioenerg Biomembr 2001,33:453458. .

18.. Fitzgerald ML, Mendez AJ, Moore KJ, Andersson LP, Panjeton HA, and Freeman MW. ATP-binding cassette transporterr Al contains an NH -terminal signal anchor sequence that translocates the protein's first hydrophiiic domainn to the exoplasmic space. J Biol Chem 2001;276:1 51 37-15145.

(20)

19.. Tanaka AR, Ikeda Y, Abe-Dohmae 5, Arakawa R, Sadanami K, Kidera A, Nakagawa S, Nagase T, Aoki R, Kioka N, Amach,, T, Yokoyama S, and Ueda K Human ABCA1 contains a large amino-terminal extracellular domain homologouss to an epitope of Sjogren's Syndrome. Biochem Biophys Res Commun 2001 ;283:1019-1025 200 Dean M HamonY andChiminiC, The human ATP-hindinn rasspttPi'ARr1, tran^pnrtpr ^iir-prfflmily 11 ipid RPC

-2001;42:1007-1017. .

2 1 .. Brooks-Wilson A, Marcil M, Clee SM, Zhang L, Roomp K, van Dam M, Yu L, Brewer C, Collins JA, Molhuizen HOF, Loubserr O, Ouellette BFF, Fichte, K, Ashbourne Excoffon KJD, Sensen CW, Scherer S, Mott S, Denis M, Martindale D,, Frohlich J, Morgan K, Koop B, Pimstone SN, Kastelein JJP, Genest JJr., and Hayden MR. Mutations in ABC1 in Tangierr disease and familial high-density lipoprotein deficiency. Nat Genet 1999;22:336-345.

22.. Allikmets R, Singh N, Sun H, Shroyer NF, Hutchinson A, Chidambaram A, Gerrard B, Baird L, Stauffer D, Peiffer A,, Rattner A, Smallwood P, Li Y, Anderson KL, Lewis RA, Nathans J, Leppert M, Dean M, and Lupski JR. A photoreceptorr cell-specific ATP-binding transporter gene (ABCR) is mutated in recessive Stargardt macular dystrophy.. Nat Genet 1997; 15:236-245.

23.. Martinez-MirA, BayesM, Vilageliu L, Grinberg D, AyusoC, del RioT, Garcia-Sandoval B, Bussaglia E, Baiget M, Gonzalez-Duartee R, and Balcells S. A new locus for autosomal recessive retinitis pigmentosa (RP19) maps to 11 p i 3-1 p21. Genomics 1997;40:142-146.

24.. Dixon PH, Weerasekera N, Linton KJ, Donaldson O, Chambers J, Egginton E, Weaver J, Nelson-Piercy C, de Swiett M, Warnes G, Elias E, Higgins CF, Johnston DG; McCarthy Ml, and Williamson C. Heterozygous MDR3

mi5sensee mutation associated with intrahepatic cholestasis of pregnancy: evidence for a defect in protein trafficking.. Hum Mol Genet 2000;9:1209-1217.

25.. Allikmets R, Raskind W H , Hutchinson A, Schueck ND, Dean M, and Koeller DM. Mutation of a putative mitochondriall iron transporter gene (ABC7) in X-linked sideroblastic anemia and ataxia (XLSA/A). Hum Mol Genett 1999;8:743-749.

26.. Strautnieks SS, Bull LN, Knisely AS, Kocoshis SA, Dahl N, Arnell H, Sokal E, Dahan K, Childs S, Ling V, Tanner MS, Kagalwallaa AF, Nemeth A, Pawlowska J, Baker A, Mieli-Vergani G, Freimer NB, Gardiner RM, and Thompson RJ. AA gene encoding a liver-specific ABC transporter is mutated in progressive familial intrahepatic cholestasis. Nat Genett 1998;20:233-238.

27.. Jansen PL, Strautnieks SS, Jacquemin E, Hadchouel M, Sokal EM, Hooiveld GJ, Koning JH, Jager-Krikken A, Kuiperss F, Stellaard F, Bijleveid CM, Gouw A, Van Goor H, Thompson RJ,and Muller M. Hepatocanalicularbile saltt export pump deficiency in patients with progressive familial intrahepatic cholestasis. Gastroenterology 1999;117:1370-1379. .

