ArticlePDF Available

Molecular characterization of exosomes and their microRNA cargo in human follicular fluid: Bioinformatic analysis reveals that exosomal microRNAs control pathways involved in follicular maturation

Authors:

Abstract and Figures

Objective: To characterize well-represented microRNAs in human follicular fluid (FF) and to ascertain whether they are cargo of FF exosomes and whether they are involved in the regulation of follicle maturation. Design: FF exosomes were characterized by nanosight, flow cytometry, and exosome-specific surface markers. Expression microRNA profiles from total and exosomal FF were compared with those from plasma of the same women. Setting: University laboratory and an IVF center. Patient(s): Fifteen healthy women who had undergone intracytoplasmic sperm injection. Intervention(s): None. Main outcome measure(s): TaqMan low-density array to investigate the expression profile of 384 microRNAs; DataAssist and geNorm for endogenous control identification; significance analysis of microarrays to identify differentially expressed microRNAs; nanosight, flow-cytometry, and bioanalyzer for exosome characterization; bioinformatic tools for microRNAs target prediction, gene ontology, and pathway analysis. Result(s): We identified 37 microRNAs upregulated in FF as compared with plasma from the same women. Thirty-two were carried by microvesicles that showed the well-characterized exosomal markers CD63 and CD81. These FF microRNAs are involved in critically important pathways for follicle growth and oocyte maturation. Specifically, nine of them target and negatively regulate mRNAs expressed in the follicular microenvironment encoding inhibitors of follicle maturation and meiosis resumption. Conclusion(s): This study identified a series of exosomal microRNAs that are highly represented in human FF and are involved in follicular maturation. They could represent noninvasive biomarkers of oocyte quality in assisted reproductive technology.
Content may be subject to copyright.
Molecular characterization
of exosomes and their microRNA
cargo in human follicular uid:
bioinformatic analysis reveals that
exosomal microRNAs control
pathways involved in
follicular maturation
Manuela Santonocito, Ph.D.,
a
Marilena Vento, Ph.D.,
b
Maria Rosa Guglielmino, Ph.D.,
a
Rosalia Battaglia, M.Sc.,
a
Jessica Wahlgren, Ph.D.,
c
Marco Ragusa, Ph.D.,
a
Davide Barbagallo, Ph.D.,
a
Placido Borzì, M.D.,
b
Simona Rizzari, M.Sc.,
b
Marco Maugeri, Ph.D.,
a
Paolo Scollo, M.D.,
b
Carla Tatone, Ph.D.,
d
Hadi Valadi, Ph.D.,
c
Michele Purrello, M.D., Ph.D.,
a
and Cinzia Di Pietro, Ph.D.
a
a
Department Gian Filippo Ingrassia, Biologia Cellulare e Molecolare, Genetica, Genomica Giovanni Sichel, Universit
adi
Catania;
b
Servizio di PMA/Azienda Ospedaliera Cannizzaro, Catania, Italy;
c
Department of Rheumatology and
Inammation Research, University of Gothenburg, Gothenburg, Sweden; and
d
Department of Life, Health and
Environmental Sciences, University of L'Aquila, L'Aquila, Italy
Objective: To characterize well-represented microRNAs in human follicular uid (FF) and to ascertain whether they are cargo of FF
exosomes and whether they are involved in the regulation of follicle maturation.
Design: FF exosomes were characterized by nanosight, ow cytometry, and exosome-specic surface markers. Expression microRNA
proles from total and exosomal FF were compared with those from plasma of the same women.
Setting: University laboratory and an IVF center.
Patient(s): Fifteen healthy women who had undergone intracytoplasmic sperm injection.
Intervention(s): None.
Main Outcome Measure(s): TaqMan low-density array to investigate the expression prole of 384 microRNAs; DataAssist and
geNorm for endogenous control identication; signicance analysis of microarrays to identify differentially expressed microRNAs;
nanosight, ow-cytometry, and bioanalyzer for exosome characterization; bioinformatic tools for microRNAs target prediction,
gene ontology, and pathway analysis.
Result(s): We identied 37 microRNAs upregulated in FF as compared with plasma from the same women. Thirty-two were
carried by microvesicles that showed the well-characterized exosomal markers CD63 and CD81. These FF microRNAs are
involved in critically important pathways for follicle growth and oocyte maturation. Specically, nine of them target and
negatively regulate mRNAs expressed in the follicular microenvironment encoding inhibitors of follicle maturation and meiosis
resumption.
Received February 25, 2014; revised August 4, 2014; accepted August 5, 2014.
M.S. has nothing to disclose. M.V. has nothing to disclose. M.R.G. has nothing to disclose. R.B. has nothing to disclose. J.W. has nothing to disclose. M.R. has
nothing to disclose. D.B. has nothing to disclose. P.B. has nothing to disclose. S.R. has nothing to disclose. M.M. has nothing to disclose. P.S. has nothing
to disclose. C.T. has nothing to disclose. H.V. has nothing to disclose. M.P. has nothing to disclose. C.D.P. has nothing to disclose.
This work was supported by Ministero dellUniversit
a e della Ricerca Scientica e Tecnologica (to C.D.P) and by Farmitalia SRL and LJ Pharma SRL. M. San-
tonocito acknowledges the support of the EMBO-European Molecular Biology Organization Germany and Ente Regionale per il diritto allo Studio Uni-
versitario for the assignment of a fellowship that allowed her to perform the experiments on exosome isolation and characterization with Dr. Hadi
Valadi's group at the University of Gothenburg, Sweden.
Reprint requests: Cinzia Di Pietro, Ph.D., Department Gian Filippo Ingrassia, Biologia Cellulare e Molecolare, Genetica, Genomica Giovanni Sichel, Universit
a
di Catania, Via Santa Soa 87, Comparto 10, Edicio C, 95123 Catania, Italy (E-mail: dipietro@unict.it).
Fertility and Sterility® Vol. -, No. -,-2014 0015-0282/$36.00
Copyright ©2014 American Society for Reproductive Medicine, Published by Elsevier Inc.
http://dx.doi.org/10.1016/j.fertnstert.2014.08.005
VOL. -NO. -/-2014 1
ORIGINAL ARTICLE: REPRODUCTIVE BIOLOGY
Conclusion(s): This study identied a series of exosomal microRNAs that are highly repre-
sented in human FF and are involved in follicular maturation. They could represent noninvasive
biomarkers of oocyte quality in assisted reproductive technology. (Fertil Steril
Ò
2014;-:--.
Ó2014 by American Society for Reproductive Medicine.)
Key Words: Follicular uid, exosomes, microRNA prole, intrafollicle molecular
communication
Discuss: You can discuss this article with its authors and with other ASRM members at http://
fertstertforum.com/santonocitom-exosomes-microrna-cargo-follicular-uid/
Use your smartphone
to scan this QR code
and connect to the
discussion forum for
this article now.*
* Download a free QR code scanner by searching for QR
scannerin your smartphones app store or app marketplace.
MicroRNAs (miRNAs) are small, noncoding RNAs
that repress gene expression by targeting specic
mRNAs, most commonly in their 30untranslated
region. Accordingly, they play a very important role in
post-transcriptional gene regulation (1). miRNAs regulate
the activity of most protein-coding genes; a single miRNA
may target hundreds of mRNAs, while a specic mRNA mole-
cule may be regulated by several different miRNAs (1, 2).
Many miRNAs have been conserved throughout evolution
and perform important roles in cell differentiation and the
development of invertebrates, plants, and animals,
including humans (1, 2). Over the past few years it has
become evident that alterations of their expression are
important in the pathogenesis of human diseases such as
cancers and degenerative diseases (2, 3). Another avenue of
current research is the study of circulating miRNAs in body
uids. It has been demonstrated that miRNAs are present in
most (if not all) biological uids, such as serum, plasma,
saliva, urine, and breast milk (2, 4). In these uids, miRNAs
display remarkable stability and resistance to degradation,
possibly because of their association with protein
complexes (e.g., argonaute RISC catalytic component 2,
nucleophosmin [nucleolar phosphoprotein B23, numatrin])
or because they are enclosed within membrane-bound nano-
vesicles, termed exosomes (5). Exosomes are vesicles of endo-
cytic origin between 30 and 100 nm in diameter; they are
formed through inward budding of endosomal membranes,
which gives rise to intracellular multivesicular bodies. These
later fuse with the plasma membrane and release the intralu-
menal vesicles into the extracellular environment as exo-
somes (5). Their unique chemical composition enables
exosome identication; specically, tetraspanins such as
CD9, CD63, CD81, and CD82 are dened exosomal marker
proteins (6, 7). Recent studies have demonstrated that
exosomes are bioactive vesicles that promote intercellular
communication by shuttling proteins, mRNAs, and miRNAs
among cells (5, 8). The discovery that miRNAs might act
not only within cells but also may be involved in
intercellular communication led to the hypothesis that
miRNAs could represent the oldest hormones (2). Similar to
cellular miRNAs, the proles of circulating miRNAs may be
associated with specic diseases. Accordingly, their role as
noninvasive biomarkers of pathologies and also as
potential targets of therapy has been investigated in
different neoplastic and degenerative diseases (e.g., colon
cancer, diabetes mellitus) (9, 10). Follicular uid (FF)
consists of a complex mixture of nucleic acids, proteins,
metabolites, and ions, which are secreted by the oocyte,
granulose, and thecal cells; it also comprises plasma
components that cross the blood-follicle barrier via thecal
capillaries (11, 12). It represents a critically important
microenvironment for the development of oocytes: its
biochemical composition, related to the metabolic activity
of ovarian cells, reects the physiological status of the
follicle. It is common opinion that the analysis of FF
components may provide useful information on oocyte
quality and the pathways involved in follicle differentiation
and development (11). The discovery of FF miRNAs could
open up the possibility to use these molecules as
biomarkers of oocyte quality. Some recent papers
demonstrated the presence of miRNA in FF in human and
in animal models, but they did not show their exact origin
and the role that they could perform inside ovarian follicles
(1315). The aims of this paper were [1] to nd well-
represented miRNAs in human FF whose detection was not
affected by plasma or cellular contamination; and [2] to
ascertain whether these miRNAs could regulate biologically
important pathways inside follicular cells. To identify upre-
gulated miRNAs in FF, we compared by real-time reverse
transcriptasepolymerase chain reaction (PCR) the expres-
sion prole of 384 miRNAs from FF and plasma collected
from the same women. We then isolated nanosized vesicles
and their miRNA cargo from FF and conrmed that [1] they
showed surface exosomal markers; [2] they carried miRNAs
upregulated in total FF. By using a computational approach,
we veried that nine FF miRNAs are able to regulate path-
ways involved in follicular maturation. We propose that the
identied miRNAs perform an important role inside ovarian
follicles and that their altered expression could be associated
with reproductive disorders, as has been demonstrated for
intracellular ovarian miRNAs (16). Accordingly, we propose
that these miRNAs represent useful new molecular markers
of oocyte quality.
