Exogenous plant mir168a specifically targets mammalian ldlrap1: evidence of cross-kingdom regulation by microrna
Cell Research (2012) 22:107-126.
2012 IBCB, SIBS, CAS All rights reserved 1001-0602/12 $ 32.00
Exogenous plant MIR168a specifically targets mammalian
LDLRAP1: evidence of cross-kingdom regulation by
Lin Zhang1, *, Dongxia Hou1, *, Xi Chen1, *, Donghai Li1, *, Lingyun Zhu1, 2, Yujing Zhang1, Jing Li1, Zhen Bian1,
Xiangying Liang1, Xing Cai1, Yuan Yin1, Cheng Wang1, Tianfu Zhang1, Dihan Zhu1, Dianmu Zhang1, Jie Xu1,
Qun Chen1, Yi Ba3, Jing Liu1, Qiang Wang1, Jianqun Chen1, Jin Wang1, Meng Wang1, Qipeng Zhang1,
Junfeng Zhang1, Ke Zen1, Chen-Yu Zhang1
1Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Bio-technology, School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing, Jiangsu 210093, China; 2Department of Chem-istry and Biology, School of Science, National University of Defense Technology, Changsha, Hunan 410073, China; 3Tianjin Medi-cal University Cancer Institute and Hospital, Huanhuxi Road, Tiyuanbei, Tianjin 300060, China
Our previous studies have demonstrated that stable microRNAs (miRNAs) in mammalian serum and plasma are
actively secreted from tissues and cells and can serve as a novel class of biomarkers for diseases, and act as signaling
molecules in intercellular communication. Here, we report the surprising finding that exogenous plant miRNAs are
present in the sera and tissues of various animals and that these exogenous plant miRNAs are primarily acquired
orally, through food intake. MIR168a is abundant in rice and is one of the most highly enriched exogenous plant
miRNAs in the sera of Chinese subjects. Functional studies in vitro and in vivo demonstrated that MIR168a could
bind to the human/mouse low-density lipoprotein receptor adapter protein 1 (LDLRAP1) mRNA, inhibit LDLRAP1
expression in liver, and consequently decrease LDL removal from mouse plasma. These findings demonstrate that ex-
ogenous plant miRNAs in food can regulate the expression of target genes in mammals.
Keywords: microRNA; MIR168a; LDLRAP1; low-density lipoprotein; microvesicle; cross-kingdom
Cell Research (2012)
22:107-126. doi:10.1038/cr.2011.158; published online 20 September 2011
mune response, and the maintenance of cell and tissue
identity [1, 2]. Dysregulation of miRNAs has been linked
MicroRNAs (miRNAs), a class of 19-24 nucleotide
to cancer and other diseases [3, 4]. Recently, we and oth-
long non-coding RNAs derived from hairpin precursors,
ers found that mammalian miRNAs exist stably in the
mediate the post-transcriptional silencing of an estimated
sera and plasma of humans and animals [5, 6]. Specific
30% of protein-coding genes in mammals by pairing
serum miRNA expression profiles show a great potential
with complementary sites in the 3′ untranslated regions
to serve as a novel class of biomarkers for the diagnosis
(UTRs) of target genes [1, 2]. miRNAs have been widely
of cancer and other diseases [5, 6]. Numerous reports
shown to modulate various critical biological processes,
have subsequently shown that unique expression patterns
including differentiation, apoptosis, proliferation, the im-
of circulating miRNAs reflect various physiological and
pathological conditions [7-12].
We next characterized the possible carrier of circulat-
*These four authors contributed equally to this work.
ing miRNAs. Microvesicles (MVs) are small vesicles
Correspondence: Chen-Yu Zhanga, Ke Zenb, Junfeng Zhangc
that are shed from almost all cell types under both nor-
mal and pathological conditions [13, 14]. They bear sur-
bE-mail:
[email protected]
face receptors/ligands of the original cells and have the
Received 11 August 2011; revised 23 August 2011; accepted 26 August
potential to selectively interact with specific target cells
2011; published online 20 September 2011
and mediate intercellular communication by transporting
npg Plant MIR168a downregulates mammalian LDLRAP1108
bioactive lipids, mRNA, or proteins between cells [13,
was rice. Plant miRNAs were also detected in the sera
14]. Our recent results demonstrated that MVs from hu-
of other animals, such as calves, whose diet was mainly
man plasma are a mixture of microparticles, exosomes,
green rich fodder (Supplementary information, Table
and other vesicular structures and that many types of
S2). The significant differences in plant miRNA profiles
MVs in human plasma contain miRNAs [15]. These find-
between human and calf sera are shown in Supplemen-
ings were in agreement with the recent reports by other
tary information, Figure S1B, with a Pearson's correla-
investigators that exosomes from cultured cells served as
tion coefficient (
R) of 0.3951. Furthermore, although the
physiological carriers of miRNAs [16, 17]. Our further
number of plant miRNAs was about 5% of mammalian
studies demonstrated that miRNAs could be selectively
miRNAs in human and calf sera (Supplementary infor-
packaged into MVs and actively delivered into recipient
mation, Figure S1C and S1E), there were fewer absolute
cells where the exogenous miRNAs can regulate target
sequencing reads from the plant miRNAs (Supplementary
gene expression and recipient cell function [15]. Thus,
information, Figure S1D and S1F). To confirm the Sol-
secreted miRNAs can serve as a novel class of signaling
exa data, the levels of MIR168a and MIR156a, the two
molecules in mediating intercellular communication [15].
plant miRNAs with the highest levels in the sera of Chi-
The novel and important functions of the secreted miR-
nese subjects, and MIR166a, a plant miRNA with modest
NAs were also reported by many other groups [18-21].
level, were assessed by a stem-loop quantitative reverse
The identification of circulating miRNAs, mainly deliv-
transcription polymerase chain reaction (qRT-PCR) as-
ered by cell-secreted MVs, as stable and active signaling
say. MIR161, whose expression level was undetectable,
molecules opens a new field of research in intercellular
served as a negative control. First, the dynamic range
and interorganelle signal transduction.
and sensitivity of the qRT-PCR assay for measuring plant
In the present study, we were surprised to detect ex-
miRNAs in serum were evaluated. A series of synthetic
ogenous plant miRNAs in the serum and plasma of hu-
plant miRNA oligonucleotides with known concentra-
man and animals. Over half of plant miRNAs detected
tions were reverse transcribed and amplified to generate
in serum and plasma are present in MVs. Further
in vitro
a standard curve. As shown in Supplementary informa-
and
in vivo analysis demonstrated for the first time that
tion, Figure S1G, these plant miRNAs had a linear semi-
food-derived exogenous plant MIR168a can pass through
logarithmic plot in a range from 1 fM to 104 fM. In other
the mouse gastrointestinal (GI) track and enter the cir-
words, plant miRNAs at a level as low as 1 fM can be
culation and various organs especially the liver where
efficiently examined by this assay. The absolute concen-
it cross-kingdomly regulates mouse LDLRAP1 protein
tration of each miRNA was then calculated according
expression and physiological condition.
to the standard curve. As can be seen in Figure 1B, the
tested plant miRNAs were clearly present in sera from
humans, mice, and calves. Moreover, when compared
to the endogenous mammalian miRNAs known to be
Plant miRNAs are present in human and animal sera and
stably present in animal serum [5, 15], these plant miR-
NAs were relatively lower, but in a similar concentration
Upon investigation of the global miRNA expres-
range. The levels of plant miRNAs in various serum
sion profile in human serum, we found that exogenous
samples were about one-tenth of that of endogenous
plant miRNAs were consistently present in the serum of
miR-16 (Figure 1B, insert), one of the major endogenous
healthy Chinese men and women. As shown in Figure
miRNAs in animal serum [5, 6]. Furthermore, the pres-
1A and Supplementary information, Table S1, Solexa se-
ence of plant miRNAs in human and animal serum was
quencing revealed 30 known plant miRNAs in Chinese
confirmed by semi-quantitative RT-PCR and northern
healthy donors, among which MIR156a and MIR168a
blotting (Figure 1C and 1D). To determine whether the
showed considerable levels of expression (the mean Sol-
MIR156a, MIR168a, and MIR166a identified in serum
exa reads/total mammalian miRNAs > 0.005). A compar-
are genuine plant miRNAs, we treated the total small
ison of the Solexa reads of plant miRNAs between male
RNA isolated from human serum with sodium perio-
and female sera revealed no significant difference for the
date (oxidizing agent). Plant miRNAs are 2′-O-methyl
majority of plant miRNAs. As shown in the Pearson's
modified on their terminal nucleotide, which renders
correlation scatter plot comparing plant miRNAs in male
them resistant to periodate [22]. In contrast, mammalian
and female sera, the Pearson's correlation coefficient
miRNAs with free 2′ and 3′ hydroxyls are sensitive to
(
R)
was close to 1 (
R = 0.9002; Supplementary informa-
periodate. Since oxidized terminal nucleotides can not
tion, Figure S1A). These healthy Chinese women and
be ligated to the cloning adapter, a comparison of deep
men had no metabolic dysfunctions, and their main diet
sequencing libraries before and after oxidation allows
Cell Research Vol 22 No 1 January 2012
Lin Zhang
et al. npg
www.cell-research.com Cell Research
npg Plant MIR168a downregulates mammalian LDLRAP1110
Figure 1 Plant miRNAs are present in human and animal sera and organs.
(A) The levels (Solexa reads) of 10 plant miRNAs
detected by Solexa sequencing in sera from healthy Chinese men and women, and eight pooled samples (each pooled from
10 healthy Chinese subjects). For normalization, the sequencing frequency of each plant miRNA was normalized to the total
amount of mammalian miRNAs.
(B) The absolute levels of plant miRNAs in the sera of various mammals detected by qRT-
PCR (
n = 6). Endogenous animal miRNAs, miR-16, and miR-25 serve as controls (insert).
(C) Semi-quantitative RT-PCR
analysis of the indicated miRNAs in the serum from human, mouse, rat, calf, horse, and sheep. Accurate amplification of
each miRNA was confirmed by Sanger-based method to sequence the PCR products.
(D) Northern blotting analysis of the
expression levels of MIR168a, MIR156a, and miR-16 in human serum (100 ml) and calf serum (40 ml). Synthetic MIR168a,
MIR156a, and miR-16 (1 pmol) served as positive controls.
(E) Equal amount of total small RNAs (each from 100 ml of hu-
man serum) were treated with/without sodium periodate. After the reactions, the RNAs were purified and then subjected to
Solexa sequencing. Solexa reads of the plant miRNAs in oxidized and unoxidized groups were compared. Total and indi-
vidual mammalian miRNAs were compared to serve as controls (insert). The absolute Solexa reads of miRNAs are indicated.
