Tải bản đầy đủ (.pdf) (14 trang)

Báo cáo khoa học: Function and regulation of ABCA1 – membrane meso-domain organization and reorganization pptx

Bạn đang xem bản rút gọn của tài liệu. Xem và tải ngay bản đầy đủ của tài liệu tại đây (262.17 KB, 14 trang )

MINIREVIEW
Function and regulation of ABCA1 – membrane
meso-domain organization and reorganization
Kohjiro Nagao, Maiko Tomioka and Kazumitsu Ueda
Institute for Integrated Cell–Material Sciences (iCeMS), Kyoto University, Japan
Introduction
The plasma membrane is critical for the life of the cell,
not only as the boundary maintaining the cytosolic
environment differently from the extracellular environ-
ment, but also as a platform for protein assembly,
which converts extracellular stimuli into intracellular
signals. The skin lipid barrier prevents water loss from
our body, lipids in the myelin sheath functions as an
insulator of nerve fibers, and pulmonary surfactant lip-
ids at the air–water interface decrease alveolar surface
tension. These lipids are appropriately transported in
our body and within cells, and their abnormal deposi-
tion causes cell death and various diseases (Table 1).
Several ATP-binding cassette protein A (ABCA) sub-
family proteins are involved in lipid transport between
organs and between intracellular compartments
(Table 1). Furthermore, recent studies suggest that
ABCA1 moves lipids not only between different mem-
branes but also within the same membrane to organize
and reorganize submicrometre (meso-scale) membrane
domains such as lipid rafts. In this review, the function
and regulation of ABCA1 are summarized.
Cholesterol homeostasis and ABCA1
Cholesterol is a key component of the cell membrane
and is required for cell proliferation; however, excess
accumulation of cholesterol is toxic to cells and


its excess deposition in peripheral tissues causes
Keywords
ATP-binding cassette protein; cholesterol
homeostasis; lipid raft; membranes;
phospholipids
Correspondence
K. Ueda, Institute for integrated
Cell–Material Sciences (iCeMS), Kyoto
University, Kyoto 606-8502, Japan
Fax: 81 75 753 6104
Tel: 81 75 753 6124
E-mail:
(Received 17 December 2010, revised 27
February 2011, accepted 6 May 2011)
doi:10.1111/j.1742-4658.2011.08170.x
The ATP-binding cassette protein A1 (ABCA1) mediates the secretion of
cellular-free cholesterol and phospholipids to an extracellular acceptor,
apolipoprotein A-I, to form high-density lipoprotein. Because ABCA1 is a
key factor in cholesterol homeostasis, elaborate transcriptional and post-
transcriptional regulations of ABCA1 have evolved to maintain cholesterol
homeostasis. Recent studies suggest that ABCA1 moves lipids not only
between membranes but also within membranes to organize and reorganize
membrane meso-domains to modulate cell proliferation and immunity.
Abbreviations
ABCA1, ATP-binding cassette protein A1; apoA-I, apolipoprotein A-I; CK2, casein kinase 2; ECD, extracellular domain; HDL, high-density
lipoprotein; JAK2, Janus kinase 2; LXR, liver X receptor; LXRE, liver X response element; MbCD, methyl-b-cyclodextrin; NBD, nucleotide
binding domain; PC, phosphatidylcholine; PKA, protein kinase A; PKC, protein kinase C; PS, phosphatidylserine; RXR, retinoid X receptor;
SM, sphingomyelin; SREBP-2, sterol regulatory element binding protein 2.
3190 FEBS Journal 278 (2011) 3190–3203 ª 2011 The Authors Journal compilation ª 2011 FEBS
atherosclerosis. Excess cholesterol in peripheral tissues

is reversely transported as high-density lipoprotein
(HDL) to the liver. Because cholesterol is not catabo-
lized in peripheral tissues, HDL formation is the only
pathway by which excess cholesterol is removed from
peripheral cells. The inverse relationship between
plasma HDL levels and the risk of coronary artery dis-
ease is demonstrated [1], although genetically low
HDL per se may not predict the increased risk [2]. At
least 70 mutations have been identified in the ABCA1
gene, leading to Tangier disease and familial hypoal-
phalipoproteinaemia, in which patients have a near
absence of or decrease in circulating HDL [3–8]
(Fig. 1). More than 15 mutations examined show high
correlations between phospholipids, preferentially
phosphatidylcholine (PC) and cholesterol efflux [9–11],
indicating that ABCA1 influences the efflux of both
PC and free cholesterol. Indeed, ABCA1, expressed in
cultured cells, mediates the secretion of both types of
lipids when lipid-free apolipoprotein A-I (apoA-I), an
Table 1. Possible substrates, localization and related diseases of ABCA subfamily proteins. PE, phosphatidylethanolamine; PG, phosphatidyl-
glycerol.
Possible substrates
Subcellular
localization Tissue localization Related diseases
ABCA1 (similarity
with ABCA1)
PS [81]
PC and cholesterol [54]
Oxysterols [105]
Cell surface,

intracellular
vesicles [11,31]
Macrophages, liver,
small intestine,
brain [5,106–108]
Tangier’s disease,
atherosclerosis
[5–7,13]
ABCA2 (59.2%) SM, gangliosides [109] Endosome [110] Brain [111,112] Alzheimer’s
disease
[113–115]
ABCA3 (57.3%) PC, PG, PE [116–120] Intracellular vesicles,
lamellar bodies
[121–123]
Lung alveolar type II
cells [121,123]
Neonatal fatal
surfactant deficiency
and chronic interstitial
lung disease [124,125]
ABCA4 (66.7%) All-trans-retinal,
N-retinylidene-PE
[126–128]
Intracellular disc
membrane [129]
Photoreceptor cells
(rod cells, cone cells)
[129–131]
Stargardt muscular
dystrophy, age-related

macular degeneration
[132–135]
ABCA5 (50.3%) Unknown Lysosome, late
endosome [136]
Brain, lung, heart and
thyroid gland [136]
Dilated cardiomyopathy [136]
ABCA6 (48.6%) Unknown Unknown Unknown Unknown
ABCA7 (69.3%) PC, SM,
cholesterol
[77,79,137]
Plasma membrane
and intracellular
membranes
[77,79,137,138]
Spleen, lung, adrenal,
brain, liver, kidney (proximal
tubule), peritoneal
macrophages [79,137]
Unknown
ABCA8 (46.1%) Oestradiol-b-glucuronide,
taurocholate, LTC4,
p-aminohippuric acid,
ochratoxin-A [139]
Unknown Unknown Unknown
ABCA9 (48.2%) Unknown Unknown Macrophages [140] Unknown
ABCA10 (49.5%) Unknown Unknown Macrophages [141] Unknown
ABCA12 (56.3%) Glucosylceramide [142] Lamellar bodies [143] Keratinocyte [143] Harlequin ichthyosis [144]
ABCA13 (51.1%) Unknown Unknown Brain [145] Schizophrenia, bipolar
disorder, depression [145]

Fig. 1. Putative secondary structure of human ABCA1. Five cyste-
ine residues (C75, C309, C1463, C1465 and C1477) involved in the
two intramolecular disulfide bonds between ECD1 and ECD2 [73]
are indicated. The PEST sequence [59], the 1-5-8-14, the putative
calmodulin binding site [29], the PDZ binding motif [27,28] and PKA
[38] and CK2 [43] phosphorylation sites are indicated. (Not to
scale.)
K. Nagao et al. Function and regulation of ABCA1
FEBS Journal 278 (2011) 3190–3203 ª 2011 The Authors Journal compilation ª 2011 FEBS 3191
extracellular lipid acceptor in the plasma, is added to
the medium [11,12].
Transcriptional regulation of ABCA1
ABCA1-mediated cholesterol efflux is highly regulated
at the transcriptional level. In peripheral cells, such as
macrophages and fibroblasts, ABCA1 gene expression is
enhanced by loading cholesterol [13]. This response
is mediated by the nuclear receptors LXRa and
LXRb, whose ligands are sterol metabolites such as
22-(R)-hydroxycholesterol, 24-(S)-hydroxycholesterol,
27-hydroxycholesterol and 24-(S),25-epoxycholesterol
[14,15]. LXRb is ubiquitously expressed, whereas LXRa
is restricted to the liver, adipose tissue, adrenal glands,
intestine, lungs, kidneys and cells of myeloid origin.
Human LXRa expression is highly regulated and can be
autoregulated by itself, whereas human LXRb is stably
expressed even in the absence of excess cholesterol. In
the basal state, LXR b and retinoid X receptor (RXR)
heterodimers are bound to liver X response elements
(LXREs) in the promoters of target genes [16] (Fig. 2).
When cholesterol accumulates in cells, intracellular con-

centrations of oxysterols increase; subsequently, LXRb,
activated via the binding of oxysterols, stimulates the
transcription of ABCA1 [17–19] and also of LXRa.
Interestingly, cholesterol feeding of mice or rats has
failed to show a significant increase in hepatic ABCA1
mRNA expression [20]. A promoter region, which
responds to sterol regulatory element binding protein 2
(SREBP-2), was identified in the first intron of the
ABCA1 gene and was reported to be involved in the reg-
ulation of ABCA1 expression in the liver [21]. Recently,
MiR-33, an intronic microRNA located within the gene
encoding SREBP-2, a transcriptional regulator of cho-
lesterol synthesis, was found to modulate the expression
of ABCA1 at the post-transcriptional level [22,23]. An
elaborate network of regulations has evolved to modu-
late cholesterol synthesis and efflux to maintain choles-
terol homeostasis.
Post-translational regulation of ABCA1
activity
ABCA1-mediated cholesterol efflux is also highly regu-
lated at the post-translational level. Because cholesterol
is an essential component of cells, excessive elimination
of cholesterol can result in cell death. Consequently, the
ability to rapidly degrade ABCA1 in order to prevent
excessive elimination is also important. Indeed, ABCA1
protein turns over rapidly, with a half-life of 1–2 h
[24–28]. Several proteins, including a1-syntrophin,
A In the absence of excess cholesterol B When cholesterol accumulates
PC
Chol

