Quantitative and stoichiometric analysis of the microRNA ... · to testing the validity of any...

6
Quantitative and stoichiometric analysis of the microRNA content of exosomes John R. Chevillet a , Qing Kang a,b , Ingrid K. Ruf a,1 , Hilary A. Briggs a,1 , Lucia N. Vojtech c,1 , Sean M. Hughes c,1 , Heather H. Cheng a,d , Jason D. Arroyo a , Emily K. Meredith a , Emily N. Gallichotte a , Era L. Pogosova-Agadjanyan e , Colm Morrissey f , Derek L. Stirewalt e , Florian Hladik c,d,g , Evan Y. Yu d , Celestia S. Higano d,e,f , and Muneesh Tewari a,b,e,h,i,j,k,2 Divisions of a Human Biology, e Clinical Research, g Vaccine and Infectious Disease, and i Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109; Departments of b Internal Medicine and h Biomedical Engineering, j Biointerfaces Institute, and k Center for Computational Medicine, University of Michigan, Ann Arbor, MI 48109; and Departments of c Obstetrics and Gynecology, d Medicine, and f Urology, Division of Oncology, University of Washington, Seattle, WA 98195 Edited* by Vishva M. Dixit, Genentech, San Francisco, CA, and approved August 29, 2014 (received for review May 18, 2014) Exosomes have been proposed as vehicles for microRNA (miRNA) -based intercellular communication and a source of miRNA bio- markers in bodily fluids. Although exosome preparations contain miRNAs, a quantitative analysis of their abundance and stoi- chiometry is lacking. In the course of studying cancer-associated extracellular miRNAs in patient blood samples, we found that exosome fractions contained a small minority of the miRNA content of plasma. This low yield prompted us to perform a more quantitative assessment of the relationship between miRNAs and exosomes using a stoichiometric approach. We quantified both the number of exosomes and the number of miRNA molecules in replicate samples that were isolated from five diverse sources (i.e., plasma, seminal fluid, dendritic cells, mast cells, and ovarian cancer cells). Regardless of the source, on average, there was far less than one molecule of a given miRNA per exosome, even for the most abundant miRNAs in exosome preparations (mean ± SD across six exosome sources: 0.00825 ± 0.02 miRNA molecules/exosome). Thus, if miRNAs were distributed homogenously across the exo- some population, on average, over 100 exosomes would need to be examined to observe one copy of a given abundant miRNA. This stoichiometry of miRNAs and exosomes suggests that most individual exosomes in standard preparations do not carry biolog- ically significant numbers of miRNAs and are, therefore, individually unlikely to be functional as vehicles for miRNA-based communica- tion. We propose revised models to reconcile the exosome-mediated, miRNA-based intercellular communication hypothesis with the ob- served stoichiometry of miRNAs associated with exosomes. microvesicle | circulating E xosomes have been classically described as 40- to 100-nm vesicles (1, 2) that are secreted by a broad range of cell types and have been identified in diverse body fluids (e.g., plasma, saliva, lymph, ascites, semen, amniotic fluid, cerebrospinal fluid, etc.) (ref. 3 and references therein). More recently, microRNAs (miRNAs) have been reported to be present in exosome prep- arations (4), suggesting that these vesicles may function as a ve- hicle for intercellular miRNA transfer [a message in a bottle(5)] and a mode of intercellular communication (6). This hy- pothesis has been tantalizing, given that miRNAs bind to and repress the activity of specific target mRNAs, and it is estimated that >60% of all mRNAs are regulated by miRNAs (7). In ad- dition, exosomes typically display cell surface proteins derived from their cell of origin, which can be recognized by cell surface receptors (e.g., proteoglycans) and internalized by recipient cells (8), resulting in transfer of the exosome contents. In- tercellular transfer of miRNAs through exosomes would bypass recipient cell transcriptional controls (9), providing a relatively direct means of regulation. Studies delivering purified exo- somes to recipient cells have reported transfer of miRNAs in experimental settings (1012), and the use of exosomes as small RNA delivery vehicles is being studied as a potential therapeutic strategy (13, 14). Although the possibility of exosome-mediated miRNA trans- fer as a mode of intercellular communication is an attractive concept (6, 15, 16), current mechanistic models lack detail, and the physiologic significance of this paradigm is not yet established (17). Quantitative evaluation of key components is fundamental to testing the validity of any model (18), and knowledge of the stoichiometry of miRNAs and exosomes (i.e., how many mole- cules of a given miRNA are carried by an exosome) would provide insight into the requirements and limits of miRNA- based intercellular communication. This question has historically been challenging to address, because exosomes are subdiffraction limit particles and therefore, cannot be directly enumerated by light microscopy or flow cytometric methods (19). Although exo- somes are typically visualized by electron microscopy (EM) (20), this approach is nonquantitative because of the variation introduced by the complex sample preparation process. One method that can circumvent these limitations is nanoparticle tracking analysis (NTA). NTA quantifies particles under 300 nm by visualizing the light scattering generated by the particles and using it to track them during Brownian motion (19). In this study, we set out to determine the degree to which extracellular biomarker miRNAs are associated with exosomes prepared from the plasma of prostate cancer patients given that Significance Exosomes have been a subject of great interest in recent years, especially in the context of the microRNAs (miRNAs) that they contain. Exosome-mediated miRNA transfer between cells has been proposed to be a mechanism for intercellular signaling and exosome-associated miRNAs in biofluids have been suggested as potential minimally invasive biomarkers for multiple human disease states. Remarkably, we show here that most exosomes derived from standard preparations do not harbor many copies of miRNA molecules. These findings suggest a reevaluation of current models of the mechanism of exosome-mediated miRNA communication and indicate that stoichiometric analysis will be valuable for the study of other populations of extracellular vesicles and their associated RNAs as well. Author contributions: J.R.C., H.H.C., and M.T. designed research; J.R.C., Q.K., I.K.R., H.A.B., S.M.H., and E.N.G. performed research; L.N.V., S.M.H., H.H.C., J.D.A., E.K.M., E.L.P.-A., C.M., D.L.S., F.H., E.Y.Y., and C.S.H. contributed new reagents/specimens/analytic tools; J.R.C., Q.K., I.K.R., H.A.B., E.N.G., and M.T. analyzed data; and J.R.C. and M.T. wrote the paper. The authors declare no conflict of interest. *This Direct Submission article had a prearranged editor. Freely available online through the PNAS open access option. 1 I.K.R., H.A.B., L.N.V., and S.M.H. contributed equally to this work. 2 To whom correspondence should be addressed. Email: [email protected]. This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. 1073/pnas.1408301111/-/DCSupplemental. 1488814893 | PNAS | October 14, 2014 | vol. 111 | no. 41 www.pnas.org/cgi/doi/10.1073/pnas.1408301111 Downloaded by guest on September 23, 2020

Transcript of Quantitative and stoichiometric analysis of the microRNA ... · to testing the validity of any...

Page 1: Quantitative and stoichiometric analysis of the microRNA ... · to testing the validity of any model (18), and knowledge of the stoichiometry of miRNAs and exosomes (i.e., how many

Quantitative and stoichiometric analysis of themicroRNA content of exosomesJohn R. Chevilleta, Qing Kanga,b, Ingrid K. Rufa,1, Hilary A. Briggsa,1, Lucia N. Vojtechc,1, Sean M. Hughesc,1,Heather H. Chenga,d, Jason D. Arroyoa, Emily K. Mereditha, Emily N. Gallichottea, Era L. Pogosova-Agadjanyane,Colm Morrisseyf, Derek L. Stirewalte, Florian Hladikc,d,g, Evan Y. Yud, Celestia S. Higanod,e,f, and Muneesh Tewaria,b,e,h,i,j,k,2

Divisions of aHuman Biology, eClinical Research, gVaccine and Infectious Disease, and iPublic Health Sciences, Fred Hutchinson Cancer Research Center, Seattle,WA 98109; Departments of bInternal Medicine and hBiomedical Engineering, jBiointerfaces Institute, and kCenter for Computational Medicine, Universityof Michigan, Ann Arbor, MI 48109; and Departments of cObstetrics and Gynecology, dMedicine, and fUrology, Division of Oncology, University ofWashington, Seattle, WA 98195