(F) Equal amount of synthetic plant miRNAs (without 2′-O-methylated 3′ ends) and total small RNAs isolated from rice and
human serum were treated with/without sodium periodate. After the reactions, the RNAs were subjected to qRT-PCR with
the miScript PCR system. Synthetic miR-16 and total small RNAs isolated from mouse liver and human serum were treated
as above. MiR-16 expression levels with/without oxidation were compared to serve as controls (insert).
(G) The levels of
plant miRNAs detected by qRT-PCR in MVs isolated from C57BL/6J mouse plasma (
n = 4).
(H) The levels of plant miRNAs
detected by Solexa sequencing in various organs of C57BL/6J mice.
(I) The levels of plant miRNAs detected by qRT-PCR
in various organs of C57BL/6J mice (normalized to U6;
n = 6). As before, miR-16 and miR-25 serve as controls (insert).
(J,
K) The levels of plant miRNAs detected by Solexa sequencing
(J) and qRT-PCR
(K) in mouse liver after oxidation. Similarly,
mammalian miRNAs serve as controls (insert). The absolute Solexa reads of miRNAs are indicated.
assessing whether the small RNAs tested bear 2′-O-
In contrast, MIR156a, MIR168a, and MIR166a in human
methylated 3′ ends [23, 24]. Indeed, as shown in Figure
serum remained unchanged with this treatment and were
1E, most mammalian miRNAs in human serum, such
sequenced by Solexa, suggesting that they bear 2′-O-
as miR-423-5p, miR-320a, miR-483-5p, miR-16, and
methylated 3′ ends and are therefore genuine plant miR-
miR-221, had an unmodified 2′, 3′ hydroxyls and were
NAs. Moreover, the methylation status of small RNAs
therefore oxidized and failed to be sequenced by Solexa.
can be evaluated by qRT-PCR with miScript PCR system
Cell Research Vol 22 No 1 January 2012
Lin Zhang
et al. npg
after treating the small RNAs with sodium periodate.
and 0.66 fmol/g (miRNA level/weight of chow diet),
This method blocks ligation of a poly(A) tail to small
respectively. However, the levels of these three miRNAs
RNAs bearing 2′,3′ hydroxy termini, preventing them
were 6-, 10- and 3-fold higher, respectively, in fresh
from being reverse transcribed and amplified. Consistent
rice than in chow diet (Figure 2A). In contrast, mammal
with the results of oxidized deep sequencing, MIR156a
miRNAs were expressed in chow diet but were undetect-
and MIR168a in rice and serum showed high levels of
able in fresh rice (Figure 2A, insert). It is worth noting
resistance to periodate and could be amplified, whereas
that these three plant miRNAs, MIR168a, MIR156a, and
miR-16 in liver and serum was oxidized and unable to be
MIR166a, were detected in other foods, including Chi-
detected (Figure 1F). We next determined the portion of
nese cabbage (
Brassica rapa pekinensis), wheat (
Triticum
circulating plant miRNAs in MVs compared to MV-free
aestivum), and potato (
Solanum tuberosum) (Supplemen-
plasma. Similar to most endogenous miRNAs, such as
tary information, Table S3). Interestingly, plant miRNAs
miR-16, miR-21, and miR-150 (Figure 1G, insert), plant
were stable in cooked foods (Supplementary information,
MIR168a and MIR156a were primarily detected in MVs
Table S3). The levels of MIR168a and MIR156a were
in C57BL/6J mouse plasma (Figure 1G). Further study
further assessed in the sera and tissues of mice fed with
revealed that plant MIR168a and MIR156a were detected
either a chow diet or fresh rice following 12 h of fasting.
in various mouse tissues, including liver, small intestine,
As shown in Figure 2B and 2C and Supplementary in-
and lung (Figure 1H and 1I). Interestingly, MIR166a was
formation, Figure S2C, the levels of plant MIR168a and
nearly undetectable in various mouse tissues, although it
MIR156a were not elevated in the serum or liver at 0.5, 3,
was present in the serum. In these studies, the levels of
and 6 h after chow diet feeding. In contrast, the levels of
miRNAs were normalized to U6 snRNA and other small
these two miRNAs were significantly increased in both
RNAs such as snoRNA146 and snoRNA251. As shown
the sera and the livers of mice fed with fresh rice for 6 h
in Supplementary information, Figure S2A, the expres-
(Figure 2B and 2C and Supplementary information, Fig-
sion levels of U6 and small RNAs were approximately
ure S2C). We further compared the levels of MIR168a
equivalent between different mouse tissues. Moreover,
in perfused and non-perfused organs after rice feeding
no plant pre-miRNA was detected by semi-quantitative
and found no difference (Supplementary information,
RT-PCR in human and animal sera and tissues (Supple-
Figure S2D), which indicates that the elevation of plant
mentary information, Figure S2B). The presence of
miRNAs in mouse organs was not due to contamination
genuine plant miRNAs in mouse tissues was further
by blood cells. In agreement with the hypothesis that
demonstrated by oxidized deep sequencing and validated
exogenous plant miRNAs are derived from food intake,
by qRT-PCR with miScript PCR system. As shown in
the levels of MIR168a were increased in the stomach and
Figure 1J and 1K, mammalian miRNAs in mouse liver
small intestine, but not in the kidney, of mice following
were oxidized and undetectable by both Solexa sequenc-
feeding with a chow diet or fresh rice (Supplementary
ing and qRT-PCR assay with miScript PCR system. In
information, Figure S2E-S2G). Furthermore, when mice
contrast, MIR168a and MIR156a in mouse liver were
were gavage fed with total RNA extracted from fresh
resistant to oxidation by periodate.
rice, the levels of MIR168a in mouse serum and liver
were elevated 3 h after feeding (Figure 2D and 2E). Ad-
The exogenous mature plant miRNAs in food can pass
ditionally, mice were gavage fed with synthetic MIR168a
through mouse GI tract and enter the sera and organs
(single-stranded mature MIR168a) or synthetic methy-
Next, we sought to identify the source of these exog-
lated MIR168a because it has been reported that plant
enous plant miRNAs in the sera and organs of mammals.
miRNAs are methylated [27]. The levels of MIR168a in
Given that plant MIR168a and MIR156a have been
mouse serum and liver were elevated 3 h after feeding
reported to be enriched in various plants, including rice
(Figure 2F and 2G). To identify the form in which MI-
(
Oryza sativa) and crucifers (
Brassicaceae) [25, 26],
R168a in a food such as rice was taken up by C57BL/6J
and that rice is the main stable food in China, we hy-
mice, mice were gavage fed with various forms of MI-
pothesized that plant miRNAs in the sera and tissues of
R168a, including double-stranded miRNA (dsMIR168a),
humans and animals are primarily a result of food intake.
precursor miRNA (pre-MIR168a), single-stranded DNA
To test this hypothesis, the expression levels and stabil-
miRNA (ssDNA-MIR168a), and DNA precursor miRNA
ity of plant miRNAs in various foods were investigated.
(pre-DNA-MIR168a), and the level of each form of MI-
As shown in Supplementary information, Table S3,
R168a in mouse serum was assessed after 6 h. Interest-
plant MIR168a, MIR156a, and MIR166a were detected
ingly, we only detected single-stranded mature MIR168a
in chow diet (a mouse diet with a relatively low fat and
by qRT-PCR (Supplementary information, Figure S2H-
cholesterol content) in the concentrations of 0.43, 0.54
S2K), strongly suggesting that food-derived MIR168a is
www.cell-research.com Cell Research
npg Plant MIR168a downregulates mammalian LDLRAP1112
Figure 2 The exogenous mature plant miRNAs in food can pass through gastrointestinal (GI) tract and enter the sera and or-
gans.
(A) The levels of MIR168a, MIR156a, and MIR166a detected by qRT-PCR in fresh rice and chow diet (
n = 6). Two en-
dogenous animal miRNAs, miR-16, and miR-150, served as controls (insert). UD, undetectable.
(B,
C) The levels of MIR168a
in mouse serum
(B) and liver
(C) after feeding with fresh rice or chow diet for 0.5 h, 3 h, or 6 h (
n = 8). The control group (named
0 h) was euthanized after a 12-h of fasting.
(D,
E) The levels of MIR168a in mouse serum
(D) and liver
(E) following gavage
feeding with RNA extracted from fresh rice (
n = 8). After 0.5 h, 3 h, or 6 h, MIR168a levels were detected by qRT-PCR. The
control group (0 h) was euthanized after a 12-h of fasting.
(F,
G) The levels of MIR168a in mouse serum
(F) and liver
(G) fol-
lowing gavage feeding with synthetic MIR168a and synthetic methylated MIR168a (
n = 8). The control group was gavage fed
with ncRNA. *
P < 0.05; **
P < 0.01.
taken up in its mature form. To mimic GI tract environ-
4.0-fold induction of secreted endogenous monocytic
ment, the effect of acidification on the stability of plant
miR-150 in the plasma of patients with atherosclerosis
miRNAs and mammalian miRNAs was examined. Total
could sufficiently decrease the protein level of its target
RNA isolated from rice or mouse liver was adjusted to
gene, c-Myb, and increase cell migration in recipient en-
pH 2.0 and kept at 37 °C for several hours. As shown in
dothelial cells [15]. Given that structurally functional ex-
Supplementary information, Figure S2L, acidification did
ogenous plant miRNAs can enter human and animal sera
not significantly affect the yield and quality of miRNAs.
and tissues through food intake and can be maintained
The majority of plant miRNAs and mammalian miRNAs
at elevated levels, we hypothesized that they may play a
can survive under acidic condition for at least 6 h. Inter-
role in regulating the functions of mammalian cells and
estingly, the degradation rate of mammalian miRNAs un-
organs. Because most plant miRNAs can act like RNA
der acidic condition was similar to that of their synthetic
interference (RNAi), which requires a high degree of
form (without 2′-O-methylated 3′ ends), whereas plant
complementarity between miRNAs and target RNAs [28],
miRNAs had a much slower degradation rate compared
we performed bioinformatic analysis to identify any se-
with their synthetic form (without 2′-O-methylated 3′
quences in the human, mouse, or rat genome with perfect
ends), suggesting that methylation had a protective effect
or near-perfect match to MIR168a.
Approximately 50
on the stability of plant miRNAs.
putative target genes were identified as the target genes
of MIR168a (Supplementary information, Table S4).