28.. Wang L, Soroka CJ, and Boyer JL. The role of bile salt export pump mutations in progressive familial intrahepatic cholestasiss type II. J Clin Invest 2002; 110:965-972.

29.. Bergen AA, Plomp AS, Schuurman EJ, Terry S, Breuning M, Dauwerse H, Swart J, Kool M, van Soest S, Baas F, ten Brinkk JB, and de Jong PT. Mutations in ABCC6 cause pseudoxanthoma elasticum. Nat Genet 2000;25:228-231. 30.. Nichols CG, Shyng SL, Nestorowicz A, Glaser B, Clement JP, Gonzalez G, Aguilar-Bryan L, Permutt MA, and Bryan

J.. Adenosine diphosphate as an intracellular regulator of insulin secretion. Science 1996;272:1785-1 787. 3 1 .. Smith KD, Kemp S, Braiterman LT, Lu JF, Wei HM, Geraghty M, Stetten G, Bergin JS, Pevsner J, and Watkins

PA.. X-linked adrenoleukodystrophy: genes, mutations, and phenotypes. Neurochem Res 1999,24:521-535. 32.. Paulusma CC, Kool M, Bosma PJ, Scheffer GL, ter Borg F, Scheper RJ, Tytgat GN, Borst P, Baas F, and Oude

Elferink,, RP. A mutation in the human canalicular multispecific organic anion transporter gene causes the Dubin-Johnsonn syndrome. Hepatology 1997,25:1 539-1 542.

(21)

33.. Riordan JR, Rommens JM, Kerem B, Alon N, Rozmahel R, Grzelczak Z, Zielenski J, Lok S, Plavsic N, Chou JL, Drummm ML, lannuzzi MC, Collins FC, and Tsui LC. Identification of the cystic fibrosis gene: cloning and characterizationn of complementary DNA. Science 1989;245:1066-1073.

34.. Kerem B, Rommens JM, Buchanan JA, Markiewicz D, Cox TK, Chakravarti A, Buchwald M, and Tsui LC. Identificationn of the cystic fibrosis gene: genetic analysis. Science 1989;245:1073-1080.

35.. Berge KE, Tian H, Graf GA, Yu L, Grishin NV, Schultz J, Kwiterovich P, Shan B, Barnes R, and Hobbs HH Accumulationn of dietary cholesterol in sitosterolemia caused by mutations in adjacent ABC transporters. Sciencee 2000;290:1771-1775.

36.. Broccardo C, Luciani M.F, and Chimin! G. The ABCA subclass of mammalian transporters. Biochim Biophys Actaa 1999,1461:395-404.

37.. Peelman F, Labeur C, Vanloo B, Roosbeek S, Devaud C, Duverger N, Denefle P, Rosier M, Vandekerckhove J, andd Rosseneu M. Characterization of the ABCA transporter subfamily: identification of prokaryotic and eukaryoticc members, phylogeny and topology. J Mol Biol 2003,325:259-274.

38.. Hong SH, Riley W, Rhyne J, Friel G, and Miller M. Lack of association between increased carotid intima-media thickeningg and decreased HDL-cholesterol in a family with a novel ABCA1 variant, G2265T. Clin Chem 2002;48:2066-2070. .

39.. Rust S, Rosier M, Funke H, Amoura Z, Piette J-C, Deleuze J-F, Brewer HB Jr., Duverger N, Denèfle P, and Assmannn G. Tangier Disease is caused by mutations in the gene encoding ATP-binding cassette transporter 1. Natt Genet 1999;22:352-355.