MATERIALS AND METHODS
Patients
FF and plasma samples were collected from 15 healthy
women selected by an IVF center (Servizio di PMA, Azienda
Ospedaliera Cannizzaro, Catania) who had undergone intra-
cytoplasmic sperm injection. We selected women younger
than 35 years whose primary infertility was due to a male
2VOL. -NO. -/-2014
ORIGINAL ARTICLE: REPRODUCTIVE BIOLOGY
factor; this excluded pathologies that could inuence oocyte
quality (e.g., endometriosis, polycystic ovaries, and ovarian
insufciency). Moreover, we excluded from the study heavy
smokers and overweight women. Among the 15 enrolled
women, there were six pregnancies: two miscarriages and
ve live births (one twin birth). The patients signed an
informed consent to participate in the research project, which
included the use of collected FF and plasma. The study was
exempted from Institutional Review Board approval because
patients were included in the IVF program. Accordingly,
there were no identiers linking individuals to the samples
and no additional treatment or use of personal data was
necessary.
Ovarian Stimulation and Sample Collection
Hormone stimulation was performed by treatment with
GnRH agonists (triptorelin or buserelin), followed by ovarian
stimulation with recombinant FSH and hMG. Stimulation
was monitored using both serum and E
2
concentrations as
well as ultrasound measurements of follicle numbers and di-
ameters. When follicles had reached a diameter >18 mm and
serum E
2
concentration per follicle reached 150200 ng/L,
ovulation was induced with 10,000 IU of hCG. Transvaginal
ultrasoundguided aspiration of ovarian follicles was per-
formed 3436 hours after hCG injection. FF samples were
centrifuged for 200at 2,800 rpm at 4C to remove follicular
cell residues and any blood traces; the supernatant was
immediately transferred into a clean polypropylene tube
and stored at 20C for further analysis. Samples with
massive blood contamination were excluded from further
analysis. Blood samples were collected in commercially
available EDTA-containing tubes. Cells were removed from
plasma by centrifugation for 100at 1,800 rpm at 4C. The
resulting supernatant was immediately transferred into a
clean polypropylene tube using a Pasteur pipette and stored
at 20C. We recovered about 20 mL of FF from each
woman. FF of individual follicles was kept separated until
decumulation of the oocytes to collect only the FF in which
nuclear mature oocytes (metaphase II) had been identied. To
obtain a homogenous pool of samples, 18 mL of FF from
each woman was mixed and 270 mL of pooled FF was
obtained. We used three aliquots of 400 mL for miRNA puri-
cation from total FF, while two aliquots of 120 mL for exo-
some characterization and exosomal miRNA purication.
From the same cohort of 15 women, 5 mL of blood from
each woman was collected, 2 mL of plasma was obtained,
and 1 mL was used to make a pool of 15 mL. We used three
aliquots of 400 mL for miRNA purication.
Exosome Purication and Characterization
FF exosomes were isolated and characterized according to a
previously published protocol with minor modications
(17). Two different isolations were performed: the rst for
exosome molecular characterization and the second for
miRNA extraction. For each, a volume of 120 mL of pooled
samples from the 15 women was aliquoted in 50-mL tubes
and centrifuged at 3,000 rpm for 15 minutes at 4C to pellet
debris. The supernatant was transferred to ultracentrifuge
tubes and ultracentrifuged at 16,500 gfor 30 minutes at
4C, followed by ltration through a 0.2-mm syringe lter.
Finally, exosomes were pelleted by ultracentrifugation at
120,000 gfor 70 minutes at 4C in a Beckman Optima
L-100 XP ultracentrifuge using a Ti70 rotor (Beckman
Coulter). Exosome pellets were resuspended in Trizol (Invitro-
gen) for RNA isolation or in phosphate-buffered saline (PBS)
for nanosight and uorescence-activated cell sorter analysis.
For the nanoparticle tracking analysis (NTA), samples were
diluted with sterile PBS following the manufacturer's instruc-
tions. Samples (approximately 300 mL) were injected into the
LM10 unit with a 1-mL sterile syringe. Capturing and
analyzing settings were manually set according to the proto-
col. Using the NanoSight LM10 instrument, vesicles were
visualized by laser light scattering, and Brownian motion of
these vesicles was captured on video. The number of tracks al-
ways exceeded 200, and ve size distribution measurements
were taken for each sample. Recorded videos were then
analyzed with NTA software, which provided high-
resolution particle size distribution proles and concentration
measurements of the vesicles in solution. Flow cytometry
analysis was performed according to a previously published
protocol (18). Aldehyde/sulfate latex beads of 4 mm (Invitro-
gen) were serially diluted with PBS to reach a nal concentra-
tion of 2,900 beads/mL. Vesicles from FF samples were
incubated with 25 mL of diluted latex beads at 37C for 30 mi-
nutes and then overnight at 4C with gentle agitation. After
one wash in PBS, exosome-coated beads were incubated
with 20 mL 1 M glycine (Sigma-Aldrich) for 30 minutes at
20C to block any remaining available binding sites. After
two washes in PBS with 1% fetal bovine serum (FBS),
exosome-coated beads were stained with phycoerythrin-
(PE-) conjugated CD63, CD81, or CD9 antibodies or isotype
control (BD Biosciences Pharmingen) and incubated for
1 hour at 4C with gentle agitation. After the third wash in
PBS/FBS 1% solution, samples were resuspended in PBS
and analyzed on FACSCantoII (Becton Dickinson) with
FlowJo software (TreeStar, Inc.).
miRNA Isolation
All experiments were performed in triplicate. Three aliquots of
400 mL of FF and plasma samples were thawed completely on
ice. miRNA isolation was performed by using a Qiagen miR-
Neasy Mini Kit (Qiagen GmbH), according to the Qiagen Sup-
plementary Protocol for the purication of small RNAs from
serum and plasma and nally eluted in a 30-mL volume of
elution buffer. Total RNA was isolated from three aliquots
of FF exosomes using Trizol reagent (Invitrogen), according
to the manufacturer's instructions. Before precipitating the
RNA with isopropyl alcohol, 20 mg RNase-free glycogen (In-
vitrogen) as carrier was added to the aqueous phase and the
samples were stored for 16 hours at 80C. RNA pellets
were dissolved in RNase-free water. Detection and quality
control of RNA were performed using an Agilent 2100 Bio-
analyzer (Agilent Technologies Sweden AB) and 2100 Expert
software.
VOL. -NO. -/-2014 3
Fertility and Sterility®
Reverse Transcription, Preamplication and
MicroRNA Proling
Expression proles of 384 miRNAs were investigated by using
TaqMan low-density array technology (Applied Biosystems)
in a 7900HT fast real time PCR system (Applied Biosystems).
This technology is highly specic, and it allows the amplica-
tion of only mature miRNAs (19).
RNA (3 mL of each sample) was reverse-transcribed (RT)
by using the TaqMan MicroRNA Reverse Transcription Kit
and Megaplex RT Primer Pool A (Applied Biosystems). Four
microliters of RT products were preamplied with TaqMan
PreAmp Master Mix 2and Megaplex PreAmp Primers
Pool A (Applied Biosystems). Preamplication products
were subsequently diluted in 75 mL of RNase free puried wa-
ter. Nine microliters of each amplied product was mixed
with Universal Master Mix II, no uracil-N glycoslyase
(Applied Biosystems), and loaded into TaqMan Human Micro-
RNA Array A cards (Applied Biosystems), according to the
manufacturer's instructions.
Expression Data Analysis
Housekeeping gene detection. No data are available on reli-
able reference genes to be used for miRNA normalization in
FF. Accordingly, we used DataAssist version 3 software
(Applied Biosystems) and the geNorm algorithm (http://
medgen.ugent.be/jvdesomp/genorm/) to identify appro-
priate endogenous controls. GeNorm is based on the geomet-
ric average of multiple internal control genes, and it is
especially useful for normalization of data from a large and
unbiased set of genes (e.g., miRNA expression proling) (20,
21). DataAssist is a data analysis tool that is useful to
compare samples when using the comparative CT (DDCT)
method for calculating relative quantitation of gene
expression. In a rst step, three miRNAs common to two
programs and having a low score in both (the lower the
score, the better the normalization) were selected as
housekeeping genes. The miRNAs miR-25, miR-28-3p, and
miR-145 were identied for comparison between FF and
plasma, and miR-126, miR-28-3p, and miR-145 were identi-
ed for exosomes versus plasma. The DCt values were
independently calculated by using the three selected house-
keeping genes, for each sample.
Identication of differentially expressed miRNAs. To iden-
tify differentially expressed (DE) miRNAs, three different
signicance analysis of microarrays (SAM; http://www.
tm4.org) tests (Tusher, 5th percentile and minimum S value)
were used, applying a two-class unpaired test among DCt
and using a Pvalue based on all possible permutations:
imputation engine, K-nearest neighbors (10 neighbors); false
discovery rate, <.05. We considered as DE miRNAs only
those with three housekeeping genes in common after at least
two SAM tests.
Relative quantication and selection of upregulated
miRNAs. The DCt values of DE miRNAs were used to calcu-
late relative quantity (RQ) values as a natural logarithm of
2
DDCt
and the average of DCt of each plasma sample as cali-
brator. miRNAs with a natural logarithm of RQ values R2.5
were considered as upregulated (highly expressed) in FF or
exosomes compared with plasma. For each DE miRNA, nine
RQ values (three for each housekeeping gene) were obtained
that were reciprocally comparable, and RQ values obtained
with the housekeeping miR145 were used to perform addi-
tional statistical analysis according to Livak and Schmittgen
(22). The mean fold change was calculated as a natural loga-
rithm of RQ values, and the error was estimated by evaluating
the 2
DDCt
equation using DDCt plus SD and DDCt minus
SD (22).
miRNA Target Prediction
Targets of upregulated miRNAs were explored by using a
combination of different approaches. By interpolation among
the highest numbers of 11 prediction tools, the rst series of
experimentally validated and predicted targets were extracted
from miRecords (http://mirecords.biolead.org)(23).To
improve prediction, TarBase, version 5.0 (http://www.dia
na.pcbi.upenn.edu/tarbase), and miRTarBase, version 3.5
(http://mirtarbase.mbc.nctu.edu.tw), were also used. These
are two databases that have accumulated more than 50,000
experimentally supported targets. An additional ltering
was performed by using starBase, version 2.0, a database
for predicted miRNA-target interactions that are overlapped
with data from Argonaute cross-linked immunoprecipita-
tion-sequencing (CLIP-Seq; http://starbase.sysu.edu.cn).