apoA-I HDL
ABCA1
ABCA1
LXRβ/RXR
LXRα
RXR
Oxysterol
A
LXRβ
RXR
ABCA1
Cholesterol
BCA1
Fig. 2. LXR regulates ABCA1 not only in transcriptional level but also in post-translational level by direct binding. In addition to its well-
defined role in transcription, LXRb directly binds the C-terminal region of ABCA1 to mediate its post-translational regulation. (A) In the
absence of cholesterol accumulation, LXRb ⁄ RXR heterodimer binds to the C-terminal region of ABCA1. The ABCA1–LXRb ⁄ RXR complex sta-
bly localizes to the plasma membrane, but is inactive in HDL formation (30,147). (B) When excess cholesterol accumulates, oxysterols bind
to LXRb leading to its dissociation from ABCA1. Because ABCA1 turns over rapidly with a half-life of 1–2 h, and because the transcription,
splicing, translation and maturation of ABCA1, at more than 2000 amino acid residues, takes several hours after transcriptional activation,
cells cannot cope with an acute accumulation of cholesterol for several hours. This post-translational regulation allows ABCA1 to cause an
immediate early response against acute cholesterol accumulation. LXRb has at least two distinct roles in controlling cholesterol homeostasis
(modified from [148]).
Function and regulation of ABCA1 K. Nagao et al.
3192 FEBS Journal 278 (2011) 3190–3203 ª 2011 The Authors Journal compilation ª 2011 FEBS
b1-syntrophin, calmodulin and apoA-I have been
reported to interact with ABCA1 and reduce the rate of
ABCA1 protein degradation [25–29].
The degradation of ABCA1 is regulated [27,28,30]
and is carried out via several pathways: (a) cell-surface
ABCA1 is endocytosed and recycled back to the

plasma membrane or delivered to the lysosomes
through early and late endosomes for degradation
[31,32]; (b) calpain protease degrades ABCA1 on the
plasma membrane [25,26] and intracellularly, especially
when apoA-I does not bind to ABCA1 [33]. ABCA1 is
also degraded through the ubiquitin–proteasome path-
way [34,35]. COP9 signalosome complex, which plays
an important role in the degradation of various
proteins such as IjBa, associates with ABCA1 and
controls the ubiquitinylation and deubiquitinylation of
ABCA1 [36].
Several protein kinases including protein kinase A
(PKA), protein kinase C (PKC), Janus kinase 2 (JAK2)
and casein kinase (CK2) are involved in the regulation
of ABCA1 activity and stability by apoA-I. The inter-
action of apoA-I with ABCA1 increases the cellular
cAMP content and ABCA1 phosphorylation [37]. This
phosphorylation is important for ABCA1 activity, as
apoA-I-dependent phospholipid efflux is decreased sig-
nificantly by the mutation of the PKA phosphorylation
site, Ser-2054, of ABCA1 [38]. ApoA-I also activates
PKCa and phosphorylation of ABCA1 [39,40]. This
reaction leads to the protection of ABCA1 from its
degradation by calpain. On the other hand, phosphory-
lation of Thr-1286 and Thr-1305 in the PEST sequence
(rich in proline, glutamic acid, serine and threonine)
(Fig. 1) within the cytoplasmic domain of ABCA1 pro-
motes calpain degradation and is reversed by apoA-I
[41]. Unsaturated fatty acids destabilize ABCA1 by
phosphorylation through a PKCd pathway [42]. Phos-

phorylation of Thr-1242, Thr-1243 and Ser-1255 by
CK2 decreases the ABCA1 flippase activity, apoA-I
binding and lipid efflux [43]. Calmodulin interacts with
ABCA1 at a close or overlapping position to the CK2
phosphorylation site and this interaction regulates
calpain-mediated ABCA1 degradation [29]. The inter-
action of apoA-I with ABCA1 for only minutes stimu-
lates autophosphorylation of JAK2, which in turn
activates ABCA1-dependent lipid efflux [44]. Interest-
ingly, the apoA-I-mediated activation of JAK2 also
activates STAT3, which is independent of the lipid
efflux activity of ABCA1. The apoA-I ⁄ ABCA1 path-
way in macrophages is proposed to function as an anti-
inflammatory receptor through activation of JAK2 ⁄
STAT3 [45]. Cyclosporine A and FK506 were reported
to abolish ABCA1-dependent lipid efflux by inhibiting
the Ca
2+
-dependent calcineurin ⁄ JAK2 pathway [46].
CDC42, a member of the Rho GTPase family, is
reported to interact with ABCA1 and the interaction
enhances apoA-I-mediated cholesterol efflux [47]. Palm-
itoylation of ABCA1 is also involved in the trafficking
and function of ABCA1 [48].
We have reported [30] that the LXRb ⁄ RXR complex
binds to ABCA1 when the intracellular concentration
of oxysterols is low, and the ABCA1–LXRb ⁄ RXR
complex is distributed on the plasma membrane but is
inert in terms of cholesterol efflux (Fig. 2). When cho-
lesterol accumulates and the intracellular concentration

of oxysterols increases, oxysterols bind to LXRb and
the LXRb ⁄ RXR complex dissociates from ABCA1.
Once free from LXRb ⁄ RXR, ABCA1 is active in the
formation of HDL and decreases the local cholesterol
concentration immediately. Upon binding to oxysterols,
LXRb ⁄ RXR activates the transcription of ABCA1 and
other genes. Consequently, LXRb can cause a post-
translational response, as well as a transcriptional
response, to maintain cholesterol homeostasis. This
novel mechanism is an immediate early response to
cope with rapid increases in intracellular cholesterol,
such as when macrophages consume apoptotic cells.
Effects of Tangier mutations on ABCA1
From patients with Tangier disease and familial hypo-
alphalipiproteinaemia, more than 70 mutations have
been identified in the ABCA1 gene. Most mutations
reside in extracellular domains (ECDs) (putative apoA-
I binding site) and nucleotide binding domains (NBDs)
(driving-force-supplying site) of ABCA1 [3] (Fig. 1).
Mutations can be categorized into three groups
(Table 2). The first is the ‘maturation defect mutant’
and the majority of mutations belong to this group.
Wild-type ABCA1, modified with complex oligosaccha-
rides, is mainly localized to the plasma membrane and
sometimes in the intracellular compartments [11,31];
however, maturation defect mutants of ABCA1, modi-
fied with high mannose type oligosaccharides, have
impaired trafficking and are localized inappropriately
to the endoplasmic reticulum [10,11,49]. Because
apoA-I interacts with ABCA1 on the cell surface,

mutants which do not reach the plasma membrane are
unable to mediate apoA-I-dependent lipid efflux. The
second group is the ‘apoA-I binding defect mutant’.
This group is represented by the C1477R mutant of the
second ECD (Table 2). Although C1477R mutant is
expressed in the plasma membrane like wild-type
ABCA1, apoA-I binding is abolished [9,10,49,50]. The
third group is the ‘lipid translocation defect mutant’,
represented by the W590S mutation in the first ECD of
ABCA1 (Table 2). The subcellular distribution of
K. Nagao et al. Function and regulation of ABCA1
FEBS Journal 278 (2011) 3190–3203 ª 2011 The Authors Journal compilation ª 2011 FEBS 3193
W590S is indistinguishable from that of wild-type
ABCA1. Furthermore, W590S mutation does not affect
apoA-I binding [9,10,49,50]; however, W590S mutation
reduced phosphatidylserine (PS) flopping activity of
ABCA1, detected with the annexin V binding assay
[49], and impaired sodium taurocholate-dependent cho-
lesterol and phospholipid efflux by ABCA1 [51]. These
results suggest that W590S mutation affects the lipid
translocation activity of ABCA1 and that the two
activities of ABCA1 (apoA-I binding and lipid translo-
cation) can be separable (Fig. 3A). Additionally,
W590S mutation retarded the dissociation of apoA-I
from ABCA1 [51]. Lipid translocation by ABCA1 is
supposed to facilitate the dissociation of apoA-I from
ABCA1. ApoA-I is reported to undergo a conforma-
tional transition in response to lipid [52], and lipidated
apoA-I does not interact with ABCA1 [53,54]. The con-
formational transition of apoA-I caused by lipid load-

ing during binding to ABCA1 may facilitate the
dissociation of apoA-I from ABCA1 [55] (Fig. 3A).
Subcellular localization and function of
ABCA1
ABCA1 localizes mainly to the plasma membrane but
sometimes also localizes in the intracellular compart-
ments. Two distinct mechanisms have been proposed
for ABCA1-mediated HDL formation. One is that
ABCA1 mediates the complex formation of apoA-I
with phospholipids and cholesterol on the cell surface
(cell-surface model), and the other is that apoA-I binds
to ABCA1 on the cell surface and ABCA1 ⁄ apoA-I
complexes are subsequently internalized. ApoA-I⁄ lipid
complexes are formed (probably via ABCA1 activity)
in late endosomes and re-secreted by exocytosis (retro-
endocytosis model). Takahashi and Smith [56] first
showed that, following internalization, apoA-I is
recycled back to the cell surface to be re-secreted. The
internalized ABCA1 and apoA-I were reported to
co-localize within late endosomes, and ABCA1 rapidly
shuttled between intracellular compartments and the
plasma membrane [31,57]. Trapping ABCA1 on the
plasma membrane by cyclosporine A treatment reduces
apoA-I-mediated cholesterol efflux [58]. Deletion of the
PEST sequence blocks its endocytosis and decreases
apoA-I-mediated efflux of cholesterol after loading
the late endosome ⁄ lysosome pool of cholesterol by
Fig. 3. Membrane meso-domain organization and reorganization by
ABCA1. (A) Two proposed mechanisms for HDL formation. (i)
Membrane phospholipids and cholesterol are translocated by

