Accepted Manuscript Involvement of scavenger receptor class B type 1 and low-density lipoprotein receptor in the internalization of liposomes into HepG2 cells
Kumiko Sakai-Kato, Mari Sakurai, Yuki Takechi-Haraya, Kunie Nanjo, Yukihiro Goda PII: DOI: Reference:
S0005-2736(17)30279-1 doi: 10.1016/j.bbamem.2017.09.005 BBAMEM 82578
To appear in: Received date: Revised date: Accepted date:
16 January 2017 6 August 2017 5 September 2017
Please cite this article as: Kumiko Sakai-Kato, Mari Sakurai, Yuki Takechi-Haraya, Kunie Nanjo, Yukihiro Goda , Involvement of scavenger receptor class B type 1 and low-density lipoprotein receptor in the internalization of liposomes into HepG2 cells, (2017), doi: 10.1016/j.bbamem.2017.09.005
This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT
Involvement of scavenger receptor class B type 1 and low-density lipoprotein receptor in the internalization of liposomes into HepG2 cells
RI
PT
Kumiko Sakai-Kato*, Mari Sakurai, Yuki Takechi-Haraya, Kunie Nanjo, Yukihiro Goda
SC
Division of Drugs, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya, Tokyo
MA
NU
158-8501, Japan
*Corresponding author
D
Kumiko Sakai-Kato, PhD
CE
Tokyo 158-8501, Japan
PT E
Division of Drugs, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku,
Phone: +81-3-3700-9662
AC
Fax: +81-3-3700-9662
E-mail address:
[email protected]
1
ACCEPTED MANUSCRIPT Abstract In this study, HepG2 cells, an in vitro model system for human hepatocytes, were used to evaluate the interaction of lipoprotein receptors with liposomes carrying fluorescently labeled cholesterol and their subsequent intracellular uptake. In these experiments, two
PT
lipoprotein receptors, scavenger receptor class B type 1 (SR-B1) and low-density lipoprotein
RI
receptor (LDLR), accounted for approximately 20% and 10%, respectively, of the
SC
intracellular uptake of the labeled liposomes. These findings indicate that additional
NU
mechanisms contributed to liposomal internalization. Liposomes modified with both
MA
apolipoproteins A-I and E were internalized in HepG2 cells in FBS-depleted culture medium at the same levels as unmodified liposomes in FBS-containing culture medium, which
D
indicates that apolipoproteins A-I and E were the major serum components involved in
PT E
liposomal binding to SR-B1 or LDLR (or both). These results increase our understanding of
CE
the disposition of liposomes, processes that can directly affect the efficacy and safety of drug
AC
products.
Keywords: liposome; lipoprotein receptor; apolipoprotein; internalization; HepG2 cell
Abbreviations:
DMEM,
Dulbecco's
Modified
Eagle's
medium;
EDTA,
ethylenediaminetetraacetic acid; FBS, fetal bovine serum; HBSS, Hanks' Balanced Salt Solution; HDL, high-density lipoprotein; HSPC, hydrogenated soy phosphatidylcholine; LDL, 2
ACCEPTED MANUSCRIPT
low-density lipoprotein receptor; LDLR, low-density lipoprotein receptor; mAbs, monoclonal antibodies;
NBD-Chol,
25-[N-[(7-nitro-2-1,3-benzoxadiazol-4-yl)methyl]amino]-27-norcholesterol; PEG2000-DSPE, 2000)-1,2-distearoyl-sn-glycero-3-phosphoethanolamine;
PT
N-(carbonyl-methoxyPEG
PBS,
RI
phosphate buffered saline; siRNA, small inhibitory RNA; SR-B1, scavenger receptor class B
AC
CE
PT E
D
MA
NU
SC
type 1
3
ACCEPTED MANUSCRIPT
1. Introduction Nanotechnology-based drug formulations have been developed to improve the in vivo stability and pharmacokinetics of the encapsulated active substances or to modulate
PT
their release rate or active-site targeting [1,2]. Lipid-based formulations, such as
RI
liposomes, are being developed worldwide, and some are in clinical use [3–5]. Various
SC
types of drugs, including anticancer drugs, can be encapsulated within liposomes or in
of
liposomes
is
very
important
because
it
determines
their
MA
disposition
NU
their intramembrane space and then delivered to the target location in the body. The
blood-concentration profile, which in turn influences the safety and efficacy of
D
encapsulated drug products. Moreover, for drugs with repeated dosing, the accumulation
PT E
of lipid carriers needs to be considered because it can affect drug safety. Although the
CE
disposition of encapsulated drugs and liposomal uptake by macrophages have been elucidated in detail [6–9], little is known about the disposition of liposomes or
AC
liposomal constituents, such as cholesterol, by hepatocytes. According to one of the few reports available, a commercially available cholesterol-containing liposomal product is eliminated primarily through the feces (33%, compared with <2% in urine), presumably as a result of biliary excretion of intact liposomes or cholesterol (or both) [10]. Cholesterol appears to be especially important for stabilizing liposomes against the effects of various plasma components [11]. 4
ACCEPTED MANUSCRIPT
Low-density lipoproteins (LDLs), which are endogenous lipid-based particles, are incorporated into cells by binding to the low-density lipoprotein receptor (LDLR), a cell-surface glycoprotein that removes LDL from plasma by receptor-mediated endocytosis