CLIP-Seq experiments are based on cross-linking between
RNA and proteins, followed by immunoprecipitation coupled
with high-throughput sequencing. The application of this
technique is useful for the identication of miRNA binding
sites. Finally, our list was enriched by retrieving from the
literature specic information on validated miRNA targets.
Gene Ontology (GO) and Pathway Analysis
The analysis was independently performed on miRNAs upre-
gulated in total FF and miRNAs upregulated in exosomes. The
GO functional classication of miRNA targets was focused
on the biological processes, in which target mRNAs are
involved. For this analysis, the g:GOSt (GO statistics) tool
of g:Proler (http://biit.cs.ut.ee/gproler/) was used and
Bonferroni correction for a Pvalue of .05 was applied.
Pathway analysis was performed by using the prediction soft-
ware DIANA-microT-4.0 (beta version; http://diana.cslab.
ece.ntua.gr/pathways/). DIANA-miRPath uses miRNA targets
that have been predicted with high accuracy based on
DIANA-microT-CDS. In our analysis, we input upregulated
miRNAs, and the software performs an enrichment analysis
of multiple miRNA targets genes comparing the set of miRNA
targets to all known KEGG pathways. It retrieved signaling
pathways enriched with gene targets of the miRNAs sorting
them by Pvalues (P-value threshold .05 and microthreshold
0.8) (24).
RESULTS
Exosome Characterization and miRNA Isolation
The nanovesicles isolated from human FF had an average
size of 40 nm in diameter, which is consistent with the
4VOL. -NO. -/-2014
ORIGINAL ARTICLE: REPRODUCTIVE BIOLOGY
characteristic size range of exosomes (25) (Fig. 1A). More-
over, our samples were positive for the tetraspanin proteins
CD63 and CD81, which are known to be enriched in exo-
somes (25) (Fig. 1B). RNAs isolated from exosomes were
small-sized RNAs and lacked bands corresponding to
cellular 18S and 28S ribosomal RNAs (Fig. 1C).
Expression Prole of mRNAs
We identied 37 miRNAs upregulated in human FF compared
with in plasma (Fig. 2). Specically, 15 miRNAs were found to
be upregulated in total FF compared with in plasma; 10 miR-
NAs were carried by exosomes, while ve were not (Fig. 2).
Twenty-two miRNAs were present exclusively in exosomes.
FIGURE 1
Characterization of exosomes. (A) Nanoparticle tracking measurement of particle size and concentration. Pelleted fractions from FF are vesicles
whose diameter size ranged from 10 to 100 nm, with a peak size between 30 and 50 nm. Average vesicle size was 40 nm. Particle size is
consistent with exosome size range. Error bars represent SDs obtained from ve measurements of the same sample. (B) Flow cytometry
detection of surface molecules on exosomes from FF. Exosomes, bound to latex beads, were immunostained by using monoclonal antibodies
against the tetraspanins CD9, CD63, and CD81 and analyzed by ow cytometry. The antibodies were compared with their appropriate isotype
control IgG1. FF exosomes were positive for CD63 and CD81. (C) Exosomal RNA analyzed using a Bioanalyzer. RNA pattern, isolated from FF
exosomes, is visualized in Bioanalyzer as electrophoretic data and a gel-like image. The results show that exosomes are enriched in small RNAs
and contain no 18S and 28S ribosomal RNAs. Abbreviations: SSC-H ¼side scatter height; FU ¼uorescence; nt ¼nucleotides.
Santonocito. Exosomal microRNAs in human follicular uid. Fertil Steril 2014.
VOL. -NO. -/-2014 5
Fertility and Sterility®
FIGURE 2
MicroRNA expression in total FF and exosomes compared with plasma. Black bars show the 10 upregulated miRNAs in total FF and in exosomes.
Gray bars represent ve upregulated miRNAs exclusively in the total FF, and light gray bars represent 22 upregulated miRNAs in exosomes only. Fold
change is shown as the natural logarithm of RQ values, which were calculated through the 2
DDCt
method (by using miR-145 as an endogenous
control and plasma as the calibrator sample) and respective SD values.
Santonocito. Exosomal microRNAs in human follicular uid. Fertil Steril 2014.
6VOL. -NO. -/-2014
ORIGINAL ARTICLE: REPRODUCTIVE BIOLOGY
They were undetectable in total FF because their relative con-
centration was higher in puried exosomes compared with
total FF (Fig. 2). The ve miRNAs detected only in total FF
and absent in exosomes could be free circulating miRNAs
associated with protein complexes (Fig. 2). Interestingly,
miR-508-3p, whose fold change value was among the high-
est, seems to be follicle specic, since we could not detect it
in plasma (Fig. 2). Among the 37 miRNAs previously cited,
we found miR-10b, miR-29a, miR-99a, miR-125b, miR-132,
miR-202, miR-212, and miR-874, which had been reported
to be highly expressed in granulose or in cumulus cells in hu-
mans and in mice (2628).
Genomic Analysis
Genomic analysis showed that 22 miRNAs (miR-29a, miR-99a,
miR-100, miR-125b, miR-132, miR-134, miR-193b,miR-203,
miR-212, miR-214, miR-323-3p, miR-337-5p, miR-365,
miR-370, miR-410, miR-449a, miR-489, miR-493, miR-503,
miR-508-3p, miR-542-5p, and miR-654-3p) are localized in
13 different clusters; a number of the upregulated miRNAs
were part of the same cluster (Supplemental Table 1). Interest-
ingly, miR-134, miR-323-3p, miR-410, and miR-654-3p were
localized within the large miR-379/miR-656 cluster, which is
exclusively present in placental mammals and involvedin em-
bryonic development (29). We found that genes encoding 13
miRNAs were located inside genes encoding proteins, while
those for the other 24 miRNAs were located in extragenic re-
gions or inside noncoding genes (Supplemental Table 1).
GO and Pathway Analysis
GO analysis of validated and predicted targets was indepen-
dently performed on miRNAs upregulated in total FF and
miRNAs upregulated in exosomes. GOs were used to func-
tionally categorize miRNA target genes that were identied
in a range of biological processes. The GO terms ranged
from one to three levels of detail, and only signicant GO
terms are reported. The maximum Pvalue was .048.
Figure 3A shows the signicant GOs, and in the x-axis the
negative log
10
(Pvalue) is reported. The most signicant bio-
logical processes involving the targets of our upregulated
miRNAs are developmental, regulation of cellular process,
cell differentiation, and cell communication (Fig. 3A).
Pathway analysis showed that WNT, MAPK, ErbB, and
TGFbsignaling pathways, which are shared by total FF and
exosome miRNAs, are the most signicant, with a negative
log
10
(Pvalue) between 11.04 and 4.12 (Fig. 3B). Figure 3C
specically shows the miRNAs involved in the regulation of
the signicant pathways presented in Figure 3B. It can be
observed that WNT and MAPK signaling pathways are clearly
targeted by most miRNAs with a very small Pvalue. On the
other hand, it can be seen that some miRNAs (e.g., miR-132,
miR-212, and miR-214) could regulate at least eight different
FIGURE 3
GO analysis based on highly expressed miRNA targets and heat-map representations of signaling pathways. (A) GO terms within the biological
process category for highly expressed miRNA targets in total FF (red bars) and in exosomes (blue bars) are shown. The x-axis represents the
log
10
(Pvalue); the signicance was determined by the adjusted Bonferroni correction. (B) Signaling pathway heat map regulated by total FF
and exosomal miRNAs. Gray boxes indicate that the pathway is not signicant. (C) miRNAs versus pathways. The probability values are reported
as log
10
(Pvalue) for both panels. Abbreviations: EXO ¼exosomes; P ¼plasma.
Santonocito. Exosomal microRNAs in human follicular uid. Fertil Steril 2014.
VOL. -NO. -/-2014 7
Fertility and Sterility®
pathways (Fig. 3C). To perform a more accurate analysis, our
computational approach was validated using literature data.
Its application conrmed that miR-29a, miR-99a, miR-100,
miR-132, miR-212, miR-214, miR-218, miR-508-3p, and
miR-654-3p are particularly interesting. In fact, their expres-
sion inside granulose or cumulus cells and their regulatory
role were demonstrated by their targeting mRNAs involved
in ovarian physiology and pathology (14, 2641).
DISCUSSION
In this paper, we describe the identication of 37 miRNAs up-
regulated in human FF compared with plasma; our data
demonstrate that 32 of them are carried by exosomes
(Fig. 2). In fact, the average size of isolated microvescicles
was 40 nm in diameter, and they were positive for the tetra-
spanin proteins CD63 and CD81, which are known to be en-
riched in exosomes (Fig. 1A and B). The absence of marker
CD9 could be due to the fact that exosomes may express
different surface markers, based on their cellular source of
origin and their specic physiological state (6, 17). The
presence of miRNAs within FF vesicles was recently
described in humans and in animal models, but no data
were presented to support their identication as exosomes;
furthermore, absence of miRNA target identication
precluded characterization of downstream networks and
corresponding biomolecular functions (1315). We studied
miRNAs upregulated in FF compared with plasma; this
allowed us to hypothesize that they were not released by
blood cells but specically by ovarian follicle cells. The
presence of exosome miRNAs means that an unexplored
communication tool could exist inside ovarian cells. By
applying many different computational tools, we performed
multiple searches in public databases and literature data to
conrm that identied miRNAs could carry out their
biological role inside ovarian follicles. This approach
allowed us to conrm that miR-29a, miR-99a, miR-100,
miR-132, miR-212, miR-214, miR-218, miR-508-3p, and
miR-654-3p are novel molecular regulators of ovarian follicle
pathways. In fact, as we will explain more in detail later, some
of these miRNAs or their host genes are expressed in granulose
or cumulus cells, and some of their validated targets per-
formed their role inside follicular cells (2641). Genomic
analysis showed that some miRNAs belong to the same
cluster, supporting the fact that they are all highly
expressed in FF at the same time (Supplemental Table 1). In
fact, clustered miRNAs arise from a common primary
transcript and are expected to have similar expression
proles. Interestingly, miR-132 and miR-212, located in a
cluster at 17p13.3, were found highly upregulated in mouse
mural granulose cells after LH/hCG induction (26). In addi-
tion, intronic miRNAs are usually expressed together with
the host gene: miR-218 is located within the slit homolog 2
(Drosophila, SLIT2) gene, which encodes the secreted SLIT
glycoprotein roundabout (ROBO) receptors ligand. SLIT and
ROBO have been proposed to be regulated by steroid hor-
mones and to modulate physiological cell functions in the
ovary (39). GO and pathway analysis demonstrated that the
targets of upregulated miRNAs could perform their role inside
ovarian follicles during follicular maturation. In fact, we iden-
tied biological processes involved in development, in cell
differentiation, and in cell communication (Fig. 3A). We
found that most of the upregulated miRNAs are involved in
the regulation of the WNT, MAPK, ErbB, and TGFbsignaling
pathways (Fig. 3B and C). These pathways have been described
inside the different cell compartments of the ovarian follicle
and perform a critically important role for follicular develop-
ment, meiotic resumption, and subsequent ovulation (4245).
