ArticlePDF Available

Extracellular Vesicle-Mediated Modulation of Stem-like Phenotype in Breast Cancer Cells under Fluid Shear Stress

MDPI
Biomolecules
Authors:

Abstract and Figures

Circulating tumor cells (CTCs) are some of the key culprits that cause cancer metastasis and metastasis-related deaths. These cells exist in a dynamic microenvironment where they experience fluid shear stress (FSS), and the CTCs that survive FSS are considered to be highly metastatic and stem cell-like. Biophysical stresses such as FSS are also known to cause the production of extracellular vesicles (EVs) that can facilitate cell–cell communication by carrying biomolecular cargos such as microRNAs. Here, we hypothesized that physiological FSS will impact the yield of EV production, and that these EVs will have biomolecules that transform the recipient cells. The EVs were isolated using direct flow filtration with and without FSS from the MDA-MB-231 cancer cell line, and the expression of key stemness-related genes and microRNAs was characterized. There was a significantly increased yield of EVs under FSS. These EVs also contained significantly increased levels of miR-21, which was previously implicated to promote metastatic progression and chemotherapeutic resistance. When these EVs from FSS were introduced to MCF-7 cancer cells, the recipient cells had a significant increase in their stem-like gene expression and CD44⁺/CD24⁻ cancer stem cell-like subpopulation. There was also a correlated increased proliferation along with an increased ATP production. Together, these findings indicate that the presence of physiological FSS can directly influence the EVs’ production and their contents, and that the EV-mediated transfer of miR-21 can have an important role in FSS-existing contexts, such as in cancer metastasis.
This content is subject to copyright.
Citation: Brown, S.R.; Radcliffe, M.E.;
Danner, J.T.; Andújar Cruz, W.J.;
Lackey, K.H.; Park, H.-A.; Weinman,
S.T.; Kim, Y. Extracellular
Vesicle-Mediated Modulation of
Stem-like Phenotype in Breast Cancer
Cells under Fluid Shear Stress.
Biomolecules 2024,14, 757. https://
doi.org/10.3390/biom14070757
Academic Editor: Tracey Martin
Received: 20 May 2024
Revised: 21 June 2024
Accepted: 22 June 2024
Published: 25 June 2024
Copyright: © 2024 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
biomolecules
Article
Extracellular Vesicle-Mediated Modulation of Stem-like
Phenotype in Breast Cancer Cells under Fluid Shear Stress
Spenser R. Brown 1, , Margaret E. Radcliffe 1, , Joseph T. Danner 1, Wilmer J. Andújar Cruz 1,
Kimberly H. Lackey 2, Han-A Park 3, Steven T. Weinman 1and Yonghyun Kim 1 ,*
1
Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487, USA;
stweinman@eng.ua.edu (S.T.W.)
2
Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA; lacke003@ua.edu
3Department of Human Nutrition and Hospitality Management, The University of Alabama,
Tuscaloosa, AL 35487, USA; hpark36@ches.ua.edu
*Correspondence: ykim@eng.ua.edu
Current address: Oak Ridge National Laboratory, Oak Ridge, TN 37830, USA.
Current address: Graduate Biomedical Sciences, The University of Alabama at Birmingham,
Birmingham, AL 35294, USA.
Abstract: Circulating tumor cells (CTCs) are some of the key culprits that cause cancer metastasis and
metastasis-related deaths. These cells exist in a dynamic microenvironment where they experience
fluid shear stress (FSS), and the CTCs that survive FSS are considered to be highly metastatic and
stem cell-like. Biophysical stresses such as FSS are also known to cause the production of extracellular
vesicles (EVs) that can facilitate cell–cell communication by carrying biomolecular cargos such as
microRNAs. Here, we hypothesized that physiological FSS will impact the yield of EV production,
and that these EVs will have biomolecules that transform the recipient cells. The EVs were isolated
using direct flow filtration with and without FSS from the MDA-MB-231 cancer cell line, and the
expression of key stemness-related genes and microRNAs was characterized. There was a significantly
increased yield of EVs under FSS. These EVs also contained significantly increased levels of miR-
21, which was previously implicated to promote metastatic progression and chemotherapeutic
resistance. When these EVs from FSS were introduced to MCF-7 cancer cells, the recipient cells had
a significant increase in their stem-like gene expression and CD44
+
/CD24
cancer stem cell-like
subpopulation. There was also a correlated increased proliferation along with an increased ATP
production. Together, these findings indicate that the presence of physiological FSS can directly
influence the EVs’ production and their contents, and that the EV-mediated transfer of miR-21 can
have an important role in FSS-existing contexts, such as in cancer metastasis.
Keywords: extracellular vesicles; breast cancer metastasis; cancer stem cells; fluid shear stress
1. Introduction
Metastasis is the leading cause of cancer deaths worldwide [
1
], with metastatic breast
cancer being the second deadliest cancer among women, behind lung cancer [
2
]. Metastatic
breast cancer presents additional treatment challenges since the cells at the secondary
site have evolved from the phenotype of the primary tumor. These cells have traveled
as circulating tumor cells (CTCs), which affects their ability to adapt to the dynamic
environment during circulation and in secondary site colonization. While in circulation,
CTCs can encounter fluid shear stress (FSS) and immunological responses [
3
]. Although
most CTCs die in circulation, some of them survive with the potential to form distant
metastases [
4
] and are thus targeted for diagnostics [
5
,
6
]. We and others theorized that this
subset of CTCs overlaps with the cancer stem cell (CSC) subpopulation [
7
10
], which can
help explain how these cells are adaptable to dynamic environments. CSCs are known
to be capable of self-renewal, differentiation, tumor initiation, invasion, and metastatic
Biomolecules 2024,14, 757. https://doi.org/10.3390/biom14070757 https://www.mdpi.com/journal/biomolecules
Biomolecules 2024,14, 757 2 of 14
cancer propagation [
11
]. They are known to possess chemotherapeutic resistance through
efflux pumps that remove would-be toxins from the cytosol and via extracellular vesicles
(EVs) [
12
,
13
]. CSCs are also plastic, being able to convert from stem cell state to a non-
stem state when the need arises [
14
]. It is lesser known how such plasticity is conferred
and passed to other differentiated cells, especially in stressful dynamic environments.
Since stem-like behavior enables metastasis, it is of significant research interest to better
understand the mechanisms of perpetuating stemness qualities.
EVs facilitate cell-to-cell communication through the transfer of biomolecules. EVs are
lipid bilayer vesicles that contain membrane-penetrating proteins, receptors, and nucleic
acids [
13
]. They are formed by cells as a result of inward budding during endosomal matu-
ration [
15
]. There are several subsets of EVs including exosomes, endosomes, and apoptotic
bodies. Exosomes are 50–150 nm in diameter and are capable of influencing angiogenesis,
facilitating movement through tissues, and promoting cancer cell
growth [16,17].
Further,
EVs have demonstrated the abilities to confer cancer-like qualities on normal cells such
as fibroblasts and epithelial cells [
18
] and carry the integrins that direct organ-specific
metastases [
19
]. These abilities are attributed to the transfer of genetic cargo from the
EVs to recipient cells. EVs are also thought to nurture the “soil” in the “seed-and-soil”
hypothesis [
20
] for metastatic spread [
21
,
22
]. Various stresses including hypoxia, serum
starvation, and thermal stress applied to EV-producing cells have induced aberrations in
the amounts of protein and DNA expressed within the EVs produced under stress [
23
26
].
Others have acknowledged that EV-mediated preconditioning could be a key factor in
tumor progression by conveying stress-induced responses [27].
It has been demonstrated that EVs (namely exosomes) derived from stem cells have a
variant microRNA (miRNA) profile that is distinct from that of EVs isolated from non-stem
cells [
28
]. Additionally, miRNAs that are found in EVs can enhance tumor invasiveness
and metastasis [
29
]. Notably, miR-21 and miR-29a were identified as promoting breast
CSC phenotypes and being potential biomarkers of breast and lung cancer cells [
29
32
].
Though it has been shown that the application of FSS has induced the rapid production
of microparticles [
33
], to the best of our knowledge, the effects of FSS on the composition
of EVs has not been extensively reported. We hypothesize that (1) FSS influences the
composition of EVs and (2) EVs from CSC-like cells (i.e., cells with a high percentage of
CD44
+
/CD24
subpopulation) will endow a stem-like phenotype on the recipient cells.
Illuminating the relationship between EVs and the commutation of stem-like traits will
provide insight for developing targeted therapeutics. To elucidate the impact that FSS
has on stem-like phenotype via EVs, we focus on miRNAs that correlate to stemness:
miR-21 induces targeted therapy resistance, mediates metastasis, and is upregulated in
stem cell-derived EVs [
13
,
16
,
28
]; miR-16 is involved in endocrine and targeted therapy
resistances [
13
]; miR-17 has been implicated in mediating metastasis [
13
,
16
]; and miR-29a
is correlated with tumorigenicity, drug resistance, and metastasis, promoting prolifera-
tion, and has been shown to be upregulated in stem cell-derived EVs [
28
,
32
]. Here, we
present evidence that FSS directly impacts the production and composition of EVs, which
subsequently increases the stem-like phenotype and proliferation of recipient cells.
2. Materials & Methods
2.1. Cell Culture
MDA-MB-231 and MCF-7 cells were obtained from ATCC (Manassas, VA, USA).
MCF-7 was cultured in phenol red-free Dulbecco’s modified Eagle’s medium (DMEM)
(Gibco, Grand Island, NY, USA) supplemented with 10% fetal bovine serum (Gibco), 1%
penicillin/streptomycin (Corning, Corning, NY, USA), and 1% amphotericin B (Cytiva,
Marlborough, MA, USA). For EV incubation studies, the FBS was pre-depleted of native
EVs by ultracentrifugation (100,000
×
gfor 14 h). MDA-MB-231 was cultured in DMEM/F12
(Gibco) supplemented with 1X B-27 (Gibco), 1% penicillin/streptomycin (Corning), 5 mg/L
insulin (Thermo Scientific, Waltham, MA, USA), 20
µ
g/L bFGF (Shenandoah Inc., Warwick,
PA, USA), 20
µ
g/L EGF (Shenandoah Inc.), 0.5 mg/L hydrocortisone (Sigma-Aldrich,
Biomolecules 2024,14, 757 3 of 14
St. Louis, MO, USA), 8
µ
g/mL heparin (Akron Biotech, Boca Raton, FL, USA), and 1%
amphotericin B (Cytiva). Cell viability was measured using trypan blue staining (Amresco,
Solon, OH, USA) and counted using a T20 automated cell counter (Bio-Rad, Hercules,
CA, USA).
