ArticlePDF AvailableLiterature Review

Abstract

Microparticles (MPs) or ectosomes are small enclosed fragments (from 0.2 to 2 μm in diameter) released from the cellular plasma membrane. Several oncogenic molecules have been identified inside MPs, including soluble proteins XIAP, survivin, metalloproteinases, CX3CL1, PYK2 and other microRNA-related proteins; membrane proteins EGFR, HER-2, integrins and efflux pumps; and messenger RNAs and microRNAs miR-21, miR-27a, let-7, miR-451, among others. Studies have shown that MPs transfer their cargo to neoplastic or non-malignant cells and thus contribute to activation of oncogenic pathways, resulting in cell survival, drug resistance and cancer dissemination. This review summarizes recent findings on MP biogenesis and the role of the MPs cargo in cancer and discusses some of the RNAs and proteins involved. In addition, the discussion covers evidence of (1) how and which signaling pathways can be activated by MPs in recipient cells; (2) recipient cell-type selectivity in incorporation of proteins and RNAs transported by MPs; and (3) how upon stimulation, stromal cells release MPs, promoting resistance to chemotherapeutics and invasiveness in cancer cells.
1 3
J Cancer Res Clin Oncol (2016) 142:1395–1406
DOI 10.1007/s00432-015-2029-8
REVIEW – CANCER RESEARCH
Membrane microparticles: shedding new light into cancer cell
communication
Paloma Silva de Souza1 · Roberta Soares Faccion1 · Paula Sabbo Bernardo2 ·
Raquel Ciuvalschi Maia1
Received: 12 June 2015 / Accepted: 5 August 2015 / Published online: 19 August 2015
© Springer-Verlag Berlin Heidelberg 2015
Keywords Microparticles · Multidrug resistance ·
Cancer · Intercellular communication · MicroRNAs · Akt
and ERK signaling pathways
Introduction
A comprehensive classification of the different types of
shed vesicles is a main concern in the intercellular com-
munication research field. However, the complexity of this
task begins with confusing nomenclature, which includes
oncosomes, microvesicles (MVs), microparticles (MPs),
endosomes, exosomes and ectosomes. Usually, microvesi-
cle is considered to be the collective term for both MPs and
exosomes. MPs may be called ectosomes or, as recently
proposed, oncosomes, in the case of cancer-derived MPs.
The diameter of MPs may range between 0.2 and 2 µm
(Piccin et al. 2007; Simak et al. 2004), and exosomes are
usually approximately 50–100 nm in diameter. The meth-
odologies available to distinguish MPs from exosomes
by size may result in overlapping pools of the different
microvesicles. Biochemically, MPs are derived from the
plasma membrane and thus usually display membrane bio-
markers such as PS or lipid raft proteins, whereas exosomes
are derived from intracellular endosomes and present dis-
tinct biomarkers. In this review, we focus on studies that
specifically isolate MPs to describe their role in cancer
communication and their content.
Membrane microparticles: What are they?
MPs are small vesicles formed from the cytoplasmic mem-
brane that can be released from normal or cancer cells
(termed oncosomes) (El Andaloussi et al. 2013; Hugel
Abstract
Background Microparticles (MPs) or ectosomes are small
enclosed fragments (from 0.2 to 2 μm in diameter) released
from the cellular plasma membrane. Several oncogenic
molecules have been identified inside MPs, including solu-
ble proteins XIAP, survivin, metalloproteinases, CX3CL1,
PYK2 and other microRNA-related proteins; membrane
proteins EGFR, HER-2, integrins and efflux pumps; and
messenger RNAs and microRNAs miR-21, miR-27a,
let-7, miR-451, among others. Studies have shown that
MPs transfer their cargo to neoplastic or non-malignant
cells and thus contribute to activation of oncogenic path-
ways, resulting in cell survival, drug resistance and cancer
dissemination.
Discussion and Conclusion This review summarizes
recent findings on MP biogenesis and the role of the MPs
cargo in cancer and discusses some of the RNAs and pro-
teins involved. In addition, the discussion covers evidence
of (1) how and which signaling pathways can be activated
by MPs in recipient cells; (2) recipient cell-type selectivity
in incorporation of proteins and RNAs transported by MPs;
and (3) how upon stimulation, stromal cells release MPs,
promoting resistance to chemotherapeutics and invasive-
ness in cancer cells.
Paloma Silva de Souza, Roberta Soares Faccion and Paula Sabbo
Bernardo have contributed equally to this work.
* Raquel Ciuvalschi Maia
rcmaia@inca.gov.br
1 Laboratório de Hemato-Oncologia Celular e Molecular,
Programa de Hemato-Oncologia Molecular, Brazilian
National Cancer Institute (INCA), Rio de Janeiro, Brazil
2 Programa de Pós-Graduação em Oncologia, INCA, Rio de
Janeiro, Brazil
1396 J Cancer Res Clin Oncol (2016) 142:1395–1406
1 3
et al. 2005). Commonly, MPs are characterized by expo-
sure of phosphatidylserine (PS) residues on their outer sur-
faces. However, because MPs are preferentially released
from membrane lipid rafts, they may also contain raft-
related proteins, such as P-selectin glycoprotein ligand-1
(PSGL-1) and tissue factor (TF) in monocyte-derived MPs
(Del Conde et al. 2005) and other markers, depending on
the cell type, such as CD44 for breast cancer cell-derived
MPs (BCC-MPs) and ezrin for leukemic cell-derived MPs
and BCC-MPs (Jaiswal et al. 2013). Phospholipids are
distributed in a specific manner in the bilayer membrane,
but during MP shedding, an energy-dependent mechanism
rearranges the PS to outer membrane. In this process, the
flippase, floppase and scramblase enzymes respond to
increasing intracellular levels of Ca2+ and promote phos-
pholipid redistribution. In parallel, MP maturation is initi-
ated by membrane restructuring and cytoplasmic protru-
sions with cytoskeletal rearrangement and degradation.
These events are mediated by proteins such as gelsolin
and calpain allowing MP shedding (Del Conde et al. 2005;
Enjeti et al. 2008; Salzer et al. 2002).
In tumor cell-derived MPs, others proteins also contrib-
ute to MP shedding. The ADP-ribosylation factor (ARF),
which belongs to ARF family of small GTPases, has been
associated with membrane traffic, including internaliza-
tion of ligands and the organization of the actin cytoskel-
eton (D’Souza-Schorey and Chavrier 2006). Muralidharan-
Chari and colleagues demonstrated that ARF6 modulates
MP shedding via the phospholipase D, extracellular-sig-
nal-regulated kinases (ERK) and myosin light chain path-
way. They also observed that ARF6 inhibition blocks MP
shedding (Muralidharan-Chari et al. 2009). In addition,
Pasquier and colleagues showed the importance of ARF6
upregulation in MPs in mediating a vascular metastatic
microenvironment. Inhibition or deletion of ARF6 trig-
gered a reduction in MP shedding by endothelial and can-
cer cells (Pasquier et al. 2014). Furthermore, Schllienger
et al. (2014) demonstrated that ARF1 activity is required
for the shedding of MPs, invadopodia maturation and
MMP-9 activity, suggesting that ARF1 is important not
only to MP biogenesis but also to MP-mediated cancer cell
invasiveness. They showed that ARF1 modulates RhoA and
RhoC activity, which in turn activate myosin light chain
(MLC) phosphorylation.
Another important protein is P2X7, a non-selective cat-
ion channel belonging to the family of P2X receptors. This
family is associated with several cellular processes such as
cell-to-cell communication, secretory activity and mem-
brane excitability. Its activation induces a sustained Ca2+
influx and consequently changes in phospholipid asym-
metry followed by the release of MPs (Roger et al. 2014).
Several groups have previously demonstrated that P2X7
stimulation induces MP release in normal hematopoietic
cells (Qu and Dubyak 2009). Qu et al. (2007) even sug-
gested that P2X7 itself might be carried by MPs. In the
context of cancer, Constantinescu et al. (2010) further dem-
onstrated that P2X7 activity stimulates MP release from
murine erythroleukemia cells, while inhibition of P2X7
activity significantly decreases MP release. Although they
did not describe the contents of the MPs, they provided the
first evidence that P2X7 is important for cancer-derived MP
release.
Other proteins described as being involved in MP bio-
genesis are transglutaminase 2 (TG2) and neurokinin 1
receptor (NK1R). Transglutaminases (TGs) are enzymes
involved in protein transamidation. TG2, also called tis-
sue TG (tTG), is a member of the TG family that plays a
role in cellular signaling and in cytoskeletal organization.
TG2 mediates cell adhesion in association with integrins
and fibronectin (Lorand and Graham 2003). In addition,
van der Akker et al. (2012) suggested that cross-linking of
TG2 with fibronectin and the association of this complex
with cytoskeletal elements are required to form a structural
foundation for the MP in smooth muscle cells. Conversely,
Antonyak and colleagues (2011) demonstrated that TG2
was not necessary for cancer MP biogenesis and release,
on the basis that its inhibition did not alter MP shedding
(Antonyak et al. 2011). Regarding NK1R, activation of this
protein induces rapid cell shape changes (membrane bleb-
bing) (Meshki et al. 2009). Chen et al. (2012) observed that
activation of NK1R in human embryonic kidney cells, but
not in human glioblastoma cells, induces membrane bleb-
bing followed by MP release via a ROCK and dynamic
activity-dependent mechanism. Together, these studies sug-
gest that tumor cell-derived MP biogenesis deserves further
analysis. As few reports have investigated the role of TG2
and NK1R, at this point it is not clear whether tumor cells
may present different mechanisms of MP biogenesis than
normal cells.
Although the mechanism of MP release has not been
clearly characterized, the endosomal sorting complex
required for transport (ESCRT) machinery has been shown
to be required. Like other membrane-related processes, MP
membrane fission and consequently MP release involve the
core ESCRT machinery, assembled in three types of com-
plexes on the membrane. These complexes are recruited by
site-specific adaptors as well as other associated proteins
such as the ATPase and vacuolar protein sorting 4 (VPS4).
Mechanistic models have been proposed in which ESCRT-
III filaments and VPS4 interact to catalyze membrane fis-
sion (Jouvenet 2012; McCullough et al. 2013).
The interaction between MPs and recipient cells is a
fundamental process in cancer cell communication. Some
studies have hypothesized that the exposed PS on the MP
surface may bind to cellular PS receptors, leading to MP
recognition and consequently, cargo transfer. Binding of
1397J Cancer Res Clin Oncol (2016) 142:1395–1406
1 3
PS to PS receptors has been described as an important pro-
cess for the identification and removal of apoptotic cells
and apoptotic bodies (large apoptotic vesicles) (Fadok et al.
1998; Hoffmann et al. 2001). Park and colleagues demon-
strated that BAI1 receptor binds to PS on apoptotic cells
and then promotes the phagocytic removal of their apop-
totic bodies (Park et al. 2007). Tim4 and Tim1 were also
identified as PS receptors associated with engulfment of
cells undergoing apoptosis (Miyanishi et al. 2007).
Usually, membrane fusion events are dependent on
ligand–receptor interactions. However, studies have sug-
gested that MP may also be engulfed by the endosome
pathway or the cargo may be transferred by a physical
interaction between MP and the recipient cell, as reviewed
by (Mause and Weber 2010).