WNT proteins are secreted extracellular signaling molecules
that activate the G proteincoupled receptors Frizzled (Fz). It
has been demonstrated that WNTs and Fz are expressed at
specic stages of follicular growth and luteinization, and
their role could be important for the growth and
development of ovarian follicles (42). The MAPK pathway,
activated by FSH and LH trigger, stimulates granulose cell
proliferation and cumulus expansion (43). Its activation in
cumulus cells appears to require the permissive effect of
TGFbfamily factors produced by the oocyte, such as growth
differentiation factor 9 and bone morphogenetic protein 15.
On the other hand, the MAPK pathway and the ErbB
pathway promote meiosis resumption in the oocyte by a
decrease of cAMP (44, 45). The outcome of the complex
interaction among the different molecules involved in these
pathways is the elimination of meiosis-inhibiting factors
and/or the accumulation or activation of oocyte maturation
signals (44). Interestingly, we found that miR-29a, miR-99a,
miR-100, miR-132, miR-212, miR-214, miR-218, miR-508-
3p, and miR-654-3p could trigger meiosis resumption by
negatively regulating genes encoding for follicle maturation
inhibiting factors (46, 47) (Fig. 4). We present a schematic
view of the pathways inside follicular cells and oocytes, and
we pinpoint where our miRNAs could perform their action
(Fig. 4).
miR-132, miR-212, and miR-214 have phosphatidylino-
sitol 3,4,5-trisphosphate 3-phosphatase (PTEN) as a validated
target (37, 38). PTEN performs an important role in the
regulation of primordial follicle survival and activation,
cyclic follicular recruitment, ovulation, and meiosis
resumption; nevertheless, the inhibitory mechanisms that
are involved in PTEN silencing remain unknown (47). Our
miRNAs represent the best candidates for its silencing and
the resulting MAPK activation mediated by v-akt murine
thymoma viral oncogene homolog 1 (AKT1). Intriguingly, in
ovarian cancer, miR-214 induces cell survival and cisplatin
resistance by targeting PTEN (38). Deregulation of PTEN in-
hibitors could be associated with reproductive disorders, as
it has been shown that PTEN knockout mice exhibit follicular
depletion in young adulthood and polycystic ovarian failure
(48). It was recently demonstrated that PTEN downregulation
may inhibit the formation of polycystic ovaries in a rat model
(49). Moreover, PTEN inhibition by miR-132, miR-212, and
miR-214 could be used as a potential tool to trigger the acti-
vation and enhance the survival rate of primordial follicles in
cultured ovarian cortical tissue in vitro (50).
Mechanistic target of rapamycin (MTOR) is a validated
target of miR-99a and miR-100, and tuberous sclerosis 1
(TSC1) is a predicted target of miR-508. The altered regulation
of miR-99a (in cluster with let-7c, which is highly expressed
8VOL. -NO. -/-2014
ORIGINAL ARTICLE: REPRODUCTIVE BIOLOGY
in cumulus cells [27]), and miR-100 is involved in ovarian
cancer through MTOR regulation (33, 34). MTOR inhibition,
one of the necessary steps for follicular maturation, could
be mediated by these miRNAs (47). miR-654-3p inhibits
cyclin-dependent kinase inhibitor 1 (P21), a negative regu-
lator of cell cycle progression (40). It has been demonstrated
that P21 inhibition induces oocyte maturation (41). Another
brake for cell cycle progression and meiosis resumption is
FIGURE 4
Ovarian follicle, signaling pathways, and FF miRNAs. Model summarizing crucial LH- and FSH-induced signaling pathways resulting in oocyte
meiosis resumption. Hormone-receptor binding activates the AKT,PKA, and MAPK pathways and then induces the EGF-like factors' expression
in granulosa cells. EGF-like factors act on EGFR to activate the RAS-MAPK pathway, contributing to phosphorylation of Cx-43 and the resulting
decrease in gap junction permeability. These events would reduce NPR2 activity, induce PDE1 activation, and decrease cGMP levels in oocytes.
Within the oocyte, activated PDE3A degrades cAMP and meiosis resumes. Identied miRNAs could act along these pathways and regulate the
processes of follicular development and meiotic resumption. Abbreviations: FSHR ¼FSH receptor; LHR ¼LH receptor; AC ¼adenylate cyclase;
cAMP ¼cyclic adenosine 30,50-monophosphate; PKA ¼cAMP-dependent protein kinase; CREB ¼cAMP responsive element binding protein;
MAPK ¼mitogen-activated protein kinase; EGF ¼epidermal growth factor; EGFR ¼EGF receptor; PI3K ¼phosphatidylinositol 3 kinase; PIP2 ¼
phosphatidylinositol 4, 5-bisphosphate; PIP3 ¼phosphatidylinositol-3,4,5-trisphosphate; AKT ¼v-akt murine thymoma viral oncogene
homolog (also known as PKB); CDK ¼cyclin-dependent kinase; E2F ¼E2F transcription factor; PLC ¼phospholipase C; IP3 ¼inositol 1,4,5-
trisphosphate; DAG ¼1,2-diacylglycerol; PKC ¼protein kinase C; Cx-43 ¼connexin 43; NPR2 ¼natriuretic peptide receptor B/guanylate
cyclase B; cGMP ¼cyclic guanosine 30,50-monophosphate; PDE3A ¼phosphodiesterase 3A, cGMP-inhibited.
Santonocito. Exosomal microRNAs in human follicular uid. Fertil Steril 2014.
VOL. -NO. -/-2014 9
Fertility and Sterility®
represented by retinoblastoma protein (RB1). We found
that miR-132 and miR-212 have as a validated target this
tumor suppressor (36) (Fig. 4). We found that DNA
(cytosine-5-)-methyltransferase 3 alfa (DNMT3A) and DNA
(cytosine-5-)-methyltransferase 3 beta (DNMT3B) are regu-
lated by miR-29a and miR-132 (30, 37), both of which are
found highly expressed in granulose and cumulus cells (26,
27). It is known that DNA methylation establishment is
dynamically controlled during oocyte growth and that it
may contribute to gene regulation in the oocyte by marking
specic genes for activity in the embryo (51). DNMT3
silencing, mediated by miR-29a and miR-132, could explain
the hypomethylation found in preimplantation embryos and
also in the inner-cell mass (52). Recent studies have suggested
a link between the use of assisted reproductive technology
(ART) and the increased incidence of normally rare imprinting
disorders (52). In agreement with literature data about the
complexity of gene expression regulation mediated by miR-
NAs, we found that several miRNAs could act on the same
mRNA (e.g., miR-132, miR-212, and miR214 are able to regu-
late PTEN) and that a single miRNA may target different
mRNAs (miR-132 is able to regulate PTEN, RB1, and
DNMT3A; Fig. 4). We propose that identied miRNAs are syn-
thesized by ovarian follicle cells and that they could be trans-
ferred to recipient cells through FF exosomes. It is known that
exocytosis and endocytosis mechanisms have been described
in granulose cells and oocytes (5356), but further studies are
necessary to identify specic exosome surface markers able to
discriminate the vesicles released by somatic follicular cells or
by the oocyte and to ascertain whether their cargo acts in an
autocrine or paracrine manner.
Conclusions
This study identied exosomal miRNAs highly represented in
human FF and involved in the regulation of ovarian follicular
pathways. We believe that, as it has been demonstrated for
intracellular ovarian miRNAs, their altered expression could
be associated with reproductive disorders. We propose that
these miRNAs could represent noninvasive molecular
markers of oocyte quality in ART.
Acknowledgments: The authors thank Professor Antony
Bridgewood for language revision of the manuscript.
REFERENCES
1. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell
2004;116:28197.
2. Cortez MA, Bueso-Ramos C, Ferdin J, Lopez-Berestein G, Sood AK,
Calin GA. MicroRNAs in body uidsthe mix of hormones and biomarkers.
Nat Rev Clin Oncol 2011;8:46777.
3. Croce CM. Causes and consequences of microRNA dysregulation in cancer.
Nat Rev Genet 2009;10:70414.
4. Weber JA, Baxter DH, Zhang S, Huang DY, Huang KH, Lee MJ, et al. The
microRNA spectrum in 12 body uids. Clin Chem 2010;56:173341.
5. Valadi H, Ekstr
om K, Bossios A, Sj
ostrand M, Lee JJ, L
otvall JO. Exosome-
mediated transfer of mRNAs and microRNAs is a novel mechanism of
genetic exchange between cells. Nat Cell Biol 2007;9:6549.
6. Escola JM, Kleijmeer MJ, Stoorvogel W, Grifth JM, Yoshie O, Geuze HJ.
Selective enrichment of tetraspan proteins on the internal vesicles of
multivesicular endosomes and on exosomes secreted by human B-lympho-
cytes. J Biol Chem 1998;273:201217.
7. Urbanelli L, Magini A, Buratta S, Brozzi A, Sagini K, Polchi A, et al. Signaling
pathways in exosomes biogenesis, secretionand fate. Genes 2013;4:15270.
8. Hu G, Drescher KM, Chen XM. Exosomal miRNAs: biological properties and
therapeutic potential. Front Genet 2012;3:56.
9. Ragusa M, Statello L, Maugeri M, Majorana A, Barbagallo D, Salito L, et al.
Specic alterations of the microRNA transcriptome and global network
structure in colorectal cancer after treatment with MAPK/ERK inhibitors. J
Mol Med (Berl) 2012;90:142138.
10. Barbagallo D, Piro S, Condorelli AG, Mascali LG, Urbano F, Parrinello N, et al.
miR-296-3p, miR-298-5p and their downstream networks are causally
involved in the higher resistance of mammalian pancreatic acells to
cytokine-induced apoptosis as compared to bcells. BMC Genomics 2013;
14:62.
11. Revelli A, Delle Piane L, Casano S, Molinari E, Massobrio M, Rinaudo P. Follic-
ular uid content and oocyte quality: from single biochemical markers to
metabolomics. Reprod Biol Endocrinol 2009;7:40.
12. Rodgers RJ, Irving-Rodgers HF. Formation of the ovarian follicular antrum
and follicular uid. Biol Reprod 2010;82:10219.
13. da Silveira JC, Veeramachaneni DN, Winger QA, Carnevale EM, Bouma GJ.