Edited* by Vishva M. Dixit, Genentech, San Francisco, CA, and approved August 29, 2014 (received for review May 18, 2014)

Exosomes have been proposed as vehicles for microRNA (miRNA)-based intercellular communication and a source of miRNA bio-markers in bodily fluids. Although exosome preparations containmiRNAs, a quantitative analysis of their abundance and stoi-chiometry is lacking. In the course of studying cancer-associatedextracellular miRNAs in patient blood samples, we found thatexosome fractions contained a small minority of the miRNAcontent of plasma. This low yield prompted us to perform amore quantitative assessment of the relationship between miRNAsand exosomes using a stoichiometric approach. We quantified boththe number of exosomes and the number of miRNA molecules inreplicate samples that were isolated from five diverse sources (i.e.,plasma, seminal fluid, dendritic cells, mast cells, and ovarian cancercells). Regardless of the source, on average, there was far less thanone molecule of a given miRNA per exosome, even for the mostabundant miRNAs in exosome preparations (mean ± SD across sixexosome sources: 0.00825 ± 0.02 miRNA molecules/exosome).Thus, if miRNAs were distributed homogenously across the exo-some population, on average, over 100 exosomes would need tobe examined to observe one copy of a given abundant miRNA.This stoichiometry of miRNAs and exosomes suggests that mostindividual exosomes in standard preparations do not carry biolog-ically significant numbers of miRNAs and are, therefore, individuallyunlikely to be functional as vehicles for miRNA-based communica-tion. We propose revised models to reconcile the exosome-mediated,miRNA-based intercellular communication hypothesis with the ob-served stoichiometry of miRNAs associated with exosomes.

microvesicle | circulating

Exosomes have been classically described as 40- to 100-nmvesicles (1, 2) that are secreted by a broad range of cell types

and have been identified in diverse body fluids (e.g., plasma,saliva, lymph, ascites, semen, amniotic fluid, cerebrospinal fluid,etc.) (ref. 3 and references therein). More recently, microRNAs(miRNAs) have been reported to be present in exosome prep-arations (4), suggesting that these vesicles may function as a ve-hicle for intercellular miRNA transfer [“a message in a bottle”(5)] and a mode of intercellular communication (6). This hy-pothesis has been tantalizing, given that miRNAs bind to andrepress the activity of specific target mRNAs, and it is estimatedthat >60% of all mRNAs are regulated by miRNAs (7). In ad-dition, exosomes typically display cell surface proteins derivedfrom their cell of origin, which can be recognized by cell surfacereceptors (e.g., proteoglycans) and internalized by recipientcells (8), resulting in transfer of the exosome contents. In-tercellular transfer of miRNAs through exosomes would bypassrecipient cell transcriptional controls (9), providing a relativelydirect means of regulation. Studies delivering purified exo-somes to recipient cells have reported transfer of miRNAs inexperimental settings (10–12), and the use of exosomes as small

RNA delivery vehicles is being studied as a potential therapeuticstrategy (13, 14).Although the possibility of exosome-mediated miRNA trans-

fer as a mode of intercellular communication is an attractiveconcept (6, 15, 16), current mechanistic models lack detail, andthe physiologic significance of this paradigm is not yet established(17). Quantitative evaluation of key components is fundamentalto testing the validity of any model (18), and knowledge of thestoichiometry of miRNAs and exosomes (i.e., how many mole-cules of a given miRNA are carried by an exosome) wouldprovide insight into the requirements and limits of miRNA-based intercellular communication. This question has historicallybeen challenging to address, because exosomes are subdiffractionlimit particles and therefore, cannot be directly enumerated bylight microscopy or flow cytometric methods (19). Although exo-somes are typically visualized by electron microscopy (EM) (20),this approach is nonquantitative because of the variation introducedby the complex sample preparation process. One method that cancircumvent these limitations is nanoparticle tracking analysis (NTA).NTA quantifies particles under 300 nm by visualizing the lightscattering generated by the particles and using it to track themduring Brownian motion (19).In this study, we set out to determine the degree to which

extracellular biomarker miRNAs are associated with exosomesprepared from the plasma of prostate cancer patients given that

Significance

Exosomes have been a subject of great interest in recent years,especially in the context of the microRNAs (miRNAs) that theycontain. Exosome-mediated miRNA transfer between cells hasbeen proposed to be a mechanism for intercellular signaling andexosome-associated miRNAs in biofluids have been suggestedas potential minimally invasive biomarkers for multiple humandisease states. Remarkably, we show here that most exosomesderived from standard preparations do not harbor many copiesof miRNA molecules. These findings suggest a reevaluation ofcurrent models of the mechanism of exosome-mediated miRNAcommunication and indicate that stoichiometric analysis willbe valuable for the study of other populations of extracellularvesicles and their associated RNAs as well.

Author contributions: J.R.C., H.H.C., and M.T. designed research; J.R.C., Q.K., I.K.R., H.A.B.,S.M.H., and E.N.G. performed research; L.N.V., S.M.H., H.H.C., J.D.A., E.K.M., E.L.P.-A.,C.M., D.L.S., F.H., E.Y.Y., and C.S.H. contributed new reagents/specimens/analytic tools;J.R.C., Q.K., I.K.R., H.A.B., E.N.G., and M.T. analyzed data; and J.R.C. and M.T. wrotethe paper.

The authors declare no conflict of interest.

*This Direct Submission article had a prearranged editor.

Freely available online through the PNAS open access option.1I.K.R., H.A.B., L.N.V., and S.M.H. contributed equally to this work.2To whom correspondence should be addressed. Email: [email protected].

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1408301111/-/DCSupplemental.

14888–14893 | PNAS | October 14, 2014 | vol. 111 | no. 41 www.pnas.org/cgi/doi/10.1073/pnas.1408301111

Dow

nloa

ded

by g

uest

on

Sep

tem

ber

23, 2

020

Page 2: Quantitative and stoichiometric analysis of the microRNA ... · to testing the validity of any model (18), and knowledge of the stoichiometry of miRNAs and exosomes (i.e., how many

exosomes have been proposed as a significant packaging mech-anism for miRNAs released from cancer cells (21–25). We foundthat only a small minority (median = 2.5%) of the extracellularmiRNA content of plasma is associated with the exosomalfraction prepared by standard differential centrifugation meth-ods, prompting us to use quantitative methods to more directlydetermine the stoichiometry of miRNAs and exosomes fromcancer patient plasma as well as diverse other biological sources.We show here that the stoichiometry of a given miRNA andexosome is consistently much less than one, even for the mostabundant miRNAs identified in our exosome preparations. Thus,most individual exosomes in standard preparations do not carrybiologically significant numbers of miRNAs. These results haveimplications for exosome-mediated miRNA communication(6, 15) and indicate that current mechanistic models for thisphenomenon may need to be revised to be reconciled with theobserved miRNA–exosome stoichiometry.

ResultsStandard Exosome Preparations Recover Only a Minority of miRNAsfrom Cancer Patient Plasma. Multiple independent studies havereported that exosomes contain miRNA, and exosomes havebeen proposed to be treasure chests for biomarker applications(22, 26). However, we and other groups have found that, in healthyindividuals, many cell-free circulating miRNAs are present insoluble form and complexed with proteins, such as Argonaute2(Ago2) (27, 28). Although circulating miRNA biomarkers forcancer have been observed in exosome preparations (22, 29),a quantitative analysis of the physical state (i.e., exosome-asso-ciated vs. soluble) of cancer-associated miRNAs has not beenreported. Given that cancer cells are reported to abundantlyshed exosomes, we hypothesized that cancer-associated cir-culating miRNAs (and consequently, those most suitable asbiomarkers) would be packaged primarily in exosomes.To determine the physical state of cancer-associated circulat-