PT
[12–15]. In addition to expressing LDLR, hepatocytes also express scavenger receptor class
RI
B type 1 (SR-B1), a multiligand receptor that binds a variety of lipoproteins, including
SC
high-density lipoprotein (HDL) and LDL [16,17]. Furthermore, SR-B1 is presumed to transfer additional cellular cholesterol to growing lipoprotein particles [18]. Apolipoproteins,
NU
which are ligands for several lipoprotein receptors, are thought to be involved in liposomal
MA
internalization, but the mechanism underlying the involvement of LDLR, SR-B1, and
D
apolipoproteins in this process has not yet been clarified [18–22]. Therefore, in this study, we through which lipoprotein receptors
and
PT E
investigated the molecular mechanism
apolipoproteins are involved in the internalization of PEGylated liposomes. The PEGylated
CE
liposomes that we used were approximately 100 nm in diameter and were composed of lipids,
AC
PEGylated lipids, cholesterol, and fluorescently labeled cholesterol (to enable the liposomes to be traced). The lipid composition and particle size, which were approximately the same as those of liposomal products in clinical use [3], minimized recognition of the liposomes by macrophages in the liver or spleen, thus prolonging their circulation [3,8]. Using HepG2 cells as an in vitro model system for human hepatocytes, we studied the interaction of our PEGylated liposomes with LDLR and SR-B1. 5
ACCEPTED MANUSCRIPT
2. Materials and Methods 2.1. Materials and cells Hydrogenated soy phosphatidylcholine (HSPC) and N-(carbonyl-methoxyPEG
NOF
Corporation
(Tokyo,
Japan);
RI
from
PT
2000)-1,2-distearoyl-sn-glycero-3-phosphoethanolamine (PEG2000-DSPE) were purchased
SC
25-[N-[(7-nitro-2-1,3-benzoxadiazol-4-yl)methyl]amino]-27-norcholesterol (NBD-Chol) was purchased from Avanti Polar Lipids (Alabaster, AL, USA); Dulbecco's Modified Eagle's
NU
medium (DMEM), RPMI-1640, penicillin–streptomycin, and Opti-MEM I were purchased
MA
from Life Technologies (Brooklyn, NY, USA); and fetal bovine serum (FBS) was obtained
D
from Nichirei Biosciences (Tokyo, Japan). Recombinant human apolipoproteins A-I and E
PT E
were purchased from Wako Pure Chemical Industries (Osaka, Japan). Apolipoprotein B from human plasma was purchased from Sigma-Aldrich (Tokyo, Japan). All other chemicals used
CE
in this study were of the highest purity available. HepG2 cells (American Type Culture
AC
Collection, Manassas, VA, USA) were cultured in DMEM supplemented with 10% FBS and 100 U/mL penicillin–streptomycin. Cells were grown in a humidified incubator at 37 °C and 5% CO2.
2.2. Preparation of liposomes The
lipid
composition
of
the
liposomes 6
used
in
our
experiments
was
ACCEPTED MANUSCRIPT
HSPC/PEG2000-DSPE/Chol/NBD-Chol
(56.3/5.3/37.4/1
mol
%).
NBD-Chol–labeled
liposomes were prepared by using a modified extrusion method [23]. Briefly, the desired amounts of lipids (60 µmol total lipids) was mixed in chloroform, and the mixture was dried
PT
by evaporation at 70 °C to create a thin homogeneous lipid film. The film was further dried
RI
overnight under vacuum desiccation to remove residual solvent. The dried film was hydrated
SC
with 2 mL of 5% (w/w) glucose aqueous solution under mechanical agitation at 70 °C for 5–10 min, and the hydrated film was dispersed by sonication for 5–10 min in a bath type
NU
sonicator (Sharp, Tokyo, Japan). The resulting dispersion was sonicated for an additional 1
MA
min with a tip-type sonicator (Sonics, Newtown, CT, USA). After sonication, the dispersion
D
was passed 21 times through a mini-extruder (Avanti Polar Lipids) equipped with
PT E
polycarbonate filters with 100-nm pores. The particle size, polydispersity index, and ζ-potential of the resulting liposomes were determined (Zetasizer Nano ZS, Malvern
CE
Instruments, Malvern, Worcestershire, United Kingdom). Hereafter, we refer to these
AC
NBD-Chol–labeled, PEGylated liposomes simply as “liposomes.”
2.3 Confocal microscopy The intracellular trafficking of the liposomes in HepG2 cells was followed by using confocal microscopy (A1, Nikon, Tokyo, Japan). Data were collected by using the manufacturer-supplied software and were exported as tagged image files. HepG2 cells (5 × 7
ACCEPTED MANUSCRIPT 105 cells per well) were plated in 35-mm glass-bottom dishes coated with poly-L-lysine (Matsunami, Osaka, Japan), each containing 1 mL of DMEM supplemented with 10% FBS (2 mL each well). After incubation for 72 h (37 °C, 5% CO2), the cells were exposed to
PT
liposomes (final lipid concentration, 50 μg/mL) in culture medium. After 4 h, the cells were
RI
washed with and kept in Hank’s balanced salt solution (HBSS) (Invitrogen, Carlsbad, CA,
SC
USA) for confocal imaging. The nuclei were labeled with Hoechst 33342 (Life Technologies)
NU
according to the manufacturer’s instructions.