40.. Bodzioch M, Orsó E, Klucken J, Langmann T, Böttcher A, Diederich W, Drobnik W,, Barlage S, Buchler C, Porsch-Özcurümezz M, Kaminski WE, Hahmann HW, Oette K, Rothe G, Aslanidis C, Lackner KJ, and Schmitz G. The gene encodingg ATP-binding cassette transporter 1 is mutated in Tangier Disease. Nat Genet 1999;22:347-351 4 1 .. Wang J, Burnett JR, Near S, Young K, Zinman B, Hanley AJ, Connelly PW, Harris SB, and Hegele RA. Common and

raree ABCA1 variants affecting plasma HDL cholesterol. Arterioscler Thromb Vase Biol 2000;20:1983-1989. 42.. Nishida Y, Hirano K, Tsukamoto K, Nagano M, Ikegami C, Roomp K, Ishihara M, Sakane N, Zhang Z, Tsujii KK,

Matsuyamaa A, Ohama T, Matsuura F, Ishigami M, Sakai N, Hiraoka H, Hattori H, Wellington C, Yoshida Y, Misugii S, Hayden MR, Egashira T, Yamashita S, and Matsuzawa Y, Expression and functional analyses of novel mutationss of ATP-binding cassette transporter-1 in Japanese patients with high-density lipoprotein deficiency. Biochemm Biophys Res Commun 2002;290:713-721

43.. Brousseau ME, Shaefer EJ, Dupuis J, Eustace B, Van Eerdewegh P, Goldkamp AL, Thurston LM, FitzGerald MG, Yasek-McKennaa D, O'Neill G, Eberhart GP, Weiffenbach B, Ordovas JM, Freeman MW, Brown RH Jr., and Gu JZ.. Novel mutations in the gene encoding ATP-binding cassette 1 in four Tangier disease kindreds J Lipid Res 2000;41:433-441. .

444 Lawn RM, Wade DP, Garvin MR, Wang X, Schwartz K, Porter JG, Seilhamer JJ, Vaughan A M , and Oram JF. The Tangierr disease gene product ABC 1 controls the cellular apolipoprotein-mediated lipid removal pathway. J Clin Investt 1999;104:R25-R31.

455 Lapicka-Bodzioch K, Bodzioch M, Krull M, Kielar D, Probst M, Kiec B, Andnkovics H, Bottcher A, Hubacek J, Aslanidiss C, SuttorpN, and Schmitz G Homogeneous assay based on 52 primer sets to scan for mutations of thee ABCA1 gene and its application in genetic analysis of a new patient with familial high-density lipoprotein deficiencyy syndrome. Biochim Biophys Acta 2001,1 537:42-48

(22)

46.. Huang W, Moriyama K, Koga T, Hua H, Ageta M, Kawabata S, Mawatan K, Imamura T, Eto T, Kawamura M, Teramotoo T, and Sasaki J Novel mutations in ABCA1 gene in Japanese patients with Tangier disease and familial highh density lipoprotein deficiency with coronary heart disease. Biochim Biophys Acta 2001; 1 537:71-78. 47.. Ishu J, Nagano M, Kujiraoka T, Ishihara 171. Fgashira T, Takada D, Tsuji M, Hattori H. and Emi M ninirnl variant

off I angier disease in Japan: mutation of the ABCA1 gene in hypoalphalipoprotememia with corneal lipidosis. JJ Hum Genet 2002;47:366-369.

48.. Altilia S, Pisciotta L, Garuti R, Tarugi P, Cantafora A, Calabresi L, Tagliabue J, Maccari S, Bernini F, Zanotti I, Verganii C, Bertolini S, and Calandra S. Abnormal splicing of ABCA1 pre-mRNA in Tangier disease due to a IVS2 +5G>CC mutation in ABCA1 gene. J Lipid Res 2003;44:254-264.

49.. Guo Z, Inazu A, Yu W, Suzumura T, Okamoto M, Nohara A, Higashikata T, Sano R, Wakasugi K, Hayakawa T, Yoshidaa K, Suehiro T, Schmitz G, and Mabuchi H. Double deletions and missense mutations in the first nucleotide-bmdingg fold of the ATP-binding cassette transporter A1 (ABCA1) gene in Japanese patients with Tangierr disease. J Hum Genet 2002;47:325-329.

50.. Bertolini 5, Pisciotta L, Seri M, Cusano R, Cantafora A, Calabresi L, Franceschini G, Ravazzolo R, and Calandra S.. A point mutation in ABC1 gene in a patient with severe premature coronary heart disease and mild clinical phenotypee of Tangier disease. Atheroscler 2001 , 1 54:599-605.

51.. Hong SH, Rhyne J, Zeiler K, and Miller M. Novel ABCA1 compound variant associated with HDL cholesterol deficiency.. Biochim Biophys Acta 2002,1 587:60-64.

