Exosomes: natural nanoparticles as bio shuttles for RNAi delivery

Exosomes: natural nanoparticles as bio shuttles for RNAi delivery

Accepted Manuscript Exosomes: natural nanoparticles as bio shuttles for RNAi delivery Saber Ghazizadeh Darband, Mohammad Mirza-AghazadehAttari, Mojta...

2MB Sizes 0 Downloads 44 Views

Accepted Manuscript Exosomes: natural nanoparticles as bio shuttles for RNAi delivery

Saber Ghazizadeh Darband, Mohammad Mirza-AghazadehAttari, Mojtaba Kaviani, Ainaz Mihanfar, Shirin Sadighparvar, Bahman Yousefi, Maryam Majidinia PII: DOI: Reference:

S0168-3659(18)30572-8 doi:10.1016/j.jconrel.2018.10.001 COREL 9484

To appear in:

Journal of Controlled Release

Received date: Revised date: Accepted date:

3 July 2018 30 September 2018 1 October 2018

Please cite this article as: Saber Ghazizadeh Darband, Mohammad Mirza-AghazadehAttari, Mojtaba Kaviani, Ainaz Mihanfar, Shirin Sadighparvar, Bahman Yousefi, Maryam Majidinia , Exosomes: natural nanoparticles as bio shuttles for RNAi delivery. Corel (2018), doi:10.1016/j.jconrel.2018.10.001

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT Exosomes: natural nanoparticles as bio shuttles for RNAi delivery Saber Ghazizadeh Darbanda, Mohammad Mirza-Aghazadeh-Attarib,c, , Mojtaba Kavianid, Ainaz Mihanfare, Shirin Sadighparvarf, Bahman Yousefig,h,* [email protected], Maryam Majidiniai,j,**,1 [email protected] a

IP

T

Danesh Pey Hadi Co., Health Technology Development Center, Urmia University of Medical

CR

Sciences, Urmia, Iran; b

Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran;

d

School of Nutrition and Dietetics, Acadia University, Wolfville, Nova Scotia, Canada;

e

Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences,

AN

US

c

f

M

Urmia, Iran;

Neurophysiology Research Center, Urmia University of Medical Sciences, Urmia, Iran;

g

Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran;

PT

h

ED

Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran;

i

Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran; j

*

CE

Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran;

AC

Correspondence to: Bahman Yousefi, Immunology Research Center, Tabriz University of Medical

Sciences, Tabriz, Iran. **

Correspondence to: Maryam Majidinia, Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran. 1

These authors contributed equally to this work

Abstract Application of exosomes, natural nanoscale vesicles, as specific delivery vehicles has received considerable attention in recent past years. The presence of various adhesive proteins on the

ACCEPTED MANUSCRIPT surface of these lipid bilayers, gives them the ability to interact with cellular membranes. Although the function of exosomes was not known for some time, further researches have suggested that they are effective in multiple cellular pathways, as well as pathogenesis of a broad range of diseases including neurodegenerative diseases, cardiovascular diseases, and more

T

importantly, multiple human malignancies. As erstwhile research brought to light more aspects

IP

of exosomes’ biogenesis and functions, researchers sought to benefit from their effects in

CR

therapeutic and diagnostic purposes. Gene therapy is one of these fields that has seen many endeavors made. The present review article looks at gene therapy and its latest advancements,

US

structure and function of exosomes and their role as bio shuttles in various clinical contexts

AN

relating to gene therapy.

AC

CE

PT

ED

M

Keywords: Exosomes; miRNA, siRNA, gene transferring, vehicle

ACCEPTED MANUSCRIPT

Introduction The past century is hallmarked with giant steps taken towards diagnosis and management of infectious and non-infectious diseases, which led to increased quality and duration of life in virtually all societies. Notwithstanding, many diseases and pathologies such as cancers, chronic

T

inflammatory diseases, single gene defects and degenerative diseases of organs have been largely

IP

immune to the mentioned effect, and classic therapies and diagnostic methods have limited

CR

application (1). Various new modalities have been introduced to further the treatment and

US

diagnosis of these diseases. One of these relatively new concepts is gene therapy in which genetic material is transferred to where it is needed (2). One crucial step of gene therapy is the

AN

medium by which the load is delivered, or the so called vector. Finding an efficient vector is one

M

of the main hurdles of development in gene therapy (3). Fortunately, progress in cellular biochemistry and physiology has resulted in better understanding of cellular mechanisms and

ED

discovery of various mediators. One of these mediators is exosomes, which was introduced in

PT

1983. The function of exosomes was not known for some time, but further research suggested that exosomes play roles in multiple cellular pathways and pathologies (4). As new functions of

CE

these lipid bilayers were unraveled, researchers sought to benefit from their application for

AC

therapeutic and diagnostic purposes, for example, gene therapy (5). Accordingly, the present review article explores gene therapy and its latest advancements. It further delineates the structure and function of exosomes, and, finally, investigates exosomes’ role as bio shuttles in various clinical contexts relating to gene therapy.