2.2. Fluid Shear Stress
Cells were inoculated into a CLS-1450 Series spinner flask (Chemglass Life Sciences,
Vineland, NJ, USA). The flasks were incubated on a Dura-Mag magnetic stirrer (Chemglass
Life Sciences) for 4 h at a wall FSS of 5 dyn/cm2. FSS was calculated using the correlation
equations of power number for the propeller [
34
]. For each FSS, 6
×
10
6
cells were inocu-
lated in 60 mL of media and cultured in T-75 flasks for 24 h post-FSS to allow cell recovery
prior to media collection for EV isolation.
2.3. EV Isolation
EVs collected via ultracentrifugation followed a differential centrifugation protocol.
Conditioned cell medium was centrifuged using a high-speed centrifuge (Avanti J-30I;
Beckman Coulter, Brea, CA, USA). The conditioned media were centrifuged at 300
×
g
for 5 min,
1000×g
for 5 min, 4000
×
gfor 10 min, 20,000
×
gfor 20 min, and, finally, at
100,000×g
for 60 min. The supernatant was transferred to a new tube following each spin,
until, ultimately, the pelleted EVs were collected after the final step.
EVs were collected by direct flow filtration (DFF), where conditioned cell culture
medium was centrifuged at 1000
×
gfor 10 min and, subsequently, at 10,000
×
gfor 10 min
to remove remaining cell debris, extracellular proteins, and apoptotic bodies. Synder LY
(PES, 100 kDa, Synder Filtration, Vacaville, CA, USA) membranes were soaked in DI H
2
O
in a 4
C refrigerator overnight. Each membrane coupon (active area = 14.6 cm
2
) was cut
and loaded into an HP4750 direct flow filtration cell (Sterlitech, Auburn, WA, USA). The
filtration cell was filled with 50 mL of DI H
2
O and pressurized to 6.9 bar
g
for 1 min to
facilitate pore wetting. Afterwards, excess water was removed from the filtration cell and
replaced with 20 mL of conditioned cell media. A complete filtration (i.e., no stirring) of
the cell media was performed at a pressure of 2 bar
g
for approximately 2.5 h. Remaining
cell media (<1 mL) were removed with a pipettor prior to disassembling the filtration cell.
The membrane coupon was removed from the filtration cell and placed in a sterilized petri
dish where the EVs were eluted from the membrane in 5 mL of 1X PBS following a 45-min
incubation at 4
C. Lastly, the collected EVs were filtered using a 200 nm Avanti extruder
five times.
2.4. Scanning Electron Microscopy (SEM)
Membrane top surface morphology was imaged with an Apreo S field-emission
scanning electron microscope (FE-SEM, Thermo Fisher Scientific, Waltham, MA, USA).
Pristine samples were dehydrated using ethanol solutions consisting of 25, 50, 75, and
~100% ethanol for 5 min apiece in a stepwise manner. Each membrane was stored in ~100%
ethanol until the sample was imaged. Prior to mounting, the dehydrated membrane was
transferred to a solution containing 50% HMDS/50% ethanol (v/v) for 15 min, and then it
was soaked in a 100% HMDS solution for at least 15 min to prevent pore collapse [
35
]. Next,
the membranes were dried in air for 5 min and mounted to an aluminum stub mount with
carbon tape, followed by placing a piece of conductive copper tape across the sample and
stub mount to form a direct connection. Samples were sputter coated with an MCM-200 Ion
Sputter Coater (SEC, Hwaseong-si, Republic of Korea) with a ~5 nm layer of gold before
they were placed inside the FE-SEM. Images were acquired using the Optiplan lens and
T1 segment A detector at a working distance of 3–5 mm, a magnification of 65,000
×
, an
accelerating voltage of 5.00 kV, a beam current of 25 pA, and a scan speed of 5.00–10.00
µ
s.
Top surface FE-SEM images were analyzed using Fiji image processing package
(version 2.14.0) [
36
] and ImageJ image processing software (version 1.54f) [
37
] to estimate
the pore size distribution of the membrane [
38
,
39
]. Similar to our previous work [
40
],
Biomolecules 2024,14, 757 4 of 14
each image was converted to an 8-bit format and the software was calibrated using the
micrograph scale bar before further processing. The color threshold was adjusted to provide
adequate contrast for the analyze particles function. The analyze particles function was
used to calculate Feret’s diameter for top surface pores between a range of 10 and 1000 nm.
Feret’s diameter provides the approximate circular pore diameter based on the longest
distance between any two points of each pore [
41
]. Results were exported to OriginPro
2023 (version 10.0.0.154) to generate the pore size distribution.
2.5. EV Verification and Quantification
The presence of EVs was qualitatively verified by transmission electron microscopy
(TEM) on a FEI TECNAI F20 TEM. Samples were fixed in 4% paraformaldehyde in PBS
(Fisher Scientific, Hampton, NH, USA) for 30 min and loaded onto a carbon coated copper
grid. Samples were allowed to dry for at least 20 min, stained with 2% uranyl acetate for
5 min, and washed with ultra-filtered DI H2O.
The presence of EVs was verified by protein analysis using the commercial Exo-
Check Exosome Array kit (System Biosciences, Palo Alto, CA, USA) according to the
manufacturer’s protocol. For imaging, the membrane was treated with the SuperSignal
West Pico PLUS Chemiluminescent Substrate system (Thermo Scientific) for imaging on
the ChemiDoc MP Imaging System with ImageLab software (v.5.1).
For total protein quantifications, EVs isolated with methods described above were
resuspended 9:1 (v/v) in 100% (200 proof) ethanol at
80
C for a minimum of 2 h to
precipitate the proteins. The precipitated protein was centrifuged at 3220
×
gfor 1 h
to concentrate the precipitate. The ethanolic supernatant was aspirated and the pellet
allowed to air dry before being resuspended in RIPA lysis buffer. BSA standards were
used for calibration and all protein samples were quantified via BCA assay measured
at 562 nm. EV protein collected was normalized to the cell number that produced the
conditioned medium.
2.6. Nanoparticle Tracking Analysis (NTA)
To determine the size and quantity of the EVs, NTA was performed with a NanoSight
NS-300 instrument running the NTA 3.4 software (Malvern Panalytical, Malvern, UK).
Samples were kept in PBS on ice until NTA was performed. In volumes of 1 mL, samples
were loaded in the sample chamber and the camera was focused. For all recordings, a
cameral level of 9 and screen gain of 9 was used. Five 60 s videos were recorded for each
sample and each sample was loaded at a flow rate of 50. Averages of particle size and
quantity from the recordings were calculated (n= 5).
2.7. qRT-PCR
EV miRNAs were isolated via Qiagen’s (Hilden, Germany) miRNeasy Serum/Plasma
kit according to the manufacturer’s protocol. Briefly, 1 mL of QIAzol Lysis Reagent was
added to 200
µ
L suspended EVs in 1X PBS and vortexed to mix. After a 5-min incubation,
200
µ
L chloroform was added to the mixture and shaken thoroughly for 15 s. After a 3 min
incubation, the lysate was centrifuged at 12,000
×
gfor 15 min at 4
C. The upper aqueous
solution was transferred to a new collection tube and supplemented with 1.5 volumes of
100% ethanol. The sample was loaded into a RNeasy MinElute spin column and centrifuged
at 10,000
×
gfor 30 s until all of the lysate had passed through the column. The spin column
was washed with two separate provided buffers followed by 80% ethanol. The column was
centrifuged at maximum speed for 5 min to dry the membrane prior to eluting the miRNA
in 14 µL RNase-free water. miRNA was quantified on a NanoDrop 2000. Complementary
DNA (cDNA) was synthesized (35 ng/
µ
L miRNA) using the Qiagen miRCURY LNA RT
kit according to the manufacturer’s protocol. qRT-PCR was performed using the Qiagen
miRCURY LNA miRNA SYBR Green PCR kit according to the manufacturer’s protocol.
Primers for specific miRNA were acquired from Qiagen via the GeneGlobe database: miR-
21 (GeneGlobe ID YP-00204230); miR-16 (GeneGlobe ID YP-00205702); miR-17 (GeneGlobe
Biomolecules 2024,14, 757 5 of 14
ID YP-02119304); miR-29a (GeneGlobe ID YP-00204698). Expressions of miRNA of interest
were normalized to hsa-miR-30e-5p (GeneGlobe ID YP-204714) as a reference marker [
42
].
Cell-derived RNA was isolated with a GeneJet RNA Purification kit (Thermo Scien-
tific) according to the manufacturer’s protocol for mammalian cultured cells. RNA was
quantified on a NanoDrop 2000. cDNA was synthesized with the qScript cDNA SuperMix
(Quanta Biosciences, Gaithersburg, MD, USA) and Mastercycler Nexus Gradient (Eppen-
dorf, Hauppauge, NY, USA) according to the manufacturer’s protocol. Applied Biosystems
PowerUp SYBR Green Master Mix was used in concordance with the manufacturer’s
instructions for real-time PCR. A StepOnePlus Real-Time PCR System was used for all
qRT-PCR and the StepOne Software (v2.3) was used for data analysis. Data were processed
using the
Ct method to measure the changes in gene and miRNA expression. The NCBI
gene database was used to select the primer sequences as shown in Table 1.
Table 1. Primer sequences used in this study.