52.. Hong SH, Rhyne J, Zeiler K, and Miller M. ABCA1 (Alabama): a novel variant associated with HDL deficiency and prematuree coronary artery disease. Atheroscler 2002; 164:245-250.

53.. RemaleyAT, RustS, Rosier M, Knapper C, NaudmL, BroccardoC, Peterson KM, KochC, Arnould I, PradesC, Duvergerr N, Funke H, Assman G, Dinger M, Dean M, Chimini G, Santamanna-Fojo S, Fredrickson DS, Denefle P,, and Brewer HB Jr. Human ATP-binding cassette transporter 1 (ABC1): genomic organization and identification off the genetic defect in the original Tangier disease kindred. Proc Natl Acad Sci U S A 1999;96:12685-12690. 54.. Asztalos BF, Brousseau ME, McNamara JR, Horvath KV, Roheim PS, and Schaefer EJ. Subpopulations of high

densityy lipoproteins in homozygous and heterozygous Tangier disease Atheroscler 2001; 1 56:217-225. 55.. Blaxter M. Caenorhabditis elegans is a nematode. Science 1998;282:2041 -2046

56.. Feng DF, Cho G, and Doolittle RF. Determining divergence times with a protein clock: update and reevaluation. Procc Natl Acad So U S A 1997,94:13028-13033

577 Bungert S, Molday LL, and Molday RS. Membrane topology of the ATP binding cassette transporter ABCR and itss relationship to ABC1 and related ABCA transporters: identification of N-linked glycosylation sites. J Biol Chemm 2001 ;276:23539-23546.

38.. Fitzyeidld ML, Morris AL, Rhee JS, Andersson LF, MendezAJ, and Freeman MW. Naturally occurring mutations inn the largest extracellular loops of ABCA1 can disrupt its direct interaction with apolipoprotem A-l. J Biol Chem 2002,2777 33178-33187.

59.. Tanaka AR, Abe-Dohmae S, Ohnishi T, Aoki R, Mormaga G, Okuhira KI, Ikeda Y, Kano F, Matsuo M, Kioka N, Amachii T, Murata M, Yokoyama S, and Ueda K. Effects of mutations of ABCA1 in the first extracellular domain onn subcellular trafficking and ATP binding/hydrolysis. J Biol Chem 2002;278:8815-8819.

60.. Rigot V, Hamon Y, Chambenoit O, Alibert M, Duverger N, and Chimini G Distinct sites on ABCA1 control distinctt steps required for cellular release of phospholipids J Lipid Res 2002,43:2077-2086.

(23)

6 1 .. Sun, H Smallwood PM, and Nathans J Biochemical detects in ABCR protein variants associated with human retinopathies.. Nat Genet 2000;26:242-246

622 Biswas EE. Nucleotide binding domain 1 of the human retinal ABC transporter functions as a genera! ribonucleotidasee Biochemistry 2001 ;40:8181 -8187

63.. Ko YH and Pedersen PL The first nucleotide binding fold of the cystic fibrosis transmembrane conductance regulatorr can function as an active ATPase. J Biol Chem 1995;270:22093-22096.

644 Randak C, Neth P, Auerswald EA, Eckerskorn C, Assfalg-Machleidt i, and Machleidt W. A recombinant polypeptide modell of the second nucleotide binding fold of the cystic fibrosis transmembrane conductance regulator functionss as an active ATPase, GTPase and adenylate kinase FEBS Lett 1997;41 0:180-186.

65.. Hou YX, Riordan JR, and Chang XB. ATP binding, not hydrolysis, at the first nucleotide-binding domain of multidrugg resistance-associated protein MRP1 enhances ADP.Vi trapping at the second domain J Biol Chem 2003,2783599-3605 5

66.. Swiatecka-Urban A, Duhaime M, Coutermarsh B, Karlson KH, Collawn J, Milewski M, Cutting GR, Guggino WB, Langfordd G, and Stanton BA PDZ domain interaction controls the endocytic recycling of the cystic fibrosis transmembranee conductance regulator. J Biol Chem 2002,277 40099-40105.