ACCEPTED MANUSCRIPT Gene therapy Gene therapy or the replacement of a defective gene with a corrected one, is an attractive and promising strategy for scientists, and more importantly patients (6). Many scholars believe that the foundations of gene therapy were upheld in 1928, when a British scientist called Griffith

IP

T

made a shocking observation-non-virulent bacteria had the capability to transform into virulent

CR

specimen. Future studies proved that the transforming agent in Griffith’s observation was part of the cell-free extract and would precipitate by adding alcohol, which would be called

US

deoxyribonucleic acid (DNA). Over the years, other mechanisms of sharing DNA between bacteria were understood, and new principles emerged in the field of genetics. In the wake of

AN

these advances, the first case of gene therapy on a human was conducted on an adenosine

M

deaminase (ADA) patient (7). Gene therapy can be divided to two distinct variants with the ability of passing the therapeutic effect on to the generations; one targeting somatic cells and the

ED

other targeting germ line cells,. Somatic gene therapy is the dominant form and is classified into

PT

three types of ex-vivo, in-situ and in-vivo delivery (8-10). One important obstacle in the way of introducing gene therapy into clinics is gene delivery, as many specialists express concerns about

CE

the safety and efficiency of the vectors used (11). Various vectors have been introduced to

AC

facilitate the entry of genes into cells, including viral and non-viral vectors. Viral vectors include retroviral, adenoviral, herpes simplex, lentivirus, poxvirus and Ebstein-Barr virus (EBV) vectors. Viral vectors are the most common vectors used, but have their share of limitations as causing immune responses, mutagenesis and limited capacity to transfer genes. The non-viral methods comprise a wide array of chemical and physical methods. Although they are not as efficient as viral counterparts, they do not elicit any immune response.(12). Since the first human gene therapy, various uses have been developed for gene therapy, such as cystic fibrosis and other

ACCEPTED MANUSCRIPT single gene diseases (13, 14), chronic neurologic diseases including Alzheimer’s and Parkinson’s (15), craniofacial regeneration (16), orthopedic applications (17) and the emergency field of gene therapy in cancer. Numerous clinical studies have studied the efficacy of gene therapy in cancer such as prostate cancer (18, 19), melanoma (20), glioblastoma (21), to name a few. Gene therapy

T

in cancer is divided into three general subtypes: immunotherapy or enhancing the function of the

IP

native immune system to target cancer cells and eliminate them, has led to the emergence of

CR

vaccines against cancers, oncolytic agents, which are viruses that have been modified to target cancer cells, and gene transfer, which is the new concept of introducing specific genes into

US

cancerous cells of their surrounding environment in order to destroy or incapacitate them (22).

AN

This third subtype can be further divided into three subtypes consisting of introduction of anti-

M

angiogenic agents, mutation compensation and molecular chemotherapy (23).

ED

Recent progress in gene therapy

A lot has changed since the composition of the concept of gene therapy, around a century ago, to

PT

the first approved gene therapy in 1989, and the first approved clinical trial in the European

CE

Union in 2012. Despite limitation in the number of gene therapies available to the public, many clinical trials are being done annually in the context of gene therapy. One staggering study

AC

showed that from 1989 in which only one clinical trial was conducted, the number had increased to 163 trials in the year 2015 (24). Interestingly, originally the trials mainly dealt with single gene defect diseases currently, however, the majority of clinical trials performed are concerned with cancer therapy (more than 64%) and single gene disease accounts for only 10% of new trials (24, 25). A longstanding challenge facing gene therapy is gene delivery which has caused countless considerable achievements. Classically, viruses have been used as vectors, but gave their way to

ACCEPTED MANUSCRIPT newer ones owing to various toxic and immunological side-effects. One of the relatively newer vectors is the exosome (discussed in the next section). Other newly introduced vectors are bacteria. These have been used specially in tumor-targeting therapies, by mechanisms such as growth in hypoxic regions, in environment in which tumors are active, or the chemotaxis of

T

bacteria to compounds found in necrosis (26). Recent studies have implicated various species of

IP

bacteria such as clostridium, salmonella, listeria, E-coli, Bifidobacterium in gene therapy via the

CR

aforementioned mechanisms of gene therapy in cancer. One of the uses of bacteria has been the advent of new imaging techniques such as optical imaging in which luciferase enzymes produce

