Unraveling The Multifaceted Roles of Extracellular Vesicles Insights Into Biology, Pharmacology, and Pharmaceutical Applications For Drug Delivery
Unraveling The Multifaceted Roles of Extracellular Vesicles Insights Into Biology, Pharmacology, and Pharmaceutical Applications For Drug Delivery
Molecular Sciences
Review
Unraveling the Multifaceted Roles of Extracellular Vesicles:
Insights into Biology, Pharmacology, and Pharmaceutical
Applications for Drug Delivery
Ali Al-Jipouri 1 , Àuria Eritja 2 and Milica Bozic 1,2, *
Abstract: Extracellular vesicles (EVs) are nanoparticles released from various cell types that have
emerged as powerful new therapeutic option for a variety of diseases. EVs are involved in the trans-
mission of biological signals between cells and in the regulation of a variety of biological processes,
highlighting them as potential novel targets/platforms for therapeutics intervention and/or delivery.
Therefore, it is necessary to investigate new aspects of EVs’ biogenesis, biodistribution, metabolism,
and excretion as well as safety/compatibility of both unmodified and engineered EVs upon adminis-
tration in different pharmaceutical dosage forms and delivery systems. In this review, we summarize
the current knowledge of essential physiological and pathological roles of EVs in different organs
and organ systems. We provide an overview regarding application of EVs as therapeutic targets,
therapeutics, and drug delivery platforms. We also explore various approaches implemented over
the years to improve the dosage of specific EV products for different administration routes.
cell surface receptor activation, and/or (b) recipient cell plasma membrane fusion and
delivery of effectors (proteins and nucleic acids) [30–33], playing critical roles in stem cell
maintenance [34], tissue repair [35], immunosurveillance [20], and blood coagulation [36].
Body fluid (urine, saliva, synovial, bile, cerebrospinal, bronchoalveolar, nasal, uterine,
amniotic, breast, blood, feces, and seminal)-derived EVs are a mixture of vesicles that
originate from various sources, such as cells in body fluids and/or cells that line extruded
body fluid cavities. Thus, the contents of these EVs act as a source of physiological and
pathological information, which can be transmitted over a long distance. In this section,
we will focus on important physiological roles of EVs in maintaining the homeostasis of
different organs.
barrier disruption, have been found in fecal samples of healthy subjects, while in vitro and
in vivo studies have shown their beneficial effect in intestinal barrier integrity [58].
In addition, placenta-derived EVs have been shown to reduce cytotoxicity of CD4+, CD8+,
and NK cells through Ig-like receptors and the NKG2D NK cell receptors, respectively [82].
During the late stages of pregnancy, syncytiotrophoblast (STB)-EVs are released directly
into the maternal circulation and can be uniquely recognized by a placental alkaline
phosphatase [83]. STB-EVs have been shown to carry several proteins including endoglin,
plasminogen activator inhibitor, soluble fms-like kinase (sFlt), and endothelial nitric oxide
synthase [84,85], as well as miRNAs [86,87], tRNA [88], and DNA [81,89]. The physiological
functions of peripherally circulating STB-EVs in both in vitro and in vivo investigations
are broad and their molecular contents as well as surface markers are powerful and cannot
be underestimated.
elongated EVs [106], to smaller EVs (70–150 nm) resembling exosomes. Their load includes
proteins from the plasma membrane, cytosol, organelles, adhesion receptors, coagulation
and transcription factors, growth factors, active enzymes, cytokines, and chemokines [105].
Also, the main mediators of interaction with circulatory cells and matrices (fibrin) in-
clude GP IIa/IIIa (CD41/CD61), GP Ib (CD42b), P-selectin (CD62P), and CD40L (CD154),
as well as unique exosomal markers (CD9, CD63, CD81, HSP70, TSG101 [103]). The
content of platelet EVs can be confusing, as factors with opposing functions, e.g., pro-
and anti-coagulant substances, can be detected [107]. In addition, they contain small
metabolites [108], and RNAome comprises microRNAs (miRNAs), YRNAs, and circular-
RNAs (ciRNAs) [31] originating from parent megakaryocytes. Although the lifespan of
platelets is about 10 days, exogenously injected EVs are cleared from the circulation within
10–60 min [103]. The most unique physiological role of platelet EVs is their ability to
promote coagulation and thus participate in hemostasis. Human Scott syndrome abol-
ishes platelet phosphatidylserine (PS) exposure, microvesiculation (i.e., EV formation),
and thrombosis, and results in a mild bleeding disorder [22]. Upon platelet activation,
disruption of the membrane phospholipid bilayer resulted in impaired PS externalization
and decreased procoagulant activity, abolishing fibrin formation at sites of vascular dam-
age [109]. The relationship between membrane phospholipid bilayer scrambling and EV
formation in human Scott syndrome was found to be due to a gene defect encoding the
transmembrane protein 16F (TMEM16F), an occult Ca2+ -activated phospholipid scramblase
(CaPLSase) that passively transports phospholipids down their chemical gradients and
mediates blood coagulation [110]. Physiological hypoxic exercise training increased the
level of pro-coagulant EVs and thus increased thrombin production [111]. Larger EVs
(microparticles) from different cellular sources, rather than exosomes [112], show pro-
coagulant activity in relation to tissue factor (TF+ ) EVs [113], which also act in other body
fluids such as saliva and urine [114].
were able directly to stimulate CD8+ and CD4+ T-cells via binding their respective plasma
membrane receptors [127]. Moreover, a comparative study showed that DCs pulsed with
tumor peptides released EVs that could induce a stronger anti-tumor CD4+ T-cell response
than T-cells incubated with the peptides alone, which is evidence for the theory of enhanced
immunity [128].
In terms of homeostatic ability, while some EVs are able to activate the immune system,
other EVs are able to suppress the immune system. For example, natural killer-derived
EVs (NK-EVs) show their cytotoxic activity only on activated, but not resting cells [129].
The suppressive effect of EVs is important during pregnancy, as the pro-inflammatory state
can be harmful. Placental EVs are shed in large quantities during pregnancy [130,131]
and have been associated with TNF-family ligands FasL and TRAIL, leading to apoptosis
in activated lymphocytes [132,133]. In the same context, FasL plus plasma EVs have
been shown to induce apoptosis of CD4+ T-cells [134]. Immunomodulatory EVs play an
important role in the prevention of autoimmunity and chronic inflammation. The term
tolerosomes was coined to demonstrate EV-mediated immune modulation by epithelial
cells [135], which represents the key to the effective development of an allergic response.
The same was demonstrated for EVs isolated from bronchoalveolar lavage fluid (BALF) of
mice immunized against olive pollen allergen [136], and adoptively transferred into naïve
mice. Immunized mice upon exposure to the allergen showed suppression of the immune
response and production of Th2 cytokines. Similarly, responding to ovalbumin-loaded
dendritic cell (DC)-derived EVs from MHC−/− mice stimulated antigen-specific T cells
at the same magnitude as wild type EVs, i.e., MHC-independent immune response [137].
This stimulation of immune tolerance has been exploited therapeutically in diseases such
as post-transplant graft vs. host [138].
kidney damage. Indeed, EVs derived from TGF-β1-treated tubular cells exacerbated kidney
injury and fibrosis [144]. This result was supported by the demonstration that EVs gener-
ated in vivo from animal kidney tissue with ischemia reperfusion injury transferred TGF-β1
into fibroblasts in vitro [149]. Besides communication between vasculature and glomerulus,
as well as tubules and interstitial renal compartments, communication between proximal
and distal tubular cells has also been demonstrated in vitro. For example, proximal tubule
EVs have been shown to reduce sodium channel function in distal cells by transferring
nucleic acid [150,151].
downstream effects on cells and organ systems. For example, synovial fibroblasts from
patients with rheumatoid arthritis (RA) produce EVs containing the inflammatory protein
TNF-α and stimulate NFkB production [168]. In contrast, synovial fluid EVs from men and
women with OA showed an ability to reduce cellular metabolic activity [169]. These studies
include the identification of the miRNA content of EVs that have shown enrichment of
targeting of sex-specific signaling pathways and shed light on the importance of combining
clinical data with pre-clinical research.
Recent work has demonstrated that familial Alzheimer’s-induced pluripotent stem cell
(IPSC)-derived EVs are able to induce tau pathology in naïve mice [193]. In Parkinson’s
patients, EVs have been proposed as a link for α-synuclein aggregation, as they also have
the potential to be used as diagnostic tools for stratifying patients [194,195].
pro-inflammatory cytokines have been shown to increase intestinal permeability [230]. Other
studies have linked EV-mediated inflammation to CVDs [206,231–234], MS [189,235–237], and
more. Autoimmune diseases can stem from EV-presenting self-antigens that auto-activate
lymphocytes and trigger immune cells with antigen specificity for EVs’ own parent cells.
Circulating EVs in systemic lupus erythematosus (SLE) that are enriched for antigenic
DNA may act as an autoantigen to accelerate disease progression [238]. Similarly, the
interaction between EVs and autoreactive T- and B-cells has been shown to trigger pancre-
atic inflammation and lead to the development of diabetes in non-obese diabetic (NOD)
mice [239–241]. In contrast to activation of the immune system, EV-mediated suppression
can be devastating. Currently, there is a large body of evidence showing the participation
of EVs in the development of the metastatic niche by suppressing the circulating immune
response to migrate tumor cells [242,243]. An active inflammatory response to cellular
growth is a key mechanism to reduce tumor burden, which is currently being exploited, as
EVs play an important role in this process. In vitro and in vivo studies showed that tumor-
derived EVs expressing FasL and TRAIL activated regulatory T-cells and myeloid-derived
suppressor cells (MDSCs), which prevented CD8+ T-cell from targeting the tumor [244].
FasL+ EVs were found in the sera of oral squamous cell carcinoma patients, and their level
correlated with tumor burden and nodal involvement [245]. In addition, EVs suppress
the immune system by reducing cytotoxic NK cells circulating in the lung and spleen,
allowing metastatic niches to form in those organs. It has been demonstrated that neu-
trophil mobilization is required for tumor growth; thus, targeting EV release by GTPase
RAB27A/B blockade in mice decreased primary mammary carcinoma tumor growth and
its dissemination into the lung [246].
target cells, and by release of soluble factors (including EVs) as a means of modulating their
targets remotely [299–301]. The anti-apoptotic, pro-angiogenic, anti-inflammatory, prolif-
erative, and trophic nature of MSC-EVs provide potent intrinsic regenerative properties
that have been demonstrated in numerous organs [302–306]. Thus, mouse bone marrow
endothelial progenitor cell-derived EVs improved the hemodynamic status of murine mod-
els of myocardial infarction (MI), showing significant pro-angiogenic effects [307]. Bone
marrow, adipose tissue, and umbilical cord MSC-EVs inhibit cardiomyocyte apoptosis and
promote angiogenesis, thus improving cardiac function and protecting myocardium [308].
The mechanism by which natural umbilical cord MSC-EVs alleviate liver injury after is-
chemia/reperfusion is due to the binding of miR-20a and two upregulated genes, Beclin-1
and FAS 3′ UTRs, thereby inhibiting apoptosis [309]. Injecting MSC-EVs locally into a
murine model of retinal detachment (RD) significantly reduces levels of inflammatory
cytokines TNF-α and IL-1β, Atg5 cleavage, and apoptosis of photoreceptor cells, thereby
preserving the normal structure of the retina [310]. The existence of miRNAs targeting
TLR4/NF-kB within MSC-EVs suppresses inflammation associated with peripheral neu-
ropathy in a murine diabetic model by decreasing the expression of inflammatory cytokines
and regulating the ratio of M1 and M2 macrophages, thereby improving neurovascular
architecture [311].
In contrast to the aforementioned natural pharmacodynamics of MSC-EVs, engineer-
ing EV-producing MSCs leverages them towards producing stronger specialized EVs. The
methods that can be employed range from stimulating EV-producing cells with, e.g., drugs,
cytokines, growth factors, altering cell culture conditions (e.g., hypoxic vs. normoxic, 3D
vs. 2D culture), and genetic engineering with genetic constructs (e.g., plasmids) [312]. The
goals of these methods may be to increase the amount(s) of a specific molecule(s) in the EVs,
alter the sorting of small RNAs, or even add/delete a specific gene in the final structure
of the released EVs. Natural regenerative effects of human adipose tissue derived-MSC
secretome in the lungs have been revealed to include proteins and lipids essential for main-
taining protease/anti-protease homeostasis and anti-microbial activity. In vitro stimulation
of MSCs with dexamethasone and IL-1β along with starvation leads to an increment in
Alpha-1 antitrypsin (AAT), the major elastase-inhibitory enzyme in the lung [313]. Al-
though MSC-EVs carrying miR-20a partially alleviated liver IR-induced injuries in rats,
boosting these EVs with mimics of this miRNA resulted in the complete alleviation of the
injury [314]. Similarly, MSC-EVs transfected with a miR-20b-3p mimic reduced calcium
oxalate accumulation in rat kidneys, with downregulation of oxalate-induced autophagy
and inflammation as responsible therapeutic effects [315].
response, the original tumor antigens, and the overexpression of specific molecules and
receptors that are essential for antigen sampling by APCs [327]. In this sense, the use
of TC-EVs as natural samples of tumor antigens may be a viable option for developing
effective antigen-based immunotherapies. Although the components of TC-EVs are diverse,
one of the many strategies is to employ TC-EVs in ex vivo maturation and induction of
DCs to induce robust CTL responses rather than additional activation stimuli in DCs, as
well as to combine different oncolytic peptides to increase coverage of designed thera-
pies/vaccines [324]. Like MSCs, tumor cells can be primed to produce EVs with enhanced
therapeutic activities that, when presented to the host directly, elicit effects of interest, such
as targeting the delivery of specific molecules. The complex composition of TC-EVs could
be a major drawback of this strategy, which could lead to undesirable effects. This can
be solved by exploiting the surface properties of TC-EVs without undesirable effects to
produce biomimetics in which the drug-carrying core is coated with the outer covering of
the TC-EVs [328,329]. One study showed HepG2 and SKBR3 TC-EVs transfected with a
therapeutic anti-miR-21 to activate synthetic gold iron oxide nanoparticles and harness their
potential in tumor targeting [330]. Thus, the targeting behavior of TC-EVs depends on the
type of cancer cell from which they were isolated. The larger amounts of TC-EVs produced
by tumor cells compared to their non-malignant counterparts [331] and their targeting
behaviors based on their parent cells show that TC-EVs may be unusual candidates for
serving as novel targeting therapies [332].
candidate trigger immune responses via the toll-like receptor (TLR) pathway in Staphylococ-
cus aureus-induced pneumonia in mice [344]. Although microbial EV lipopolysaccharide
(LPS) mediate immunomodulation, further study of the therapeutic opportunities of other
immunomodulatory components and their potential risks is needed.
poor predictability of the process across different cell types [381,383,384]. Similarly, much
remains to be understood about the transmission of EVs in the blood and the crossing of
the endothelial layers of blood vessels. However, recent studies on the passage of EVs
across the blood–brain barrier (BBB) have indicated that transcytosis is the most widely
accepted mechanism for transporting EVs across the endothelium [385]. Breast-cancer
derived EVs are taken up by endothelial cells via clathrin-mediated endocytosis, sorted by
Rab11 for exocytosis at the basolateral membrane, and finally secreted from the cell through
interactions between EVs v-SNARE and VAMP-3 and membrane-associated t-SNAREs
SNAP23 and syntaxin 4 [386]. Although it is not clear whether endothelial cells are involved
in the internalization or attachment of EVs, surface heparan sulfate proteoglycans have
been shown to be involved in cellular endocytosis of EVs [387]. Another mechanism that
facilitates this is EVs’ adsorptive transcytosis through interactions between positive and
negative molecular charges [388].
carries 30–60 eGFP molecules on average [408]. This approach can similarly be exploited to
label other EV sorting domains, such as CD9, CD81, syntenin, and Gag [408,409]. However,
not all EV protein domains, such as ALIX, SIMPLE, and syndecan, can be engineered and
characterized because the efficiency is relatively low [408]. Generally, genetic engineering
approaches provide an effective way to tag a specific group of EVs, either with fluores-
cent proteins, e.g., GFP, RFP, etc., or bioluminescent proteins, e.g., Gaussia-, Firefly-, and
Nano-luciferase. Disadvantages of these approaches include failure to label all EVs and
the requirement for genetic engineering of the produced cells, which is challenging for
some cell sources. Furthermore, overexpression of specific EV-sorting proteins may change
EV biogenesis and/or proteome, thereby changing the biodistribution of EVs. Because
there is no perfect EV reporter or labeling method, and because each method has a certain
degree of advantages and disadvantages, the choice of labeling method should be based on
indication and feasibility.
Figure 2. Challenges of administration of lipid bilayer vesicles (LBVs: LPs, EVs, and LOs) in vivo.
Figure 2. Challenges of administration of lipid bilayer vesicles (LBVs: LPs, EVs, and LOs) in vivo.
The formulation of LBVs ranges from free suspensions to bulk composites and 3D scaffolds within
The formulation
functional of LBVsThe
biomaterials. ranges
routefrom free suspensions
of administration to bulk the
determines composites and 3D scaffolds
most appropriate LBV formu-within
functional biomaterials.
lation to use Theintended
to achieve the route ofeffect.
administration
By adjustingdetermines the most
the composition and appropriate
administration LBV
of for-
LBVs, it is
mulation topossible to facilitate
use to achieve the their delivery
intended across
effect. By normal physiological
adjusting barriers.
the composition andEpithelial and
administration
ofendothelial
LBVs, it iscells liningtothese
possible barriers
facilitate theirinternalize LBVs by
delivery across different
normal mechanisms
physiological (createdEpithelial
barriers. with
https://app.biorender.com/ (accessed on 25 December 2023)).
and endothelial cells lining these barriers internalize LBVs by different mechanisms (created with
https://app.biorender.com/ (accessed on 25 December 2023)).
4.1. Applications of Lipid Bilayer Vesicles for Drug Delivery
4.1. Applications
Compared of toLipid Bilayer
several Vesicles systems,
NP delivery for Drug lipid
Delivery
bilayer vesicles (LBVs) are versatile
platforms for drug
Compared packaging
to several NPand delivery.
delivery Based on
systems, their
lipid origin,vesicles
bilayer they are(LBVs)
classified
areinto
versa-
synthetically originated LPs, biologically originated EVs, as well as hybrid LOs
tile platforms for drug packaging and delivery. Based on their origin, they are classified originat-
ing synthetically
into from the fusion of LPs and
originated EVsbiologically
LPs, [253]. LPs are self-assembled
originated EVs, assynthetic
well as NPs thatLOs
hybrid pro-orig-
vide a prominent platform consisting of fatty acids and lipids centered
inating from the fusion of LPs and EVs [253]. LPs are self-assembled synthetic NPs in a spherical bi-that
layer membrane surrounding an aqueous chamber [422] in which both hydrophilic
provide a prominent platform consisting of fatty acids and lipids centered in a spherical and
hydrophobic
bilayer molecules
membrane can be encapsulated.
surrounding LPs improve
an aqueous chamber [422] the pharmacokinetics
in which of the and
both hydrophilic
incorporated molecules by increasing their circulation time and overcoming
hydrophobic molecules can be encapsulated. LPs improve the pharmacokinetics barriers, suchof the
incorporated molecules by increasing their circulation time and overcoming barriers, such
as the BBB [423]. LPs were classified based on structure into unilamellar, multilamellar,
and multivesicular constructs. The LP preparation method determines the structure and
Int. J. Mol. Sci. 2024, 25, 485 22 of 51
size of the LPs produced, which in turn determines the encapsulation capacity and the
drug release [424]. The membrane fluidity of LPs associated with their composition has
also been the basis for their classification into either non-flexible (non-deformable, clas-
sical, and conventional) or flexible (deformable and elastic) LPs. The charge of the LPs’
phospholipid head groups depends on the characteristics of the surrounding environment,
including pH, temperature, and ionic strength. Thus, LPs’ ζ-potential is a key parameter
affecting the stability of liposomal dispersions and plays a role in the interaction between
LPs and the biological environment [425,426]. Studying these interactions is crucial for
predicting the biological fate of LPs, including corona formation and adsorption onto the
cell membrane [427].
In the context of drug delivery, all advantages of LPs reported in the literature also
apply to EVs. EVs have revealed great potential for integrating many small molecules [428],
proteins [429], nucleic acids [430], and theranostics [431] to be loaded and transported
via EVs for therapeutic and diagnostic applications. Moreover, EVs in a hybrid plat-
form [432,433] incorporate other nanovesicles [434,435] that provide them with superior
biomimetic or drug loading purposes. In contrast, diverse combinations of LPs with chemi-
cal and biological entities improve their physicochemical properties and stability, which in
turn enable controlled drug release and optimize their interactions with the biological envi-
ronment [436]. These combination approaches include modification with polymers [437],
peptides and proteins [438], and nucleic acids [439], as well as coating with [440] or en-
capsulating other [441] nano-entities and hybridizing with the cell membrane [442] or
EVs [443]. Apart from these approaches, intact EVs are more complex due to their biolog-
ical origin, and thus meet the complexity requirements of the optimal biological level of
nanomedicine. This ideality is due to the hundreds of different types of lipids, proteins, and
carbohydrates as well as internal cargoes and surface-bound molecules [253]. In addition,
further design of EVs can be accomplished using EV parent cells engineering [444–446].
Although very simple LP systems can be produced on a large scale, EVs may offer the
possibility to design more complex membrane nanovesicles. EVs outperform LPs through
their remarkable similarity to the cell membrane and are thus more biocompatible and
safer [447]. Employing patient-derived EVs makes it a very promising tool in the context
of personalized medicine [448]. From a pharmacokinetic point of view, EVs, compared
to LPs, have superb circulation time [393,449,450] in crossing biological barriers and ex-
ert physiological, pathological, and therapeutic effects [388,451]. In order to bridge the
bench-to-market gap in the clinical translation of both LBV (LP and EV) drug delivery
products, a range of hurdles must be overcome. These barriers include (1) fully disclosing
the physicochemical properties of the interaction of the lipid bilayer with the biological
environment [452], (2) employing smart strategies to control drug release and concentration
at the site of action [453], (3) advanced production techniques with the highest levels of
particle homogeneity, drug content uniformity, and batch reproducibility, scalability, and
sterility [454], (4) preserving storage stability through innovative formulations [427], and
(5) ensuring clinical trial success by fitting in silico, in vitro, and in vivo models to provide
the highest simulation of the PK-PD of the human body in in vivo studies [253,455]. The
following parts will deal with LP-based dosage forms for various routes of administration
and how they may pave the way for better EV-based dosage forms.
both locally and systemically. The fact that most water and nutrient absorption takes place
in the gut could also be true for orally delivered EVs [465]. The digestion stability of bovine
milk EVs containing miRNAs was evaluated in vitro by subsequent incubation in three
solutions simulating oral, gastric, and intestinal phases of digestion, respectively. It has
been shown that about 50% of all miRNAs survive the oral and gastric phases of digestion.
Moreover, in in vivo oral administration in mice, EVs were detected in various distant
tissues [466]. These results provide indirect evidence of the digestive stability of EVs, al-
lowing them to reach the intestine after oral administration. Furthermore, after absorption
through the intestine [467], therapeutic EVs can exert predictable effects at distant sites. The
present observations suggest the involvement of the “neonatal” Fc receptor in the uptake
of intact EVs [468] and the role of integrins in both tissue trafficking [469] and subsequent
EV uptake by cells [470]. Attempts to evaluate orally administered fluorochrome-labeled
EV bioavailability and tissue biodistribution in mice after oral gavage have demonstrated
the presence of vesicles in the intestine, liver, spleen, kidney, lung, heart, and brain [471].
However, these in vivo attempts have failed to estimate the precise efficacy of EVs’ passage
through the GIT, [472] because it relied on the detection of EV-containing miRNAs rather
than the vesicles themselves [473]. Oral drug administration is the preferred route for clini-
cians and patients. EVs’ superiority to LPs in oral delivery is due to features including fast
internalization, low immunogenicity even at repeated doses, physiological stability, and
feasibility of modification of internal and surface components, which generates specific and
controlled release of internalized or loaded therapeutic molecules. In general, nanocarriers
(including LPs and EVs) are a colloidal delivery system for drugs with a particle size of
less than 500 nm [474]. Different types of nanocarriers cross the intestinal epithelium using
different mechanisms [475]. One such mechanism is paracellular transport, which covers
the diffusion of particles between 0.5 and 20 nm across the intestinal epithelial barrier and
is therefore impractical due to the limited physical dimensions between cells [476,477].
Conversely, disruption of the intestinal barrier, either due to inflammatory diseases or treat-
ments that reduce the tightness of the epithelial barrier, allows the passage of larger EVs
over 200 nm [478]. The other mechanism is transcellular transport, mainly via endocytosis
by epithelial cells and phagocytosis by M cells, where the former accounts for 90–95% and
the latter 1% of the total cells of the GIT [475]. Wu et al., 2022 found insulin-loaded bovine
milk EVs showing efficient internalization through multiple active endocytic pathways into
the epithelium [479]. The authors, as well as Betker et al., 2019, suggested that since milk
is a nutrient, milk EV uptake is mediated by peptides, amino acids, glucose transporters,
and the neonatal Fc receptor (FcRn) [468,479]. In contrast to several studies demonstrating
a rapid clearance rate of circulating exogenous EVs after IV injection (~2–30 min) medi-
ated by the reticuloendothelial system (RES), mainly macrophages [480], Munagala et al.,
2016 found that bovine milk EVs remained circulating for at least 24 h after oral admin-
istration in nude mice [481]. The same group tested milk EVs for oral paclitaxel (PTX)
administration in a lung tumor xenograft model, demonstrating that orally administered
PTX-EVs significantly inhibited tumor growth compared to the same dose administering
PTX intraperitoneally. These PTX-EVs showed significantly less systemic and immuno-
logic toxicities compared to IV PTX [482]. Soo Kim et al., 2016 showed that murine RAW
264.7 macrophage-derived EVs loaded with PTX are more than 50-fold cytotoxic to drug-
resistant MDCKMDR1 (Pgp+ ) cells in vitro [483].