67.. Short, DB Trotter KW, Reczek D, Kreda SM, Bretscher A, Boucher RC, Stutts MJ, and Milgram SL An apical PDZ proteinn anchors the cystic fibrosis transmembrane conductance regulator to the cytoskeleton J Biol Chem

1998;273:19797-19801 1

688 Buechler C, Boettcher A, Bared SM, Probst MC, and Schmitz G. The carboxytermmus of the ATP-binding cassettee transporter A1 interacts with a beta2-syntrophin/utrophm complex. Biochem Biophys Res Commun 2002,293:759-765 5

69.. Wellington CL, Yang YZ, Zhou S, Clee SM, Tan B, Hirano K, Zwarts K, Kwok A, Gelfer A, Marcil M, Newman S, Roompp K, Smgaraja R, Collins J, Zhang LH, Groen AK, Hovingh K, Brownlie A, Tafun S, Genest J Jr., Kastelein JJ,, andHayden MR. Truncation mutations in ABCA1 suppress normal upregulationof full-length ABCA1 by 9-cis-retinoicc acid and 22-R-hydroxycholesterol. J Lipid Res 2002,43:1939-1949.

70.. Clee SM, Kastelein JJ, van Dam M, Marcil M, Roomp K, Zwarts KY, Collins JA, Roelants R, Tamasawa N, Stulc T, Sudaa T, Ceska R, Boucher B, Rondeau C, DeSouich C, Brooks-Wilson A, Molhuizen HO, Frohlich J, Genest J Jr., andd Hayden MR Age and residual cholesterol efflux affect HDL cholesterol levels and coronary artery disease mABCAll heterozygotes J Clin invest 2000; 106:1 263-1 270

7 1 .. Assmann G, Simantke O, Schaefer HE, and Smootz E. Characterization of high density lipoproteins in patients neterozygousforr Tangiei disease J Clin invest 1977,60:102 5-1035

722 Schaefer EJ, Zech LA, Schwartz DE, and Brewer HB, Jr. Coronary heart disease prevalence and other clinica featuress in familial high-density lipoprotein deficiency (Tangier disease). Ann Intern Med 1980;93:261-266 733 van Dam MJ, de Groot E, Clee SM, Hovingh GK, Roelants R, Brooks-Wilson A, Zwinderman AH, Smit AJ, Smelt

AH,, Groen AK, Hayden MR, and Kastelein JJ. Association between increased arterial-wall thickness and impairment inn ABCA1-driven cholesterol efflux: an observational study. Lancet 2002,359:37-42

744 Bisoendia' R

J, Hovingh GK, Levels JHM, I erch PG, Andersen I, Hayden MR, Kastelein J, and St'oes ESG Impairmentt of Vascular Function is Rapidly Restored upon HDL increase in Isolated L O W - H D L Subjects Circulationn 2003;

(24)

75.. Clee SM, Zwindemnan AH, Engert JC, Zwarts KY, Molhmzen HO, Roomp K, Jukema JW, van Wijland M, van Damm M, Hudson TJ, Brooks-Wilson A, Genest J Jr., Kastelein JJ, and Hayden MR. Common genetic variation in ABCA11 is associated with altered lipoprotein levels and a modified risk for coronary artery disease. Circulation 2001,103:1198-1205 5

76.. Lutucuta S, Ballantyne CM, Elghannam H, Gotto A M Jr., and Marian AJ. Novel polymorphisms in promoter regionn of ATP binding cassette transporter gene and plasma lipids, severity, progression, and regression of coronaryy atherosclerosis and response to therapy. Circ Res 2001 ;88:969-973

77.. Brousseau ME, Bodzioch M, Schaefer EJ, Goldkamp AL, Kielar D, Probst M, Ordovas JM, Aslanidis C, Lackner KJ,, Rubins HB, Collins D, Robins SJ, Wilson PWF, and Schmitz G. Common variants in the gene encoding ATP-bindingg cassette transporter 1 in men with low HDL cholesterol levels and coronary heart disease. Atheroscler 2001;154:607-611. .

78.. Zwarts KY, Clee SM, Zwinderman AH, Engert JC, Smgaraja R, Loubser O, James E, Roomp K, Hudson TJ, Jukema JW,, Kastelein JJ, and Hayden MR. ABCA1 regulatory variants influence coronary artery disease independent of effectss on plasma lipid levels, Clin Genet 2002;61:11 5-125.