Plant MIR168a binds to exon 4 of mammalian LDLRAP1
The most highly conserved sequence of a putative bind-
and decreases LDLRAP1 protein level in vitro
ing site among various species is located in exon 4 of
In a previous study, it was demonstrated that a 2.5-
the low-density lipoprotein receptor adapter protein 1
Cell Research Vol 22 No 1 January 2012
Lin Zhang
et al. npg
(LDLRAP1) (Figure 3A). We calculated the minimum
phenomenon, the LDLRAP1 ORF was GFP tagged at
free energy of binding to be −29.2 kcal/mol. LDLRAP1
its carboxyl terminus (Figure 3L). Compared to cells
is a liver-enriched gene that plays a critical role in facili-
transfected with ncRNA, the number of cells expressing
tating the removal of LDL from the circulatory system
GFP-tagged LDLRAP1 was drastically decreased in cells
[29]. As shown in Figure 3B, transfection of HepG2
transfected with pre-MIR168a (Figure 3M and 3N).
cells (a hepatocyte carcinoma cell line) with MIR168a
Although overexpression of pre-miRNA is the con-
precursor (pre-MIR168a) resulted in a 1 000-fold eleva-
ventional method to assess the function of a miRNA, we
tion in MIR168a, suggesting that plant pre-MIR168a
determined the direct effect of a single-stranded mature
can be properly processed in mammalian HepG2 cells.
plant MIR168a on LDLRAP1 expression in HepG2 cells
Additionally, the LDLRAP1 protein level in these cells
for two reasons. First, neither plant pri-miRNA nor plant
was significantly reduced (Figure 3C and 3D), whereas
pre-miRNA has been detected in the sera and tissues
the LDLRAP1 mRNA level was not affected (Figure
of human and animals. Second, and more importantly,
3E and 3F). A luciferase reporter assay was employed
single-stranded mature MIR168a is likely to be the form
to demonstrate the direct binding of MIR168a to exon 4
taken up by animals via the GI tract. By transfecting
of LDLRAP1. The same strategy has been used by oth-
HepG2 cells with equivalent amounts of single-stranded
ers in studying miRNA targeting in coding regions [30-
mature plant MIR168a, we increased the level of mature
32]. In this experiment, the wild-type (WT) or mutant
MIR168a in HepG2 cells by 10 000-fold (Supplemen-
(MUT) MIR168a complementary site (CS), WT or MUT
tary information, Figure 3C) and observed a concomitant
LDLRAP1 binding site (BS), the human LDLRAP1
reduction in LDLRAP1 (Supplementary information,
exon 4, and human LDLRAP1 coding sequence (CDS)
Figure S3D and S3E). Similar to the results from trans-
were cloned into a luciferase reporter plasmid (Figure
fection with pre-MIR168a, the overexpression of single-
3G) and transfected into HepG2 cells combined with
stranded mature MIR168a had no effect on LDLRAP1
pre-MIR168a. As expected, luciferase reporter activity
mRNA levels (Supplementary information, Figure S3F
for WT was significantly reduced following transfection
and S3G). A luciferase reporter assay also demonstrated
with pre-MIR168a, whereas the MUT luciferase reporter
that high levels of mature MIR168a decreased luciferase
was unaffected by transfection with pre-MIR168a (Fig-
activity (Supplementary information, Figure S3H), in
ure 3H). Because the predicted MIR168a binding site is
agreement with the observation that mature MIR168a
not in the 3′ UTR but in the open reading frame (ORF)
could affect LDLRAP1 expression despite a relatively
of LDLRAP1, we further compared the efficiency of our
low affinity for the putative binding site of LDLRAP1.
luciferase reporter assay with those of typical reporters
in which the endogenous miRNAs bind to the 3′ UTR
AGO2-associated mature MIR168a in Caco-2 cel -derived
of target genes. We found that the rate of the inhibition
MVs sufficiently reduces mammalian LDLRAP1 protein
of luciferase activity by MIR168a was dependent on
level in recipient HepG2 cells
the ratio of pre-MIR168a to BS-containing luciferase
In this study, we hypothesized that epithelial cells, par-
reporter (Supplementary information, Figure S3A) and
ticularly in the small intestine, might take up plant miR-
that the efficiency of MIR168a inhibition of LDLRAP1
NAs in food, then package them into MVs and release
expression was similar to the inhibition of target genes
them into the circulatory system. The secreted MVs from
by endogenous miRNAs (miR-16, miR-21, and miR-
small intestinal epithelial cells could then deliver exog-
150; Supplementary information, Figure S3B). To deter-
enous plant miRNA to target cells of other organs and
mine whether MIR168a could directly target LDLRAP1
regulate recipient cell function. To test this hypothesis,
within its CDS, we followed the strategy proposed by
human intestinal epithelial Caco-2 cells were transfected
others [30-32] and created a construct that expressed the
with single-stranded mature MIR168a. As indicated by
full-length ORF of LDLRAP1 (Figure 3I). Furthermore,
Figure 4A, the MVs released by the Caco-2 cells were
we used site-directed mutagenesis to create a variant of
collected and used to treat HepG2 cells. The LDLRAP1
the LDLRAP1 construct in which the MIR168a target
protein level in HepG2 cells was then assessed. As
site was mutated while leaving the LDLRAP1 amino-
shown in Figure 4B, the levels of mature MIR168a were
acid sequence intact (Figure 3I). These constructs were
elevated by 200-fold in MVs released by Caco-2 cells
co-transfected with pre-MIR168a or pre-ncRNA into
transfected with MIR168a and by 100-fold in Caco-2
293T cells. WT LDLRAP1 was strongly downregulated
MV-treated HepG2 cells. Plant MIR168a delivered into
by MIR168a, whereas MIR168a had no effect on the
HepG2 cells via Caco-2 MVs significantly decreased
expression of LDLRAP1 when the MIR168a target site
the LDLRAP1 protein level in the recipient HepG2
was mutated (Figure 3J and 3K). To further explore this
cells (Figure 4C and 4D), while it had no effect on LD-
www.cell-research.com Cell Research
npg Plant MIR168a downregulates mammalian LDLRAP1114
Cell Research Vol 22 No 1 January 2012
Lin Zhang
et al. npg
Figure 3 Plant MIR168a binds to exon 4 of mammalian LDLRAP1 and decreases LDLRAP1 protein level
in vitro.
(A) Sche-
matic description of the hypothesized duplexes formed by interactions between the exon 4 of LDLRAP1 and MIR168a. Paired
bases are indicated by a black oval and G:U pairs are indicated by two dots. The predicted free energy of the hybrid is indi-
cated. Note that the potential binding site of MIR168a to LDLRAP1 mRNA is highly conserved across species.
(B) qRT-PCR
analysis of MIR168a levels in pre-MIR168a-transfected HepG2 cells (
n = 9).
(C) Western blot analysis of LDLRAP1 protein
levels in pre-MIR168a-transfected HepG2 cells.
(D) The quantification of the LDLRAP1 protein expression in
C (
n = 9).
(E,
F)
Semi-quantitative RT-PCR
(E) and qRT-PCR
(F) analysis of LDLRAP1 mRNA levels in pre-MIR168a-transfected HepG2 cells (
n
= 5).
(G) Diagram of the luciferase reporter plasmid carrying the firefly luciferase-coding sequence attached with the wild-type
(WT) or mutant (MUT) MIR168a complementary site (CS), WT, or MUT LDLRAP1 BS, LDLRAP1 exon 4, and the LDLRAP1
CDS.
(H) Luciferase activities in HepG2 cells co-transfected with luciferase reporters described in G and pre-MIR168a or pre-
ncRNA (
n = 9).
(I) Constructs for the expression of the WT or MUT ORFs of LDLRAP1.
(J) LDLRAP1 level in 293T cells co-
transfected with WT or MUT LDLRAP1 ORFs and pre-MIR168a or pre-ncRNA.
(K) The quantification of the LDLRAP1 protein
expression in
J (
n = 3).
(L) The LDLRAP1 ORF tagged with GFP at its carboxyl terminus.
(M) 293T cells were co-transfected
with the GFP-tagged LDLRAP1 ORF (top) and pre-MIR168a or pre-ncRNA. GFP-positive cells were analyzed by fluores-
cence microscopy at 24 h of post-transfection.
(N) The percentage of GFP-positive cells in
M (
n = 6). *
P < 0.05; **
P < 0.01.
LRAP1 mRNA levels (Figure 4E and 4F). Interestingly,
(Figure 4M and 4N). The association of MIR168a with
repression of LDLRAP1 protein levels in HepG2 cells
AGO2 was additionally detected in HepG2 cells directly
by Caco-2 MVs was dose dependent, as Caco-2 MVs
transfected with pre-MIR168a or mature MIR168a (Sup-
containing more MIR168a decreased the expression of
plementary information, Figure S3I and S3J). In separate
LDLRAP1 more effectively (Figure 4G-4I). A luciferase
experiments, we tested whether plant miRNAs can be
assay further demonstrated that plant MIR168a secreted
processed and packaged into secreted MVs by cell types
by Caco-2 cells could effectively enter HepG2 cells
other than colonic epithelial Caco-2 cells. As can be
and recognize exon 4 of the LDLRAP1 transcripts. WT
seen in Supplementary information, Figure S4, similar to
luciferase reporter activity was significantly reduced fol-
Caco-2 cells, 293T cells were able to package MIR168a
lowing incubation with MVs derived from Caco-2 cells
into MVs and then deliver MIR168a into HepG2 cells.
transfected with mature MIR168a, whereas the MUT
luciferase reporter was unaffected (Figure 4J). In 293T
Anti-MIR168a ASO reverses rice feeding-induced reduc-
cells transfected with WT LDLRAP1 ORF, treatment
tion of mouse liver LDLRAP1 protein at 6 h feeding
with MIR168a-containing Caco-2 MVs but not ncRNA-
In order to determine the specificity of the inhibitory
containing Caco-2 MVs significantly reduced the level of
effect of MIR168a on LDLRAP1 and exclude the possi-
LDLRAP1 expression (Figure 4K and 4L). In contrast,
bility that the reduction of LDLRAP1 protein was caused
MIR168a-containing Caco-2 MVs had no effect on the
by factors other than MIR168a, an anti-MIR168a anti-
expression of the MUT LDLRAP1 ORF (Figure 4K and
sense oligonucleotide (ASO) was injected intravenously
4L). These results clearly indicate a possible mechanism
into mice during rice feeding. The injection of anti-
for the action of exogenous plant miRNAs in an animal
MIR168a ASO not only significantly reduced MIR168a
model. Moreover, because Argonaute2 (AGO2) is present
levels in the livers of fresh rice-fed mice (Figure 5A),
in MVs and facilitates miRNA binding to its target gene
but also remarkably reversed the reduction of mouse
via the RNA-induced silencing complex (RISC) [33], we
liver LDLRAP1 by fresh rice-derived MIR168a (Figure
performed an immunoprecipitation experiment to assess
5B and 5C). Taken together, these results indicate that
whether plant MIR168a was associated with mammalian
exogenous plant miRNAs are able to enter the serum
AGO2 and LDLRAP1. As can be seen in Figure 4M-4O,
and organs of mammals via food intake, and that plant
both MIR168a (Figure 4N) and LDLRAP1 mRNA (Fig-
MIR168a can bind to the nucleotide sequence located in
ure 4O) were detected in the product precipitated by an
exon 4 of mammalian LDLRAP1, leading to the inhibi-
anti-AGO2 antibody from the HepG2 cells treated with
tion of LDLRAP1 expression
in vivo.