To understand the true clinical potential of oral administration of EVs, the question
that remains to be answered is why EVs absorbed from the GIT have a longer circulating
half-life than observed in systemically injected EVs. Bardonnet et al., 2006 suggested that
NP size is necessary for gastric retention, as particles < 7 mm are evacuated efficiently [484].
Thus, the size range of EVs of 50–200 nm [5] is unlikely to have any biological effect in
the stomach due to poor gastric retention. However, modifying EVs with mucoadhesion
strategies using polymers or phospholipids in their surface membrane could give them
time to induce the desired GIT biological changes [475] as well as drug delivery. In
accordance with bovine milk EVs, the addition of casein has been shown to enhance the
Int. J. Mol. Sci. 2024, 25, 485 24 of 51
uptake of EVs derived from human cardiosphere stromal cells. Modification of EVs with
casein also presented an increased biological effect compared to unmodified EVs in cardiac
dysfunction [485]. Munagala et al., 2016 showed that the addition of folic acid to the surface
of bovine milk EVs loaded with withaferin A resulted in a reduction in tumor size in a
murine model of lung cancer. This response was attributed to folic acid, which either
enhanced the stability of EVs in the GIT or targeted cancer cells after systemic circulation
was reached [481]. These data indirectly support bovine milk EVs as nanocarriers for
oral drug delivery. Similar to PEGylated LPs, Warren et al., 2021 modified the surface
of milk EVs with PEG, thereby decreasing hydrophobic interactions with mucin lining
the intestinal lumen, increasing uptake by epithelial cells, and delivering siRNA loaded
in vitro [486]. Although oral delivery of EVs offers various physiological and practical
advantages compared to other routes, there is still a need for further investigation into their
safety, stability, pharmacokinetics, and biodistribution features before they can be widely
used as drug vehicles or nutritional supplements.
room temperature for three months outperforms the requirement of mRNA-loaded lipid
NP vaccines for cold chain transportation [516]. The use of lung EVs as nanovesicles for
inhaled drug delivery may increase drug retention and efficacy by more efficiently avoiding
immune clearance and targeting pneumocytes. Besides drug delivery, lung EVs themselves
have demonstrated therapeutic benefits. In a rodent model of idiopathic pulmonary fibro-
sis, lung EVs better restore lung function and reduce the severity of fibrosis compared to
their MSC-EV counterpart [517]. Furthermore, inhaled EV therapeutics are superior and
outperform LPs, as they are naturally optimized to distribute and retain mRNA and protein
cargo components to the lung after inhaled delivery [518].
Targeting EVs to a specific organ presents the challenge of rapid clearance after sys-
temic administration, mainly via the liver. A study to minimize liver clearance was per-
formed by Cober et al., 2023, which is based on the ability of porous microgels to engraft
and increase the survival of transplanted cells. They encapsulated EVs and showed that
lung targeting was improved, thanks to EVs’ size-based retention within the pulmonary
microcirculation [519]. The existence of lung EVs in human airway mucus and their less
obstructed movement facilitates crosstalk between lung-resident parenchymal cells and/or
immune cells. This concession was used by Kwak et al., 2023, who demonstrated that
Adeno-associated virus serotype 6 (AAV6) associated with EVs and secreted from vector-
producing HEK-293 cells was a safe and effective platform for inhaled gene delivery. In
contrast, standard preparations of AAV6 alone as well as physical mixtures of individu-
ally prepared EVs and AAV6 failed to mediate EV-AAV6 interaction or to improve gene
transfer efficacy [520]. EVs, as cell-free therapeutics naturally loaded with various bioactive
molecules, offer several advantages for clinical respiratory applications. First, small-sized
EVs facilitate their inhalation and deposit within the small bronchioles and alveoli. Second,
the lipid bilayer structure of EVs grants them stability in tissues and body fluids. Third,
EVs show lower levels of immunogenicity and toxicity compared to cell therapies.
loaded with anticancer drugs can improve their solubility and reduce toxicity, while the
use of ligands grafted onto the surface of engineered EVs can improve their targeting and
efficacy [531]. Similar to chemotherapies’ poor pharmacokinetics, the susceptibility of
current mRNA therapeutics (Pfizer–BioNTech’s mRNA-BNT162b2 and Moderna’s mRNA-
1273 COVID-19 vaccines) to degradation [532] increases the need for an effective delivery
system. Although lipid NPs could efficiently deliver mRNA intracellularly, a portion of the
internalized mRNA continued to function through EV secretion, containing more molecules
with similar biological functions. Thus, EVs can be considered a functional expansion of
lipid NPs [533] and are best used to protect mRNAs as their loading vehicle.
Zhao et al., 2020 [556] incorporated human umbilical vein endothelial cell-derived EVs
(HUVEC-EVs) into well-designed gelatin methacryloyl (GelMA) hydrogels, and completely
dressed skin wounds with them. They demonstrated in vivo and in vitro that GelMA
hydrogel dressings not only helped repair injured tissue, but also achieved prolonged
release of loaded HUVEC-EVs.
5. Conclusions
EVs play crucial roles in various biological processes and diverse cellular activities
by mediating intercellular communications. Conversely, in pathological conditions, they
contribute to the initiation, worsening, and resilience in various diseases. Understanding
EVs’ regulatory mechanisms and function in different biological processes is deemed crucial
for unlocking their clinical potential and applications. This, indeed, involves developing
new therapeutic strategies or interventions based on regulatory mechanisms of EVs.
EVs derived from MSCs, specific tumor cells, dendritic cells, B lymphocytes, and
macrophages hold significant therapeutic potential, without the necessity for specific
manipulation, and they elicit anti-inflammatory, anti-apoptotic, pro-angiogenic, and prolif-
erative effects. Additionally, engineering EV-producing cells will enhance their therapeutic
potential, and this can be achieved through genetic engineering, changing cell culture con-
ditions, and stimulating cells with factors such as drugs and cytokines. Specific examples,
such as the regenerative effects of MSCs in the lungs and breast milk-derived EVs, with
anti-inflammatory and immunomodulatory properties, highlight the diverse applications
of EVs in treating various disorders. The modulation of EV content, including proteins,
small RNAs, and lipids, emerges as a key strategy for tailoring therapeutic effects, showing
promise for future developments in regenerative medicine and disease treatment.
Besides their use as therapeutics, EVs hold a great potential as therapeutic nano-
biocarriers for drug delivery. Namely, EVs can encapsulate drugs or be decorated with
specific ligands for targeted delivery. The pharmacokinetics of intrinsic and extrinsic
EVs make them good candidates for drug delivery platforms owing to their in vivo toler-
ance and ability to cross biological barriers. Thus, different EV labeling and engineering
strategies were developed to understand the in vivo pharmacokinetics of exogenously
administered EVs. Labeling EVs can be performed with fluorescent dyes, radiotracers, or
employing genetically engineered producer cells that express a reporter protein fused to
the EV sorting. However, evident flaws of labeling using covalent conjugations include
the alteration of the surface proteome of EVs, which affects their interactions with other
proteins. Furthermore, these dyes can label non-vesicular proteins, which makes them im-
precise. Additionally, EV engineering strategies, including the use of surface proteins like
CD47, albumin, and polyethylene glycols (PEGs) to enhance circulation half-life, prevent
clearance, and improve extrahepatic delivery, could also enhance the potential of EVs for
drug delivery and targeting. Genetic engineering approaches also have drawbacks, such
as the inability to label all EVs, as well as problems related with genetic engineering of
producing cells. Additionally, the overexpression of specific EV-sorting proteins has the
potential to alter EV biogenesis and/or proteome, impacting the biodistribution of EVs.
Pharmaceutical application of EVs in drug delivery is currently in the pre-clinical stage.
EVs present challenges in designing suitable dosage forms for specific applications. De-
spite challenges, EVs demonstrate great potential in integrating small molecules, proteins,
nucleic acids, and theranostics for therapeutic and diagnostic purposes. Being biologi-
cally derived, EVs offer complexity requirements ideal for nanomedicine. Importantly,
patient-derived EVs hold promise for personalized medicine due to their biocompatibility
and safety. From a pharmacokinetic point of view, EVs exhibit superior circulation time
compared to LPs, enabling them to cross biological barriers effectively. Overcoming ob-
stacles in clinical translation involves disclosing physicochemical properties, controlling
drug release, employing advanced production techniques, ensuring storage stability, and
fitting models for successful clinical trials. The exploration of LP-based dosage forms for
various administration routes may pave the way for improved EV-based dosage forms in
the future.
Author Contributions: Conceptualization, A.A.-J. and M.B.; literature review, A.A.-J., À.E. and M.B.;
writing—original draft preparation, A.A.-J., À.E. and M.B.; writing—review and editing, A.A.-J., À.E.
and M.B.; scientific illustrations, figures and tables, A.A.-J.; supervision and project administration,
M.B. All authors have read and agreed to the published version of the manuscript.
Int. J. Mol. Sci. 2024, 25, 485 30 of 51
Funding: M.B. was supported by the Miguel Servet contract from the Instituto de Salud Carlos III
(ISCIII) (CP19/00027), ESF “Investing in your future”, and MV22/00017 grant.
Data Availability Statement: Not applicable.
Conflicts of Interest: The authors declare no conflicts of interest.
References
1. Buzas, E.I.; György, B.; Nagy, G.; Falus, A.; Gay, S. Emerging role of extracellular vesicles in inflammatory diseases. Nat. Rev.
Rheumatol. 2014, 10, 356–364. [CrossRef] [PubMed]
2. Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.;
Atkin-Smith, G.K. Minimal information for studies of extracellular vesicles 2018(MISEV2018): A position statement of the
International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2018, 7, 1535750.
[CrossRef] [PubMed]
3. De Sousa, K.P.; Rossi, I.; Abdullahi, M.; Ramirez, M.I.; Stratton, D.; Inal, J.M. Isolation and characterization of extracellular vesicles
and future directions in diagnosis and therapy. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnology 2023, 15, e1835. [CrossRef]
[PubMed]
4. Van Niel, G.; d’Angelo, G.; Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2018, 19,
213–228. [CrossRef] [PubMed]
5. Kalluri, R.; LeBleu, V.S. The biology, function, and biomedical applications of exosomes. Science 2020, 367, eaau6977. [CrossRef]
6. Couch, Y.; Buzàs, E.I.; Di Vizio, D.; Gho, Y.S.; Harrison, P.; Hill, A.F.; Lötvall, J.; Raposo, G.; Stahl, P.D.; Théry, C. A brief history of
nearly EV-erything–The rise and rise of extracellular vesicles. J. Extracell. Vesicles 2021, 10, e12144. [CrossRef]
7. van Niel, G.; Carter, D.R.; Clayton, A.; Lambert, D.W.; Raposo, G.; Vader, P. Challenges and directions in studying cell–cell
communication by extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2022, 23, 369–382. [CrossRef]
8. Stahl, P.D.; Raposo, G. Extracellular vesicles: Exosomes and microvesicles, integrators of homeostasis. Physiology 2019, 34, 169–177.
[CrossRef]
9. Kosanović, M.; Milutinovic, B.; Glamočlija, S.; Morlans, I.M.; Ortiz, A.; Bozic, M. Extracellular Vesicles and Acute Kidney Injury:
Potential Therapeutic Avenue for Renal Repair and Regeneration. Int. J. Mol. Sci. 2022, 23, 3792. [CrossRef]
10. Kosanović, M.; Milutinović, B.; Kutzner, T.J.; Mouloud, Y.; Bozic, M. Clinical Prospect of Mesenchymal Stromal/Stem Cell-Derived
Extracellular Vesicles in Kidney Disease: Challenges and the Way Forward. Pharmaceutics 2023, 15, 1911. [CrossRef]
11. Jin, Y.; Ma, L.; Zhang, W.; Yang, W.; Feng, Q.; Wang, H. Extracellular signals regulate the biogenesis of extracellular vesicles. Biol.
Res. 2022, 55, 1–16. [CrossRef] [PubMed]
12. Dixson, A.C.; Dawson, T.R.; Di Vizio, D.; Weaver, A.M. Context-specific regulation of extracellular vesicle biogenesis and cargo
selection. Nat. Rev. Mol. Cell Biol. 2023, 1–23. [CrossRef] [PubMed]
13. Muraoka, S.; Hirano, M.; Isoyama, J.; Nagayama, S.; Tomonaga, T.; Adachi, J. Comprehensive proteomic profiling of plasma and
serum phosphatidylserine-positive extracellular vesicles reveals tissue-specific proteins. Iscience 2022, 25, 104012. [CrossRef]
[PubMed]
14. Kehrloesser, S.; Cast, O.; Elliott, T.S.; Ernst, R.J.; Machel, A.C.; Chen, J.-X.; Chin, J.W.; Miller, M.L. Cell-of-origin–specific
proteomics of extracellular vesicles. PNAS Nexus 2023, 2, pgad107. [CrossRef] [PubMed]
15. Donoso-Quezada, J.; Ayala-Mar, S.; González-Valdez, J. The role of lipids in exosome biology and intercellular communication:
Function, analytics and applications. Traffic 2021, 22, 204–220. [CrossRef] [PubMed]
16. Buratta, S.; Urbanelli, L.; Tognoloni, A.; Latella, R.; Cerrotti, G.; Emiliani, C.; Chiaradia, E. Protein and Lipid Content of Milk
Extracellular Vesicles: A Comparative Overview. Life 2023, 13, 401. [CrossRef]
17. Clarke-Bland, C.E.; Bill, R.M.; Devitt, A. Emerging roles for AQP in mammalian extracellular vesicles. Biochim. Et Biophys.
Acta(BBA)-Biomembr. 2022, 1864, 183826. [CrossRef]
18. Roussin, L. Effect of Gut Microbiota from Children with Autism Spectrum Disorder(ASD) on Behavior and ASD-Related Biological
Markers in Germ-Free Mice. Ph.D. Thesis, Université Paris-Saclay, Paris, France, 2023.
19. Yedigaryan, L.; Sampaolesi, M. Extracellular vesicles and Duchenne muscular dystrophy pathology: Modulators of disease
progression. Front. Physiol. 2023, 14, 204. [CrossRef]
20. Buzas, E.I. The roles of extracellular vesicles in the immune system. Nat. Rev. Immunol. 2023, 23, 236–250. [CrossRef]
21. Coulter, M.E.; Dorobantu, C.M.; Lodewijk, G.A.; Delalande, F.; Cianferani, S.; Ganesh, V.S.; Smith, R.S.; Lim, E.T.; Xu, C.S.; Pang, S.
The ESCRT-III protein CHMP1A mediates secretion of sonic hedgehog on a distinctive subtype of extracellular vesicles. Cell Rep.
2018, 24, 973–986. [CrossRef]
22. Agbani, E.O.; Hers, I.; Poole, A.W. Platelet procoagulant membrane dynamics: A key distinction between thrombosis and
hemostasis? Blood Adv. 2023, 7, 1615–1619. [CrossRef] [PubMed]
23. Kim, H.J.; Kim, G.; Lee, J.; Lee, Y.; Kim, J.-H. Secretome of stem cells: Roles of extracellular vesicles in diseases, stemness,
differentiation, and reprogramming. Tissue Eng. Regen. Med. 2022, 19, 19–33. [CrossRef] [PubMed]
24. Wei, W.; Riley, N.M.; Yang, A.C.; Kim, J.T.; Terrell, S.M.; Li, V.L.; Garcia-Contreras, M.; Bertozzi, C.R.; Long, J.Z. Cell type-selective
secretome profiling in vivo. Nat. Chem. Biol. 2021, 17, 326–334. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2024, 25, 485 31 of 51
25. He, N.; Zhang, Y.; Zhang, S.; Wang, D.; Ye, H. Exosomes: Cell-free therapy for cardiovascular diseases. J. Cardiovasc. Transl. Res.
2020, 13, 713–721. [CrossRef] [PubMed]
26. Quaglia, M.; Merlotti, G.; Colombatto, A.; Bruno, S.; Stasi, A.; Franzin, R.; Castellano, G.; Grossini, E.; Fanelli, V.; Cantaluppi, V.
Stem Cell-Derived Extracellular Vesicles as Potential Therapeutic Approach for Acute Kidney Injury. Dysfunct. Immune Syst. Dur.
Acute Kidney Inj. 2022, 13, 849891. [CrossRef] [PubMed]
27. Liu, Y.; Zheng, Y.; Yang, Y.; Liu, K.; Wu, J.; Gao, P.; Zhang, C. Exosomes in liver fibrosis: The role of modulating hepatic stellate
cells and immune cells, and prospects for clinical applications. Front. Immunol. 2023, 14, 1133297. [CrossRef]
28. Hering, C.; Shetty, A.K. Extracellular Vesicles Derived From Neural Stem Cells, Astrocytes, and Microglia as Therapeutics for
Easing TBI-Induced Brain Dysfunction. Stem Cells Transl. Med. 2023, 12, 140–153. [CrossRef]
29. Bian, D.; Wu, Y.; Song, G.; Azizi, R.; Zamani, A. The application of mesenchymal stromal cells(MSCs) and their derivative
exosome in skin wound healing: A comprehensive review. Stem Cell Res. Ther. 2022, 13, 24. [CrossRef]
30. Ghanam, J.; Chetty, V.K.; Barthel, L.; Reinhardt, D.; Hoyer, P.-F.; Thakur, B.K. DNA in extracellular vesicles: From evolution to its
current application in health and disease. Cell Biosci. 2022, 12, 37. [CrossRef]
31. Dellar, E.R.; Hill, C.; Melling, G.E.; Carter, D.R.; Baena-Lopez, L.A. Unpacking extracellular vesicles: RNA cargo loading and
function. J. Extracell. Biol. 2022, 1, e40. [CrossRef]
32. Vaka, R.; Parent, S.; Risha, Y.; Khan, S.; Courtman, D.; Stewart, D.J.; Davis, D.R. Extracellular vesicle microRNA and protein cargo
profiling in three clinical-grade stem cell products reveals key functional pathways. Mol. Ther. Nucleic Acids 2023, 32, 80–93.
[CrossRef] [PubMed]
33. Man, K.; Brunet, M.Y.; Fernandez-Rhodes, M.; Williams, S.; Heaney, L.M.; Gethings, L.A.; Federici, A.; Davies, O.G.; Hoey, D.;
Cox, S.C. Epigenetic reprogramming enhances the therapeutic efficacy of osteoblast-derived extracellular vesicles to promote
human bone marrow stem cell osteogenic differentiation. J. Extracell. Vesicles 2021, 10, e12118. [CrossRef] [PubMed]
34. Hur, Y.H.; Feng, S.; Wilson, K.F.; Cerione, R.A.; Antonyak, M.A. Embryonic stem cell-derived extracellular vesicles maintain ESC
stemness by activating FAK. Dev. Cell 2021, 56, 277–291.e6. [CrossRef] [PubMed]
35. Ekram, S.; Khalid, S.; Ramzan, F.; Salim, A.; Bashir, I.; Durrieu, M.C.; Khan, I. Mesenchymal Stem Cell–Derived Extracellular
Vesicles Protect Rat Nucleus Pulposus Cells from Oxidative Stress. Cartilage 2023. [CrossRef] [PubMed]
36. Alasztics, B.; Kovács, Á.F.; Molvarec, A.; Koller, Á.; Szabó, G.; Fekete, N.; Buzás, E.I.; Pállinger, É.; Rigó Jr, J. Platelet-derived
extracellular vesicles may contribute to the hypercoagulable state in preeclampsia. J. Reprod. Immunol. 2021, 148, 103380.
[CrossRef] [PubMed]
37. Delrue, C.; De Bruyne, S.; Speeckaert, R.; Speeckaert, M.M. Urinary Extracellular Vesicles in Chronic Kidney Disease: From Bench
to Bedside? Diagnostics 2023, 13, 443. [CrossRef] [PubMed]
38. Wang, Y.; Zhang, M. Urinary Exosomes: A Promising Biomarker for Disease Diagnosis. Lab. Med. 2023, 54, 115–125. [CrossRef]
39. Jain, G.; Das, P.; Ranjan, P.; Valderrama, F.; Cieza-Borrella, C. Urinary extracellular vesicles miRNA—A new era of prostate cancer
biomarkers. Front. Genet. 2023, 14, 61. [CrossRef]
40. Spanu, S.; van Roeyen, C.; Denecke, B.; Floege, J.; Mühlfeld, A. Urinary Exosomes: A Novel Means to Non-Invasively Assess
Changes in Renal Gene. PLoS ONE 2014, 9, e109631. [CrossRef]
41. Erdbrügger, U.; Blijdorp, C.J.; Bijnsdorp, I.V.; Borràs, F.E.; Burger, D.; Bussolati, B.; Byrd, J.B.; Clayton, A.; Dear, J.W.; Falcón-Pérez,
J.M. Urinary extracellular vesicles: A position paper by the Urine Task Force of the International Society for Extracellular Vesicles.
J. Extracell. Vesicles 2021, 10, e12093. [CrossRef]
42. Oshikawa-Hori, S.; Yokota-Ikeda, N.; Sonoda, H.; Sasaki, Y.; Ikeda, M. Reduced urinary release of AQP1-and AQP2-bearing
extracellular vesicles in patients with advanced chronic kidney disease. Physiol. Rep. 2021, 9, e15005. [CrossRef] [PubMed]
43. Liu, Z.Z.; Jose, P.A.; Yang, J.; Zeng, C. Importance of extracellular vesicles in hypertension. Exp. Biol. Med. 2021, 246, 342–353.
[CrossRef] [PubMed]
44. Lee, S.A.; Choi, C.; Yoo, T.-H. Extracellular vesicles in kidneys and their clinical potential in renal diseases. Kidney Res. Clin. Pract.
2021, 40, 194. [CrossRef] [PubMed]
45. Kwon, S.H. Extracellular vesicles in renal physiology and clinical applications for renal disease. Korean J. Intern. Med. 2019,
34, 470. [CrossRef] [PubMed]
46. Ilea, A.; Andrei, V.; Feurdean, C.N.; Băbt, an, A.-M.; Petrescu, N.B.; Câmpian, R.S.; Bos, ca, A.B.; Ciui, B.; Tertis, , M.; Săndulescu, R.
Saliva, a magic biofluid available for multilevel assessment and a mirror of general health—A systematic review. Biosensors 2019,
9, 27. [CrossRef] [PubMed]
47. Palanisamy, V.; Sharma, S.; Deshpande, A.; Zhou, H.; Gimzewski, J.; Wong, D.T. Nanostructural and transcriptomic analyses of
human saliva derived exosomes. PLoS ONE 2010, 5, e8577. [CrossRef] [PubMed]
48. Lässer, C.; Seyed Alikhani, V.; Ekström, K.; Eldh, M.; Torregrosa Paredes, P.; Bossios, A.; Sjöstrand, M.; Gabrielsson, S.; Lötvall, J.;
Valadi, H. Human saliva, plasma and breast milk exosomes contain RNA: Uptake by macrophages. J. Transl. Med. 2011, 9, 1–8.
[CrossRef]
49. Berckmans, R.J.; Sturk, A.; van Tienen, L.M.; Schaap, M.C.; Nieuwland, R. Cell-derived vesicles exposing coagulant tissue factor
in saliva. Blood J. Am. Soc. Hematol. 2011, 117, 3172–3180. [CrossRef]
50. Yu, Y.; Gool, E.; Berckmans, R.; Coumans, F.; Barendrecht, A.; Maas, C.; van der Wel, N.; Altevogt, P.; Sturk, A.; Nieuwland,
R. Extracellular vesicles from human saliva promote hemostasis by delivering coagulant tissue factor to activated platelets. J.
Thromb. Haemost. 2018, 16, 1153–1163. [CrossRef]
Int. J. Mol. Sci. 2024, 25, 485 32 of 51
51. Ogawa, Y.; Kanai-Azuma, M.; Akimoto, Y.; Kawakami, H.; Yanoshita, R. Exosome-like vesicles with dipeptidyl peptidase IV in
human saliva. Biol. Pharm. Bull. 2008, 31, 1059–1062. [CrossRef]
52. Bajinka, O.; Tan, Y.; Darboe, A.; Ighaede-Edwards, I.G.; Abdelhalim, K.A. The gut microbiota pathway mechanisms of diabetes.
AMB Express 2023, 13, 16. [CrossRef] [PubMed]
53. Villa, T.; Sánchez-Pérez, A. The Gut Microbiome Affects Human Mood and Behavior. In Developmental Biology in Prokaryotes and
Lower Eukaryotes; Springer: Cham, Switzerland, 2021; pp. 541–565.
54. Wang, Y.; Liu, T.; Wan, Z.; Wang, L.; Hou, J.; Shi, M.; Tsui, S.K.W. Investigating causal relationships between the gut microbiota
and allergic diseases: A mendelian randomization study. Front. Genet. 2023, 14, 541–565. [CrossRef] [PubMed]
55. Jin, Q.; Ren, F.; Dai, D.; Sun, N.; Qian, Y.; Song, P. The causality between intestinal flora and allergic diseases: Insights from a
bi-directional two-sample Mendelian randomization analysis. Front. Immunol. 2023, 14, 1121273. [CrossRef] [PubMed]
56. Ñahui Palomino, R.A.; Vanpouille, C.; Costantini, P.E.; Margolis, L. Microbiota–host communications: Bacterial extracellular
vesicles as a common language. PLoS Pathog. 2021, 17, e1009508. [CrossRef] [PubMed]
57. Wang, X.; Lin, S.; Wang, L.; Cao, Z.; Zhang, M.; Zhang, Y.; Liu, R.; Liu, J. Versatility of bacterial outer membrane vesicles in
regulating intestinal homeostasis. Sci. Adv. 2023, 9, eade5079. [CrossRef] [PubMed]
58. Chelakkot, C.; Choi, Y.; Kim, D.-K.; Park, H.T.; Ghim, J.; Kwon, Y.; Jeon, J.; Kim, M.-S.; Jee, Y.-K.; Gho, Y.S. Akkermansia
muciniphila-derived extracellular vesicles influence gut permeability through the regulation of tight junctions. Exp. Mol. Med.