ing miRNAs, we chose to examine the abundance of three estab-lished circulating prostate cancer biomarkers [hsa-miR-141 (30),-210 (31), and -375 (22, 31)] and a broadly expressed controlmiRNA found to be nondiagnostic in plasma [hsa-miR-16 (30)]in exosome preparations from plasma collected from patientswith metastatic prostate cancer (n = 9). Plasma samples werefractionated by differential centrifugation, a typical method usedin the field to prepare exosomes (Fig. 1A and SI Materials andMethods) (20, 32, 33). Transmission EM (TEM) was used toconfirm the recovery of vesicles that were consistent with exo-somes by size and morphology (Fig. 1B). miRNA was extractedfrom aliquots of both the supernatant and exosome fractions,and the abundance of the four miRNAs listed above was quan-tified by real-time PCR. The exosome fraction contained littlemiRNA relative to the supernatant (mean values ranging from2.7% for miR-375 to 5.6% for miR-141) (Fig. 1C and Table S1).Comparable results were obtained from the analysis of exosomefractions prepared from patient serum (n = 3) (Fig. S1) and fresh(i.e., never frozen) plasma (n = 6) (Fig. S2), indicating that ourresults were not specific to plasma and not an artifact of freezing.To determine if the observed distribution of cancer biomarkermiRNAs was representative of the global profile of miRNAs inplasma from these patients, we measured the relative abundanceof 375 individual miRNAs in the plasma fractions using real-timePCR array analysis of pooled samples (n = 3 individuals per poolin three pools) (Fig. 1D). On average, we detected 137 miRNAsin the pools by real-time PCR, and only 2.5% (median) (TableS2) of the miRNA content of these miRNAs was present in theexosome fractions.

Exosomes from Diverse Biologic Sources Contain Less than One Copy,on Average, of Their Most Abundant miRNAs.Although we observedthat the proportion of total circulating miRNA associated withexosome fractions was small, we hypothesized that individualexosomes may still carry a significant number of miRNA moleculesand thereby, provide potency for intercellular communication (34).

To test this hypothesis, we sought to quantify the number ofmiRNA molecules (of a given sequence) that are contained withinan exosome. To provide data that would be broadly repre-sentative, we prepared exosomes from diverse sources, includingdifferent human biofluids (healthy donor plasma, n = 3 donors;prostate cancer patient plasma, n = 3 donors; healthy donorseminal fluid, n = 3 donors), as well as in vitro sources (humandendritic cells, n = 3 donors; ovarian cancer cells, n = 3 prepa-rations; mast cells, n = 2 preparations) to also evaluate morehomogenous exosomes (i.e., produced by single-cell types) (4).Exosomes were prepared using typical ultracentrifugation-basedprotocols (SI Materials and Methods). TEM was used to confirmthat the protocols yielded vesicles that were consistent withexosomes by size and morphology (Fig. 2).We then measured the number of exosomes in each of the

samples using NTA (Fig. 3 and SI Materials and Methods). Exo-some concentrations ranged over two orders of magnitude from6.06 × 107 exosomes/μL in one dendritic cell-derived preparationto 7.79 × 109 exosomes/μL in a semen exosome preparation. Sizedistribution profile analysis of the NTA data confirmed pop-ulations with particle sizes consistent with exosomes, with an

D

A Bplasma

cleared plasma

supernatant

centrifugation(12,000 g)

ultracentrifugation(120,000 g)

exosomes

C

case pool 1 case pool 3case pool 2

hsa-miR-337-5phsa-miR-454hsa-miR-629hsa-miR-374ahsa-miR-142-3phsa-let-7fhsa-miR-194hsa-miR-30chsa-miR-20b*hsa-miR-26ahsa-miR-30bhsa-miR-181ahsa-let-7ehsa-miR-148ahsa-miR-720hsa-miR-374bhsa-let-7ahsa-miR-32hsa-miR-885-5phsa-miR-1979hsa-miR-15bhsa-miR-144hsa-miR-151-5phsa-let-7chsa-miR-30e*hsa-let-7dhsa-miR-192hsa-miR-215hsa-let-7ghsa-miR-590-5phsa-miR-378hsa-miR-101hsa-miR-99bhsa-miR-106bhsa-miR-27ahsa-miR-20ahsa-miR-19ahsa-miR-122hsa-miR-191hsa-miR-23bhsa-miR-660hsa-miR-425hsa-miR-130ahsa-miR-126hsa-miR-15ahsa-miR-451hsa-miR-106ahsa-miR-185hsa-miR-486-5phsa-miR-16hsa-miR-29bhsa-miR-23ahsa-miR-126*hsa-miR-940hsa-miR-27bhsa-miR-92ahsa-miR-21hsa-miR-222hsa-miR-223hsa-miR-19bhsa-miR-93hsa-miR-146b-5phsa-miR-484hsa-miR-24hsa-miR-342-3phsa-miR-146ahsa-miR-30dhsa-miR-320ahsa-miR-25hsa-let-7d*hsa-miR-151-3phsa-miR-197hsa-let-7bhsa-let-7ihsa-miR-125a-5phsa-miR-125bhsa-miR-130bhsa-miR-132hsa-miR-140-3phsa-miR-140-5phsa-miR-141hsa-miR-142-5phsa-miR-143hsa-miR-145hsa-miR-150hsa-miR-152hsa-miR-154hsa-miR-186hsa-miR-18bhsa-miR-193a-5phsa-miR-195hsa-miR-20bhsa-miR-22hsa-miR-221hsa-miR-26bhsa-miR-29ahsa-miR-324-3phsa-miR-324-5phsa-miR-328hsa-miR-335hsa-miR-369-5phsa-miR-375hsa-miR-376ahsa-miR-376chsa-miR-379hsa-miR-409-3phsa-miR-421hsa-miR-423-5phsa-miR-432hsa-miR-532-5phsa-miR-574-3phsa-miR-584hsa-miR-625*hsa-miR-628-3phsa-miR-744hsa-miR-99a

hsa-miR-142-3phsa-miR-454hsa-let-7ehsa-let-7fhsa-miR-30chsa-let-7dhsa-miR-1979hsa-let-7chsa-miR-30bhsa-miR-144hsa-miR-940hsa-miR-720hsa-miR-107hsa-miR-154hsa-miR-32hsa-miR-374ahsa-miR-27ahsa-miR-766hsa-miR-374bhsa-miR-369-5phsa-miR-125a-5phsa-let-7ahsa-miR-22hsa-miR-26ahsa-miR-140-5phsa-miR-98hsa-miR-106bhsa-miR-660hsa-miR-99ahsa-miR-15ahsa-miR-99bhsa-miR-181ahsa-let-7ihsa-miR-194hsa-miR-324-5phsa-miR-150hsa-let-7ghsa-miR-451hsa-miR-185hsa-miR-26bhsa-miR-191hsa-let-7bhsa-miR-151-5phsa-miR-425hsa-miR-145hsa-miR-126hsa-miR-532-5phsa-miR-21hsa-miR-199a-3phsa-miR-15bhsa-miR-20bhsa-miR-29bhsa-miR-195hsa-miR-126*hsa-miR-484hsa-miR-192hsa-miR-744hsa-miR-222hsa-miR-378hsa-miR-23bhsa-miR-27bhsa-miR-130ahsa-miR-20ahsa-miR-92ahsa-miR-101hsa-miR-421hsa-miR-186hsa-miR-19bhsa-miR-93hsa-miR-16hsa-miR-106ahsa-miR-146ahsa-miR-148ahsa-miR-122hsa-miR-342-3phsa-miR-215hsa-miR-19ahsa-miR-140-3phsa-miR-223hsa-miR-320ahsa-miR-151-3phsa-miR-590-5phsa-miR-375hsa-miR-409-3phsa-miR-24hsa-miR-23ahsa-miR-25hsa-miR-146b-5phsa-miR-221hsa-miR-625*hsa-miR-486-5phsa-miR-152hsa-miR-30dhsa-miR-20b*hsa-let-7d*hsa-miR-197hsa-miR-1hsa-miR-103hsa-miR-125bhsa-miR-130bhsa-miR-132hsa-miR-133ahsa-miR-134hsa-miR-139-5phsa-miR-141hsa-miR-142-5phsa-miR-143hsa-miR-148bhsa-miR-17hsa-miR-181dhsa-miR-18ahsa-miR-18bhsa-miR-193a-5phsa-miR-196bhsa-miR-199a-5phsa-miR-200bhsa-miR-200chsa-miR-210hsa-miR-214hsa-miR-22*hsa-miR-29ahsa-miR-29chsa-miR-301ahsa-miR-30ahsa-miR-30e*hsa-miR-328hsa-miR-335hsa-miR-337-3phsa-miR-363hsa-miR-365hsa-miR-376ahsa-miR-376chsa-miR-379hsa-miR-382hsa-miR-423-5phsa-miR-424hsa-miR-432hsa-miR-452hsa-miR-495hsa-miR-574-3phsa-miR-584hsa-miR-598hsa-miR-628-3phsa-miR-629hsa-miR-652hsa-miR-671-5phsa-miR-7hsa-miR-877hsa-miR-885-5p