Cell-secreted vesicles in equine ovarian follicular uid contain miRNAs and
proteins: a possible new form of cell communication within the ovarian fol-
licle. Biol Reprod 2012;86:71.
14. Sang Q, Yao Z, Wang H, Feng R, Wang H, Zhao X, et al. Identication of mi-
croRNAs in human follicular uid: characterization of microRNAs that
govern steroidogenesis in vitro and are associated with polycystic ovary syn-
drome in vivo. J Clin Endocrinol Metab 2013;98:306879.
15. Sohel MM, Hoelker M, Noferesti SS, Salilew-Wondim D, Tholen E, Looft C,
et al. Exosomal and non-exosomal transport of extra-cellular microRNAs in
follicular uid: implications for bovine oocyte developmental competence.
PLoS One 2013;8:e78505.
16. Baley J, Li J. MicroRNAs and ovarian function. J Ovarian Res 2012;5:8.
17. L
asser C, Eldh M, L
otvall J. Isolation and characterization of RNA-containing
exosomes. J Vis Exp 2012;59:e3037.
18. Wahlgren J, Karlson TD, Brisslert M, Vaziri Sani F, Telemo E, Sunnerhagen P,
et al. Plasma exosomes can deliver exogenous short interfering RNA to
monocytes and lymphocytes. Nucleic Acids Res 2012;40:e130.
19. Chen C, Ridzon DA, Broomer AJ, Zhou Z, Lee DH, Nguyen JT, et al. Real-time
quantication of microRNAs by stem-loop RT-PCR. Nucleic Acids Res 2005;
33:e179.
20. Vandesompele J, De Preter K, Pattyn F, Poppe B, Van Roy N, De Paepe A,
et al. Accurate normalization of real-time quantitative RT-PCR data by geo-
metric averaging of multiple internal control genes. Genome Biol 2002;3:
R34.
21. Mestdagh P, Van Vlierberghe P, De Weer A, Muth D, Westermann F,
Speleman F, et al. A novel and universal method for microRNA RT-qPCR
data normalization. Genome Biol 2009;10:R64.
22. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-
time quantitative PCR and the 2(delta delta C(T)) method. Methods 2001;
25:4028.
23. Xiao F, Zuo Z, Cai G, Kang S, Gao X, Li T. miRecords: an integrated resource
for microRNA-target interactions. Nucleic Acids Res 2009;37:D10510.
24. Vlachos IS, Kostoulas N, Vergoulis T, Georgakilas G, Reczko M,
Maragkakis M, et al. DIANA miRPath v.2.0: investigating the combinatorial
effect of microRNAs in pathways. Nucleic Acids Res 2012;40:W498504.
25. Th
ery C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and
function. Nat Rev Immunol 2002;2:56979.
26. Fiedler SD, Carletti MZ, Hong X, Christenson LK. Hormonal regulation of
MicroRNA expression in periovulatory mouse mural granulosa cells. Biol Re-
prod 2008;79:10307.
27. Assou S, Al-edani T, Haouzi D, Philippe N, Lecellier CH, Piquemal D, et al.
MicroRNAs: new candidates for the regulation of the human cumulus-
oocyte complex. Hum Reprod 2013;28:303849.
28. Velthut-Meikas A, Simm J, Tuuri T, Tapanainen JS, Metsis M, Salumets A.
Research resource: small RNA-seq of human granulosa cells reveals miRNAs
in FSHR and aromatase genes. Mol Endocrinol 2013;27:112841.
10 VOL. -NO. -/-2014
ORIGINAL ARTICLE: REPRODUCTIVE BIOLOGY
29. Glazov EA, McWilliam S, Barris WC, Dalrymple BP. Origin, evolution, and
biological role of miRNA cluster in DLK-DIO3 genomic region in placental
mammals. Mol Biol Evol 2008;25:93948.
30. Denis H, Ndlovu MN, Fuks F. Regulation of mammalian DNA methyltrans-
ferases: a route to new mechanisms. EMBO Rep 2011;12:64756.
31. Takada S, Berezikov E, Choi YL, Yamashita Y, Mano H. Potential role of miR-
29b in modulation of Dnmt3a and Dnmt3b expression in primordial germ
cells of female mouse embryos. RNA 2009;15:150714.
32. Doghman M, El Wakil A, Cardinaud B, Thomas E, Wang J, Zhao W, et al.
Regulation of insulin-like growth factor-mammalian target of rapamycin
signaling by microRNA in childhood adrenocortical tumors. Cancer Res
2010;70:466675.
33. Nam EJ, Yoon H, Kim SW, Kim H, Kim YT, Kim JH, et al. MicroRNA expression
proles in serous ovarian carcinoma. Clin Cancer Res 2008;14:26905.
34. Nagaraja AK, Creighton CJ, Yu Z, Zhu H, Gunaratne PH, Reid JG, et al. A link
between mir-100 and FRAP1/mTOR in clear cell ovarian cancer. Mol Endo-
crinol 2010;24:44763.
35. Wong HK, Veremeyko T, Patel N, Lemere CA, Walsh DM, Esau C, et al. De-
repression of FOXO3a death axis by microRNA-132 and -212 causes
neuronal apoptosis in Alzheimer's disease. Hum Mol Genet 2013;22:
307792.
36. Park JK, Henry JC, Jiang J, Esau C, Gusev Y, Lerner MR, et al. miR-132 and
miR-212 are increased in pancreatic cancer and target the retinoblastoma
tumor suppressor. Biochem Biophys Res Commun 2011;406:51823.
37. Miller BH, Zeier Z, Xi L, Lanz TA, Deng S, Strathmann J, et al. MicroRNA-132
dysregulation in schizophrenia has implications for both neurodevelopment
and adult brain function. Proc Natl Acad Sci U S A 2012;109:312530.
38. Yang H, Kong W, He L, Zhao JJ, ODonnell JD, Wang J, et al. MicroRNA
expression proling in human ovarian cancer: miR-214 induces cell survival
and cisplatin resistance by targeting PTEN. Cancer Res 2008;68:42533.
39. Dickinson RE, Duncan WC. The SLIT-ROBO pathway: a regulator of cell func-
tion with implications for the reproductive system. Reproduction 2010;139:
697704.
40. Wu S, Huang S, Ding J, Zhao Y, Liang L, Liu T, et al. Multiple microRNAs
modulate p21Cip1/Waf1 expression by directly targeting its 3' untranslated
region. Oncogene 2010;29:23028.
41. Kovac C, Clie L, Morin J, Friedman FK, Robinson R, Chung DL, et al. Plasmid
expression of a peptide that selectively blocks oncogenic ras-p21-induced
oocyte maturation. Cancer Chemother Pharmacol 2001;48:914.
42. Richards JS, Russell DL, Ochsner S, Hsieh M, Doyle KH, Falender AE, et al.
Novel signaling pathways that control ovarian follicular development, ovula-
tion, and luteinization. Recent Prog Horm Res 2002;57:195220.
43. Conti M, Hsieh M, Zamah AM, Oh JS. Novel signaling mechanisms in the
ovary during oocyte maturation and ovulation. Mol Cell Endocrinol 2012;
356:6573.
44. Zhang M, Ouyang H, Xia G. The signal pathway of gonadotrophins-induced
mammalian oocyte meioticresumption. Mol Hum Reprod 2009;15:399409.
45. Russell DL, Robker RL. Molecular mechanisms of ovulation: co-ordination
through the cumulus complex. Hum Reprod Update 2007;13:289312.
46. Sun QY, Miao YL, Schatten H. Towards a new understanding on the regu-
lation of mammalian oocyte meiosis resumption. Cell Cycle 2009;8:27417.
47. Reddy P, Zheng W, Liu K. Mechanisms maintaining the dormancy and sur-
vival of mammalian primordial follicles. Trends Endocrinol Metab 2010;21:
96103.
48. Reddy P, Liu L, Adhikari D, Jagarlamudi K, Rajareddy S, Shen Y, et al. Oocyte-
specic deletion of Pten causes premature activation of the primordial folli-
cle pool. Science 2008;319:6113.
49. Ouyang JX, Luo T, Sun HY, Huang J, Tang DF, Wu L, et al. RNA interference
mediated pten knock-down inhibit the formation of polycystic ovary. Mol
Cell Biochem 2013;380:195202.
50. Picton HM, Harris SE, Muruvi W, Chambers EL. The in vitro growth and
maturation of follicles. Reproduction 2008;136:70315.
51. Tomizawa S, Nowacka-Woszuk J, Kelsey G. DNA methylation establishment
during oocyte growth: mechanisms and signicance. Int J Dev Biol 2012;56:
86775.
52. Swales AK, Spears N. Genomic imprinting and reproduction. Reproduction
2005;130:38999.
53. Grant B, Hirsh D. Receptor-mediated endocytosis in the Caenorhabditis ele-
gans oocyte. Mol Biol Cell 1999;10:431126.
54. Krishnamurthy H, Kishi H, Shi M, Galet C, Bhaskaran RS, Hirakawa T, et al.
Postendocytotic trafcking of the follicle-stimulating hormone (FSH)-FSH re-
ceptor complex. Mol Endocrinol 2003;17:216276.
55. Shimada M, Yanai Y, Okazaki T, Yamashita Y, Sriraman V, Wilson MC, et al.
Synaptosomal-associated protein 25 gene expression is hormonally regu-
lated during ovulation and is involved in cytokine/chemokine exocytosis
from granulosa cells. Mol Endocrinol 2007;21:2487502.
56. Liu M. The biology and dynamics of mammalian cortical granules. Reprod
Biol Endocrinol 2011;9:149.
VOL. -NO. -/-2014 11
Fertility and Sterility®
SUPPLEMENTAL TABLE 1
Upregulated FF miRNAs and their genomic localization.
Extragenic miRNAs
a
miRNAs located in protein encoding genes
miRNA Chromosomal position Clustered miRNAs miRNA Chromosomal position Clustered miRNAs Host gene
miR-886-5p 5q31.1 miR-214 1q24.3 miR-199a-2
miR-3120
DNM3
miR-339-3p 7p22.3 miR-135b 1q32.1 LEMD1
miR-29a 7q32.3 miR-29b-1 miR-10b 2q31.1 HOXD3
miR-31 9p21.3 miR-95 4 ABLIM2
miR-202 10q26.3 miR-218 4p15.31 SLIT2
miR-210 11p15.5 miR-449a 5q11.2 miR-449b
miR-449c
CDC20 B
miR-100 11q24.1 let-7a-2 miR-887 5p15.1 FBXL7
miR-125b 21q21.1 miR-99a
miR-125b
let-7c
miR-874 5q31.2 KLHL3
miR-99a miR-489 7q21.3 miR-653 CALCR
miR-134 14q32.31 Cluster
miR-379/miR-656
miR-204 9q21.12 TRPM3
miR-323-3p miR-455-5p 9q32 COL27A1
miR-410 miR-483-5p 11p15.5 IGF2
miR-654-3p miR-140-3p 16q22.1 WWP2
miR-203 14q32.33 miR-203b
miR-203a
miR-337-5p 14q32.2 miR-493
miR-665
miR-431
miR-433
miR-127
miR-432
miR-337
miR-370
miR-493
miR-193b 16p13.12 miR-365a
miR-193bmiR-365
miR-212 17p13.3 miR-132
miR-212miR-132
miR-503 Xq26.3 miR-450a-2
miR-542
miR-450a-1
miR-450b
miR-503
miR-424
miR-542-5p
miR-508-3p Xq27.3 miR-507
miR-506
a
Or located in noncoding genes.