Microparticles and receptor proteins
EGFR
The epidermal growth factor receptor (EGFR) is a trans-
membrane glycoprotein member of the ErbB family, which
consists of four members: EGFR (also known as ErbB1
or HER1), ErbB2 (HER2), ErbB3 (HER3) and ErbB4
(HER4). Similar to the other family members, EGFR acti-
vation culminates in changes in gene expression, rearrange-
ment of the cytoskeleton, increased cell proliferation and
inhibition of apoptosis. In several epithelial tumors, EGFR
is constitutively activated. The EGFRvIII mutation pro-
duces a constitutively active receptor, which is associated
with an unfavorable cancer prognosis. The EGFRvIII muta-
tion is the most frequent genetic alteration in glioblastoma
(GBM), accounting for 34–63 % of the cases. However,
it has been shown to arise in a variety of other epithelial
tumor types such as breast (20–78 % of cases) and lung
carcinomas (16–39 % of cases). The EGFRvIII mutated
protein can activate a variety of signaling pathways, such
as Src/Stat3, PI3K/Akt, Raf/MEK1/2/ERK1/2, Beclin and
NFκB, among others (Gan et al. 2013; Jutten and Rouschop
2014).
Al-Nedawi et al. (2008) were the first to detect EGR-
FvIII in GBM-derived MP. They observed that: (1)
EGFRvIII expression in glioma cells induces the release
of microvesicles containing flotillin-1, which is a com-
mon lipid raft protein. This observation indicates that
these microvesicles are indeed MPs derived from lipid
rafts-rich membrane regions, (2) these MP merge with
the plasma membrane of cells that express the wild-type
EGFR, and (3) this fusion further confers the oncogenic
activity of EGFRvIII to the recipient cells, which begin to
display activation of MAPK and Akt pathways, enhanced
expression of EGFRvIII-upregulated genes, morphological
transformation and increased anchorage-independent
growth ability. The authors concluded that these glioma-
derived MPs may be loaded with EGFRvIII and that
blockage of MP exchange (for instance, using annexin
derivatives) may have therapeutic potential (Al-Nedawi
et al. 2008).
In a subsequent study, Al-Nedawi et al. (2009) fur-
ther demonstrated that these EGFR-containing MPs may
also be taken up by other cell types, such as non-malig-
nant endothelial cells, and showed that (1) the recipient
cells begin to exhibit EGFR-dependent signaling includ-
ing MAPK and Akt phosphorylation and (2) MP transfer
can indeed be inhibited by PS blockers such as annexin V.
Furthermore, they observed that EGFR transfer through
glioma-derived MPs is associated with the onset of VEGF
(vascular endothelial growth factor) expression and auto-
crine activation of its receptor (VEGFR), consistent with
previous findings that microvascular density was reduced
by PS blockers. Diannexin (an annexin analogous) treat-
ment even resulted in reduced tumor growth. Thus, they
concluded that the uptake of glioma-derived MPs by nor-
mal endothelial cells renders these cells cooperative with
the tumor, which in turn provides the endothelial cells with
an angiogenic stimulus that favors tumor development (Al-
Nedawi et al. 2009).
Together, these studies provided great insight into
tumor cell communication within the tumor microenviron-
ment because they demonstrated the lack of a requirement
for cell–cell contact in order to transfer the EGFRvIII
mutation phenotype horizontally. They argued that glioma
cells can share EGFRvIII with wild-type tumor cells, via
MPs converting them to a more aggressive phenotype,
and with stromal cells, switching them to a tumor-prone
phenotype.
HER2
Human epidermal growth factor receptor 2 (HER2/neu/
Erb2) is also a member of the EGFR family. It shares many
of its properties with EGFR. HER2 overexpression is a
common feature of breast (15–30 % of cases) and ovarian
(20–30 % of cases) cancer, but it also occurs in a number of
others malignancies (head and neck, esophagus, stomach,
colon, bladder, lung, etc.), and it may be associated with
gene amplification (Iqbal and Iqbal 2014).
Recently, Liebhardt et al. (2010) identified HER2
expression in MPs from breast cancer patients. Although
it did not reach statistical significance, the level of HER2-
positive MPs were elevated in patients with lymph node
metastasis, which suggests that the presence of this Erb
family member may be involved in the metastatic ability of
breast cancer cells (Liebhardt et al. 2010). This observation
deserves further investigation.
1398 J Cancer Res Clin Oncol (2016) 142:1395–1406
1 3
Mac‑1 integrins
Macrophage1 antigen, or Mac-1, is an αMβ2-integrin, also
known as complement receptor 3 (CR3) or CD11/CD18b.
It is a leukocyte adhesion receptor of the β2-integrin fam-
ily, which consists of major players in the leukocyte adhe-
sion cascade (Mitroulis et al. 2015). Although normally
expressed only in neutrophils and monocytes, Mac-1 has
been found to play an important role in MP-driven metastasis
of murine hepatocarcinoma cells (Ma et al. 2013). Ma et al.
(2013) observed that upon activation with PMA (phorbol
myristate acetate), myeloid splenic cells produce and release
MPs loaded with a variety of immune cell markers, including
CD3, CD19, F4/80 and Gr-1. When co-incubated with hep-
atocarcinoma cells, these MPs are taken up by tumor cells,
but the MPs do not co-localize with lysosomes, ER or Golgi,
suggesting an endosome-independent pathway of uptake
that preserves the MPs in the cytoplasm. Moreover, tumor
cells that received MPs derived from PMA-treated tumor
cells (PMA-MP) grow in several tissues and yield a worse
prognosis. Furthermore, PMA-MP-treatment of both mela-
noma and hepatocarcinoma cells causes a tenfold increase in
migration and invasion, suggesting that the MPs derived from
activated immune cells could enhance the invasiveness of
diverse tumor cell types. Blocking either the CD11b or CD18
subunits of Mac-1 inhibits transwell migration of most cells,
diminishes adhesion of tumor cells to the endothelium and
improves the overall survival of mice, suggesting that Mac-1
is a fundamental player in the metastasis promoted by acti-
vated immune cell-derived MPs. Thus, the authors suggested
that activated immune cell-derived MPs may be taken up by
tumor cells, providing the tumor with immune cells typical
migration features, and facilitating metastasis. Interestingly,
Mitroulis et al. (2015) recently reviewed endogenous inhibi-
tors that modify integrin functions and could thus be impli-
cated as promising therapeutic targets (Mitroulis et al. 2015).
CXCR4
CXCR4 is a membrane receptor for CXCL12 (stromal
cell-derived factor-1), which is a potent chemoattractant
for human progenitor cells. CXCL12 mediates homing to
the bone marrow, survival, proliferation and even egress
to the circulation (Lapidot et al. 2005). Overexpression of
CXCR4 on human CD34+ progenitors increases their pro-
liferation, migration and repopulation in mice (Kahn et al.
2004), whereas disruption of CXCL12/CXCR4-mediated
cell anchorage induces the release of hematopoietic progen-
itors as well as mature cells from the bone marrow into the
circulation (Lapidot et al. 2005). In cancer, the CXCL12/
CXCR4 interaction has been implicated in acute myeloid
leukemia (AML) progression and in solid tumor growth
and metastasis (Juarez and Bendall 2004; Zlotnik 2004).
Kalinkovich et al. (2006) described CXCR4 as well as
its ligand, CXCL12, in acute myeloid leukemia (AML)-
derived MP. CXCR4 molecules could be transferred from
MPs to AML cells, enhancing migration toward CXCL12
in vitro and homing to the bone marrow of mice. CXCR4
inhibition diminished these effects, suggesting that
CXCR4-loaded MPs are involved in the progression of
AML. The levels of CXCR4 and CXCL12 in MPs were
also enhanced in the serum of AML patients compared
with normal subjects, indicating that these proteins could
be used as biomarkers for the disease (Kalinkovich et al.
2006).
Osteopontin
Osteopontin (OPN) is a small integrin-binding ligand
N-linked glycoprotein (SIBLING). SIBLING proteins bind
to cell surface integrins and CD44 in normal tissues and
function as signal transducers to promote cell adhesion,
motility and survival through the activation of kinase cas-
cades and transcription factors. They can also regulate pro-
cesses such as inflammation, immune response and cancer
(Bellahcene et al. 2008).
In both non-malignant and cancer cells, OPN has been
shown to activate the PI-3K/AKT/NFκB pathway, which
promotes cellular proliferation and differentiation, and
inhibits apoptosis (Gimba and Tilli 2013). Interaction with
CD44 and integrins has been shown to activate this OPN-
dependent signaling. In cancer, OPN is also involved in
invasion, extracellular matrix degradation, metastasis,
inflammation/complement evasion and angiogenesis (Bel-
lahcene et al. 2008; Ramchandani and Weber 2015).
Recently, Fremder et al. (2014) detected a high OPN
content in tumor cell-derived MP. Although OPN deple-
tion from MP is not sufficient to reduce tumor growth, they
observed that when bone marrow-derived pro-angiogenic
cells (BMDC) from paclitaxel-treated mice were injected
into OPN-depleted tumor-bearing mice, tumor growth and
BMDC infiltration were greatly inhibited (Fremder et al.
2014). These data suggest that OPN-loaded MPs may play
important roles in tumor homing and BMDC mobilization,
which facilitates angiogenesis.
Microparticles and cancer resistance
Multidrug resistance (MDR) is one of the major obstacles
for chemotherapy in several types of neoplasia. The main
mechanism of MDR is the overexpression of P-glycopro-
tein (Pgp/ABCB1), a transporter protein involved in the
efflux of anticancer drugs. Pgp belongs to the ABC super-
family of transporters, and others members of this family
including multidrug resistance-associated protein 1 (MRP1
1399J Cancer Res Clin Oncol (2016) 142:1395–1406
1 3
or ABCC1) and breast cancer resistance protein (BCRP or
ABCG2) are also associated with the efflux of chemical
compounds in cancer (Gottesman et al. 2002; Vasconcelos
et al. 2011).
In 2009, Bebawy and colleagues demonstrated that Pgp
is carried by MPs derived from drug-resistant cancer cells
and can be transferred to drug-sensitive cells. Their work
also showed transfer of functional Pgp and consequently
MDR phenotype acquisition in recipient cells after MPs
interaction (Bebawy et al. 2009). In another study, the same
group identified the Pgp mRNA and also ABCC1 mRNA in
MPs derived from resistant cancer cells and their transfer
to recipient cells (Lu et al. 2013). They also showed MDR-
related miRNAs in the contents of drug-resistant cell-
derived MPs (Jaiswal et al. 2012a).
Jaiswal and co-workers (2013) hypothesized that Pgp
transfer by MPs can occur selectively. They showed that
MPs derived from resistant breast cancer cells could only
transfer their MDR protein content to malignant breast
cells and not to non-malignant cells, while resistant leu-
kemic cell-derived MPs could transfer MDR proteins to
both malignant and non-malignant cells. These results sug-
gest MPs derived from breast cancer cells display cell-type
selectivity on the transfer of MDR features. In this regard,
the authors suggested that the CD44 carried by MPs derived
from breast cancer cells could contribute to the horizontal
transfer of MDR proteins (Jaiswal et al. 2013). Addition-
ally, our group showed that Pgp protein and mRNA as well
as miRNAs related to the MDR phenotype are carried in
MPs derived from myeloid leukemia cells and that these
molecules can be transferred to sensitive breast and lung
cancer cells, displaying no cell-type selectivity (de Souza
et al. 2015). Moreover, Pasquier et al. (2014) showed that
endothelial cells are recipient cells for the MPs derived
from both epithelial and mesenchymal cancer cells, sug-
gesting that endothelial cells might be targets for MPs of
both origins.