hsa-let-7fhsa-miR-142-3phsa-let-7chsa-let-7ahsa-let-7ehsa-miR-374ahsa-miR-30chsa-miR-210hsa-miR-454hsa-miR-30bhsa-miR-19ahsa-miR-374bhsa-miR-125bhsa-miR-125a-5phsa-miR-144hsa-miR-30ahsa-miR-26ahsa-miR-1979hsa-miR-181ahsa-miR-148ahsa-miR-194hsa-miR-629hsa-let-7dhsa-miR-122hsa-miR-145hsa-miR-146b-5phsa-miR-26bhsa-miR-720hsa-miR-940hsa-miR-192hsa-miR-151-5phsa-miR-625*hsa-let-7ghsa-miR-15bhsa-miR-99bhsa-miR-15ahsa-miR-126hsa-miR-425hsa-miR-101hsa-miR-20bhsa-let-7bhsa-miR-484hsa-miR-126*hsa-miR-16hsa-miR-21hsa-miR-27ahsa-miR-27bhsa-miR-590-5phsa-miR-451hsa-miR-197hsa-miR-23bhsa-miR-146ahsa-miR-199a-3phsa-miR-22hsa-miR-20ahsa-miR-342-3phsa-miR-152hsa-miR-660hsa-let-7d*hsa-let-7ihsa-miR-223hsa-miR-375hsa-miR-20b*hsa-miR-92ahsa-miR-151-3phsa-miR-25hsa-miR-106ahsa-miR-486-5phsa-miR-106bhsa-miR-215hsa-miR-140-3phsa-miR-24hsa-miR-186hsa-miR-93hsa-miR-222hsa-miR-409-3phsa-miR-130ahsa-miR-191hsa-miR-23ahsa-miR-378hsa-miR-320ahsa-miR-19bhsa-miR-185hsa-miR-221hsa-miR-30dhsa-miR-1hsa-miR-100hsa-miR-107hsa-miR-130bhsa-miR-132hsa-miR-133ahsa-miR-133bhsa-miR-134hsa-miR-140-5phsa-miR-141hsa-miR-142-5phsa-miR-143hsa-miR-148bhsa-miR-150hsa-miR-154hsa-miR-17hsa-miR-181dhsa-miR-18ahsa-miR-18a*hsa-miR-18bhsa-miR-193a-5phsa-miR-195hsa-miR-196bhsa-miR-199a-5phsa-miR-200bhsa-miR-200chsa-miR-29ahsa-miR-29bhsa-miR-29chsa-miR-301ahsa-miR-30e*hsa-miR-32hsa-miR-324-3phsa-miR-324-5phsa-miR-328hsa-miR-335hsa-miR-337-3phsa-miR-363hsa-miR-369-5phsa-miR-376ahsa-miR-376chsa-miR-379hsa-miR-382hsa-miR-421hsa-miR-423-3phsa-miR-423-5phsa-miR-425*hsa-miR-432hsa-miR-495hsa-miR-532-5phsa-miR-574-3phsa-miR-584hsa-miR-627hsa-miR-628-3phsa-miR-652hsa-miR-7hsa-miR-744hsa-miR-766hsa-miR-877hsa-miR-885-5phsa-miR-99a

1000 20 40 60 80

individual donor

1 2 3 4 5 6 7 8 90.0

0.2

0.4

0.6

0.8

1.0

1 2 3 4 5 6 7 8 90.0

0.2

0.4

0.6

0.8

1.0

1 2 3 4 5 6 7 8 90.0

0.2

0.4

0.6

0.8

1.0

1 2 3 4 5 6 7 8 90.0

0.2

0.4

0.6

0.8

1.0re

lativ

e co

pies

rela

tive

copi

esre

lativ

e co

pies

rela

tive

copi

es

miR-141

miR-375

miR-210

miR-16

1 2 3 4 5 6 7 8 9

exosomes supernatant

1.0

0.00.2

0.80.60.4

1 2 3 4 5 6 7 8 9

1 2 3 4 5 6 7 8 9

1 2 3 4 5 6 7 8 9

1.0

0.00.2

0.80.60.4

1.0

0.00.2

0.80.60.4

1.0

0.00.2

0.80.60.4

Fig. 1. Exosome preparations from cancer patient plasma have low miRNAabundance. (A) Differential centrifugation workflow used to prepare exo-some fractions. (B) Representative TEM image of exosome preparations.(Scale bars: Left, 0.5 μm; Right, 200 nm.) (C) Relative abundance of selectedbiomarker (miR-141, -210, and -375) and nonbiomarker control (miR-16)miRNAs in plasma exosome fractions (blue bars) and postultracentrifugationsupernatant (gray bars) across prostate cancer patient plasmas (n = 9). (D)Heat map of relative quantity values (relative percentage of abundance) formiRNAs detected by real-time PCR array profiling of exosome and super-natant fractions derived from prostate cancer patient plasmas (n = 3 poolscomposed of N = 3 individuals each). miRNAs are ranked (top to bottom)according to relative proportion of each miRNA in exosome vs. supernatantfractions. Consistent with our previous study using healthy donor plasma, asmall but notable minority of miRNAs was enriched in the exosome fraction,including the hematopoietic-specific miRNA miR-142–3p.

Chevillet et al. PNAS | October 14, 2014 | vol. 111 | no. 41 | 14889

MED

ICALSC

IENCE

S

Dow

nloa

ded

by g

uest

on

Sep

tem

ber

23, 2

020

Page 3: Quantitative and stoichiometric analysis of the microRNA ... · to testing the validity of any model (18), and knowledge of the stoichiometry of miRNAs and exosomes (i.e., how many

average modal size ranging from 92 (for mast cell exosomes)to 122 nm (for seminal fluid exosomes) (Fig. 3), reflective oftheir appearance by EM (Fig. 2).Although the accuracy of exosome counts derived from NTA

has been independently established (19, 35), we empirically vali-dated this approach by comparing NTA-derived counts with thoseproduced using an independent method of quantification: com-parative fluorescence microscopy analysis (SI Materials and Methodsand Table S3). This method is based on the quantification ofdye-labeled exosomes relative to fluorescent beads of a com-parable size. Results produced by NTA were similar to thoseobtained by comparative fluorescence in triplicate analysis of thesame exosome sample (8.9 × 1010 vs. 1.3 × 1011 exosomes/mL,respectively; P = 0.5619).To identify the most abundant miRNAs in each exosome type,

we extracted total RNA from aliquots of each sample, pooled thesamples corresponding to a given exosome type, and analyzedeach pool by real-time PCR array analysis. Abundant miRNAswere identified by ranking cycle threshold data in ascendingorder (Fig. 2 and Table S4). We then performed absolutequantification of two of five miRNAs displaying the lowest cyclethresholds for each of the individual (nonpooled) samples usinga standard real-time PCR-based absolute quantification protocolfor miRNAs (Fig. 3 B, Right, C, Right, D, Right, E, Right, F, Right,and G, Right) (36).

Using the data derived from the above experiments, we de-termined the ratio of miRNA molecules to exosomes for eachsample. In all of the samples examined, the ratio of miRNAmolecules for a given miRNA to the number of individualexosomes was substantially lower than one (Fig. 4 and TableS5). Across all samples, we observed an average of one copy ofmiRNA detected per 121 exosomes, ranging from one copy per 9exosomes (miR-720 in seminal fluid exosomes) to one copy per47,162 exosomes (miR-126 in healthy donor plasma exosomes).We found this result surprising, because it implied that mostexosomes would not contain any copies of abundant miRNAs.