Santonocito. Exosomal microRNAs in human follicular uid. Fertil Steril 2014.
11.e1 VOL. -NO. -/-2014
ORIGINAL ARTICLE: REPRODUCTIVE BIOLOGY
... These short non-coding sequences can regulate gene expression by directly targeting the mRNA and inhibiting its translation to a protein, or/and by interfering with the epigenome [26,27]. Therefore, miRNAs are capable of modulating biological pathways involved in folliculogenesis [28], and extracellular miRNAs (exmiRNAs) from FF are predicted to influence targets that regulate the epidermal growth factor receptor (EGFR), transforming growth factor beta (TGFβ), mitogen-activated protein kinase (MAPK), and Wnt signaling pathways, all of which are known to contribute to follicular development, oocyte meiotic resumption, and ovulation [29][30][31][32]. Since the majority of FF miRNAs are present within EVs [32], it is believed that follicular fluid EVs (ffEVs) play an important role in mediating cell-tocell communication within the follicular microenvironment and may modulate biological signaling pathways via transferring bioactive molecules such as miRNAs. ...
... The manner of interaction and physiological homogeneity of EVs are currently the subject of many studies in relation to folliculogenesis, gametogenesis, and embryo development in diverse animal species [29,[33][34][35]. Several studies in mice, pigs, and cattle have shown that EVs can significantly affect changes in the transcriptome during gamete and embryo development [35][36][37]. ...
Article
Full-text available
Cumulus cell (CC) expansion is pivotal for oocyte maturation, during which CCs release factors that initiate paracrine signaling within the follicular fluid (FF). The FF is abundant in extracellular vesicles (EVs) that facilitate intercellular communication. Although bovine and murine EVs can control cumulus expansion, these effects have not been observed in equines. This study aimed to assess the impact of FF-derived EVs (ffEVs) on equine CC expansion, viability, and transcriptome. Cumulus–oocyte complexes (COCs) that underwent in vitro maturation (IVM) in the presence (200 µg protein/mL) or absence (control) of ffEVs were assessed for cumulus expansion and viability. CCs were isolated after 12 h of IVM, followed by RNA extraction, cDNA library generation, and subsequent transcriptome analysis using next-generation sequencing. Confocal microscopy images illustrated the internalization of labeled ffEVs by CCs. Supplementation with ffEVs significantly enhanced cumulus expansion in both compacted (Cp, p < 0.0001) and expanded (Ex, p < 0.05) COCs, while viability increased in Cp groups (p < 0.01), but decreased in Ex groups (p < 0.05), compared to the controls. Although transcriptome analysis revealed a subtle effect on CC RNA profiles, differentially expressed genes encompassed processes (e.g., MAPK and Wnt signaling) potentially crucial for cumulus properties and, consequently, oocyte maturation.
... The proper follicular development and oocyte maturation depend on the follicular fluid (FF), which creates an essential microenvironment. Exosomal miRNAs were found in the follicular fluid of human, bovine and other species, according to previous studies [18][19][20], and they may closely relate to the development of ovarian follicles. But the contribution of follicular fluid exosomal miRNAs to female infertility hasn't been fully clarified. ...
... The transmission of cell-secreted vesicles containing miRNAs between germ cells and their surrounding somatic cells, is indispensable for germ cell maturation [27]. Otherwise, numerous researchers have also suggested that exosomal miRNA may be a possible regulator of TGFB/BMP, WNT, MAPK, ErbB, and the ubiquitin-mediated signaling pathway for modulating mammalian follicle and oocyte maturation [18][19][20]28]. It implies that these exosomal DEmiRs may be crucial in the pathological process of ovarian dysfunction in patients with DOR by regulating angiogenesis and cell maturation in the ovary. ...
Article
Full-text available
Background Diminished ovarian reserve (DOR) is defined as a reduction in ovarian reserve and oocyte quality. The pathophysiology of DOR has not been completely explained as of yet. Scholars have uncovered a large number of exosomes that have been detected in follicular fluid, and exosomal miRNAs have been proven to play a critical role in controlling ovarian disorders and follicle formation. We focused on the expression profile of follicular fluid-derived exosomal microRNAs (miRNAs) and attempted to understand if their role is connected to the pathomechanism of DOR. Methods The follicular fluid-derived differentially expressed exosomal miRNAs (DEmiRs) between patients with DOR and those with normal ovarian function were investigated using the next-generation sequencing (NGS) method. The main metabolic and signaling pathways of DEmiRs were identified using the KEGG pathway database, disease ontology (DO) analysis, and gene ontology (GO) analysis. In the end, a Protein-Protein Interaction (PPI) network was built to search for exosomal miRNAs and their target genes that were potentially strongly connected with DOR. Results In comparison to normal controls, 52 DEmiRs were discovered in follicular fluid-derived exosomes of DOR patients, of which 19 were up-regulated and 33 were down-regulated (|log2(fold change) |>2, P < 0.05). GO, DO analysis, and the KEGG pathway database revealed that many of these DEmiRs have broad biological roles that are connected to ovarian function and disorders. The top ten DEmiRs in terms of expression were then chosen for miRNA-mRNA interaction analysis. Totally, 8 experimentally supported miRNAs (hsa-miR-1246, hsa-miR-483-3p, hsa-miR-122-5p, hsa-miR-130b-3p, hsa-miR-342-3p, hsa-miR-625-3p, hsa-miR-675-3p, and hsa-miR-134-5p) and 126 target genes were filtrated by utilizing Cytoscape software. The module analysis findings of the PPI network showed that the main module cluster with a score > 6.0 (MCODE score = 15) had six hub genes, including IGFR, VEGFA, KRAS, ERBB2, RHOA, and PTEN (MCODE score = 11.472). Conclusion Our data suggested a special expression profile of follicular fluid-derived exosomal miRNAs in patients with DOR, which was probably correlated to ovarian dysfunction and follicle formation. These results may give a unique insight into a better understanding of the molecular process in the pathogenesis of DOR or other ovarian diseases.
... Hong et al. showed that plasma miR-23a levels were elevated in DOR women, and treatment with pre-miR-23a induced apoptosis in human granulosa cells (GCs) cultured in vitro [8]. A study has shown that follicular fluid (FF) harbors numerous exosomes, and its exosomal miRNAs are involved in biological events such as oocyte meiotic resumption, follicular atresia apoptosis, and epigenetic modifications [9]. However, the function of FF exosomal miRNAs in patients with DOR is yet to be deciphered. ...
Article
Full-text available
Purpose Decreased ovarian reserve function is mainly characterized by female endocrine disorders and fertility decline. Follicular fluid (FF) exosomal microRNAs (miRNAs) have been shown to regulate the function of granulosa cells (GCs). The present study explored differentially expressed miRNAs (DEmiRNAs) in patients with diminished ovarian reserve (DOR). Methods FF was collected from 12 DOR patients and 12 healthy controls. DEmiRNAs between the two groups were identified and analyzed using high-throughput sequencing technology and validated by real-time quantitative PCR (RT-qPCR). Results A total of 592 DEmiRNAs were identified using high-throughput miRNA sequencing, of which 213 were significantly upregulated and 379 were significantly downregulated. The sequencing results were further validated by RT-qPCR. These DEmiRNA target genes were mainly involved in the cancer pathway, phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway, regulation of actin cytoskeleton signaling pathway, and biological processes related to protein binding, nucleoplasm, cytoplasm, and cell membrane. Conclusion FF exosomal miRNAs are significantly differentially expressed in DOR patients versus non-DOR patients, underscoring their crucial role in regulating the pathogenesis of DOR.
... Most of the upregulated miRNAs are involved in the regulation of WNT, MAPK, ErbB, and TGF-β signaling pathways, and can contribute to the development of ovarian follicles or stimulate the proliferation of granulosa cells and expansion of cumulus. They can also start the resumption of meiosis by negatively regulating genes that encode follicle maturation-inhibiting factors [110]. Hu et al. [111] identified six exosomal miRNAs in the follicular fluid (miR-6087, miR-199a-5p, miR143-3p, miR-483-5p, miR-23b-3p, and miR-200a-3p) related to PCOS pathogenesis and predicted that these miRNAs were mainly related to amino acid biosynthesis; metabolism of glycine, serine, and threonine; glycosaminoglycan biosynthesis; monocarboxylic acid metabolism; and carbon metabolism. ...
Article
Polycystic ovarian syndrome (PCOS) is a common endocrine disorder in women of reproductive age. The clinical symptoms include hyperandrogenism, chronic anovulation, and multiple ovarian cysts. PCOS is strongly associated with obesity and insulin resistance. MicroRNAs are a group of short non-coding RNAs that play a role in the post-transcriptional regulation of gene expression and translational inhibition. They play a vital role in the regulation of multiple metabolic and hormonal processes as well as in oocyte maturation and folliculogenesis in the female reproductive system. miRNAs can be used as diagnostic biomarkers or therapeutic targets because of their stability. The encapsulation of miRNAs in extracellular vesicles or exosomes contributes to their stability. Exosomes are constantly secreted by many cells and size of about 30 to 150 nm. Enveloping miRNAs exosomes can release them for cellular communication. The induced transfer of miRNAs by exosomes is a novel process of genetic exchange between cells. Many studies have shown that along with non-exosomal miRNAs, different types of exosomal miRNAs derived from the serum and follicular fluid can play an essential role in PCOS pathogenesis. These miRNAs are involved in follicular development and various functions in granulosa cells, apoptosis, cell proliferation, and follicular atresia. The present study aimed to comprehensively review the evidence on miRNAs and their affected pathways under both non-exosomal and exosomal circumstances, primarily focusing on the pathogenesis of PCOS.