A proteomic study demonstrated that nineteen cytoskel-
etal proteins and ten proteins involved in regulation of
the actin cytoskeletal pathway were detected in the MPs
derived from MDR breast cancer cells. These results sug-
gest that these proteins are important for stabilizing Pgp in
MPs (Pokharel et al. 2014). Among the identified proteins,
CD44, ezrin, radixin and moesin appeared to be involved
in the resistance phenotype and metastasis (Donatello et al.
2012). Moreover, CD44 has been demonstrated to interact
with Pgp and also to be associated with cell migration and
invasiveness (Miletti-Gonzalez et al. 2005), suggesting that
the presence of CD44 in MPs may be required at least to
facilitate the MDR phenotype transfer from solid tumor
cells to drug-sensitive recipient cells (Pokharel et al. 2014).
Ezrin, radixin and moesin proteins were shown to
be important for the interaction between the plasma
membrane and cytoskeletal proteins (Arpin et al. 2011).
The presence of all of these proteins in MPs may facilitate
the transfer and intracellular trafficking of Pgp. Probably,
other membrane proteins are also involved in Pgp trans-
fer to drug-sensitive cancer cells via MPs (Pokharel et al.
2014). In addition, ezrin plays an important role in Pgp
membrane insertion. This protein is carried in MPs, and
its expression is unchanged in recipient cells. Therefore, it
has been suggested that ezrin is related to Pgp stabilization
in MPs and incorporation by the drug-sensitive recipient
cells (Brambilla et al. 2012; Jaiswal et al. 2013; Luciani
et al. 2002).
Pokharel et al. (2014) also observed CD73 loaded in
the MPs derived from MDR breast cancer cells. Their data
suggest that Pgp interacts with CD73 along with CD44 in
MPs, which further suggests that CD73 may also partici-
pate in the MDR phenotype transfer from solid tumor cells
to recipient cells (Pokharel et al. 2014). Increased levels of
CD73 protein expression were found in several MDR cell
lines, and inhibition of its enzymatic activity reverses the
MDR phenotype and inhibits tumor cell growth, suggesting
that this enzyme is also involved in drug resistance (Ujhazy
et al. 1996). Moreover, in human breast cancer cells, CD73
has been shown to promote tumor angiogenesis (Wang
et al. 2013), invasion, migration, adhesion and growth
(Wang et al. 2008; Zhi et al. 2007).
The aforementioned studies demonstrate that MPs
derived from cancer cells are potent disseminators of the
drug-resistant phenotype, because MPs can interact with
sensitive cancer cells and non-malignant cells as well as
with endothelial cells. Studies have also reported the pres-
ence of MPs in the plasma of cancer patients (Fleitas et al.
2012; Savasan et al. 2004; Toth et al. 2008). Liebhardt et al.
(2010) analyzed the MPs in the plasma of breast cancer
patients and identified four major subpopulations of MPs
including a group positive for BCRP (BCRP+). The authors
observed that patients with lymph node metastases showed
elevated levels of BCRP+ MPs compared to patients with
initial disease and attributed this increase to enhanced can-
cer growth and dissemination in these patients (Liebhardt
et al. 2010).
The clinical relevance of the MPs role on pathological
processes has been comprehensively discussed by several
researchers. Gong and co-workers (2015) emphasized the
clinical importance of MPs on regarding drug resistance
and metastasis. On their review, the MPs potential on can-
cer diagnosis and prognosis was also discussed (Gong et al.
2015). In addition, El Andaloussi and co-workers (2013)
largely reviewed the role of MPs in tumor biology. They
argued about the importance of MPs in promoting cancer
progression, because MPs transport and transfer molecules
associated with tumor growth, immune system escape by
tumors and angiogenesis (El Andaloussi et al. 2013).
1400 J Cancer Res Clin Oncol (2016) 142:1395–1406
1 3
Microparticles and microRNAs
MicroRNAs (miRNAs) are small non-coding RNAs (18–
24 nucleotides) that regulate gene expression by binding
to mRNAs, leading to mRNA degradation or impairment
of translation. Briefly, miRNAs are processed by a com-
plex machinery composed by the endonucleases Drosha
and Dicer, and by a protein complex called RISC (RNA
silencing complex). The central components of RISC are
Argonaute proteins (Ago), primarily Ago-2. Ago proteins
mediate the binding of mature miRNAs to mRNA (Winter
et al. 2009). Each miRNA has hundreds of predicted tar-
gets, and it is believed that 60 % of mRNAs are regulated
by miRNAs (Jansson and Lund 2012). In cancer, they are
deregulated and involved in tumorigenesis, tumor pro-
gression, metastasis and resistance to treatment. Further-
more, miRNAs have recently been proposed as promising
biomarkers for cancer diagnosis and follow-up studies on
disease progression, because they can be detected in body
fluids, carried by MPs or associated with proteins that pro-
tect them from degradation (Fujiwara et al. 2014; Inns and
James 2015; Jackson et al. 2014; Lindner et al. 2015; Liu
and Xiao 2014; Pimentel et al. 2014). Several groups are
attempting to uncover the underlying mechanisms involved
in miRNA selection, release and transport to blood flu-
ids and their role in cancer (Arroyo et al. 2011; Li et al.
2012; Turchinovich et al. 2011). However, the majority
of the studies that have attempted to elucidate the mecha-
nism of miRNAs transfer among cells did not differenti-
ate between MPs and exosomes, analyzing them together
as MVs. Arroyo et al. (2011) demonstrated that only 15 %
of circulating miRNAs, including let-7a and miR-142-3p,
are found in MVs (exosomes and MPs). The majority of
the circulating miRNAs is found in non-vesicular frac-
tions associated with ribonucleoprotein complexes contain-
ing Ago-2 protein (Arroyo et al. 2011). The association of
miRNAs and Ago-2 in MPs was also identified in cancer
cells undergoing apoptosis and in those in which differ-
entiation had been induced, suggesting that Ago-2 confers
higher stability to miRNAs molecules (Li et al. 2012).
However, distinguishing between these types of vesi-
cles is very important because they transport different car-
gos, as demonstrated by Ji and colleagues (2014). These
authors separated exosomes from MPs derived from a
colon carcinoma cell line and demonstrated that some miR-
NAs including Let-7 family members and miR-451a are
common to all vesicles, but other miRNAs are specific for
either MPs or exosomes. They found four miRNAs selec-
tively carried in MPs: miR-675-5p, miR-7704, miR-98-5p
and miR-664a-3p (Ji et al. 2014).
Some studies have investigated the role of MPs in
the resistance to cancer treatment. Using parental acute
lymphocytic leukemia (ALL) and breast cancer cells along
with their MDR counterparts, Jaiswal and colleagues
(2012a) demonstrated the effect of MDR cell-derived MPs
on their respective parental cell lines. The authors identi-
fied transcripts of Drosha, Dicer and Ago-2 in MPs derived
from both parental and MDR cell lines. This study supports
the general MVs findings that suggest that miRNA machin-
ery proteins may be important to guarantee the activity of
the miRNA molecules (Jaiswal et al. 2012a). Additionally,
the authors also identified miR-1228*, miR-1246, miR-
1308, miR-149*, miR-455-3p, miR-638 and miR-923 in
MPs derived from both MDR cell lines. These miRNAs are
transferred to drug-sensitive cells and can activate impor-
tant pathways associated with the vesiculation of MPs,
including the calcium signaling pathway and regulation of
the actin cytoskeletal pathway, as well as pathways associ-
ated with MDR phenotype and oncogenesis (Jaiswal et al.
2012a; b).
miR‑503 downregulation and the resistance phenotype
Gong et al. (2014) identified miR-22-3p, miR-185-5p,
miR-503-5p, miR-652-3p, miR-1280 in MPs associated
with the acquisition of the MDR phenotype. Among these,
miR-503 was overexpressed in drug-sensitive cells and
downregulated in drug-resistant cells. The authors observed
that MPs derived from drug-resistant cells can inhibit miR-
503 expression in recipient cells and promote migration
and invasion (Gong et al. 2014). It was shown that miR-
503 inhibits PI3Kp85 and IKKβ and consequently, Akt
phosphorylation and NFκB activation (Yang et al. 2014).
In agreement with this, our group and others have dem-
onstrated that activation of these signaling pathways by
MPs confers a resistance phenotype (de Souza et al. 2015;
Pasquier et al. 2014; Wysoczynski and Ratajczak 2009).
miRNA-503 downregulation was also observed in lung and
hepatocellular cancers and in highly invasive breast can-
cer cells, and its overexpression induces G1 arrest, reduces
cell migration and invasiveness in vitro and in vivo (Gong
et al. 2014; Yang et al. 2014; Zhou and Wang 2011). One
study demonstrated that in response to a microbial chal-
lenge, epithelial cells overexpress CX3CL1 in a NFκB- and
Dicer-dependent manner. This effect was associated with
the downregulation of miR-503, which directly inhib-
its CX3CL1 expression (Zhou et al. 2013). CX3CL1 is a
membrane-bound chemokine that can be shed in a soluble
form or through release of MPs (Bazan et al. 1997; Cas-
tellana et al. 2009). In addition, Castellana et al. (2009)
showed that fibroblasts activated by tumor MPs in turn
release MPs containing CX3CL1 and induce a migratory
phenotype in recipient prostate cancer cells. Together, these
findings suggest that miR-503 downregulation in cancer
1401J Cancer Res Clin Oncol (2016) 142:1395–1406
1 3
cells may be mediated by Dicer, CX3CL1 and probably by
other MP cargo.
PYK2 regulation by CD44, miR‑494 and miR‑303‑3p
As miR-503 was transported by MPs but was not detected
in recipient cells, the opposite situation was observed for
proline-rich tyrosine kinase 2 (PYK2) (Gong et al. 2014).
PYK2 is a cytoplasmic tyrosine kinase of the focal adhe-
sion kinase subfamily (Fan and Guan 2011). PYK2 pro-
motes cell migration and drug resistance in many tumor
types including hepatocellular and breast cancer and is acti-
vated by the PI3K/Akt signaling pathway (Datta et al. 2015;
Geng et al. 2011). PYK2 mRNA and protein were detected
in recipient cells after co-culture with the MDR counterpart
cell line, although PYK2 was not detected in MPs. These
observations suggest that MPs carry and transfer interme-
diates that result in the inhibition of miR-503 and PYK2
expression in the recipient cells (Gong et al. 2014). Jaiswal
and colleagues demonstrated that CD44 is carried by the
MPs but is not detected in the recipient cells and that CD44
promotes PYK2 phosphorylation. This observation sug-
gests that somehow the interaction with CD44 stimulates
activation of endogenous PYK2 in the recipient cells. miR-
494 and miR-330-3p are regulators of PYK2 expression
(Gong et al. 2014) and were modulated in recipient cells.
miR-494, an inhibitor of a repressor of PYK2, was trans-
ported by MPs and incorporated into the recipient cells.