US

light, which is detected by cooled charged coupled device (CCD) cameras. These enzymes are

AN

mostly derived from a firefly called Photinus pyralis and a sea pansy, Renilla luciferase. Bacteria expressing these enzymes, such as Mycobacterium smegmatis or S. Typhimurium, can be used in

M

designing a vectors to introduce the enzyme (27, 28). The other types of vectors studied

ED

extensively in the past years are mammalian mesenchymal stem cells. These cells have the advantage of eliciting no adverse immunologic response, as well as possessing easy expansion

PT

compared to other vectors. The downside of these cells, however, is that their kinetics and

CE

possibility to promote tumorigenesis is not well understood (29). Nanoparticles are the last major group of vectors introduced, which comprise four types of lipid-based, polymeric, inorganic and

AC

hybrid nanoparticles (30). While nanoparticles do not activate the immune system, as viruses and bacteria do and do not have the toxic potential of virulent bacteria and viruses, utilizing them, however, can be quite challenging as to overcoming natural barriers in the way of their absorption, targeting them to localize at wanted sites, for example the cancer environment, medicalizing the research, as many are performed on murine models, and finally, concerns regarding production in large scales (31, 32).

ACCEPTED MANUSCRIPT Current delivery methods for gene therapy Viral vectors Viruses are the result of millions of years of evolutionary progress, which has endowed them

T

many acquired abilities such as entering into host cells, long term survival in the cells via

IP

genome integration and changes in the host immune system. The first use of viruses as vectors in

CR

gene therapy in clinical contexts dates back to the late 1990s and the early 2000s when retroviruses were used to treat adenosine deaminase deficiency in patients with severe combined

US

immunodeficiency. The result of those study showed superior outcomes compared with the

AN

conventional immune boosting treatments, paving the way for further use of viral vectors (33). Since then multiple viral entities , other than retroviruses have gained attention in the field of

M

gene therapy including Adenoviruses, Lentiviruses, pox viruses adeno-associated viruses and

ED

herpes viruses (34, 35). Retroviruses are RNA-bearing viruses which depend on reverse transcription for proliferation. These enable the transfer of a transgene up to 10 Kb with a

PT

lifelong expression, because of reverse transcription and the preferred route of delivery of these

CE

viruses has been ex vivo administration (33). Adenoviruses are medium-sized non-enveloped double stranded DNA viruses which are prepared for gene therapy with the removal of some of

AC

their genes encoding primary proteins, and can deliver genes up to 30 Kb with a short period of expression, in contrast to retroviruses (34). These viruses are mostly used in a local form in instances in which an immune response is needed and preferred, such as vaccine development or targeting malignant cells (36, 37). They Lentiviruses are a genus of retroviruses which came to light later. They resemble retroviruses in terms of application and characteristics, but gained popularity in treating the conditions of the hematopoietic system and hemophilia A and B (38, 39). Adeno-associated viruses (AAV) are a member of parvo viruses bearing a single stranded

ACCEPTED MANUSCRIPT DNA. They have a rather limited capacity of gene transfer (only up to 4 Kb) with relatively less safety concernsas they have low immunogenicity and yield a long period of gene expression comparable to retroviruses. Conversely, however, AAV do not integrate into the genome of the host cell, thus the amount of expression decreases with time (40). AAVs have been used in

T

multiple trials in various clinical contexts such as hemophilia, cystic fibrosis, rheumatoid

IP

arthritis, macular dystrophies, Parkinson’s disease and more (41). Pox viruses are large double

CR

stranded DNA viruses with a famous application in producing vaccines. They can carry genes up to 20Kb and have a short period of gene transcription (42). Herpes viruses are the last group

US

discussed in this article. They are a family of DNA viruses that have a unique ability to carry a

AN

comparatively larger load of genome than other viruses (30-40 kb – 152 kb in the HSV1amplicon) (43). In addition, characteristics such as a high ability in infecting cells, a genome

M

with large segments of unnecessary DNA and easy recombinant generation make it a suitable

ED

target for gene therapy (44).

PT

Non-viral vectors

Other biologic vectors have been introduced in addition to viruses, which include bacteria,

CE

bacteriophages, virus like particles, erythrocyte ghosts and exosomes (47). The simplest way to

AC

transfer genes into a tissue has been the direct administration of the gene, usually via injection. This method has shown some success, but further studies have suggested alternative physical and chemical methods to improve the absorption of the nucleic acids. Some of the physical methods are ballistic DNA, electroporation, sonoporation, photoporation, magnetofection, hydroporation and mechanical massage. Chemical methods utilize inorganic particles, such as silica, calcium phosphate and gold, or synthetic biodegradable compounds such as cationic lipids, lipid nanoemulsions, solid lipid nano-particles, peptide based products, or polymer based vectors, such as

ACCEPTED MANUSCRIPT Polyethylenimine, chitosan, poly (DL- Lactide) (PLA) and poly ( DL-Lactide- co- glycoside) (PLGA), Dendrimers and Polymethacrylate (45, 46).