ABCA1 and (ii) are loaded to apoA-I directly bound to the ECDs of
ABCA1 to generate nascent HDL particles [55]. (iii) Membrane
phospholipid translocation via ABCA1 induces bending of the mem-
brane bilayer to create high curvature sites, to which apoA-I binds
and solubilizes membrane phospholipid and cholesterol to create
nascent HDL particles [95]. (B) Regulation of cell signalling by
ABCA1. (i) ABCA1 translocates membrane phospholipids and cho-
lesterol and (iv) changes membrane meso-damain organization,
such as lipid rafts, that lead to suppressed receptor-mediated sig-
nalling events [99].
Table 2. Effects of mutations on the functions of ABCA1.
Class
Effects of mutations Position of mutations
PM localization ApoA-I binding HDL formation ECD1 TMD1 NBD1 ECD2 TMD2 NBD2
I Defect Defect Defect R587W
a,b
Q597R
a–c
DL693
b,c
N935S
b
A1046D
b
M1091T
b
S1506L
b
N1800H
b,d

R2081W
b
II Normal Defect Defect C1477R
b,c
III Normal Normal Defect A255T
b
W590S
a–e
T929I
b
C1660R
f
a
[11].
b
[10].
c
[49].
d
N1800 is predicted to reside in the loop between TM11 and TM12.
e
[9].
f
[146].
Function and regulation of ABCA1 K. Nagao et al.
3194 FEBS Journal 278 (2011) 3190–3203 ª 2011 The Authors Journal compilation ª 2011 FEBS
acetylated low-density lipoprotein treatment [59]. These
results together support the idea that nascent lipopro-
tein particles are formed in intracellular compartments
and subsequently secreted from the cell.

There are also many reports that ABCA1-mediated
cholesterol efflux to apoA-I mainly occurs on the
cell surface and that the retroendocytosis pathway
does not contribute significantly to HDL formation
[33,60,61]. The majority of internalized apoA-I is
directly transported to late endosomes and lysosomes
for degradation, and blocking endocytosis does not
decrease apoA-I-dependent cholesterol efflux.
Although apoA-I is specifically taken up by macro-
phages, only a small fraction of apoA-I is re-secreted
from these cells. Furthermore, the majority of
re-secreted apoA-I is degraded in the medium, suggest-
ing that the mass of retroendocytosed apoA-I is not
sufficient to account for HDL formation; however,
these studies were performed using macrophages and
other cells without cholesterol loading.
As pointed out by Oram [62], the mechanism of
HDL formation is probably different whether excess
cholesterol has accumulated within cells or not.
ABCA1 and apoA-I are endocytosed via a clathrin-
and Rab5-mediated pathway and recycled rapidly back
to the cell surface, at least in part via a Rab4-mediated
route; approximately 30% of the endocytosed ABCA1
is recycled back to the cell surface [32]. When clathrin-
mediated endocytosis is inhibited, the level of ABCA1
at the cell surface increases and apoA-I internalization
is blocked. Under these conditions, apoA-I-mediated
cholesterol efflux from cells that have accumulated
lipoprotein-derived cholesterol is decreased, whereas
efflux from cells without excess cholesterol is increased

[32]. These results suggest that the retroendocytosis
pathway of ABCA1 ⁄ apoA-I contributes to HDL for-
mation when excess lipoprotein-derived cholesterol has
accumulated in cells. This study is also in agreement
with previous studies that blocking retroendocytosis of
ABCA1 does not affect cholesterol efflux from cells in
the absence of excess cholesterol [33,60,61]. ABCA1
probably follows the same constitutive recycling path-
way as the LDL receptor [63].
Lipid acceptor for ABCA1
Because cholesterol and phospholipids are very hydro-
phobic, lipid acceptors which solubilize them in aque-
ous solutions are required for lipid secretion. Under
physiological conditions, apoA-I and apoE, containing
amphipathic helices, function as acceptors of lipids
secreted into serum by ABCA1. Although other
amphipathic-helical-peptide-containing proteins (e.g.
apoA-II, apoC-I, C-II, C-III, PLTP and serum amy-
loid A) also function as lipid acceptors for ABCA1-
dependent cholesterol efflux, their physiological contri-
butions remain to be clarified [64–67]. Synthetic
amphipathic helical peptide (37pA) also promotes cho-
lesterol and phospholipid efflux from ABCA1-express-
ing cells [68,69]. Furthermore, the 37pA peptide
synthesized with d amino acids is as effective as that
with l amino acids. From these results, the amphi-
pathic helix is considered to be a key structural motif
for peptide-mediated lipid efflux from ABCA1 [68].
We reported that sodium taurocholate can also serve
as an acceptor for cholesterol and phospholipids trans-

located by ABCA1 [51]; therefore, the detergent-like
property of the amphipathic helix might be important
as a lipid acceptor.
It is proposed that apoA-I directly binds to ABCA1
in the process of HDL formation, shown by several
groups via crosslinking experiments [54,65,70–72].
Because the 3-A
˚
crosslinker can crosslink apoA-I with
ABCA1, the pair of reactive amino acids of ABCA1
and apoA-I are within a distance of £ 3A
˚
[70]. Hozoji
et al. found that two intramolecular disulfide bonds
are formed between ECD1 and ECD2 of ABCA1, and
these two disulfide bonds are necessary for apoA-I
binding and HDL formation [73] (Fig. 1). It is
reported that apoA-I is not crosslinked with the
ATPase-deficient mutant form of ABCA1 [74]. Fur-
thermore, fluorescent-labelled apoA-I does not bind to
cells expressing ATPase-deficient mutant [51,75]. These
results suggest that the large ECD of ABCA1 is the
direct binding site for apoA-I and its ATP-dependent
conformational change is required for apoA-I binding.
But, it is also proposed that apoA-I interacts with a
special domain on the plasma membrane apart from
ABCA1 and solubilizes membrane lipids.
Transport substrates
Substrates transported directly by ABCA1 are still
controversial [55] (Table 1). Several models have been

proposed for the mechanism of ABCA1-mediated HDL
formation: (a) a two-step process model in which
ABCA1 first mediates PC efflux to apoA-I, and this
apoA-I–PC complex accepts cholesterol in an ABCA1-
independent manner; (b) a concurrent process model in
which PC and cholesterol efflux by ABCA1 to apoA-I
are coupled to each other; and (c) a third model in
which ABCA1 generates a specific apoA-I binding site
on the plasma membrane with subsequent translocation
of PC and cholesterol to apoA-I. When it was proposed
[74,76], the two-step model looked the most plausible,
because photoactive PC could be crosslinked with
K. Nagao et al. Function and regulation of ABCA1
FEBS Journal 278 (2011) 3190–3203 ª 2011 The Authors Journal compilation ª 2011 FEBS 3195
ABCA1 whereas direct binding of photoactive choles-
terol to ABCA1 could not be detected [74]; however,
analysis of the functions of ABCA7 raised questions
about this model. Human ABCA7, which has the high-
est homology (69.3%) to ABCA1, mediates the apoA-I-
dependent efflux of PC and cholesterol, similar to
ABCA1 [77]; however, human ABCA7 mediates choles-
terol release much less efficiently than ABCA1 [78], and
PC but not cholesterol are loaded onto apoA-I by
mouse ABCA7 [79]. These results cannot be explained
by the two-step process model. Instead, they suggest
that ABCA1 has higher affinity for cholesterol trans-
port than ABCA7, and are consistent with the concur-
rent process model [80]. The third model was originally
that ABCA1 mediates the translocation of PS to the
outer leaflet to form a special membrane domain, where

apoA-I binds and solubilizes membrane lipids [81].
Membrane meso-domain organization
and reorganization
On the plasma membrane, various meso-scale (10–
100 nm) domains, such as lipid rafts [82], are supposed
to be dynamically organized and reorganized and
involved in various cellular functions, such as signal
transduction. ABCA1 is involved in membrane meso-
domain reorganization, as ABCA1 expression results
in a significant redistribution of cholesterol and sphin-
gomyelin (SM) from Triton X-100-resistant membrane
domains [83]. Caveolin also redistributes from punctu-
ate caveolae-like structures to the general area of the
plasma membrane [83]. Macrophages from ABCA1-
deficient mice exhibited increased lipid rafts on the cell
surface [84] and ABCA1 made cells more sensitive to
methyl-b-cyclodextrin (MbCD) treatment [80] and
generated cholesterol-oxidase-accessible membrane
domains [85].
Importantly, membrane meso-domain reorganization
by ABCA1 is independent of apoA-I [83,85]. It is possi-
ble that active lipid translocation via ABCA1, the flop
of phospholipids and cholesterol from the inner to
outer leaflet, leads to membrane destabilization. Since
ABCA1 is not associated with Triton X-100-resistant
membrane domains [86,87], the domains are destabi-
lized via lipid translocation by ABCA1 located outside
the domains. It is reported that ABCA1 is associated
with Luburol WX-resistant membranes in cholesterol-
loaded monocyte-derived macrophages [87], that