2018, 50, e450. [CrossRef]
59. Yedigaryan, L.; Sampaolesi, M. Skeletal Muscle–Extricated Extracellular Vesicles: Facilitators of Repair and Regeneration:
Facilitators of Repair and Regeneration. In Handbook of Stem Cell Therapy; Springer: Cham, Switzerland, 2022; pp. 1–25.
60. Ji, S.; Ma, P.; Cao, X.; Wang, J.; Yu, X.; Luo, X.; Lu, J.; Hou, W.; Zhang, Z.; Yan, Y. Myoblast-derived exosomes promote the repair
and regeneration of injured skeletal muscle in mice. FEBS Open Bio 2022, 12, 2213–2226. [CrossRef]
61. Florin, A.; Lambert, C.; Sanchez, C.; Zappia, J.; Durieux, N.; Tieppo, A.M.; Mobasheri, A.; Henrotin, Y. The secretome of skeletal
muscle cells: A systematic review. Osteoarthr. Cartil. Open 2020, 2, 100019. [CrossRef]
62. Lam, N.T.; Gartz, M.; Thomas, L.; Haberman, M.; Strande, J.L. Influence of microRNAs and exosomes in muscle health and
diseases. J. Muscle Res. Cell Motil. 2020, 41, 269–284. [CrossRef]
63. Coenen-Stass, A.M.; Betts, C.A.; Lee, Y.F.; Mäger, I.; Turunen, M.P.; El Andaloussi, S.; Morgan, J.E.; Wood, M.J.; Roberts, T.C.
Selective release of muscle-specific, extracellular microRNAs during myogenic differentiation. Hum. Mol. Genet. 2016, 25,
3960–3974. [CrossRef]
64. Mytidou, C.; Koutsoulidou, A.; Zachariou, M.; Prokopi, M.; Kapnisis, K.; Spyrou, G.M.; Anayiotos, A.; Phylactou, L.A. Age-related
exosomal and endogenous expression patterns of miR-1, miR-133a, miR-133b, and miR-206 in skeletal muscles. Front. Physiol.
2021, 12, 708278. [CrossRef] [PubMed]
65. Chidester, S.; Livinski, A.A.; Fish, A.F.; Joseph, P.V. The role of extracellular vesicles in β-cell function and viability: A scoping
review. Front. Endocrinol. 2020, 11, 375. [CrossRef] [PubMed]
66. Wang, W.; Li, M.; Chen, Z.; Xu, L.; Chang, M.; Wang, K.; Deng, C.; Gu, Y.; Zhou, S.; Shen, Y. Biogenesis and function of extracellular
vesicles in pathophysiological processes skeletal muscle atrophy. Biochem. Pharmacol. 2022, 114954. [CrossRef] [PubMed]
67. Yahara, Y.; Nguyen, T.; Ishikawa, K.; Kamei, K.; Alman, B.A. The origins and roles of osteoclasts in bone development, homeostasis
and repair. Development 2022, 149, dev199908. [CrossRef] [PubMed]
68. Lu, Y.; Huang, W.; Li, M.; Zheng, A. Exosome-Based Carrier for RNA Delivery: Progress and Challenges. Pharmaceutics 2023,
15, 598. [CrossRef] [PubMed]
69. Uenaka, M.; Yamashita, E.; Kikuta, J.; Morimoto, A.; Ao, T.; Mizuno, H.; Furuya, M.; Hasegawa, T.; Tsukazaki, H.; Sudo, T.
Osteoblast-derived vesicles induce a switch from bone-formation to bone-resorption in vivo. Nat. Commun. 2022, 13, 1066.
[CrossRef]
70. Yuan, F.-L.; Wu, Q.-y.; Miao, Z.-N.; Xu, M.-H.; Xu, R.-S.; Jiang, D.-L.; Ye, J.-X.; Chen, F.-h.; Zhao, M.-D.; Wang, H.-j. Osteoclast-
derived extracellular vesicles: Novel regulators of osteoclastogenesis and osteoclast–osteoblasts communication in bone remodel-
ing. Front. Physiol. 2018, 9, 628. [CrossRef]
71. Xu, L.; Qian, Z.; Wang, S.; Wang, R.; Pu, X.; Yang, B.; Zhou, Q.; Du, C.; Chen, Q.; Feng, Z. Galectin-3 Enhances Osteogenic
Differentiation of Precursor Cells From Patients With Diffuse Idiopathic Skeletal Hyperostosis via Wnt/β-Catenin Signaling. J.
Bone Miner. Res. 2022, 37, 724–739. [CrossRef]
72. Anderson, H.C. Vesicles associated with calcification in the matrix of epiphyseal cartilage. J. Cell Biol. 1969, 41, 59–72. [CrossRef]
73. Ali, S.Y.; Sajdera, S.; Anderson, H. Isolation and characterization of calcifying matrix vesicles from epiphyseal cartilage. Proc. Natl.
Acad. Sci. 1970, 67, 1513–1520. [CrossRef]
74. Simon, C.; Greening, D.W.; Bolumar, D.; Balaguer, N.; Salamonsen, L.A.; Vilella, F. Extracellular vesicles in human reproduction
in health and disease. Endocr. Rev. 2018, 39, 292–332. [CrossRef] [PubMed]
75. Park, K.-H.; Kim, B.-J.; Kang, J.; Nam, T.-S.; Lim, J.M.; Kim, H.T.; Park, J.K.; Kim, Y.G.; Chae, S.-W.; Kim, U.-H. Ca2+ signaling
tools acquired from prostasomes are required for progesterone-induced sperm motility. Sci. Signal. 2011, 4, ra31. [CrossRef]
[PubMed]
76. Aalberts, M.; Stout, T.; Stoorvogel, W. Prostasomes: Extracellular vesicles from the prostate. Reproduction 2014, 147, R1–R14.
[CrossRef] [PubMed]
Int. J. Mol. Sci. 2024, 25, 485 33 of 51
77. Carlsson, L.; Påhlson, C.; Bergquist, M.; Ronquist, G.; Stridsberg, M. Antibacterial activity of human prostasomes. Prostate 2000,
44, 279–286. [CrossRef] [PubMed]
78. Al-Dossary, A.A.; Strehler, E.E.; Martin-DeLeon, P.A. Expression and secretion of plasma membrane Ca2+-ATPase 4a(PMCA4a)
during murine estrus: Association with oviductal exosomes and uptake in sperm. PLoS ONE 2013, 8, e80181. [CrossRef] [PubMed]
79. Sun, R.; Liang, H.; Guo, H.; Wang, Z.; Deng, Q. PMCA4 gene expression is regulated by the androgen receptor in the mouse testis
during spermatogenesis. Mol. Med. Rep. 2021, 23, 1. [CrossRef] [PubMed]
80. Ravaux, B.; Favier, S.; Perez, E.; Gourier, C. Egg CD9 protein tides correlated with sperm oscillations tune the gamete fusion
ability in mammal. J. Mol. Cell Biol. 2018, 10, 494–502. [CrossRef]
81. Tannetta, D.; Masliukaite, I.; Vatish, M.; Redman, C.; Sargent, I. Update of syncytiotrophoblast derived extracellular vesicles in
normal pregnancy and preeclampsia. J. Reprod. Immunol. 2017, 119, 98–106. [CrossRef]
82. Wensveen, F.M.; Jelenčić, V.; Polić, B. NKG2D: A master regulator of immune cell responsiveness. Front. Immunol. 2018, 9, 441.
[CrossRef]
83. Rana, S.; Lemoine, E.; Granger, J.P.; Karumanchi, S.A. Preeclampsia: Pathophysiology, challenges, and perspectives. Circ. Res.
2019, 124, 1094–1112. [CrossRef]
84. Awoyemi, T.; Cerdeira, A.S.; Zhang, W.; Jiang, S.; Rahbar, M.; Logenthiran, P.; Redman, C.; Vatish, M. Preeclampsia and
syncytiotrophoblast membrane extracellular vesicles(STB-EVs). Clin. Sci. 2022, 136, 1793–1807. [CrossRef] [PubMed]
85. Guerby, P.; Swiader, A.; Augé, N.; Parant, O.; Vayssière, C.; Uchida, K.; Salvayre, R.; Negre-Salvayre, A. High glutathionylation of
placental endothelial nitric oxide synthase in preeclampsia. Redox Biol. 2019, 22, 101126. [CrossRef] [PubMed]
86. Maraghechi, P.; Aponte, M.T.S.; Ecker, A.; Lázár, B.; Tóth, R.; Szabadi, N.T.; Gócza, E. Pluripotency-Associated microRNAs in
Early Vertebrate Embryos and Stem Cells. Genes 2023, 14, 1434. [CrossRef] [PubMed]
87. Xu, P.; Ma, Y.; Wu, H.; Wang, Y.-L. Placenta-derived microRNAs in the pathophysiology of human pregnancy. Front. Cell Dev. Biol.
2021, 9, 646326. [CrossRef] [PubMed]
88. Cooke, W.R.; Cribbs, A.; Zhang, W.; Kandzija, N.; Motta-Mejia, C.; Dombi, E.; Ri, R.; Cerdeira, A.S.; Redman, C.; Vatish, M.
Maternal circulating syncytiotrophoblast-derived extracellular vesicles contain biologically active 5’-tRNA halves. Biochem.
Biophys. Res. Commun. 2019, 518, 107–113. [CrossRef] [PubMed]
89. Cooke, W.R.; Jones, G.D.; Redman, C.W.; Vatish, M. Syncytiotrophoblast derived extracellular vesicles in relation to preeclampsia.
Matern. -Fetal Med. 2021, 3, 151–160. [CrossRef]
90. Yakovlev, A.A. Neuronal Exosomes as a New Signaling System. Biochemistry 2023, 88, 457–465. [CrossRef]
91. Paolicelli, R.C.; Ferretti, M.T. Function and dysfunction of microglia during brain development: Consequences for synapses and
neural circuits. Front. Synaptic Neurosci. 2017, 9, 9. [CrossRef]
92. Tanaka, Y.; Morozumi, A.; Hirokawa, N. Nodal flow transfers polycystin to determine mouse left-right asymmetry. Dev. Cell 2023,
58, 1447–1461.e6. [CrossRef]
93. Chen, X.; He, X.; Xu, F.; Xu, N.; Sharifi, N.H.; Zhang, P.; Flores, J.J.; Wu, L.; He, Q.; Kanamaru, H. Fractalkine Enhances Hematoma
Resolution and Improves Neurological Function via CX3CR1/AMPK/PPARγ Pathway After GMH. Stroke 2023, 54, 2420–2433.
[CrossRef]
94. Brown, M.; Johnson, L.A.; Leone, D.A.; Majek, P.; Vaahtomeri, K.; Senfter, D.; Bukosza, N.; Schachner, H.; Asfour, G.; Langer,
B. Lymphatic exosomes promote dendritic cell migration along guidance cues. J. Cell Biol. 2018, 217, 2205–2221. [CrossRef]
[PubMed]
95. Wu, D.; Chen, Q.; Chen, X.; Han, F.; Chen, Z.; Wang, Y. The blood–brain barrier: Structure, regulation, and drug delivery. Signal
Transduct. Target. Ther. 2023, 8, 217. [CrossRef] [PubMed]
96. Izco, M.; Blesa, J.; Schleef, M.; Schmeer, M.; Porcari, R.; Al-Shawi, R.; Ellmerich, S.; de Toro, M.; Gardiner, C.; Seow, Y. Systemic
exosomal delivery of shRNA minicircles prevents parkinsonian pathology. Mol. Ther. 2019, 27, 2111–2122. [CrossRef] [PubMed]
97. Morales-Prieto, D.M.; Stojiljkovic, M.; Diezel, C.; Streicher, P.-E.; Röstel, F.; Lindner, J.; Weis, S.; Schmeer, C.; Marz, M. Peripheral
blood exosomes pass blood-brain-barrier and induce glial cell activation. BioRxiv 2018, 471409.
98. Luarte, A.; Nardocci, G.; Chakraborty, A.; Batiz, L.F.; Pino-Lagos, K.; Wyneken, Ú. Astrocyte-derived extracellular vesicles in
stress-associated mood disorders. Does the immune system get astrocytic? Pharmacol. Res. 2023, 194, 106833. [CrossRef]
99. Sharma, K.; Zhang, Y.; Paudel, K.R.; Kachelmeier, A.; Hansbro, P.M.; Shi, X. The Emerging Role of Pericyte-Derived Extracellular
Vesicles in Vascular and Neurological Health. Cells 2022, 11, 3108. [CrossRef]
100. Fu, S.; Zhang, Y.; Li, Y.; Luo, L.; Zhao, Y.; Yao, Y. Extracellular vesicles in cardiovascular diseases. Cell Death Discov. 2020, 6, 68.
[CrossRef]
101. Murugesan, S.; Hussey, H.; Saravanakumar, L.; Sinkey, R.G.; Sturdivant, A.B.; Powell, M.F.; Berkowitz, D.E. Extracellular vesicles
from women with severe preeclampsia impair vascular endothelial function. Anesth. Analg. 2022, 134, 713–723. [CrossRef]
102. Wolf, P. The nature and significance of platelet products in human plasma. Br. J. Haematol. 1967, 13, 269–288. [CrossRef]
103. Ayers, L.; Nieuwland, R.; Kohler, M.; Kraenkel, N.; Ferry, B.; Leeson, P. Dynamic microvesicle release and clearance within the
cardiovascular system: Triggers and mechanisms. Clin. Sci. 2015, 129, 915–931. [CrossRef]
104. Torti, M.; Vismara, M.; Manfredi, M.; Zarà, M.; Trivigno, S.; Galgano, L.; Barbieri, S.; Canobbio, I.; Guidetti, G. Proteomic and
functional profiling of platelet-derived extracellular vesicles released under physiological or tumor-associated conditions. Cell
Death Discov. 2022, 8, 467.
Int. J. Mol. Sci. 2024, 25, 485 34 of 51
105. Gasecka, A.; Nieuwland, R.; Siljander, P.R.-M. Platelet-derived extracellular vesicles. In Platelets; Elsevier: Amsterdam,
The Netherlands, 2019; pp. 401–416.
106. Brisson, A.R.; Tan, S.; Linares, R.; Gounou, C.; Arraud, N. Extracellular vesicles from activated platelets: A semiquantitative
cryo-electron microscopy and immuno-gold labeling study. Platelets 2017, 28, 263–271. [CrossRef] [PubMed]
107. Puricelli, C.; Boggio, E.; Gigliotti, C.L.; Stoppa, I.; Sutti, S.; Giordano, M.; Dianzani, U.; Rolla, R. Platelets, Protean Cells with
All-Around Functions and Multifaceted Pharmacological Applications. Int. J. Mol. Sci. 2023, 24, 4565. [CrossRef] [PubMed]
108. Puhka, M.; Takatalo, M.; Nordberg, M.-E.; Valkonen, S.; Nandania, J.; Aatonen, M.; Yliperttula, M.; Laitinen, S.; Velagapudi, V.;
Mirtti, T. Metabolomic profiling of extracellular vesicles and alternative normalization methods reveal enriched metabolites and
strategies to study prostate cancer-related changes. Theranostics 2017, 7, 3824. [CrossRef] [PubMed]
109. Reddy, E.C.; Rand, M.L. Procoagulant phosphatidylserine-exposing platelets in vitro and in vivo. Front. Cardiovasc. Med. 2020,
7, 15. [CrossRef] [PubMed]
110. Le, T.; Jia, Z.; Zhang, Y.; Le, S.C.; Chen, J.; Yang, H. An inner activation gate controls TMEM16F phospholipids scrambling.
Biophys. J. 2019, 116, 25a–26a. [CrossRef]
111. Frühbeis, C.; Helmig, S.; Tug, S.; Simon, P.; Krämer-Albers, E.-M. Physical exercise induces rapid release of small extracellular
vesicles into the circulation. J. Extracell. Vesicles 2015, 4, 28239. [CrossRef]
112. Gao, Y.; Li, X.; Qin, Y.; Men, J.; Ren, J.; Li, X.; Xu, C.; Li, Q.; Li, Y.; Cui, W. MPs-ACT, an Assay to Evaluate the Procoagulant
Activity of Microparticles. Clin. Appl. Thromb. Hemost. 2023, 29, 10760296231159374. [CrossRef]
113. Sachetto, A.T.; Archibald, S.J.; Hisada, Y.; Rosell, A.; Havervall, S.; van Es, N.; Nieuwland, R.; Campbell, R.A.; Middleton, E.A.;
Rondina, M.T. Tissue factor activity of small and large extracellular vesicles in different diseases. Res. Pract. Thromb. Haemost.
2023, 7, 100124. [CrossRef]
114. Hu, Y.; Repa, A.; Lisman, T.; Yerlikaya-Schatten, G.; Hau, C.; Pabinger, I.; Ay, C.; Nieuwland, R.; Thaler, J. Extracellular vesicles
from amniotic fluid, milk, saliva, and urine expose complexes of tissue factor and activated factor VII. J. Thromb. Haemost. 2022,
20, 2306–2312. [CrossRef]
115. Hong, C.-W. Extracellular vesicles of neutrophils. Immune Netw. 2018, 18. [CrossRef] [PubMed]
116. Zhou, X.; Xie, F.; Wang, L.; Zhang, L.; Zhang, S.; Fang, M.; Zhou, F. The function and clinical application of extracellular vesicles
in innate immune regulation. Cell. Mol. Immunol. 2020, 17, 323–334. [CrossRef] [PubMed]
117. Mehrani, Y.; Rahimi Junqani, R.; Morovati, S.; Mehrani, H.; Karimi, N.; Ghasemi, S. The Importance of Neutrophils in Osteoarthri-
tis: Current Concepts and Therapeutic Perspectives. Immuno 2023, 3, 250–272. [CrossRef]
118. Yu, Y.; Jin, H.; Li, L.; Zhang, X.; Zheng, C.; Gao, X.; Yang, Y.; Sun, B. An injectable, activated neutrophil-derived exosome
mimetics/extracellular matrix hybrid hydrogel with antibacterial activity and wound healing promotion effect for diabetic
wound therapy. J. Nanobiotechnol. 2023, 21, 308. [CrossRef] [PubMed]
119. Chen, H.-P.; Wang, X.-Y.; Pan, X.-Y.; Hu, W.-W.; Cai, S.-T.; Joshi, K.; Deng, L.-H.; Ma, D. Circulating neutrophil-derived
microparticles associated with the prognosis of patients with sepsis. J. Inflamm. Res. 2020, 1113–1124. [CrossRef] [PubMed]
120. Moon, M.J.; McFadyen, J.D.; Peter, K. Caught at the scene of the crime: Platelets and neutrophils are conspirators in thrombosis.
Arterioscler. Thromb. Vasc. Biol. 2022, 42, 63–66. [CrossRef] [PubMed]
121. Brakhage, A.A.; Zimmermann, A.-K.; Rivieccio, F.; Visser, C.; Blango, M.G. Host-derived extracellular vesicles for antimicrobial
defense. MicroLife 2021, 2, uqab003. [CrossRef]
122. Kolonics, F.; Kajdácsi, E.; Farkas, V.J.; Veres, D.S.; Khamari, D.; Kittel, Á.; Merchant, M.L.; McLeish, K.R.; Lőrincz, Á.M.; Ligeti, E.
Neutrophils produce proinflammatory or anti-inflammatory extracellular vesicles depending on the environmental conditions. J.
Leucoc. Biol. 2021, 109, 793–806. [CrossRef]
123. Lim, K.; Hyun, Y.-M.; Lambert-Emo, K.; Capece, T.; Bae, S.; Miller, R.; Topham, D.J.; Kim, M. Neutrophil trails guide influenza-
specific CD8+ T cells in the airways. Science 2015, 349, aaa4352. [CrossRef]
124. Popēna, I.; Ābols, A.; Saulı̄te, L.; Pleiko, K.; Zandberga, E.; Jēkabsons, K.; Endzelin, š, E.; Llorente, A.; Linē, A.; Riekstin, a, U. Effect
of colorectal cancer-derived extracellular vesicles on the immunophenotype and cytokine secretion profile of monocytes and
macrophages. Cell Commun. Signal. 2018, 16, 17. [CrossRef]
125. Raposo, G.; Nijman, H.W.; Stoorvogel, W.; Liejendekker, R.; Harding, C.V.; Melief, C.; Geuze, H.J. B lymphocytes secrete
antigen-presenting vesicles. J. Exp. Med. 1996, 183, 1161–1172. [CrossRef] [PubMed]
126. Zitvogel, L.; Regnault, A.; Lozier, A.; Wolfers, J.; Flament, C.; Tenza, D.; Ricciardi-Castagnoli, P.; Raposo, G.; Amigorena, S.
Eradication of established murine tumors using a novel cell-free vaccine: Dendritic cell derived exosomes. Nat. Med. 1998, 4,
594–600. [CrossRef] [PubMed]
127. Nolte-‘t Hoen, E.N.; Buschow, S.I.; Anderton, S.M.; Stoorvogel, W.; Wauben, M.H. Activated T cells recruit exosomes secreted by
dendritic cells via LFA-1. Blood J. Am. Soc. Hematol. 2009, 113, 1977–1981. [CrossRef] [PubMed]
128. Théry, C.; Duban, L.; Segura, E.; Véron, P.; Lantz, O.; Amigorena, S. Indirect activation of naïve CD4+ T cells by dendritic
cell–derived exosomes. Nat. Immunol. 2002, 3, 1156–1162. [CrossRef] [PubMed]
129. Hatami, Z.; Hashemi, Z.S.; Eftekhary, M.; Amiri, A.; Karpisheh, V.; Nasrollahi, K.; Jafari, R. Natural killer cell-derived exosomes
for cancer immunotherapy: Innovative therapeutics art. Cancer Cell Int. 2023, 23, 1–18. [CrossRef] [PubMed]
130. Gill, M.; Motta-Mejia, C.; Kandzija, N.; Cooke, W.; Zhang, W.; Cerdeira, A.S.; Bastie, C.; Redman, C.; Vatish, M. Placental
syncytiotrophoblast-derived extracellular vesicles carry active NEP(neprilysin) and are increased in preeclampsia. Hypertension
2019, 73, 1112–1119. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2024, 25, 485 35 of 51
131. Buca, D.; Bologna, G.; D’Amico, A.; Cugini, S.; Musca, F.; Febbo, M.; D’Arcangelo, D.; Buca, D.; Simeone, P.; Liberati, M.
Extracellular vesicles in feto–maternal crosstalk and pregnancy disorders. Int. J. Mol. Sci. 2020, 21, 2120. [CrossRef]
132. Huang, C.-C.; Hsueh, Y.-W.; Chang, C.-W.; Hsu, H.-C.; Yang, T.-C.; Lin, W.-C.; Chang, H.-M. Establishment of the fetal-maternal
interface: Developmental events in human implantation and placentation. Front. Cell Dev. Biol. 2023, 11, 1200330. [CrossRef]
133. Göhner, C.; Plösch, T.; Faas, M.M. Immune-modulatory effects of syncytiotrophoblast extracellular vesicles in pregnancy and
preeclampsia. Placenta 2017, 60, S41–S51. [CrossRef]
134. Ren, Y.; Yang, J.; Xie, R.; Gao, L.; Yang, Y.; Fan, H.; Qian, K. Exosomal-like vesicles with immune-modulatory features are present
in human plasma and can induce CD4+ T-cell apoptosis in vitro. Transfusion 2011, 51, 1002–1011. [CrossRef]
135. Karlsson, M.; Lundin, S.; Dahlgren, U.; Kahu, H.; Pettersson, I.; Telemo, E. “Tolerosomes” are produced by intestinal epithelial
cells. Eur. J. Immunol. 2001, 31, 2892–2900. [CrossRef] [PubMed]
136. Prado, N.; Marazuela, E.G.; Segura, E.; Fernandez-Garcia, H.; Villalba, M.; Théry, C.; Rodríguez, R.A.; Batanero, E. Exosomes
from bronchoalveolar fluid of tolerized mice prevent allergic reaction. J. Immunol. 2008, 181, 1519–1525. [CrossRef] [PubMed]
137. Hiltbrunner, S.; Larssen, P.; Eldh, M.; Martinez-Bravo, M.-J.; Wagner, A.K.; Karlsson, M.C.; Gabrielsson, S. Exosomal cancer
immunotherapy is independent of MHC molecules on exosomes. Oncotarget 2016, 7, 38707. [CrossRef] [PubMed]
138. Monguió-Tortajada, M.; Lauzurica-Valdemoros, R.; Borràs, F.E. Tolerance in organ transplantation: From conventional immuno-
suppression to extracellular vesicles. Front. Immunol. 2014, 5, 416. [CrossRef] [PubMed]
139. Xia, X.; Wang, Y.; Zheng, J.C. Extracellular vesicles, from the pathogenesis to the therapy of neurodegenerative diseases. Transl.
Neurodegener. 2022, 11, 1–31. [CrossRef] [PubMed]
140. Dalla, P.V.; Santos, J.; Milthorpe, B.K.; Padula, M.P. Selectively-packaged proteins in breast cancer extracellular vesicles involved
in metastasis. Int. J. Mol. Sci. 2020, 21, 4990. [CrossRef] [PubMed]
141. Chatterjee, E.; Rodosthenous, R.S.; Kujala, V.; Karalis, K.; Spanos, M.; Lehmann, H.I.; Oliveira, G.O.P.D.; Shi, M.; Flem-
ing, T.W.M.; Li, G. Circulating Extracellular Vesicles in Human Cardiorenal Syndrome Promote Renal Injury. medRxiv 2023.
medRxiv:2023.02.07.23285599.