Article
Full-text available
Recent studies have established that exosomes (EXs) derived from follicular fluid (FF) can promote oocyte development. However, the specific sources of these EXs and their regulatory mechanisms remain elusive. It is universally acknowledged that oocyte development requires signal communication between granulosa cells (GCs) and oocytes. However, the role of GC-secreted EXs and their functions are poorly understood. This study aimed to investigate the role of porcine granulosa-cell-derived exosomes (GC-EXs) in oocyte development. In this study, we constructed an in vitro model of porcine GCs and collected and identified GC-EXs. We confirmed that porcine GCs can secrete EXs and investigated the role of GC-EXs in regulating oocyte development by supplementing them to cumulus–oocyte complexes (COCs) cultured in vitro. Specifically, GC-EXs increase the cumulus expansion index (CEI), promote the expansion of the cumulus, alleviate reactive oxygen species (ROS), and increase mitochondrial membrane potential (MMP), resulting in improved oocyte development. Additionally, we conducted small RNA sequencing of GC-EXs and hypothesized that miR-148a-3p, the highest-expressed microRNA (miRNA), may be the key miRNA. Our study determined that transfection of miR-148a-3p mimics exerts effects comparable to the addition of EXs. Meanwhile, bioinformatics prediction, dual luciferase reporter gene assay, and RT-qPCR identified DOCK6 as the target gene of miR-148a-3p. In summary, our results demonstrated that GC-EXs may improve oocyte antioxidant capacity and promote oocyte development through miR-148a-3p by targeting DOCK6.
Article
Extracellular vesicles (EVs) play an important role in the development and function of mammalian ovarian follicles. However, the mechanisms by which they are taken up by the follicular granulosa cells remain unclear. In addition, while oocytes play a pivotal role in follicular development, the possible interactions between oocyte‐derived paracrine factors (ODPFs) and EV signals are unknown. Therefore, this study aimed to elucidate the mechanism of EV uptake and the effects of ODPFs on EV uptake by follicular somatic mural granulosa cells in mice. Fluorescence‐labeled transferrin (TRF) and cholera toxin B (CTB), substrates for clathrin‐ and caveolae‐mediated endocytosis, respectively, were taken up by mural granulosa cells in vitro. Their uptake was inhibited by Pitstop 2 and genistein, inhibitors of clathrin and caveolae pathways, respectively. Mural granulosa cells took up EVs, and this uptake was suppressed by Pitstop 2 and genistein. Moreover, ODPFs promoted the uptake of EVs and CTB, but not TRF, by mural granulosa cells. These results suggest that mural granulosa cells take up EVs through both clathrin‐ and caveolae‐mediated endocytosis and that oocytes may promote caveolae‐mediated endocytosis to facilitate the uptake of EVs.
Article
Full-text available
Evaluation of the optimal number of embryos, their quality, and the precise timing for transfer are critical determinants in reproductive success, although still remaining one of the main challenges in assisted reproduction technologies (ART). Indeed, the success of in vitro fertilization (IVF) treatments relies on a multitude of events and factors involving both the endometrium and the embryo. Despite concerted efforts on both fronts, the overall success rates of IVF techniques continue to range between 25% and 30%. The role of the endometrium in implantation has been recently recognized, leading to the hypothesis that both the “soil” and the “seed” play a central role in a successful pregnancy. In this respect, identification of the molecular signature of endometrial receptivity together with the selection of the best embryo for transfer become crucial in ART. Currently, efforts have been made to develop accurate, predictive, and personalized tests to identify the window of implantation and the best quality embryo. However, the value of these tests is still debated, as conflicting results are reported in the literature. The purpose of this review is to summarize and critically report the available criteria to optimize the success of embryo transfer and to better understand current limitations and potential areas for improvement.
Article
Full-text available
Background Assisted reproductive technology accounts for an increasing proportion of infertility treatments, and assessments to predict clinical pregnancy outcomes are desired. Extracellular vesicles exist in follicular fluid, and small non coding RNAs in extracellular vesicles underline the possibility of reflecting pregnancy potential. Methods Follicular fluid samples are collected from 20 ovarian follicles of 15 infertile patients undergoing assisted reproductive technology. Extracellular vesicles are isolated by serial centrifugation and small RNA sequencing is performed to investigate the profiles of microRNAs and P-element-induced wimpy testis-interacting RNAs. Results Small extracellular vesicles with a size range of approximately 100 nm are successfully isolated, and the small non coding RNA profiles of pregnant samples (n = 8) are different from those of non-pregnant samples (n = 12). Fourteen dysregulated small non coding RNAs are selected to identify the independent candidates [mean read count >100, area under the curve >0.8]. Among them, we find that a specific combination of small non coding RNAs (miR-16-2-3p, miR-378a-3p, and miR-483-5p) can predict the pregnant samples more precisely using a receiver operating characteristics curves analysis (area under the curve: 0.96). Furthermore, even in the same patients, the three microRNAs are differentially expressed between pregnant and non-pregnant samples. Conclusions Our results demonstrate that small non coding RNAs derived from small extracellular vesicles in follicular fluid can be potential non-invasive biomarkers for predicting pregnancy, leading to their probable application in assisted reproductive technology. Further large-scale studies are required to validate the clinical usefulness of these small non coding RNAs.
Article
BACKGROUND Peripheral nerve injury can result in significant clinical complications that have uncertain prognoses. Currently, there is a lack of effective pharmacological interventions for nerve damage, despite the existence of several small compounds, peptides, hormones, and growth factors that have been suggested as potential enhancers of neuron regeneration. Despite the objective of achieving full functional restoration by surgical intervention, the persistent challenge of inadequate functional recovery remains a significant concern in the context of peripheral nerve injuries. AIM To examine the impact of exosomes on the process of functional recovery following a complete radial nerve damage. METHODS A male individual, aged 24, who is right-hand dominant and an immigrant, arrived with an injury caused by a knife assault. The cut is located on the left arm, specifically below the elbow. The neurological examination and electrodiagnostic testing reveal evidence of left radial nerve damage. The sural autograft was utilized for repair, followed by the application of 1 mL of mesenchymal stem cell-derived exosome, comprising 5 billion microvesicles. This exosome was split into four equal volumes of 0.25 mL each and delivered microsurgically to both the proximal and distal stumps using the subepineural pathway. The patient was subjected to a period of 180 d during which they had neurological examination and electrodiagnostic testing. RESULTS The duration of the patient’s follow-up period was 180 d. An increasing Tinel’s sign and sensory-motor recovery were detected even at the 10th wk following nerve grafting. Upon the conclusion of the 6-mo post-treatment period, an evaluation was conducted to measure the extent of improvement in motor and sensory functions of the nerve. This assessment was based on the British Medical Research Council scale and the Mackinnon-Dellon scale. The results indicated that the level of improvement in motor function was classified as M5, denoting an excellent outcome. Additionally, the level of improvement in sensory function was classified as S3+, indicating a good outcome. It is noteworthy that these assessments were conducted in the absence of physical therapy. At the 10th wk post-injury, despite the persistence of substantial axonal damage, the nerve exhibited indications of nerve re-innervation as evidenced by control electromyography (EMG). In contrast to the preceding. EMG analysis revealed a significant electrophysiological enhancement in the EMG conducted at the 6th-mo follow-up, indicating ongoing regeneration. CONCLUSION Enhanced comprehension of the neurobiological ramifications associated with peripheral nerve damage, as well as the experimental and therapy approaches delineated in this investigation, holds the potential to catalyze future clinical progress.
Article
To explore whether granulosa cell (GC)‐derived exosomes (GC‐Exos) and follicular fluid‐derived exosomes (FF‐Exos) have functional similarities in follicle development and to establish relevant experiments to validate whether GC‐Exos could serve as a potential substitute for follicular fluid‐derived exosomes to improve folliculogenesis. GC‐Exos were characterized. MicroRNA (miRNA) profiles of exosomes from human GCs and follicular fluid were analyzed in depth. The signature was associated with folliculogenesis, such as phosphatidylinositol 3 kinases‐protein kinase B signal pathway, mammalian target of rapamycin signal pathway, mitogen‐activated protein kinase signal pathway, Wnt signal pathway, and cyclic adenosine monophosphate signal pathway. A total of five prominent miRNAs were found to regulate the above five signaling pathways. These miRNAs include miRNA‐486‐5p, miRNA‐10b‐5p, miRNA‐100‐5p, miRNA‐99a‐5p, and miRNA‐21‐5p. The exosomes from GCs and follicular fluid were investigated to explore the effect on folliculogenesis by injecting exosomes into older mice. The proportion of follicles at each stage is counted to help us understand folliculogenesis. Exosomes derived from GCs were isolated successfully. miRNA profiles demonstrated a remarkable overlap between the miRNA profiles of FF‐Exos and GC‐Exos. The shared miRNA signature exhibited a positive influence on follicle development and activation. Furthermore, exosomes derived from GCs and follicular fluid promoted folliculogenesis in older female mice. Exosomes derived from GCs had similar miRNA profiles and follicle‐promoting functions as follicular fluid exosomes. Consequently, GC‐Exos are promising for replacing FF‐Exos and developing new commercial reagents to improve female fertility.