Primer Sequence
ACTB forward GCC CTG GAC TTC GAG CAA GAG A
ACTB reverse ATG GTG ATG GAC CTG GCC GTC A
ATP5A1 forward TGG AGC CCA GCA AGA TTA CA
ATP5A1 reverse TGA TAG TGC CCA ACA AGG CT
ATP5B forward CGC AAA CAT CTC CTT CGC CA
ATP5B reverse AGT CCC TCA TCA AAC TGG ACG
ATP5G1 forward GTG AGT CTG TCA CCT TGA GCC
ATP5G1 reverse CTG CAC TCC TAC TAC CCT GCA A
ATP5G2 forward GTC AAG AGC ACC TCA CAG C
ATP5G2 reverse TCT GTC AGT ATC TCC GGT CGT
ATP5G3 forward TTA ATG GGG CCC AGA ATG GTG
ATP5G3 reverse CCA GCC ACT CCT ACT GTT GC
ATPAF1 forward TAT GTG CTC TGC CAA GAA GGG
ATPAF1 reverse AAG TGG AGT TCA GTA CCT GTC C
ATPAF2 forward TTG AAG AAA CTG GGC GTG TC
ATPAF2 reverse CTG CTT GAA CAT TCC TCA GCC
ATP-BL forward ACC TCA CAT CTG ACC CTG GA
ATP-BL reverse AAG CTT CCC TTC TTG GCC TC
NANOG forward AAT ACC TCA GCC TCC AGC AGA TG
NANOG reverse TGC GTC ACA CCA TTG CTA TTC TTC
OCT4 forward GAG AAC CGA GTG AGA GGC AAC C
OCT4 reverse CAT AGT CGC TGC TTG ATC GCT TG
2.8. Fluorescent Imaging
Isolated EVs were stained with PKH67 Green Fluorescent Cell Linker Kit for General
Cell Membrane Labeling (Sigma-Aldrich) following the manufacturer’s protocol. Briefly,
EVs were resuspended in Diluent C following final centrifugation. EVs were labeled
with the PKH67 dye, incubated for 5 min and the staining was stopped with 1% BSA
in PBS. Labeled EVs were washed with PBS prior to addition to MCF-7 for overnight
(>14 h) incubation. Proliferating cells were identified using a Ki-67 (sc-23900) (Santa Cruz
Biotechnology, Inc., Dallas, TX, USA) and Alexa Fluor 555 goat anti-mouse IgG (H + L)
(A21422) (Invitrogen, Waltham, MA, USA) antibodies according to the manufacturer’s
protocol. Cells were stained with DAPI for 10 min prior to imaging on a 20
×
objective
Biomolecules 2024,14, 757 6 of 14
using a Nikon C2+ confocal microscope (Nikon Instruments, Melville, NY, USA) in The
University of Alabama Optical Analysis Facility.
2.9. Flow Cytometry
Flow cytometry was performed using a Bio-Rad S3e Cell Sorter (Bio-Rad Labora-
tories, Hercules, CA, USA). CSC marker expression was tested using PE Mouse Anti-
Human CD24 Clone ML5 (RUO), FITC Mouse Anti-Human CD44 Clone G44-26 (also
known as C26) (RUO), PE Mouse IgG2a,
κ
Isotype Control Clone G155-178 (RUO), FITC
Mouse IgG2b
κ
Isotype Control Clone 27-35 (RUO) (BD Biosciences, Franklin Lakes,
NJ, USA, cat.# 555428, 555478, 555574, and 555742, respectively) antibodies according
to the manufacturer’s protocol. Proliferating cells were measured using a Ki-67 (sc-
23900) (Santa Cruz Biotechnology, Inc., Dallas, TX, USA) and Alexa Fluor 555 goat anti-
mouse IgG (H + L) (A21422) (Invitrogen, Waltham, MA, USA) antibodies according to the
manufacturer’s protocol.
2.10. Measurement of ATP Production
Cellular ATP production was measured using the ATPlite™ Luminescence Assay
System (PerkinElmer, Waltham, MA, USA) as previously described [
43
,
44
]. Briefly, cells
(0.1
×
10
6
/well) were lysed on the shaker for 5 min. Cells were incubated with the substrate
(luciferin) on the shaker for 10 min. The reaction between ATP, luciferase and luciferin
produced bioluminescence. ATP-induced luminescence was measured using a fluorescence
microplate reader (CLARIOstar, BMG Labtech, Mornington, Australia).
2.11. Statistical Analyses
Results were analyzed using the Student’s unpaired t-test and one-way ANOVA
(GraphPad Prism, La Jolla, CA, USA). Tukey and Fisher tests were used to determine
significance, with p-values < 0.05 presenting as significant.
3. Results
3.1. Direct Flow Filtration Produces Higher EV Recovery Than Differential Ultracentrifugation
To compare a traditional EV-isolation method with our novel approach, we collected
EVs produced in MDA-MB-231 serum-free media by differential ultracentrifugation (UC)
and direct flow filtration (DFF; Figure 1A). The results of the top surface image analysis
indicate that almost all of the pores are <100 nm. This confirmed that the membrane PES
pore sizes are smaller than those of EVs (>150 nm and <200 nm, Figure 2), allowing the
EVs to be collected and concentrated as the retentate. We characterized the EVs from both
isolation methods by TEM and protein analysis. The size and morphology of the EVs
derived was consistent across both methods (Figure 1B). To further support successful
isolation of EVs, we performed protein analysis to assess the absence of contaminating
cellular proteins and the presence of established exosome markers. Both the UC-and DFF-
derived EVs were negative for the cell marker GM130 and showed comparable amounts
of the cytosolic exosome marker ALIX, but the DFF EVs demonstrated a relatively higher
concentration of CD81 (Figure 1C). For more substantive evidence of increased EV recovery,
we measured the total protein isolated from both methods (Figure 1D). DFF EVs produced
a higher mass of total protein, suggesting an overall higher recovery. This is supportive of
using DFF as a novel method of EV isolation since the qualities of the isolated particles are
consistent with the traditional centrifugation method, but with the advantages of a higher
recovery and a significantly less laborious, varied, and time-consuming process.
Biomolecules 2024,14, 757 7 of 14
Biomolecules2024,14,xFORPEERREVIEW7of15
Figure1.Ver icationofisolationofextracellularvesicles(EVs)producedbyserum-freeMDA-MB-
231cells.(A)Representativetopsurfacescanningelectronmicrograph(FE-SEM)ofapre-ltration
SynderLYmembraneusedforEVisolation(left;scalebar:100nm)andhistogramdemonstrating
sizedistributionofporesizes(right).(B)Representativetransmissionelectronmicrograph(TEM)
imageofEVsderivedusingultracentrifugation(UC;left)andofEVsderivedusingdirectowl-
tration(DFF;right).(C)ExosomeAntibodyArrayofMDA-MB-231wholecelllysate,UC-derived
EVproteins,andDFF-derivedEVproteins(50µgeach).Numbersrepresentrelativeintensityofthe
bandscomparedtothepositivecontrol.OriginalWesternblotimagesareavailableinSupplemen-
taryMaterials.(D)TotalproteinisolatedfromEVsperonemillioncells.n≥3;mean±standarderror;
***p<0.001viaStudent’st-testwithWelchcorrections.
Figure2.ImpactofFSSonEVsfromserum-freeMDA-MB-231cells.(A)TEMimagesofEVsderived
fromstaticandFSScultures.Scalebars:200nm.(B)MeanparticlesizeofEVsbasedonNTA.(C)
ConcentrationofEVsderivedfromstaticandFSSculturesdeterminedthroughNTA.(D)EVprotein
isolatedfollowingethanolprecipitation.n≥3;mean±standarderror;*p<0.05;ns=notsignicant
viaStudent’st-testwithWelch corrections.
Figure 1. Verification of isolation of extracellular vesicles (EVs) produced by serum-free MDA-MB-
231 cells. (A) Representative top surface scanning electron micrograph (FE-SEM) of a pre-filtration
Synder LY membrane used for EV isolation (left; scale bar: 100 nm) and histogram demonstrating
size distribution of pore sizes (right). (B) Representative transmission electron micrograph (TEM)
image of EVs derived using ultracentrifugation (UC; left) and of EVs derived using direct flow
filtration (DFF; right). (C) Exosome Antibody Array of MDA-MB-231 whole cell lysate, UC-derived
EV proteins, and DFF-derived EV proteins (50
µ
g each). Numbers represent relative intensity of the
bands compared to the positive control. Original Western blot images are available in Supplementary
Materials. (D) Total protein isolated from EVs per one million cells. n
3; mean
±
standard error;
*** p< 0.001 via Student’s t-test with Welch corrections.
Biomolecules2024,14,xFORPEERREVIEW7of15
Figure1.Ver icationofisolationofextracellularvesicles(EVs)producedbyserum-freeMDA-MB-
231cells.(A)Representativetopsurfacescanningelectronmicrograph(FE-SEM)ofapre-ltration
SynderLYmembraneusedforEVisolation(left;scalebar:100nm)andhistogramdemonstrating
sizedistributionofporesizes(right).(B)Representativetransmissionelectronmicrograph(TEM)
imageofEVsderivedusingultracentrifugation(UC;left)andofEVsderivedusingdirectowl-
tration(DFF;right).(C)ExosomeAntibodyArrayofMDA-MB-231wholecelllysate,UC-derived
EVproteins,andDFF-derivedEVproteins(50µgeach).Numbersrepresentrelativeintensityofthe
bandscomparedtothepositivecontrol.OriginalWeste rnblotimagesareavailableinSupplemen-
taryMaterials.(D)TotalproteinisolatedfromEVsperonemillioncells.n≥3;mean±standarderror;
***p<0.001viaStudent’st-testwithWelchcorrections.
Figure2.ImpactofFSSonEVsfromserum-freeMDA-MB-231cells.(A)TEMimagesofEVsderived
fromstaticandFSScultures.Scalebars:200nm.(B)MeanparticlesizeofEVsbasedonNTA.(C)
ConcentrationofEVsderivedfromstaticandFSSculturesdeterminedthroughNTA.(D)EVprotein
isolatedfollowingethanolprecipitation.n≥3;mean±standarderror;*p<0.05;ns=notsignicant
viaStudent’st-testwithWelchcorrections.
Figure 2. Impact of FSS on EVs from serum-free MDA-MB-231 cells. (A) TEM images of EVs derived
from static and FSS cultures. Scale bars: 200 nm. (B) Mean particle size of EVs based on NTA.