142. Wang, C.; Li, S.-W.; Zhong, X.; Liu, B.-C.; Lv, L.-L. An update on renal fibrosis: From mechanisms to therapeutic strategies with a
focus on extracellular vesicles. Kidney Res. Clin. Pract. 2023, 42, 174. [CrossRef]
143. Mazzariol, M.; Camussi, G.; Brizzi, M.F. Extracellular vesicles tune the immune system in renal disease: A focus on systemic
lupus erythematosus, antiphospholipid syndrome, thrombotic microangiopathy and ANCA-Vasculitis. Int. J. Mol. Sci. 2021,
22, 4194. [CrossRef]
144. Lu, Y.; Zhang, R.; Gu, X.; Wang, X.; Xi, P.; Chen, X. Exosomes from tubular epithelial cells undergoing epithelial-to-mesenchymal
transition promote renal fibrosis by M1 macrophage activation. FASEB BioAdv. 2023, 5, 101–113. [CrossRef]
145. Shen, A.-R.; Lv, L.-L. Tubule epithelial cells and fibroblasts communication: Vicious cycle of renal fibrosis. EBioMedicine 2022,
86, 104360. [CrossRef] [PubMed]
146. Kosanović, M.; Llorente, A.; Glamočlija, S.; Valdivielso, J.M.; Bozic, M. Extracellular Vesicles and Renal Fibrosis: An Odyssey
toward a New Therapeutic Approach. Int. J. Mol. Sci. 2021, 22, 3887. [CrossRef] [PubMed]
147. Tian, Z.-y.; Li, Z.; Chu, L.; Liu, Y.; He, J.-r.; Xin, Y.; Li, A.-m.; Zhang, H. Iron metabolism and chronic inflammation in IgA
nephropathy. Ren. Fail. 2023, 45, 2195012. [CrossRef] [PubMed]
148. Wang, D.; Zhao, T.; Zhao, Y.; Yin, Y.; Huang, Y.; Cheng, Z.; Wang, B.; Liu, S.; Pan, M.; Sun, D. PPARγ Mediates the Anti-Epithelial-
Mesenchymal Transition Effects of FGF1∆HBS in Chronic Kidney Diseases via Inhibition of TGF-β1/SMAD3 Signaling. Front.
Pharmacol. 2021, 12, 690535. [CrossRef]
149. Gomes, R.N.; Manuel, F.; Nascimento, D.S. The bright side of fibroblasts: Molecular signature and regenerative cues in major
organs. NPJ Regen. Med. 2021, 6, 43. [CrossRef]
150. Martinez-Arroyo, O.; Ortega, A.; Redon, J.; Cortes, R. Therapeutic potential of extracellular vesicles in hypertension-associated
kidney disease. Hypertension 2021, 77, 28–38. [CrossRef]
151. Rigalli, J.P.; Barros, E.R.; Sommers, V.; Bindels, R.J.; Hoenderop, J.G. Novel aspects of extracellular vesicles in the regulation of
renal physiological and pathophysiological processes. Front. Cell Dev. Biol. 2020, 8, 244. [CrossRef]
152. Zhu, L.; Huang, Y.; Li, H.; Shao, S. Helicobacter pylori promotes gastric cancer progression through the tumor microenvironment.
Appl. Microbiol. Biotechnol. 2022, 106, 4375–4385. [CrossRef]
153. Usui, Y.; Taniyama, Y.; Endo, M.; Koyanagi, Y.N.; Kasugai, Y.; Oze, I.; Ito, H.; Imoto, I.; Tanaka, T.; Tajika, M. Helicobacter pylori,
homologous-recombination genes, and gastric cancer. N. Engl. J. Med. 2023, 388, 1181–1190. [CrossRef]
154. Baj, J.; Forma, A.; Sitarz, M.; Portincasa, P.; Garruti, G.; Krasowska, D.; Maciejewski, R. Helicobacter pylori virulence factors—
Mechanisms of bacterial pathogenicity in the gastric microenvironment. Cells 2020, 10, 27. [CrossRef]
155. Bucci, P.; Barbaglia, Y.; Tedeschi, F.; Zalazar, F. Helicobacter pylori infection: A balance between bacteria and host. Rev. Argent. De
Microbiol. 2023, 55, 60–67. [CrossRef] [PubMed]
156. Choi, H.-I.; Choi, J.-P.; Seo, J.; Kim, B.J.; Rho, M.; Han, J.K.; Kim, J.G. Helicobacter pylori-derived extracellular vesicles increased in
the gastric juices of gastric adenocarcinoma patients and induced inflammation mainly via specific targeting of gastric epithelial
cells. Exp. Mol. Med. 2017, 49, e330. [CrossRef] [PubMed]
157. Sun, D.; Chen, P.; Xi, Y.; Sheng, J. From trash to treasure: The role of bacterial extracellular vesicles in gut health and disease.
Front. Immunol. 2023, 14, 1274295. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2024, 25, 485 36 of 51
158. Navashenaq, J.G.; Shabgah, A.G.; Banach, M.; Jamialahmadi, T.; Penson, P.E.; Johnston, T.P.; Sahebkar, A. The interaction of
Helicobacter pylori with cancer immunomodulatory stromal cells: New insight into gastric cancer pathogenesis. Semin. Cancer
Biol. 2022, 86, 951–959. [CrossRef] [PubMed]
159. Zhao, L.; Ye, Y.; Gu, L.; Jian, Z.; Stary, C.M.; Xiong, X. Extracellular vesicle-derived miRNA as a novel regulatory system for
bi-directional communication in gut-brain-microbiota axis. J. Transl. Med. 2021, 19, 1–12. [CrossRef]
160. Kotsiliti, E. Gut microbiome and autism spectrum disorder. Nat. Rev. Gastroenterol. Hepatol. 2022, 19, 6. [CrossRef]
161. Taniya, M.A.; Chung, H.-J.; Al Mamun, A.; Alam, S.; Aziz, M.A.; Emon, N.U.; Islam, M.M.; Podder, B.R.; Ara Mimi, A.; Aktar
Suchi, S. Role of gut microbiome in autism spectrum disorder and its therapeutic regulation. Front. Cell. Infect. Microbiol. 2022,
12, 998. [CrossRef]
162. Cuesta, C.M.; Guerri, C.; Ureña, J.; Pascual, M. Role of microbiota-derived extracellular vesicles in gut-brain communication. Int.
J. Mol. Sci. 2021, 22, 4235. [CrossRef]
163. Zaidi, S.; Ali, K.; Khan, A.U. It’s all relative: Analyzing microbiome compositions, its significance, pathogenesis and microbiota
derived biofilms: Challenges and opportunities for disease intervention. Arch. Microbiol. 2023, 205, 257. [CrossRef]
164. Argilés, J.M.; López-Soriano, F.J.; Stemmler, B.; Busquets, S. Cancer-associated cachexia—Understanding the tumour macroenvi-
ronment and microenvironment to improve management. Nat. Rev. Clin. Oncol. 2023, 20, 250–264. [CrossRef]
165. Vitucci, D.; Martone, D.; Alfieri, A.; Buono, P. Muscle-derived exosomes and exercise in cancer prevention. Front. Mol. Med. 2023,
3, 16. [CrossRef]
166. Catapano, F.; Scaglioni, D.; Maresh, K.; Ala, P.; Domingos, J.; Selby, V.; Ricotti, V.; Phillips, L.; Servais, L.; Seferian, A. Novel
free-circulating and extracellular vesicle-derived miRNAs dysregulated in Duchenne muscular dystrophy. Epigenomics 2020, 12,
1899–1915. [CrossRef] [PubMed]
167. Matsuzaka, Y.; Hirai, Y.; Hashido, K.; Okada, T. Therapeutic Application of Extracellular Vesicles-Capsulated Adeno-Associated
Virus Vector via nSMase2/Smpd3, Satellite, and Immune Cells in Duchenne Muscular Dystrophy. Int. J. Mol. Sci. 2022, 23, 1551.
[CrossRef] [PubMed]
168. Zhao, J.; Zhang, B.; Meng, W.; Hu, J. Elucidating a fresh perspective on the interplay between exosomes and rheumatoid arthritis.
Front. Cell Dev. Biol. 2023, 11, 1177303. [CrossRef] [PubMed]
169. Liu, Z.; Zhuang, Y.; Fang, L.; Yuan, C.; Wang, X.; Lin, K. Breakthrough of extracellular vesicles in pathogenesis, diagnosis and
treatment of osteoarthritis. Bioact. Mater. 2023, 22, 423–452. [CrossRef] [PubMed]
170. Scheck, S.; Paterson, E.S.; Henry, C.E. A promising future for endometriosis diagnosis and therapy: Extracellular vesicles-a
systematic review. Reprod. Biol. Endocrinol. 2022, 20, 174. [CrossRef] [PubMed]
171. Che, X.; Jian, F.; Chen, C.; Liu, C.; Liu, G.; Feng, W. PCOS serum-derived exosomal miR-27a-5p stimulates endometrial cancer
cells migration and invasion. J. Mol. Endocrinol. 2020, 64, 1–12. [CrossRef]
172. Chen, Y.; Huang, P.; Han, C.; Li, J.; Liu, L.; Zhao, Z.; Gao, Y.; Qin, Y.; Xu, Q.; Yan, Y. Association of placenta-derived extracellular
vesicles with pre-eclampsia and associated hypercoagulability: A clinical observational study. BJOG: Int. J. Obstet. Gynaecol. 2021,
128, 1037–1046. [CrossRef]
173. Reyes, A.B.; Burger, D. Small extracellular vesicles: A new player in GDM pathogenesis. Clin. Sci. 2022, 136, 1873–1875. [CrossRef]
174. Kulkarni, R.; Teves, M.E.; Han, A.X.; McAllister, J.M.; Strauss III, J.F. Colocalization of polycystic ovary syndrome candidate gene
products in theca cells suggests novel signaling pathways. J. Endocr. Soc. 2019, 3, 2204–2223. [CrossRef]
175. Cui, C.; Wang, J.; Han, X.; Wang, Q.; Zhang, S.; Liang, S.; Li, H.; Meng, L.; Zhang, C.; Chen, H. Identification of small extracellular
vesicle-linked miRNA specifically derived from intrafollicular cells in women with polycystic ovary syndrome. Reprod. Biomed.
Online 2021, 42, 870–880. [CrossRef] [PubMed]
176. Koiou, E.; Tziomalos, K.; Katsikis, I.; Papadakis, E.; Kandaraki, E.A.; Panidis, D. Platelet-derived microparticles in over-
weight/obese women with the polycystic ovary syndrome. Gynecol. Endocrinol. 2013, 29, 250–253. [CrossRef] [PubMed]
177. Willis, G.; Connolly, K.; Ladell, K.; Davies, T.; Guschina, I.; Ramji, D.; Miners, K.; Price, D.; Clayton, A.; James, P. Young women
with polycystic ovary syndrome have raised levels of circulating annexin V-positive platelet microparticles. Hum. Reprod. 2014,
29, 2756–2763. [CrossRef] [PubMed]
178. Steegers, E.A.; Von Dadelszen, P.; Duvekot, J.J.; Pijnenborg, R. Pre-eclampsia. Lancet 2010, 376, 631–644. [CrossRef] [PubMed]
179. Awoyemi, T.; Jiang, S.; Bjarkadottir, B.; Rahbar, M.; Logenthiran, P.; Collett, G.; Zhang, W.; Cribbs, A.; Cerdeira, A.S.; Vatish, M.
Identification of Novel Syncytiotrophoblast Membrane Extracellular Vesicles Derived Protein Biomarkers in Preeclampsia: A
Cross-Sectional Study. medRxiv 2023. medRxiv:2023.06.03.23290935.
180. Han, C.; Wang, C.; Chen, Y.; Wang, J.; Xu, X.; Hilton, T.; Cai, W.; Zhao, Z.; Wu, Y.; Li, K. Placenta-derived extracellular vesicles
induce preeclampsia in mouse models. Haematologica 2020, 105, 1686. [CrossRef] [PubMed]
181. Salomon, C.; Guanzon, D.; Scholz-Romero, K.; Longo, S.; Correa, P.; Illanes, S.E.; Rice, G.E. Placental exosomes as early biomarker
of preeclampsia: Potential role of exosomal microRNAs across gestation. J. Clin. Endocrinol. Metab. 2017, 102, 3182–3194.
[CrossRef]
182. James-Allan, L.B.; Rosario, F.J.; Barner, K.; Lai, A.; Guanzon, D.; McIntyre, H.D.; Lappas, M.; Powell, T.L.; Salomon, C.; Jansson, T.
Regulation of glucose homeostasis by small extracellular vesicles in normal pregnancy and in gestational diabetes. FASEB J. 2020,
34, 5724–5739. [CrossRef]
183. Bathla, T.; Abolbaghaei, A.; Reyes, A.B.; Burger, D. Extracellular vesicles in gestational diabetes mellitus: A scoping review.
Diabetes Vasc. Dis. Res. 2022, 19, 14791641221093901. [CrossRef]
Int. J. Mol. Sci. 2024, 25, 485 37 of 51
184. Liu, X.; Zhang, L.; Cao, Y.; Jia, H.; Li, X.; Li, F.; Zhang, S.; Zhang, J. Neuroinflammation of traumatic brain injury: Roles of
extracellular vesicles. Front. Immunol. 2023, 13, 1088827. [CrossRef]
185. Lin, H.; Chen, H.; Qi, B.; Jiang, Y.; Lian, N.; Zhuang, X.; Yu, Y. Brain-derived extracellular vesicles mediated coagulopathy,
inflammation and apoptosis after sepsis. Thromb. Res. 2021, 207, 85–95. [CrossRef] [PubMed]
186. Marostica, G.; Gelibter, S.; Gironi, M.; Nigro, A.; Furlan, R. Extracellular vesicles in neuroinflammation. Front. Cell Dev. Biol. 2021,
8, 623039. [CrossRef] [PubMed]
187. Gill, J.; Mustapic, M.; Diaz-Arrastia, R.; Lange, R.; Gulyani, S.; Diehl, T.; Motamedi, V.; Osier, N.; Stern, R.A.; Kapogiannis, D.
Higher exosomal tau, amyloid-beta 42 and IL-10 are associated with mild TBIs and chronic symptoms in military personnel.
Brain Inj. 2018, 32, 1359–1366. [CrossRef] [PubMed]
188. Li, L.; Li, F.; Bai, X.; Jia, H.; Wang, C.; Li, P.; Zhang, Q.; Guan, S.; Peng, R.; Zhang, S. Circulating extracellular vesicles from
patients with traumatic brain injury induce cerebrovascular endothelial dysfunction. Pharmacol. Res. 2023, 192, 106791. [CrossRef]
[PubMed]
189. Pistono, C.; Osera, C.; Cuccia, M.; Bergamaschi, R. Roles of Extracellular Vesicles in Multiple Sclerosis: From Pathogenesis to
Potential Tools as Biomarkers and Therapeutics. Sclerosis 2023, 1, 91–112. [CrossRef]
190. Yas, A. Preparation and characterization of L–Dopa loaded chitosan–based dry powder: Rescue/continuous supplement in
Parkinson’s disease via inhalation. World J. Pharm. Sci. 2016, 23–36.
191. Iba, M.; Guo, J.L.; McBride, J.D.; Zhang, B.; Trojanowski, J.Q.; Lee, V.M.-Y. Synthetic tau fibrils mediate transmission of
neurofibrillary tangles in a transgenic mouse model of Alzheimer’s-like tauopathy. J. Neurosci. 2013, 33, 1024–1037. [CrossRef]
192. Ruan, Z.; Pathak, D.; Venkatesan Kalavai, S.; Yoshii-Kitahara, A.; Muraoka, S.; Bhatt, N.; Takamatsu-Yukawa, K.; Hu, J.; Wang, Y.;
Hersh, S. Alzheimer’s disease brain-derived extracellular vesicles spread tau pathology in interneurons. Brain 2021, 144, 288–309.
[CrossRef]
193. Aulston, B.; Liu, Q.; Mante, M.; Florio, J.; Rissman, R.A.; Yuan, S.H. Extracellular vesicles isolated from familial Alzheimer’s
disease neuronal cultures induce aberrant tau phosphorylation in the wild-type mouse brain. J. Alzheimer’s Dis. 2019, 72, 575–585.
[CrossRef]
194. Alvarez-Erviti, L.; Seow, Y.; Yin, H.; Betts, C.; Lakhal, S.; Wood, M.J. Delivery of siRNA to the mouse brain by systemic injection of
targeted exosomes. Nat. Biotechnol. 2011, 29, 341–345. [CrossRef]
195. Stuendl, A.; Kunadt, M.; Kruse, N.; Bartels, C.; Moebius, W.; Danzer, K.M.; Mollenhauer, B.; Schneider, A. Induction of α-synuclein
aggregate formation by CSF exosomes from patients with Parkinson’s disease and dementia with Lewy bodies. Brain 2016, 139,
481–494. [CrossRef] [PubMed]
196. Liu, Q.; Piao, H.; Wang, Y.; Zheng, D.; Wang, W. Circulating exosomes in cardiovascular disease: Novel carriers of biological
information. Biomed. Pharmacother. 2021, 135, 111148. [CrossRef] [PubMed]
197. Zhang, X.; Wu, Y.; Cheng, Q.; Bai, L.; Huang, S.; Gao, J. Extracellular vesicles in cardiovascular diseases: Diagnosis and therapy.
Front. Cell Dev. Biol. 2022, 10, 875376. [CrossRef] [PubMed]
198. Reiss, A.B.; Ahmed, S.; Johnson, M.; Saeedullah, U.; De Leon, J. Exosomes in Cardiovascular Disease: From Mechanism to
Therapeutic Target. Metabolites 2023, 13, 479. [CrossRef] [PubMed]
199. Buffolo, F.; Monticone, S.; Camussi, G.; Aikawa, E. Role of extracellular vesicles in the pathogenesis of vascular damage.
Hypertension 2022, 79, 863–873. [CrossRef] [PubMed]
200. Fujioka, Y.; Otani, K.; Okada, M.; Yamawaki, H. Plasma small extracellular vesicles in hypertensive rats impair reactivity of
isolated blood vessels. J. Vet. Med. Sci. 2020, 82, 897–902. [CrossRef] [PubMed]
201. Drożdż, D.; Drożdż, M.; Wójcik, M. Endothelial dysfunction as a factor leading to arterial hypertension. Pediatr. Nephrol. 2022, 38,
2973–2985. [CrossRef]
202. Barsom, S.H.; Glasstetter, L.M.; Siddiqi, S.; Rajagopalan, K.S.; Eirin, A.; Lerman, L.O. Emergent players in renovascular disease.
Clin. Sci. 2022, 136, 239–256. [CrossRef]
203. Rui, R.; Yang, H.; Liu, Y.; Zhou, Y.; Xu, X.; Li, C.; Liu, S. Effects of berberine on atherosclerosis. Front. Pharmacol. 2021,
3074, 764175. [CrossRef]
204. Zhang, J. Biomarkers of endothelial activation and dysfunction in cardiovascular diseases. Rev. Cardiovasc. Med. 2022, 23, 73.
[CrossRef]
205. Sheng, L.; Zhang, M.; Lu, Y.; Han, X.; Yu, L.; Zhang, W.; Liu, S.; Liu, Y. Advances in Molecular Pathology of Obstructive Sleep
Apnea. Molecules 2022, 27, 8422.
206. Jang, S.; Palzer, E.F.; Rudser, K.D.; Fox, C.K.; Hebbel, R.P.; Dengel, D.R.; Milbauer, L.; Kelly, A.S.; Ryder, J.R. Relationship of
Endothelial Microparticles to Obesity and Cardiovascular Disease Risk in Children and Adolescents. J. Am. Heart Assoc. 2022,
11, e026430. [CrossRef] [PubMed]
207. Nik Ibrahim, N.N.I.; Abdul Rahman, R.; Azlan, M.; Abd Aziz, A.; Ghulam Rasool, A.H. Endothelial Microparticles as Potential
Biomarkers in the Assessment of Endothelial Dysfunction in Hypercholesterolemia. Medicina 2022, 58, 824. [CrossRef]
208. Grange, C.; Bussolati, B. Extracellular vesicles in kidney disease. Nat. Rev. Nephrol. 2022, 18, 499–513. [CrossRef] [PubMed]
209. Cao, W.; Zhao, B.; Gui, L.; Sun, X.; Zhang, Z.; Huang, L. The role and mechanism of miR-92a in endothelial autophagy.
Res. Sq. 2023. [CrossRef]
210. Cheng, P.; Wang, X.; Liu, Q.; Yang, T.; Qu, H.; Zhou, H. Extracellular vesicles mediate biological information delivery: A
double-edged sword in cardiac remodeling after myocardial infarction. Front. Pharmacol. 2023, 14, 1067992. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2024, 25, 485 38 of 51
211. Gong, S.; Wang, C.; Xiong, J.; Zhao, J.; Yang, K. Activated platelets, the booster of chronic kidney disease and cardiovascular
complications. Kidney Dis. 2022, 8, 297–307. [CrossRef] [PubMed]
212. Figuer, A.; Alique, M.; Valera, G.; Serroukh, N.; Ceprían, N.; de Sequera, P.; Morales, E.; Carracedo, J.; Ramírez, R.; Bodega, G.
New mechanisms involved in the development of cardiovascular disease in chronic kidney disease. Nefrología 2023, 43, 63–80.
[CrossRef]
213. Li, N.; Wu, B.; Wang, J.; Yan, Y.; An, P.; Li, Y.; Liu, Y.; Hou, Y.; Qing, X.; Niu, L. Differential proteomic patterns of plasma
extracellular vesicles show potential to discriminate β-thalassemia subtypes. Iscience 2023, 26, 106048. [CrossRef]
214. Carandina, A.; Favero, C.; Sacco, R.M.; Hoxha, M.; Torgano, G.; Montano, N.; Bollati, V.; Tobaldini, E. The Role of Extracellular
Vesicles in Ischemic Stroke Severity. Biology 2022, 11, 1489. [CrossRef]
215. Suades, R.; Padró, T.; Vilahur, G.; Badimon, L. Platelet-released extracellular vesicles: The effects of thrombin activation. Cell. Mol.
Life Sci. 2022, 79, 190. [CrossRef] [PubMed]
216. Abdelmaksoud, M.F.; Abdelmaksoud, S.S.; Abdelsamee, H.F.; Ezzelregal, H.G.; Alfeky, M.A. Platelets derived microparticles in
COVID-19: Correlation to inflammatory and coagulation state. J. Appl. Hematol. 2021, 12, 195.
217. Ebeyer-Masotta, M.; Eichhorn, T.; Weiss, R.; Lauková, L.; Weber, V. Activated platelets and platelet-derived extracellular vesicles
mediate COVID-19-associated immunothrombosis. Front. Cell Dev. Biol. 2022, 10, 914891. [CrossRef] [PubMed]
218. Ferreira, P.; Bozbas, E.; Tannetta, S.; Alroqaiba, N.; Zhou, R.; Crawley, J.; Gibbins, J.; Jones, C.; Ahnström, J.; Yaqoob, P. Mode
of induction of platelet-derived extracellular vesicles is a critical determinant of their phenotype and function. Sci. Rep. 2020,
10, 18061. [CrossRef] [PubMed]
219. Zong, Y.; Pruner, I.; Antovic, A.; Taxiarchis, A.; Vila, Z.P.; Soutari, N.; Mobarrez, F.; Chaireti, R.; Widengren, J.; Piguet, J.
Phosphatidylserine positive microparticles improve hemostasis in in-vitro hemophilia A plasma models. Sci. Rep. 2020, 10, 7871.
[CrossRef] [PubMed]
220. Litvinov, R.I.; Nabiullina, R.M.; Zubairova, L.D.; Shakurova, M.A.; Andrianova, I.A.; Weisel, J.W. Lytic susceptibility, structure,
and mechanical properties of fibrin in systemic lupus erythematosus. Front. Immunol. 2019, 10, 1626. [CrossRef] [PubMed]
221. Chaudhary, P.K.; Kim, S.; Kim, S. Shedding Light on the Cell Biology of Platelet-Derived Extracellular Vesicles and Their
Biomedical Applications. Life 2023, 13, 1403. [CrossRef]
222. Eustes, A.S.; Dayal, S. The role of platelet-derived extracellular vesicles in immune-mediated thrombosis. Int. J. Mol. Sci. 2022,
23, 7837. [CrossRef]
223. Mabrouk, M.; Guessous, F.; Naya, A.; Merhi, Y.; Zaid, Y. The Pathophysiological Role of Platelet-Derived Extracellular Vesicles. In
Seminars in Thrombosis and Hemostasis; Thieme Medical Publishers, Inc.: New York, NY, USA, 2022.
224. Garcia, C.; Compagnon, B.; Poëtte, M.; Gratacap, M.-P.; Lapébie, F.-X.; Voisin, S.; Minville, V.; Payrastre, B.; Vardon-Bounes, F.;
Ribes, A. Platelet versus megakaryocyte: Who is the real bandleader of thromboinflammation in sepsis? Cells 2022, 11, 1507.