Conversely, miR-330-3p is a repressor of PYK2, and its
expression was inhibited after co-culture, suggesting that
PYK2 expression might be regulated by these miRNAs in
association with CD44 through cellular signaling pathways
(Jaiswal et al. 2013). In conclusion, the regulatory role of
MPs in cancer cells is a remarkable, complex and modestly
understood intercellular communication mechanism.
microRNAs involved with Efflux pumps
Likewise, the miRNAs involved with Pgp and MRP1 regu-
lation are transferred by MPs to drug-sensitive cancer cells.
miR-27a and miR-451 indirectly promote Pgp overexpres-
sion, while miR-326 inhibits MRP1 expression (Liang
et al. 2010). Zhu et al. (2008) demonstrated that miR-27a
and miR-451 positively regulate Pgp/ABCB1 expression.
However, other reports have demonstrated that miR-27a is
downregulated in Pgp-positive leukemia cells (Feng et al.
2011). Jaiswal et al. (2012b) demonstrated that a MDR
leukemia cell line transfers miR-326 to the sensitive coun-
terpart which is associated with MRP1 downregulation in
the cells. Moreover, the breast cancer MDR-derived MPs
transfer miR-326 and miR-27a, but the miR-326 expres-
sion did not correlate with MRP1 downregulation. MPs
carry miRNAs from the donor cells, but not all of them are
incorporated by the recipient cells. These results suggest
that the MP regulatory mechanisms that control MP for-
mation and incorporation differ among cell types (Jaiswal
et al. 2012b). Corroborating these findings, our group dem-
onstrated that the chronic myeloid leukemia (CML) cell
lines K562 and Lucena (a MDR-positive line derived from
K562 by continuous exposure to vincristine) release MPs
and that the Lucena-derived MPs carried Pgp protein and
mRNA (ABCB1). Furthermore, the co-cultures of Lucena
cell line or the Lucena-derived MP with recipient cells
(breast and lung cancer models) transferred miR-27a and
miR-451. However, an important oncogenic miRNA, miR-
21, was transferred only to the breast cancer cells, rein-
forcing the concept that transfer of cargo by MPs differs
among the recipient cell lines because it occurs in a cell-
type-dependent manner (de Souza et al. 2015; Jaiswal et al.
2013).
Signaling pathways and resistance mediated
by MPs
Several reports have demonstrated that the molecules car-
ried in cancer cell vesicles can activate diverse signaling
pathways including STAT, PI3K-Akt and MAPK, and pro-
mote angiogenesis, metastasis and treatment resistance in
the recipient cells (Lam et al. 2013). However, the major-
ity of these studies do not differentiate between MPs and
other vesicles. Here, we focus specifically on MP-related
mechanisms.
Uptream pathways: STAT, PI3K‑Akt and MAPK
STAT activation has been strongly associated with the
induction of angiogenesis, and this activation may be medi-
ated by MPs containing miRNAs. MPs released by pancre-
atic adenocarcinoma, colorectal adenocarcinoma and lung
carcinoma cells induce STAT3 activation in monocytes, an
important pathway for cytokine production and activation
of these cells (Baj-Krzyworzeka et al. 2007).
A non-pure fraction of MPs derived from human and
murine lung cancer cell lines activates the Akt and MAPK
pathways in human umbilical vein endothelial cells
(HUVEC) and in tumor-associated fibroblasts (TAF) induc-
ing the expression of pro-angiogenic factors, cytokines,
adhesion molecules and metalloproteinases (MMPs), with-
out inducing HUVEC proliferation. Once stimulated by
MPs, endothelial cells and fibroblasts, in turn, influence
tumor cells by inducing STAT3, Akt and MAPK activation
and by enhancing the metastatic potential of lung tumor
cells in vivo (Wysoczynski and Ratajczak 2009). Corrob-
orating these findings, MPs derived from prostate cancer
cells transfer MMPs to fibroblasts, activate ERK1/2 and
1402 J Cancer Res Clin Oncol (2016) 142:1395–1406
1 3
consequently induce an increase in active MMP-9. MMPs
are involved in extracellular matrix degradation allow-
ing cancer cell migration and dissemination. Additionally,
MP-induced chemoresistance in fibroblasts was shown to
be reversed by ERK1/2 inhibitors, which demonstrates the
role of this pathway in the acquired resistance. In turn, the
activated fibroblasts induce cancer cell migration and inva-
sion in a fibroblast MP-dependent manner (Castellana et al.
2009).
In a similar way, Antonyak and colleagues (2011) co-
cultured glioblastoma- and breast cancer cell-derived MPs
with non-tumoral cells. Cancer cell-derived MPs induced
Akt and ERK activation, promoting enhanced cellular
survival, growth in medium with low serum concentra-
tion and anchorage-independent growth, all of which are
characteristics of transformed cells. TG2 was found in the
outer leaflet of the MP membranes. The authors associ-
ated the oncogenic transformation of the non-tumoral
cells with the transfer of TG2 by the MPs. Nevertheless,
TG2 alone was not sufficient for cell transformation;
TG2 cross-linking with fibronectin was required. In addi-
tion, the breast cancer cell-derived MPs promoted aber-
rant growth of non-malignant mammary epithelial cells.
Finally, breast cancer cells from murine tumors secrete
MPs and induce transformation of fibroblasts in vivo
(Antonyak et al. 2011).
Pasquier et al. (2014) observed that MPs derived from
cells with a mesenchymal phenotype (M–MPs) induce pro-
liferation, motility and activation of endothelial HUVEC
cells, while MPs derived from cells with an epithelial phe-
notype (E-MPs) could not induce the same effect. M–MPs
induced Akt phosphorylation and activation of this path-
way by upregulation of Arf-6 expression. In addition, using
an endothelial cell model in which Akt was constitutively
activated, the authors demonstrated that MPs induced cel-
lular resistance to doxorubicin and taxol. The M–MPs also
induced the formation of spheres and epithelial-mesenchy-
mal transition (EMT) and as a consequence, an increase in
the pro-metastatic phenotype of tumor cells (Pasquier et al.
2014). In agreement with this study, we have previously
observed that after co-culture with MPs derived from an
MDR cell line, breast or lung cancer recipient cells become
resistant to paclitaxel and cisplatin. To further clarify the
pathways modulated in recipient cells by the MDR-posi-
tive MPs, activation of Akt pathway in breast cancer cells
and NFκB transcription factor nuclear localization in both
recipient cell lines were demonstrated (de Souza et al.
2015). Collectively, these studies strengthen the evidence
for a role of Akt pathway in the acquired resistance, dem-
onstrating once more that tumor-derived MPs can induce
a drug-resistant phenotype (de Souza et al. 2015; Pasquier
et al. 2014).
Inhibitor of apoptosis proteins
Furthermore, MPs derived from fibrosarcoma cells activate
the NFκB signaling pathway in recipient mesenchymal
stem cells. This activation is associated with the upregula-
tion of several targets of this pathway that are involved in
cell survival including MMP-1, MMP-3, BIRC3, NFκB1
and TNF. It is noteworthy that the authors of this work
studied MPs in a plasmatic membrane vesicle-enriched
fraction, containing approximately 25 % exosomes. Among
the activated genes, BIRC3 encodes a member of the inhib-
itor of apoptosis protein (IAP) family that is important for
cancer cell survival (Lozito and Tuan 2014; Verhagen et al.
2001).
Survivin and XIAP are the most studied and best char-
acterized IAP family members. These proteins are overex-
pressed in several types of cancer and are associated with
treatment resistance and poor prognosis (Faccion et al.
2012; Fulda and Vucic 2012). Our group was the first to
demonstrate that MPs transfer IAP proteins and mRNAs
including survivin, XIAP and cIAP1 to recipient cells (de
Souza et al. 2015). Honegger and colleagues demonstrated
that cervical cancer-derived extracellular vesicles carry
IAPs including survivin, cIAP1, XIAP and livin. However,
these vesicles were not a pure MP fraction. (Honegger et al.
2013). Our group also observed that after co-culture, lung
cancer cells expressed higher amounts of survivin, XIAP
and cIAP protein and mRNA (BIRC5, BIRC4 and BIRC2,
respectively), while breast cancer cells expressed higher
levels of XIAP with no increase in cIAP1 mRNA (BIRC2),
suggesting a selectivity with respect to MP cargo transfer
(de Souza et al. 2015). In addition, because XIAP mRNA
(BIRC4) was not detected in the MDR cell line-derived
MP, but was detected in recipient cells, others intermediates
carried by MPs may regulate the gene expression in recipi-
ent cells.
Activation of cellular signaling pathways by MPs is
also associated with miRNAs. The oncomiR (oncogenic
miRNA) miR-21 inhibits proteins associated with cell
survival and apoptosis pathways, including PDCD4 (pro-
grammed cell death 4) and PTEN. This inhibition induces
Akt and NFκB activation and results in cellular transfor-
mation (Iliopoulos et al. 2010). Our group identified miR-
21 as a content of leukemic MDR cell-derived MPs. We
analyzed miR-21 in recipient cells and observed an asso-
ciation of this miRNA with NFκB activation mediated by
Akt phosphorylation in these cells (de Souza et al. 2015).
In summary, MPs can mediate activation of signaling path-
ways associated with cancer survival in recipient cells by
carrying and transferring miRNAs or other regulatory mol-
ecules that modify the cellular gene/protein expression
patterns.
1403J Cancer Res Clin Oncol (2016) 142:1395–1406
1 3
Conclusion and future perspectives
In this review, we have described recent findings regarding
the contents of MPs and how these vesicles influence the
acquisition of an MDR or metastatic phenotype in recipi-
ent cells (Fig. 1). Gaining better understanding of MPs role
in cancer cell communication and its distinct role regarding
each cell type will provide future tools for cancer therapy.
Although MPs study and understanding are developing
fast, many issues concerning its role and properties have
not being clarified yet.
First, a notable and major challenge in the study of MPs
is the similarity between MPs and other cellular vesicles,
such as exosomes; because these are all small microvesi-
cles, their profiles may overlap, depending on the meth-
odology employed to study them. Thus, properly differ-
entiating the types of MVs is primordial to improve our
understanding of their specific roles. However, up until
now few studies analyze microvesicles properly, and in
most, a mixture of very different types of microvesicles
are included in the same analysis. In addition, terms such
as oncosomes, microvesicles, microparticles, endosomes,
Fig. 1 Schematic representa-
tion of a membrane microparti-
cles (MPs) budding from donor
cell and cargo content transfer-
ence to recipient cell. MPs are
released from donor cells after
increase in intracellular Ca2+
which promotes phospholip-
ids translocases response and
consequently cytoskeleton
disruption. MPs cargo can
include membrane and solubles
proteins, and microRNAs. MPs
content can be transferred to
recipient cells and promote
oncogenic pathways activation
1404 J Cancer Res Clin Oncol (2016) 142:1395–1406
1 3
exosomes and ectosomes present a challenge, as they often
appear in the nomenclature as synonyms when indeed they
are not. Thus, many controversial data are obtained because
the literature lacks standardized nomenclature. Mark-
ers to identify and differentiate these vesicles and refined
protocols for extraction and purification are also urgently
needed. To move forward and to fill the empty gaps with
reliable results, it is required to properly differentiate MVs
structures. Cellular MVs are in fact the collective of MPs
and exosomes, and they may have some common cargo
molecules. In this review, we made an effort to select stud-
ies that exclusively focus on MPs on the basis that there
are major biological differences between these and other
MVs and that studies on single forms of microvesicles are
required to properly understand the contribution of each
microvesicle type to cellular communication.