Caco-2 MVs, while only MIR168a (Figure 4M) but not
LDLRAP1 mRNA (Figure 4O) was detected in the anti-
Exogenous MIR168a inhibits mouse liver LDLRAP1 ex-
AGO2 antibody-precipitated product from the Caco-2
pression and elevates plasma LDL-cholesterol level at 3
MVs. As a control, endogenous mammalian miR-16 was
days after food intake
detected with the anti-AGO2 antibody-precipitated prod-
LDL is the major cholesterol-carrying lipoprotein of
uct from both Caco-2 MVs and MV-treated HepG2 cells,
human plasma and plays an essential role in the patho-
but its level was not affected by MIR168a transfection
genesis of atherosclerosis [34]. Downregulation of
www.cell-research.com Cell Research
npg Plant MIR168a downregulates mammalian LDLRAP1116
Cell Research Vol 22 No 1 January 2012
Lin Zhang
et al. npg
Figure 4 AGO2-associated mature MIR168a in Caco-2 cell-derived MVs sufficiently reduces mammalian LDLRAP1 protein
level in recipient HepG2 cells.
(A) A flow chart of the experimental design.
(B) The elevation of MIR168a in Caco-2 MVs after
transfection with mature MIR168a (left) and in HepG2 cells after treatment with Caco-2 MVs (right;
n = 9). Caco-2 MVs were
harvested after transfecting the cells with mature MIR168a or ncRNA.
(C) Western blot analysis of LDLRAP1 protein lev-
els in HepG2 cells treated with or without Caco-2 MVs. Caco-2 MVs were harvested after transfecting the cells with mature
MIR168a or ncRNA.
(D) The quantification of the LDLRAP1 protein expression in
C (
n = 6).
(E,
F) Semi-quantitative RT-PCR
(E) and qRT-PCR
(F) analysis of the LDLRAP1 mRNA levels in HepG2 cells treated with or without Caco-2 MVs (
n = 5).
(G,
H) The levels of MIR168a
(G) and LDLRAP1 protein
(H) in HepG2 cells treated with Caco-2 MVs derived from Caco-2 cells
transfected with different doses (10, 50, or 100 pmol/105 cells) of mature MIR168a or ncRNA.
(I) The quantification of the
LDLRAP1 level in
H (
n = 3).
(J) The luciferase activities in luciferase reporter-transfected HepG2 cells treated with or without
Caco-2 MVs (
n = 9).
(K) LDLRAP1 protein level in 293T cells transfected with WT or MUT LDLRAP1 ORF and then treated
with Caco-2 MVs.
(L) The quantification of the LDLRAP1 protein expression in
K (
n = 3).
(M,
N) The association of MIR168a
with AGO2 in Caco-2 MVs
(M) and HepG2 cells treated with Caco-2 MVs
(N). The levels of MIR168a and miR-16 (control) in
anti-AGO2 immunoprecipitated products detected by qRT-PCR (
n = 9).
(O) The association of LDLRAP1 mRNA with AGO2
in Caco-2 MV-treated HepG2 cells or Caco-2 MVs. The LDLRAP1 mRNA in anti-AGO2 immunoprecipitated products from
HepG2 cells (lanes 1 and 2) and Caco-2 MVs (lanes 3 and 4) was detected by semi-quantitative RT-PCR with 25-30 cycles.
*
P < 0.05; **
P < 0.01.
LDLRAP1 in the liver causes decreased endocytosis of
the chow diet group (Figure 6C). Likewise, levels of
LDL by liver cells and impairs the removal of LDL from
MIR168a in the liver were increased in fresh rice-fed
plasma [35, 36]. To assess the physiological function
mice compared to chow diet-fed mice after 1 day (Figure
of food-enriched MIR168a in mammals, mice were fed
6D). Supplementary information, Figure S5A to S5C
with fresh rice and chow diets for 7 days after a 12-h of
shows the level of MIR168a in other mouse organs after
fasting. Body weight was not different between the two
a long period of feeding. Although miR168a was ini-
groups of mice during the experimental period (Figure
tially increased in the stomach of mice fed with rice, no
6A), although the fresh rice-fed group had an increased
significant difference in MIR168a levels in the stomach,
food intake (Figure 6B). In addition, the serum MIR168a
intestine, or kidney were found between chow diet- and
levels of the chow diet-fed group were not significantly
rice-fed mice. Concomitant with a significant elevation in
altered during the experimental period (Figure 6C).
MIR168a levels in the livers of mice after 1 day of fresh
In contrast, there was a significant induction of serum
rice feeding (Figure 6D), LDLRAP1 expression dramati-
MIR168a levels in mice after 1 day of feeding with fresh
cally decreased in the group of fresh rice-fed mice (Figure
rice, and these levels remained elevated over those of
6E and 6F). In these experiments, LDLRAP1 levels in
Figure 5 Anti-MIR168a ASO reverses rice feeding-induced reduction of mouse liver LDLRAP1 protein at 6 h feeding.
(A,
B)
The levels of MIR168a
(A) and LDLRAP1 protein
(B) in mouse liver after feeding with chow diet, fresh rice, or fresh rice ac-
companying an intravenous injection of anti-MIR168a ASO or anti-ncRNA for 6 h (
n = 8).
(C) The quantification of the LDL-
RAP1 level in
B (
n = 8). *
P < 0.05; **
P < 0.01.
www.cell-research.com Cell Research
npg Plant MIR168a downregulates mammalian LDLRAP1118
Cell Research Vol 22 No 1 January 2012
Lin Zhang
et al. npg
Figure 6 Exogenous MIR168a inhibits mouse liver LDLRAP1 expression and elevates plasma LDL-cholesterol level at 3
days after food intake.
(A) The weight changes of mice fed with chow diet or fresh rice (
n = 8).
(B) The food intake of chow
diet or fresh rice (
n = 8).
(C,
D) The levels of MIR168a in mouse sera
(C) and livers
(D) after chow diet or fresh rice feeding (
n
= 8).
(E) The LDLRAP1 protein levels in mouse livers after chow diet or fresh rice feeding.
(F) The quantification of LDLRAP1
protein expression in
E (
n = 8).
(G) The levels of LDL-cholesterol in mouse plasma after chow diet or fresh rice feeding (
n = 8).
(H,
I) The levels of MIR168a
(H) and LDLRAP1 protein
(I) in mouse livers after 3 days of feeding with chow diet, fresh rice,
or fresh rice accompanying an injection of anti-MIR168a ASO or anti-ncRNA (
n = 8).
(J) The quantification of LDLRAP1 pro-
tein expression in
I (
n = 8).
(K) The levels of LDL-cholesterol in mouse plasma after 3 days of feeding with chow diet, fresh
rice, or fresh rice accompanying an injection of anti-MIR168a ASO or anti-ncRNA (
n = 8).
(L) The association of MIR168a
with AGO2 in mouse livers after 3 days of feeding with chow diet, fresh rice, or fresh rice plus injection of anti-MIR168a ASO
or anti-ncRNA. The levels of MIR168a and miR-16 (control) in anti-AGO2 immunoprecipitated products were detected by
qRT-PCR.
(M) The association of LDLRAP1 mRNA with AGO2 in mouse livers after 3-day feeding. The LDLRAP1 mRNA in
anti-AGO2 immunoprecipitated products was detected by semi-quantitative RT-PCR.
(N,
O) The levels of MIR168a
(N) and
LDLRAP1 protein
(O) in mouse livers after 3 days of feeding with chow diet and mature MIR168a or ncRNA (
n = 8).
(P) The
quantification of LDLRAP1 protein expression in
O (
n = 8).
(Q) The levels of LDL-cholesterol in mouse plasma after 3 days of
feeding with chow diet and mature MIR168a or ncRNA (
n = 8). *
P < 0.05; **
P < 0.01.
the control group (0 d) were obtained from mice that had
creased compared to those in chow diet-fed mice and this
undergone a 12-h period of fasting. It has been found
rice feeding-induced elevation of liver AGO2-associated
that fasting decreases LDLRAP1 expression in mouse
MIR168a and LDLRAP1 mRNA could be blocked by
livers, and re-feeding was sufficient to rescue this reduc-
administration of anti-MIR168a ASO. Furthermore, by
tion (Supplementary information, Figure S5D and S5E).
adding mature MIR168a into chow diet and feeding
In agreement with the reduction of LDLRAP1 in the
mice, we found that the MIR168a levels in chow diet
mouse liver, LDL levels in mouse plasma were signifi-
(Supplementary information, Figure S5F), sera (Supple-
cantly elevated on days 3 and 7 after fresh rice feeding
mentary information, Figure S5G) and mouse liver (Fig-
(Figure 6G). The rice-induced elevation of mouse liver
ure 6N) were increased but the LDLRAP1 protein level
MIR168a (Figure 6H) and, more importantly, MIR168a-
in the mouse liver (Figure 6O and 6P) was decreased.
mediated reduction of liver LDLRAP1 (Figure 6I and
In contrast, there is no difference of MIR156a levels
6J) was largely blocked by the administration of anti-
between the mice after chow diet feeding with addition
MIR168a ASO but not negative control oligonucleotides.
of MIR168a or ncRNA (Supplementary information,
Consistently, the rice-induced elevation of LDL levels in
Figure S5G). Additionally, decreased liver LDLRAP1
mouse plasma was blocked by the anti-MIR168a ASO
levels led to an elevation in the mouse plasma LDL level
(Figure 6K). Furthermore, the association of MIR168a
(Figure 6Q). In separate experiments, we tested the link
with LDLRAP1 mRNA through AGO2 in mouse liver
between the downregulation of mouse liver LDLRAP1
was tested. For this experiment, AGO2 protein in mouse
and the elevation of mouse plasma LDL level by directly
liver following different treatment was immunoprecipi-
decreasing liver LDLRAP1 by administration of LDL-
tated by anti-AGO2 antibody and the levels of MIR168a
RAP1 siRNA. Downregulation of liver LDLRAP1 via
(Figure 6L) and LDLRAP1 mRNA (Figure 6M) in anti-
LDLRAP1 siRNA (Supplementary information, Figure
AGO2 antibody IP product were assessed. As can be
S6A and S6B) resulted in the elevation of plasma LDL
seen, in rice-fed mice, AGO2-associated MIR168a and
level (Supplementary information, Figure S6C), sug-
LDLRAP1 mRNA in mouse livers were significantly in-
gesting that LDLRAP1 is responsible for plasma LDL
www.cell-research.com Cell Research
npg Plant MIR168a downregulates mammalian LDLRAP1120
clearance. However, the level of liver LDLRAP1 was not
did not significantly affect the stability of plant miR-
related to the levels of plasma cholesterol or triglycer-
NAs, and that methylation may have a protective effect
ides (Supplementary information, Figure S6D and S6E).