[CrossRef]
225. Li, X.; Wang, Q. Platelet-Derived Microparticles and Autoimmune Diseases. Int. J. Mol. Sci. 2023, 24, 10275. [CrossRef]
226. Krajewska-Włodarczyk, M.; Owczarczyk-Saczonek, A.; Żuber, Z.; Wojtkiewicz, M.; Wojtkiewicz, J. Role of microparticles in the
pathogenesis of inflammatory joint diseases. Int. J. Mol. Sci. 2019, 20, 5453. [CrossRef] [PubMed]
227. Li, Y.; Wang, H.; Zhao, Z.; Yang, Y.; Meng, Z.; Qin, L. Effects of the interactions between platelets with other cells in tumor growth
and progression. Front. Immunol. 2023, 14, 1165989. [CrossRef] [PubMed]
228. Mackman, N.; Hisada, Y. Therapeutic potential of granulocyte microvesicles in sepsis. Blood J. Am. Soc. Hematol. 2022, 139,
2269–2271. [CrossRef] [PubMed]
229. Xu, M.; Du, R.; Xing, W.; Chen, X.; Wan, J.; Wang, S.; Xiong, L.; Nandakumar, K.S.; Holmdahl, R.; Geng, H. Platelets derived
citrullinated proteins and microparticles are potential autoantibodies ACPA targets in RA patients. Front. Immunol. 2023,
14, 1084283. [CrossRef] [PubMed]
230. Mitsuhashi, S.; Feldbrügge, L.; Csizmadia, E.; Mitsuhashi, M.; Robson, S.C.; Moss, A.C. Luminal extracellular vesicles (EVs) in
inflammatory bowel disease(IBD) exhibit proinflammatory effects on epithelial cells and macrophages. Inflamm. Bowel Dis. 2016,
22, 1587–1595. [CrossRef] [PubMed]
231. Akbar, N.; Digby, J.E.; Cahill, T.J.; Tavare, A.N.; Corbin, A.L.; Saluja, S.; Dawkins, S.; Edgar, L.; Rawlings, N.; Ziberna, K.
Endothelium-derived extracellular vesicles promote splenic monocyte mobilization in myocardial infarction. JCI Insight 2017,
2, e93344. [CrossRef]
232. Jansen, F.; Nickenig, G.; Werner, N. Extracellular vesicles in cardiovascular disease: Potential applications in diagnosis, prognosis,
and epidemiology. Circ. Res. 2017, 120, 1649–1657. [CrossRef]
233. Atehortúa, L.; Rojas, M.; Vásquez, G.; Muñoz-Vahos, C.H.; Vanegas-García, A.; Posada-Duque, R.A.; Castaño, D. Endothelial
activation and injury by microparticles in patients with systemic lupus erythematosus and rheumatoid arthritis. Arthritis Res.
Ther. 2019, 21, 1–15. [CrossRef]
234. Wang, Y.; Xie, Y.; Zhang, A.; Wang, M.; Fang, Z.; Zhang, J. Exosomes: An emerging factor in atherosclerosis. Biomed. Pharmacother.
2019, 115, 108951. [CrossRef]
235. Blonda, M.; Amoruso, A.; Martino, T.; Avolio, C. New insights into immune cell-derived extracellular vesicles in multiple sclerosis.
Front. Neurol. 2018, 9, 604. [CrossRef]
Int. J. Mol. Sci. 2024, 25, 485 39 of 51
236. Casella, G.; Colombo, F.; Finardi, A.; Descamps, H.; Ill-Raga, G.; Spinelli, A.; Podini, P.; Bastoni, M.; Martino, G.; Muzio, L.
Extracellular vesicles containing IL-4 modulate neuroinflammation in a mouse model of multiple sclerosis. Mol. Ther. 2018, 26,
2107–2118. [CrossRef] [PubMed]
237. Kimura, K.; Hohjoh, H.; Yamamura, T. The role for exosomal microRNAs in disruption of regulatory T cell homeostasis in
multiple sclerosis. J. Exp. Neurosci. 2018, 12, 1179069518764892. [CrossRef] [PubMed]
238. Pisetsky, D.S. Evolving story of autoantibodies in systemic lupus erythematosus. J. Autoimmun. 2020, 110, 102356. [CrossRef]
[PubMed]
239. Pang, H.; Luo, S.; Xiao, Y.; Xia, Y.; Li, X.; Huang, G.; Xie, Z.; Zhou, Z. Emerging roles of exosomes in T1DM. Front. Immunol. 2020,
11, 593348. [CrossRef]
240. Wu, W.-C.; Song, S.-J.; Zhang, Y.; Li, X. Role of extracellular vesicles in autoimmune pathogenesis. Front. Immunol. 2020,
11, 579043. [CrossRef] [PubMed]
241. Liu, J.; Zhang, Y.; Tian, Y.; Huang, W.; Tong, N.; Fu, X. Integrative biology of extracellular vesicles in diabetes mellitus and diabetic
complications. Theranostics 2022, 12, 1342. [CrossRef] [PubMed]
242. Kimoto, A.; Kadoi, Y.; Tsuruda, T.; Kim, Y.-S.; Miyoshi, M.; Nomoto, Y.; Nakata, Y.; Miyake, M.; Miyashita, K.; Shimizu, K.
Exosomes in ascites from patients with human pancreatic cancer enhance remote metastasis partially through endothelial-
mesenchymal transition. Pancreatology 2023, 23, 377–388. [CrossRef] [PubMed]
243. Li, M.; Cai, H.; Deng, R.; Cheng, J.; Shi, Y. Effects of exosomes on tumor immunomodulation and their potential clinical
applications. Int. J. Oncol. 2022, 61, 1–14. [CrossRef]
244. Kurt, F.G.O.; Lasser, S.; Arkhypov, I.; Utikal, J.; Umansky, V. Enhancing immunotherapy response in melanoma: Myeloid-derived
suppressor cells as a therapeutic target. J. Clin. Investig. 2023, 133, e170762.
245. Law, Z.-J.; Khoo, X.H.; Lim, P.T.; Goh, B.H.; Ming, L.C.; Lee, W.-L.; Goh, H.P. Extracellular vesicle-mediated chemoresistance in
oral squamous cell carcinoma. Front. Mol. Biosci. 2021, 8, 629888. [CrossRef]
246. Li, Z.; Fang, R.; Fang, J.; He, S.; Liu, T. Functional implications of Rab27 GTPases in cancer. Cell Commun. Signal. 2018, 16, 44.
[CrossRef] [PubMed]
247. ICH S7A Safety Pharmacology Studies for Human Pharmaceuticals; European Medicines Agency: London, UK, 2000.
248. FDA. Preclinical Safety Evaluation of Biotechnology-Derived Pharmaceuticals S6(R1). In Proceedings of the International Conference
on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use, Prague, Czech Republic, 31 October–1
November 2023; FDA: Silver Spring, MD, USA, 2011.
249. Committee for Human Medicinal Products (CHMP). 2009. Available online: https://www.ema.europa.eu/en/committees/
committee-medicinal-products-human-use-chmp (accessed on 25 December 2023).
250. Lener, T.; Gimona, M.; Aigner, L.; Börger, V.; Buzas, E.; Camussi, G.; Chaput, N.; Chatterjee, D.; Court, F.A.; Portillo, H.A.D.
Applying extracellular vesicles based therapeutics in clinical trials–an ISEV position paper. J. Extracell. Vesicles 2015, 4, 30087.
[CrossRef] [PubMed]
251. Decision of the Gdansk Court of Appeal 26 June 2012—Case No. I ACa 320/12. “Bolar Exemption–Poland”; Directive 2001/83/EC
of the European Parliament and of the Council of 6 November 2001 on the Community Code Relating to Medicinal Products for
Human Use. IIC Int. Rev. Intellect. Prop. Compet. Law 2013, 44, 366–368.
252. Ramati, Y. Accelerated Access of Advanced Regenerative Therapies: An Industry Perspective. Cell Gene Ther. Insights 2018, 4,
555–561. [CrossRef]
253. Al-Jipouri, A.; Almurisi, S.H.; Al-Japairai, K.; Bakar, L.M.; Doolaanea, A.A. Liposomes or extracellular vesicles: A comprehensive
comparison of both lipid bilayer vesicles for pulmonary drug delivery. Polymers 2023, 15, 318. [CrossRef] [PubMed]
254. Li, Y.; Liu, C.; Guo, N.; Cai, L.; Wang, M.; Zhu, L.; Li, F.; Jin, L.; Sui, C. Extracellular vesicles from human Fallopian tubal fluid
benefit embryo development in vitro. Hum. Reprod. Open 2023, 2023, hoad006. [CrossRef] [PubMed]
255. Billing, A.M.; Dib, S.S.; Bhagwat, A.M.; da Silva, I.T.; Drummond, R.D.; Hayat, S.; Al-Mismar, R.; Ben-Hamidane, H.; Goswami, N.;
Engholm-Keller, K. A Systems-level Characterization of the Differentiation of Human Embryonic Stem Cells into Mesenchymal
Stem Cells*[S]. Mol. Cell. Proteom. 2019, 18, 1950–1966. [CrossRef]
256. de Alcântara-Neto, A.S.; Cuello, C.; Uzbekov, R.; Bauersachs, S.; Mermillod, P.; Almiñana, C. Oviductal extracellular vesicles
enhance porcine in vitro embryo development by modulating the embryonic transcriptome. Biomolecules 2022, 12, 1300. [CrossRef]
257. Avni, D.; Avni, O. Extracellular vesicles: Schistosomal long-range precise weapon to manipulate the immune response. Front.
Cell. Infect. Microbiol. 2021, 11, 196. [CrossRef]
258. Soltani, S.; Mansouri, K.; Emami Aleagha, M.S.; Moasefi, N.; Yavari, N.; Shakouri, S.K.; Notararigo, S.; Shojaeian, A.; Pociot, F.;
Yarani, R. Extracellular vesicle therapy for type 1 diabetes. Front. Immunol. 2022, 1574, 865782. [CrossRef]
259. Bonetto, V.; Grilli, M. Neural stem cell-derived extracellular vesicles: Mini players with key roles in neurogenesis, immunomodu-
lation, neuroprotection and aging. Front. Mol. Biosci. 2023, 10, 1187263. [CrossRef] [PubMed]
260. Misawa, T.; Hitomi, K.; Miyata, K.; Tanaka, Y.; Fujii, R.; Chiba, M.; Loo, T.M.; Hanyu, A.; Kawasaki, H.; Kato, H. Identification of
Novel Senescent Markers in Small Extracellular Vesicles. Int. J. Mol. Sci. 2023, 24, 2421. [CrossRef] [PubMed]
261. Loric, S.; Denis, J.A.; Desbene, C.; Sabbah, M.; Conti, M. Extracellular Vesicles in Breast Cancer: From Biology and Function to
Clinical Diagnosis and Therapeutic Management. Int. J. Mol. Sci. 2023, 24, 7208. [CrossRef] [PubMed]
262. Gregory, C.D.; Rimmer, M.P. Extracellular vesicles arising from apoptosis: Forms, functions, and applications. J. Pathol. 2023.
[CrossRef] [PubMed]
Int. J. Mol. Sci. 2024, 25, 485 40 of 51
263. Li, P.; Hong, J.; Liang, C.; Li, Y.; Gao, L.; Wu, L.; Yao, R.; Zhang, Y. Endothelial cell-released extracellular vesicles trigger pyroptosis
and vascular inflammation to induce atherosclerosis through the delivery of HIF1A-AS2. FASEB J. 2023, 37, e22942. [CrossRef]
[PubMed]
264. Tan, S.; Yang, Y.; Yang, W.; Han, Y.; Huang, L.; Yang, R.; Hu, Z.; Tao, Y.; Liu, L.; Li, Y. Exosomal cargos-mediated metabolic
reprogramming in tumor microenvironment. J. Exp. Clin. Cancer Res. 2023, 42, 1–28. [CrossRef] [PubMed]
265. Kamerkar, S.; Leng, C.; Burenkova, O.; Jang, S.C.; McCoy, C.; Zhang, K.; Dooley, K.; Kasera, S.; Zi, T.; Sisó, S. Exosome-mediated
genetic reprogramming of tumor-associated macrophages by exoASO-STAT6 leads to potent monotherapy antitumor activity. Sci.
Adv. 2022, 8, eabj7002. [CrossRef]
266. Ma, F.; Vayalil, J.; Lee, G.; Wang, Y.; Peng, G. Emerging role of tumor-derived extracellular vesicles in T cell suppression and
dysfunction in the tumor microenvironment. J. Immunother. Cancer 2021, 9, e003217. [CrossRef]
267. Di Bella, M.A. Overview and update on extracellular vesicles: Considerations on exosomes and their application in modern
medicine. Biology 2022, 11, 804. [CrossRef]
268. Ross, T.J.; Zhang, J. The Microbiome-TIME Axis: A Host of Possibilities. Microorganisms 2023, 11, 288. [CrossRef]
269. Sanz-Ros, J.; Mas-Bargues, C.; Romero-García, N.; Huete-Acevedo, J.; Dromant, M.; Borrás, C. Extracellular vesicles as therapeutic
resources in the clinical environment. Int. J. Mol. Sci. 2023, 24, 2344. [CrossRef] [PubMed]
270. Duong, A.; Parmar, G.; Kirkham, A.M.; Burger, D.; Allan, D.S. Registered clinical trials investigating treatment with cell-derived
extracellular vesicles: A scoping review. Cytotherapy 2023, 11, 288. [CrossRef]
271. Amarasinghe, I.; Phillips, W.; Hill, A.F.; Cheng, L.; Helbig, K.J.; Willms, E.; Monson, E.A. Cellular communication through
extracellular vesicles and lipid droplets. J. Extracell. Biol. 2023, 2, e77. [CrossRef]
272. Ma, Z.-J.; Yang, J.-J.; Lu, Y.-B.; Liu, Z.-Y.; Wang, X.-X. Mesenchymal stem cell-derived exosomes: Toward cell-free therapeutic
strategies in regenerative medicine. World J. Stem Cells 2020, 12, 814. [CrossRef] [PubMed]
273. Lian, M.Q.; Chng, W.H.; Liang, J.; Yeo, H.Q.; Lee, C.K.; Belaid, M.; Tollemeto, M.; Wacker, M.G.; Czarny, B.; Pastorin, G.
Plant-derived extracellular vesicles: Recent advancements and current challenges on their use for biomedical applications. J.
Extracell. Vesicles 2022, 11, 12283. [CrossRef] [PubMed]
274. Nikfarjam, S.; Rezaie, J.; Zolbanin, N.M.; Jafari, R. Mesenchymal stem cell derived-exosomes: A modern approach in translational
medicine. J. Transl. Med. 2020, 18, 1–21. [CrossRef]
275. Huang, D.; Rao, D.; Xi, X.; Zhang, Z.; Zhong, T. Application of extracellular vesicles proteins in cancer diagnosis. Front. Cell Dev.
Biol. 2022, 10, 1007360. [CrossRef]
276. Bhatia, R.; Chang, J.; Munoz, J.L.; Walker, N.D. Forging New Therapeutic Targets: Efforts of Tumor Derived Exosomes to Prepare
the Pre-Metastatic Niche for Cancer Cell Dissemination and Dormancy. Biomedicines 2023, 11, 1614. [CrossRef]
277. Gangadaran, P.; Madhyastha, H.; Madhyastha, R.; Rajendran, R.L.; Nakajima, Y.; Watanabe, N.; Velikkakath, A.K.G.; Hong, C.M.;
Gopi, R.V.; Muthukalianan, G.K. The emerging role of exosomes in innate immunity, diagnosis and therapy. Front. Immunol. 2023,
13, 1085057. [CrossRef]
278. Fu, M.; Li, J.; Battulga, T.; Li, X.; Xu, M. Biological Functions and Applications of Exosomes in Drug Research. Int. J. Drug Discov.
Pharmacol. 2023, 2, 1–9. [CrossRef]
279. Kang, F.; Yan, Y.; Liu, Y.; Liang, Q.; Xu, Z.; Zhu, W.; Thakur, A. Unraveling the significance of exosomal circRNAs in cancer
therapeutic resistance. Front. Pharmacol. 2023, 14, 1093175. [CrossRef] [PubMed]
280. Mezzasoma, L.; Bellezza, I.; Romani, R.; Talesa, V.N. Extracellular Vesicles and the Inflammasome: An Intricate Network
Sustaining Chemoresistance. Front. Oncol. 2022, 1602, 888135. [CrossRef] [PubMed]
281. de Visser, K.E.; Joyce, J.A. The evolving tumor microenvironment: From cancer initiation to metastatic outgrowth. Cancer Cell
2023, 41, 374–403. [CrossRef] [PubMed]
282. Zhang, H.; Wang, X.; Yu, Y.; Yang, Z. Progression of Exosome-Mediated Chemotherapy Resistance in Cancer. Oncologie 2022, 24,
247–259. [CrossRef]
283. Krylova, S.V.; Feng, D. The Machinery of Exosomes: Biogenesis, Release, and Uptake. Int. J. Mol. Sci. 2023, 24, 1337. [CrossRef]
[PubMed]
284. Moloudizargari, M.; Redegeld, F.; Asghari, M.H.; Mosaffa, N.; Mortaz, E. Long-chain polyunsaturated omega-3 fatty acids reduce
multiple myeloma exosome-mediated suppression of NK cell cytotoxicity. DARU J. Pharm. Sci. 2020, 28, 647–659. [CrossRef]
285. Aslan, C.; Maralbashi, S.; Kahroba, H.; Asadi, M.; Soltani-Zangbar, M.S.; Javadian, M.; Shanehbandi, D.; Baradaran, B.; Darabi, M.;
Kazemi, T. Docosahexaenoic acid(DHA) inhibits pro-angiogenic effects of breast cancer cells via down-regulating cellular and
exosomal expression of angiogenic genes and microRNAs. Life Sci. 2020, 258, 118094. [CrossRef]
286. Ghaffari-Makhmalbaf, P.; Sayyad, M.; Pakravan, K.; Razmara, E.; Bitaraf, A.; Bakhshinejad, B.; Goudarzi, P.; Yousefi, H.;
Pournaghshband, M.; Nemati, F. Docosahexaenoic acid reverses the promoting effects of breast tumor cell-derived exosomes on
endothelial cell migration and angiogenesis. Life Sci. 2021, 264, 118719. [CrossRef]
287. Datta, A.; Kim, H.; McGee, L.; Johnson, A.E.; Talwar, S.; Marugan, J.; Southall, N.; Hu, X.; Lal, M.; Mondal, D. High-throughput
screening identified selective inhibitors of exosome biogenesis and secretion: A drug repurposing strategy for advanced cancer.
Sci. Rep. 2018, 8, 8161. [CrossRef]
288. Zhang, H.; Lu, J.; Liu, J.; Zhang, G.; Lu, A. Advances in the discovery of exosome inhibitors in cancer. J. Enzym. Inhib. Med. Chem.
2020, 35, 1322–1330. [CrossRef]
Int. J. Mol. Sci. 2024, 25, 485 41 of 51
289. Kim, J.H.; Lee, C.-H.; Baek, M.-C. Dissecting exosome inhibitors: Therapeutic insights into small-molecule chemicals against
cancer. Exp. Mol. Med. 2022, 54, 1833–1843. [CrossRef] [PubMed]
290. Datta, A.; Kim, H.; Lal, M.; McGee, L.; Johnson, A.; Moustafa, A.A.; Jones, J.C.; Mondal, D.; Ferrer, M.; Abdel-Mageed, A.B.
Manumycin A suppresses exosome biogenesis and secretion via targeted inhibition of Ras/Raf/ERK1/2 signaling and hnRNP
H1 in castration-resistant prostate cancer cells. Cancer Lett. 2017, 408, 73–81. [CrossRef] [PubMed]
291. Moloudizargari, M.; Asghari, M.H.; Mortaz, E. Inhibiting exosomal MIC-A and MIC-B shedding of cancer cells to overcome
immune escape: New insight of approved drugs. DARU J. Pharm. Sci. 2019, 27, 879–884. [CrossRef] [PubMed]
292. Morimoto, Y.; Yamashita, N.; Daimon, T.; Hirose, H.; Yamano, S.; Haratake, N.; Ishikawa, S.; Bhattacharya, A.; Fushimi, A.;
Ahmad, R. MUC1-C is a master regulator of MICA/B NKG2D ligand and exosome secretion in human cancer cells. J. Immunother.
Cancer 2023, 11, e006238. [CrossRef] [PubMed]
293. Jia, Z.; Jia, J.; Yao, L.; Li, Z. Crosstalk of exosomal non-coding RNAs in the tumor microenvironment: Novel frontiers. Front.
Immunol. 2022, 13. [CrossRef] [PubMed]
294. Yáñez-Mó, M.; Siljander, P.R.-M.; Andreu, Z.; Bedina Zavec, A.; Borràs, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho,
J. Biological properties of extracellular vesicles and their physiological functions. J. Extracell. Vesicles 2015, 4, 27066. [CrossRef]
[PubMed]
295. Edelmann, M.J.; Kima, P.E. Current understanding of extracellular vesicle homing/tropism. Zoonoses (Burlingt. Mass.) 2022, 2, 14.
[CrossRef]
296. Ragni, E.; Parolini, O.; Silini, A.R. MSC-Derived Extracellular Vesicles and Secreted Factors as “Cell-Free” Therapeutic Alternatives
in Regenerative Medicine. Front. Bioeng. Biotechnol. 2022, 10, 56. [CrossRef]
297. Yang, G.; Fan, X.; Liu, Y.; Jie, P.; Mazhar, M.; Liu, Y.; Dechsupa, N.; Wang, L. Immunomodulatory Mechanisms and Therapeutic
Potential of Mesenchymal Stem Cells. Stem Cell Rev. Rep. 2023, 19, 1214–1231. [CrossRef]
298. Kou, M.; Huang, L.; Yang, J.; Chiang, Z.; Chen, S.; Liu, J.; Guo, L.; Zhang, X.; Zhou, X.; Xu, X. Mesenchymal stem cell-derived
extracellular vesicles for immunomodulation and regeneration: A next generation therapeutic tool? Cell Death Dis. 2022, 13, 580.
[CrossRef]
299. Zhao, R.; Zhao, T.; He, Z.; Cai, R.; Pang, W. Composition, isolation, identification and function of adipose tissue-derived exosomes.
Adipocyte 2021, 10, 587–604. [CrossRef] [PubMed]
300. Crum, R.J.; Capella-Monsonís, H.; Badylak, S.F.; Hussey, G.S. Extracellular vesicles for regenerative medicine applications. Appl.
Sci. 2022, 12, 7472. [CrossRef]
301. Ju, Y.; Fang, B. Research advances on the mechanism of extracellular vesicles of adipose-derived mesenchymal stem cells in
promoting wound angiogenesis. Zhonghua Shao Shang Za Zhi = Zhonghua Shaoshang Zazhi = Chin. J. Burn. 2023, 39, 85–90.
302. van de Wakker, S.I.; Meijers, F.M.; Sluijter, J.P.G.; Vader, P. Extracellular vesicle heterogeneity and its impact for regenerative
medicine applications. Pharmacol. Rev. 2023, 75, 1043–1061. [CrossRef] [PubMed]
303. Jin, Y.; Li, S.; Yu, Q.; Chen, T.; Liu, D. Application of stem cells in regeneration medicine. MedComm 2023, 4, e291. [CrossRef]
[PubMed]
304. Namjoo, A.R.; Abrbekoh, F.N.; Saghati, S.; Amini, H.; Saadatlou, M.A.E.; Rahbarghazi, R. Tissue engineering modalities in skeletal
muscles: Focus on angiogenesis and immunomodulation properties. Stem Cell Res. Ther. 2023, 14, 1–24. [CrossRef] [PubMed]
305. Byun, S.-E.; Sim, C.; Chung, Y.; Kim, H.K.; Park, S.; Kim, D.K.; Cho, S.; Lee, S. Skeletal muscle regeneration by the exosomes of
adipose tissue-derived mesenchymal stem cells. Curr. Issues Mol. Biol. 2021, 43, 1473–1488. [CrossRef]
306. Asgarpour, K.; Shojaei, Z.; Amiri, F.; Ai, J.; Mahjoubin-Tehran, M.; Ghasemi, F.; ArefNezhad, R.; Hamblin, M.R.; Mirzaei,
H. Exosomal microRNAs derived from mesenchymal stem cells: Cell-to-cell messages. Cell Commun. Signal. 2020, 18, 1–16.
[CrossRef]
307. Chen, K.; Li, Y.; Xu, L.; Qian, Y.; Liu, N.; Zhou, C.; Liu, J.; Zhou, L.; Xu, Z.; Jia, R. Comprehensive insight into endothelial
progenitor cell-derived extracellular vesicles as a promising candidate for disease treatment. Stem Cell Res. Ther. 2022, 13, 238.
[CrossRef]
308. Yang, S.; Li, J.; Tang, M.; Gao, X.; Liu, W.; Wei, S. Mesenchymal Stem Cell-Derived Exosomes in Cardioprotection: A Novel
Application to Prevent Myocardial Injury. Rev. Cardiovasc. Med. 2022, 23, 310. [CrossRef]
309. Mao, B.; Yuan, W.; Wu, F.; Yan, Y.; Wang, B. Autophagy in hepatic ischemia–reperfusion injury. Cell Death Discov. 2023, 9, 115.
[CrossRef] [PubMed]
310. Wu, K.Y.; Ahmad, H.; Lin, G.; Carbonneau, M.; Tran, S.D. Mesenchymal Stem Cell-Derived Exosomes in Ophthalmology: A
Comprehensive Review. Pharmaceutics 2023, 15, 1167. [CrossRef] [PubMed]
311. Yang, M.; Chen, J.; Chen, L. The roles of mesenchymal stem cell-derived exosomes in diabetes mellitus and its related complica-
tions. Front. Endocrinol. 2022, 13, 1027686. [CrossRef] [PubMed]
312. Tsiapalis, D.; O’Driscoll, L. Mesenchymal stem cell derived extracellular vesicles for tissue engineering and regenerative medicine
applications. Cells 2020, 9, 991. [CrossRef] [PubMed]
313. Bari, E.; Ferrarotti, I.; Perteghella, S.; Di Silvestre, D.; Rossi, R.; Mauri, P.; Grisoli, P.; Barzon, V.; Balderacchi, A.; Corsico, A.
Mesenchymal Extracellular Vesicles as Alpha-1-antitrypsin physiological delivery systems for lung regeneration. In Proceedings
of the 19th Advanced School in Pharmaceutical Technology, Soverato, Italy, 9–12 September 2019.