Second, modulation of MPs release, uptake and its surface
molecules have emerged as a promising therapeutic option
and several groups are trying to reduce MPs cancer cells
release and their uptake by recipient cells in order to impair
cancer progression as recently reviewed by El Andaloussi
(El Andaloussi et al. 2013). Furthermore, cellular engineer-
ing using MVs as a drug delivery system can be a less toxic
treatment option because MVs are biocompatible, can cross
biological barriers such as the blood–brain barrier (BBB), do
not activate immune responses against them and can deliver
nucleic acids and drugs, becoming an promissing tool for
gene therapy (Alvarez-Erviti et al. 2011; Zhuang et al. 2011).
Finally, further understanding the role of these vesicles in
the communication between cancer cells and the microenvi-
ronment (such as extracellular matrix elements, stromal and
immune cells) will contribute to elucidate an important gap in
our current understanding of the cancer cells network. How-
ever, there is still much more to be learnt in each one of these
fields. In conclusion, MPs play a remarkable, complex and
incompletely understood role in cancer cell communication.
Acknowledgments This work was supported by Conselho Nacional
de Desenvolvimento Científico e Tecnológico (CNPq) and Fundação
para Amparo a Pesquisa do Estado do Rio de Janeiro (FAPERJ). PSS
and RSF were supported by postdoctoral fellowships from Ministé-
rio da Saúde/Instituto Nacional de Câncer. PSB was supported by a
“Nota 10” Ph.D. scholarship from (FAPERJ).
Compliance with ethical standards
Conflict of interest The authors declare to have no conflict of inter-
est.
References
Al-Nedawi K, Meehan B, Micallef J, Lhotak V, May L, Guha A,
Rak J (2008) Intercellular transfer of the oncogenic receptor
EGFRvIII by microvesicles derived from tumour cells. Nat Cell
Biol 10(5):619–624
Al-Nedawi K, Meehan B, Kerbel RS, Allison AC, Rak J (2009)
Endothelial expression of autocrine VEGF upon the uptake of
tumor-derived microvesicles containing oncogenic EGFR. Proc
Natl Acad Sci USA 106(10):3794–3799
Alvarez-Erviti L, Seow Y, Yin H, Betts C, Lakhal S, Wood MJ (2011)
Delivery of siRNA to the mouse brain by systemic injection of
targeted exosomes. Nat Biotechnol 29(4):341–345
Antonyak MA et al (2011) Cancer cell-derived microvesicles
induce transformation by transferring tissue transglutaminase
and fibronectin to recipient cells. Proc Natl Acad Sci USA
108(12):4852–4857
Arpin M, Chirivino D, Naba A, Zwaenepoel I (2011) Emerging role
for ERM proteins in cell adhesion and migration. Cell Adhes
Migr 5(2):199–206
Arroyo JD et al (2011) Argonaute2 complexes carry a population
of circulating microRNAs independent of vesicles in human
plasma. Proc Natl Acad Sci USA 108(12):5003–5008
Baj-Krzyworzeka M, Szatanek R, Weglarczyk K, Baran J, Zembala
M (2007) Tumour-derived microvesicles modulate biological
activity of human monocytes. Immunol Lett 113(2):76–82
Bazan JF et al (1997) A new class of membrane-bound chemokine
with a CX3C motif. Nature 385(6617):640–644
Bebawy M, Combes V, Lee E, Jaiswal R, Gong J, Bonhoure A,
Grau GE (2009) Membrane microparticles mediate trans-
fer of P-glycoprotein to drug sensitive cancer cells. Leukemia
23(9):1643–1649
Bellahcene A, Castronovo V, Ogbureke KU, Fisher LW, Fedarko NS
(2008) Small integrin-binding ligand N-linked glycoproteins
(SIBLINGs): multifunctional proteins in cancer. Nat Rev Can-
cer 8(3):212–226
Brambilla D et al (2012) P-glycoprotein binds to ezrin at amino acid
residues 149–242 in the FERM domain and plays a key role in
the multidrug resistance of human osteosarcoma. Int J Cancer
130(12):2824–2834
Castellana D, Zobairi F, Martinez MC, Panaro MA, Mitolo V, Frey-
ssinet JM, Kunzelmann C (2009) Membrane microvesicles
as actors in the establishment of a favorable prostatic tumoral
niche: a role for activated fibroblasts and CX3CL1–CX3CR1
axis. Cancer Res 69(3):785–793
Chen P, Douglas SD, Meshki J, Tuluc F (2012) Neurokinin 1 recep-
tor mediates membrane blebbing and sheer stress-induced
microparticle formation in HEK293 cells. PLoS ONE
7(9):e45322
Constantinescu P, Wang B, Kovacevic K, Jalilian I, Bosman GJ,
Wiley JS, Sluyter R (2010) P2X7 receptor activation induces
cell death and microparticle release in murine erythroleukemia
cells. Biochim Biophys Acta 1798(9):1797–1804
Datta A et al (2015) Selective targeting of FAK-Pyk2 axis by alpha-
naphthoflavone abrogates doxorubicin resistance in breast can-
cer cells. Cancer Lett 362(1):25–35
de Souza PS, Cruz AL, Viola JP, Maia RC (2015) Microparticles
induce multifactorial resistance through oncogenic pathways
independently of cancer cell type. Cancer Sci 106(1):60–68
Del Conde I, Shrimpton CN, Thiagarajan P, Lopez JA (2005) Tis-
sue-factor-bearing microvesicles arise from lipid rafts and
fuse with activated platelets to initiate coagulation. Blood
106(5):1604–1611
Donatello S, Babina IS, Hazelwood LD, Hill AD, Nabi IR, Hopkins
AM (2012) Lipid raft association restricts CD44-ezrin interac-
tion and promotion of breast cancer cell migration. Am J Pathol
181(6):2172–2187
D’Souza-Schorey C, Chavrier P (2006) ARF proteins: roles in mem-
brane traffic and beyond. Nat Rev Mol Cell Biol 7(5):347–358
El Andaloussi S, Mager I, Breakefield XO, Wood MJ (2013) Extracel-
lular vesicles: biology and emerging therapeutic opportunities.
Nat Rev Drug Discov 12(5):347–357
1405J Cancer Res Clin Oncol (2016) 142:1395–1406
1 3
Enjeti AK, Lincz LF, Seldon M (2008) Microparticles in health and
disease. Semin Thromb Hemost 34(7):683–691
Faccion RS, Rezende LM, Romano Sde O, Bigni Rde S, Mendes GL,
Maia RC (2012) Centroblastic diffuse large B cell lymphoma
displays distinct expression pattern and prognostic role of apop-
tosis resistance related proteins. Cancer Invest 30(5):404–414
Fadok VA, Bratton DL, Frasch SC, Warner ML, Henson PM (1998)
The role of phosphatidylserine in recognition of apoptotic cells
by phagocytes. Cell Death Differ 5(7):551–562
Fan H, Guan JL (2011) Compensatory function of Pyk2 protein in
the promotion of focal adhesion kinase (FAK)-null mammary
cancer stem cell tumorigenicity and metastatic activity. J Biol
Chem 286(21):18573–18582
Feng DD et al (2011) Down-regulated miR-331-5p and miR-27a are
associated with chemotherapy resistance and relapse in leukae-
mia. J Cell Mol Med 15(10):2164–2175
Fleitas T et al (2012) Circulating endothelial cells and microparticles
as prognostic markers in advanced non-small cell lung cancer.
PLoS ONE 7(10):e47365
Fremder E et al (2014) Tumor-derived microparticles induce bone
marrow-derived cell mobilization and tumor homing: a process
regulated by osteopontin. Int J Cancer 135(2):270–281
Fujiwara T, Kunisada T, Takeda K, Uotani K, Yoshida A, Ochiya T,
Ozaki T (2014) MicroRNAs in soft tissue sarcomas: overview
of the accumulating evidence and importance as novel biomark-
ers. BioMed Res Int. Article ID 592868
Fulda S, Vucic D (2012) Targeting IAP proteins for therapeutic inter-
vention in cancer. Nat Rev Drug Discov 11(2):109–124
Gan HK, Cvrljevic AN, Johns TG (2013) The epidermal growth factor
receptor variant III (EGFRvIII): where wild things are altered.