79.. Takagi S, Iwai N, Miyazaki S, Nonogi H, and Goto Y. Relationship between ABCA1 genetic variation and HDL cholesteroll level in subjects with ischemic heart diseases in Japanese. Thromb Haemost 2002;88:369-370. 80.. lida A, Saito S, Sekme A, Kitamura Y, Kondo K, Mishima C, Osawa S, Harigae 5, and Nakamura Y. High-density

single-nucleotidee polymorphism (SNP) map of the 1 50-kb region corresponding to the human ATP-bmding cassettee transporter A1 (ABCA1) gene. J Hum Genet 2001 ;46:522-528.

8 1 .. Lutucuta S, Ballantyne CM, Gotto A M , and Marian AJ. A Common Promoter Haplotype of ABCA1 Gene Reducess Promoter Activity, Alters Transcriptional Factors Binding and Is Associated with Progression of Coronaryy Atherosclerosis [abstract]. Circulation 2002; 106:11-168-11-168.

82.. Evans D, and Beil F-U. The association of the R219K polymorphism in the ATP-bindmg cassette transporter 1 (ABCA1)) gene with coronary artery disease and hyperlipidemia. J Mol Med 2003;81:264-270.

83.. Kakko S, Kelloniemi J, Kelloniemi J, von Rohr P, Hoeschele I, Tamminen M, Brousseau ME, Kesaniemi YA, and Savolainenn MJ. ATP-binding cassette transporter A1 locus is not a major determinant of HDL-C levels in a populationn at high risk for coronary heart disease. Atheroscler 2003,166:285-290.

84.. Cenarro A, Artieda M, Castillo S, Mozas P, Reyes G, Tejedor D, Alonso R, Mata P, Pocovi M, and Civeira F. A commonn variant in the ABCA1 gene is associated with a lower risk for premature coronary heart disease in familiall hypercholesterolemia. J Med Genet 2003;40:163-168

85.. Aiello RJ, Brees D, Bourassa PA, Royer L, Lmdsey S, Coskran T, Haghpassand M, and Francone OL. Increased atherosclerosiss in hyperlipidemic mice with inactivation of ABCA1 in macrophages. Arterioscler Thromb Vase Bioll 2002;22:630-637.

86.. Haghpassand M, Bourassa PA, Francone OL, and Aiello RJ. Monocyte/macrophage expression of ABCA1 has minimall contribution to plasma HDL levels. J Clin Invest 2001; 108:131 5-1320.

87.. Smgaraja RR, Bocher V, James ER, Clee SM, Zhang LH, Leavitt BR, Tan B, Brooks-Wilson A, Kwok A, Bissada N, Yangg YZ, Liu G, Tafuri SR, Fievet C, Wellington CL, Staels B, and Hayden MR. Human ABCA1 BAC transgenic micee show increased high density lipoprotein cholesterol and ApoAI-dependent efflux stimulated by an nternall promoter containing liver X receptor response elements in intron 1. J Biol Chem 2001,276:33969-33979. .

(25)

Referenties

GERELATEERDE DOCUMENTEN

clinical outcome. In patients with a poor prognosis, endovascular thrombolysis can be considered, but this procedure does not appear to be beneficial in patients with

ties en ondersteuning) en komt tot de conclusie dat beide hypothesen empirisch ondersteund worden: terwijl aan het begin van de levens- loop het mannelijke ‘overwicht’

If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, stating your reasons. In case of

Glazen inktpotten kwamen voor in de noord- westelijke provincies van het Romeinse rijk vanaf het midden van de eerste eeuw tot het begin van de tweede eeuw n.Chr. Het is opval-

The beginning passage of the Eighth Maḥberet goes as follows: “I wan- dered from the days of my youth through the outsides, going around in doing business, from people to people,

On this track segment four train types can be distinquished (Intercity trains, Inter-regional train service 8800, Regional train service 9800 and freight trains)?.

This thesis presents an innovative approach based on the theory of the Semi-Markovian Decision Processes and examines its potential as a dynamic delay management mechanism. The

If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, stating your reasons.. In case of