Int. J. Mol. Sci. 2024, 25, 485 42 of 51
314. Zhang, L.; Song, Y.; Chen, L.; Li, D.; Feng, H.; Lu, Z.; Fan, T.; Chen, Z.; Livingston, M.J.; Geng, Q. MiR-20a-containing exosomes
from umbilical cord mesenchymal stem cells alleviates liver ischemia/reperfusion injury. J. Cell. Physiol. 2020, 235, 3698–3710.
[CrossRef] [PubMed]
315. Li, H.; Zhou, Y.; Xu, W.; Liu, J.; Wang, S.; Jiang, H. The role of autophagy in calcium oxalate kidney stone: A systematic review of
the literature. Front. Physiol. 2022, 1981. [CrossRef] [PubMed]
316. Gonzalez-Melero, L.; Hernandez, R.M.; Santos-Vizcaino, E.; Igartua, M. Tumour-derived extracellular vesicle based vaccines for
melanoma treatment. Drug Deliv. Transl. Res. 2023, 13, 1520–1542. [CrossRef] [PubMed]
317. Oliveira, M.M.; D’Aulerio, R.; Yong, T.; He, M.; Baptista, M.A.; Nylén, S.; Westerberg, L.S. Increased cross-presentation by
dendritic cells and enhanced anti-tumour therapy using the Arp2/3 inhibitor CK666. Br. J. Cancer 2023, 128, 982–991. [CrossRef]
318. Iglesias-Escudero, M.; Arias-González, N.; Martínez-Cáceres, E. Regulatory cells and the effect of cancer immunotherapy. Mol.
Cancer 2023, 22, 26. [CrossRef]
319. Tay, R.E.; Richardson, E.K.; Toh, H.C. Revisiting the role of CD4+ T cells in cancer immunotherapy—New insights into old
paradigms. Cancer Gene Ther. 2021, 28, 5–17. [CrossRef]
320. Axelrod, M.L.; Cook, R.S.; Johnson, D.B.; Balko, J.M. Biological Consequences of MHC-II Expression by Tumor Cells in
CancerBiological Consequences of MHC-II Expression by Tumor Cells. Clin. Cancer Res. 2019, 25, 2392–2402. [CrossRef]
[PubMed]
321. Xie, N.; Shen, G.; Gao, W.; Huang, Z.; Huang, C.; Fu, L. Neoantigens: Promising targets for cancer therapy. Signal Transduct.
Target. Ther. 2023, 8, 9. [CrossRef] [PubMed]
322. Stephens, A.J.; Burgess-Brown, N.A.; Jiang, S. Beyond just peptide antigens: The complex world of peptide-based cancer vaccines.
Front. Immunol. 2021, 12, 696791. [CrossRef] [PubMed]
323. Kim, C.-G.; Sang, Y.-B.; Lee, J.-H.; Chon, H.-J. Combining cancer vaccines with immunotherapy: Establishing a new immunological
approach. Int. J. Mol. Sci. 2021, 22, 8035. [CrossRef] [PubMed]
324. Liu, W.; Tang, H.; Li, L.; Wang, X.; Yu, Z.; Li, J. Peptide-based therapeutic cancer vaccine: Current trends in clinical application.
Cell Prolif. 2021, 54, e13025. [CrossRef] [PubMed]
325. Nin, D.S.; Deng, L.-W. Biology of Cancer-Testis Antigens and Their Therapeutic Implications in Cancer. Cells 2023, 12, 926.
[CrossRef] [PubMed]
326. Wolfers, J.; Lozier, A.; Raposo, G.; Regnault, A.; Théry, C.; Masurier, C.; Flament, C.; Pouzieux, S.; Faure, F.; Tursz, T. Tumor-derived
exosomes are a source of shared tumor rejection antigens for CTL cross-priming. Nat. Med. 2001, 7, 297–303. [CrossRef]
327. Naseri, M.; Bozorgmehr, M.; Zöller, M.; Ranaei Pirmardan, E.; Madjd, Z. Tumor-derived exosomes: The next generation of
promising cell-free vaccines in cancer immunotherapy. Oncoimmunology 2020, 9, 1779991. [CrossRef]
328. Raguraman, R.; Bhavsar, D.; Kim, D.; Ren, X.; Sikavitsas, V.; Munshi, A.; Ramesh, R. Tumor-targeted exosomes for delivery of
anticancer drugs. Cancer Lett. 2023, 558, 216093. [CrossRef]
329. Martínez-Santillán, A.; González-Valdez, J. Novel Technologies for Exosome and Exosome-like Nanovesicle Procurement and
Enhancement. Biomedicines 2023, 11, 1487. [CrossRef]
330. Shao, M.; Lopes, D.; Lopes, J.; Yousefiasl, S.; Macário-Soares, A.; Peixoto, D.; Ferreira-Faria, I.; Veiga, F.; Conde, J.; Huang, Y.
Exosome membrane-coated nanosystems: Exploring biomedical applications in cancer diagnosis and therapy. Matter 2023, 6,
761–799. [CrossRef]
331. Gulati, R.; Nandi, D.; Sarkar, K.; Venkataraman, P.; Ramkumar, K.; Ranjan, P.; Janardhanan, R. Exosomes as theranostic targets:
Implications for the clinical prognosis of aggressive cancers. Front. Mol. Biosci. 2022, 9, 890768. [CrossRef] [PubMed]
332. Kar, R.; Dhar, R.; Mukherjee, S.; Nag, S.; Gorai, S.; Mukerjee, N.; Mukherjee, D.; Vatsa, R.; Chandrakanth Jadhav, M.; Ghosh, A.
Exosome-Based Smart Drug Delivery Tool for Cancer Theranostics. ACS Biomater. Sci. Eng. 2023, 9, 577–594. [CrossRef] [PubMed]
333. Nikfarjam, S.; Rezaie, J.; Kashanchi, F.; Jafari, R. Dexosomes as a cell-free vaccine for cancer immunotherapy. J. Exp. Clin. Cancer
Res. 2020, 39, 1–20. [CrossRef]
334. Rädler, J.; Gupta, D.; Zickler, A.; Andaloussi, S.E. Exploiting the biogenesis of EVs for bioengineering and therapeutic cargo
loading. Mol. Ther. 2023, 31, 1231–1250. [CrossRef] [PubMed]
335. Rademacher, D.J. Potential for Therapeutic-Loaded Exosomes to Ameliorate the Pathogenic Effects of α-Synuclein in Parkinson’s
Disease. Biomedicines 2023, 11, 1187. [CrossRef] [PubMed]
336. Wu, T.-H.; Hsieh, S.-C.; Li, T.-H.; Lu, C.-H.; Liao, H.-T.; Shen, C.-Y.; Li, K.-J.; Wu, C.-H.; Kuo, Y.-M.; Tsai, C.-Y. Molecular Basis
for Paradoxical Activities of Polymorphonuclear Neutrophils in Inflammation/Anti-Inflammation, Bactericide/Autoimmunity,
Pro-Cancer/Anticancer, and Antiviral Infection/SARS-CoV-II-Induced Immunothrombotic Dysregulation. Biomedicines 2022,
10, 773. [CrossRef]
337. Zhang, M.; Hu, S.; Liu, L.; Dang, P.; Liu, Y.; Sun, Z.; Qiao, B.; Wang, C. Engineered exosomes from different sources for
cancer-targeted therapy. Signal Transduct. Target. Ther. 2023, 8, 124. [CrossRef]
338. Hussen, B.M.; Faraj, G.S.H.; Rasul, M.F.; Hidayat, H.J.; Salihi, A.; Baniahmad, A.; Taheri, M.; Ghafouri-Frad, S. Strategies to
overcome the main challenges of the use of exosomes as drug carrier for cancer therapy. Cancer Cell Int. 2022, 22, 1–23. [CrossRef]
339. Toyofuku, M.; Schild, S.; Kaparakis-Liaskos, M.; Eberl, L. Composition and functions of bacterial membrane vesicles. Nat. Rev.
Microbiol. 2023, 21, 415–430. [CrossRef]
340. Macion, A.; Wyszyńska, A.; Godlewska, R. Delivery of toxins and effectors by bacterial membrane vesicles. Toxins 2021, 13, 845.
[CrossRef] [PubMed]
Int. J. Mol. Sci. 2024, 25, 485 43 of 51
341. Yang, J.; Kim, E.K.; McDowell, A.; Kim, Y.-K. Microbe-derived extracellular vesicles as a smart drug delivery system. Transl. Clin.
Pharmacol. 2018, 26, 103. [CrossRef] [PubMed]
342. Croatti, V.; Parolin, C.; Giordani, B.; Foschi, C.; Fedi, S.; Vitali, B. Lactobacilli extracellular vesicles: Potential postbiotics to support
the vaginal microbiota homeostasis. Microb. Cell Factories 2022, 21, 1–11. [CrossRef] [PubMed]
343. Hosseini-Giv, N.; Basas, A.; Hicks, C.; El-Omar, E.; El-Assaad, F.; Hosseini-Beheshti, E. Bacterial extracellular vesicles and their
novel therapeutic applications in health and cancer. Front. Cell. Infect. Microbiol. 2022, 1661, 962216. [CrossRef] [PubMed]
344. Clegg, J.; Soldaini, E.; McLoughlin, R.M.; Rittenhouse, S.; Bagnoli, F.; Phogat, S. Staphylococcus aureus vaccine research and
development: The past, present and future, including novel therapeutic strategies. Front. Immunol. 2021, 12, 705360. [CrossRef]
[PubMed]
345. Ahlberg, E.; Al-Kaabawi, A.; Thune, R.; Simpson, M.R.; Pedersen, S.A.; Cione, E.; Jenmalm, M.C.; Tingö, L. Breast milk microRNAs:
Potential players in oral tolerance development. Front. Immunol. 2023, 14, 1154211. [CrossRef]
346. Kondracka, A.; Gil-Kulik, P.; Kondracki, B.; Fraszczak,
˛ K.; Oniszczuk, A.; Rybak-Krzyszkowska, M.; Staniczek, J.; Kwaśniewska,
A.; Kocki, J. Occurrence, Role, and Challenges of MicroRNA in Human Breast Milk: A Scoping Review. Biomedicines 2023, 11, 248.
[CrossRef]
347. Mecocci, S.; Trabalza-Marinucci, M.; Cappelli, K. Extracellular vesicles from animal milk: Great potentialities and critical issues.
Animals 2022, 12, 3231. [CrossRef]
348. Tong, L.; Hao, H.; Zhang, Z.; Lv, Y.; Liang, X.; Liu, Q.; Liu, T.; Gong, P.; Zhang, L.; Cao, F. Milk-derived extracellular vesicles
alleviate ulcerative colitis by regulating the gut immunity and reshaping the gut microbiota. Theranostics 2021, 11, 8570. [CrossRef]
349. Jiang, X.; You, L.; Zhang, Z.; Cui, X.; Zhong, H.; Sun, X.; Ji, C.; Chi, X. Biological properties of milk-derived extracellular vesicles
and their physiological functions in infant. Front. Cell Dev. Biol. 2021, 9, 693534. [CrossRef]
350. Mun, D.; Oh, S.; Kim, Y. Perspectives on bovine milk-derived extracellular vesicles for therapeutic applications in gut health. Food
Sci. Anim. Resour. 2022, 42, 197. [CrossRef] [PubMed]
351. Matic, S.; D’Souza, D.H.; Wu, T.; Pangloli, P.; Dia, V.P. Bovine milk exosomes affect proliferation and protect macrophages against
cisplatin-induced cytotoxicity. Immunol. Investig. 2020, 49, 711–725. [CrossRef] [PubMed]
352. Krupova, Z.; Leroux, C.; Péchoux, C.; Bevilacqua, C.; Martin, P. Comparison of goat and cow milk-derived extracellular vesicle
miRNomes. Sci. Data 2023, 10, 465. [CrossRef] [PubMed]
353. Badawy, A.A.; El-Magd, M.A.; AlSadrah, S.A. Therapeutic effect of camel milk and its exosomes on MCF7 cells in vitro and
in vivo. Integr. Cancer Ther. 2018, 17, 1235–1246. [CrossRef] [PubMed]
354. Woodman, P.G.; Futter, C.E. Multivesicular bodies: Co-ordinated progression to maturity. Curr. Opin. Cell Biol. 2008, 20, 408–414.
[CrossRef] [PubMed]
355. O’sullivan, M.J.; Lindsay, A.J. The endosomal recycling pathway—At the crossroads of the cell. Int. J. Mol. Sci. 2020, 21, 6074.
[CrossRef] [PubMed]
356. Kinoshita, M.; Matsumori, N. Inimitable Impacts of Ceramides on Lipid Rafts Formed in Artificial and Natural Cell Membranes.
Membranes 2022, 12, 727. [CrossRef]
357. Redpath, G.M.; Betzler, V.M.; Rossatti, P.; Rossy, J. Membrane heterogeneity controls cellular endocytic trafficking. Front. Cell Dev.
Biol. 2020, 8, 757. [CrossRef]
358. Xu, M.; Ji, J.; Jin, D.; Wu, Y.; Wu, T.; Lin, R.; Zhu, S.; Jiang, F.; Ji, Y.; Bao, B. The biogenesis and secretion of exosomes and
multivesicular bodies(MVBs): Intercellular shuttles and implications in human diseases. Genes Dis. 2022, 10, 1894–1907. [CrossRef]
359. Gurung, S.; Perocheau, D.; Touramanidou, L.; Baruteau, J. The exosome journey: From biogenesis to uptake and intracellular
signalling. Cell Commun. Signal. 2021, 19, 1–19. [CrossRef]
360. Lürick, A.; Kümmel, D.; Ungermann, C. Multisubunit tethers in membrane fusion. Curr. Biol. 2018, 28, R417–R420. [CrossRef]
[PubMed]
361. Vats, S.; Galli, T. Role of SNAREs in Unconventional Secretion—Focus on the VAMP7-Dependent Secretion. Front. Cell Dev. Biol.
2022, 10, 884020. [CrossRef] [PubMed]
362. Pilliod, J.; Desjardins, A.; Pernègre, C.; Jamann, H.; Larochelle, C.; Fon, E.A.; Leclerc, N. Clearance of intracellular tau protein
from neuronal cells via VAMP8-induced secretion. J. Biol. Chem. 2020, 295, 17827–17841. [CrossRef] [PubMed]
363. Ren, H.; Elgner, F.; Himmelsbach, K.; Akhras, S.; Jiang, B.; Medvedev, R.; Ploen, D.; Hildt, E. Identification of syntaxin 4 as an
essential factor for the hepatitis C virus life cycle. Eur. J. Cell Biol. 2017, 96, 542–552. [CrossRef]
364. Bunz, M.; Ritter, M.; Schindler, M. HCV egress–unconventional secretion of assembled viral particles. Trends Microbiol. 2022, 30,
364–378. [CrossRef]
365. Yu, Z.; Shi, M.; Stewart, T.; Fernagut, P.-O.; Huang, Y.; Tian, C.; Dehay, B.; Atik, A.; Yang, D.; De Giorgi, F. Reduced oligodendrocyte
exosome secretion in multiple system atrophy involves SNARE dysfunction. Brain 2020, 143, 1780–1797. [CrossRef]
366. Lu, Z.; Luo, W.; Ding, L.; Wang, H.; Li, Y.; wen Yang, B.; Ren, L.; Zheng, Q.; Xie, H.; Wang, R. Exosomes in Genitourinary Cancers:
Emerging Mediators of Drug Resistance and Promising Biomarkers. Int. J. Biol. Sci. 2023, 19, 167. [CrossRef]
367. Very, N.; Yazidi-Belkoura, E. Targeting O-GlcNAcylation to overcome resistance to anti-cancer therapies. Front. Oncol. 2022,
12, 960312. [CrossRef]
368. Arya, S.B.; Collie, S.P.; Parent, C.A. The ins-and-outs of exosome biogenesis, secretion, and internalization. Trends Cell Biol. 2023.
[CrossRef]
Int. J. Mol. Sci. 2024, 25, 485 44 of 51
369. Fu, X.; Song, J.; Yan, W.; Downs, B.M.; Wang, W.; Li, J. The biological function of tumor-derived extracellular vesicles on
metabolism. Cell Commun. Signal. 2023, 21, 150. [CrossRef]
370. Sun, C.; Wang, P.; Dong, W.; Liu, H.; Sun, J.; Zhao, L. LncRNA PVT1 promotes exosome secretion through YKT6, RAB7, and
VAMP3 in pancreatic cancer. Aging (Albany NY) 2020, 12, 10427. [CrossRef] [PubMed]
371. Raju, G.S.R.; Pavitra, E.; Bandaru, S.S.; Varaprasad, G.L.; Nagaraju, G.P.; Malla, R.R.; Huh, Y.S.; Han, Y.-K. HOTAIR: A potential
metastatic, drug-resistant and prognostic regulator of breast cancer. Mol. Cancer 2023, 22, 1–15. [CrossRef] [PubMed]
372. Liu, Y.-J.; Wang, C. A review of the regulatory mechanisms of extracellular vesicles-mediated intercellular communication. Cell
Commun. Signal. 2023, 21, 1–12. [CrossRef] [PubMed]
373. Schwarzenbach, H.; Gahan, P.B. MicroRNA shuttle from cell-to-cell by exosomes and its impact in cancer. Non-Coding RNA 2019,
5, 28. [CrossRef]
374. Joshi, B.S.; de Beer, M.A.; Giepmans, B.N.; Zuhorn, I.S. Endocytosis of extracellular vesicles and release of their cargo from
endosomes. ACS Nano 2020, 14, 4444–4455. [CrossRef]
375. Gonda, A.; Kabagwira, J.; Senthil, G.N.; Wall, N.R. Internalization of exosomes through receptor-mediated endocytosis. Mol.
Cancer Res. 2019, 17, 337–347. [CrossRef]
376. Zhao, M.; Nanbo, A.; Sun, L.; Lin, Z. Extracellular vesicles in Epstein-Barr virus’ life cycle and pathogenesis. Microorganisms 2019,
7, 48. [CrossRef]
377. Lischnig, A.; Bergqvist, M.; Ochiya, T.; Lässer, C. Quantitative proteomics identifies proteins enriched in large and small
extracellular vesicles. Mol. Cell. Proteom. 2022, 21, 100273. [CrossRef]
378. Sung, P.-S.; Hsieh, S.-L. C-type lectins and extracellular vesicles in virus-induced NETosis. J. Biomed. Sci. 2021, 28, 1–12. [CrossRef]
379. Somiya, M. Where does the cargo go?: Solutions to provide experimental support for the “extracellular vesicle cargo transfer
hypothesis”. J. Cell Commun. Signal. 2020, 14, 135–146. [CrossRef]
380. Ramos-Andrade, I.; Moraes, J.; Brandão-Costa, R.M.; da Silva, S.V.; de Souza, A.; da Silva, C.; Renovato-Martins, M.; Barja-
Fidalgo, C. Obese adipose tissue extracellular vesicles raise breast cancer cell malignancy. Endocr.-Relat. Cancer 2020, 27, 571–582.
[CrossRef] [PubMed]
381. O’Brien, K.; Ughetto, S.; Mahjoum, S.; Nair, A.V.; Breakefield, X.O. Uptake, functionality, and re-release of extracellular vesicle-
encapsulated cargo. Cell Rep. 2022, 39, 110651. [CrossRef] [PubMed]
382. Polanco, J.C.; Götz, J. Exosomal and vesicle-free tau seeds—Propagation and convergence in endolysosomal permeabilization.
FEBS J. 2022, 289, 6891–6907. [CrossRef] [PubMed]
383. Oshchepkova, A.; Zenkova, M.; Vlassov, V. Extracellular Vesicles for Therapeutic Nucleic Acid Delivery: Loading Strategies and
Challenges. Int. J. Mol. Sci. 2023, 24, 7287. [CrossRef] [PubMed]
384. O’Brien, K.; Breyne, K.; Ughetto, S.; Laurent, L.C.; Breakefield, X.O. RNA delivery by extracellular vesicles in mammalian cells
and its applications. Nat. Rev. Mol. Cell Biol. 2020, 21, 585–606. [CrossRef] [PubMed]
385. Ramos-Zaldívar, H.M.; Polakovicova, I.; Salas-Huenuleo, E.; Corvalán, A.H.; Kogan, M.J.; Yefi, C.P.; Andia, M.E. Extracellular
vesicles through the blood–brain barrier: A review. Fluids Barriers CNS 2022, 19, 1–15. [CrossRef] [PubMed]
386. Morad, G.; Carman, C.V.; Hagedorn, E.J.; Perlin, J.R.; Zon, L.I.; Mustafaoglu, N.; Park, T.-E.; Ingber, D.E.; Daisy, C.C.; Moses,
M.A. Tumor-derived extracellular vesicles breach the intact blood–brain barrier via transcytosis. ACS Nano 2019, 13, 13853–13865.
[CrossRef]
387. Joshi, B.S.; Zuhorn, I.S. Heparan sulfate proteoglycan-mediated dynamin-dependent transport of neural stem cell exosomes in an
in vitro blood–brain barrier model. Eur. J. Neurosci. 2021, 53, 706–719. [CrossRef]
388. Banks, W.A.; Sharma, P.; Bullock, K.M.; Hansen, K.M.; Ludwig, N.; Whiteside, T.L. Transport of extracellular vesicles across the
blood-brain barrier: Brain pharmacokinetics and effects of inflammation. Int. J. Mol. Sci. 2020, 21, 4407. [CrossRef]
389. Wang, L.; Wang, D.; Ye, Z.; Xu, J. Engineering Extracellular Vesicles as Delivery Systems in Therapeutic Applications. Adv. Sci.
2023, 10, e2300552. [CrossRef]
390. Toh, W.S.; Yarani, R.; El Andaloussi, S.; Cho, B.S.; Choi, C.; Corteling, R.; De Fougerolles, A.; Gimona, M.; Herz, J.; Khoury, M. A
report on the International Society for Cell & Gene Therapy 2022 Scientific Signature Series,“Therapeutic advances with native
and engineered human extracellular vesicles”. Cytotherapy 2023, 25, 810–814. [PubMed]
391. Bao, C.; Xiang, H.; Chen, Q.; Zhao, Y.; Gao, Q.; Huang, F.; Mao, L. A Review of Labeling Approaches Used in Small Extracellular
Vesicles Tracing and Imaging. Int. J. Nanomed. 2023, 18, 4567–4588. [CrossRef] [PubMed]
392. Verweij, F.J.; Balaj, L.; Boulanger, C.M.; Carter, D.R.; Compeer, E.B.; D’angelo, G.; El Andaloussi, S.; Goetz, J.G.; Gross, J.C.;
Hyenne, V. The power of imaging to understand extracellular vesicle biology in vivo. Nat. Methods 2021, 18, 1013–1026. [CrossRef]
[PubMed]
393. Kang, M.; Jordan, V.; Blenkiron, C.; Chamley, L.W. Biodistribution of extracellular vesicles following administration into animals:
A systematic review. J. Extracell. Vesicles 2021, 10, e12085. [CrossRef]
394. Dehghani, M.; Gulvin, S.M.; Flax, J.; Gaborski, T.R. Systematic evaluation of PKH labelling on extracellular vesicle size by
nanoparticle tracking analysis. Sci. Rep. 2020, 10, 9533. [CrossRef]
395. Cha, M.; Jeong, S.H.; Bae, S.; Park, J.H.; Baeg, Y.; Han, D.W.; Kim, S.S.; Shin, J.; Park, J.E.; Oh, S.W. Efficient Labeling of Vesicles
with Lipophilic Fluorescent Dyes via the Salt-Change Method. Anal. Chem. 2023, 95, 5843–5849. [CrossRef]
396. Ashique, S.; Anand, K. Radiolabelled Extracellular Vesicles as Imaging Modalities for Precise Targeted Drug Delivery. Pharmaceu-
tics 2023, 15, 1426. [CrossRef]
Int. J. Mol. Sci. 2024, 25, 485 45 of 51
397. Kooijmans, S.A.; Gitz-Francois, J.J.; Schiffelers, R.M.; Vader, P. Recombinant phosphatidylserine-binding nanobodies for targeting
of extracellular vesicles to tumor cells: A plug-and-play approach. Nanoscale 2018, 10, 2413–2426. [CrossRef]
398. Han, C.; Qin, G. Reporter systems for assessments of extracellular vesicle transfer. Front. Cardiovasc. Med. 2022, 9, 922420.