ABCA1 and flotillin-1 are co-localized in these deter-
gent-resistant membranes and can be co-precipitated
[88], and that expression of caveolin-1 enhances apoA-
I-dependent cholesterol efflux in hepatic cells [89].
ABCA1 may localize just outside Triton X-100-resistant
membrane domains. But ABCA1 is not recovered from
either Triton X-100- or Luburol WX-resistant mem-
branes in fibroblasts [87]. It is not clear how lipid trans-
location by ABCA1 in non-raft regions reorganizes
other domains of the plasma membrane.
We analysed the effects of the cellular SM level on
the function of ABCA1 using a CHO-K1 mutant cell
line, LY-A [90], which has a missense mutation in the
ceramide transfer protein CERT, and reported that the
decrease in SM content in the plasma membrane stim-
ulates apoA-I-dependent cholesterol efflux by ABCA1
[91]. The amount of cholesterol available to cold
MbCD extraction is increased when SM content is
reduced. However, apoA-I-dependent cholesterol efflux
is not observed without ABCA1 expression even when
SM content is reduced, suggesting that the cholesterol
available to cold MbCD cannot be loaded onto apoA-
I spontaneously. When ABCA1 is expressed, the cho-
lesterol available to cold MbCD is increased by 40%.
This effect of ABCA1 is independent of apoA-I. These
results suggest that ABCA1 translocates membrane
lipids in detergent-soluble domains and makes (acti-
vates [92] or projects [55]) cholesterol available to cold
MbCD. The effect of the reduction of plasma mem-
brane SM content on the function of ABCA1 is quite

different from that on the function of ABCG1, since
ABCG1-mediated cholesterol efflux decreases when
cellular SM content is reduced [93]. This is consistent
with previous reports that lipids loaded onto lipid-poor
apoA-I by ABCA1 are provided by Triton X-100-sen-
sitive membrane domains, whereas lipids loaded onto
HDL, the lipid acceptor for ABCG1, are derived from
Triton X-100-resistant membrane domains [86,87], and
that treating rat fibroblasts with SMase increased
apoA-I- or apo-E-dependent cholesterol efflux [94].
Several studies suggest that ABCA1 generates spe-
cial membrane meso-domains. Membrane phospho-
lipid translocation via ABCA1 induces bending of the
membrane bilayer to create high curvature sites, such
as is created in 20-nm-diameter small unilamellar vesi-
cles, to which apoA-I binds and solubilizes membrane
phospholipid and cholesterol to create nascent HDL
particles [95] (Fig. 3A). In agreement with this concept,
plasma membrane protrusions [54] and apoA-I binding
to protruding plasma membrane domains [96] have
been observed in cells expressing ABCA1. Such apoA-I
binding could be enhanced by PS molecules translocated
to the exofacial leaflet by ABCA1 [81], because the
presence of PS enhances vesicle curvature [97]. How-
ever, there are still clear differences in the dependence
on apoA-I concentration between ABCA1-mediated
lipid efflux and in vitro solubilization of phospholipid
vesicles [95]. It has been calculated that only a portion
Function and regulation of ABCA1 K. Nagao et al.
3196 FEBS Journal 278 (2011) 3190–3203 ª 2011 The Authors Journal compilation ª 2011 FEBS

of the apoA-I associated with the cell surface binds
directly to ABCA1 [72,98]. One plausible explanation
may be that direct interaction with ABCA1 causes
conformational changes in apoA-I to make it open
and receptive to lipids. Activated apoA-I may interact
with the special meso-domains created by ABCA1.
Recently it was reported that LXR signalling at a
metabolic checkpoint modulates T cell proliferation
and immunity [99]. T cell activation is accompanied by
the downregulation of LXR target genes, ABCA1 and
ABCG1 [100]. Loss of LXR expression confers a pro-
liferative advantage to lymphocytes, resulting in
enhanced homeostatic and antigen-driven responses.
Conversely, ligand activation of LXR inhibits mitogen-
driven T cell expansion. Cellular cholesterol is required
for increased membrane synthesis during cell prolifera-
tion. Additional mechanisms may also be involved,
such as changes in membrane meso-domain organiza-
tion, e.g. lipid rafts, that lead to enhanced receptor-
mediated signalling events. Functional deficiencies of
ABCA1 and ABCG1 cause enhanced inflammatory
responses of macrophages, especially after treatment
with lipopolysaccharide or other toll-like receptor
ligands [84,101–103]. ABCA1 and ABCG1 may pro-
mote membrane lipid redistribution [83,104], which
modulates raft-dependent cell signalling (Fig. 3B).
Acknowledgements
This study was supported by a Grant-in-aid for Scien-
tific Research (S) from the Ministry of Education, Cul-
ture, Sports, Science and Technology of Japan, the

Bio-oriented Technology Research Advancement Insti-
tution, and the World Premier International Research
Center Initiative, MEXT, Japan.
References
1 van Dam MJ, de Groot E, Clee SM, Hovingh GK,
Roelants R, Brooks-Wilson A, Zwinderman AH, Smit
AJ, Smelt AH, Groen AK et al. (2002) Association
between increased arterial-wall thickness and impair-
ment in ABCA1-driven cholesterol efflux: an observa-
tional study. Lancet 359, 37–42.
2 Frikke-Schmidt R (2010) Genetic variation in the
ABCA1 gene, HDL cholesterol, and risk of ischemic
heart disease in the general population. Atherosclerosis
208, 305–316.
3 Singaraja RR, Brunham LR, Visscher H, Kastelein JJ
& Hayden MR (2003) Efflux and atherosclerosis: the
clinical and biochemical impact of variations in the
ABCA1 gene. Arterioscler Thromb Vasc Biol 23,
1322–1332.
4 Oram JF & Vaughan AM (2006) ATP-binding cassette
cholesterol transporters and cardiovascular disease.
Circ Res 99, 1031–1043.
5 Bodzioch M, Orso E, Klucken J, Langmann T, Bott-
cher A, Diederich W, Drobnik W, Barlage S, Buchler
C, Porsch-Ozcurumez M et al. (1999) The gene encod-
ing ATP-binding cassette transporter 1 is mutated in
Tangier disease. Nat Genet 22, 347–351.
6 Brooks-Wilson A, Marcil M, Clee S, Zhang L, Roomp
K, van Dam M, Yu L, Brewer C, Collins J, Molhuizen
H et al. (1999) Mutations in ABC1 in Tangier disease

and familial high-density lipoprotein deficiency. Nat
Genet 22, 336–345.
7 Rust S, Rosier M, Funke H, Real J, Amoura Z, Piette
J, Deleuze J, Brewer H, Duverger N, Denefle P et al.
(1999) Tangier disease is caused by mutations in the
gene encoding ATP-binding cassette transporter 1.
Nat Genet 22, 352–355.
8 Brunham LR, Singaraja RR & Hayden MR (2006)
Variations on a gene: rare and common variants in
ABCA1 and their impact on HDL cholesterol levels
and atherosclerosis. Annu Rev Nutr 26, 105–129.
9 Fitzgerald ML, Morris AL, Rhee JS, Andersson LP,
Mendez AJ & Freeman MW (2002) Naturally occur-
ring mutations in ABCA1’s largest extracellular loops
can disrupt its direct interaction with apolipoprotein
A-I. J Biol Chem 277, 33178–33187.
10 Singaraja RR, Visscher H, James ER, Chroni A,
Coutinho JM, Brunham LR, Kang MH, Zannis VI,
Chimini G & Hayden MR (2006) Specific mutations
in ABCA1 have discrete effects on ABCA1 function
and lipid phenotypes both in vivo and in vitro. Circ
Res 99, 389–397.
11 Tanaka AR, Abe-Dohmae S, Ohnishi T, Aoki R,
Morinaga G, Okuhira KI, Ikeda Y, Kano F, Matsuo
M, Kioka N et al. (2003) Effects of mutations of
ABCA1 in the first extracellular domain on subcellular
trafficking and ATP binding ⁄ hydrolysis. J Biol Chem
278, 8815–8819.
12 Yokoyama S (2000) Release of cellular cholesterol:
molecular mechanism for cholesterol homeostasis in

cells and in the body. Biochim Biophys Acta 1529,
231–244.
13 Lawn RM, Wade DP, Garvin MR, Wang X, Schwartz
K, Porter JG, Seilhamer JJ, Vaughan AM & Oram JF
(1999) The Tangier disease gene product ABC1 con-
trols the cellular apolipoprotein-mediated lipid removal
pathway. J Clin Invest 104, R25–31.
14 Janowski BA, Willy PJ, Devi TR, Falck JR &
Mangelsdorf DJ (1996) An oxysterol signalling
pathway mediated by the nuclear receptor LXR alpha.
Nature 383, 728–731.
15 Lehmann JM, Kliewer SA, Moore LB, Smith-Oliver
TA, Oliver BB, Su JL, Sundseth SS, Winegar DA, Blan-
chard DE, Spencer TA et al. (1997) Activation of the
K. Nagao et al. Function and regulation of ABCA1
FEBS Journal 278 (2011) 3190–3203 ª 2011 The Authors Journal compilation ª 2011 FEBS 3197
nuclear receptor LXR by oxysterols defines a new hor-
mone response pathway. J Biol Chem 272, 3137–3140.
16 Bischoff ED, Daige CL, Petrowski M, Dedman H,
Pattison J, Juliano J, Li AC & Schulman IG (2010)
Non-redundant roles for LXRalpha and LXRbeta in
atherosclerosis susceptibility in low density lipoprotein
receptor knockout mice. J Lipid Res 51, 900–906.
17 Venkateswaran A, Laffitte BA, Joseph SB, Mak PA,
Wilpitz DC, Edwards PA & Tontonoz P (2000) Con-
trol of cellular cholesterol efflux by the nuclear oxys-
terol receptor LXR alpha. Proc Natl Acad Sci USA
97, 12097–12102.
18 Repa JJ, Turley SD, Lobaccaro J-MA, Medina J,
Li L, Lustig K, Shan B, Heyman RA, Dietschy JM &