20

25

30

35

40

healthy plasma

PCapatient plasma

dendriticcells

mast cells

OvCa cells

exosomesRNA

extractionmiRNA ID abundant

miRNAs

miR-223miR-126

A

B

C

D

E

F

G

Ct

20

25

30

35

40

20

25

30

35

40

15

20

25

30

35

40

30

35

40

30

35

40

1004035

202530

200 300

3530

152025

40

35

25

30

40100 200 300

100 200 300

50 150 250

100 200 300

50 100 150

4035

202530

4035

202530

40

35

25

30

Ct

Ct

Ct

Ct

Ct

100 200

miR-223miR-126

miR-720let-7b

miR-720miR-223

miR-223miR-720

let-7bmiR-720

number of miRNAs detected

seminal

Fig. 2. Identification of abundant miRNAs in exosome preparations fromdiverse sources. (A) Schematic of the workflow used to identify (ID) abun-dant miRNAs. (B–G) TEM images of exosomes prepared from (B) healthy(donor) plasma, (C) prostate cancer (PCa) patient plasma, (D) healthy donorseminal fluid, (E) dendritic cells (in vitro differentiated Langerhans cells), (F)mast cells (HMC-1 cell line), and (G) ovarian carcinoma (OvCa) cells (2008 cellline). Real-time PCR array profiling results for all detected miRNAs [sorted byabundance as represented by cycle threshold (Ct) on a reverse y axis] aredisplayed in Right for the corresponding samples in Left. Each bar representsa different miRNA. miRNAs selected for absolute copy number quantifica-tion from the top five most abundant (as defined by the lowest Ct values)are indicated by arrows. Primary miRNA profiling data are also presented inTable S4. (Scale bars: B, D, and F, 100 nm; C, E, and G, 200 nm.)

Prepara

tion 1

Prepara

tion 2

Prepara

tion 3

0

2 1008

4 1008

6 1008

8 1008

0 100 200 300 400 5000.0

0.5

1.0

1.5

Donor

1

Donor

2

Donor

30

1 1009

2 1009

3 1009

4 1009

serum

free

exos

ome f

ree0

1 1009

2 1009

3 1009

4 1009

exosomepreparationcount and measure

exosome size (NTA)

miRNA absolute

(real-time PCR)

concentration

healthy plasma

PCapatient plasma

seminal

dendritic cells

mast cells

OvCa cells

A

B

C

D

E

F

G

1 109

4 109

3 109

2 109

part

icle

s/µL

1 2 30

0 100 200 300 400 5000.0

0.5

1.0

1.5

patient 1patient 2patient 3

0 0

0.5

1.0

1.5

donor 1donor 2donor 3

0 100 200 300 400 5000.0

0.5

1.0

1.5

2.0

donor 1donor 2donor 3

0 0

0.5

1.0

1.5

1.5donor 1donor 2donor 30.5

1.0

% o

f cou

nts

particle diameter (nm)

0 100 200 300 400 5000.0

0.5

1.0

1.5

prep 1prep 2prep 3

0.0

1.5

0.5

1.0

% o

f cou

nts

0.0

1.5

0.5

1.0

% o

f cou

nts

0.0

2.01.5

% o

f cou

nts

0.0

1.5

0.5

1.0

% o

f cou

nts

0.0

1.00.5

1.5

0.5

1.0

% o

f cou

nts

0.0

Donor

1

Donor

2

Donor

30

2 1009

4 1009

6 1009

Donor

1

Donor

2

Donor

30

2 1009

4 1009

6 1009

8 1009

1 1010

donor

Donor

1

Donor

2

Donor

30

2 1008

4 1008

6 1008

0

6 109

4 109

2 109

part

icle

s/µL

4 109

1 1010

8 109

6 109

part

icle

s/µL

0

6 108

4 108

2 108

part

icle

s/µL

prep 1prep 2

0

4 109

2 109

1 109

part

icle

s/µL

0

8 108

6 108

4 108

part

icle

/µL

1 2 3patient

2 109

0

3 109

2 108

1 2 3donor

1 2 3donor

1 2prep

1 2 3prep

miRNA (copies/µL) donor miR-126 miR-223

1.57 105 4.44 105

5.93 104 1.40 105

1.22 105 2.43 105

123

miRNA (copies/µL) patient miR-126 miR-223

9.51 104 1.60 106

4.78 105 1.59 106

4.21 105 1.48 106

miRNA (copies/µL) donor let-7b miR-720

1.32 107 8.61 107

1.38 107 9.98 107

1.80 107 6.73 107

miRNA (copies/µL) donor miR-223 miR-720

3.41 105 3.38 106

1.20 104 4.45 105

2.12 104 2.91 105

miRNA (copies/µL) prep miR-223 miR-720

4.85 106 5.01 106

8.08 106 3.31 106

miRNA (copies/µL) prep let-7b miR-720

2.90 106 2.61 106

2.52 106 2.54 106

1.02 105 5.52 105

123

123

123

12

123

copy number

Fig. 3. Quantification of exosome number, size distribution, and miRNAcontent. Aliquots of exosome preparations described in Fig. 2 were countedand sized by NTA. In parallel, total RNA was extracted from additionalexosome aliquots, and the concentrations of abundant exosomal miRNAsidentified in Fig. 2 were determined by real-time PCR. (A) Workflow to de-termine the size distribution, exosome concentration, and absolute quanti-fication of miRNAs in each exosome sample. (B–G) Exosome size distributionhistograms (representing the percentage of total counts found within each1-nm-sized bin), total particle concentrations, and miRNA concentrations inexosome preparations for (B) healthy (donor) plasma (n = 3 donors), (C)prostate cancer (PCa) patient plasma (n = 3 patients), (D) healthy donorseminal fluid (n = 3 donors), (E) dendritic cells (in vitro differentiatedLangerhans cells; n = 3 donors), (F) mast cells (HMC-1 cell line; n = 2 pre-parative replicates: preparation 1 from serum-free conditioned medium andpreparation 2 from exosome-depleted serum containing medium), and (G)ovarian carcinoma (OvCa) cells (2008 cell line; n = 3 preparative replicates).Values represent the concentrations in each exosome preparation (i.e., notthe concentration in the crude starting specimen).

14890 | www.pnas.org/cgi/doi/10.1073/pnas.1408301111 Chevillet et al.

Dow

nloa

ded

by g

uest

on

Sep

tem

ber

23, 2

020

Page 4: Quantitative and stoichiometric analysis of the microRNA ... · to testing the validity of any model (18), and knowledge of the stoichiometry of miRNAs and exosomes (i.e., how many

We sought to exclude the possibility that confounding tech-nical factors may have impacted our results, resulting in an un-derestimate of miRNA abundance and/or an overestimate ofexosome number. First, we sought to determine whether miRNAamplification efficiency might be reduced in the exosome sam-ples (e.g., because of the copurification and enrichment of a re-verse transcription or PCR inhibitor in the specimens). Thisreduced amplification efficiency could artificially lower our es-timation of miRNA abundance. However, there was no statisti-cally significant difference (P = 0.1038) in the measured valuesof the spiked-in synthetic cel-miR-39 miRNA oligoribonucleo-tide control (SI Materials and Methods) (36) in RNA extractsprepared from exosomes vs. those prepared from the post-ultracentrifugation supernatant. This result indicated that therewas no overall reduction in sensitivity for miRNA detection inexosome samples relative to the supernatant (Materials andMethods and Fig. S3). Likewise, there was no difference (P =0.7587) between exosome and supernatant samples with respectto measured values of synthetic oligoribonucleotide controls(UniSp6) (SI Materials and Methods) that were spiked into thereverse transcription reaction mixtures (Fig. S3), consistent withno difference in the abundance of potential reverse tran-scription or PCR inhibitors.Second, we considered the possibility that our exosome prepa-