[CrossRef]
399. Chuo, S.T.-Y.; Chien, J.C.-Y.; Lai, C.P.-K. Imaging extracellular vesicles: Current and emerging methods. J. Biomed. Sci. 2018, 25,
1–10. [CrossRef]
400. Cilliers, C.; Liao, J.; Atangcho, L.; Thurber, G.M. Residualization rates of near-infrared dyes for the rational design of molecular
imaging agents. Mol. Imaging Biol. 2015, 17, 757–762. [CrossRef] [PubMed]
401. Varga, Z.; Gyurkó, I.; Pálóczi, K.; Buzás, E.I.; Horváth, I.; Hegedűs, N.; Máthé, D.; Szigeti, K. Radiolabeling of extracellular vesicles
with 99mTc for quantitative in vivo imaging studies. Cancer Biother. Radiopharm. 2016, 31, 168–173. [CrossRef] [PubMed]
402. Morishita, M.; Takahashi, Y.; Nishikawa, M.; Sano, K.; Kato, K.; Yamashita, T.; Imai, T.; Saji, H.; Takakura, Y. Quantitative analysis
of tissue distribution of the B16BL6-derived exosomes using a streptavidin-lactadherin fusion protein and iodine-125-labeled
biotin derivative after intravenous injection in mice. J. Pharm. Sci. 2015, 104, 705–713. [CrossRef] [PubMed]
403. Smyth, T.; Kullberg, M.; Malik, N.; Smith-Jones, P.; Graner, M.W.; Anchordoquy, T.J. Biodistribution and delivery efficiency of
unmodified tumor-derived exosomes. J. Control. Release 2015, 199, 145–155. [CrossRef] [PubMed]
404. Kutchy, N.A.; Ma, R.; Liu, Y.; Buch, S.; Hu, G. Extracellular vesicle-mediated delivery of ultrasmall superparamagnetic iron oxide
nanoparticles to mice brain. Front. Pharmacol. 2022, 13, 819516. [CrossRef] [PubMed]
405. Davidson, S.M.; Boulanger, C.M.; Aikawa, E.; Badimon, L.; Barile, L.; Binder, C.J.; Brisson, A.; Buzas, E.; Emanueli, C.; Jansen,
F. Methods for the identification and characterization of extracellular vesicles in cardiovascular studies: From exosomes to
microvesicles. Cardiovasc. Res. 2023, 119, 45–63. [CrossRef]
406. Santelices, J.; Ou, M.; Hui, W.W.; Maegawa, G.H.; Edelmann, M.J. Fluorescent labeling of small extracellular vesicles(EVs) isolated
from conditioned media. Bio-Protoc. 2022, 12, e4447. [CrossRef]
407. Kwon, Y.; Park, J. Methods to analyze extracellular vesicles at single particle level. Micro Nano Syst. Lett. 2022, 10, 14. [CrossRef]
408. Corso, G.; Heusermann, W.; Trojer, D.; Görgens, A.; Steib, E.; Voshol, J.; Graff, A.; Genoud, C.; Lee, Y.; Hean, J. Systematic charac-
terization of extracellular vesicle sorting domains and quantification at the single molecule–single vesicle level by fluorescence
correlation spectroscopy and single particle imaging. J. Extracell. Vesicles 2019, 8, 1663043. [CrossRef]
409. Silva, A.M.; Lázaro-Ibáñez, E.; Gunnarsson, A.; Dhande, A.; Daaboul, G.; Peacock, B.; Osteikoetxea, X.; Salmond, N.; Friis, K.P.;
Shatnyeva, O. Quantification of protein cargo loading into engineered extracellular vesicles at single-vesicle and single-molecule
resolution. J. Extracell. Vesicles 2021, 10, e12130. [CrossRef]
410. Li, Y.; Wu, Y.; Federzoni, E.A.; Wang, X.; Dharmawan, A.; Hu, X.; Wang, H.; Hawley, R.J.; Stevens, S.; Sykes, M. CD47 cross-
dressing by extracellular vesicles expressing CD47 inhibits phagocytosis without transmitting cell death signals. eLife 2022,
11, e73677. [CrossRef] [PubMed]
411. Cowzer, D.; Zameer, M.; Conroy, M.; Kolch, W.; Duffy, A.G. Targeting KRAS in pancreatic cancer. J. Pers. Med. 2022, 12, 1870.
[CrossRef] [PubMed]
412. Cieślik, M.; Bryniarski, K.; Nazimek, K. Biodelivery of therapeutic extracellular vesicles: Should mononuclear phagocytes always
be feared? Front. Cell Dev. Biol. 2023, 11, 1211833. [CrossRef] [PubMed]
413. Li, S.-Y.; Guo, Y.-L.; Tian, J.-W.; Zhang, H.-J.; Li, R.-F.; Gong, P.; Yu, Z.-L. Anti-Tumor Strategies by Harnessing the Phagocytosis of
Macrophages. Cancers 2023, 15, 2717. [CrossRef] [PubMed]
414. Rahimizadeh, P.; Yang, S.; Lim, S.I. Albumin: An emerging opportunity in drug delivery. Biotechnol. Bioprocess Eng. 2020, 25,
985–995. [CrossRef]
415. Liang, X.; Niu, Z.; Galli, V.; Howe, N.; Zhao, Y.; Wiklander, O.P.; Zheng, W.; Wiklander, R.J.; Corso, G.; Davies, C. Extracellular
vesicles engineered to bind albumin demonstrate extended circulation time and lymph node accumulation in mouse models. J.
Extracell. Vesicles 2022, 11, e12248. [CrossRef]
416. Suk, J.S.; Xu, Q.; Kim, N.; Hanes, J.; Ensign, L.M. PEGylation as a strategy for improving nanoparticle-based drug and gene
delivery. Adv. Drug Deliv. Rev. 2016, 99, 28–51. [CrossRef]
417. Mitchell, M.J.; Billingsley, M.M.; Haley, R.M.; Wechsler, M.E.; Peppas, N.A.; Langer, R. Engineering precision nanoparticles for
drug delivery. Nat. Rev. Drug Discov. 2021, 20, 101–124. [CrossRef]
418. Patras, L.; Ionescu, A.E.; Munteanu, C.; Hajdu, R.; Kosa, A.; Porfire, A.; Licarete, E.; Rauca, V.F.; Sesarman, A.; Luput, L. Trojan
horse treatment based on PEG-coated extracellular vesicles to deliver doxorubicin to melanoma in vitro and in vivo. Cancer Biol.
Ther. 2022, 23, 1–16. [CrossRef]
419. Gu, W.; Andrews, G.P.; Tian, Y. Recent Clinical Successes in Liposomal Nanomedicines. Int. J. Drug Discov. Pharmacol. 2023, 2,
52–59. [CrossRef]
420. Han, L.; Zhao, Z.; He, C.; Li, J.; Li, X.; Lu, M. Removing the stumbling block of exosome applications in clinical and translational
medicine: Expand production and improve accuracy. Stem Cell Res. Ther. 2023, 14, 57. [CrossRef] [PubMed]
421. Ducrot, C.; Loiseau, S.; Wong, C.; Madec, E.; Volatron, J.; Piffoux, M. Hybrid extracellular vesicles for drug delivery. Cancer Lett.
2023, 558, 216107. [CrossRef] [PubMed]
422. Large, D.E.; Abdelmessih, R.G.; Fink, E.A.; Auguste, D.T. Liposome composition in drug delivery design, synthesis, characteriza-
tion, and clinical application. Adv. Drug Deliv. Rev. 2021, 176, 113851. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2024, 25, 485 46 of 51
423. Raju, R.; Abuwatfa, W.H.; Pitt, W.G.; Husseini, G.A. Liposomes for the Treatment of Brain Cancer—A Review. Pharmaceuticals
2023, 16, 1056. [CrossRef] [PubMed]
424. Liu, P.; Chen, G.; Zhang, J. A review of liposomes as a drug delivery system: Current status of approved products, regulatory
environments, and future perspectives. Molecules 2022, 27, 1372. [CrossRef]
425. Nagy, N.A.; Castenmiller, C.; Vigario, F.L.; Sparrius, R.; van Capel, T.M.; de Haas, A.M.; van Kooyk, Y.; van Ree, R.; Tas, S.W.;
Geijtenbeek, T.B. Uptake kinetics of liposomal formulations of differing charge influences development of in vivo dendritic cell
immunotherapy. J. Pharm. Sci. 2022, 111, 1081–1091. [CrossRef] [PubMed]
426. Montizaan, D.; Yang, K.; Reker-Smit, C.; Salvati, A. Comparison of the uptake mechanisms of zwitterionic and negatively charged
liposomes by HeLa cells. Nanomed. Nanotechnol. Biol. Med. 2020, 30, 102300. [CrossRef]
427. Jyothi, V.G.S.; Bulusu, R.; Rao, B.V.K.; Pranothi, M.; Banda, S.; Bolla, P.K.; Kommineni, N. Stability characterization for
pharmaceutical liposome product development with focus on regulatory considerations: An update. Int. J. Pharm. 2022,
624, 122022.
428. Zhao, Y.; Zheng, Y.; Zhu, Y.; Li, H.; Zhu, H.; Liu, T. Docetaxel-loaded M1 macrophage-derived exosomes for a safe and efficient
chemoimmunotherapy of breast cancer. J. Nanobiotechnol. 2022, 20, 1–12. [CrossRef]
429. Ilahibaks, N.F.; Ardisasmita, A.I.; Xie, S.; Gunnarsson, A.; Brealey, J.; Vader, P.; de Jong, O.G.; de Jager, S.; Dekker, N.; Peacock, B.
TOP-EVs: Technology of Protein delivery through Extracellular Vesicles is a versatile platform for intracellular protein delivery. J.
Control. Release 2023, 355, 579–592. [CrossRef]
430. Hade, M.D.; Suire, C.N.; Suo, Z. An Effective Peptide-Based Platform for Efficient Exosomal Loading and Cellular Delivery of a
microRNA. ACS Appl. Mater. Interfaces 2023, 15, 3851–3866. [CrossRef] [PubMed]
431. Cao, T.G.N.; Kang, J.H.; Kim, W.; Lim, J.; Kang, S.J.; You, J.Y.; Hoang, Q.T.; Kim, W.J.; Rhee, W.J.; Kim, C. Engineered extracellular
vesicle-based sonotheranostics for dual stimuli-sensitive drug release and photoacoustic imaging-guided chemo-sonodynamic
cancer therapy. Theranostics 2022, 12, 1247.
432. Zhou, X.; Miao, Y.; Wang, Y.; He, S.; Guo, L.; Mao, J.; Chen, M.; Yang, Y.; Zhang, X.; Gan, Y. Tumour-derived extracellular vesicle
membrane hybrid lipid nanovesicles enhance siRNA delivery by tumour-homing and intracellular freeway transportation. J.
Extracell. Vesicles 2022, 11, e12198. [CrossRef] [PubMed]
433. Xu, X.; Xu, L.; Wen, C.; Xia, J.; Zhang, Y.; Liang, Y. Programming assembly of biomimetic exosomes: An emerging theranostic
nanomedicine platform. Mater. Today Bio 2023, 22, 100760. [CrossRef] [PubMed]
434. Filipe, L.; de Sousa, T.; Silva, D.; Santos, M.M.; Ribeiro Carrott, M.; Poeta, P.; Branco, L.C.; Gago, S. In Vitro Antimicrobial Studies
of Mesoporous Silica Nanoparticles Comprising Anionic Ciprofloxacin Ionic Liquids and Organic Salts. Pharmaceutics 2023,
15, 1934. [CrossRef] [PubMed]
435. Sancho-Albero, M.; Martín-Pardillos, A.; Lujan, L.; Sebastian, V.; Santamaria, J.; Martín-Duque, P. Exosomes loaded with
ultrasmall Pt nanoparticles: A novel low-toxicity alternative to cisplatin. J. Nanobiotechnol. 2022, 20, 473. [CrossRef] [PubMed]
436. Tang, J.; Rakshit, M.; Chua, H.M.; Darwitan, A.; Nguyen, L.T.; Muktabar, A.; Venkatraman, S.; Ng, K.W. Liposome interaction with
macrophages and foam cells for atherosclerosis treatment: Effects of size, surface charge and lipid composition. Nanotechnology
2021, 32, 505105. [CrossRef]
437. Badran, M.M.; Alouny, N.N.; Aldosari, B.N.; Alhusaini, A.M.; Abou El Ela, A.E.S. Transdermal Glipizide Delivery System Based
on Chitosan-Coated Deformable Liposomes: Development, Ex Vivo, and In Vivo Studies. Pharmaceutics 2022, 14, 826. [CrossRef]
438. Mejia, F.; Khan, S.; Omstead, D.T.; Minetos, C.; Bilgicer, B. Identification and optimization of tunable endosomal escape parameters
for enhanced efficacy in peptide-targeted prodrug-loaded nanoparticles. Nanoscale 2022, 14, 1226–1240. [CrossRef]
439. Manoochehri, H.; Jalali, A.; Tanzadehpanah, H.; Taherkhani, A.; Najafi, R. Aptamer-conjugated nanoliposomes containing
COL1A1 siRNA sensitize CRC cells to conventional chemotherapeutic drugs. Colloids Surf. B Biointerfaces 2022, 218, 112714.
[CrossRef]
440. Koga, K.; Tagami, T.; Ozeki, T. Gold nanoparticle-coated thermosensitive liposomes for the triggered release of doxorubicin, and
photothermal therapy using a near-infrared laser. Colloids Surf. A Physicochem. Eng. Asp. 2021, 626, 127038. [CrossRef]
441. Li, R.-T.; Zhu, Y.-D.; Li, W.-Y.; Hou, Y.-K.; Zou, Y.-M.; Zhao, Y.-H.; Zou, Q.; Zhang, W.-H.; Chen, J.-X. Synergistic photothermal-
photodynamic-chemotherapy toward breast cancer based on a liposome-coated core–shell AuNS@ NMOFs nanocomposite
encapsulated with gambogic acid. J. Nanobiotechnol. 2022, 20, 1–22. [CrossRef] [PubMed]
442. Liu, L.; Pan, D.; Chen, S.; Martikainen, M.-V.; Kårlund, A.; Ke, J.; Pulkkinen, H.; Ruhanen, H.; Roponen, M.; Käkelä, R. Systematic
design of cell membrane coating to improve tumor targeting of nanoparticles. Nat. Commun. 2022, 13, 6181. [CrossRef] [PubMed]
443. Chan, M.-H.; Chang, Z.-X.; Huang, C.-Y.F.; Lee, L.J.; Liu, R.-S.; Hsiao, M. Integrated therapy platform of exosomal system: Hybrid
inorganic/organic nanoparticles with exosomes for cancer treatment. Nanoscale Horiz. 2022, 7, 352–367. [CrossRef] [PubMed]
444. Xing, Z.; Zhao, C.; Wu, S.; Yang, D.; Zhang, C.; Wei, X.; Wei, X.; Su, H.; Liu, H.; Fan, Y. Hydrogel Loaded with VEGF/TFEB-
Engineered Extracellular Vesicles for Rescuing Critical Limb Ischemia by a Dual-Pathway Activation Strategy. Adv. Healthc. Mater.
2022, 11, 2100334. [CrossRef] [PubMed]
445. Zheng, W.; He, R.; Liang, X.; Roudi, S.; Bost, J.; Coly, P.M.; van Niel, G.; Andaloussi, S.E. Cell-specific targeting of extracellular
vesicles though engineering the glycocalyx. J. Extracell. Vesicles 2022, 11, 12290. [CrossRef]
446. Richter, M.; Vader, P.; Fuhrmann, G. Approaches to surface engineering of extracellular vesicles. Adv. Drug Deliv. Rev. 2021, 173,
416–426. [CrossRef]
Int. J. Mol. Sci. 2024, 25, 485 47 of 51
447. Huang, H.; Yi, X.; Wei, Q.; Li, M.; Cai, X.; Lv, Y.; Weng, L.; Mao, Y.; Fan, W.; Zhao, M. Edible and cation-free kiwi fruit derived
vesicles mediated EGFR-targeted siRNA delivery to inhibit multidrug resistant lung cancer. J. Nanobiotechnol. 2023, 21, 1–14.
[CrossRef]
448. Villa, A.; Garofalo, M.; Crescenti, D.; Rizzi, N.; Brunialti, E.; Vingiani, A.; Belotti, P.; Sposito, C.; Franzè, S.; Cilurzo, F. Transplanta-
tion of autologous extracellular vesicles for cancer-specific targeting. Theranostics 2021, 11, 2034. [CrossRef]
449. Lázaro-Ibáñez, E.; Faruqu, F.N.; Saleh, A.F.; Silva, A.M.; Tzu-Wen Wang, J.; Rak, J.; Al-Jamal, K.T.; Dekker, N. Selection of
fluorescent, bioluminescent, and radioactive tracers to accurately reflect extracellular vesicle biodistribution in vivo. ACS Nano
2021, 15, 3212–3227. [CrossRef]
450. Skotland, T.; Iversen, T.G.; Llorente, A.; Sandvig, K. Biodistribution, pharmacokinetics and excretion studies of intravenously
injected nanoparticles and extracellular vesicles: Possibilities and challenges. Adv. Drug Deliv. Rev. 2022, 186, 114326. [CrossRef]
[PubMed]
451. Haney, M.J.; Klyachko, N.L.; Harrison, E.B.; Zhao, Y.; Kabanov, A.V.; Batrakova, E.V. TPP1 delivery to lysosomes with extracellular
vesicles and their enhanced brain distribution in the animal model of batten disease. Adv. Healthc. Mater. 2019, 8, 1801271.
[CrossRef] [PubMed]
452. Zahednezhad, F.; Saadat, M.; Valizadeh, H.; Zakeri-Milani, P.; Baradaran, B. Liposome and immune system interplay: Challenges
and potentials. J. Control. Release 2019, 305, 194–209. [CrossRef] [PubMed]
453. Fu, Y.; Saraswat, A.; Vartak, R.; Patki, M.; Patel, K. Liposomal formulation: Opportunities, challenges, and industrial applicability.
In Multifunctional Nanocarriers; Elsevier: Amsterdam, The Netherlands, 2022; pp. 79–102.
454. Jensen, G.M.; Hodgson, D.F. Opportunities and challenges in commercial pharmaceutical liposome applications. Adv. Drug Deliv.
Rev. 2020, 154, 2–12. [CrossRef] [PubMed]
455. He, H.; Yuan, D.; Wu, Y.; Cao, Y. Pharmacokinetics and pharmacodynamics modeling and simulation systems to support the
development and regulation of liposomal drugs. Pharmaceutics 2019, 11, 110. [CrossRef] [PubMed]
456. He, H.; Lu, Y.; Qi, J.; Zhao, W.; Dong, X.; Wu, W. Biomimetic thiamine-and niacin-decorated liposomes for enhanced oral delivery
of insulin. Acta Pharm. Sin. B 2018, 8, 97–105. [CrossRef]
457. Arrieta-Molero, J.F.; Aleck, K.; Sinha, M.K.; Brownscheidle, C.M.; Shapiro, L.J.; Sperling, M.A. Orally Administered Liposome-
Entrapped Insulin in Diabetic Animals: A Critical Assessment. Horm. Res. Paediatr. 1982, 16, 249–256. [CrossRef]
458. Song, Z.; Lin, Y.; Zhang, X.; Feng, C.; Lu, Y.; Gao, Y.; Dong, C. Cyclic RGD peptide-modified liposomal drug delivery system
for targeted oral apatinib administration: Enhanced cellular uptake and improved therapeutic effects. Int. J. Nanomed. 2017, 12,
1941–1958. [CrossRef]
459. Liu, Y.; Luo, X.; Xu, X.; Gao, N.; Liu, X. Preparation, characterization and in vivo pharmacokinetic study of PVP-modified
oleanolic acid liposomes. Int. J. Pharm. 2017, 517, 1–7. [CrossRef]
460. Chen, W.-l.; Yuan, Z.-Q.; Liu, Y.; Yang, S.-d.; Zhang, C.-g.; Li, J.-z.; Zhu, W.-j.; Li, F.; Zhou, X.-f.; Lin, Y.-m. Liposomes coated with
N-trimethyl chitosan to improve the absorption of harmine in vivo and in vitro. Int. J. Nanomed. 2016, 11, 325–336.
461. Jensen, S.M.; Christensen, C.J.; Petersen, J.M.; Treusch, A.H.; Brandl, M. Liposomes containing lipids from Sulfolobus islandicus
withstand intestinal bile salts: An approach for oral drug delivery? Int. J. Pharm. 2015, 493, 63–69. [CrossRef] [PubMed]
462. Zhang, Y.; Xiong, G.M.; Ali, Y.; Boehm, B.O.; Huang, Y.Y.; Venkatraman, S. Layer-by-layer coated nanoliposomes for oral delivery
of insulin. Nanoscale 2021, 13, 776–789. [CrossRef] [PubMed]
463. Wu, H.; Nan, J.; Yang, L.; Park, H.J.; Li, J. Insulin-loaded liposomes packaged in alginate hydrogels promote the oral bioavailability
of insulin. J. Control. Release 2023, 353, 51–62. [CrossRef] [PubMed]
464. Tome-Carneiro, J.; Fernandez-Alonso, N.; Tomas-Zapico, C.; Visioli, F.; Iglesias-Gutierrez, E.; Davalos, A. Breast milk microRNAs
harsh journey towards potential effects in infant development and maturation. Lipid encapsulation can help. Pharmacol. Res.
2018, 132, 21–32. [CrossRef] [PubMed]
465. Ghiasi, M.R.; Rahimi, E.; Amirkhani, Z.; Salehi, R. Leucine-rich repeat-containing G-protein coupled receptor 5 gene overexpres-
sion of the rat small intestinal progenitor cells in response to orally administered grape exosome-like nanovesicles. Adv. Biomed.
Res. 2018, 7, 125.
466. López de las Hazas, M.-C.; del Pozo-Acebo, L.; Hansen, M.S.; Gil-Zamorano, J.; Mantilla-Escalante, D.C.; Gómez-Coronado,
D.; Marín, F.; Garcia-Ruiz, A.; Rasmussen, J.T.; Dávalos, A. Dietary bovine milk miRNAs transported in extracellular vesicles
are partially stable during GI digestion, are bioavailable and reach target tissues but need a minimum dose to impact on gene
expression. Eur. J. Nutr. 2022, 61, 1043–1056. [CrossRef] [PubMed]
467. Samuel, M.; Fonseka, P.; Sanwlani, R.; Gangoda, L.; Chee, S.H.; Keerthikumar, S.; Spurling, A.; Chitti, S.V.; Zanker, D.; Ang, C.-S.
Oral administration of bovine milk-derived extracellular vesicles induces senescence in the primary tumor but accelerates cancer
metastasis. Nat. Commun. 2021, 12, 3950. [CrossRef]
468. Betker, J.L.; Angle, B.M.; Graner, M.W.; Anchordoquy, T.J. The potential of exosomes from cow milk for oral delivery. J. Pharm. Sci.
2019, 108, 1496–1505. [CrossRef]
469. Charoenviriyakul, C.; Takahashi, Y.; Morishita, M.; Nishikawa, M.; Takakura, Y. Role of extracellular vesicle surface proteins in
the pharmacokinetics of extracellular vesicles. Mol. Pharm. 2018, 15, 1073–1080. [CrossRef]
470. Nolte, M.A.; Nolte, E.N.; Margadant, C. Integrins control vesicular trafficking; new tricks for old dogs. Trends Biochem. Sci. 2021,
46, 124–137. [CrossRef]
Int. J. Mol. Sci. 2024, 25, 485 48 of 51
471. Manca, S.; Upadhyaya, B.; Mutai, E.; Desaulniers, A.T.; Cederberg, R.A.; White, B.R.; Zempleni, J. Milk exosomes are bioavailable
and distinct microRNA cargos have unique tissue distribution patterns. Sci. Rep. 2018, 8, 11321. [CrossRef] [PubMed]
472. Akuma, P.; Okagu, O.D.; Udenigwe, C.C. Naturally occurring exosome vesicles as potential delivery vehicle for bioactive
compounds. Front. Sustain. Food Syst. 2019, 3, 23. [CrossRef]
473. Liang, G.; Zhu, Y.; Sun, B.; Shao, Y.; Jing, A.; Wang, J.; Xiao, Z. Assessing the survival of exogenous plant microRNA in mice. Food
Sci. Nutr. 2014, 2, 380–388. [CrossRef] [PubMed]
474. Din, F.U.; Aman, W.; Ullah, I.; Qureshi, O.S.; Mustapha, O.; Shafique, S.; Zeb, A. Effective use of nanocarriers as drug delivery
systems for the treatment of selected tumors. Int. J. Nanomed. 2017, 12, 7291–7309. [CrossRef] [PubMed]
475. Hua, S. Advances in oral drug delivery for regional targeting in the gastrointestinal tract-influence of physiological, pathophysio-
logical and pharmaceutical factors. Front. Pharmacol. 2020, 11, 524. [CrossRef] [PubMed]
476. Chen, M.-C.; Mi, F.-L.; Liao, Z.-X.; Hsiao, C.-W.; Sonaje, K.; Chung, M.-F.; Hsu, L.-W.; Sung, H.-W. Recent advances in chitosan-
based nanoparticles for oral delivery of macromolecules. Adv. Drug Deliv. Rev. 2013, 65, 865–879. [CrossRef] [PubMed]
477. Mikušová, V.; Mikuš, P. Advances in chitosan-based nanoparticles for drug delivery. Int. J. Mol. Sci. 2021, 22, 9652. [CrossRef]
478. Tulkens, J.; Vergauwen, G.; Van Deun, J.; Geeurickx, E.; Dhondt, B.; Lippens, L.; De Scheerder, M.-A.; Miinalainen, I.; Rappu,
P.; De Geest, B.G. Increased levels of systemic LPS-positive bacterial extracellular vesicles in patients with intestinal barrier
dysfunction. Gut 2020, 69, 191–193. [CrossRef]
479. Wu, L.; Wang, L.; Liu, X.; Bai, Y.; Wu, R.; Li, X.; Mao, Y.; Zhang, L.; Zheng, Y.; Gong, T. Milk-derived exosomes exhibit versatile
effects for improved oral drug delivery. Acta Pharm. Sin. B 2022, 12, 2029–2042. [CrossRef]
480. Parada, N.; Romero-Trujillo, A.; Georges, N.; Alcayaga-Miranda, F. Camouflage strategies for therapeutic exosomes evasion from
phagocytosis. J. Adv. Res. 2021, 31, 61–74. [CrossRef]
481. Munagala, R.; Aqil, F.; Jeyabalan, J.; Gupta, R.C. Bovine milk-derived exosomes for drug delivery. Cancer Lett. 2016, 371, 48–61.
[CrossRef] [PubMed]
482. Agrawal, A.K.; Aqil, F.; Jeyabalan, J.; Spencer, W.A.; Beck, J.; Gachuki, B.W.; Alhakeem, S.S.; Oben, K.; Munagala, R.; Bondada, S.
Milk-derived exosomes for oral delivery of paclitaxel. Nanomed. Nanotechnol. Biol. Med. 2017, 13, 1627–1636. [CrossRef] [PubMed]
483. Kim, M.S.; Haney, M.J.; Zhao, Y.; Mahajan, V.; Deygen, I.; Klyachko, N.L.; Inskoe, E.; Piroyan, A.; Sokolsky, M.; Okolie, O.