MA
2.4. Inhibition of endocytosis
Endocytosis was inhibited with 10 μg/mL chlorpromazine (an inhibitor of
PT E
D
clathrin-mediated endocytosis), 150 μM genistein (which inhibits caveolae-mediated endocytosis), and 50 μM 5-(N-ethyl-N-isopropyl)amiloride (an inhibitor of macropinocytosis)
CE
[24,25]. We confirmed that these inhibitors did not cause significant toxicity (Supplementary
AC
Figure 1a) and that each effectively inhibited the appropriate endocytosis pathway (Supplementary Figure 1b–d). HepG2 cells (5 × 105 per well) were seeded in 12-well plates in DMEM containing 10% FBS (1 mL per well). After incubation for 48 h (37 °C, 5% CO2), one of the endocytosis inhibitors was added to the culture medium; 30 min later, liposomes in 500 μL of DMEM containing 10% FBS were added to the treated cells (final lipid concentration, 50 μg/mL), and the cells were incubated for 2 h (37 °C, 5% CO2). After 8
ACCEPTED MANUSCRIPT
incubation, the medium was replaced with PBS. Cells were trypsinized with 0.25% (v/v) trypsin–EDTA (Life Technologies), washed with HBSS three times, and suspended in lysis buffer (1.0% [v/v] Triton X-100 in HBSS). The resulting cell suspension was sonicated for 10
PT
min in a bath-type sonicator (Sharp), vortexed for 5 min, and centrifuged (15,000 × g, 4 °C,
RI
10 min).
SC
The fluorescence intensity of the supernatant was measured with a fluorescence spectrophotometer (F-7000, Hitachi High-Technologies, Tokyo, Japan). The excitation and
NU
emission wavelengths were 490 and 540 nm, respectively. The fluorescence intensity was
MA
normalized to the protein content of the cell lysate (Protein Assay Kit, Bio-Rad Laboratories,
PT E
D
Hercules, CA, USA).
2.5 LDLR and SR-B1 blocking studies
CE
Monoclonal antibodies (mAbs) were used to block the ligand-binding domains of
AC
LDLR (located in the extracellular region of the N terminus [26]) and SR-B1. The mAbs to the two receptors were obtained from Calbiochem (San Diego, CA, USA) and Abcam (Cambridge, UK), respectively. HepG2 cells (5 × 105 per well) were seeded on 12-well plates in DMEM containing 10% FBS (1 mL per well) and incubated for 48 h (37 °C, 5% CO2). Cells were incubated with or without 5 μg/mL anti-LDLR or anti-SR-B1 mAb for 1 h at 4 °C [20,27,28], and then NBD-Chol–labeled liposomes (final lipid concentration, 50 μg/mL) in 9
ACCEPTED MANUSCRIPT 500 μL of culture medium were added to the treated cells, which were incubated for 1 h (37 °C, 5% CO2). In preliminary experiments, we evaluated mAb doses from 2.5 to 10.0 μg/mL (LDLR) and from 2.5 to 5.0 μg/mL (SR-B1). Anti-LDLR mAb doses from 5.0 to 10.0
PT
μg/mL yielded similar inhibitory efficiencies, as did anti-SRB1 mAb doses from 2.5 to 5.0
RI
μg/mL. We therefore decided to use a dose of 5 μg/mL for both mAbs in the experiments. We
SC
chose the 1-h incubation time because LDLR recycling occurs—reportedly reaching a steady state, during which the amounts of receptor-bound and intracellular LDL remain
NU
constant—within 2 h [28]. Therefore, we used a shorter incubation time (i.e., 1 h) to capture
MA
the data before anti-LDLRs that were bound to LDLRs were replaced by the liposomes. Cells
D
incubated with an isotype control (anti-LDLR, purified mouse IgG2a [BioLegend, San Diego,
PT E
CA, USA]; anti-SR-B1, normal rabbit IgG [Santa Cruz Biotechnology, Dallas, TX, USA]) were used as reference controls. After incubation, the medium was replaced with PBS, and
AC
CE
the fluorescence intensity of the cells was measured as described in section 2.4.
2.6 Treatment with small inhibitory RNAs Small inhibitory RNA (siRNA) 25mer oligonucleotides for SR-B1 and LDLR and their negative controls were obtained from Life Technologies (Stealth RNAi and Stealth RNAi Negative Control Medium GC Duplex, respectively). The siRNAs were transfected into cells by using Lipofectamine RNAiMAX (Life Technologies) according to the 10
ACCEPTED MANUSCRIPT manufacturer’s protocols; 48 h after transfection, the culture medium was replaced with fresh medium containing liposomes, and incubation was continued for a further 2 h. After incubation, the medium was replaced with PBS, and the fluorescence intensity of the cells
RI
PT
was measured as described in section 2.4.
SC
2.7 Effect of FBS on internalization of liposomes
HepG2 cells (5 × 105 per well) were seeded in 12-well plates in DMEM containing
NU
10% FBS (1 mL per well). After incubation for 48 h (37 °C, 5% CO2), the cells were washed
MA
with PBS; liposomes (final lipid concentration, 50 μg/mL) either in 500 μL of culture
D
medium supplemented with 10% FBS or in 500 μL of FBS-depleted culture medium were
PT E
added; and the cultures were incubated for an additional 2 h at 37 °C. Similarly, to clarify the role of apolipoproteins in the internalization of liposome into the cells, the liposomes (final
CE
lipid concentration, 50 μg/mL) were preincubated with apolipoprotein A-I, E, or B (final
AC
concentration of each apolipoprotein, 33 μg/mL) at 4 °C overnight [29]. Then the apolipoprotein-modified liposomes were added to the HepG2 cells in 500 μL of FBS-depleted culture medium, and the cells were incubated for an additional 2 h at 37 °C. After incubation, the intracellular fluorescence intensity (derived from NBD-Chol) was quantified as described in section 2.4.
11
ACCEPTED MANUSCRIPT
2.8 Western blotting The expression of human LDLR and SR-B1 was detected by western blotting, as
PT
described previously [30], by using mAbs to LDLR and SR-B1 (both obtained from Abcam).
RI
2.9 Statistical analyses
SC
Results are presented as means ± SDs of at least three experiments. Two-group comparisons were performed by using Student’s t-test. A P value less than 0.05 was
MA
NU
considered significant.