Mangelsdorf DJ (2000) Regulation of absorption
and ABC1-mediated efflux of cholesterol by RXR
heterodimers. Science 289, 1524–1529.
19 Costet P, Luo Y, Wang N & Tall AR (2000) Sterol-
dependent transactivation of the ABC1 promoter by
the liver X receptor ⁄ retinoid X receptor. J Biol Chem
275, 28240–28245.
20 Engelking LJ, Kuriyama H, Hammer RE, Horton JD,
Brown MS, Goldstein JL & Liang G (2004) Overex-
pression of Insig-1 in the livers of transgenic mice
inhibits SREBP processing and reduces insulin-stimu-
lated lipogenesis. J Clin Invest 113, 1168–1175.
21 Tamehiro N, Shigemoto-Mogami Y, Kakeya T, Okuh-
ira K, Suzuki K, Sato R, Nagao T & Nishimaki-Mo-
gami T (2007) Sterol regulatory element-binding
protein-2- and liver X receptor-driven dual promoter
regulation of hepatic ABC transporter A1 gene expres-
sion: mechanism underlying the unique response to cel-
lular cholesterol status. J Biol Chem 282, 21090–21099.
22 Najafi-Shoushtari SH, Kristo F, Li Y, Shioda T, Co-
hen DE, Gerszten RE & Naar AM (2010) MicroRNA-
33 and the SREBP host genes cooperate to control
cholesterol homeostasis. Science 328, 1566–1569.
23 Rayner KJ, Suarez Y, Davalos A, Parathath S,
Fitzgerald ML, Tamehiro N, Fisher EA, Moore KJ &
Fernandez-Hernando C (2010) MiR-33 contributes to
the regulation of cholesterol homeostasis. Science 328,
1570–1573.
24 Wang Y & Oram JF (2002) Unsaturated fatty acids
inhibit cholesterol efflux from macrophages by increas-

ing degradation of ATP-binding cassette transporter
A1. J Biol Chem 277, 5692–5697.
25 Arakawa R & Yokoyama S (2002) Helical apolipopro-
teins stabilize ATP-binding cassette transporter A1 by
protecting it from thiol protease-mediated degradation.
J Biol Chem 277, 22426–22429.
26 Wang N, Chen W, Linsel-Nitschke P, Martinez LO,
Agerholm-Larsen B, Silver DL & Tall AR (2003) A
PEST sequence in ABCA1 regulates degradation by
calpain protease and stabilization of ABCA1 by
apoA-I. J Clin Invest 111, 99–107.
27 Munehira Y, Ohnishi T, Kawamoto S, Furuya A,
Shitara K, Imamura M, Yokota T, Takeda S, Amachi
T, Matsuo M et al. (2004) Alpha1-syntrophin
modulates turnover of ABCA1. J Biol Chem 279,
15091–15095.
28 Okuhira K, Fitzgerald ML, Sarracino DA, Manning
JJ, Bell SA, Goss JL & Freeman MW (2005) Purifica-
tion of ATP-binding cassette transporter A1 and asso-
ciated binding proteins reveals the importance of
beta1-syntrophin in cholesterol efflux. J Biol Chem
280, 39653–39664.
29 Iwamoto N, Lu R, Tanaka N, Abe-Dohmae S &
Yokoyama S (2010) Calmodulin interacts with ATP
binding cassette transporter A1 to protect from
calpain-mediated degradation and upregulates high-
density lipoprotein generation. Arterioscler Thromb
Vasc Biol 30, 1446–1452.
30 Hozoji M, Munehira Y, Ikeda Y, Makishima M,
Matsuo M, Kioka N & Ueda K (2008) Direct inter-

action of nuclear liver X receptor-beta with ABCA1
modulates cholesterol efflux. J Biol Chem 283, 30057–
30063.
31 Neufeld EB, Remaley AT, Demosky SJ, Stonik JA,
Cooney AM, Comly M, Dwyer NK, Zhang M,
Blanchette-Mackie J, Santamarina-Fojo S et al. (2001)
Cellular localization and trafficking of the human
ABCA1 transporter. J Biol Chem
276, 27584–27590.
32 Azuma Y, Takada M, Shin H-W, Kioka N, Nakay-
ama K & Ueda K (2009) Retroendocytosis pathway of
ABCA1 ⁄ apoA-I contributes to HDL formation. Genes
Cells 14, 191–204.
33 Lu R, Arakawa R, Ito-Osumi C, Iwamoto N &
Yokoyama S (2008) ApoA-I facilitates ABCA1
recycle ⁄ accumulation to cell surface by inhibiting its
intracellular degradation and increases HDL genera-
tion. Arterioscler Thromb Vasc Biol 28, 1820–1824.
34 Feng B & Tabas I (2002) ABCA1-mediated cholesterol
efflux is defective in free cholesterol-loaded macro-
phages. Mechanism involves enhanced ABCA1 degra-
dation in a process requiring full NPC1 activity. J Biol
Chem 277, 43271–43280.
35 Tanaka AR, Kano F, Yamamoto A, Ueda K & Murata
M (2008) Formation of cholesterol-enriched structures
by aberrant intracellular accumulation of ATP-binding
cassette transporter A1. Genes Cells 13, 889–904.
36 Azuma Y, Takada M, Maeda M, Kioka N & Ueda K
(2009) The COP9 signalosome controls ubiquitinyla-
tion of ABCA1. Biochem Biophys Res Commun 382,

145–148.
37 Haidar B, Denis M, Marcil M, Krimbou L & Genest J
Jr (2004) Apolipoprotein A-I activates cellular cAMP
signaling through the ABCA1 transporter. J Biol Chem
279, 9963–9969.
38 See RH, Caday-Malcolm RA, Singaraja RR, Zhou S,
Silverston A, Huber MT, Moran J, James ER, Janoo
Function and regulation of ABCA1 K. Nagao et al.
3198 FEBS Journal 278 (2011) 3190–3203 ª 2011 The Authors Journal compilation ª 2011 FEBS
R, Savill JM et al. (2002) Protein kinase A site-specific
phosphorylation regulates ATP-binding cassette A1
(ABCA1)-mediated phospholipid efflux. J Biol Chem
277, 41835–41842.
39 Yamauchi Y, Hayashi M, Abe-Dohmae S & Yokoy-
ama S (2003) Apolipoprotein A-I activates protein
kinase C alpha signaling to phosphorylate and stabilize
ATP binding cassette transporter A1 for the high den-
sity lipoprotein assembly. J Biol Chem 278, 47890–
47897.
40 Yamauchi Y, Chang CC, Hayashi M, Abe-Dohmae S,
Reid PC, Chang TY & Yokoyama S (2004) Intracellu-
lar cholesterol mobilization involved in the
ABCA1 ⁄ apolipoprotein-mediated assembly of high
density lipoprotein in fibroblasts. J Lipid Res 45,
1943–1951.
41 Martinez LO, Agerholm-Larsen B, Wang N, Chen W
& Tall AR (2003) Phosphorylation of a PEST
sequence in ABCA1 promotes calpain degradation and
is reversed by ApoA-I. J Biol Chem 278, 37368–37374.
42 Wang Y & Oram JF (2007) Unsaturated fatty acids

phosphorylate and destabilize ABCA1 through a pro-
tein kinase C delta pathway. J Lipid Res 48, 1062–
1068.
43 Roosbeek S, Peelman F, Verhee A, Labeur C, Caster
H, Lensink MF, Cirulli C, Grooten J, Cochet C, Van-
dekerckhove J et al. (2004) Phosphorylation by protein
kinase CK2 modulates the activity of the ATP binding
cassette A1 transporter. J Biol Chem 279, 37779–
37788.
44 Tang C, Vaughan AM & Oram JF (2004) Janus kinase
2 modulates the apolipoprotein interactions with
ABCA1 required for removing cellular cholesterol.
J Biol Chem 279, 7622–7628.
45 Tang C, Liu Y, Kessler PS, Vaughan AM & Oram JF
(2009) The macrophage cholesterol exporter ABCA1
functions as an anti-inflammatory receptor. J Biol
Chem 284, 32336–32343.
46 Karwatsky J, Ma L, Dong F & Zha X (2010) Choles-
terol efflux to apoA-I in ABCA1-expressing cells is
regulated by Ca
2+
-dependent calcineurin signaling.
J Lipid Res 51, 1144–1156.
47 Nofer JR, Remaley AT, Feuerborn R, Wolinnska I,
Engel T, von Eckardstein A & Assmann G (2006)
Apolipoprotein A-I activates Cdc42 signaling through
the ABCA1 transporter. J Lipid Res 47, 794–803.
48 Singaraja RR, Kang MH, Vaid K, Sanders SS, Vilas
GL, Arstikaitis P, Coutinho J, Drisdel RC, El-Husse-
ini Ael D, Green WN et al. (2009) Palmitoylation of

ATP-binding cassette transporter A1 is essential for its
trafficking and function. Circ Res 105, 138–147.
49 Rigot V, Hamon Y, Chambenoit O, Alibert M, Duver-
ger N & Chimini G (2002) Distinct sites on ABCA1
control distinct steps required for cellular release of
phospholipids. J Lipid Res 43, 2077–2086.
50 Vaughan AM, Tang C & Oram JF (2009) ABCA1
mutants reveal an interdependency between lipid
export function, apoA-I binding activity, and Janus
kinase 2 activation. J Lipid Res 50, 285–292.
51 Nagao K, Zhao Y, Takahashi K, Kimura Y & Ueda
K (2009) Sodium taurocholate-dependent lipid efflux
by ABCA1: effects of W590S mutation on lipid trans-
location and apolipoprotein A-I dissociation. J Lipid
Res 50, 1165–1172.
52 Davidson WS & Thompson TB (2007) The structure
of apolipoprotein A-I in high density lipoproteins.
J Biol Chem 282, 22249–22253.
53 Mulya A, Lee JY, Gebre AK, Thomas MJ, Colvin PL
& Parks JS (2007) Minimal lipidation of pre-beta
HDL by ABCA1 results in reduced ability to interact
with ABCA1. Arterioscler Thromb Vasc Biol 27,
1828–1836.
54 Wang N, Silver D, Costet P & Tall A (2000) Specific
binding of ApoA-I, enhanced cholesterol efflux, and
altered plasma membrane morphology in cells express-
ing ABC1. J Biol Chem 275 , 33053–33058.
55 Nagao K, Kimura Y, Mastuo M & Ueda K (2010)
Lipid outward translocation by ABC proteins. FEBS
Lett 584, 2717–2723.