Article
Full-text available
Exosomes are vesicles of endocytic origin released by many cells. These vesicles can mediate communication between cells, facilitating processes such as antigen presentation. Here, we show that exosomes from a mouse and a human mast cell line (MC/9 and HMC-1, respectively), as well as primary bone marrow-derived mouse mast cells, contain RNA. Microarray assessments revealed the presence of mRNA from approximately 1300 genes, many of which are not present in the cytoplasm of the donor cell. In vitro translation proved that the exosome mRNAs were functional. Quality control RNA analysis of total RNA derived from exosomes also revealed presence of small RNAs, including microRNAs. The RNA from mast cell exosomes is transferable to other mouse and human mast cells. After transfer of mouse exosomal RNA to human mast cells, new mouse proteins were found in the recipient cells, indicating that transferred exosomal mRNA can be translated after entering another cell. In summary, we show that exosomes contain both mRNA and microRNA, which can be delivered to another cell, and can be functional in this new location. We propose that this RNA is called " exosomal shuttle RNA " (esRNA). Exosomes are small (50–90 nm) membrane vesicles of endocytic origin that are released into the extracellular environment on fusion of multivesicular bodies (MVB) with the plasma membrane 1. Many cells have the capacity to release exosomes, including reticulo-cytes 2 , dendritic cells 3 , B cells 4 , T cells 5 , mast cells 6 , epithelial cells 7 and tumour cells 8. The functions of exosomes are not completely understood, although it has been shown that exosomes can participate in the signalling events contributing to antigen presentation to T cells 4 and the development of tolerance 9. Several mechanisms have been hypothesized describing the interactions of exosomes and recipient cells. Exosomes can bind to cells through recep-tor–ligand interactions, similar to cell–cell communication mediating , for example, antigen presentation 4. Alternatively, exosomes could putatively attach or fuse with the target-cell membrane, delivering exosomal surface proteins and perhaps cytoplasm to the recipient cell 10,11. Finally, exosomes may also be internalized by the recipient cells by mechanisms such as endocytosis 12. Exosomes were isolated from a mast-cell line (MC/9), primary bone marrow-derived mast cells (BMMC) and a human mast-cell line (HMC-1) through a series of microfiltration and ultracentrifugation steps modified from what has been previously described 4. To confirm that the structures studied indeed are exosomes, they were examined by electron microscopy (Fig. 1a), flow cytometric analysis (FACS; Fig. 1b), and proteomic analysis (see Supplementary Information, Table S1). The electron micrographs of the exosomes revealed rounded structures with a size of approximately 50–80 nm, similar to previously described exo-somes 4,13–15. The identity of the studied vesicles was further confirmed as exosomes by FACS analysis (Fig. 1b), which show the presence of the surface protein CD63 — a commonly used marker of exosomes. Finally, extensive protein analysis of the MC/9 exosomes was performed on multiple samples using LC-MS/MS technology. A total of 271 proteins were identified (see Supplementary Information, Table S1) from three preparations of the isolated vesicles, of which 47 proteins were present in all three samples. More importantly, a large number of the proteins found in the preparations were the same as proteins previously identified in exosomes produced by other cells (that is, exosomes derived from intestinal epithelial cells, urine, dendritic cells, microglia, melanoma, T-cells and B-cells). In particular, 60% of the 47 proteins found in all samples of mast-cell exosomes have been previously found in other types of exosomes. Moreover, 39% of the 271 total proteins found in the analysed exosome samples have also been previously found in other types of exosomes. Thus, the electron microscopy, the FACS, and the detailed protein analyses each provided significant evidence in favour of the identification of the isolated vesicles as exosomes. The presence of nucleic acids was examined in exosomes derived from MC/9, BMMC and HMC-1 cells to define a potential mechanism by which exosomes may mediate cell–cell communication. These assessments showed that isolated exosomes contain no DNA (see Supplementary Information, Fig. S1). However, substantial amounts of RNA were detected by agarose gel electrophoresis, spectrophotometry
Article
Full-text available
Background: Gene-expression analysis is increasingly important in biological research, with real-time reverse transcription PCR (RT-PCR) becoming the method of choice for high-throughput and accurate expression profiling of selected genes. Given the increased sensitivity, reproducibility and large dynamic range of this methodology, the requirements for a proper internal control gene for normalization have become increasingly stringent. Although housekeeping gene expression has been reported to vary considerably, no systematic survey has properly determined the errors related to the common practice of using only one control gene, nor presented an adequate way of working around this problem. Results: We outline a robust and innovative strategy to identify the most stably expressed control genes in a given set of tissues, and to determine the minimum number of genes required to calculate a reliable normalization factor. We have evaluated ten housekeeping genes from different abundance and functional classes in various human tissues, and demonstrated that the conventional use of a single gene for normalization leads to relatively large errors in a significant proportion of samples tested. The geometric mean of multiple carefully selected housekeeping genes was validated as an accurate normalization factor by analyzing publicly available microarray data. Conclusions: The normalization strategy presented here is a prerequisite for accurate RT-PCR expression profiling, which, among other things, opens up the possibility of studying the biological relevance of small expression differences.
Article
Full-text available
Exosomes are small extracellular vesicles (30-100 nm) derived from the endosomal system, which have raised considerable interest in the last decade. Several studies have shown that they mediate cell-to-cell communication in a variety of biological processes. Thus, in addition to cell-to-cell direct interaction or secretion of active molecules, they are now considered another class of signal mediators. Exosomes can be secreted by several cell types and retrieved in many body fluids, such as blood, urine, saliva and cerebrospinal fluid. In addition to proteins and lipids, they also contain nucleic acids, namely mRNA and miRNA. These features have prompted extensive research to exploit them as a source of biomarkers for several pathologies, such as cancer and neurodegenerative disorders. In this context, exosomes also appear attractive as gene delivery vehicles. Furthermore, exosome immunomodulatory and regenerative properties are also encouraging their application for further therapeutic purposes. Nevertheless, several issues remain to be addressed: exosome biogenesis and secretion mechanisms have not been clearly understood, and physiological functions, as well as pathological roles, are far from being satisfactorily elucidated.
Article
Full-text available
Cell-cell communication within the follicle involves many signaling molecules, and this process may be mediated by secretion and uptake of exosomes that contain several bioactive molecules including extra-cellular miRNAs. Follicular fluid and cells from individual follicles of cattle were grouped based on Brilliant Cresyl Blue (BCB) staining of the corresponding oocytes. Both Exoquick precipitation and differential ultracentrifugation were used to separate the exosome and non-exosomal fraction of follicular fluid. Following miRNA isolation from both fractions, the human miRCURY LNA™ Universal RT miRNA PCR array system was used to profile miRNA expression. This analysis found that miRNAs were present in both exosomal and non-exosomal fraction of bovine follicular fluid. We found 25 miRNAs differentially expressed (16 up and 9 down) in exosomes and 30 miRNAs differentially expressed (21 up and 9 down) in non-exosomal fraction of follicular fluid in comparison of BCB- versus BCB+ oocyte groups. Expression of selected miRNAs was detected in theca, granulosa and cumulus oocyte complex. To further explore the potential roles of these follicular fluid derived extra-cellular miRNAs, the potential target genes were predicted, and functional annotation and pathway analysis revealed most of these pathways are known regulators of follicular development and oocyte growth. In order to validate exosome mediated cell-cell communication within follicular microenvironment, we demonstrated uptake of exosomes and resulting increase of endogenous miRNA level and subsequent alteration of mRNA levels in follicular cells in vitro. This study demonstrates for the first time, the presence of exosome or non-exosome mediated transfer of miRNA in the bovine follicular fluid, and oocyte growth dependent variation in extra-cellular miRNA signatures in the follicular environment.
Article
Full-text available
What is the expression pattern of microRNAs (miRNAs) in human cumulus-oocyte complexes (COCs)? Several miRNAs are enriched in cumulus cells (CCs) or oocytes, and are predicted to target genes involved in biological functions of the COC. The transcriptional profiles of human MII oocytes and the surrounding CCs are known. However, very limited data are available about post-transcriptional regulators, such as miRNAs. This is the first study focussing on the identification and quantification of small RNAs, including miRNAs, in human oocytes and CCs using a deep-sequencing approach. MII oocytes and CCs were collected from women who underwent IVF. Using the Illumina/deep-sequencing technology, we analyzed the small RNAome of pooled MII oocytes (n = 24) and CC samples (n = 20). The mRNA targets of CC and MII oocyte miRNAs were identified using in silico prediction algorithms. Using oligonucleotide microarrays, genome-wide gene expression was studied in oocytes (10 pools of 19 ± 3 oocytes/each) and 10 individual CC samples. TaqMan miRNA assays were used to confirm the sequencing results in independent pools of MII oocytes (3 pools of 8 ± 3 oocytes/each) and CC samples (3 pools of 7 ± 3 CCs/each). The functional role of one miRNA, MIR23a, was assessed in primary cultures of human CCs. Deep sequencing of small RNAs yielded more than 1 million raw reads. By mapping reads with a single location to the human genome, known miRNAs that were abundant in MII oocytes (MIR184, MIR100 and MIR10A) or CCs (MIR29a, MIR30d, MIR21, MIR93, MIR320a, MIR125a and the LET7 family) were identified. Predicted target genes of the oocyte miRNAs were associated with the regulation of transcription and cell cycle, whereas genes targeted by CC miRNAs were involved in extracellular matrix and apoptosis. Comparison of the predicted miRNA target genes and mRNA microarray data resulted in a list of 224 target genes that were differentially expressed in MII oocytes and CCs, including PTGS2, CTGF and BMPR1B that are important for cumulus-oocyte communication. Functional analysis using primary CC cultures revealed that BCL2 and CYP19A1 mRNA levels were decreased upon MIR23a overexpression. Only known miRNAs were investigated in the present study on COCs. Moreover, the source of the material is MII oocytes that failed to fertilize. The present findings suggest that miRNA could play a role in the regulation of the oocyte and CC crosstalk. This work was partially supported by a grant from Ferring Pharmaceuticals. The authors of the study have no conflict of interest to report. Not applicable.
Article
Full-text available
Pten (phosphatase and tensin homolog deleted on chromosome 10), a kind of tumor suppressor gene, plays important roles in female reproductive system. But its expression and roles in the formation of polycystic ovaries are yet to be known. In this study, we constructed a rat model of PCOS using norethindrone and HCG injections and found the expressions of pten mRNA and PTEN protein increased significantly in the polycystic ovary tissue by immunohistochemistry, RT-PCR, and western blot. Furthermore, the results showed that in vivo ovaries could be effectively transfected by lentiviral vectors through the ovarian microinjection method and indicated that pten shRNA may inhibit the formation of polycystic ovaries by pten down-regulation. Our study provides new information regarding the role of PTEN in female reproductive disorders, such as polycystic ovary syndrome.
Chapter
In this chapter, issues in the thermal management of LED in packaging level were discussed. The basic concepts of heat transfer were briefly introduced covering conduction, convention, radiation, and thermal resistance. Models for system level thermal resistance for typical LED packages were briefly discussed. Various ways to decrease thermal resistance in terms of packaging forms were discussed, including packaging materials. System in packaging (SiP) and chip on board (COB) were discussed and some examples were given.
Article
Over the past several years it has become clear that alterations in the expression of microRNA (miRNA) genes contribute to the pathogenesis of most if not all human malignancies. These alterations can be caused by various mechanisms, including deletions, amplifications or mutations involving miRNA loci, epigenetic silencing or the dysregulation of transcription factors that target specific miRNAs. Because malignant cells show dependence on the dysregulated expression of miRNA genes, which in turn control or are controlled by the dysregulation of multiple protein-coding oncogenes or tumour suppressor genes, these small RNAs provide important opportunities for the development of future miRNA-based therapies.
Article
The two most commonly used methods to analyze data from real-time, quantitative PCR experiments are absolute quantification and relative quantification. Absolute quantification determines the input copy number, usually by relating the PCR signal to a standard curve. Relative quantification relates the PCR signal of the target transcript in a treatment group to that of another sample such as an untreated control. The 2(-DeltaDeltaCr) method is a convenient way to analyze the relative changes in gene expression from real-time quantitative PCR experiments. The purpose of this report is to present the derivation, assumptions, and applications of the 2(-DeltaDeltaCr) method. In addition, we present the derivation and applications of two variations of the 2(-DeltaDeltaCr) method that may be useful in the analysis of real-time, quantitative PCR data. (C) 2001 Elsevier science.