D
3. Results and Discussion
PT E
3.1 Physicochemical properties of liposomes The mean particle size of the liposomes was 115 nm; the polydispersity index value
CE
was 0.12, indicating a homogenous size distribution; and the ζ-potential was –2.3 mV. We
AC
confirmed that the mean particle size and ζ-potential of the liposomes in culture medium containing 10% FBS were 117 nm and –2.6 mV, which are equivalent to the values measured in PBS without FBS. Furthermore, the liposome particles were stable even in FBS-containing culture medium at 37 °C for 3 h (Figure 1a).
3.2 Intracellular distribution of liposomes 12
ACCEPTED MANUSCRIPT
We used confocal microscopy to study the intracellular distribution of the inherently fluorescent liposomes in HepG2 cells. The expected dot-like staining pattern was localized to the perinuclear regions but not to the nucleus (Figure 1b,c). In addition, the plasma
PT
membrane remained unstained, thus demonstrating that nonspecific binding of the liposomes
SC
RI
to the plasma membrane was negligible.
3.3 Mechanism of liposome internalization into HepG2 cells
NU
Treatment with chlorpromazine, an inhibitor of clathrin-mediated endocytosis,
MA
significantly suppressed the fluorescence intensity of the liposomes in HepG2 cells (P < 0.01;
D
Figure 2). In contrast, treatment with either 5-(N-ethyl-N-isopropyl)amiloride or genistein did
PT E
not alter the amount of intracellular liposomes. Given that the contributions of caveolae-mediated endocytosis and macropinocytosis to liposomal internalization were
transport
into
the
hepatocytes
was
clathrin-mediated
endocytosis.
AC
liposomes
CE
negligible in our system (Figure 2), these observations suggest that a key mechanism of
Clathrin-mediated endocytosis reportedly is the primary mechanism by which nanoparticles are internalized into cells [31]. In particular, clathrin-mediated endocytosis is reported to be involved in the uptake of nanometer-sized liposomes into A549 and HeLa cells [32]. In contrast, cationic liposomes have been reported to be internalized in HepG2 cells by lipid-raft-mediated endocytosis [33]. Because the endocytotic pathway can vary depending 13
ACCEPTED MANUSCRIPT on the nanoparticle’s physicochemical properties (e.g., size, charge) [34], we cannot compare our current findings with the previous studies in detail, but our results are consistent with
PT
those supporting clathrin-mediated endocytosis of liposomes into hepatocytes.
RI
3.4 Binding of liposomes to lipoprotein-related receptors on HepG2 cells
SC
Clathrin-mediated endocytosis is mediated by cell-surface receptors [35]. To examine the involvement of lipoprotein-related receptors in the endocytosis of liposomes, we
NU
first confirmed the expression of LDLR and SR-B1 in the HepG2 cells used in our study. As
MA
shown in Figure 3a, western blotting confirmed that both proteins were expressed in HepG2
D
cells, as indicated by an LDLR-specific band at approximately 120 kDa and an
PT E
SR-B1-specific band at approximately 76 kDa; these values are close to the reported values [20,36].
CE
We then examined the involvement of LDLR and SR-B1 in the intracellular transfer
AC
of the liposomes. Both receptors are known to be involved in the internalization of cholesterol to maintain intrinsic cholesterol homeostasis [12–15,36]. The intracellular fluorescence intensity derived from NBD-Chol decreased to 92% of that of the control when anti-LDLR mAb was used and to 84% when anti-SR-B1 mAb was used; the reduction due to the anti-SR-B1 mAb was significant (P < 0.01) (Figure 3b). These results show that LDLR and SR-B1 were involved in the intracellular transfer of the liposomes. When both receptors 14
ACCEPTED MANUSCRIPT
were inhibited simultaneously, the inhibition efficiency was 63% (P < 0.05, Figure 3c). These data show that LDLR and SR-B1 acted cooperatively; as reported previously, SR-B1 mediates LDL association with and uptake by cells that do not express any LDLR [37].
interactions
[32,34].
Our
findings
imply
that
in
addition
to
RI
nonspecific
PT
Nanoparticles reportedly are internalized through multiple mechanisms, including
SC
lipoprotein-receptor–mediated endocytosis, other processes (such as clathrin- and caveolae-independent endocytosis through receptor–ligand binding, and endocytosis through
NU
nonspecific interactions [34]) were involved in the internalization of the liposomes by
MA
hepatocytes.
D
We used receptor-specific siRNAs to confirm the significant inhibitory effect of
PT E
knockdown of lipoprotein receptor production on intracellular fluorescence due to NBD-Chol. The efficiency of siRNA knockdown was confirmed through ImageJ analysis of the
CE
individual bands in the western blots [38] (as described in section 2.8), which indicated that
AC
LDLR and SR-B1 expression levels were decreased to 28% and 23%, respectively (Figure 4a). Treatment with the LDLR-specific siRNA decreased the fluorescence inside HepG2 cells to 91% of that when an siRNA negative control was used (P < 0.01, Figure 4b). When HepG2 cells were exposed to the SR-B1-specific siRNA, NBD-Chol–derived intracellular fluorescence decreased to 66% of the control value (P < 0.01, Figure 4b). These results confirm those of the receptor-blocking experiment (Figure 3) and indicate that both LDLR 15
ACCEPTED MANUSCRIPT
and SR-B1 were specifically involved in the internalization of the liposomes into hepatocytes. The difference between the inhibition levels observed in the antibody blocking and siRNA knockdown experiments (Figures 3 and 4, respectively) likely reflects the differing
PT
mechanisms of antibody inhibition and siRNA knockdown. Regardless, both methods of
RI
receptor inhibition show that the internalization of the liposomes by HepG2 cells was
SC
mediated specifically by LDLR and SR-B1.