on plant miRNAs (Supplementary information, Figure
Decreased levels of plasma cholesterol (Supplementary
S2L). Interestingly, pre-MIR168a and pre-MIR156a can
information, Figure S7A) but unchanged plasma ApoA
be detected in fresh rice but not in either cooked rice
(Supplementary information, Figure S7B) and triglycer-
or mouse serum (Supplementary information, Figure
ide levels (Supplementary information, Figure S7C) fur-
S2B), implicating that these pre-miRNAs may not be
ther supported the hypothesis that the elevation of fresh
stable and able to pass through GI tract. In support of
rice-derived MIR168a in the mouse liver specifically
this, the gavage feeding experiment using equal amount
decreased liver LDLRAP1 expression and thus caused
of single-stranded miRNA (ssRNA), double-stranded
an elevated LDL level in mouse plasma. Interestingly,
miRNA (dsRNA), precursor miRNA (preRNA), single-
no significant differences in the levels of plasma cho-
stranded DNA form of miRNA (ssDNA), and DNA form
lesterol (Supplementary information, Figure S7D) and
of precursor miRNA (preDNA) clearly showed that only
triglycerides (Supplementary information, Figure S7E)
mature miRNA was detected in mouse serum and tis-
were observed in mice after 3 days of feeding with chow
sues while other forms of RNA/DNA were not detected
diet, fresh rice, or fresh rice accompanying an injection
(probably degraded; Supplementary information, Figure
of anti-MIR168a or anti-ncRNA oligonucleotides. As a
S2H-S2K). The exclusive presence of mature MIR168a
control, mature mammalian miR-150 was added into the
in mouse serum and tissues maybe largely dependent on
chow diet to feed mice. In a similar fashion, miR-150
its high stability compared to other forms of RNA/DNA
levels in mouse liver (Supplementary information, Fig-
in mouse GI to serum uptake pathway, although it cannot
ure S7F) and serum (Supplementary information, Figure
rule out the possibility that there is a specific pathway for
S7G) were significantly increased after 3 days of feed-
mature miRNA uptake. Our results clearly demonstrate
ing, leading to downregulation of liver c-Myb expression
that exogenous plant mature miRNAs in food can pass
(Supplementary information, Figure S7H).
through the GI tract and be transferred into the blood-
stream and tissues. To our knowledge, this is the first re-
port indicating that nucleotides with complete functional
structure are resistant to digestion in the GI tract and can
Mature single-stranded plant miRNAs are stable
be delivered to other tissues.
Our previous study demonstrated that mature single-
stranded mammalian miRNAs are stably present in
Plant miRNAs execute their function in mammalian cells
serum or plasma of human and other animals [5] and
in a fashion of mammalian miRNA
that the circulating miRNAs can serve both as a class
Generally, mammalian miRNAs execute its func-
of biomarkers of diseases [8, 12] and as active signal
tion by base pairing to the complementary sites in the
molecules [15]. In the present study, we have detected
3′ UTR of its target genes, thus blocking the translation
stable mature single-stranded plant miRNAs in serum
or triggering the degradation of the target mRNAs [1,
and tissues of mammals using both Solexa sequencing
2]. There are only a few reports showing that mamma-
and qRT-PCR (Figure 1A, 1B, 1H and 1I). By perform-
lian miRNA can also bind to exon of the target genes
ing Solexa sequencing and qRT-PCR after oxidation of
[30-32]. Unlike mammalian miRNAs, it is well known
small RNAs with periodate, we have found that the plant
that plant miRNAs act through RNAi, which requires a
miRNAs identified in serum and tissues showed high
complete match of their nucleotide sequence with that of
levels of resistance to periodate, demonstrating that they
their target genes [28]. Plant MIR168 has been reported
bear 2′-O-methylated 3′ ends and are therefore genu-
to be expressed widely in various species of plant [25,
ine plant miRNAs (Figure 1E, 1F, 1J and 1K). Given
26]. In plants, expression of ARGONAUTE1 (AGO1),
the fact that plant miRNAs are completely exogenous,
the catalytic subunit of the RISC responsible for post-
plant miRNAs detected in serum and organs of mam-
transcriptional gene silencing, is controlled through a
mals should only come from food intake. Food-derived
feedback loop involving the MIR168 [37-39]. Here, our
mature plant miRNAs such as MIR168a, which can be
data showed that MIR168a decreased the LDLRAP1
detected even after cooking the food (Supplementary
protein level but did not affect the mRNA level (Figure
information, Table S3), are quite stable in animal serum
3C-3F), suggesting that plant MIR168a regulates mam-
(Figure 1A-1D), suggesting that they may be also resis-
malian LDLRAP1 translation in a fashion of mammalian
tant to enzymatic digest in GI. Indeed, we have shown
functional miRNA. Results of luciferase reporter assays
that acidification mimicking the GI tract environment
using multiple plasmids including the plasmid contain-
Cell Research Vol 22 No 1 January 2012
Lin Zhang
et al. npg
ing partial or whole MIR168a complementary site,
results of association of MIR168a with AGO2 in MVs,
LDLRAP1 BS, human LDLRAP1 exon 4, and human
as well as the association of AGO2 with MIR168a and
LDLRAP1 CDS (Figure 3G and 3H) and the results that
LDLRAP mRNA in HepG2 cells treated with Caco-2
MIR168a was also able to target the artificially expressed
cell MVs (Figure 4M-4O) strongly argue that exogenous
LDLRAP1 protein in 293T cells (Figure 3I-3K) strongly
plant miRNAs cross-kindomly regulate mammalian gene
demonstrate that plant MIR168a could bind to its binding
through a MV-mediated pathway. Previous study showed
site located in exon 4 of mammalian LDLRAP1 gene,
that mature miRNA had a low affinity to AGO2 [40],
and then inhibit LDLRAP1 protein expression.
which might provide an explanation why an extreme el-
evation of mature MIR168a via the direct transfection of
AGO2-associated mature MIR168a in MVs is the func-
MIR168a was required to reduce LDLRAP1 expression.
tional form in vivo
Endogenous mammalian miRNAs have a dynamic
Although a very large amount of single-stranded
range of expression, extending from < 1 copy per cell
mature plant MIR168a can affect LDLRAP1 levels in
to > 10 000 copies per cell, and the regulation of target
HepG2 cells (Supplementary information, Figure S3C-
genes is highly dependent on miRNA expression levels
S3H), it is unlikely that such high concentrations of
[41]. It has been shown that miRNAs expressed at low
mature plant miRNAs can be achieved in serum, plasma
levels (< 100 copies per cell) did not significantly repress
and organs of humans or animals via food intake. Our
target-containing transcripts [42], since target encounter
study here suggested that plant MIR168a derived from
occurs by mass action and modestly expressed miR-
food intake might execute its function in mouse through
NAs have a low probability to meet target-containing
a novel pathway. We previously demonstrated that mam-
transcripts. Previous measurements of miRNAs in liver
malian cells could selectively pack miRNAs into MVs
samples revealed that let-7a, miR-16, miR-20, miR-21,
in response to different stimuli and then secrete these
and miR-22 were present at 700, 3 890, 130, 4 450, and
MVs into the circulation of animals or culture medium
310 copies per cell, respectively [43], while miR-181a
[15]. Cell-derived MVs could further efficiently deliver
was expressed at 810 copies per cell in DP thymocytes
miRNAs into the recipient cells where these exogenous
[44] and miR-223 and miR-451 were 828 and 1 972
miRNAs regulate the expression of target genes and the
copies per cell in CD34(+)/CD133(−) cells [45]. It has
biological functions of recipient cells [15]. In this study,
been widely demonstrated that miRNAs expressed at
we hypothesized that intestinal epithelial cells could take
these concentrations are biologically active [46-49]. We
up plant miRNAs in food, then pack them into MVs and
calculated the amount of MIR168a that was detected in
release the miRNA-containing MVs into the circulatory
1 mg of total liver RNA. The amount of MIR168a was
system. These MVs secreted from intestinal epithelial
3.2 × 10−6 fmol (1 920 copies) per 100 pg of total RNA,
cells would then deliver exogenous plant miRNAs into
equivalent to 853 copies per cell. This quantification
other organs and regulate the function of recipient cells.
revealed that MIR168a is expressed in a similar concen-
Supporting this hypothesis, more than half of the plant
tration range in liver cells as compared with endogenous
MIR168a in the serum was found in MVs (Figure 1G).
mammalian miRNAs. Besides the concentration of the
The results also showed that MVs contained much less
miRNAs, the extent of suppression depends on a number
amount of MIR168a but caused stronger effect on LD-
of factors, such as the concentration of AGO2-associated
LRAP1 level than ‘free' MIR168a did (Figure 4B-4D).
miRNAs and the complementarity between the miRNA
The efficiency of MV-contained MIR168a was even
and its target site [41]. We have identified near-perfect
higher than that of transfected pre-MIR168a (Figure
complementarity between MIR168a and LDLRAP1
3C and 3D). These results indicated that MIR168a in
mRNA. More importantly, we have shown that exog-
MVs was more functionally active compared to ‘free'
enous plant MIR168a in recipient mammalian liver cells
MIR168a. The immunoprecipitation experiments in both
is AGO2 associated (Figure 6L and 6M), which repre-
MVs and MV-treated cells clearly showed that MIR168a
sents an enriched fraction of active and functional miR-
was already associated with AGO2 in the MVs derived
NAs. Thus, MIR168a-targeting LDLRAP1 fulfills the
from Caco-2 cells (Figure 4M), from which we may
requirement for a threshold miRNA concentration and
draw two conclusions: (1) MIR168a in MVs is more
the principle for miRNA-target recognition.
functionally active because it has a higher affinity to
AGO2 although the underneath mechanism remains
Food-derived miRNAs may serve as a novel essential nu-
unknown, and (2) plant miRNAs can use mammalian
AGO2 to form AGO2-associated RISC and execute
It has been widely reported that downregulation of
their functions, similar to mammalian miRNAs. The
LDLRAP1 increases plasma LDL level [35, 36]. In the
www.cell-research.com Cell Research
npg Plant MIR168a downregulates mammalian LDLRAP1122
present study, direct reduction of LDLRAP1 in mouse
RAP1
in vitro and
in vivo, the present study significantly
liver by RNAi significantly elevated plasma LDL level
extends our understanding of the role of miRNAs. With
(Supplementary information, Figure S6A-S6C), confirm-
their robust stability and highly conserved sequences,
ing that LDLRAP1 is a gene candidate responsible for
secretory miRNAs can act not only in a cross-species,
plasma LDL removal. Interestingly, an elevated level
but also a cross-kingdom fashion. In this sense, miRNAs
of MIR168a but a decreased LDLRAP1 in mouse liver
may represent a novel class of universal modulators that
were detected after just 6 h of rice feeding (Figures 2C
play an important role in mediating animal-plant interac-
and 5B), indicating that exogenous plant MIR168a from
tions at the molecular level. Like vitamins, minerals and
food intake can quickly change mouse liver LDLRAP1
other essential nutrients derived from food sources, plant
level. Continuous downregulation of mouse liver LD-
miRNAs may serve as a novel functional component of
LRAP1 level by MIR168a through rice feeding (Figure
food and make a critical contribution to maintaining and
6E and 6F) resulted in an elevation of the plasma LDL-
shaping animal body structure and function. Extending
cholesterol level after 3 days (Figure 6G), implicating a
from this concept, the intake of certain plant miRNAs
physiological relevance of food-derived plant MIR168a.