56 Takahashi Y & Smith JD (1999) Cholesterol efflux to
apolipoprotein AI involves endocytosis and resecretion
in a calcium-dependent pathway. Proc Natl Acad Sci
USA 96, 11358–11363.
57 Neufeld EB, Stonik JA, Demosky SJ Jr, Knapper CL,
Combs CA, Cooney A, Comly M, Dwyer N,
Blanchette-Mackie J, Remaley AT et al. (2004) The
ABCA1 transporter modulates late endocytic
trafficking: insights from the correction of the genetic
defect in Tangier disease. J Biol Chem 279, 15571–
15578.
58 Le Goff W, Peng DQ, Settle M, Brubaker G, Morton
RE & Smith JD (2004) Cyclosporin A traps ABCA1
at the plasma membrane and inhibits ABCA1-medi-
ated lipid efflux to apolipoprotein A-I. Arterioscler
Thromb Vasc Biol 24, 2155–2161.
59 Chen W, Wang N & Tall AR (2005) A PEST deletion
mutant of ABCA1 shows impaired internalization and
defective cholesterol efflux from late endosomes. J Biol
Chem 280, 29277–29281.
60 Denis M, Landry YD & Zha X (2008) ATP-binding
cassette A1-mediated lipidation of apolipoprotein A-I
occurs at the plasma membrane and not in the endocy-
tic compartments. J Biol Chem 283, 16178–16186.
61 Faulkner LE, Panagotopulos SE, Johnson JD,
Woollett LA, Hui DY, Witting SR, Maiorano JN &
Davidson WS (2008) An analysis of the role of a retro-
endocytosis pathway in ABCA1-mediated cholesterol
efflux from macrophages. J Lipid Res 49, 1322–1332.
62 Oram JF (2008) The ins and outs of ABCA. J Lipid

Res 49, 1150–1151.
K. Nagao et al. Function and regulation of ABCA1
FEBS Journal 278 (2011) 3190–3203 ª 2011 The Authors Journal compilation ª 2011 FEBS 3199
63 Heuser JE & Anderson RG (1989) Hypertonic media
inhibit receptor-mediated endocytosis by blocking
clathrin-coated pit formation. J Cell Biol 108, 389–400.
64 Remaley AT, Stonik JA, Demosky SJ, Neufeld EB,
Bocharov AV, Vishnyakova TG, Eggerman TL,
Patterson AP, Duverger NJ, Santamarina-Fojo S et al.
(2001) Apolipoprotein specificity for lipid efflux by the
human ABCAI transporter. Biochem Biophys Res
Commun 280, 818–823.
65 Fitzgerald ML, Morris AL, Chroni A, Mendez AJ,
Zannis VI & Freeman MW (2004) ABCA1 and amphi-
pathic apolipoproteins form high-affinity molecular
complexes required for cholesterol efflux. J Lipid Res
45, 287–294.
66 Abe-Dohmae S, Kato KH, Kumon Y, Hu W, Ishigami
H, Iwamoto N, Okazaki M, Wu CA, Tsujita M, Ueda
K et al. (2006) Serum amyloid A generates high density
lipoprotein with cellular lipid in an ABCA1- or
ABCA7-dependent manner. J Lipid Res 47, 1542–1550.
67 Oram JF, Wolfbauer G, Vaughan AM, Tang C &
Albers JJ (2003) Phospholipid transfer protein inter-
acts with and stabilizes ATP-binding cassette trans-
porter A1 and enhances cholesterol efflux from cells.
J Biol Chem 278, 52379–52385.
68 Remaley AT, Thomas F, Stonik JA, Demosky SJ,
Bark SE, Neufeld EB, Bocharov AV, Vishnyakova
TG, Patterson AP, Eggerman TL et al. (2003)

Synthetic amphipathic helical peptides promote lipid
efflux from cells by an ABCA1-dependent and an
ABCA1-independent pathway. J Lipid Res 44,
828–836.
69 Arakawa R, Hayashi M, Remaley AT, Brewer BH,
Yamauchi Y & Yokoyama S (2004) Phosphorylation
and stabilization of ATP binding cassette transporter
A1 by synthetic amphiphilic helical peptides. J Biol
Chem 279, 6217–6220.
70 Chroni A, Liu T, Fitzgerald ML, Freeman MW &
Zannis VI (2004) Cross-linking and lipid efflux proper-
ties of apoA-I mutants suggest direct association
between apoA-I helices and ABCA1. Biochemistry 43,
2126–2139.
71 Oram J, Lawn R, Garvin M & Wade D (2000)
ABCA1 is the cAMP-inducible apolipoprotein receptor
that mediates cholesterol secretion from macrophages.
J Biol Chem 275, 34508–34511.
72 Vedhachalam C, Ghering AB, Davidson WS, Lund-
Katz S, Rothblat GH & Phillips MC (2007) ABCA1-
induced cell surface binding sites for ApoA-I.
Arterioscler Thromb Vasc Biol 27, 1603–1609.
73 Hozoji M, Kimura Y, Kioka N & Ueda K (2009)
Formation of two intramolecular disulfide bonds is
necessary for ApoA-I-dependent cholesterol efflux
mediated by ABCA1. J Biol Chem 284, 11293–11300.
74 Wang N, Silver DL, Thiele C & Tall AR (2001) ATP-
binding cassette transporter A1 (ABCA1) functions as
a cholesterol efflux regulatory protein. J Biol Chem
276, 23742–23747.

75 Chambenoit O, Hamon Y, Marguet D, Rigneault H,
Rosseneu M & Chimini G (2001) Specific docking of
apolipoprotein A-I at the cell surface requires a func-
tional ABCA1 transporter. J Biol Chem 276, 9955–9960.
76 Fielding PE, Nagao K, Hakamata H, Chimini G &
Fielding CJ (2000) A two-step mechanism for free cho-
lesterol and phospholipid efflux from human vascular
cells to apolipoprotein A-1. Biochemistry 39, 14113–
14120.
77 Abe-Dohmae S, Ikeda Y, Matsuo M, Hayashi M, Ok-
uhira K, Ueda K & Yokoyama S (2004) Human
ABCA7 supports apolipoprotein-mediated release of
cellular cholesterol and phospholipid to generate high
density lipoprotein. J Biol Chem 279, 604–611.
78 Hayashi M, Abe-Dohmae S, Okazaki M, Ueda K &
Yokoyama S (2005) Heterogeneity of high density
lipoprotein generated by ABCA1 and ABCA7. J Lipid
Res 46, 1703–1711.
79 Wang N, Lan D, Gerbod-Giannone M, Linsel-Nit-
schke P, Jehle AW, Chen W, Martinez LO & Tall AR
(2003) ATP-binding cassette transporter A7 (ABCA7)
binds apolipoprotein A-I and mediates cellular phos-
pholipid but not cholesterol efflux. J Biol Chem 278,
42906–42912.
80 Smith JD, Le Goff W, Settle M, Brubaker G, Waelde
C, Horwitz A & Oda MN (2004) ABCA1 mediates
concurrent cholesterol and phospholipid efflux to
apolipoprotein A-I. J Lipid Res 45, 635–644.
81 Hamon Y, Broccardo C, Chambenoit O, Luciani M,
Toti F, Chaslin S, Freyssinet J, Devaux P, McNeish J,

Marguet D et al. (2000) ABC1 promotes engulfment
of apoptotic cells and transbilayer redistribution of
phosphatidylserine. Nat Cell Biol 2, 399–406.
82 Simons K & Toomre D (2000) Lipid rafts and signal
transduction. Nat Rev Mol Cell Biol 1, 31–39.
83 Landry YD, Denis M, Nandi S, Bell S, Vaughan AM
& Zha X (2006) ABCA1 expression disrupts raft
membrane microdomains through its ATPase-related
functions. J Biol Chem 281, 36091–36101.
84 Koseki M, Hirano KI, Masuda D, Ikegami C, Tanaka
M, Ota A, Sandoval JC, Nakagawa-Toyama Y, Sato
SB, Kobayashi T et al. (2007) Increased lipid rafts and
accelerated lipopolysaccharide-induced tumor necrosis
factor alpha secretion in ABCa1-deficient macrophag-
es. J Lipid Res 48, 299–306.
85 Vaughan AM & Oram JF (2003) ABCA1 redistributes
membrane cholesterol independent of apolipoprotein
interactions. J Lipid Res 44, 1373–1380.
86 Mendez AJ, Lin G, Wade DP, Lawn RM & Oram JF
(2001) Membrane lipid domains distinct from choles-
terol ⁄ sphingomyelin-rich rafts are involved in the
ABCA1-mediated lipid secretory pathway. J Biol Chem
276, 3158–3166.
Function and regulation of ABCA1 K. Nagao et al.
3200 FEBS Journal 278 (2011) 3190–3203 ª 2011 The Authors Journal compilation ª 2011 FEBS
87 Drobnik W, Borsukova H, Bottcher A, Pfeiffer A, Lie-
bisch G, Schutz GJ, Schindler H & Schmitz G (2002)
Apo AI ⁄ ABCA1-dependent and HDL3-mediated lipid
efflux from compositionally distinct cholesterol-based
microdomains. Traffic 3, 268–278.