NU
3.5 Involvement of apolipoproteins in liposomal binding to receptors
MA
The binding of lipoproteins such as LDL and HDL to LDLR and SR-B1 reportedly
D
is mediated by apolipoproteins, including apolipoproteins A-I, B, and E in serum [39,40]. In
PT E
addition, PEGylated liposomes administered to mice were shown to bind to serum apolipoproteins, but whether these proteins are involved in the clearance of PEGylated
CE
liposomes from the bloodstream has not yet been clarified [41].
AC
As shown in Figure 5, when liposomes in FBS-depleted culture medium were added to HepG2 cells, the observed intracellular NBD-Chol fluorescence intensity was about 40% less than that observed when the liposomes were added in culture medium containing 10% FBS (control). However, the decrease in intensity was significantly attenuated when liposomes modified with apolipoprotein A-I (P < 0.05, Figure 5a) or apolipoprotein E (P < 0.01, Figure 5c) were added. In contrast, apolipoprotein B did not attenuate the decrease in 16
ACCEPTED MANUSCRIPT
intensity (Figure 5b). This suggests some interesting size-dependent characteristics of apolipoprotein adsorption or opsonization of liposomes and their subsequent cellular uptake. Interestingly, when liposomes modified with both apolipoproteins A-I and E were added in
PT
FBS-depleted medium, there was no decrease in fluorescence intensity relative to that in the
RI
control experiment (Figure 5d). These results show that apolipoproteins A-I and E played a
SC
key role in the internalization of the liposomes into HepG2 cells. Apolipoproteins A-I and E have high affinity for SR-B1 and LDLR, respectively [39, 42], and our results indicate that
NU
liposome-adsorbed apolipoproteins A-I and E were recognized by SR-B1 and LDLR. These
MA
results are consistent with our results indicating that cooperation between LDLR and SR-B1
D
was involved in the internalization of the liposomes (Figure 3c). Therefore, we suggest that
PT E
the liposomes were taken up by endocytic process involving LDLR and SR-B1. SR-B1 is known to promote selective uptake of HDL–cholesteryl ester by cells without particle uptake.
CE
In addition, SR-BI reportedly binds HDL with high affinity and mediates the selective uptake
AC
of HDL–cholesteryl ester into the liver [43,44]. SR-BI also mediates the bidirectional flux of free cholesterol between cells and HDL [45–47]. Therefore, some portion of the cholesterol contained in the liposomes may have entered HepG2 cells via SR-B1 in the same manner as cholesterol or cholesteryl ester in HDL.
4. Conclusions 17
ACCEPTED MANUSCRIPT
We have clarified the contributions of LDLR and SR-B1 to the internalization of liposomes into HepG2 cells. Our results suggest that apolipoproteins A-I and E are bound to the surface of liposomes and are involved in their recognition by the lipoprotein receptors on
PT
hepatocytes. Our findings imply that in addition to lipoprotein-receptor–mediated endocytosis,
RI
other processes (such as clathrin- and caveolae-independent endocytosis through
SC
receptor–ligand binding, and endocytosis through nonspecific interactions [34]) are involved in the internalization of liposomes by hepatocytes. Further research is necessary to clarify the
NU
correlation between the physicochemical properties of liposomes (including lipid
MA
composition) and receptor specificity, with the goals of controlling liposome disposition and
D
targeting liposomes to specific tissues. We surmise that the knowledge obtained from our
PT E
current study with HepG2 cells will facilitate the development of hepatocyte-targeting liposomes and will provide insights into the accumulation and metabolism of liposomes,
AC
CE
given that drug products are predominantly metabolized and excreted through the liver.
Acknowledgement
This work was supported in part by a JSPS KAKENHI (grant no. 15K07915 [K.S.-K.]) and by funding for Research on Regulatory Harmonization and Evaluation of Pharmaceuticals, Medical Devices, Regenerative and Cellular Therapy Products, Gene Therapy Products, and Cosmetics, and Research on Development of New Drugs from the 18
ACCEPTED MANUSCRIPT
Japan Agency for Medical Research and Development, AMED. The authors would like to
AC
CE
PT E
D
MA
NU
SC
RI
PT
thank R. Takizawa and T. Mano for their technical assistance with the experiments.
19
ACCEPTED MANUSCRIPT References
[1]
K. Sakai-Kato, N. Nishiyama, M. Kozaki, T. Nakanishi, Y. Matsuda, M. Hirano, H. Hanada, S. Hisada, H. Onodera, H. Harashima, Y. Matsumura, K. Kataoka, Y. Goda, H.
PT
Okuda, T. Kawanishi, General considerations regarding the in vitro and in vivo
RI
properties of block copolymer micelle products and their evaluation, J. Control. Release
[2]
SC
210 (2015) 76-83.
M. Ferrari, Cancer nanotechnology: opportunities and challenges, Nat. Rev. Cancer 5
Y. Barenholz, Doxil®-the first FDA-approved nano-drug: lessons learned, J. Control.
MA
[3]
NU
(2005) 161-171.
R. Duncan, R. Gaspar, Nanomedicine(s) under the microscope, Mol. Pharmaceutics 8 (2011) 2101-2141.
F. Perche, V.P. Torchilin, Recent trends in multifunctional liposomal nanocarriers for
CE
[5]
PT E
[4]
D
Release 160 (2012) 117-134.
[6]
AC
enhanced tumor targeting, J. Drug Deliv. (2013) 705265. A. Gabizon, R. Catane, B. Uziely, B. Kaufman, T. Safra, R. Cohen, F. Martin, A. Huang, Y. Barenholz, Prolonged circulation time and enhanced accumulation in malignant exudates of doxorubicin encapsulated in polyethylene-glycol coated liposomes, Cancer Res. 54 (1994) 987-992.