generation after generation through a particular food
Rice feeding-induced reduction of LDLRAP1 protein
source may leave an imprint on the genetic map of the
and elevation of plasma LDL-cholesterol level could
human race. In conclusion, the discovery of plant miR-
be largely reversed by anti-MIR168a ASO (Figure 6I-
NAs and their roles in the biology of mammalian cells
6K), confirming that the rice feeding-mediated physi-
and animal organs represents the first evidence of cross-
ological alteration is specifically due to the targeting of
kingdom transfer of functionally active miRNAs and
mouse liver LDLRAP1 by MIR168a. This conclusion
opens a new avenue to explore miRNA-mediated animal-
is also supported by the observation that chow diet with
plant interactions.
addition of mature MIR168a significantly enhanced the
levels of mouse liver MIR168a (Figure 6N) and plasma
Materials and Methods
LDL-cholesterol (Figure 6Q) but decreased mouse liver
LDLRAP1 protein level (Figure 6O and 6P). Interest-
Reagents, cells and antibodies
ingly, food intake, possibly via intestinal epithelia of GI
The human intestinal carcinoma cell line Caco-2 and the hepa-
track, may represent a general pathway for uptake of
toma cell line HepG2 were purchased from Institute of Biochemis-
food-derived or food-associated miRNAs. As shown in
try and Cell Biology, Shanghai Institutes for Biological Sciences,
Chinese Academy of Sciences (Shanghai, China). HepG2 cells
Supplementary information, Figure S7F-S7H, we added
were maintained at 37 °C in a humidified 5% CO
miR-150, an endogenous mammalian miRNA, into chow
2 incubator with
Dulbecco's modified eagle medium (Gibco, CA, USA) containing
diet and fed mice with miR-150-enriched chow diet and
10% fetal bovine serum (FBS, Gibco), 100 units/ml of penicil-
normal chow diet, respectively. We found that miR-150
lin, and 100 µg/ml of streptomycin. Caco-2 cells were grown in
could also enter mouse liver and downregulate its target
minimum essential medium (Gibco) containing 10% FBS (Gibco),
gene, c-Myb. Given that exogenous miRNAs in food or
100 units/ml of penicillin, and 100 µg/ml of streptomycin. Anti-
miRNAs that are ‘added' into the food can enter the cir-
LDLRAP1 (LS-C20125) antibody was purchased from Lifespan
Biosciences (Seattle, WA, USA), anti-Ago2 (ab57113) antibody
culation and various organs of animals and play a role in
was purchased from Abcam (Cambridge, MA, USA), and anti-c-
regulating the physiological or pathophysiological condi-
Myb (C19) and anti-α-tubulin (B-7) antibodies were purchased
tions, food-derived exogenous miRNAs may be qualified
from Santa Cruz Biotechnology (Santa Cruz, CA, USA). Synthetic
as a novel nutrient component, like vitamins and miner-
RNA molecules, including pre-MIR168a, anti-MIR168a and
scrambled negative control oligonucleotides (pre-ncRNA and anti-
Previous studies have reported that the transfer of ge-
ncRNA), were purchased from Ambion (Austin, TX, USA). Syn-
netic material from one species to another may modulate
thetic mature MIR168a oligonucleotides and scrambled negative
the cellular functions of the recipient species [50, 51].
control oligonucleotides (mature ncRNA) were purchased from
Takara (Dalian, China).
Such examples include human miRNAs targeting viral
genes [50] and the translocation of host plant mRNAs
Serum preparation
into dodder (a parasitic plant) [51]. However, to our
The recruitment of sera from 11 male (mean age 26.3 ± 1.9) and
knowledge, it was still unknown whether plant miRNAs
10 female (mean age 24.2 ± 1.3) healthy Chinese, and 8 pooled
could enter mammals and modulate mammalian cell
samples (each pooled from 10 healthy Chinese subjects, mean
functions. By illustrating that plant miRNAs, such as
age 50.9 ± 7.9) was conducted in the Healthy Physical Examina-
MIR168a, can be delivered into animal serum and tissues
tion Center of the Jinling Hospital. The health condition checkup
included detailed history, physical, radiological examinations,
through food intake and digestion and that exogenous
blood tests, and abdominal sonography. Subjects who showed no
MIR168a can target mammalian liver-specific LDL-
abnormalities during the medical checkup were enrolled. Written
Cell Research Vol 22 No 1 January 2012
Lin Zhang
et al. npg
informed consent was obtained from all donors prior to the study,
using TaqMan miRNA probes (Applied Biosystems, Foster City,
and the study was approved by the ethics committee of Nanjing
CA, USA) according to the manufacturer's instructions. Briefly,
University, China. Venous blood samples ( 5 ml) were collected
total RNA was reverse transcribed to cDNA using AMV reverse
from each donor and placed in a serum separator tube. Samples
transcriptase (Takara) and a stem-loop RT primer (Applied Biosys-
were processed within 1 h. Separation of the serum was accom-
tems). Real-time PCR was performed using a TaqMan PCR kit and
plished by centrifugation at 800×
g for 10 min at room tempera-
an Applied Biosystems 7300 Sequence Detection System (Applied
ture, followed by a 15 min high-speed centrifugation at 10 000×
g
at room temperature to completely remove the cell debris. The su-
All reactions, including no-template controls, were performed
pernatant serum was recovered and stored at −80 °C until analysis.
in triplicate. After the reaction, the CT values were determined us-
ing fixed threshold settings. To calculate the absolute expression
Solexa sequencing
levels of the target miRNAs, a series of synthetic miRNA oligo-
The sequencing procedure was conducted as previously de-
nucleotides at known concentrations was reverse transcribed and
scribed [5, 52]. Briefly, total RNA was extracted from 100 ml of
amplified. The absolute amount of each miRNA was then calculat-
serum using the Trizol Reagent (Invitrogen, Carlsbad, CA, USA)
ed in reference to the standard curve. In the experiments presented
according to the manufacturer's instructions. After the PAGE puri-
here, miRNA expression in cells was normalized to U6 snRNA, as
fication of small RNA molecules (under 30 base pairs) and the li-
is common in many other reports [53].
gation of a pair of Solexa adaptors to their 5′ and 3′ ends, the small
RNA molecules were amplified using the adaptor primers for 17
Deep sequencing and qRT-PCR after oxidation of small
cycles and the fragments of around 90 bp (small RNA + adaptors)
RNAs with periodate
were isolated from PAGE gels. The purified DNA was directly
The methylation status of small RNAs was evaluated by treat-
used for the cluster generation and sequencing analysis using an Il-
ment of the small RNAs with sodium periodate followed by Solexa
lumina Genome Analyzer according to the manufacturer's instruc-
sequencing or qRT-PCR with the miScript PCR system. Periodate
tions. The image files generated by the sequencer were then pro-
oxidation was performed as previously described with slight modi-
cessed to produce digital data. The subsequent procedures included
fications [22, 54]. Briefly, total RNA was extracted from 200 ml
summarizing data production, evaluating sequencing quality and
of human serum or 100 mg mouse liver using the Trizol Reagent
depth, calculating length distribution of small RNAs, and filtrat-
(Invitrogen) according to the manufacturer's instructions. Small
ing contaminated reads. After masking the adaptor sequences, the
RNA fractions (fewer than 30 base pairs) were enriched by PAGE
clean reads were aligned against the miRBase database 16.0 based
purification. A 100 µl mixture consisting of 20 µg of small RNA
on the Smith-Waterman algorithm. Only candidate with identical
fraction and 10 mM NaIO4 was incubated at 0 °C for 40 min in
sequence and length compared to reference miRNA was counted
dark. The oxidized RNA was precipitated twice by ethanol, rinsed
as miRNA matching. For normalization, the sequencing frequency
once with 80% ethanol, aired dried, dissolved in ddH2O, and then
of each plant miRNA was normalized to the total amount of mam-
subjected to Solexa sequencing or qRT-PCR assay. The qRT-PCR
malian miRNAs.
assay was conducted using the miScript PCR system (QIAGEN,
Valencia, CA, USA) according to the manufacturer's instructions.
Transfection of cells with ncRNA, pre-MIR168a, or mature
Briefly, miRNAs are polyadenylated by poly(A) polymerase and
subsequently converted into cDNA by reverse transcriptase with
HepG2 or Caco-2 cells were seeded on 12-well plates or 10-
oligo-dT. The cDNA is then used for real-time PCR quantification
mm dishes overnight and transfected the following day using
of mature miRNAs.
Lipofectamine 2000 (Invitrogen), according to the manufacturer's
instructions. For the overexpression of MIR168a, 20 pmol per 1 ×
Northern blotting analysis
105 cells of pre-MIR168a or mature MIR168a was used. Scrambled
Oligonucleotide probes complementary to mature miRNAs
negative control pre-miRNA (pre-ncRNA) and mature ncRNA
were end-labeled with γ-32P-ATP using T4 Polynucleotide Kinase
were used as controls for pre-MIR168a and mature MIR168a,
(Takara). Labeled probes were purified using a Sephadex G25 spin
respectively. Cells were harvested 24 or 48 h after transfection for
column (Roche). Total RNA was extracted from 100 ml human
semi-quantitative RT-PCR, real-time PCR analysis, and western
serum or 40 ml calf serum using TRIzol Reagent (Invitrogen) ac-
cording to the manufacturer's instructions. Synthetic oligonucle-
otides (1 pmol) were loaded as positive control. Total RNA was
MV isolation
fractionated by PAGE using a 15% denaturing polyacrylamide gel.
MVs were isolated from the cell culture medium by differential
The RNA was then transferred onto a nylon membrane (Hybond
centrifugation according to previous publications [16, 17]. Briefly,
N+, Amersham Biosciences) by electroblotting at 200 mA in 0.5×
after removing cells and other debris by centrifugation at 300×
g,
TBE (Tris-Borate-EDTA) buffer for 2 h. The membrane was dried
1 200×
g, and 10 000×
g, the supernatant was centrifuged at 110
and cross-linked. A prehybridization step was performed by in-
000×
g for 2 h (all steps were performed at 4 °C). MVs were col-
cubating the membrane with 10 ml of ULTRAhyb-Oligo solution
lected from the pellet and resuspended in FBS-free medium.