(C) Concentration of EVs derived from static and FSS cultures determined through NTA. (D) EV
protein isolated following ethanol precipitation. n
3; mean
±
standard error; * p< 0.05; ns = not
significant via Student’s t-test with Welch corrections.
Biomolecules 2024,14, 757 8 of 14
3.2. Fluid Shear Stress Increases the Quantity of EVs Produced
Cellular stress is known to affect the production and composition of EVs, so we sought
to investigate the impact of FSS on the size and quantity of EVs produced by serum-free
MDA-MB-231 cells. Although the sizes of EVs did not change (Figure 2A,B), nanoparticle
tracking analysis revealed that cells under FSS produced nearly triple the amount of EVs
than the static control (Figure 2C). The results were further supported by the total protein
quantification of EVs derived from static and FSS cultures (Figure 2D). Together, those data
suggest that FSS induced increased the production of EVs.
3.3. Fluid Shear Stress Increases Stemness-Related miRNA in EVs
Previous work showed that the application of FSS can enrich the CSC-like subpopula-
tion (CD44
+
/CD24
) in MCF-7 breast cancer cells but not in MDA-MB-231 breast cancer
cells that already have a high percentage of CSC-like subpopulation [
45
]. Here, we ob-
served a similar phenotype, where FSS did not increase the expression of stemness markers
NANOG and OCT4 in MDA-MB-231 (Figure 3A). The same was true for various miRNAs
(miR-21, miR-17, miR-16, and miR-29a) that are known to mediate stemness and/or metas-
tasis (Figure 3B). Interestingly, when analyzing for the same stemness-related miRNAs in
the isolated EVs, there was a statistically significant increased expression of them (except for
miR-29a) in those from the shear stressed cells (Figure 3C). These results suggest that FSS
EVs have increased miRNA cargos that can modulate stemness to their neighboring cells.
Biomolecules2024,14,xFORPEERREVIEW8of15
3.2.FluidShearStressIncreasestheQuantityofEVsProduced
CellularstressisknowntoaecttheproductionandcompositionofEVs,sowe
soughttoinvestigatetheimpactofFSSonthesizeandquantityofEVsproducedbyse-
rum-freeMDA-MB-231cells.AlthoughthesizesofEVsdidnotchange(Figure2A,B),na-
noparticletrackinganalysisrevealedthatcellsunderFSSproducednearlytriplethe
amountofEVsthanthestaticcontrol(Figure2C).Theresultswerefurthersupportedby
thetotalproteinquanticationofEVsderivedfromstaticandFSScultures(Figure2D).
Together,thosedatasuggestthatFSSinducedincreasedtheproductionofEVs.
3.3.FluidShearStressIncreasesStemnessRelatedmiRNAinEVs
PreviousworkshowedthattheapplicationofFSScanenrichtheCSC-likesubpopu-
lation(CD44+/CD24)inMCF-7breastcancercellsbutnotinMDA-MB-231breastcancer
cellsthatalreadyhaveahighpercentageofCSC-likesubpopulation[45].Here,weob-
servedasimilarphenotype,whereFSSdidnotincreasetheexpressionofstemnessmark-
ersNANOGandOCT4inMDA-MB-231(Figure3A).ThesamewastrueforvariousmiR-
NAs(miR-21,miR-17,miR-16,andmiR-29a)thatareknowntomediatestemnessand/or
metastasis(Figure3B).Interestingly,whenanalyzingforthesamestemness-relatedmiR-
NAsintheisolatedEVs,therewasastatisticallysignicantincreasedexpressionofthem
(exceptformiR-29a)inthosefromtheshearstressedcells(Figure3C).Theseresultssug-
gestthatFSSEVshaveincreasedmiRNAcargosthatcanmodulatestemnesstotheir
neighboringcells.
Figure3.ExpressionofstemnessmarkersandmicroRNAsinserum-freeMDA-MB-231cellsand
correspondingEVswithandwithoutFSS.(A)RNAlevelsofstemness-relatedgenesNANOGand
OCT4normalizedtoACTB.(B)miRNAlevelsinstatic-andFSS-culturedMDA-MB-231cellsand
(C)intheircorrespondingEVsnormalizedtoreferencemiRNAmarkermiR-30e.n=3biological
replicateseachwith2or3technicalreplicates;mean±standarderror;**p<0.01;****p<0.0001;ns
=notsignicantviaStudent’st-test.
Figure 3. Expression of stemness markers and microRNAs in serum-free MDA-MB-231 cells and
corresponding EVs with and without FSS. (A) RNA levels of stemness-related genes NANOG and
OCT4 normalized to ACTB. (B) miRNA levels in static- and FSS-cultured MDA-MB-231 cells and
(C) in their corresponding EVs normalized to reference miRNA marker miR-30e. n= 3 biological
replicates each with 2 or 3 technical replicates; mean
±
standard error; ** p< 0.01; **** p< 0.0001;
ns = not significant via Student’s t-test.
Biomolecules 2024,14, 757 9 of 14
3.4. MCF-7 Cells Receiving EVs from MDA-MB-231 Showed Increased Stem-Like Signature
We hypothesized that the uptake of EVs from MDA-MB-231 would transform the
recipient cells, particularly towards an increased stem-like phenotype. To test this, EVs col-
lected from static and FSS-treated MDA-MB-231 were labeled with fluorescent membrane
label PKH67 and incubated overnight with unlabeled MCF-7 cells in a serum-depleted
culture medium. We observed the successful uptake of these PKH67-labeled EVs in MCF-7
recipient cells after 24 h (Figure 4A). In particular, we observed EV agglomeration on the
same Z-plane as the cell nucleus contrary to a halo around the nucleus, suggesting cellular
uptake of EVs and their retention within the cells. We then analyzed the miRNA in the
MCF-7 cells after incubation with the EVs. Significantly, both miR-21 and miR-29a showed
increased expression in the recipient cells, especially for those with FSS EVs (Figure 4B).
We further assayed for the CSC-like CD44
+
/CD24
population. Interestingly, this sub-
population was only significantly increased in FSS EV-treated MCF-7 cells (Figure 4C),
suggesting that not all stemness-related miRNA changes directly correlate to changes in
CD44
+
/CD24
subpopulation. Nevertheless, these results suggest that the EV uptake does
indeed transform the recipient cells into an increased stem-like signature.
Biomolecules2024,14,xFORPEERREVIEW9of15
3.4.MCF7CellsReceivingEVsfromMDAMB231ShowedIncreasedStemLikeSignature
WehypothesizedthattheuptakeofEVsfromMDA-MB-231wouldtransformthe
recipientcells,particularlytowardsanincreasedstem-likephenotype.Totestthis,EVs
collectedfromstaticandFSS-treatedMDA-MB-231werelabeledwithuorescentmem-
branelabelPKH67andincubatedovernightwithunlabeledMCF-7cellsinaserum-de-
pletedculturemedium.WeobservedthesuccessfuluptakeofthesePKH67-labeledEVs
inMCF-7recipientcellsafter24h(Figure4A).Inparticular,weobservedEVagglomera-
tiononthesameZ-planeasthecellnucleuscontrarytoahaloaroundthenucleus,sug-
gestingcellularuptakeofEVsandtheirretentionwithinthecells.Wethenanalyzedthe
miRNAintheMCF-7cellsafterincubationwiththeEVs.Signicantly,bothmiR-21and
miR-29ashowedincreasedexpressionintherecipientcells,especiallyforthosewithFSS
EVs(Figure4B).WefurtherassayedfortheCSC-likeCD44+/CD24population.Interest-
ingly,thissubpopulationwasonlysignicantlyincreasedinFSSEV-treatedMCF-7cells
(Figure4C),suggestingthatnotallstemness-relatedmiRNAchangesdirectlycorrelateto
changesinCD44+/CD24subpopulation.Nevertheless,theseresultssuggestthattheEV
uptakedoesindeedtransformtherecipientcellsintoanincreasedstem-likesignature.
Figure4.ImpactofEVsderivedfromCSC-likeMDA-MB-231cellsonMCF-7cells.(A)Uptakeof
PKH67-labeledEVsinMCF-7cells.Lefttoright:brighteld,DAPInuclearstain,PKH67-labeled
EVs,andmerged.Scalebar:25µm.(B)miRNAexpressionsofstemness-relatedmarkersinMCF-7
cellsafter24hincubationwithPBScontrol(PBS),staticcell-derivedEVs(static),andFSS-derived
EVs(FSS).ExpressionswerenormalizedtoreferencemiRNAmarkermiR-30e.n=3biologicalrep-
licateseachwith2or3technicalreplicates;mean±standarderror;*p<0.05;**p<0.01;***p<0.001;
****p<0.0001;ns=notsignicantviaone-wayANOVA.(C)Cancerstemcell-likesubpopulation
(CD44+/CD24;lowerrightquadrant)ofMCF-7cellsafter24hofEVintroductionmeasuredviaow
cytometry(n=3).
Figure 4. Impact of EVs derived from CSC-like MDA-MB-231 cells on MCF-7 cells. (A) Uptake of
PKH67-labeled EVs in MCF-7 cells. Left to right: brightfield, DAPI nuclear stain, PKH67-labeled EVs,
and merged. Scale bar: 25
µ
m. (B) miRNA expressions of stemness-related markers in MCF-7 cells
after 24 h incubation with PBS control (PBS), static cell-derived EVs (static), and FSS-derived EVs
(FSS). Expressions were normalized to reference miRNA marker miR-30e. n= 3 biological replicates
each with 2 or 3 technical replicates; mean
±
standard error; * p< 0.05; ** p< 0.01; *** p< 0.001;
**** p< 0.0001; ns = not significant via one-way ANOVA. (C) Cancer stem cell-like subpopulation
(CD44
+
/CD24
; lower right quadrant) of MCF-7 cells after 24 h of EV introduction measured via
flow cytometry (n= 3).