20
ACCEPTED MANUSCRIPT
[7]
K. Mross, B. Niemann, U. Massing, J. Drevs, C. Unger, R. Bhamra, C.E. Swenson, Pharmacokinetics of liposomal doxorubicin (TLC-D99;Myocet) in patients with solid tumors: an open-label, single-dose study, Cancer Chemother. Pharmacol. 54 (2004)
S.M. Moghimi, A.C. Hunter, J.C. Murray, Long-circulating and target-specific
RI
[8]
PT
514-524.
[9]
SC
nanoparticles: theory to practice, Pharmacol. Rev. 53 (2001) 283-318. J.A. Kamps, G.L. Scherphof, Receptor versus non-receptor mediated clearance of
R.W. Townsend, A. Zutshi, I. Bekersky, Biodistribution of 4-[14C]Cholesterol-AmBisome
MA
[10]
NU
liposomes, Advanced Drug Delivery Reviews 32 (1998) 81-97.
D
following a Single Intravenous Administration to Rats, Drug Metab. Dispos. 29 (2001)
[11]
PT E
681-685.
D.C. Drummond, O. Meyer, K. Hong, D.B.
Kirpotin, D. Papahadjopoulos, Optimizing
CE
liposomes for delivery of chemotherapeutic agents to solid tumors, Pharmacol. Rev. 51
[12]
AC
(1999) 691-743.
M.S. Brown, J.L. Goldstein, Familial hypercholesterolemia: defective binding of lipoproteins
to
cultured
fibroblasts
associated
with
impaired
regulation
of
3-hydroxy-3-methylglutaryl coenzyme A reductase activity, Proc. Natl. Acad. Sci. USA. 71 (1974) 788-792.
21
ACCEPTED MANUSCRIPT
[13]
J.L. Goldstein, M.S. Brown, Binding and degradation of low density lipoproteins by cultured human fibroblasts. Comparison of cells from a normal subject and from a patient with homozygous familial hypercholesterolemia, J. Biol. Chem. 249 (1974)
M.S. Brown, J.R. Faust, J.L. Goldstein, Role of the low density lipoprotein receptor in
RI
[14]
PT
5153-5162.
SC
regulating the content of free and esterified cholesterol in human fibroblasts, J. Clin. Invest. 55 (1975) 783-793.
J.L. Goldstein, M.S. Brown, Regulation of low-density lipoprotein receptors:
NU
[15]
X.P. Yang, M.J. Amar, B. Vaisman, A.V. Bocharov, T.G. Vishnyakova, L.A. Freeman,
PT E
[16]
D
Circulation 76 (1987) 504-507.
MA
implications for pathogenesis and therapy of hypercholesterolemia and atherosclerosis,
R.J. Kurlander, A.P. Patterson, L.C. Becker, A.T. Remaley, Scavenger receptor-BI is a
S. Acton, A. Rigotti, K.T. Landschulz, S. Xu, H.H. Hobbs, M. Krieger. Identification of
AC
[17]
CE
receptor for lipoprotein(a), J. Lipid Res. 54 (2013) 2450-2457.
scavenger receptor SR-BI as a high density lipoprotein receptor, Science 271 (1996) 518-520. [18]
V.I. Zannis, A. Chroni, M. Krieger, Role of apoA-I, ABCA1, LCAT, and SR-BI in the biogenesis of HDL, J. Mol. Med. 84 (2006) 276-294.
22
ACCEPTED MANUSCRIPT
[19]
H.R. Kim, K. Andrieux, S. Gil, M. Taverna, H. Chacun, D. Desmaële, F. Taran, D. Georgin,
P.
Couvreur,
Translocation
of
poly
(ethylene
glycol-co-hexadecyl)
cyanoacrylate nanoparticles into rat brain endothelial cells: role of apolipoproteins in
H.R. Kim, S. Gil, K. Andrieux, V. Nicolas, M. Appel, H. Chacun, D. Desmaële,
RI
[20]
PT
receptor-mediated endocytosis, Biomacromolecules 8 (2007) 793-799.
SC
Low-density lipoprotein receptor-mediated endocytosis of PEGylated nanoparticles in rat brain endothelial cells, Cell. Mol. Life Sci. 64 (2007) 356-364. X. Yan, K. Poelstra, G.L. Scherphof, J.A. Kamps, A role for scavenger receptor B-I in
NU
[21]
X. Yan, F. Kuipers, L.M. Havekes, R. Havinga, B. Dontje, K. Poelstra, G.L. Scherphof, J.
PT E
[22]
D
Commun. 325 (2004) 908-914.
MA
selective transfer of rhodamine-PE from liposomes to cells, Biochem. Biophys. Res.
A. Kamps, The role of apolipoprotein E in the elimination of liposomes from blood by
Y. Takechi-Haraya, K. Sakai-Kato, Y. Abe, T. Kawanishi, H. Okuda, Y. Goda, Atomic
AC
[23]
CE
hepatocytes in the mouse, Biochem. Biophys. Res. Commun. 328 (2005) 57-62.
force microscopic analysis of the effect of lipid composition on liposome membrane rigidity, Langmuir 32 (2016) 6074-6082. [24]
J. Rejman, A. Bragonzi, M. Conese. Role of clathrin- and caveolae-mediated endocytosis in gene transfer mediated by lipo- and polyplexes, Mol. Ther. 12 (2005) 468-474.