rations may have been contaminated by cosedimentation ofother particles endogenously present in the crude samples. Theseparticles could be miscounted by NTA as exosomes, resulting inan overestimate of exosome abundance and therefore, reducingour estimate of miRNA copies per exosome. However, thediverse nature of specimen types (ranging from cell culturesupernatants to body fluids) and the use of different purificationmethods (SI Materials and Methods) make a consistent andabundant contaminant unlikely. Furthermore, TEM images arenot consistent with such a predominant contaminant. Even in thecase of exosomes prepared from a complex fluid, like plasma(Fig. 2 B and C), although there were additional backgroundparticles observed on TEM, they were predominantly very small(∼10 nm; possibly representing HDL) and generally below thesize limit of detection of NTA. In contrast, exosomes purifiedfrom mast and ovarian cancer cells in culture (Fig. 2 F and G)did not display such additional small particles but still yielded farless than one miRNA molecule per exosome. In addition, themodal distribution of the sizes of particles observed by NTA isconsistent with expectations of exosomes and TEM observations.Third, we addressed the possibility that our results could be

the product of inaccurate miRNA quantification caused by the

dependence of real-time PCR on standard curves (37). Werecently described (38) a droplet digital PCR (ddPCR) -basedapproach for absolute quantification of miRNAs from biofluidsamples. This end point method is independent of standardcurves, shows higher precision for the quantification of low-abundance miRNA targets, and is resistant to residual PCRinhibitors (39). To determine whether ddPCR would confirm ouroverall findings, we used ddPCR and real-time PCR side by sideto quantify miR-223 from healthy donor plasma exosomes aswell as let-7b and miR-720 from ovarian cancer cell exosomes(Fig. S4). Quantification of miR-223 and let-7b differed by lessthan 1% and 27% between the two methods, respectively, whichwere not statistically significant (P = 0.8861 and P = 0.1696,respectively). miR-720 measured in ovarian cancer cell exosomeswas found to be present at 70% lower abundance when mea-sured by ddPCR vs. real-time PCR (2.71 × 10−4 vs. 9.16 × 10−4copies per exosome, respectively; P = 0.0040), indicating that thismiRNA may be even more rare in exosomes than estimated byreal-time PCR.

DiscussionThe discovery that exosomes are associated with miRNAs hasspawned great interest in these vesicles as potential vehicles formiRNA-based intercellular communication as well as a source ofdiagnostic extracellular miRNA biomarkers. However, quanti-tative analyses to inform our mechanistic understanding of exo-some-mediated miRNA communication and guide our approachto the development of exosome-based molecular diagnosticshave been lacking. Exosomes are several orders of magnitudesmaller than cells (i.e., if we model both as spheres with exosomediameter = 100 nm and cellular diameter = 10 μm, an exosomewould be estimated to contain 0.0001% of the cellular volume).Although this very small size implies that exosomes have a verylimited capacity to sample the diverse RNA repertoire, somestudies have reported that certain miRNA sequences areenriched in exosomes relative to their cells of origin (4, 10). Suchenrichment could yield exosomes containing selected miRNAs atmuch higher copy numbers than would be expected from randomsampling. However, our results show that even abundant miRNAsare present at far less than one copy per exosome, indicating thatmost exosomes from standard preparations are devoid of the mostcommon sequences. This finding seems to be generalizable, be-cause the same conclusion was reached across six differentsources of exosomes derived from various body fluids as wellas conditioned media from in vitro cell cultures.We considered it important to exclude the possibility of

technical inaccuracy of our miRNA and/or exosome quantifica-tion methods, although such error would have to be on the scaleof orders of magnitude to change the overall conclusions.Therefore, we validated both our miRNA and exosome quanti-fication methods using orthogonal approaches (i.e., ddPCR andmembrane labeling/fluorescence microscopy, respectively) andconfirmed empirically that a difference in neither PCR amplifi-cation efficiency nor exosome quantification could explain ourresults. We also ruled out the possibility that freezing and sub-sequent thawing of exosome samples could be a confoundingvariable, which is consistent with an independent report con-firming the stability of exosomes and exosomal RNA underconditions of freezing–thawing (40).Multiple reports have implicated exosomes as a vehicle for

cell–cell communication through carriage and transfer of miR-NAs between cells. In general, studies of this particular functionhave typically been done with a large excess of exosomes, andwhether it is feasible for endogenous exosomes to be functionalmiRNA transfer vehicles in native physiological settings is notyet well-established. Our observed stoichiometry would suggestthat most individual exosomes are unlikely to be functional formiRNA communication. Our results refute both a high-occu-pancy/high-miRNA concentration model (Fig. 5A) and a high-occupancy/low-miRNA concentration model (Fig. 5B). However,we propose that our data are consistent with two alternative

one copy per exosome1

0.1

0.01

0.001

0.0001

0.00001

miR

NA

co

pie

s/ex

oso

me

10

Fig. 4. Abundant miRNAs in exosomes are present at much less than onecopy per exosome. Dot plot of miRNA copies per exosome determined foreach exosome source (based on data collected like in Fig. 3) and sorted bymean value (bar). Numeric values are presented in Table S5. OvCa, ovariancarcinoma; PCa, prostate cancer.

Chevillet et al. PNAS | October 14, 2014 | vol. 111 | no. 41 | 14891

MED

ICALSC

IENCE

S

Dow

nloa

ded

by g

uest

on

Sep

tem

ber

23, 2

020

Page 5: Quantitative and stoichiometric analysis of the microRNA ... · to testing the validity of any model (18), and knowledge of the stoichiometry of miRNAs and exosomes (i.e., how many

models: (i) a low-occupancy/low-miRNA concentration model(Fig. 5C), in which a small fraction of exosomes carries a lowconcentration of miRNA, or (ii) a low-occupancy/high-miRNAconcentration model (Fig. 5D), in which there are rare exosomesin the population carrying many copies of a given miRNA. Bothof these models would yield an average stoichiometry of less thanone miRNA per exosome. If exosome uptake is a selective andinfrequent event for a given cell, the low-occupancy/high-con-centration occupancy model would seem to be the most likely tosupport physiologic exosome-mediated communication throughconventional RNA-induced silencing complex-mediated target-ing of mRNAs. Our results, therefore, suggest that additionalstudy of potentially miRNA-rich subpopulations of exosomes maybe important in investigating intercellular miRNA-based com-munication. However, if cellular uptake of exosomes is rapid, low-concentration/low-occupancy miRNA may accumulate within thecell in functionally sufficient quantities. Such rapid uptake hasbeen observed in macrophages, which can internalize an equiva-lent of their cell surface in pinocytic vesicles every 33 min (41). Inaddition, it is worth noting that both low-occupancy models couldbe compatible with recently proposed nonconventional activitiesof miRNAs that presumably require much lower concentrations ofmiRNA delivery than conventional RNA-induced silencing com-plex-mediated mRNA targeting. Such activities include the elici-tation of cellular responses through binding of Toll-like receptors(42, 43) as well as potential effects of small RNAs on DNA tran-scription and/or epigenetic states (44).Exosomes isolated from plasma and serum have also been

reported as potential sources of miRNA biomarkers for mini-mally invasive disease diagnostics. We observed that only a smallminority (<3%) of cancer-associated biomarker miRNAs wererecovered with classical exosomes isolated using gold standarddifferential ultracentrifugation-based methods. Although this resultdoes not directly address the diagnostic use of these exosomes,it indicates that the majority of these established biomarkers ispresent in plasma and serum in other physical forms. Future

rigorous studies will be required to determine the diagnostic useof classical exosomes relative to extracellular vesicle prepara-tions recovered by alternate methods as well as other physicaland biochemical fractions of patient plasma. Larger vesicles,such as microvesicles and oncosomes, have also been reported tocontain miRNAs as well as larger RNAs (45, 46), and our pro-tocols were not designed to recover such vesicles. It is possiblethat such larger classes of extracellular vesicles may carry phys-iologically significant numbers of miRNA molecules and functionas an alternative vehicle for miRNA-based intercellular communi-cation as well as a source of biomarkers.In addition, whereas our study focused on miRNAs, it is

possible that other classes of RNA commonly measured in suchbiomarker studies, such as mRNAs (4, 45), may be packageddifferently than miRNAs. Future studies will be required tounderstand the relationship of other classes of RNA with dif-ferent classes of extracellular vesicles.In conclusion, our study of the stoichiometry of extracellular

vesicles and the miRNA cargo that they carry provides a funda-mental approach and a quantitative mechanistic framework todelineate the functional boundaries of extracellular vesicle-medi-ated communication. Our data warrant the specific quantitativeevaluation of exosomes that have been observed to be biologicallyactive in other settings [e.g., glioblastoma (45), placental immunityto viral infections (47), etc.] and thought to exercise their effectsthrough miRNAs, because the relevant exosome–miRNA stoi-chiometries may contrast with those reported here and providevaluable insight into their mechanism and physiologic relevance.