Biomolecules 2024,14, 757 10 of 14
3.5. MCF-7 Cells Receiving EVs from MDA-MB-231 Showed Increased Proliferation and
ATP Production
Upon collecting the MCF-7 cells, it was noted that the samples treated with the EVs
showed a higher number of live cells than the negative control (Figure 5A). Indeed, there
was a higher percentage of Ki-67-postive proliferative cells when treated with either static
EVs or FSS EV cells compared to the untreated control (Figure 5B,C). To test whether the
EV-induced changes in proliferation were associated with energy metabolism, we measured
the amounts of ATP produced in the MCF-7 cells (Figure 5D). The samples treated with the
FSS EV showed a significant increase in intracellular ATP levels compared to the untreated
control. This was further corroborated when assaying for the expressions of genes encoding
for the
α
-,
β
-, and c-subunits of F
1
Fo ATP synthase (Figure 5E), the enzyme complex that
governs cellular energy metabolism. Overall, the results showed a statistically significant
increase in the
α
- and c-subunit genes in the FSS EV-treated MCF-7 cells compared to the
untreated control.
Biomolecules2024,14,xFORPEERREVIEW10of15
3.5.MCF7CellsReceivingEVsfromMDAMB231ShowedIncreasedProliferationandATP
Production
UponcollectingtheMCF-7cells,itwasnotedthatthesamplestreatedwiththeEVs
showedahighernumberoflivecellsthanthenegativecontrol(Figure5A).Indeed,there
wasahigherpercentageofKi-67-postiveproliferativecellswhentreatedwitheitherstatic
EVsorFSSEVcellscomparedtotheuntreatedcontrol(Figure5B,C).Totestwhetherthe
EV-inducedchangesinproliferationwereassociatedwithenergymetabolism,wemeas-
uredtheamountsofATPproducedintheMCF-7cells(Figure5D).Thesamplestreated
withtheFSSEVshowedasignicantincreaseinintracellularATP levelscomparedtothe
untreatedcontrol.Thiswasfurthercorroboratedwhenassayingfortheexpressionsof
genesencodingfortheα-,β-,andc-subunitsofF1FoATPsynthase(Figure5E),theenzyme
complexthatgovernscellularenergymetabolism.Overall,theresultsshowedastatisti-
callysignicantincreaseintheα-andc-subunitgenesintheFSSEV-treatedMCF-7cells
comparedtotheuntreatedcontrol.
Figure5.ImpactonMCF-7proliferation24hpost-EVintroduction.(A)LivecellcountsofMCF-7
withPBScontrol(PBS),staticcell-derivedEVs(static),andFSS-derivedEVs(FSS).(B)Percent-posi-
tiveKi-67populationsquantiedusingowcytometry.(C)Ki-67stainingindicatingproliferating
cells.Lefttoright:DAPInuclearstain,Ki-67,andmerged.Scalebar:50µm.(D)ATP measurements
inMCF-7cellsquantiedusingluminescence.(E)RNAlevelsofF1FoATPsynthasec-subunitgenes
(ATP 5G1,ATP 5G 2,andAT P5 G3 ),α-subunitgenes(AT P5 AandAT PAF2 ),andβ-subunitgenes
(ATPA F1,ATP5 B,andATPBL)normalizedtohousekeepinggeneACTB.n=3biologicalreplicates
eachwith2or3technicalreplicates;mean±standarderror;*p<0.05;**p<0.01;***p<0.001;****p
<0.0001;ns=notsignicantviaone-wayANOVA.
4.Discussion
Inthisstudy,wecomparedtraditionalEVisolationmethodviaUCtoanovelap-
proachviaDFF.Furthermore,weexploredtheimpactthatFSShasonEVproductionand
Figure 5. Impact on MCF-7 proliferation 24 h post-EV introduction. (A) Live cell counts of MCF-7
with PBS control (PBS), static cell-derived EVs (static), and FSS-derived EVs (FSS). (B) Percent-positive
Ki-67 populations quantified using flow cytometry. (C) Ki-67 staining indicating proliferating cells.
Left to right: DAPI nuclear stain, Ki-67, and merged. Scale bar: 50
µ
m. (D) ATP measurements
in MCF-7 cells quantified using luminescence. (E) RNA levels of F
1
Fo ATP synthase c-subunit
genes (ATP5G1,ATP5G2, and ATP5G3),
α
-subunit genes (ATP5A and ATPAF2), and
β
-subunit genes
(ATPAF1,ATP5B, and ATP-BL) normalized to housekeeping gene ACTB.n= 3 biological replicates
each with 2 or 3 technical replicates; mean
±
standard error; * p< 0.05; ** p< 0.01; *** p< 0.001;
**** p< 0.0001; ns = not significant via one-way ANOVA.
4. Discussion
In this study, we compared traditional EV isolation method via UC to a novel approach
via DFF. Furthermore, we explored the impact that FSS has on EV production and on the
miRNAs contained within the EVs, as well as the translated effects of those EVs on recipient
MCF-7 cells.
We observed that DFF was superior to traditional differential UC in recovering higher
quantity EVs without impacting their quality (Figure 1). UC is a laborious and highly
variable method in isolating EVs [
46
]. In contrast, DFF allows for large-scale isolation of
EVs with higher purity [
47
,
48
], motivating our approach. Presently, there is a high interest
to adopt EVs as targeted therapeutic delivery vehicles, so it is advantageous to identify
Biomolecules 2024,14, 757 11 of 14
and validate a scalable and reproducible EV isolation process. With TEM, we showed that
the EVs isolated from both methods produced comparable particles of similar size and
morphology, consistent with previously reported imaging data [
49
]. Interestingly, protein
marker analysis indicated EVs had similar levels of ALIX, but higher levels of CD81 via
DFF than via UC. It is possible that DFF preferentially selects for CD81-positive EVs, and
this will need to be explored further in future studies.
We hypothesized that FSS would increase the production of EVs like other previously
reported stresses [
26
]. While the bulk population size of the EVs did not significantly
change (Figure 2A,B), there was a significant increased production of EVs due to the FSS
(Figure 2C). The static culture cells produced approximately 100 EVs per cell, whereas the
FSS-exposed cells produced 350 EVs per cell. Thus, our results suggest that sublethal FSS
can have beneficial effects in the biomanufacturing of EVs.
We previously demonstrated that the application of FSS preferentially enriched the
CSC-like CD44
+
/CD24
subpopulation in MCF-7 but not in the more aggressive MDA-MB-
231 [
45
,
50
]. Here, key stemness-related genes and miRNAs did not change within the cells
after FSS, but the miRNA cargos in the secreted EVs showed a significant increase (Figure 3).
To our knowledge, this is the first reporting of such an effect by FSS on EVs derived
from breast cancer cells. Previous studies showed that hypoxic and serum deprivation
stresses impact the composition of EVs [
23
26
]. Consistent with the previous literature
that demonstrated upregulation of miR-21 and miR-29a in breast cancer stem cells [
28
],
our results suggest that EVs are one of the key means by which such changes occur after
FSS. Together, these results suggest that FSS can improve the survivability of the cells via
EVs [51], while also communicating with the nearby cells through them.
It is particularly interesting that miR-21 showed such a high increase upon EV uptake
(Figure 4A,B), as it is correlated to numerous oncological processes including invasion,
migration, and targeting of tumor suppressor genes associated with proliferation [
52
,
53
].
miR-21 has previously been shown to localize in mitochondria, where it upregulates cy-
tochrome B, ultimately resulting in increased ATP production [
54
]. miR-21 also has multiple
genetic targets including tumor suppressors PTEN and PCDC4, which play critical roles in
CSC maintenance [
55
57
]. Additionally, miR-21 has been shown to play a role in the regula-
tion of cellular energy metabolism in cancer-associated fibroblasts [
58
,
59
]. Our data suggest
that increased miR-21 in receiving cells is indeed correlated to increased stem-like signature
(Figure 4C), proliferation, and energy metabolism (Figure 5), suggesting a potential role of
miR-21 favoring cancer cell energy metabolism. We further postulate that the transfer of
miR-21, using EVs as a key delivery vehicle and mechanism of communication, is an impor-
tant mechanism by which metastasizing cancer cells potentially ward off chemotherapies
and pre-condition their microenvironment to increase stem cell-like characteristics.
We have previously reported that MCF-7 cells treated with FSS, without EVs, caused
the metabolic remodeling of cancer cells by decreasing the c- and
β
-subunit of the F
1
Fo
ATP synthase [
44
]. The c-subunit is responsible for forming a leak channel [
58
]; thus,
FSS-mediated c-subunit reduction may improve metabolic efficiency. Interestingly, recent
studies suggested that the leak channel can be regulated by the conformational changes in
the F
1
Fo ATP synthase [
60
]. Notably, dissociation of the c-subunit from the F
1
complex is
shown to be associated with a large channel conductance. Therefore, it is possible that the
upregulation of the F
1
complex components such as the
α
- and
β
-subunits may prevent the
opening of the leak channel by supporting the assembly of the F
1
Fo ATP synthase. Future
studies investigating the ratio of the F
1
Fo ATP synthase subunits may help elucidate the
underlying mechanisms of altered energy metabolism in EV-exposed cancer cells.
5. Conclusions
In conclusion, we have demonstrated that DFF is an effective method for isolating
EVs at a higher yield than traditional UC. We have also shown that the application of FSS
impacts the stemness-related miRNA cargos in EVs, namely miR-21. Furthermore, the
incubation of these EVs from stem cell-like and aggressive breast cancer cells can result in
Biomolecules 2024,14, 757 12 of 14
an increased expression of miR-21 and miR-29a in the recipient cells to ultimately increase
the CSC-like population and proliferative cells. Thus, our studies highlight the importance
of physiological FSS in EV production and the critical role EV-mediated transfer of key
microRNAs may play in the “seed-and-soil” mechanisms of cancer metastasis.
Supplementary Materials: The following supporting information can be downloaded at: https:
//www.mdpi.com/article/10.3390/biom14070757/s1, Original Western blot images.