FEBS J 280(21):5350–5370
Geng W et al (2011) The role of proline rich tyrosine kinase 2 (Pyk2)
on cisplatin resistance in hepatocellular carcinoma. PLoS ONE
6(11):e27362
Gimba ER, Tilli TM (2013) Human osteopontin splicing isoforms:
known roles, potential clinical applications and activated signal-
ing pathways. Cancer Lett 331(1):11–17
Gong J, Luk F, Jaiswal R, Bebawy M (2014) Microparticles mediate
the intercellular regulation of microRNA-503 and proline-rich
tyrosine kinase 2 to alter the migration and invasion capacity of
breast cancer cells. Front Oncol 4:220
Gong J, Jaiswal R, Dalla P, Luk F, Bebawy M (2015) Microparticles
in cancer: a review of recent developments and the potential for
clinical application. Semin Cell Dev Biol 40:35–40
Gottesman MM, Fojo T, Bates SE (2002) Multidrug resistance in
cancer: role of ATP-dependent transporters. Nat Rev Cancer
2(1):48–58
Hoffmann PR et al (2001) Phosphatidylserine (PS) induces PS recep-
tor-mediated macropinocytosis and promotes clearance of
apoptotic cells. J Cell Biol 155(4):649–659
Honegger A, Leitz J, Bulkescher J, Hoppe-Seyler K, Hoppe-Seyler F
(2013) Silencing of human papillomavirus (HPV) E6/E7 onco-
gene expression affects both the contents and the amounts of
extracellular microvesicles released from HPV-positive cancer
cells. Int J Cancer 133(7):1631–1642
Hugel B, Martinez MC, Kunzelmann C, Freyssinet JM (2005)
Membrane microparticles: two sides of the coin. Physiology
20:22–27
Iliopoulos D, Jaeger SA, Hirsch HA, Bulyk ML, Struhl K (2010)
STAT3 activation of miR-21 and miR-181b-1 via PTEN and
CYLD are part of the epigenetic switch linking inflammation to
cancer. Mol Cell 39(4):493–506
Inns J, James V (2015) Circulating microRNAs for the prediction of
metastasis in breast cancer patients diagnosed with early stage
disease. Breast 24(4):364–369
Iqbal N, Iqbal N (2014) Human Epidermal Growth Factor Receptor 2
(HER2) in Cancers: Overexpression and Therapeutic Implica-
tions. Mol Biol Int 2014:852748
Jackson BL, Grabowska A, Ratan HL (2014) MicroRNA in prostate
cancer: functional importance and potential as circulating bio-
markers. BMC Cancer 14:930
Jaiswal R et al (2012a) Microparticle-associated nucleic acids medi-
ate trait dominance in cancer. FASEB J 26(1):420–429
Jaiswal R, Luk F, Gong J, Mathys JM, Grau GE, Bebawy M (2012b)
Microparticle conferred microRNA profiles–implications in the
transfer and dominance of cancer traits. Mol Cancer 11:37
Jaiswal R, Luk F, Dalla PV, Grau GE, Bebawy M (2013) Breast can-
cer-derived microparticles display tissue selectivity in the trans-
fer of resistance proteins to cells. PLoS ONE 8(4):e61515
Jansson MD, Lund AH (2012) MicroRNA and cancer. Mol Oncol
6(6):590–610
Ji H, Chen M, Greening DW, He W, Rai A, Zhang W, Simpson RJ
(2014) Deep sequencing of RNA from three different extracel-
lular vesicle (EV) subtypes released from the human LIM1863
colon cancer cell line uncovers distinct miRNA-enrichment sig-
natures. PLoS ONE 9(10):e110314
Jouvenet N (2012) Dynamics of ESCRT proteins. Cell Mol Life Sci
69(24):4121–4133
Juarez J, Bendall L (2004) SDF-1 and CXCR4 in normal and malig-
nant hematopoiesis. Histol Histopathol 19(1):299–309
Jutten B, Rouschop KM (2014) EGFR signaling and autophagy
dependence for growth, survival, and therapy resistance. Cell
Cycle 13(1):42–51
Kahn J et al (2004) Overexpression of CXCR4 on human CD34+
progenitors increases their proliferation, migration, and NOD/
SCID repopulation. Blood 103(8):2942–2949
Kalinkovich A et al (2006) Functional CXCR4-expressing micropar-
ticles and SDF-1 correlate with circulating acute myelogenous
leukemia cells. Cancer Res 66(22):11013–11020
Lam D, Barre B, Guette C, Coqueret O (2013) Circulating miRNAs
as new activators of the JAK-STAT3 pathway. JAK-STAT
2(1):e22996
Lapidot T, Dar A, Kollet O (2005) How do stem cells find their way
home? Blood 106(6):1901–1910
Li L et al (2012) Argonaute 2 complexes selectively protect the cir-
culating microRNAs in cell-secreted microvesicles. PLoS ONE
7(10):e46957
Liang Z et al (2010) Involvement of miR-326 in chemotherapy resist-
ance of breast cancer through modulating expression of mul-
tidrug resistance-associated protein 1. Biochem Pharmacol
79(6):817–824
Liebhardt S et al (2010) CEA-, Her2/neu-, BCRP- and Hsp27-pos-
itive microparticles in breast cancer patients. Anticancer Res
30(5):1707–1712
Lindner K, Haier J, Wang Z, Watson DI, Hussey DJ, Hummel R
(2015) Circulating microRNAs: emerging biomarkers for diag-
nosis and prognosis in patients with gastrointestinal cancers.
Clin Sci 128(1):1–15
Liu HS, Xiao HS (2014) MicroRNAs as potential biomarkers for gas-
tric cancer. World J Gastroenterol 20(34):12007–12017
Lorand L, Graham RM (2003) Transglutaminases: crosslinking
enzymes with pleiotropic functions. Nat Rev Mol Cell Biol
4(2):140–156
Lozito TP, Tuan RS (2014) Endothelial and cancer cells interact with
mesenchymal stem cells via both microparticles and secreted
factors. J Cell Mol Med 18(12):2372–2384
Lu JF, Luk F, Gong J, Jaiswal R, Grau GE, Bebawy M (2013)
Microparticles mediate MRP1 intercellular transfer and the
re-templating of intrinsic resistance pathways. Pharmacol Res
76:77–83
1406 J Cancer Res Clin Oncol (2016) 142:1395–1406
1 3
Luciani F et al (2002) P-glycoprotein-actin association through ERM
family proteins: a role in P-glycoprotein function in human
cells of lymphoid origin. Blood 99(2):641–648
Ma J et al (2013) Innate immune cell-derived microparticles facilitate
hepatocarcinoma metastasis by transferring integrin alpha(M)
beta(2) to tumor cells. J Immunol 191(6):3453–3461
Mause SF, Weber C (2010) Microparticles: protagonists of a novel
communication network for intercellular information exchange.
Circ Res 107(9):1047–1057
McCullough J, Colf LA, Sundquist WI (2013) Membrane fission
reactions of the mammalian ESCRT pathway. Ann Rev Bio-
chem 82:663–692
Meshki J, Douglas SD, Lai JP, Schwartz L, Kilpatrick LE, Tuluc F
(2009) Neurokinin 1 receptor mediates membrane blebbing in
HEK293 cells through a Rho/Rho-associated coiled-coil kinase-
dependent mechanism. J Biol Chem 284(14):9280–9289
Miletti-Gonzalez KE et al (2005) The CD44 receptor interacts with
P-glycoprotein to promote cell migration and invasion in can-
cer. Cancer Res 65(15):6660–6667
Mitroulis I, Alexaki VI, Kourtzelis I, Ziogas A, Hajishengallis G,
Chavakis T (2015) Leukocyte integrins: role in leukocyte
recruitment and as therapeutic targets in inflammatory disease.
Pharmacol Ther 147:123–135
Miyanishi M, Tada K, Koike M, Uchiyama Y, Kitamura T, Nagata S
(2007) Identification of Tim4 as a phosphatidylserine receptor.
Nature 450(7168):435–439
Muralidharan-Chari V, Clancy J, Plou C, Romao M, Chavrier P,
Raposo G, D’Souza-Schorey C (2009) ARF6-regulated shed-
ding of tumor cell-derived plasma membrane microvesicles.
Curr Biol 19(22):1875–1885
Park D et al (2007) BAI1 is an engulfment receptor for apoptotic
cells upstream of the ELMO/Dock180/Rac module. Nature
450(7168):430–434
Pasquier J et al (2014) Microparticles mediated cross-talk between
tumoral and endothelial cells promote the constitution of a pro-
metastatic vascular niche through Arf6 up regulation. Cancer
Microenviron 7(1–2):41–59
Piccin A, Murphy WG, Smith OP (2007) Circulating microparti-
cles: pathophysiology and clinical implications. Blood Rev
21(3):157–171
Pimentel F et al. (2014) Technology in MicroRNA Profiling: Circu-
lating MicroRNAs as Noninvasive Cancer Biomarkers in Breast
Cancer. J Lab Autom [Epub ahead of print]
Pokharel D, Padula MP, Lu JF, Tacchi JL, Luk F, Djordjevic SP,
Bebawy M (2014) Proteome analysis of multidrug-resistant,
breast cancer-derived microparticles. J Extracell Vesicles.
doi:10.3402/jev.v3.24384
Qu Y, Dubyak GR (2009) P2X7 receptors regulate multiple types of
membrane trafficking responses and non-classical secretion
pathways. Purinergic Signal 5(2):163–173
Qu Y, Franchi L, Nunez G, Dubyak GR (2007) Nonclassical IL-1
beta secretion stimulated by P2X7 receptors is dependent on
inflammasome activation and correlated with exosome release
in murine macrophages. J Immunol 179(3):1913–1925
Ramchandani D, Weber GF (2015) Interactions between osteopontin
and vascular endothelial growth factor: implications for cancer.
Biochim Biophys Acta 1855(2):202–222
Roger S, Jelassi B, Couillin I, Pelegrin P, Besson P, Jiang L (2014)
Understanding the roles of the P2X7 receptor in solid tumour
progression and therapeutic perspectives. Biochimica et Bio-
physica Acta. doi:10.1016/j.bbamem.2014.10.029
Salzer U, Hinterdorfer P, Hunger U, Borken C, Prohaska R (2002)
Ca(++)-dependent vesicle release from erythrocytes involves
stomatin-specific lipid rafts, synexin (annexin VII), and sorcin.
Blood 99(7):2569–2577
Savasan S, Buyukavci M, Buck S, Ravindranath Y (2004) Leukaemia/
lymphoma cell microparticles in childhood mature B cell neo-
plasms. J Clin Pathol 57(6):651–653
Schlienger S, Campbell S, Claing A (2014) ARF1 regulates the Rho/
MLC pathway to control EGF-dependent breast cancer cell
invasion. Mol Biol Cell 25(1):17–29
Simak J, Holada K, Risitano AM, Zivny JH, Young NS, Vostal JG
(2004) Elevated circulating endothelial membrane microparti-
cles in paroxysmal nocturnal haemoglobinuria. Br J Haematol
125(6):804–813
Toth B et al (2008) Circulating microparticles in breast cancer
patients: a comparative analysis with established biomarkers.
Anticancer Res 28(2A):1107–1112
Turchinovich A, Weiz L, Langheinz A, Burwinkel B (2011) Charac-
terization of extracellular circulating microRNA. Nucleic Acids
Res 39(16):7223–7233
Ujhazy P, Berleth ES, Pietkiewicz JM, Kitano H, Skaar JR, Ehrke MJ,
Mihich E (1996) Evidence for the involvement of ecto-5’-nucle-
otidase (CD73) in drug resistance. Int J Cancer 68(4):493–500
van den Akker J et al (2012) Transglutaminase 2 is secreted from
smooth muscle cells by transamidation-dependent microparticle
formation. Amino Acids 42(2–3):961–973
Vasconcelos FC, Silva KL, Souza PS, Silva LF, Moellmann-Coelho A,
Klumb CE, Maia RC (2011) Variation of MDR proteins expres-
sion and activity levels according to clinical status and evolu-
tion of CML patients. Cytom Part B Clin Cytom 80(3):158–166
Verhagen AM, Coulson EJ, Vaux DL (2001) Inhibitor of apoptosis
proteins and their relatives: IAPs and other BIRPs. Genome
Biol 2(7):3009
Wang L et al (2008) Ecto-5’-nucleotidase promotes invasion, migra-
tion and adhesion of human breast cancer cells. J Cancer Res
Clin Oncol 134(3):365–372
Wang L et al (2013) Ecto-5’-nucleotidase (CD73) promotes tumor
angiogenesis. Clin Exp Metastasis 30(5):671–680
Winter J, Jung S, Keller S, Gregory RI, Diederichs S (2009) Many
roads to maturity: microRNA biogenesis pathways and their
regulation. Nat Cell Biol 11(3):228–234
Wysoczynski M, Ratajczak MZ (2009) Lung cancer secreted
microvesicles: underappreciated modulators of microenviron-
ment in expanding tumors. Int J Cancer 125(7):1595–1603
Yang Y et al (2014) MiR-503 targets PI3K p85 and IKK-beta and sup-
presses progression of non-small cell lung cancer. Int J Cancer
135(7):1531–1542
Zhi X, Chen S, Zhou P, Shao Z, Wang L, Ou Z, Yin L (2007) RNA
interference of ecto-5’-nucleotidase (CD73) inhibits human
breast cancer cell growth and invasion. Clin Exp Metastasis
24(6):439–448
Zhou J, Wang W (2011) Analysis of microRNA expression profiling
identifies microRNA-503 regulates metastatic function in hepa-
tocellular cancer cell. J Surg Oncol 104(3):278–283
Zhou R, Gong AY, Chen D, Miller RE, Eischeid AN, Chen XM
(2013) Histone deacetylases and NF-kB signaling coordinate
expression of CX3CL1 in epithelial cells in response to micro-
bial challenge by suppressing miR-424 and miR-503. PLoS
ONE 8(5):e65153
Zhu H, Wu H, Liu X, Evans BR, Medina DJ, Liu CG, Yang JM (2008)
Role of MicroRNA miR-27a and miR-451 in the regulation of
MDR1/P-glycoprotein expression in human cancer cells. Bio-
chem Pharmacol 76(5):582–588
Zhuang X et al (2011) Treatment of brain inflammatory diseases by
delivering exosome encapsulated anti-inflammatory drugs from
the nasal region to the brain. Mol Ther 19(10):1769–1779
Zlotnik A (2004) Chemokines in neoplastic progression. Semin Can-
cer Biol 14(3):181–185
... A whole group of these instructions is called a genome (Sivadas et al. 2022). Human genome is the genetic set and genes inside the nucleus of human cells (de Souza et al. 2016). There are millions of genes on each of the chromosomes, each of which has a specific role in the cell. ...