88 Bared SM, Buechler C, Boettcher A, Dayoub R, Sigru-
ener A, Grandl M, Rudolph C, Dada A & Schmitz G
(2004) Association of ABCA1 with syntaxin 13 and
flotillin-1 and enhanced phagocytosis in tangier cells.
Mol Biol Cell 15, 5399–5407.
89 Fu Y, Hoang A, Escher G, Parton RG, Krozowski Z
& Sviridov D (2004) Expression of caveolin-1 enhances
cholesterol efflux in hepatic cells. J Biol Chem 279,
14140–14146.
90 Hanada K, Kumagai K, Yasuda S, Miura Y, Kawano
M, Fukasawa M & Nishijima M (2003) Molecular
machinery for non-vesicular trafficking of ceramide.
Nature 426, 803–809.
91 Nagao K, Takahashi K, Hanada K, Kioka N, Matsuo
M & Ueda K (2007) Enhanced apoA-I-dependent cho-
lesterol efflux by ABCA1 from sphingomyelin-deficient
Chinese hamster ovary cells. J Biol Chem 282, 14868–
14874.
92 Small DM (2003) Role of ABC transporters in secre-
tion of cholesterol from liver into bile. Proc Natl Acad
Sci USA 100, 4–6.
93 Sano O, Kobayashi A, Nagao K, Kumagai K, Kioka
N, Hanada K, Ueda K & Matsuo M (2007) Sphingo-
myelin-dependence of cholesterol efflux mediated by
ABCG1. J Lipid Res 48, 2377–2384.
94 Ito J, Nagayasu Y & Yokoyama S (2000) Cholesterol-
sphingomyelin interaction in membrane and apolipo-
protein-mediated cellular cholesterol efflux. J Lipid Res
41, 894–904.
95 Vedhachalam C, Duong PT, Nickel M, Nguyen D,

Dhanasekaran P, Saito H, Rothblat GH, Lund-Katz S
& Phillips MC (2007) Mechanism of ATP-binding cas-
sette transporter A1-mediated cellular lipid efflux to
apolipoprotein A-I and formation of high density lipo-
protein particles. J Biol Chem 282, 25123–25130.
96 Lin G & Oram JF (2000) Apolipoprotein binding to
protruding membrane domains during removal of
excess cellular cholesterol. Atherosclerosis 149, 359–
370.
97 Hauser H & Phillips MC (1973) Structures of aqueous
dispersions of phosphatidylserine. J Biol Chem 248,
8585–8591.
98 Hassan HH, Denis M, Lee DY, Iatan I, Nyholt D,
Ruel I, Krimbou L & Genest J (2007) Identification of
an ABCA1-dependent phospholipid-rich plasma mem-
brane apolipoprotein A-I binding site for nascent
HDL formation: implications for current models of
HDL biogenesis. J Lipid Res 48, 2428–2442.
99 Yvan-Charvet L, Wang N & Tall AR (2010) Role of
HDL, ABCA1, and ABCG1 transporters in cholesterol
efflux and immune responses. Arterioscler Thromb
Vasc Biol 30, 139–143.
100 Bensinger SJ, Bradley MN, Joseph SB, Zelcer N, Jans-
sen EM, Hausner MA, Shih R, Parks JS, Edwards
PA, Jamieson BD et al. (2008) LXR signaling couples
sterol metabolism to proliferation in the acquired
immune response. Cell 134, 97–111.
101 Francone OL, Royer L, Boucher G, Haghpassand M,
Freeman A, Brees D & Aiello RJ (2005) Increased
cholesterol deposition, expression of scavenger recep-

tors, and response to chemotactic factors in ABCA1-
deficient macrophages. Arterioscler Thromb Vasc Biol
25, 1198–1205.
102 Zhu X, Lee JY, Timmins JM, Brown JM, Boudyguina
E, Mulya A, Gebre AK, Willingham MC, Hiltbold
EM, Mishra N et al. (2008) Increased cellular free cho-
lesterol in macrophage-specific ABCA1 knock-out mice
enhances pro-inflammatory response of macrophages.
J Biol Chem 283
, 22930–22941.
103 Yvan-Charvet L, Welch C, Pagler TA, Ranalletta M,
Lamkanfi M, Han S, Ishibashi M, Li R, Wang N &
Tall AR (2008) Increased inflammatory gene
expression in ABC transporter-deficient macrophages:
free cholesterol accumulation, increased signaling via
toll-like receptors, and neutrophil infiltration of
atherosclerotic lesions. Circulation 118, 1837–1847.
104 Vaughan AM & Oram JF (2005) ABCG1 redistributes
cell cholesterol to domains removable by high density
lipoprotein but not by lipid-depleted apolipoproteins.
J Biol Chem 280, 30150–30157.
105 Tam SP, Mok L, Chimini G, Vasa M & Deeley
RG(2006) ABCA1 mediates high-affinity uptake of
25-hydroxycholesterol by membrane vesicles and rapid
efflux of the oxysterol by intact cells. Am J Physiol
Cell Physiol 291, C490–C502.
106 Schmitz G, Kaminski WE, Porsch-Ozcurumez M,
Klucken J, Orso E, Bodzioch M, Buchler C &
Drobnik W (1999) ATP-binding cassette transporter
A1 (ABCA1) in macrophages: a dual function in

inflammation and lipid metabolism? Pathobiology 67,
236–240.
107 Brunham LR, Singaraja RR, Duong M, Timmins JM,
Fievet C, Bissada N, Kang MH, Samra A, Fruchart
JC, McManus B et al. (2009) Tissue-specific roles of
ABCA1 influence susceptibility to atherosclerosis.
Arterioscler Thromb Vasc Biol 29, 548–554.
108 Karasinska JM, Rinninger F, Lutjohann D, Ruddle P,
Franciosi S, Kruit JK, Singaraja RR, Hirsch-Reinsha-
gen V, Fan J, Brunham LR et al. (2009) Specific loss
of brain ABCA1 increases brain cholesterol uptake
and influences neuronal structure and function. J Neu-
rosci 29, 3579–3589.
109 Sakai H, Tanaka Y, Tanaka M, Ban N, Yamada K,
Matsumura Y, Watanabe D, Sasaki M, Kita T &
Inagaki N (2007) ABCA2 deficiency results in
K. Nagao et al. Function and regulation of ABCA1
FEBS Journal 278 (2011) 3190–3203 ª 2011 The Authors Journal compilation ª 2011 FEBS 3201
abnormal sphingolipid metabolism in mouse brain. J
Biol Chem 282, 19692–19699.
110 Vulevic B, Chen Z, Boyd JT, Davis W Jr, Walsh ES,
Belinsky MG & Tew KD (2001) Cloning and charac-
terization of human adenosine 5¢-triphosphate-binding
cassette, sub-family A, transporter 2 (ABCA2). Cancer
Res 61, 3339–3347.
111 Zhao L-X, Zhou C-J, Tanaka A, Nakata M, Hirabay-
ashi T, Amachi T, Shioda S, Ueda K & Inagaki N
(2000) Cloning, characterization, and tissue distribu-
tion of the ABC transporter Abca2. Biochem J 350,
865–872.

112 Mack JT, Beljanski V, Soulika AM, Townsend DM,
Brown CB, Davis W & Tew KD (2007) ‘Skittish’
Abca2 knockout mice display tremor, hyperactivity,
and abnormal myelin ultrastructure in the central
nervous system. Mol Cell Biol 27, 44–53.
113 Chen ZJ, Vulevic B, Ile KE, Soulika A, Davis W Jr,
Reiner PB, Connop BP, Nathwani P, Trojanowski JQ
& Tew KD (2004) Association of ABCA2 expression
with determinants of Alzheimer’s disease. FASEB J 18,
1129–1131.
114 Mace S, Cousin E, Ricard S, Genin E, Spanakis E,
Lafargue-Soubigou C, Genin B, Fournel R, Roche S,
Haussy G et al. (2005) ABCA2 is a strong genetic risk
factor for early-onset Alzheimer’s disease. Neurobiol-
ogy Dis 18, 119–125.
115 Wollmer MA, Kapaki E, Hersberger M, Muntwyler J,
Brunner F, Tsolaki M, Akatsu H, Kosaka K, Michik-
awa M, Molyva D et al. (2006) Ethnicity-dependent
genetic association of ABCA2 with sporadic Alzhei-
mer’s disease. Am J Med Genet B Neuropsychiatr Genet
141B, 534–536.
116 Ban N, Matsumura Y, Sakai H, Takanezawa Y, Sasa-
ki M, Arai H & Inagaki N (2007) ABCA3 as a lipid
transporter in pulmonary surfactant biogenesis. J Biol
Chem 282, 9628–9634.
117 Cheong N, Madesh M, Gonzales LW, Zhao M, Yu K,
Ballard PL & Shuman H (2006) Functional and traf-
ficking defects in ATP binding cassette A3 mutants
associated with respiratory distress syndrome. J Biol
Chem 281, 9791–9800.