Materials and MethodsHuman Specimens. Written informed consent was obtained from each donor,and the study was performed with Institutional Review Board (IRB) approvalas specified below.Plasma. The research was approved by the University of Washington and FredHutchinson Cancer Research Center IRBs. Venous blood samples from indi-viduals withmetastatic prostate cancer and prostate cancer-negative controlswere collected and centrifuged for 15 min at 1,300 × g at 4 °C. Plasma wasaspirated and stored at −80 °C.Cord blood collection and dendritic (Langerhans) cell derivation. Approval for thestudy was given by the Seattle Biomedical Research Institute IRB. IsolatedCD34+ cells were differentiated as independently published (48, 49). Purifiedclusters were cultured at 5 × 105 cells/mL for 3 d, after which culture mediumwas used for exosome isolation (see below).Semen collection and preparation. Approval for the study was given by theUniversity of Washington IRB. Ejaculates from healthy HIV-negative menwere mixed with 3 mL RPMI medium and kept on ice until processing.

Isolation of Exosomes. From plasma. Plasma or serum (1 mL) was added to 1 mLice-cold PBS and centrifuged at 12,000 × g for 45 min at 4 °C. The cleareddilute plasma was then aspirated, added to 3 mL ice-cold PBS, and centri-fuged at 120,000 × g for 70 min. Supernatants were gently decanted, andexosomes were resuspended in PBS.From Langerhans cell-conditioned medium and semen. Cells were culturedfor 3 d in exosome-depleted media. Cells were removed by centrifugationat 1,000 × g for 10 min, and debris was removed by centrifugation at 2,400 × gfor 30 min followed by syringe filtration. Exosomes were then purified by ul-tracentrifugation over a sucrose cushion using a method adapted from ref. 50.From human ovarian carcinoma cells. The 2008 ovarian cancer cells (51) weregrown in DMEM with 10% (vol/vol) FBS. Cultures were then washed in tripli-cate and grown in serum-free medium for 48 h. These media were collected,centrifuged at 500 × g for 10 min, 0.22-μm syringe filtered, and then, ultra-centrifuged as above for preparation of plasma exosomes.From mast cells. Exosomes were prepared from the HMC-1 cell line essentiallyas described in ref. 4.

EM. Exosomes were processed for visualization by TEM as previouslydescribed (20).

RNA Isolation. Total RNAwas isolated from all samples using the miRNeasy Kit(Qiagen) as described in ref. 36.

Individual Real-Time PCR Assays. Individual miRNAs were detected by real-time PCR as previously described (30, 36).

A

B

C

high occupancyhigh miRNA concentration

low occupancylow miRNA concentration

low occupancyhigh miRNA concentration

D

high occupancylow miRNA concentration

Fig. 5. Stoichiometric models for exosome miRNA content. (A) High-occu-pancy/high-miRNA concentration model, in which the number of moleculesof an individual miRNA sequence would far exceed the number of exosomes.(B) High-occupancy/low-miRNA concentration model, in which the concen-tration of miRNA is lower, but most exosomes contain the miRNA. If thenumber of exosomes exceeds the number of copies of a given miRNA (asobserved in our study), the miRNA molecules may be (C ) distributedthroughout the population in a low-occupancy/low-miRNA concentra-tion distribution or (D) amassed in rare exosomes in a low-occupancy/high-miRNA concentration distribution.

14892 | www.pnas.org/cgi/doi/10.1073/pnas.1408301111 Chevillet et al.

Dow

nloa

ded

by g

uest

on

Sep

tem

ber

23, 2

020

Page 6: Quantitative and stoichiometric analysis of the microRNA ... · to testing the validity of any model (18), and knowledge of the stoichiometry of miRNAs and exosomes (i.e., how many

ddPCR. Samples were analyzed using our recently described method (38).

miRNA Profiling. Samples were profiled using miRNA Ready-to-Use PCR,Human Panel I, V2.M (Exiqon). The limit of quantification and the PCR ef-ficiency for each miRNA assay were determined as described in ref. 27.

NTA. Samples were loaded into the assembled sample chamber of a NanoSightLM10; 60-s video images were acquired by a Hamamatsu C11440 ORCA-Flash2.8 digital camera and analyzed by NanoSight NTA 2.3 software.

Statistical Analysis. Tests and parameters are indicated in the figures. Allstatistical analyses were performed using Prism 5.0c software.

Details of sample collection, processing, experimental procedures, anddata analysis are provided in SI Materials and Methods.

ACKNOWLEDGMENTS. We thank the Fred Hutchinson Cancer Research CenterElectron Microscopy Shared Resource (B. Schneider, S. MacFarlane, andS. Knecht) and the University of Washington Keck Microscopy Facility for

assistance in preparing and visualizing samples; R. Parkin, J. Noteboom,and D. Gonzales for assistance with specimen collection; J. Guenthoer,S. Freggario, J. Yan, D. Kuppers, and W. Spangenberg for advice and helpfuldiscussion; D. Adair for administrative support; and G. Powell for researchcoordination. We especially thank the volunteers who provided specimensfor this study. Fig. 5 art was prepared by Mesa Schumacher, Information Artand Design. This work was supported by Canary Foundation/American Can-cer Society (ACS) Postdoctoral Fellowship for the Early Detection of CancerPFTED-09-249-01-SEID (to J.R.C.), the Hillcrest Committee of Southern Oregon(J.R.C. and J.D.A.), an American Association for Cancer Research/AflacScholar-in-Training Award (to J.R.C.), an ACS Postdoctoral Fellowship (toJ.D.A.), National Institutes of Health (NIH) Transformative R01 Grant DK-085714 (to M.T.), Stand Up to Cancer Innovative Research Grant SU2C-AACR-IRG1109 (to M.T.), a Damon Runyon-Rachleff Innovation Award(to M.T.), a Prostate Cancer Foundation Creativity Award (to M.T.), De-partment of Defense Prostate Cancer Research Program Award W81XWH-09-1-0144 (to C.S.H.), Royalty Research Grant R21 AI095023 (to F.H.) fromthe University of Washington, NIH P01 Grant CA-85859 (to C.M.), and PacificNorthwest Prostate Cancer Specialized Program of Research Excellence GrantP50-CA-097186 (to C.M.).

1. Pan BT, Teng K, Wu C, Adam M, Johnstone RM (1985) Electron microscopic evidencefor externalization of the transferrin receptor in vesicular form in sheep reticulocytes.J Cell Biol 101(3):942–948.

2. Harding C, Heuser J, Stahl P (1984) Endocytosis and intracellular processing oftransferrin and colloidal gold-transferrin in rat reticulocytes: Demonstration of apathway for receptor shedding. Eur J Cell Biol 35(2):256–263.

3. Raposo G, Stoorvogel W (2013) Extracellular vesicles: Exosomes, microvesicles, andfriends. J Cell Biol 200(4):373–383.

4. Valadi H, et al. (2007) Exosome-mediated transfer of mRNAs and microRNAs is a novelmechanism of genetic exchange between cells. Nat Cell Biol 9(6):654–659.

5. Kharaziha P, Ceder S, Li Q, Panaretakis T (2012) Tumor cell-derived exosomes: Amessage in a bottle. Biochim Biophys Acta 1826(1):103–111.

6. Hannafon BN, Ding WQ (2013) Intercellular communication by exosome-derived mi-croRNAs in cancer. Int J Mol Sci 14(7):14240–14269.

7. Friedman RC, Farh KK, Burge CB, Bartel DP (2009) Most mammalian mRNAs areconserved targets of microRNAs. Genome Res 19(1):92–105.

8. Christianson HC, Svensson KJ, van Kuppevelt TH, Li JP, Belting M (2013) Cancer cellexosomes depend on cell-surface heparan sulfate proteoglycans for their in-ternalization and functional activity. Proc Natl Acad Sci USA 110(43):17380–17385.