... Each sample contains the information of one patient from one cell or tissue. Here, sampling has been done in the form of gene expression, which shows how many times a gene has produced itself (de Souza et al. 2016). ...
Article
Full-text available
Introduction In recent decades, many theories have been proposed about the cause of hereditary diseases such as cancer. However, most studies state genetic and environmental factors as the most important parameters. It has been shown that gene expression data are valuable information about hereditary diseases and their analysis can identify the relationships between these diseases. Objective Identification of damaged genes from various diseases can be done through the discovery of cell-to-cell biological communications. Also, extraction of intercellular communications can identify relationships between different diseases. For example, gene disorders that cause damage to the same cells in both breast and blood cancers. Hence, the purpose is to discover cell-to-cell biological communications in gene expression data. Methodology The identification of cell-to-cell biological communications for various cancer diseases has been widely performed by clustering algorithms. However, this field remains open due to the abundance of unprocessed gene expression data. Accordingly, this paper focuses on the development of a semi-supervised ensemble clustering algorithm that can discover relationships between different diseases through the extraction of cell-to-cell biological communications. The proposed clustering framework includes a stratified feature sampling mechanism and a novel similarity metric to deal with high-dimensional data and improve the diversity of primary partitions. Results The performance of the proposed clustering algorithm is verified with several datasets from the UCI machine learning repository and then applied to the FANTOM5 dataset to extract cell-to-cell biological communications. The used version of this dataset contains 108 cells and 86,427 promoters from 702 samples. The strength of communication between two similar cells from different diseases indicates the relationship of those diseases. Here, the strength of communication is determined by promoter, so we found the highest cell-to-cell biological communication between “basophils” and “ciliary.epithelial.cells” with 62,809 promoters. Conclusion The maximum cell-to-cell biological similarity in each cluster can be used to detect the relationship between different diseases such as cancer.
... Exosomes are nanosized EV, range approximately 50-100 nm in diameter, and are derived from membrane invagination of late endosomes within large multivesicular bodies [2,3]. MP or microvesicles (MV) are small EV ($200 nm to 1 μM in diameter), derived directly from plasma membrane budding upon release of intracellular Ca 2+ [4]. Apoptotic bodies are subcellular vesicles ranging from 1 to 5 μm in diameter, which derive from membrane blebbing of cells undergoing apoptosis [5]. ...
Chapter
Full-text available
Detection of tumor necrosis factor-alpha (TNF-α) is usually performed in cell cultured medium or body fluids via measurement of its soluble extracellular form. However, depending on cellular condition, TNF-α might be transported through extracellular vesicles (EV) from donor cells to recipient cells. EV are small membrane-delimited structures (∼50 nm to 10 μm) that are spontaneously released from multiple cell types. In cancer, EV arise as important mediators in intercellular communication, and their molecular content may support tumor progression. This chapter describes methods to identify protein content in EV released from the tumor cell cultures. Through this protocol, we show first how to purify EV from in vitro cell culture by using differential centrifugation technique and then we demonstrate how to identify both membrane and soluble TNF-α forms in EV by Western blotting.
... The MDR phenotype may be acquired through cell-to-cell communication via MP, and growing evidence imply the MP role in cancer promotion by mediating the crosstalk between a tumor and its microenvironment [10,31,72,73]. Accordingly, we demonstrated that Pgp is carried by C1MP, albeit it could not be transferred to recipient non-tumor cells, presumably because of C1MP's distinct biological properties ( Figure 5). ...
Article
Full-text available
P-glycoprotein (Pgp/ABCB1) overexpression is associated with multidrug resistance (MDR) phenotype and, consequently, failure in cancer chemotherapy. However, molecules involved in cell death deregulation may also support MDR. Tumor necrosis factor-alpha (TNF-α) is an important cytokine that may trigger either death or tumor growth. Here, we examined the role of cancer cells in self-maintenance and promotion of cellular malignancy through the transport of Pgp and TNF-α molecules by extracellular vesicles (membrane microparticles (MP)). By using a classical MDR model in vitro, we identified a positive correlation between endogenous TNF-α and Pgp, which possibly favored a non-cytotoxic effect of recombinant TNF-α (rTNF-α). We also found a positive feedback involving rTNF-α incubation and TNF-α regulation. On the other hand, rTNF-α induced a reduction in Pgp expression levels and contributed to a reduced Pgp efflux function. Our results also showed that parental and MDR cells spontaneously released MP containing endogenous TNF-α and Pgp. However, these MP were unable to transfer their content to non-cancer recipient cells. Nevertheless, MP released from parental and MDR cells elevated the proliferation index of non-tumor cells. Collectively, our results suggest that Pgp and endogenous TNF-α positively regulate cancer cell malignancy and contribute to changes in normal cell behavior through MP.
... Reduced levels of other circulating miRNAs, including miR-30d, miR-140-3p, miR-532-3p, and miR-190, have been observed in patients with end-stage renal disease 17,18 . Various circulating miRNAs present in plasma function as mediators of cell-to-cell communication and play important roles in renal pathogenesis associated with various disorders, including cardiovascular diseases [19][20][21] , diabetes 22 , and cancer [23][24][25] . Despite these strong associations between circulating miRNAs and kidney diseases, the causal relationship between circulating miRNAs and renal dysfunction remains unclear. ...
Article
Full-text available
Although angiotensin II (AngII) is known to cause renal injury and fibrosis, the underlying mechanisms remain poorly characterized. Here we show that hypertensive nephropathy (HN) patients and AngII-infused mice exhibit elevated levels of circulating miR103a-3p. We observe a positive correlation between miR-103a-3p levels and AngII-induced renal dysfunction. miR-103a-3p suppresses expression of the sucrose non-fermentable-related serine/threonine-protein kinase SNRK in glomerular endothelial cells, and glomeruli of HN patients and AngII-infused mice show reduced endothelial expression of SNRK. We find that SNRK exerts anti-inflammatory effects by interacting with activated nuclear factor-κB (NF-κB)/p65. Overall, we demonstrate that AngII increases circulating miR-103a-3p levels, which reduces SNRK levels in glomerular endothelial cells, resulting in the over-activation of NF-κB/p65 and, consequently, renal inflammation and fibrosis. Together, our work identifies miR-103a-3p/SNRK/NF-κB/p65 as a regulatory axis of AngII-induced renal inflammation and fibrosis.
... Cell-derived microRNAs have relatively recently been understood as gene regulators in human diseases [13,14]. Subsequent studies suggested that some platelet activation factors, e.g., LPS or Ca 2+ , could induce the release of platelet microRNAs by plateletderived tiny membrane vesicles, such as microparticles [15]. ...
Article
Full-text available
Background Patients with anti-β2GPI antibodies display significantly higher platelet activation/aggregation and vascular endothelial cell damage. The mechanism underlying the correlation between platelet activation, vascular endothelial cell dysfunctions and anti-β2GPI antibodies remains unknown. Methods In this study, we derived miR-96 and -26a from platelets activated by the anti-β2GPI/β2GPI complex and explored their role in modulating human umbilical vein endothelial cell (HUVEC) migration and tube formation. Results Anti-β2GPI/β2GPI complex induces the release of platelet-derived microparticles (p-MPs). The amounts of miR-96 and -26a in these p-MPs were also higher than for the control group. Co-incubation of HUVECs with p-MPs resulted in the transfer of miR-96 and -26a into HUVECs, where they inhibited migration and tube formation. The targeting role of these miRNAs was further validated by directly downregulating targeted selectin-P (SELP) and platelet-derived growth factor receptor alpha (PDGFRA) via luciferase activity assay. Conclusion Our study suggests that miR-96 and -26a in p-MPs can inhibit HUVEC behavior by targeting SELP and PDGFRA.
... Thus, both normal and cancer cells express CD146 at their surface. Likewise, microparticles contained in the bloodstream and generated from both endothelial [25] and CD146-positive cancer cells [26] exhibit CD146 at their surface. It appears to be very difficult to specifically target CD146 expressed in cancer cells or their derivatives (circulating cancer cells or microparticles), leading to major problems for diagnosis and/or targeted therapy. ...
Article
Full-text available
CD146 is an adhesion molecule present on many tumors (melanoma, kidney, pancreas, breast, ...). In addition, it has been shown to be expressed on vascular endothelial and smooth muscle cells. Generating an antibody able to specifically recognize CD146 in cancer cells (designated as tumor CD146), but not in normal cells, would thus be of major interest for targeting tumor CD146 without affecting the vascular system. We thus generated antibodies against the extracellular domain of the molecule produced in cancer cells and selected an antibody that specifically recognizes tumor CD146. This antibody (TsCD146 mAb) was able to detect CD146-positive tumors in human biopsies and in vivo, by PET imaging, in a murine xenograft model. In addition, TsCD146 mAb antibody was able to specifically detect CD146-positive cancer microparticles in the plasma of patients. TsCD146 mAb displayed also therapeutic effects since it was able to reduce the growth of human CD146-positive cancer cells xenografted in nude mice. This effect was due to a decrease in the proliferation and an increase in the apoptosis of CD146-positive cancer cells after TsCD146-mediated internalization of the cell surface CD146. Thus, TsCD146 mAb could be of major interest for diagnostic and therapeutic strategies against CD146-positive tumors in a context of personalized medicine.
... In general, they are formed by outward budding and fission of the plasma membrane. Apoptotic signal is also known to promote the release of these particles via the initiation of actinmyosin sliding, resulting in budding and detachment of the membrane from the cytoskeleton [2,21,22]. In addition, hypoxia has been shown to promote MV release via HIFdependent expression of RAB22A [23]. ...
Article
Extracellular vesicles (EVs), a heterogenous group of membrane-bound particles, are virtually secreted by all cells and play important roles in cell–cell communication. Loaded with proteins, mRNAs, non-coding RNAs and membrane lipids from their donor cells, these vesicles participate in normal physiological and pathogenic processes. In addition, these sub-cellular vesicles are implicated in the progression of neurodegenerative disorders. Accumulating evidence suggests that intercellular communication via EVs is responsible for the propagation of key pathogenic proteins involved in the pathogenesis of amyotrophic lateral sclerosis, Parkinson’s diseases, Alzheimer’s diseases and other neurodegenerative disorders. For therapeutic perspective, EVs present advantage over other synthetic drug delivery systems or cell therapy; ability to cross biological barriers including blood brain barrier (BBB), ability to modulate inflammation and immune responses, stability and longer biodistribution with lack of tumorigenicity. In this review, we summarized the current state of EV research in central nervous system in terms of their values in diagnosis, disease pathology and therapeutic applications.