(Ambion) pre-heated to 65 °C. Prehybridization was performed for
1 h at 37 °C in a standard rotating hybridization oven. The radio-
RNA isolation and qRT-PCR of mature miRNAs
labeled probe was added directly to the ULTRAhyb-Oligo solution
Total RNA was extracted from the serum, cells, or tissues using
and the membrane was incubated overnight at 37 °C with rotation
TRIzol Reagent or Trizol LS Reagent (Invitrogen) according to the
in a hybridization oven. After hybridization, the membrane was
manufacturer's instructions. Quantitative RT-PCR was performed
washed 3 × 10 min at room temperature in 2× SSC (Saline-sodium
www.cell-research.com Cell Research
npg Plant MIR168a downregulates mammalian LDLRAP1124
citrate), 0.5% sodium dodecyl sulfate (SDS) and then 1 × 15 min
according to the manufacturer's instructions or by semi-quantita-
at 42 °C in 2 × SSC, 0.5% SDS. The membrane was wrapped in
tive RT-PCR using primers specific for human LDLRAP1. Primer
plastic wrap and exposed to an X-ray film at −80 °C.
sequences for human LDLRAP1 were as follows: 5′-AGAGC-
CAGCACAACCAGA-3′ (forward primer) and 5′-CTTGGACAC-
Western blot analysis
CTGCCAAAA-3′ (reverse primer). Primer sequences for mouse
Samples of tissues and cultured cells were lysed in a buffer
LDLRAP1 were as follows: 5′-AAGTATCTTGGTATGACGCT-
(50 mM Tris-HCl pH 7.4, 150 mM NaCl, 1% NP-40, 0.1% SDS),
GGTG-3′ (forward primer) and 5′-TCCTGGTTGGCTTTCTC-
sonicated (6 × 1.5 s, 30% power), and centrifuged at 12 000×
g
CCT-3′ (reverse primer).
for 10 min at 4 °C. The supernatant fraction was removed, and
the protein concentration was determined by BCA assay (Pierce,
Rockford, USA). Aliquots of proteins (60 to 100 µg) were separat-
All experimental animals were maintained on a C57BL/6J
ed on 10% SDS-polyacrylamide gels (SDS-PAGE) and transferred
background on a 12-h light/dark cycle in a pathogen-free animal
to polyvinylidene difluoride (PVDF) membranes. The membranes
facility at Nanjing University. The Institutional Review Board of
were blocked for 1 h at room temperature with 5% non-fat milk in
Nanjing University approved all housing and surgical procedures.
Tris-buffered saline (TBS) plus Tween 20 (TBST), followed by an
At 10 weeks of age, each mouse was fed fresh rice total RNA
overnight incubation at 4 °C with antibodies (diluted in blocking
(80 µg), synthetic MIR168a (300 pmol), or synthetic methylated
buffer) against LDLRAP1 and α-tubulin. Following 3 × 10 min
MIR168a (300 pmol) by gavage after fasting overnight. After a
washes with TBST, the blots were incubated at room tempera-
fixed time interval (i.e., 0.5 h, 3 h, or 6 h), serum and tissues were
ture for 1 h with the appropriate secondary antibody conjugated
collected and total RNA was extracted. In a separate experiment,
to horseradish peroxidase (HRP) and detected with an enhanced
after fasting overnight, two groups of 10-week-old male mice were
chemiluminescence reagent (Cell Signaling Technology Inc.,
placed on a diet of either mouse chow (the fundamental ingre-
USA). The autoradiographic intensity of each band was scanned
dients of the chow diet are listed in Supplementary information,
and quantified using BandScan software (Glyko Inc., Novato, CA,
Table S5) or fresh rice. Fresh food was administered every 3 days
USA). Values were normalized to α-tubulin and the ratio to the
from a supply stored at −20 °C, and food consumption and body
control values was calculated.
weight were measured. The mice were maintained on the diets for
a fixed time interval (i.e., 0.5, 3, or 6 h, or 1, 3, or 7 days), after
Plasmid construction and luciferase assay
which serum and tissues were collected. Furthermore, the mice fed
A mammalian expression vector encoding the human LDL-
on rice received tail vein injections of ncRNA or anti-MIR168a
RAP1 ORF (pReceiver-M02-LDLRAP1) and its C-terminally
(10 nmol each). After a fixed time interval (i.e., 0.5, 3, or 6 h,
GFP-tagged form (pReceiver-M03-LDLRAP1) were purchased
or 1, 3, or 7 days), sera and tissues were collected. Several mice
from GeneCopoeia (Germantown, MD, USA). To introduce muta-
were euthanized directly after overnight fasting, and their sera
tions into the MIR168a target site in the LDLRAP1 coding region,
and tissues were collected as a control. In a separate experiment,
primers were designed for site-directed mutagenesis that resulted
synthetic MIR168a or ncRNA (100 pmol) were mixed evenly with
in the destruction of the MIR168a target site without altering the
5 g of mouse chow. To avoid miRNA degradation and contamina-
amino-acid sequence of LDLRAP1. The sites (LCTKR) were
tion, this food was prepared before feeding. qRT-PCR was used
mutated as follows: prior to mutagenesis, CTC TGC ACC AAG
to determine the level of MIR168a in the chow diet with the addi-
CGG; following mutagenesis, CTg TGt ACg AAa CGc. To gener-
tion of MIR168a or ncRNA. After fasting overnight, two groups
ate luciferase reporters, the amplified fragments were cloned into
of 10-week-old male mice were placed on a diet of mouse chow
the 3′ UTR region of the pMIR-report plasmid (Ambion). Efficient
plus either MIR168a or ncRNA. Fresh food was administered ev-
insertion was confirmed by sequencing. For luciferase reporter
ery day, and food consumption and body weight were measured.
assays, 0.2 µg of firefly luciferase reporter plasmid, 0.1 µg of
Generally, mouse consumes 5-7 g of food per day. The mice were
β-galactosidase expression vector (Ambion), and equal amounts
maintained on the diets for 3 days, after which serum and tissues
(20 pmol) of pre-MIR168a, mature MIR168a or scrambled nega-
were collected.
tive control RNA were transfected into cells in 24-well plates. The
β-galactosidase vector was used as a transfection control. At 24 h
Analysis of serum lipids and lipoproteins
post-transfection, cells were analyzed using a luciferase assay kit
Serum lipids and lipoproteins were assayed using commercially
available kits and a clinical chemistry analyzer (HITACHI 7600,
Hitachi Koki Co. Ltd., Hitachinaka City, Japan). Reagent kits for
total cholesterol and triglycerides were obtained from Randox
Immunoprecipitation assays were performed using a Chromatin
Laboratories, Ltd. (Crumlin, UK) and reagent kits for LDL choles-
Immunoprecipitation (ChIP) Assay Kit (Millipore) according to
terol and lipoprotein A were obtained from Daiichi Pure Chemicals
the manufacturer's instructions. Briefly, cells were washed three
times with cold PBS (4 °C), scraped from each dish and then col-
lected by centrifugation at 1 000 rpm for 5 min at 4 °C. Cells were
then resuspended in an appropriate volume of complete RIP lysis
Full-length cDNAs of the human genes were obtained from the
buffer. Mouse monoclonal anti-AGO2 antibody (5 µg) was used
NCBI Genebank database. A program was developed and imple-
to immunoprecipitate RNA-binding proteins. After purification,
mented to identify MIR168a-matched sites in the entire CDS/UTR
immunoprecipitated RNA was analyzed by real-time RT-PCR for
of the transcripts. This program used several common criteria to
MIRNA168a using TaqMan miRNA probes (Applied Biosystems)
determine whether a transcript was a target for MIR168a. The first
Cell Research Vol 22 No 1 January 2012
Lin Zhang
et al. npg
criterion for target recognition named "seed rules" was base pair-
ing between the "seed" (the core sequence that encompassed the
10 Resnick KE, Alder H, Hagan JP, Richardson DL, Croce CM,
first 2 to 8 bases of the mature miRNA) and the target [55]. Sec-
Cohn DE. The detection of differentially expressed microR-
ond, the free energy of the hybrid was expected to be within the
NAs from the serum of ovarian cancer patients using a novel
range of the authentic miRNA-target pairs, typically lower than
real-time PCR platform.
Gynecol Oncol 2009;
112:55-59.
−17 kcal/mol. Third, an optional rule for target prediction required
11 Wang K, Zhang S, Marzolf B,
et al. Circulating microRNAs,
interspecies conservation of the putative binding sites. Using
potential biomarkers for drug-induced liver injury.
Proc Natl
these rules, 50 human genes were identified as putative targets of
Acad Sci USA 2009;
106:4402-4407.
12 Zhang C, Wang C, Chen X,
et al. Expression profile of mi-
croRNAs in serum: a fingerprint for esophageal squamous
cell carcinoma.
Clin Chem 2010;
56:1871-1879.
All photo images of western blotting and semi-quantitative RT-
13 Cocucci E, Racchetti G, Meldolesi J. Shedding microvesicles:
PCR are representative of at least three independent experiments.
artefacts no more.
Trends Cell Biol 2009;
19:43-51.
Real-time PCR were performed in triplicate and each experiment
14 Thery C, Zitvogel L, Amigorena S. Exosomes: composition,
was repeated several times. Data shown are presented as the mean
biogenesis and function.
Nat Rev Immunol 2002;
2:569-579.
± SEM of at least three independent experiments; differences are
15 Zhang Y, Liu D, Chen X,
et al. Secreted monocytic miR-150
considered statistically significant at
P < 0.05, using a Student's
enhances targeted endothelial cell migration.
Mol Cell 2010;
16 Skog J, Wurdinger T, van Rijn S,
et al. Glioblastoma mi-
crovesicles transport RNA and proteins that promote tumour
growth and provide diagnostic biomarkers.
Nat Cell Biol
We thank Dr Daniel Wu (Yale University, USA) and Dr Zheng-
Gang Liu (Cancer Research Institute, NIH, USA) for critical
17 Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ, Lotvall
reading of the manuscript and discussion of data. This work was
JO. Exosome-mediated transfer of mRNAs and microRNAs
supported by grants from the National Natural Science Foundation
is a novel mechanism of genetic exchange between cells.
Nat
of China (90813035, 30890044, 30771036, 30772484, 30725008,
Cell Biol 2007;
9:654-659.
30890032, 31071232, 31000323, and 90608010), the National
18 Collino F, Deregibus MC, Bruno S,
et al. Microvesicles
Basic Research Program of China (973 Program 2006CB503909,
derived from adult human bone marrow and tissue specific
2007CB815701, 2007CB815703, 2007CB815705, and
mesenchymal stem cells shuttle selected pattern of miRNAs.
PLoS One 2010;
5:e11803.
2006AA10A121), and the Natural Science Foundation of Jiangsu
19 Meckes DG Jr, Shair KH, Marquitz AR, Kung CP, Edwards
RH, Raab-Traub N. Human tumor virus utilizes exosomes for
intercellular communication.