9. Huan J, et al. (2013) RNA trafficking by acute myelogenous leukemia exosomes.Cancer Res 73(2):918–929.

10. Montecalvo A, et al. (2012) Mechanism of transfer of functional microRNAs betweenmouse dendritic cells via exosomes. Blood 119(3):756–766.

11. Kosaka N, et al. (2010) Secretory mechanisms and intercellular transfer of microRNAsin living cells. J Biol Chem 285(23):17442–17452.

12. Umezu T, Ohyashiki K, Kuroda M, Ohyashiki JH (2013) Leukemia cell to endothelialcell communication via exosomal miRNAs. Oncogene 32(22):2747–2755.

13. Alvarez-Erviti L, et al. (2011) Delivery of siRNA to the mouse brain by systemic in-jection of targeted exosomes. Nat Biotechnol 29(4):341–345.

14. Ohno S, et al. (2013) Systemically injected exosomes targeted to EGFR deliver anti-tumor microRNA to breast cancer cells. Mol Ther 21(1):185–191.

15. Ramachandran S, Palanisamy V (2012) Horizontal transfer of RNAs: Exosomes asmediators of intercellular communication. Wiley Interdiscip Rev RNA 3(2):286–293.

16. Stoorvogel W (2012) Functional transfer of microRNA by exosomes. Blood 119(3):646–648.

17. Leslie M (2013) Cell biology. NIH effort gambles on mysterious extracellular RNAs.Science 341(6149):947.

18. Sverdlov ED (2012) Amedeo Avogadro’s cry: What is 1 μg of exosomes? BioEssays34(10):873–875.

19. Dragovic RA, et al. (2011) Sizing and phenotyping of cellular vesicles using Nano-particle Tracking Analysis. Nanomedicine (Lond Print) 7(6):780–788.

20. Thery C, Amigorena S, Raposo G, Clayton A (2006) Isolation and characterization ofexosomes from cell culture supernatants and biological fluids. Curr Protoc Cell Biol3(2006):3.22.

21. Hessvik NP, Phuyal S, Brech A, Sandvig K, Llorente A (2012) Profiling of microRNAs inexosomes released from PC-3 prostate cancer cells. Biochim Biophys Acta 1819(11-12):1154–1163.

22. Bryant RJ, et al. (2012) Changes in circulating microRNA levels associated with pros-tate cancer. Br J Cancer 106(4):768–774.

23. Ogata-Kawata H, et al. (2014) Circulating exosomal microRNAs as biomarkers of coloncancer. PLoS ONE 9(4):e92921.

24. Gajos-Michniewicz A, Duechler M, Czyz M (2014) MiRNA in melanoma-derived exo-somes. Cancer Lett 347(1):29–37.

25. Tadokoro H, Umezu T, Ohyashiki K, Hirano T, Ohyashiki JH (2013) Exosomes derivedfrom hypoxic leukemia cells enhance tube formation in endothelial cells. J Biol Chem288(48):34343–34351.

26. Duijvesz D, Luider T, Bangma CH, Jenster G (2011) Exosomes as biomarker treasurechests for prostate cancer. Eur Urol 59(5):823–831.

27. Arroyo JD, et al. (2011) Argonaute2 complexes carry a population of circulating mi-croRNAs independent of vesicles in human plasma. Proc Natl Acad Sci USA 108(12):5003–5008.

28. Turchinovich A, Weiz L, Langheinz A, Burwinkel B (2011) Characterization of extra-cellular circulating microRNA. Nucleic Acids Res 39(16):7223–7233.

29. Cheng L, Sharples RA, Scicluna BJ, Hill AF (2014) Exosomes provide a protective andenriched source of miRNA for biomarker profiling compared to intracellular and cell-free blood. J Extracell Vesicles, 10.3402/jec.v3.23743.

30. Mitchell PS, et al. (2008) Circulating microRNAs as stable blood-based markers forcancer detection. Proc Natl Acad Sci USA 105(30):10513–10518.

31. Cheng HH, et al. (2013) Circulating microRNA profiling identifies a subset of meta-static prostate cancer patients with evidence of cancer-associated hypoxia. PLoS ONE8(7):e69239.

32. Momen-Heravi F, et al. (2013) Current methods for the isolation of extracellularvesicles. Biol Chem 394(10):1253–1262.

33. Witwer KW, et al. (2013) Standardization of sample collection, isolation and analysismethods in extracellular vesicle research. J Extracell Vesicles, 10.3402/jev.v2i0.20360.

34. Redis RS, Calin S, Yang Y, You MJ, Calin GA (2012) Cell-to-cell miRNA transfer: Frombody homeostasis to therapy. Pharmacol Ther 136(2):169–174.

35. Filipe V, Hawe A, Jiskoot W (2010) Critical evaluation of Nanoparticle TrackingAnalysis (NTA) by NanoSight for the measurement of nanoparticles and proteinaggregates. Pharm Res 27(5):796–810.

36. Kroh EM, Parkin RK, Mitchell PS, Tewari M (2010) Analysis of circulating microRNAbiomarkers in plasma and serum using quantitative reverse transcription-PCR(qRT-PCR). Methods 50(4):298–301.

37. Bustin SA, Nolan T (2004) Pitfalls of quantitative real-time reverse-transcriptionpolymerase chain reaction. J Biomol Tech 15(3):155–166.

38. Hindson CM, et al. (2013) Absolute quantification by droplet digital PCR versus analogreal-time PCR. Nat Methods 10(10):1003–1005.

39. Dingle TC, Sedlak RH, Cook L, Jerome KR (2013) Tolerance of droplet-digital PCRvs real-time quantitative PCR to inhibitory substances. Clin Chem 59(11):1670–1672.

40. Kalra H, et al. (2013) Comparative proteomics evaluation of plasma exosome isolationtechniques and assessment of the stability of exosomes in normal human bloodplasma. Proteomics 13(22):3354–3364.

41. Steinman RM, Brodie SE, Cohn ZA (1976) Membrane flow during pinocytosis. Astereologic analysis. J Cell Biol 68(3):665–687.

42. Fabbri M, et al. (2012) MicroRNAs bind to Toll-like receptors to induce prometastaticinflammatory response. Proc Natl Acad Sci USA 109(31):E2110–E2116.

43. Park CK, et al. (2014) Extracellular microRNAs activate nociceptor neurons to elicitpain via TLR7 and TRPA1. Neuron 82(1):47–54.

44. Gagnon KT, Li L, Chu Y, Janowski BA, Corey DR (2014) RNAi factors are present andactive in human cell nuclei. Cell Reports 6(1):211–221.

45. Skog J, et al. (2008) Glioblastoma microvesicles transport RNA and proteins that promotetumour growth and provide diagnostic biomarkers. Nat Cell Biol 10(12):1470–1476.

46. Morello M, et al. (2013) Large oncosomes mediate intercellular transfer of functionalmicroRNA. Cell Cycle 12(22):3526–3536.

47. Delorme-Axford E, et al. (2013) Human placental trophoblasts confer viral resistanceto recipient cells. Proc Natl Acad Sci USA 110(29):12048–12053.

48. Strobl H, et al. (1997) flt3 ligand in cooperation with transforming growth factor-beta1 potentiates in vitro development of Langerhans-type dendritic cells and allows sin-gle-cell dendritic cell cluster formation under serum-free conditions. Blood 90(4):1425–1434.

49. Gatti E, et al. (2000) Large-scale culture and selective maturation of human Langer-hans cells from granulocyte colony-stimulating factor-mobilized CD34+ progenitors.J Immunol 164(7):3600–3607.

50. Lamparski HG, et al. (2002) Production and characterization of clinical grade exo-somes derived from dendritic cells. J Immunol Methods 270(2):211–226.

51. Andrews PA, Velury S, Mann SC, Howell SB (1988) cis-Diamminedichloroplatinum(II)accumulation in sensitive and resistant human ovarian carcinoma cells. Cancer Res48(1):68–73.

Chevillet et al. PNAS | October 14, 2014 | vol. 111 | no. 41 | 14893

MED

ICALSC

IENCE

S

Dow

nloa

ded

by g

uest

on

Sep

tem

ber

23, 2

020