Article
Full-text available
Brain diseases, such as brain tumors, neurodegenerative diseases, cerebrovascular diseases and brain injuries, are caused by various pathophysiological changes such as excessive or impaired angiogenesis, neuroinflammation, immune activation or suppression, neurodegenerative disorders, and neurovirulent protein deposition, which poses a serious health threat. Brain disorders are often difficult to treat due to the presence of the blood‐brain barrier (BBB), which hinders the delivery of drugs to the brain. Biomimetic nanovesicles (BNVs), including endogenous extracellular vesicles (EVs) derived from various cells and artificial nanovesicles (ANVs), possess the ability to penetrate the BBB and thus can be utilized for drug delivery to the brain. BNVs, especially endogenous EVs, are widely distributed in body fluids and usually carry various disease‐related signal molecules such as proteins, RNA and DNA, and may therefore also be analyzed to understand the etiology and pathogenesis of brain diseases. This review will cover the exhaustive classification and characterization of BNVs and pathophysiological roles involved in various brain diseases, and emphatically focus on nanotechnology‐integrated BNVs for brain disease theranostics, including various diagnosis strategies and precise therapeutic regulations (e.g., immunity regulation, disordered protein clearance, anti‐neuroinflammation, neuro‐regeneration, angiogenesis and the gut‐brain axis regulation). We also discuss and outline the remaining challenges and future perspectives regarding the nanotechnology‐integrated BNVs for the diagnosis and treatment of brain diseases. This article is protected by copyright. All rights reserved
Article
Spectrin is a ubiquitous cytoskeletal protein that provides structural stability and supports membrane integrity. In erythrocytes, spectrin proteolysis leads to the biogenesis of plasma membrane extracellular vesicles (EVs). However, its role in non‐erythroid or cancer‐derived plasma membrane EVs biogenesis is unknown. This study aims to examine the role of αII‐spectrin in malignant and non‐malignant plasma membrane vesiculation. We developed a custom, automated cell segmentation plugin for the image processor, Fiji, that provides an unbiased assessment of high resolution confocal microscopy images of the subcellular distribution of αII‐spectrin. We show that, in low vesiculating non‐malignant MBE‐F breast cells, prominent cortical spectrin localises to the cell periphery at rest. In comparison, cortical spectrin is diminished in high vesiculating malignant MCF‐7 breast cells at rest. A cortical distribution of spectrin correlates with increased biomechanical stiffness as measured by Atomic Force Microscopy. Furthermore, cortical spectrin can be induced in malignant MCF‐7 cells by treatment with known vesiculation modulators including the calcium chelator, BAPTA‐AM or the calpain inhibitor II (ALLM). These results demonstrate that the subcellular localisation of spectrin is distinctly different in malignant and non‐malignant cells at rest and shows that the redistribution of cortical αII‐spectrin to the cytoplasm supports plasma membrane‐derived EV biogenesis in malignant cells. This article is protected by copyright. All rights reserved
Article
Full-text available
The introduction of imatinib (IM), a BCR-ABL1 tyrosine kinase inhibitor (TKI), has represented a significant advance in the first-line treatment of chronic myeloid leukemia (CML). However, approximately 30% of patients need to discontinue IM due to resistance or intolerance to this drug. Both resistance and intolerance have also been observed in treatment with the second-generation TKIs—dasatinib, nilotinib, and bosutinib—and the third-generation TKI—ponatinib. The mechanisms of resistance to TKIs may be BCR-ABL1-dependent and/or BCR-ABL1-independent. Although the role of efflux pump P-glycoprotein (Pgp), codified by the ABCB1 gene, is unquestionable in drug resistance of many neoplasms, a longstanding question exists about whether Pgp has a firm implication in TKI resistance in the clinical scenario. The goal of this review is to offer an overview of ABCB1/Pgp expression/activity/polymorphisms in CML. Understanding how interactions, associations, or cooperation between Pgp and other molecules—such as inhibitor apoptosis proteins, microRNAs, or microvesicles—impact IM resistance risk may be critical in evaluating the response to TKIs in CML patients. In addition, new non-TKI compounds may be necessary in order to overcome the resistance mediated by Pgp in CML.
Article
Full-text available
P-glycoprotein is a 170-kd glycosylated transmembrane protein, expressed in a variety of human cells and belonging to the adenosine triphosphate–binding cassette transporter family, whose membrane expression is functionally associated with the multidrug resistance phenotype. However, the mechanisms underlying the regulation of P-glycoprotein functions remain unclear. On the basis of some evidence suggesting P-glycoprotein–actin cytoskeleton interaction, this study investigated the association of P-glycoprotein with ezrin, radixin, and moesin, a class of proteins that cross-link actin filaments with plasma membrane in a human cell line of lymphoid origin and that have been shown to link other ion-pump–related proteins. To this purpose, a multidrug-resistant variant of CCRF-CEM cells (CEM-VBL100) was used as a model to investigate the following: (1) the cellular localizations of P-glycoprotein and ezrin, radixin, and moesin and their molecular associations; and (2) the effects of ezrin, radixin, and moesin antisense oligonucleotides on multidrug resistance and P-glycoprotein function. The results showed that: (1) P-glycoprotein colocalized and coimmunoprecipitated with ezrin, radixin, and moesin; and (2) treatment with antisense oligonucleotides for ezrin, radixin, and moesin restored drug susceptibility consistently with inhibition of both drug efflux and actin–P-glycoprotein association and induction of cellular redistribution of P-glycoprotein. These data suggest that P-glycoprotein association with the actin cytoskeleton through ezrin, radixin, and moesin is key in conferring to human lymphoid cells a multidrug resistance phenotype. Strategies aimed at inhibiting P-glycoprotein–actin association may be helpful in increasing the efficiency of both antitumor and antiviral therapies.
Article
Full-text available
This non-systematic review article aims to summarise the progress made in understanding the functional consequences of microRNA (miRNA) dysregulation in prostate cancer development, and the identification of potential miRNA targets as serum biomarkers for diagnosis or disease stratification. A number of miRNAs have been shown to influence key cellular processes involved in prostate tumourigenesis, including apoptosis-avoidance, cell proliferation and migration and the androgen signalling pathway. An overlapping group of miRNAs have shown differential expression in the serum of patients with prostate cancer of varying stages compared with unaffected individuals. The majority of studies thus far however, involve small numbers of patients and have shown variable and occasionally conflicting results CONCLUSION: MiRNAs show promise as potential circulating biomarkers in prostate cancer, but larger prospective studies are required to validate particular targets and better define their clinical utility.
Article
Full-text available
Multidrug resistance (MDR) is considered a multifactorial event that favors cancer cells become resistant to several chemotherapeutic agents. Numerous mechanisms contribute to MDR, such as P-glycoprotein (Pgp/ABCB1) activity that promotes drug efflux, overexpression of inhibitors of apoptosis proteins (IAPs) that contribute to evasion of apoptosis, and oncogenic pathway activation that favors cancer cells survival. MDR molecules have been identified in membrane microparticles (MPs) and can be transferred to sensitive cancer cells. By co-culturing MPs derived from MDR-positive cells with recipient cells, we showed that sensitive cells accumulated Pgp, IAPs proteins and mRNA. Additionally, MPs promoted microRNA transfer and NFκB and Yb-1 activation. Therefore, our results indicate that MPs can induce a multifactorial phenotype in sensitive cancer cells.This article is protected by copyright. All rights reserved.
Article
Full-text available
P2X7 is an intriguing ionotropic receptor for which the activation by extracellular ATP induces rapid inward cationic currents and intracellular signalling pathways associated with numerous physiological processes such as the induction of the inflammatory cascade, the survival and proliferation of cells. In contrast, long-term stimulation of P2X7 is generally associated with membrane permeabilisation and cell death. Recently, P2X7 has attracted great attention in the cancer field, and particularly in the neoplastic transformation and the progression of solid tumours. A growing number of studies were published; however they often appeared contradictory in their results and conclusions. As such, the involvement of P2X7 in the oncogenic process remains unclear so far. The present review aims to discuss the current knowledge and hypotheses on the involvement of the P2X7 receptor in the development and progression of solid tumours, and highlight the different aspects that require further clarification in order to decipher whether P2X7 could be considered as a cancer biomarker or as a target for pharmacological intervention. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.
Article
Breast cancer is the second most common malignancy diagnosed in women worldwide. The greatest cause of breast cancer mortality is development of metastasis. For many women metastasis is an early event in breast cancer which goes undetected until its presentation, thus there is an urgent need for the development of biomarkers to predict those patients at greatest risk. The expression of a group of small non-coding RNAs, termed microRNAs, has been shown to be altered in tumours. Furthermore, microRNAs identified as being highly expressed in breast cancer tumours can also be detected in the circulation. Circulating microRNAs are an emerging field of biomarker research which have the benefit of being able to be obtained non-invasively and analysed rapidly and relatively cheaply. Here the potential use of circulating miRNAs to detect metastasis in discussed and the current barriers to their progression to the clinic. Copyright © 2015 Elsevier Ltd. All rights reserved.
Article
Despite an initial positive response, breast cancer cells inevitably acquire resistance to doxorubicin (Dox). Alpha-naphthoflavone (ANF) is a well-known chemopreventive agent; however, its anti-cancer properties have not been established. We examined the therapeutic efficacy of ANF in doxorubicin-resistant MCF-7 (MCF-7/Dox) breast cancer cells and investigated its underlying molecular mechanisms of action. MCF-7/Dox cells expressed constitutively active forms of the tyrosine kinases: focal adhesion kinase (FAK-Y397) and protein tyrosine kinase 2 beta (Pyk2- Y579/580) compared with parental MCF-7 cells. ANF significantly enhanced the sensitivity of MCF-7/Dox cells to Dox cytotoxicity in vitro and when co-administered in vivo. This ANF-mediated chemosensitization has dual mechanisms of action: (a) intracellular Dox retention via suppression of P-glycoprotein pump activity, and (b) inhibition of clonogenic cell survival via de-phosphorylation of FAK, Pyk2, and EGF-induced Akt in MCF-7/Dox cells and tumor xenografts. Because of its strong chemosensitization action, broad safety profile, and health benefits, ANF is an attractive anti-cancer drug with therapeutic implications to circumvent drug resistance in breast cancer patients. Copyright © 2015. Published by Elsevier Ireland Ltd.
Article
This report describes technologies to identify and quantify microRNAs (miRNAs) as potential cancer biomarkers, using breast cancer as an example. Most breast cancer patients are not diagnosed until the disease has advanced to later stages, which decreases overall survival rates. Specific miRNAs are up- or downregulated in breast cancer patients at various stages, can be detected in plasma and serum, and have shown promising preliminary clinical sensitivity and specificity for early cancer diagnosis or staging. Nucleic acid testing methods to determine relative concentrations of selected miRNAs include reverse transcription, followed by quantitative PCR (RT-qPCR), microarrays, and next-generation sequencing (NGS). Of these methods, NGS is the most powerful approach for miRNA biomarker discovery, whereas RT-qPCR shows the most promise for eventual clinical diagnostic applications. © 2014 Society for Laboratory Automation and Screening.