Proc Natl Acad Sci USA 2010;
20 Ogawa R, Tanaka C, Sato M,
et al. Adipocyte-derived mi-
crovesicles contain RNA that is transported into macrophages
1 Bartel DP. MicroRNAs: genomics, biogenesis, mechanism,
and might be secreted into blood circulation.
Biochem Bio-
and function.
Cell 2004;
116:281-297.
phys Res Commun 2010;
398:723-729.
2 He L, Hannon GJ. MicroRNAs: small RNAs with a big role
21 Pegtel DM, Cosmopoulos K, Thorley-Lawson DA,
et al.
in gene regulation.
Nat Rev Genet 2004;
5:522-531.
Functional delivery of viral miRNAs via exosomes.
Proc Natl
3 Calin GA, Croce CM. MicroRNA signatures in human can-
Acad Sci USA 2010;
107:6328-6333.
cers.
Nat Rev Cancer 2006;
6:857-866.
22 Yu B, Yang Z, Li J,
et al. Methylation as a crucial step in plant
4 Esquela-Kerscher A, Slack FJ. Oncomirs – microRNAs with
microRNA biogenesis.
Science 2005;
307:932-935.
a role in cancer.
Nat Rev Cancer 2006;
6:259-269.
23 Grimson A, Srivastava M, Fahey B,
et al. Early origins and
5 Chen X, Ba Y, Ma L,
et al. Characterization of microRNAs
evolution of microRNAs and Piwi-interacting RNAs in ani-
in serum: a novel class of biomarkers for diagnosis of cancer
mals.
Nature 2008;
455:1193-1197.
and other diseases.
Cell Res 2008;
18:997-1006.
24 Hartig JV, Forstemann K. Loqs-PD and R2D2 define indepen-
6 Mitchell PS, Parkin RK, Kroh EM,
et al. Circulating microR-
dent pathways for RISC generation in
Drosophila.
Nucleic
NAs as stable blood-based markers for cancer detection.
Proc
Acids Res 2011;
39:3836-3851.
Natl Acad Sci USA 2008;
105:10513-10518.
25 Gazzani S, Li M, Maistri S,
et al. Evolution of MIR168 paral-
7 Gilad S, Meiri E, Yogev Y,
et al. Serum microRNAs are
ogs in Brassicaceae.
BMC Evol Biol 2009;
9:62.
promising novel biomarkers.
PLoS One 2008;
3:e3148.
26 Jia X, Mendu V, Tang G. An array platform for identification
8 Li LM, Hu ZB, Zhou ZX,
et al. Serum microRNA profiles
of stress-responsive microRNAs in plants.
Methods Mol Biol
serve as novel biomarkers for HBV infection and diagnosis of
HBV-positive hepatocarcinoma.
Cancer Res 2010;
70:9798-
27 Chen X. MicroRNA biogenesis and function in plants.
FEBS
Lett 2005;
579:5923-5931.
9 Ng EK, Chong WW, Jin H,
et al. Differential expression of
28 Du T, Zamore PD. microPrimer: the biogenesis and function
microRNAs in plasma of patients with colorectal cancer: a
of microRNA.
Development 2005;
132:4645-4652.
potential marker for colorectal cancer screening.
Gut 2009;
29 Garcia CK, Wilund K, Arca M,
et al. Autosomal recessive
www.cell-research.com Cell Research
npg Plant MIR168a downregulates mammalian LDLRAP1126
hypercholesterolemia caused by mutations in a putative LDL
according to tissue, lineage and differentiation state.
Nat Bio-
receptor adaptor protein.
Science 2001;
292:1394-1398.
technol 2007;
25:1457-1467.
30 Huang S, Wu S, Ding J,
et al. MicroRNA-181a modulates
43 Chen C, Ridzon DA, Broomer AJ,
et al. Real-time quantifica-
gene expression of zinc finger family members by directly tar-
tion of microRNAs by stem-loop RT-PCR.
Nucleic Acids Res
geting their coding regions.
Nucleic Acids Res 2010;
38:7211-
44 Neilson JR, Zheng GX, Burge CB, Sharp PA. Dynamic regu-
31 Qin W, Shi Y, Zhao B,
et al. miR-24 regulates apoptosis by
lation of miRNA expression in ordered stages of cellular de-
targeting the open reading frame (ORF) region of FAF1 in
velopment.
Genes Dev 2007;
21:578-589.
cancer cells.
PLoS One 2010;
5:e9429.
45 Bissels U, Wild S, Tomiuk S,
et al. Absolute quantification
32 Tay Y, Zhang J, Thomson AM, Lim B, Rigoutsos I. MicroR-
of microRNAs by using a universal reference.
RNA 2009;
NAs to Nanog, Oct4 and Sox2 coding regions modulate
embryonic stem cell differentiation.
Nature 2008;
455:1124-
46 Meng F, Henson R, Wehbe-Janek H, Ghoshal K, Jacob ST,
Patel T. MicroRNA-21 regulates expression of the PTEN tu-
33 Gibbings DJ, Ciaudo C, Erhardt M, Voinnet O. Multivesicular
mor suppressor gene in human hepatocellular cancer.
Gastro-
bodies associate with components of miRNA effector com-
enterology 2007;
133:647-658.
plexes and modulate miRNA activity.
Nat Cell Biol 2009;
47 Shimizu S, Takehara T, Hikita H,
et al. The let-7 family
of microRNAs inhibits Bcl-xL expression and potentiates
34 Goldstein JL, Brown MS. The low-density lipoprotein path-
sorafenib-induced apoptosis in human hepatocellular carci-
way and its relation to atherosclerosis.
Annu Rev Biochem
noma.
J Hepatol 2010;
52:698-704.
48 Tomimaru Y, Eguchi H, Nagano H,
et al. MicroRNA-21
35 Wilund KR, Yi M, Campagna F,
et al. Molecular mechanisms
induces resistance to the anti-tumour effect of interferon-
of autosomal recessive hypercholesterolemia.
Hum Mol Genet
alpha/5-fluorouracil in hepatocellular carcinoma cells.
Br J
Cancer 2010;
103:1617-1626.
36 Zuliani G, Arca M, Signore A,
et al. Characterization of a new
49 Zhang J, Yang Y, Yang T,
et al. microRNA-22, downregulated
form of inherited hypercholesterolemia: familial recessive
in hepatocellular carcinoma and correlated with prognosis,
hypercholesterolemia.
Arterioscler Thromb Vasc Biol 1999;
suppresses cell proliferation and tumourigenicity.
Br J Cancer
37 Mallory AC, Vaucheret H. ARGONAUTE 1 homeostasis
50 Jopling CL, Yi M, Lancaster AM, Lemon SM, Sarnow P.
invokes the coordinate action of the microRNA and siRNA
Modulation of hepatitis C virus RNA abundance by a liver-
pathways.
EMBO Rep 2009;
10:521-526.
specific MicroRNA.
Science 2005;
309:1577-1581.
38 Vaucheret H, Mallory AC, Bartel DP. AGO1 homeostasis en-
51 Roney JK, Khatibi PA, Westwood JH. Cross-species translo-
tails coexpression of MIR168 and AGO1 and preferential sta-
cation of mRNA from host plants into the parasitic plant dod-
bilization of miR168 by AGO1.
Mol Cell 2006;
22:129-136.
der.
Plant Physiol 2007;
143:1037-1043.
39 Vaucheret H, Vazquez F, Crete P, Bartel DP. The action of
52 Chen X, Li Q, Wang J,
et al. Identification and characteriza-
ARGONAUTE1 in the miRNA pathway and its regulation by
tion of novel amphioxus microRNAs by Solexa sequencing.
the miRNA pathway are crucial for plant development.
Genes
Genome Biol 2009;
10:R78.
Dev 2004;
18:1187-1197.
53 Jiang J, Lee EJ, Gusev Y, Schmittgen TD. Real-time expres-
40 Lima WF, Wu H, Nichols JG, Sun H, Murray HM, Crooke
sion profiling of microRNA precursors in human cancer cell
ST. Binding and cleavage specificities of human Argonaute2.
lines.
Nucleic Acids Res 2005;
33:5394-5403.
J Biol Chem 2009;
284:26017-26028.
54 Ohara T, Sakaguchi Y, Suzuki T, Ueda H, Miyauchi K. The 3′
41 Brown BD, Naldini L. Exploiting and antagonizing microR-
termini of mouse Piwi-interacting RNAs are 2′-O-methylated.
NA regulation for therapeutic and experimental applications.
Nat Struct Mol Biol 2007;
14:349-350.
Nat Rev Genet 2009;
10:578-585.
55 Smalheiser NR, Torvik VI. Complications in mammalian mi-
42 Brown BD, Gentner B, Cantore A,
et al. Endogenous microR-
croRNA target prediction.
Methods Mol Biol 2006;
342:115-
NA can be broadly exploited to regulate transgene expression
(
Supplementary information is linked to the online version of
the paper on the
Cell Research website.)
Cell Research Vol 22 No 1 January 2012
Source: http://www.thetherapybook.net/Exogenous%20plant%20MIR168a%20by%20Lin%20Zhang%20et%20al.pdf
Einstufungstest (B2): 1. Leseverstehen Lesen Sie zuerst die 10 Überschriften. Lesen Sie dann die fünf Texte und entscheiden Sie, welcher Text (1–5) am besten zu welcher Überschrift (a–j) passt. Tragen Sie Ihre Lösungen in den Antwortbogen bei den Aufgaben 1–5 ein. Neue Richtlinie für Zertifizierung von Medikamenten für Kinder
Management of agitation and aggression associated with Alzheimer diseaseClive G. Ballard, Serge Gauthier, Jeffrey L. Cummings, Henry Brodaty, George T. Grossberg, Philippe Robert and Constantine G. Lyketsos Abstract Agitation and aggression are frequently occurring and distressing behavioral and psychological symptoms of dementia (BPsD). These symptoms are disturbing for individuals with Alzheimer disease, commonly confer risk to the patient and others, and present a major management challenge for clinicians. The most widely prescribed pharmacological treatments for these symptoms—atypical antipsychotics—have a modest but significant beneficial effect in the short-term treatment (over 6–12 weeks) of aggression but limited benefits in longer term therapy. Benefits are less well established for other symptoms of agitation. in addition, concerns are growing over the potential for serious adverse outcomes with these treatments, including stroke and death. A detailed consideration of other pharmacological and nonpharmacological approaches to agitation and aggression in patients with Alzheimer disease is, therefore, imperative. This article reviews the increasing evidence in support of psychological interventions or alternative therapies (such as aromatherapy) as a first-line management strategy for agitation, as well as the potential pharmacological alternatives to atypical antipsychotics—preliminary evidence for memantine, carbamazepine, and citalopram is encouraging.