Author Contributions: Conceptualization, S.R.B. and Y.K.; methodology, S.R.B., J.T.D., H.-A.P.,
S.T.W., and Y.K.; validation, S.R.B., H.-A.P., K.H.L., and Y.K.; formal analysis, S.R.B., M.E.R., J.T.D.,
H.-A.P., K.H.L., S.T.W., and Y.K.; investigation, S.R.B., M.E.R., J.T.D., and H.-A.P.; resources, H.-A.P.,
K.H.L., S.T.W., and Y.K.; writing—original draft preparation, S.R.B., M.E.R., J.T.D., H.-A.P., S.T.W.,
and Y.K.; writing—review and editing, S.R.B., J.T.D., W.J.A.C., K.H.L., H.-A.P., S.T.W., and Y.K.;
visualization, S.R.B., M.E.R., J.T.D., and W.J.A.C.; supervision, H.-A.P., K.H.L., S.T.W., and Y.K.;
project administration, Y.K.; funding acquisition, S.T.W. and Y.K. All authors have read and agreed to
the published version of the manuscript.
Funding: This work was supported by the National Science Foundation (CBET/EBMS #2000053 to
Y.K.). S.R.B. was supported by the U.S. Department of Education as a GAANN Fellow (P200A210069).
J.T.D was supported by funding from the National Science Foundation (EFMA #2029387 to S.T.W.
and others). Any opinions, findings, and conclusions or recommendations expressed in this material
are those of the authors and do not necessarily reflect the views of the National Science Foundation
or the Department of Education.
Institutional Review Board Statement: Not applicable.
Informed Consent Statement: Not applicable.
Data Availability Statement: The original contributions presented in the study are included in
the article/Supplementary Materials, and further inquiries can be directed to the corresponding
author. The raw data supporting the conclusions of this article will be made available by the authors
on request.
Acknowledgments: We would like to thank Yuping Bao and Shahriar Habib for help with initial
study designs, Yifei Sun for help with protein analyses, Johnny Goodwin (The University of Alabama
Analytical Research Center) for assistance with TEM imaging, and Shawn Williams (The Univer-
sity of Alabama at Birmingham’s High Resolution Imaging Facility) for assistance with the NTA
analysis. S.D.G.
Conflicts of Interest: The authors have no conflicts of interest to declare.
References
1. Chaffer, C.L.; Weinberg, R.A. A perspective on cancer cell metastasis. Science 2011,331, 1559–1564. [CrossRef] [PubMed]
2.
DeSantis, C.E.; Ma, J.; Gaudet, M.M.; Newman, L.A.; Miller, K.D.; Goding Sauer, A.; Jemal, A.; Siegel, R.L. Breast cancer statistics,
2019. CA Cancer J. Clin. 2019,69, 438–451. [CrossRef] [PubMed]
3.
Fidler, I.J.; Yano, S.; Zhang, R.-D.; Fujimaki, T.; Bucana, C.D. The seed and soil hypothesis: Vascularisation and brain metastases.
Lancet Oncol. 2002,3, 53–57. [CrossRef] [PubMed]
4.
Wirtz, D.; Konstantopoulos, K.; Searson, P.C. The physics of cancer: The role of physical interactions and mechanical forces in
metastasis. Nat. Rev. Cancer 2011,11, 512–522. [CrossRef] [PubMed]
5. Muller, V.; Riethdorf, S.; Rack, B.; Janni, W.; Fasching, P.A.; Solomayer, E.; Aktas, B.; Kasimir-Bauer, S.; Pantel, K.; Fehm, T.; et al.
Prognostic impact of circulating tumor cells assessed with the CellSearch System and AdnaTest Breast in metastatic breast cancer
patients: The DETECT study. Breast Cancer Res. 2012,14, R118. [CrossRef] [PubMed]
6.
Gao, Y.; Wang, Y.; He, B.; Pan, Y.; Zhou, D.; Xiong, M.; Song, Y. An Enzyme-Loaded Metal-Organic Framework-Assisted
Microfluidic Platform Enables Single-Cell Metabolite Analysis. Angew. Chem. Int. Ed. Engl. 2023,62, e202302000. [CrossRef]
[PubMed]
7.
Aktas, B.; Tewes, M.; Fehm, T.; Hauch, S.; Kimmig, R.; Kasimir-Bauer, S. Stem cell and epithelial-mesenchymal transition markers
are frequently overexpressed in circulating tumor cells of metastatic breast cancer patients. Breast Cancer Res. 2009,11, R46.
[CrossRef] [PubMed]
8.
Kasimir-Bauer, S.; Hoffmann, O.; Wallwiener, D.; Kimmig, R.; Fehm, T. Expression of stem cell and epithelial-mesenchymal
transition markers in primary breast cancer patients with circulating tumor cells. Breast Cancer Res. 2012,14, R15. [CrossRef]
[PubMed]
Biomolecules 2024,14, 757 13 of 14
9.
Theodoropoulos, P.A.; Polioudaki, H.; Agelaki, S.; Kallergi, G.; Saridaki, Z.; Mavroudis, D.; Georgoulias, V. Circulating tumor cells
with a putative stem cell phenotype in peripheral blood of patients with breast cancer. Cancer Lett. 2010,288, 99–106. [CrossRef]
10.
Yang, M.H.; Imrali, A.; Heeschen, C. Circulating cancer stem cells: The importance to select. Chin. J. Cancer Res. 2015,27, 437–449.
[CrossRef]
11. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011,144, 646–674. [CrossRef]
12.
Clarke, M.F.; Hass, A.T. Cancer stem cells. In Reviews in Cell Biology and Molecular Medicine; Meyers, R.A., Ed.; John Wiley & Sons,
Inc.: Hoboken, NJ, USA, 2006. [CrossRef]
13.
Najminejad, H.; Kalantar, S.M.; Abdollahpour-Alitappeh, M.; Karimi, M.H.; Seifalian, A.M.; Gholipourmalekabadi, M.; Sheikhha,
M.H. Emerging roles of exosomal miRNAs in breast cancer drug resistance. IUBMB Life 2019,71, 1672–1684. [CrossRef]
14.
Plaks, V.; Kong, N.; Werb, Z. The cancer stem cell niche: How essential is the niche in regulating stemness of tumor cells? Cell
Stem Cell 2015,16, 225–238. [CrossRef] [PubMed]
15.
Van Niel, G.; Porto-Carreiro, I.; Simoes, S.; Raposo, G. Exosomes: A common pathway for a specialized function. J. Biochem. 2006,
140, 13–21. [CrossRef]
16. Sharma, A. Role of stem cell derived exosomes in tumor biology. Int. J. Cancer 2018,142, 1086–1092. [CrossRef] [PubMed]
17.
Sung, B.H.; Ketova, T.; Hoshino, D.; Zijlstra, A.; Weaver, A.M. Directional cell movement through tissues is controlled by exosome
secretion. Nat. Commun. 2015,6, 7164. [CrossRef]
18.
Antonyak, M.A.; Li, B.; Boroughs, L.K.; Johnson, J.L.; Druso, J.E.; Bryant, K.L.; Holowka, D.A.; Cerione, R.A. Cancer cell-derived
microvesicles induce transformation by transferring tissue transglutaminase and fibronectin to recipient cells. Proc. Natl. Acad.
Sci. USA 2011,108, 4852–4857. [CrossRef]
19.
Hoshino, A.; Costa-Silva, B.; Shen, T.L.; Rodrigues, G.; Hashimoto, A.; Tesic Mark, M.; Molina, H.; Kohsaka, S.; di Giannatale, A.;
Ceder, S.; et al. Tumour exosome integrins determine organotropic metastasis. Nature 2015,527, 329–335. [CrossRef]
20. Fidler, I.J.; Poste, G. The “seed and soil” hypothesis revisited. Lancet Oncol. 2008,9, 808. [CrossRef] [PubMed]
21.
Dos Anjos Pultz, B.; Andrés Cordero da Luz, F.; Socorro Faria, S.; Peixoto Ferreira de Souza, L.; Cristina Brígido Tavares, P.;
Alonso Goulart, V.; Fontes, W.; Ricardo Goulart, L.; JoséBarbosa Silva, M. The multifaceted role of extracellular vesicles in
metastasis: Priming the soil for seeding. Int. J. Cancer 2017,140, 2397–2407. [CrossRef]
22.
Yang, J.; Pan, B.; Zeng, F.; He, B.; Gao, Y.; Liu, X.; Song, Y. Magnetic Colloid Antibodies Accelerate Small Extracellular Vesicles
Isolation for Point-of-Care Diagnostics. Nano Lett. 2021,21, 2001–2009. [CrossRef]
23.
De Jong, O.G.; Verhaar, M.C.; Chen, Y.; Vader, P.; Gremmels, H.; Posthuma, G.; Schiffelers, R.M.; Gucek, M.; van Balkom, B.W.
Cellular stress conditions are reflected in the protein and RNA content of endothelial cell-derived exosomes. J. Extracell. Vesicles
2012,1, 18396. [CrossRef] [PubMed]
24.
Domenis, R.; Cifu, A.; Curcio, F. The Influence of a Stressful Microenvironment on Tumor Exosomes: A Focus on the DNA Cargo.
Int. J. Mol. Sci. 2020,21, 8728. [CrossRef]
25.
Hedlund, M.; Nagaeva, O.; Kargl, D.; Baranov, V.; Mincheva-Nilsson, L. Thermal- and oxidative stress causes enhanced release
of NKG2D ligand-bearing immunosuppressive exosomes in leukemia/lymphoma T and B cells. PLoS ONE 2011,6, e16899.
[CrossRef]
26.
Li, J.; Lee, Y.; Johansson, H.J.; Mager, I.; Vader, P.; Nordin, J.Z.; Wiklander, O.P.; Lehtio, J.; Wood, M.J.; Andaloussi, S.E. Serum-free
culture alters the quantity and protein composition of neuroblastoma-derived extracellular vesicles. J. Extracell. Vesicles 2015,
4, 26883. [CrossRef] [PubMed]
27.
Kucharzewska, P.; Belting, M. Emerging roles of extracellular vesicles in the adaptive response of tumour cells to microenviron-
mental stress. J. Extracell. Vesicles 2013,2, 20304. [CrossRef] [PubMed]
28.
Li, Q.; Eades, G.; Yao, Y.; Zhang, Y.; Zhou, Q. Characterization of a stem-like subpopulation in basal-like ductal carcinoma in situ
(DCIS) lesions. J. Biol. Chem. 2014,289, 1303–1312. [CrossRef] [PubMed]
29.