Development of exosome-encapsulated paclitaxel to overcome MDR in cancer cells. Nanomed. Nanotechnol. Biol. Med. 2016, 12,
655–664. [CrossRef] [PubMed]
484. Bardonnet, P.; Faivre, V.; Pugh, W.; Piffaretti, J.; Falson, F. Gastroretentive dosage forms: Overview and special case of Helicobacter
pylori. J. Control. Release 2006, 111, 1–18. [CrossRef]
485. Aminzadeh, M.A.; Fournier, M.; Akhmerov, A.; Jones-Ungerleider, K.C.; Valle, J.B.; Marbán, E. Casein-enhanced uptake and
disease-modifying bioactivity of ingested extracellular vesicles. J. Extracell. Vesicles 2021, 10, e12045. [CrossRef]
486. Warren, M.R.; Zhang, C.; Vedadghavami, A.; Bokvist, K.; Dhal, P.K.; Bajpayee, A.G. Milk exosomes with enhanced mucus
penetrability for oral delivery of siRNA. Biomater. Sci. 2021, 9, 4260–4277. [CrossRef]
487. Gorzelanny, C.; Mess, C.; Schneider, S.W.; Huck, V.; Brandner, J.M. Skin barriers in dermal drug delivery: Which barriers have to
be overcome and how can we measure them? Pharmaceutics 2020, 12, 684. [CrossRef]
488. Chacko, I.A.; Ghate, V.M.; Dsouza, L.; Lewis, S.A. Lipid vesicles: A versatile drug delivery platform for dermal and transdermal
applications. Colloids Surf. B: Biointerfaces 2020, 195, 111262. [CrossRef]
489. Kim, H.; Lee, J.W.; Han, G.; Kim, K.; Yang, Y.; Kim, S.H. Extracellular vesicles as potential theranostic platforms for skin diseases
and aging. Pharmaceutics 2021, 13, 760. [CrossRef]
490. Lu, K.-J.; Wang, W.; Xu, X.-L.; Jin, F.-Y.; Qi, J.; Wang, X.-J.; Kang, X.-Q.; Zhu, M.-L.; Huang, Q.-L.; Yu, C.-H. A dual deformable
liposomal ointment functionalized with retinoic acid and epidermal growth factor for enhanced burn wound healing therapy.
Biomater. Sci. 2019, 7, 2372–2382. [CrossRef]
491. Sakdiset, P.; Okada, A.; Todo, H.; Sugibayashi, K. Selection of phospholipids to design liposome preparations with high skin
penetration-enhancing effects. J. Drug Deliv. Sci. Technol. 2018, 44, 58–64. [CrossRef]
492. Kim, Y.-J.; mi Yoo, S.; Park, H.H.; Lim, H.J.; Kim, Y.-L.; Lee, S.; Seo, K.-W.; Kang, K.-S. Exosomes derived from human umbilical
cord blood mesenchymal stem cells stimulates rejuvenation of human skin. Biochem. Biophys. Res. Commun. 2017, 493, 1102–1108.
[CrossRef] [PubMed]
493. Zhang, B.; Lai, R.C.; Sim, W.K.; Choo, A.B.H.; Lane, E.B.; Lim, S.K. Topical application of mesenchymal stem cell exosomes
alleviates the imiquimod induced psoriasis-like inflammation. Int. J. Mol. Sci. 2021, 22, 720.
494. Zhang, K.; Yu, L.; Li, F.-R.; Li, X.; Wang, Z.; Zou, X.; Zhang, C.; Lv, K.; Zhou, B.; Mitragotri, S. Topical application of exosomes
derived from human umbilical cord mesenchymal stem cells in combination with sponge spicules for treatment of photoaging.
Int. J. Nanomed. 2020, 15, 2859–2872. [CrossRef] [PubMed]
495. Jun, S.; Lee, J.; Kim, S.; Choi, C.; Park, T.; Jung, H.; Cho, J.; Kim, S.; Lee, J. Staphylococcus aureus-derived membrane vesicles
exacerbate skin inflammation in atopic dermatitis. Clin. Exp. Allergy 2017, 47, 85–96. [CrossRef] [PubMed]
496. Zhang, Y.-J.; Han, Y.; Sun, Y.-Z.; Jiang, H.-H.; Liu, M.; Qi, R.-Q.; Gao, X.-H. Extracellular vesicles derived from Malassezia furfur
stimulate IL-6 production in keratinocytes as demonstrated in in vitro and in vivo models. J. Dermatol. Sci. 2019, 93, 168–175.
[CrossRef]
497. Gu, T.-W.; Wang, M.-Z.; Niu, J.; Chu, Y.; Guo, K.-R.; Peng, L.-H. Outer membrane vesicles derived from E. coli as novel vehicles
for transdermal and tumor targeting delivery. Nanoscale 2020, 12, 18965–18977. [CrossRef]
Int. J. Mol. Sci. 2024, 25, 485 49 of 51
498. Peng, L.-H.; Wang, M.-Z.; Chu, Y.; Zhang, L.; Niu, J.; Shao, H.-T.; Yuan, T.-J.; Jiang, Z.-H.; Gao, J.-Q.; Ning, X.-H. Engineering
bacterial outer membrane vesicles as transdermal nanoplatforms for photo-TRAIL–programmed therapy against melanoma. Sci.
Adv. 2020, 6, eaba2735. [CrossRef]
499. Wen, H.; Jung, H.; Li, X. Drug delivery approaches in addressing clinical pharmacology-related issues: Opportunities and
challenges. AAPS J. 2015, 17, 1327–1340. [CrossRef]
500. Lombardo, D.; Kiselev, M.A.; Caccamo, M.T. Smart nanoparticles for drug delivery application: Development of versatile
nanocarrier platforms in biotechnology and nanomedicine. J. Nanomater. 2019, 2019, e3702518. [CrossRef]
501. Balouch, M.; Storchmannová, K.I.; Štěpánek, F.E.; Berka, K. Computational Prodrug Design Methodology for Liposome Formula-
bility Enhancement of Small-Molecule APIs. Mol. Pharm. 2023, 20, 2119–2127. [CrossRef] [PubMed]
502. de Jong, O.G.; Kooijmans, S.A.; Murphy, D.E.; Jiang, L.; Evers, M.J.; Sluijter, J.P.; Vader, P.; Schiffelers, R.M. Drug delivery with
extracellular vesicles: From imagination to innovation. Acc. Chem. Res. 2019, 52, 1761–1770. [CrossRef] [PubMed]
503. Rayamajhi, S.; Nguyen, T.D.T.; Marasini, R.; Aryal, S. Macrophage-derived exosome-mimetic hybrid vesicles for tumor targeted
drug delivery. Acta Biomater. 2019, 94, 482–494. [CrossRef] [PubMed]
504. Du, S.; Guan, Y.; Xie, A.; Yan, Z.; Gao, S.; Li, W.; Rao, L.; Chen, X.; Chen, T. Extracellular vesicles: A rising star for therapeutics
and drug delivery. J. Nanobiotechnol. 2023, 21, 231. [CrossRef] [PubMed]
505. Resnier, P.; Mottais, A.; Sibiril, Y.; Le Gall, T.; Montier, T. Challenges and successes using nanomedicines for aerosol delivery to
the airways. Curr. Gene Ther. 2016, 16, 34–46. [CrossRef]
506. Anderson, C.F.; Grimmett, M.E.; Domalewski, C.J.; Cui, H. Inhalable nanotherapeutics to improve treatment efficacy for common
lung diseases. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2020, 12, e1586. [CrossRef]
507. Wang, W.; Huang, Z.; Huang, Y.; Zhang, X.; Huang, J.; Cui, Y.; Yue, X.; Ma, C.; Fu, F.; Wang, W. Pulmonary delivery nanomedicines
towards circumventing physiological barriers: Strategies and characterization approaches. Adv. Drug Deliv. Rev. 2022, 185, 114309.
[CrossRef]
508. Rogers, T.D.; Ostrowski, L.E.; Livraghi-Butrico, A.; Button, B.; Grubb, B.R. Mucociliary clearance in mice measured by tracking
trans-tracheal fluorescence of nasally aerosolized beads. Sci. Rep. 2018, 8, 14744. [CrossRef]
509. Zhang, N.-N.; Li, X.-F.; Deng, Y.-Q.; Zhao, H.; Huang, Y.-J.; Yang, G.; Huang, W.-J.; Gao, P.; Zhou, C.; Zhang, R.-R. A thermostable
mRNA vaccine against COVID-19. Cell 2020, 182, 1271–1283.e16. [CrossRef]
510. Corbett, K.S.; Edwards, D.K.; Leist, S.R.; Abiona, O.M.; Boyoglu-Barnum, S.; Gillespie, R.A.; Himansu, S.; Schäfer, A.; Ziwawo,
C.T.; DiPiazza, A.T. SARS-CoV-2 mRNA vaccine design enabled by prototype pathogen preparedness. Nature 2020, 586, 567–571.
[CrossRef]
511. Polack, F.P.; Thomas, S.J.; Kitchin, N.; Absalon, J.; Gurtman, A.; Lockhart, S.; Perez, J.L.; Pérez Marc, G.; Moreira, E.D.; Zerbini, C.
Safety and efficacy of the BNT162b2 mRNA Covid-19 vaccine. New Engl. J. Med. 2020, 383, 2603–2615. [CrossRef] [PubMed]
512. Walsh, E.E.; Frenck Jr, R.W.; Falsey, A.R.; Kitchin, N.; Absalon, J.; Gurtman, A.; Lockhart, S.; Neuzil, K.; Mulligan, M.J.; Bailey, R.
Safety and immunogenicity of two RNA-based Covid-19 vaccine candidates. New Engl. J. Med. 2020, 383, 2439–2450. [CrossRef]
[PubMed]
513. Chang, R.Y.K.; Chan, H.-K. Lipid nanoparticles for the inhalation of mRNA. Nat. Biomed. Eng. 2021, 5, 949–950. [CrossRef]
[PubMed]
514. Li, Z.; Wang, Z.; Dinh, P.-U.C.; Zhu, D.; Popowski, K.D.; Lutz, H.; Hu, S.; Lewis, M.G.; Cook, A.; Andersen, H. Cell-mimicking
nanodecoys neutralize SARS-CoV-2 and mitigate lung injury in a non-human primate model of COVID-19. Nat. Nanotechnol.
2021, 16, 942–951. [CrossRef] [PubMed]
515. Popowski, K.D.; de Juan Abad, B.L.; George, A.; Silkstone, D.; Belcher, E.; Chung, J.; Ghodsi, A.; Lutz, H.; Davenport, J.; Flanagan,
M. Inhalable exosomes outperform liposomes as mRNA and protein drug carriers to the lung. Extracell. Vesicle 2022, 1, 100002.
[CrossRef] [PubMed]
516. Wang, Z.; Popowski, K.D.; Zhu, D.; de Juan Abad, B.L.; Wang, X.; Liu, M.; Lutz, H.; De Naeyer, N.; DeMarco, C.T.; Denny, T.N.
Exosomes decorated with a recombinant SARS-CoV-2 receptor-binding domain as an inhalable COVID-19 vaccine. Nat. Biomed.
Eng. 2022, 6, 791–805. [CrossRef] [PubMed]
517. Dinh, P.-U.C.; Paudel, D.; Brochu, H.; Popowski, K.D.; Gracieux, M.C.; Cores, J.; Huang, K.; Hensley, M.T.; Harrell, E.; Vandergriff,
A.C. Inhalation of lung spheroid cell secretome and exosomes promotes lung repair in pulmonary fibrosis. Nat. Commun. 2020,
11, 1064. [CrossRef] [PubMed]
518. Popowski, K.D.; Moatti, A.; Scull, G.; Silkstone, D.; Lutz, H.; de Juan Abad, B.L.; George, A.; Belcher, E.; Zhu, D.; Mei, X. Inhalable
dry powder mRNA vaccines based on extracellular vesicles. Matter 2022, 5, 2960–2974. [CrossRef]
519. Cober, N.D.; Rowe, K.; Deng, Y.; Benavente-Babace, A.; Courtman, D.W.; Godin, M.; Stewart, D.J. Targeting extracellular vesicle
delivery to the lungs by microgel encapsulation. J. Extracell. Biol. 2023, 2, e94. [CrossRef]
520. Kwak, G.; Gololobova, O.; Sharma, N.; Caine, C.; Mazur, M.; Mulka, K.; West, N.E.; Solomon, G.M.; Cutting, G.R.; Witwer,
K.W. Extracellular vesicles enhance pulmonary transduction of stably associated adeno-associated virus following intratracheal
administration. J. Extracell. Vesicles 2023, 12, 12324. [CrossRef]
521. Silva, A.K.; Morille, M.; Piffoux, M.; Arumugam, S.; Mauduit, P.; Larghero, J.; Bianchi, A.; Aubertin, K.; Blanc-Brude, O.; Noël, D.
Development of extracellular vesicle-based medicinal products: A position paper of the group “Extracellular Vesicle translatiOn
to clinicaL perspectiVEs–EVOLVE France”. Adv. Drug Deliv. Rev. 2021, 179, 114001. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2024, 25, 485 50 of 51
522. Ju, Y.; Hu, Y.; Yang, P.; Xie, X.; Fang, B. Extracellular vesicle-loaded hydrogels for tissue repair and regeneration. Mater. Today Bio
2022, 18, 100522. [CrossRef] [PubMed]
523. Zhou, W.; Zhao, L.; Mao, Z.; Wang, Z.; Zhang, Z.; Li, M. Bidirectional Communication Between the Brain and Other Organs: The
Role of Extracellular Vesicles. Cell. Mol. Neurobiol. 2023, 43, 2675–2696. [CrossRef] [PubMed]
524. Betzer, O.; Perets, N.; Angel, A.; Motiei, M.; Sadan, T.; Yadid, G.; Offen, D.; Popovtzer, R. In vivo neuroimaging of exosomes using
gold nanoparticles. ACS Nano 2017, 11, 10883–10893. [CrossRef] [PubMed]
525. Han, Y.; Zhu, Y.; Youngblood, H.A.; Almuntashiri, S.; Jones, T.W.; Wang, X.; Liu, Y.; Somanath, P.R.; Zhang, D. Nebulization
of extracellular vesicles: A promising small RNA delivery approach for lung diseases. J. Control. Release 2022, 352, 556–569.
[CrossRef] [PubMed]
526. Li, D.; Wu, N. Mechanism and application of exosomes in the wound healing process in diabetes mellitus. Diabetes Res. Clin.
Pract. 2022, 187, 109882. [CrossRef]
527. Murali, V.P.; Holmes, C.A. Biomaterial-based extracellular vesicle delivery for therapeutic applications. Acta Biomater. 2021, 124,
88–107. [CrossRef]
528. Liu, B.; Lee, B.W.; Nakanishi, K.; Villasante, A.; Williamson, R.; Metz, J.; Kim, J.; Kanai, M.; Bi, L.; Brown, K. Cardiac recovery via
extended cell-free delivery of extracellular vesicles secreted by cardiomyocytes derived from induced pluripotent stem cells. Nat.
Biomed. Eng. 2018, 2, 293–303. [CrossRef]
529. Wang, Y.; Cao, Z.; Wei, Q.; Ma, K.; Hu, W.; Huang, Q.; Su, J.; Li, H.; Zhang, C.; Fu, X. VH298-loaded extracellular vesicles released
from gelatin methacryloyl hydrogel facilitate diabetic wound healing by HIF-1α-mediated enhancement of angiogenesis. Acta
Biomater. 2022, 147, 342–355. [CrossRef]
530. Kwak, G.; Cheng, J.; Kim, H.; Song, S.; Lee, S.J.; Yang, Y.; Jeong, J.H.; Lee, J.E.; Messersmith, P.B.; Kim, S.H. Sustained Exosome-
Guided Macrophage Polarization Using Hydrolytically Degradable PEG Hydrogels for Cutaneous Wound Healing: Identification
of Key Proteins and MiRNAs, and Sustained Release Formulation. Small 2022, 18, 2200060. [CrossRef]
531. Wang, C.; Li, N.; Li, Y.; Hou, S.; Zhang, W.; Meng, Z.; Wang, S.; Jia, Q.; Tan, J.; Wang, R. Engineering a HEK-293T exosome-based
delivery platform for efficient tumor-targeting chemotherapy/internal irradiation combination therapy. J. Nanobiotechnol. 2022,
20, 247. [CrossRef] [PubMed]
532. Kis, Z. Stability modelling of mRNA vaccine quality based on temperature monitoring throughout the distribution chain.
Pharmaceutics 2022, 14, 430. [CrossRef] [PubMed]
533. Nawaz, M.; Heydarkhan-Hagvall, S.; Tangruksa, B.; González-King Garibotti, H.; Jing, Y.; Maugeri, M.; Kohl, F.; Hultin, L.;
Reyahi, A.; Camponeschi, A. Lipid nanoparticles deliver the therapeutic VEGFA mRNA in vitro and in vivo and transform
extracellular vesicles for their functional extensions. Adv. Sci. 2023, 10, 2206187. [CrossRef] [PubMed]
534. Joy, R.; George, J.; John, F. Brief outlook on polymeric nanoparticles, micelles, niosomes, hydrogels and liposomes: Preparative
methods and action. ChemistrySelect 2022, 7, e202104045. [CrossRef]
535. Chabria, Y.; Duffy, G.P.; Lowery, A.J.; Dwyer, R.M. Hydrogels: 3D drug delivery systems for nanoparticles and extracellular
vesicles. Biomedicines 2021, 9, 1694. [CrossRef] [PubMed]
536. Sun, J.; Yin, Z.; Wang, X.; Su, J. Exosome-laden hydrogels: A novel cell-free strategy for in-situ bone tissue regeneration. Front.
Bioeng. Biotechnol. 2022, 10, 866208. [CrossRef]
537. Yang, S.; Jiang, H.; Qian, M.; Ji, G.; Wei, Y.; He, J.; Tian, H.; Zhao, Q. MSC-derived sEV-loaded hyaluronan hydrogel promotes
scarless skin healing by immunomodulation in a large skin wound model. Biomed. Mater. 2022, 17, 034104. [CrossRef]
538. Khayambashi, P.; Iyer, J.; Pillai, S.; Upadhyay, A.; Zhang, Y.; Tran, S.D. Hydrogel encapsulation of mesenchymal stem cells and
their derived exosomes for tissue engineering. Int. J. Mol. Sci. 2021, 22, 684. [CrossRef]
539. Li, J.; Mooney, D.J. Designing hydrogels for controlled drug delivery. Nat. Rev. Mater. 2016, 1, 16071. [CrossRef]
540. Mardpour, S.; Ghanian, M.H.; Sadeghi-Abandansari, H.; Mardpour, S.; Nazari, A.; Shekari, F.; Baharvand, H. Hydrogel-mediated
sustained systemic delivery of mesenchymal stem cell-derived extracellular vesicles improves hepatic regeneration in chronic
liver failure. ACS Appl. Mater. Interfaces 2019, 11, 37421–37433. [CrossRef]
541. Shirakura, T.; Kelson, T.J.; Ray, A.; Malyarenko, A.E.; Kopelman, R. Hydrogel nanoparticles with thermally controlled drug
release. ACS Macro Lett. 2014, 3, 602–606. [CrossRef] [PubMed]
542. Zhang, S.; Bellinger, A.M.; Glettig, D.L.; Barman, R.; Lee, Y.-A.L.; Zhu, J.; Cleveland, C.; Montgomery, V.A.; Gu, L.; Nash, L.D. A
pH-responsive supramolecular polymer gel as an enteric elastomer for use in gastric devices. Nat. Mater. 2015, 14, 1065–1071.
[CrossRef] [PubMed]
543. Hurler, J.; Žakelj, S.; Mravljak, J.; Pajk, S.; Kristl, A.; Schubert, R.; Škalko-Basnet, N. The effect of lipid composition and liposome
size on the release properties of liposomes-in-hydrogel. Int. J. Pharm. 2013, 456, 49–57. [CrossRef] [PubMed]
544. Palac, Z.; Hurler, J.; Škalko-Basnet, N.; Filipović-Grčić, J.; Vanić, Ž. Elastic liposomes-in-vehicle formulations destined for skin
therapy: The synergy between type of liposomes and vehicle. Drug Dev. Ind. Pharm. 2015, 41, 1247–1253. [CrossRef] [PubMed]
545. Lenzini, S.; Bargi, R.; Chung, G.; Shin, J.-W. Matrix mechanics and water permeation regulate extracellular vesicle transport. Nat.
Nanotechnol. 2020, 15, 217–223. [CrossRef] [PubMed]
546. Zhu, Y.; Zeng, Q.; Zhang, Q.; Li, K.; Shi, X.; Liang, F.; Han, D. Temperature/near-infrared light-responsive conductive hydrogels
for controlled drug release and real-time monitoring. Nanoscale 2020, 12, 8679–8686. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2024, 25, 485 51 of 51
547. Ma, G.; Lin, W.; Yuan, Z.; Wu, J.; Qian, H.; Xu, L.; Chen, S. Development of ionic strength/pH/enzyme triple-responsive
zwitterionic hydrogel of the mixed l-glutamic acid and l-lysine polypeptide for site-specific drug delivery. J. Mater. Chem. B 2017,
5, 935–943. [CrossRef] [PubMed]
548. Yeruva, T.; Lee, C.H. Enzyme Responsive Delivery of Anti-Retroviral Peptide via Smart Hydrogel. AAPS PharmSciTech 2022,
23, 234. [CrossRef]
549. Li, Z.; Zhu, D.; Hui, Q.; Bi, J.; Yu, B.; Huang, Z.; Hu, S.; Wang, Z.; Caranasos, T.; Rossi, J. Injection of ROS-responsive hydrogel
loaded with basic fibroblast growth factor into the pericardial cavity for heart repair. Adv. Funct. Mater. 2021, 31, 2004377.
[CrossRef]
550. Kuang, L.; Huang, J.; Liu, Y.; Li, X.; Yuan, Y.; Liu, C. Injectable hydrogel with NIR light-responsive, dual-mode PTH release for
osteoregeneration in osteoporosis. Adv. Funct. Mater. 2021, 31, 2105383. [CrossRef]
551. Kubota, T.; Kurashina, Y.; Zhao, J.; Ando, K.; Onoe, H. Ultrasound-triggered on-demand drug delivery using hydrogel microbeads
with release enhancer. Mater. Des. 2021, 203, 109580. [CrossRef]
552. Qu, J.; Liang, Y.; Shi, M.; Guo, B.; Gao, Y.; Yin, Z. Biocompatible conductive hydrogels based on dextran and aniline trimer as
electro-responsive drug delivery system for localized drug release. Int. J. Biol. Macromol. 2019, 140, 255–264. [CrossRef] [PubMed]
553. Tang, J.; Qiao, Y.; Chu, Y.; Tong, Z.; Zhou, Y.; Zhang, W.; Xie, S.; Hu, J.; Wang, T. Magnetic double-network hydrogels for tissue
hyperthermia and drug release. J. Mater. Chem. B 2019, 7, 1311–1321. [CrossRef] [PubMed]
554. Brumberg, V.; Astrelina, T.; Malivanova, T.; Samoilov, A. Modern wound dressings: Hydrogel dressings. Biomedicines 2021,
9, 1235. [CrossRef] [PubMed]
555. Vowden, K.; Vowden, P. Wound dressings: Principles and practice. Surgery 2017, 35, 489–494. [CrossRef]
556. Zhao, D.; Yu, Z.; Li, Y.; Wang, Y.; Li, Q.; Han, D. GelMA combined with sustained release of HUVECs derived exosomes for
promoting cutaneous wound healing and facilitating skin regeneration. J. Mol. Histol. 2020, 51, 251–263. [CrossRef]
557. Guan, G.; Zhang, Q.; Jiang, Z.; Liu, J.; Wan, J.; Jin, P.; Lv, Q. Multifunctional silk fibroin methacryloyl microneedle for diabetic
wound healing. Small 2022, 18, 2203064. [CrossRef]
558. Ma, W.; Zhang, X.; Liu, Y.; Fan, L.; Gan, J.; Liu, W.; Zhao, Y.; Sun, L. Polydopamine decorated microneedles with Fe-MSC-derived
nanovesicles encapsulation for wound healing. Adv. Sci. 2022, 9, 2103317. [CrossRef]
559. Yuan, M.; Liu, K.; Jiang, T.; Li, S.; Chen, J.; Wu, Z.; Li, W.; Tan, R.; Wei, W.; Yang, X. GelMA/PEGDA microneedles patch loaded
with HUVECs-derived exosomes and Tazarotene promote diabetic wound healing. J. Nanobiotechnol. 2022, 20, 147. [CrossRef]
560. Bertsch, P.; Diba, M.; Mooney, D.J.; Leeuwenburgh, S.C. Self-healing injectable hydrogels for tissue regeneration. Chem. Rev. 2022,
123, 834–873. [CrossRef]
561. Zhang, Y.; Bi, J.; Huang, J.; Tang, Y.; Du, S.; Li, P. Exosome: A review of its classification, isolation techniques, storage, diagnostic
and targeted therapy applications. Int. J. Nanomed. 2020, 22, 6917–6934. [CrossRef] [PubMed]
562. Cao, Y.; Xu, Y.; Chen, C.; Xie, H.; Lu, H.; Hu, J. Local delivery of USC-derived exosomes harboring ANGPTL3 enhances spinal
cord functional recovery after injury by promoting angiogenesis. Stem Cell Res. Ther. 2021, 12, 20. [CrossRef] [PubMed]
563. Halevi, O.; Chen, J.; Thangavel, G.; Morris, S.A.; Uliel, T.B.; Tischler, Y.R.; Lee, P.S.; Magdassi, S. Synthesis through 3D printing:
Formation of 3D coordination polymers. RSC Adv. 2020, 10, 14812–14817. [CrossRef] [PubMed]
564. Born, L.J.; McLoughlin, S.T.; Dutta, D.; Mahadik, B.; Jia, X.; Fisher, J.P.; Jay, S.M. Sustained released of bioactive mesenchymal
stromal cell-derived extracellular vesicles from 3D-printed gelatin methacrylate hydrogels. J. Biomed. Mater. Res. Part A 2022, 110,
1190–1198. [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.