23
ACCEPTED MANUSCRIPT
[25]
A.P. Perez, M.L. Cosaka, E.L. Romero, M.J. Morilla, Uptake and intracellular traffic of siRNA dendriplexes in glioblastoma cells and macrophages, Int. J. Nanomedicine 6 (2011) 2715-2728. H.J. Kwon, T.A. Lagace, M.C. McNutt, J.D. Horton, J. Deisenhofer, Molecular basis for
PT
[26]
RI
LDL receptor recognition by PCSK9, Proc. Natl. Acad. Sci. USA. 105 (2008)
[27]
SC
1820-1825.
M.F. Lopes-Virella, N. Binzafar, S. Rackley, A. Takei, M. La Via, G. Virella, The uptake
NU
of LDL-IC by human macrophages: predominant involvement of the Fc gamma RI
J.L. Goldstein, S.K. Basu, M.S. Brown, Receptor-mediated endocytosis of low-density
D
[28]
MA
receptor, Atherosclerosis 135 (1997) 161-170.
M. Tamaru, H. Akita, T. Nakatani, K. Kajimoto, Y. Sato, H. Hatakeyama, H. Harashima,
CE
[29]
PT E
lipoprotein in cultured cells, Methods Enzymol. 98 (1983) 241-260.
AC
Application of apolipoprotein E-modified liposomal nanoparticles as a carrier for delivering DNA and nucleic acid in the brain, Int. J. Nanomedicine 9 (2014) 4267–4276. [30]
K. Sakai-Kato, K. Ishikura, Y. Oshima, M. Tada, T. Suzuki, A. Ishii-Watabe, T. Yamaguchi, N. Nishiyama, K. Kataoka, T. Kawanishi, H. Okuda, Evaluation of intracellular trafficking and clearance from HeLa cells of doxorubicin-bound block copolymers, Int. J. Pharm. 423 (2012) 401- 409.
24
ACCEPTED MANUSCRIPT
[31]
Y. Li, J. Wang, M.G. Wientjes, J.L. Au, Delivery of nanomedicines to extracellular and intracellular compartments of a solid tumor, Adv. Drug Deliv. Rev. 64 (2012) 29-39.
[32]
J. Rejman, A. Bragonzi, M. Conese, Role of clathrin- and caveolae-mediated endocytosis
Y. Li, L. Gao, X. Tan, F. Li, M. Zhao, S. Peng, Lipid rafts-mediated endocytosis and
RI
[33]
PT
in gene transfer mediated by lipo- and polyplexes, Mol. Ther. 12 (2005) 468-474
SC
physiology-based cell membrane traffic models of doxorubicin liposomes, BBA.
S. Zhang, H. Gao, G. Bao, Physical Principles of Nanoparticle Cellular Endocytosis, ACS. Nano. 9 (2015) 8655-8671.
D
H.T. McMahon, E. Boucrot, Molecular mechanism and physiological functions of
PT E
[35]
MA
[34]
NU
Biomembranes 1858 (2016) 1801-1811.
clathrin-mediated endocytosis, Nat. Rev. Mol. Cell Biol. 12 (2011) 517-533. D. Rhainds, L. Brissette, The role of scavenger receptor class B type I (SR-BI) in lipid
CE
[36]
39-77. [37]
AC
trafficking. defining the rules for lipid traders, Int. J. Biochem. Cell. Biol. 36 (2004)
T.A. Pagler, S. Rhode, A. Neuhofer, H. Laggner, W. Strobl, C. Hinterndorfer, I. Volf, M. Pavelka, E.R. Eckhardt, D.R. van der Westhuyzen, G.J. Schütz, H. Stangl, SR-BI-mediated high density lipoprotein (HDL) endocytosis leads to HDL resecretion facilitating cholesterol efflux, J. Biol. Chem. 281(2006) 11193-11204.
25
ACCEPTED MANUSCRIPT
[38]
C.A. Schneider, W.S. Rasband, K.W. Eliceiri, NIH Image to ImageJ: 25 years of image analysis, Nat. Methods 9 (2012) 671-675.
[39]
M. Krieger, Charting the fate of the "good cholesterol": identification and
PT
characterization of the high-density lipoprotein receptor SR-BI, Annu. Rev. Biochem. 68
A. Chait, The role of lipoprotein receptors in lipid transport and in the pathogenesis of
SC
[40]
RI
(1999) 523-558.
the hyperlipoproteinemias, Spec. Top. Endocrinol. Metab. 5 (1983) 1-53. M. Kawanishi, Y. Hashimoto, T. Shimizu, I. Sagawa, T. Ishida, H. Kiwada,
NU
[41]
MA
Comprehensive analysis of PEGylated liposome-associated proteins relating to the
D
accelerated blood clearance phenomenon by combination with shotgun analysis and
[42]
PT E
conventional methods, Biotechnol. Appl. Biochem. 62 (2015) 547-555. D.M. Hatters, C.A. Peters-Libeu, K.H. Weisgraber, Apolipoprotein E structure: insights
B. Trigatti, A. Rigotti, M. Krieger, The role of the high-density lipoprotein receptor
AC
[43]
CE
into function, Trends Biochem. Sci. 31 (2006) 445-454
SR-BI in cholesterol metabolism, Curr. Opin. Lipidol. 11 (2000) 123–131. [44]
D.L. Silver, X.C.Jiang, T. Arai, C. Bruce, and A.R. Tall, Receptors and lipid transfer proteins in HDL metabolism, Ann. N.Y. Acad. Sci. 902 (2000) 103–111.
26
ACCEPTED MANUSCRIPT
[45]
M.F. Linton, H. Tao, E.F. Linton, P.G. Yancey. SR-BI: A Multifunctional Receptor in Cholesterol Homeostasis and Atherosclerosis, Trends Endocrinol. Metab. 28 (2017) 461-472. G. Kellner-Weibel, M. de La Llera-Moya, M.A. Connelly, G. Stoudt, A.E. Christian, M.P.