Xu, D.; Di, K.; Fan, B.; Wu, J.; Gu, X.; Sun, Y.; Khan, A.; Li, P.; Li, Z. MicroRNAs in extracellular vesicles: Sorting mechanisms,
diagnostic value, isolation, and detection technology. Front. Bioeng. Biotechnol. 2022,10, 948959. [CrossRef]
30.
Humphries, B.; Wang, Z.; Yang, C. MicroRNA Regulation of Breast Cancer Stemness. Int. J. Mol. Sci. 2021,22, 3756. [CrossRef]
[PubMed]
31.
Joyce, D.P.; Kerin, M.J.; Dwyer, R.M. Exosome-encapsulated microRNAs as circulating biomarkers for breast cancer. Int. J. Cancer
2016,139, 1443–1448. [CrossRef]
32.
Wang, J.Y.; Zhang, Q.; Wang, D.D.; Yan, W.; Sha, H.H.; Zhao, J.H.; Yang, S.J.; Zhang, H.D.; Hou, J.C.; Xu, H.Z.; et al. MiR-29a: A
potential therapeutic target and promising biomarker in tumors. Biosci. Rep. 2018,38, BSR20171265. [CrossRef] [PubMed]
33.
Miyazaki, Y.; Nomura, S.; Miyake, T.; Kagawa, H.; Kitada, C.; Taniguchi, H.; Komiyama, Y.; Fujimura, Y.; Ikeda, Y.; Fukuhara, S.
High Shear Stress Can Initiate Both Platelet Aggregation and Shedding of Procoagulant Containing Microparticles. Blood 1996,88,
3456–3464. [CrossRef]
34.
Furukawa, H.; Kato, Y.; Inoue, Y.; Kato, T.; Tada, Y.; Hashimoto, S. Correlation of Power Consumption for Several Kinds of Mixing
Impellers. Int. J. Chem. Eng. 2012,2012, 106496. [CrossRef]
35.
Weinman, S.T.; Bass, M.; Pandit, S.; Herzberg, M.; Freger, V.; Husson, S.M. A switchable zwitterionic membrane surface chemistry
for biofouling control. J. Membr. Sci. 2018,548, 490–501. [CrossRef]
36.
Schindelin, J.; Arganda-Carreras, I.; Frise, E.; Kaynig, V.; Longair, M.; Pietzsch, T.; Preibisch, S.; Rueden, C.; Saalfeld, S.; Schmid,
B.; et al. Fiji: An open-source platform for biological-image analysis. Nat. Methods 2012,9, 676–682. [CrossRef] [PubMed]
Biomolecules 2024,14, 757 14 of 14
37.
Schneider, C.A.; Rasband, W.S.; Eliceiri, K.W. NIH Image to ImageJ: 25 years of image analysis. Nat. Methods 2012,9, 671–675.
[CrossRef]
38.
Masselin, I.; Durand-Bourlier, L.; Laine, J.-M.; Sizaret, P.-Y.; Chasseray, X.; Lemordant, D. Membrane characterization using
microscopic image analysis. J. Membr. Sci. 2001,186, 85–96. [CrossRef]
39.
Wang, L.; Wang, X. Study of membrane morphology by microscopic image analysis and membrane structure parameter model.
J. Membr. Sci. 2006,283, 109–115. [CrossRef]
40.
Danner, J.T.; Papier, L.G.; Callahan, M.L.; Jarrell, B.J.; Weinman, S.T. 2-Methylpyrazine: A Greener Solvent for Nonsolvent Induced
Phase Separation (NIPS) Membrane Fabrication. Ind. Eng. Chem. Res. 2024,63, 10735–10747. [CrossRef]
41.
Ferreira, T.; Rasband, W. ImageJ User Guide. Available online: https://imagej.net/ij/docs/guide/146- 30.html#toc-Subsection- 30
.2 (accessed on 13 May 2024).
42.
Liang, Y.; Ridzon, D.; Wong, L.; Chen, C. Characterization of microRNA expression profiles in normal human tissues. BMC
Genom. 2007,8, 166. [CrossRef]
43.
Park, H.A.; Licznerski, P.; Mnatsakanyan, N.; Niu, Y.; Sacchetti, S.; Wu, J.; Polster, B.M.; Alavian, K.N.; Jonas, E.A. Inhibition of
Bcl-xL prevents pro-death actions of DeltaN-Bcl-xL at the mitochondrial inner membrane during glutamate excitotoxicity. Cell
Death Differ. 2017,24, 1963–1974. [CrossRef] [PubMed]
44.
Park, H.A.; Brown, S.R.; Jansen, J.; Dunn, T.; Scott, M.; Mnatsakanyan, N.; Jonas, E.A.; Kim, Y. Fluid shear stress enhances
proliferation of breast cancer cells via downregulation of the c-subunit of the F(1)F(O) ATP synthase. Biochem. Biophys. Res.
Commun. 2022,632, 173–180. [CrossRef] [PubMed]
45.
Brown, S.R.; Bates, J.C.; Avera, A.D.; Kim, Y. Relationship between Stemness, Reactive Oxygen Species, and Epithelial-to-
Mesenchymal Transition in Model Circulating Tumor Cells. Cells Tissues Organs 2022,211, 282–293. [CrossRef] [PubMed]
46.
Nordin, J.Z.; Lee, Y.; Vader, P.; Mager, I.; Johansson, H.J.; Heusermann, W.; Wiklander, O.P.; Hallbrink, M.; Seow, Y.; Bultema, J.J.;
et al. Ultrafiltration with size-exclusion liquid chromatography for high yield isolation of extracellular vesicles preserving intact
biophysical and functional properties. Nanomedicine 2015,11, 879–883. [CrossRef] [PubMed]
47.
McNamara, R.P.; Caro-Vegas, C.P.; Costantini, L.M.; Landis, J.T.; Griffith, J.D.; Damania, B.A.; Dittmer, D.P. Large-scale, cross-flow
based isolation of highly pure and endocytosis-competent extracellular vesicles. J. Extracell. Vesicles 2018,7, 1541396. [CrossRef]
[PubMed]
48.
Mansur, S.; Habib, S.; Hawkins, M.; Brown, S.R.; Weinman, S.T.; Bao, Y. Preparation of Nanoparticle-Loaded Extracellular Vesicles
Using Direct Flow Filtration. Pharmaceutics 2023,15, 1551. [CrossRef] [PubMed]
49.
Wu, Y.; Deng, W.; Klinke, D.J., 2nd. Exosomes: Improved methods to characterize their morphology, RNA content, and surface
protein biomarkers. Analyst 2015,140, 6631–6642. [CrossRef]
50.
Triantafillu, U.L.; Park, S.; Klaassen, N.L.; Raddatz, A.D.; Kim, Y. Fluid shear stress induces cancer stem cell-like phenotype in
MCF7 breast cancer cell line without inducing epithelial to mesenchymal transition. Int. J. Oncol. 2017,50, 993–1001. [CrossRef]
[PubMed]
51.
Park, H.A.; Brown, S.R.; Kim, Y. Cellular Mechanisms of Circulating Tumor Cells During Breast Cancer Metastasis. Int. J. Mol. Sci.
2020,21, 5040. [CrossRef]
52. Feng, Y.H.; Tsao, C.J. Emerging role of microRNA-21 in cancer. Biomed. Rep. 2016,5, 395–402. [CrossRef]
53.
Yang, Y.; Chaerkady, R.; Beer, M.A.; Mendell, J.T.; Pandey, A. Identification of miR-21 targets in breast cancer cells using a
quantitative proteomic approach. Proteomics 2009,9, 1374–1384. [CrossRef] [PubMed]
54.
Liu, J.; Li, W.; Li, J.; Song, E.; Liang, H.; Rong, W.; Jiang, X.; Xu, N.; Wang, W.; Qu, S.; et al. A Novel Pathway of Functional
microRNA Uptake and Mitochondria Delivery. Adv. Sci. 2023,10, e2300452. [CrossRef] [PubMed]
55. Jenike, A.E.; Halushka, M.K. miR-21: A non-specific biomarker of all maladies. Biomark. Res. 2021,9, 18. [CrossRef] [PubMed]
56.
Luongo, F.; Colonna, F.; Calapa, F.; Vitale, S.; Fiori, M.E.; de Maria, R. PTEN Tumor-Suppressor: The Dam of Stemness in Cancer.
Cancers 2019,11, 1076. [CrossRef] [PubMed]
57.
Wang, Q.; Yang, H.S. The role of Pdcd4 in tumour suppression and protein translation. Biol. Cell 2018,110, 169–177. [CrossRef]
[PubMed]
58.
Alavian, K.N.; Beutner, G.; Lazrove, E.; Sacchetti, S.; Park, H.A.; Licznerski, P.; Li, H.; Nabili, P.; Hockensmith, K.; Graham, M.;
et al. An uncoupling channel within the c-subunit ring of the F1FO ATP synthase is the mitochondrial permeability transition
pore. Proc. Natl. Acad. Sci. USA 2014,111, 10580–10585. [CrossRef] [PubMed]
59.
Chen, S.; Chen, X.; Shan, T.; Ma, J.; Lin, W.; Li, W.; Kang, Y. MiR-21-mediated Metabolic Alteration of Cancer-associated Fibroblasts
and Its Effect on Pancreatic Cancer Cell Behavior. Int. J. Biol. Sci. 2018,14, 100–110. [CrossRef]
60.
Mnatsakanyan, N.; Park, H.A.; Wu, J.; He, X.; Llaguno, M.C.; Latta, M.; Miranda, P.; Murtishi, B.; Graham, M.; Weber, J.; et al.
Mitochondrial ATP synthase c-subunit leak channel triggers cell death upon loss of its F(1) subcomplex. Cell Death Differ. 2022,29,
1874–1887. [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual
author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to
people or property resulting from any ideas, methods, instructions or products referred to in the content.