118 Matsumura Y, Sakai H, Sasaki M, Ban N & Inagaki
N (2007) ABCA3-mediated choline-phospholipids
uptake into intracellular vesicles in A549 cells. FEBS
Lett 581, 3139–3144.
119 Fitzgerald ML, Xavier R, Haley KJ, Welti R, Goss JL,
Brown CE, Zhuang DZ, Bell SA, Lu N, McKee M et al.
(2007) ABCA3 inactivation in mice causes respiratory
failure, loss of pulmonary surfactant, and depletion of
lung phosphatidylglycerol. J Lipid Res 48, 621–632.
120 Cheong N, Zhang H, Madesh M, Zhao M, Yu K,
Dodia C, Fisher AB, Savani RC & Shuman H (2007)
ABCA3 is critical for lamellar body biogenesis in vivo.
J Biol Chem 282, 23811–23817.
121 Mulugeta S, Gray JM, Notarfrancesco KL, Gonzales
LW, Koval M, Feinstein SI, Ballard PL, Fisher AB &
Shuman H (2002) Identification of LBM180, a lamellar
body limiting membrane protein of alveolar type II
cells, as the ABC transporter protein ABCA3. J Biol
Chem 277, 22147–22155.
122 Nagata K, Yamamoto A, Ban N, Tanaka AR, Matsuo
M, Kioka N, Inagaki N & Ueda K (2004) Human
ABCA3, a product of a responsible gene for abca3 for
fatal surfactant deficiency in newborns, exhibits unique
ATP hydrolysis activity and generates intracellular
multilamellar vesicles. Biochem Biophys Res Commun
324, 262–268.
123 Yamano G, Funahashi H, Kawanami O, Zhao LX,
Ban N, Uchida Y, Morohoshi T, Ogawa J, Shioda S
& Inagaki N (2001) ABCA3 is a lamellar body mem-
brane protein in human lung alveolar type II cells.

FEBS Lett 508
, 221–225.
124 Shulenin S, Nogee LM, Annilo T, Wert SE, Whitsett
JA & Dean M (2004) ABCA3 gene mutations in new-
borns with fatal surfactant deficiency. N Engl J Med
350, 1296–1303.
125 Brasch F, Schimanski S, Muhlfeld C, Barlage S, Lang-
mann T, Aslanidis C, Boettcher A, Dada A, Schroten
H, Mildenberger E et al. (2006) Alteration of the pul-
monary surfactant system in full-term infants with
hereditary ABCA3 deficiency. Am J Respir Crit Care
Med 174, 571–580.
126 Sun H, Molday R & Nathans J (1999) Retinal stimu-
lates ATP hydrolysis by purified and reconstituted
ABCR, the photoreceptor-specific ATP-binding
cassette transporter responsible for Stargardt disease.
J Biol Chem 274, 8269–8281.
127 Ahn J, Wong JT & Molday RS (2000) The effect of
lipid environment and retinoids on the ATPase activity
of ABCR, the photoreceptor ABC transporter respon-
sible for Stargardt macular dystrophy. J Biol Chem
275, 20399–20405.
128 Beharry S, Zhong M & Molday RS (2004) N-retinylid-
ene-phosphatidylethanolamine is the preferred retinoid
substrate for the photoreceptor-specific ABC trans-
porter ABCA4 (ABCR). J Biol Chem 279, 53972–
53979.
129 Illing M, Molday LL & Molday RS (1997) The
220-kDa rim protein of retinal rod outer segments is a
member of the ABC transporter superfamily. J Biol

Chem 272, 10303–10310.
130 Molday LL, Rabin AR & Molday RS (2000) ABCR
expression in foveal cone photoreceptors and its role in
Stargardt macular dystrophy. Nat Genet 25, 257–258.
131 Sun H & Nathans J (1997) Stargardt’s ABCR is local-
ized to the disc membrane of retinal rod outer seg-
ments. Nat Genet 17, 15–16.
132 Allikmets R, Shroyer N, Singh N, Seddon J, Lewis R,
Bernstein P, Peiffer A, Zabriskie N, Li Y, Hutchinson
Function and regulation of ABCA1 K. Nagao et al.
3202 FEBS Journal 278 (2011) 3190–3203 ª 2011 The Authors Journal compilation ª 2011 FEBS
A et al. (1997) Mutation of the Stargardt disease gene
(ABCR) in age-related macular degeneration. Science
277, 1805–1807.
133 Allikmets R, Singh N, Sun H, Shroyer N, Hutchinson
A, Chidambaram A, Gerrard B, Baird L, Stauffer D,
Peiffer A et al. (1997) A photoreceptor cell-specific
ATP-binding transporter gene (ABCR) is mutated in
recessive Stargardt macular dystrophy. Nat Genet 15,
236–246.
134 Shroyer NF, Lewis RA, Yatsenko AN, Wensel TG &
Lupski JR (2001) Cosegregation and functional
analysis of mutant ABCR (ABCA4) alleles in fami-
lies that manifest both Stargardt disease and age-
related macular degeneration. Hum Mol Genet 10,
2671–2678.
135 Weng J, Mata NL, Azarian SM, Tzekov RT, Birch
DG & Travis GH (1999) Insights into the function of
Rim protein in photoreceptors and etiology of Star-
gardt’s disease from the phenotype in abcr knockout

mice. Cell 98, 13–23.
136 Kubo Y, Sekiya S, Ohigashi M, Takenaka C, Tamura
K, Nada S, Nishi T, Yamamoto A & Yamaguchi A
(2005) ABCA5 resides in lysosomes, and ABCA5
knockout mice develop lysosomal disease-like symp-
toms. Mol Cell Biol 25, 4138–4149.
137 Linsel-Nitschke P, Jehle AW, Shan J, Cao G, Bacic D,
Lan D, Wang N & Tall AR (2005) Potential role of
ABCA7 in cellular lipid efflux to apoA-I. J Lipid Res
46, 86–92.
138 Ikeda Y, Abe-Dohmae S, Munehira Y, Aoki R,
Kawamoto S, Furuya A, Shitara K, Amachi T, Kioka
N, Matsuo M et al. (2003) Posttranscriptional regula-
tion of human ABCA7 and its function for the
apoA-I-dependent lipid release. Biochem Biophys Res
Commun 311, 313–318.
139 Tsuruoka S, Ishibashi K, Yamamoto H, Wakaumi M,
Suzuki M, Schwartz GJ, Imai M & Fujimura A (2002)
Functional analysis of ABCA8, a new drug trans-
porter. Biochem Biophys Res Commun 298, 41–45.
140 Piehler A, Kaminski WE, Wenzel JJ, Langmann T &
Schmitz G (2002) Molecular structure of a novel
cholesterol-responsive A subclass ABC transporter,
ABCA9. Biochem Biophys Res Commun 295, 408–416.
141 Wenzel JJ, Kaminski WE, Piehler A, Heimerl S,
Langmann T & Schmitz G (2003) ABCA10, a novel
cholesterol-regulated ABCA6-like ABC transporter.
Biochem Biophys Res Commun 306, 1089–1098.
142 Mitsutake S, Suzuki C, Akiyama M, Tsuji K, Yanagi
T, Shimizu H & Igarashi Y (2010) ABCA12 dysfunc-

tion causes a disorder in glucosylceramide accumula-
tion during keratinocyte differentiation. J Dermatol
Sci 60, 128–129.
143 Yamanaka Y, Akiyama M, Sugiyama-Nakagiri Y,
Sakai K, Goto M, McMillan JR, Ota M, Sawamura D
& Shimizu H (2007) Expression of the keratinocyte
lipid transporter ABCA12 in developing and reconsti-
tuted human epidermis. Am J Path 171, 43–52.
144 Akiyama M, Sugiyama-Nakagiri Y, Sakai K,
McMillan JR, Goto M, Arita K, Tsuji-Abe Y, Tabata
N, Matsuoka K, Sasaki R et al. (2005) Mutations in
lipid transporter ABCA12 in harlequin ichthyosis and
functional recovery by corrective gene transfer. J Clin
Invest 115, 1777–1784.
145 Knight HM, Pickard BS, Maclean A, Malloy MP,
Soares DC, McRae AF, Condie A, White A, Hawkins
W, McGhee K et al. (2009) A cytogenetic abnormality
and rare coding variants identify ABCA13 as a candi-
date gene in schizophrenia, bipolar disorder, and
depression. Am J Hum Genet 85, 833–846.
146 Maekawa M, Kikuchi J, Kotani K, Nagao K, Odgerel
T, Ueda K, Kawano M, Furukawa Y &
Sakurabayashi I (2009) A novel missense mutation of
ABCA1 in transmembrane alpha-helix in a Japanese
patient with Tangier disease. Atherosclerosis
206,
216–222.
147 Hozoji-Inada M, Munehira Y, Nagao K, Kioka N &
Ueda K (2011) LXRb directly interacts with ABCA1
to promote HDL formation during acute cholesterol

accumulation. J Biol Chem in press.
148 Ueda K (2011) ABC proteins protect the human body
and maintain optimal health. Biosci Biotechnol
Biochem 75, 401–409.
K. Nagao et al. Function and regulation of ABCA1
FEBS Journal 278 (2011) 3190–3203 ª 2011 The Authors Journal compilation ª 2011 FEBS 3203

×