PT
[46]
RI
Haynes, D.L. Williams, G.H. Rothblat, Expression of scavenger receptor BI in COS-7
P.G. Yancey, M. de la Llera-Moya, S. Swarnakar, P. Monzo, S.M. Klein, M.A. Connelly,
NU
W.J. Johnson, D.L. Williams, G.H. Rothblat, High density lipoprotein phospholipid
MA
composition is a major determinant of the bi-directional flux and net movement of
D
cellular free cholesterol mediated by scavenger receptor BI, J. Biol. Chem. 275 (2000)
CE
PT E
36596–36604.
AC
[47]
SC
cells alters cholesterol content and distribution, Biochemistry 39 (2000) 221–229.
27
ACCEPTED MANUSCRIPT
Figure Legends
Figure 1. (a) Temporal dependence of liposomal particle size during incubation of liposomes
PT
in culture medium containing 10% FBS.
RI
(b and c) Confocal images showing the intracellular distribution of fluorescently labeled
SC
liposomes (final lipid concentration, 50 μg/mL) in HepG2 cells: (b) negative control (no
MA
nuclei were labeled with Hoechst dye (blue).
NU
liposomes) and (c) liposomes added. Liposomes were labeled with NBD-Chol (green), and
D
Figure 2. Effect of endocytosis inhibitors on the intensity of intracellular NBD-Chol
PT E
fluorescence. HepG2 cells were treated with each inhibitor for 30 min before the labeled liposomes were added; intracellular uptake of the labeled liposomes was evaluated 2 h after
CE
they were added to the cells. **, P < 0.01 compared with the corresponding untreated control.
AC
Data are presented as means SDs (n = 4).
Figure 3. Effect of pretreatment with anti-LDLR and anti-SR-B1 monoclonal antibodies (mAbs) on intracellular NBD-Chol fluorescence. (a) Detection of LDLR and SR-B1 in HepG2 cells by western blotting.
28
ACCEPTED MANUSCRIPT (b) HepG2 cells were pretreated for 1 h at 4 °C with 5 μg/mL of either anti-LDLR or anti-SR-B1 mAb before labeled liposomes were added. Intracellular NBD-Chol fluorescence was measured 1 h after the addition of the liposomes to the cells. **, P < 0.01 compared with
PT
the respective isotype control. Data are presented as means SDs (n = 4).
RI
(c) HepG2 cells were pretreated for 1 h at 4 °C with 5 μg/mL anti-LDLR and 5 μg/mL
SC
anti-SR-B1 mAb before labeled liposomes were added. Intracellular NBD-Chol fluorescence was measured 1 h after the addition of the liposomes to the cells. *, P < 0.05 compared with
MA
NU
the mixture containing both isotype controls. Data are presented as means SDs (n = 4).
D
Figure 4. Effect of LDLR and SR-B1 knockdown on the intensity of intracellular NBD-Chol
(a)
PT E
fluorescence.
Knockdown of LDLR and SR-B1 expression by using specific siRNAs. HepG2 cells
Effect of LDLR and SR-B1 knockdown on the intensity of intracellular NBD-Chol
AC
(b)
CE
were also treated with siRNA negative controls. The arrows indicate the target bands.
fluorescence. Fluorescence was evaluated 2 h after labeled liposomes were added to HepG2 cells. **, P < 0.01 compared with the control group (that is, cells treated with negative-control siRNAs). Data are presented as means SDs (n = 4).
Figure 5. Effect of apolipoproteins on the intensity of intracellular NBD-Chol fluorescence. 29
ACCEPTED MANUSCRIPT
Intracellular NBD-Chol fluorescence was evaluated 2 h after the addition of labeled liposomes to HepG2 cells in culture medium containing 10% FBS (control group) and in culture medium without FBS (FBS(-)). Intracellular NBD-Chol fluorescence was also
PT
evaluated 2 h after the addition of apolipoprotein-modified labeled liposomes to HepG2 cells
RI
in culture medium without FBS (FBS(-) + Apo).
SC
(a) Apolipoprotein A-I, (b) apolipoprotein B, (c) apolipoprotein E, and (d) apolipoproteins A-I and E.
NU
**, P < 0.01 compared with the corresponding groups. *, P < 0.05 compared with the
AC
CE
PT E
D
MA
corresponding groups. Data are presented as means SDs (n = 3).
30
AC
CE
PT E
D
MA
NU
SC
RI
PT
ACCEPTED MANUSCRIPT
31
AC
CE
PT E
D
MA
NU
SC
RI
PT
ACCEPTED MANUSCRIPT
32
AC
CE
PT E
D
MA
NU
SC
RI
PT
ACCEPTED MANUSCRIPT
33
AC
CE
PT E
D
MA
NU
SC
RI
PT
ACCEPTED MANUSCRIPT
34
AC
CE
PT E
D
MA
NU
SC
RI
PT
ACCEPTED MANUSCRIPT
35
AC
CE
PT E
D
MA
NU
SC
RI
PT
ACCEPTED MANUSCRIPT
36
ACCEPTED MANUSCRIPT
AC
CE
PT E
D
MA
NU
SC
RI
PT
Graphical abstract
37
ACCEPTED MANUSCRIPT Highlights We used fluorescent NBD-Chol-liposomes to study liposomal transit into hepatocytes.
SR-B1 and LDLR participated in liposome uptake by HepG2 cells.
Apolipoprotein A-I and E mediated the interactions among SR-B1, LDLR, and liposomes.
AC
CE
PT E
D
MA
NU
SC
RI
PT
38