Article
Full-text available
The existence of cancer stem cells (CSCs) in various tumors has become increasingly clear in addition to their prominent role in therapy resistance, metastasis, and recurrence. For early diagnosis, disease progression monitoring, and targeting, there is a high demand for clinical-grade methods for quantitative measurement of CSCs from patient samples. Despite years of active research, standard measurement of CSCs has not yet reached clinical settings, especially in the case of solid tumors. This is because detecting this plastic heterogeneous population of cells is not straightforward. This review summarizes various techniques, highlighting their benefits and limitations in detecting CSCs from patient samples. In addition, methods designed to detect CSCs based on secreted and niche-associated signaling factors are reviewed. Spatial and single-cell methods for analyzing patient tumor tissues and noninvasive techniques such as liquid biopsy and in vivo imaging are discussed. Additionally, methods recently established in laboratories, preclinical studies, and clinical assays are covered. Finally, we discuss the characteristics of an ideal method as we look toward the future.
Article
Full-text available
Extracellular microRNAs (miRNAs) play a critical role in horizontal gene regulation. Uptake of extracellular miRNAs by recipient cells and their intracellular transport, however, remains elusive. Here RNA phase separation is shown as a novel pathway of miRNA uptake. In the presence of serum, synthetic miRNAs rapidly self‐assembly into ≈110 nm discrete nanoparticles, which enable miRNAs’ entry into different cells. Depleting serum cationic proteins prevents the formation of such nanoparticles and thus blocks miRNA uptake. Different from lipofectamine‐mediated miRNA transfection in which majority of miRNAs are accumulated in lysosomes of transfected cells, nanoparticles‐mediated miRNA uptake predominantly delivers miRNAs into mitochondria in a polyribonucleotide nucleotidyltransferase 1(PNPT1)‐dependent manner. Functional assays further show that the internalized miR‐21 via miRNA phase separation enhances mitochondrial translation of cytochrome b (CYB), leading to increase in adenosine triphosphate (ATP) and reactive oxygen species (ROS) reduction in HEK293T cells. The findings thus reveal a previously unrecognized mechanism for uptake and delivery functional extracellular miRNAs into mitochondria.
Article
Full-text available
Extracellular vesicles (EVs) have shown great potential as cell-free therapeutics and biomimetic nanocarriers for drug delivery. However, the potential of EVs is limited by scalable, reproducible production and in vivo tracking after delivery. Here, we report the preparation of quercetin-iron complex nanoparticle-loaded EVs derived from a breast cancer cell line, MDA-MB-231br, using direct flow filtration. The morphology and size of the nanoparticle-loaded EVs were characterized using transmission electron microscopy and dynamic light scattering. The SDS-PAGE gel electrophoresis of those EVs showed several protein bands in the range of 20–100 kDa. The analysis of EV protein markers by a semi-quantitative antibody array confirmed the presence of several typical EV markers, such as ALIX, TSG101, CD63, and CD81. Our EV yield quantification suggested a significant yield increase in direct flow filtration compared with ultracentrifugation. Subsequently, we compared the cellular uptake behaviors of nanoparticle-loaded EVs with free nanoparticles using MDA-MB-231br cell line. Iron staining studies indicated that free nanoparticles were taken up by cells via endocytosis and localized at a certain area within the cells while uniform iron staining across cells was observed for cells treated with nanoparticle-loaded EVs. Our studies demonstrate the feasibility of using direct flow filtration for the production of nanoparticle-loaded EVs from cancer cells. The cellular uptake studies suggested the possibility of deeper penetration of the nanocarriers because the cancer cells readily took up the quercetin-iron complex nanoparticles, and then released nanoparticle-loaded EVs, which can be further delivered to regional cells.
Article
Full-text available
Colonization of cancer cells at secondary sites, a decisive step in tumor metastasis, is strongly dependent on the formation of metastatic microenvironments regulated by intrinsic single‐cell metabolism traits. Herein, we report a single‐cell microfluidic platform for high‐throughput dynamic monitoring of tumor cell metabolites to evaluate tumor malignancy. This microfluidic device empowers efficient isolation of single cells (>99 %) in a squashed state similar to tumor extravasation, and employs enzyme‐packaged metal–organic frameworks to catalyze tumor cell metabolites for visualization. The microfluidic evaluation was confirmed by in vivo assays, suggesting that the platform allowed predicting the tumorigenicity of captured tumor cells and screening metabolic inhibitors as anti‐metastatic drugs. Furthermore, the platform efficiently detected various aggressive cancer cells in unprocessed whole blood samples with high sensitivity, showing potential for clinical application.
Article
Full-text available
MicroRNAs (miRNAs) are a class of short, single-stranded, noncoding RNAs, with a length of about 18–22 nucleotides. Extracellular vesicles (EVs) are derived from cells and play a vital role in the development of diseases and can be used as biomarkers for liquid biopsy, as they are the carriers of miRNA. Existing studies have found that most of the functions of miRNA are mainly realized through intercellular transmission of EVs, which can protect and sort miRNAs. Meanwhile, detection sensitivity and specificity of EV-derived miRNA are higher than those of conventional serum biomarkers. In recent years, EVs have been expected to become a new marker for liquid biopsy. This review summarizes recent progress in several aspects of EVs, including sorting mechanisms, diagnostic value, and technology for isolation of EVs and detection of EV-derived miRNAs. In addition, the study reviews challenges and future research avenues in the field of EVs, providing a basis for the application of EV-derived miRNAs as a disease marker to be used in clinical diagnosis and even for the development of point-of-care testing (POCT) platforms.
Article
Full-text available
Mitochondrial ATP synthase is vital not only for cellular energy production but also for energy dissipation and cell death. ATP synthase c-ring was suggested to house the leak channel of mitochondrial permeability transition (mPT), which activates during excitotoxic ischemic insult. In this present study, we purified human c-ring from both eukaryotic and prokaryotic hosts to biophysically characterize its channel activity. We show that purified c-ring forms a large multi-conductance, voltage-gated ion channel that is inhibited by the addition of ATP synthase F1 subcomplex. In contrast, dissociation of F1 from FO occurs during excitotoxic neuronal death suggesting that the F1 constitutes the gate of the channel. mPT is known to dissipate the osmotic gradient across the inner membrane during cell death. We show that ATP synthase c-subunit knock down (KD) prevents the osmotic change in response to high calcium and eliminates large conductance, Ca2+ and CsA sensitive channel activity of mPT. These findings elucidate the gating mechanism of the ATP synthase c-subunit leak channel (ACLC) and suggest how ACLC opening is regulated by cell stress in a CypD-dependent manner.
Article
Full-text available
Recent advances in our understanding of breast cancer have demonstrated that cancer stem-like cells (CSCs, also known as tumor-initiating cell (TICs)) are central for progression and recurrence. CSCs are a small subpopulation of cells present in breast tumors that contribute to growth, metastasis, therapy resistance, and recurrence, leading to poor clinical outcome. Data have shown that cancer cells can gain characteristics of CSCs, or stemness, through alterations in key signaling pathways. The dysregulation of miRNA expression and signaling have been well-documented in cancer, and recent studies have shown that miRNAs are associated with breast cancer initiation, progression, and recurrence through regulating CSC characteristics. More specifically, miRNAs directly target central signaling nodes within pathways that can drive the formation, maintenance, and even inhibition of the CSC population. This review aims to summarize these research findings specifically in the context of breast cancer. This review also discusses miRNAs as biomarkers and promising clinical therapeutics, and presents a comprehensive summary of currently validated targets involved in CSC-specific signaling pathways in breast cancer.
Article
Full-text available
miRNA-21 is among the most abundant and highly conserved microRNAs (miRNAs) recognized. It is expressed in essentially all cells where it performs vital regulatory roles in health and disease. It is also frequently claimed to be a biomarker of diseases such as cancer and heart disease in bodily-fluid based miRNA studies. Here we dissociate its contributions to cellular physiology and pathology from its potential as a biomarker. We show how it has been claimed as a specific predictive or prognostic biomarker by at least 29 diseases. Thus, it has no specificity to any one disease. As a result, it should not be considered a viable candidate to be a biomarker, despite its continued evaluation as such. This theme of multiple assignments of a miRNA as a biomarker is shared with other common, ubiquitous miRNAs and should be concerning for them as well.
Article
The presence of circulating cancer cells in the bloodstream is positively correlated with metastasis. We hypothesize that fluid shear stress (FSS) occurring during circulation alters mitochondrial function, enhancing metastatic behaviors of cancer cells. MCF7 and MDA-MB-231 human breast cancer cells subject to FSS exponentially increased proliferation. Notably, FSS-treated cells consumed more oxygen but were resistant to uncoupler-mediated ATP loss. We found that exposure to FSS downregulated the F1Fo ATP synthase c-subunit and overexpression of the c-subunit arrested cancer cell migration. Approaches that regulate c-subunit abundance may reduce the likelihood of breast cancer metastasis.
Article
Most cancer deaths are caused by secondary metastasized tumors. The cells that spread these tumors are known as circulating tumor cells (CTCs). They exist in a dynamic environment, including exposure to fluid shear stress (FSS) that makes them susceptible to reactive oxygen species (ROS) generation. There are questions about the similarities of CTCs to cancer stem cells (CSCs) and whether the stem cell-like characteristics of CTCs allow them to proliferate and spread despite the biophysical obstacles during the metastatic process. One of those qualities is the ability to undergo the epithelial-to-mesenchymal transition (EMT). Here, MDA-MB-231 and MCF7 were modeled as CTCs by prolonged exposure to FSS using a spinner flask. They were tested for ROS generation, CSC, EMT, and Hippo pathway gene and protein markers using qRT-PCR and flow cytometry. MDA-MB-231 did not show significant changes in CSC markers, but did show significant changes in ROS, EMT, and Hippo markers ( p < 0.05). Similarly, MCF7 showed significant changes in ROS and EMT markers ( p < 0.05). Furthermore, both cell lines demonstrated the reverse mesenchymal-to-epithelial transition signature when allowed to recover after FSS. These results suggest that the degree of their stemness or aggressiveness affects their responses to externally applied biophysical forces and demonstrates a potential link between mechanotransduction, the Hippo pathway, and the induction of EMT in breast cancer cells.