Challenges So far, multiple types of vectors with different properties have been introduced to the field of

IP

T

gene therapy, but still there are many challenges to be overcome concerning safe and efficient

CR

delivery of genetic material. Regarding safety, some vectors can cause severe immunologic reactions, even may lead to anaphylaxis and death. Vectors such as adenoviruses, pox viruses,

US

bacteria, virus like particles and bacteriophages are among the most immunogenic of the vectors (47-49). In addition, the use of some vectors, especially those of viral origins, has the potential of

AN

causing malignancies. The first studies concerning the use of retroviruses for the treatment of

M

severe combined immunodeficiency showed development of leukemia in some patients after the

ED

treatment. This was associated with the unwanted and unpredicted increase in the expression of some proto-oncogenes after viral treatment (50), a phenomenon commonly referred to as

PT

insertional mutagenesis, which still is a major challenge to the use of viral vectors (51). One other safety issue regarding vector selection is the possibility of systemic inflammatory

CE

responses resulting from serious infections seen in biologic vectors. The odds of this happening

AC

can be minimalized by attenuating the viruses or bacteria used in gene therapy, although it does not eradicate the risk (52). The next major challenge to gene therapy is the biological limitation of conventional vectors such as viruses in transferring genetic material. Most viruses used for this purpose can only transfer segment up to 40 Kb in size (53). Currently, some researchers are trying to use less investigated vectors such as lentiviruses, or using methods such as luminal charge alteration to increase the packaging capacity, but no series of methods is yet sufficiently studied (54, 55). Another major challenge to gene therapy is the specificity of transgene delivery.

ACCEPTED MANUSCRIPT As particles, vectors of gene therapy will most likely enter the cites proximal to the circulation, and not always the desired location. Multiple methods have been introduced to overcome this problem, specifically in viral vectors, including changes in the capsid proteins, using tissue specific anti-bodies, tissue‐specific promoters or serial testing of multiple serotypes, but the

T

tissue specificity of other vectors and also ways to further make them more organ or tissue

CR

IP

specific is less studied (56, 57).

Exosomes

US

First described over 30 years ago, exosome are extracellular vesicles that are excreted to the

AN

extracellular space by the conjugation of intermediate endocytic bodies to the plasma membrane. The name of this intermediate structure is “Multivesicular body”, which can also connect to

M

lysosomes (58). Multivesicular body releases intraluminal vesicles to the extracellular space.

ED

Intraluminal vesicles or exosomes are generated by two pathways, one dependent on the endosomal sorting complexes required for transport (ESCRT), which are complex of cytosolic

PT

proteins binding to membrane proteins marked, usually but not necessarily by ubiquitination

CE

(59), and a non-dependent pathway (60). Exosomes are excreted from a wide array of living mammalian cells, such as B cells, dendritic cells, T cells, mastocytes, platelets as well as

AC

reticulocytes and tumors, and proteins from various families are involved in their structure , including integrins, tetraspanins, MHC molecules, membrane fusion peptides such as RAB5 and 7, RAP1B and annexins, cytoskeleton associated molecules and many other undiscovered components (figure 1) (61). As noted, exosomes are present in various cell lines, and with diversity of origin comes diversity of function. One important function of exosomes concerns maturation of reticulocytes, more specifically, dumping the excess proteins in the process of maturation (62).. Exosomes are a key

ACCEPTED MANUSCRIPT effector of inflammation, and diseases such as multiple sclerosis, Parkinson’s and Alzheimer’s, signaling the future use of exosomes as biomarkers in these pathologies (64-67). Exosomes also have established roles in cancer, as they are frequently encountered in the micro-environment wherein the tumors are active(68), being effective in adaptive immunity towards malignant cells,

T

possibly both as a pro and anti-cancer agent (69). In addition, they are an important mediator of

IP

angiogenesis and vascular integrity and permeability. Uptake of exosome secreted by cancers is

CR

effective in increasing angiogenesis, and promoting leakage of vessels, leading to more metastasis (70). The entities also contribute to the interaction of cancer-associated fibroblasts

US

with neoplastic cells; Exosome secreted by fibroblasts adjacent to the tumoral tissue causes

AN

chemo-resistance in colorectal cancer cells (71). Further discussion is done in the next

cancer. advances

in

knowledge

of exosomes

including

their

many

roles

in

cellular

ED

Recent

M

paragraphs regarding the function of exosomes in normal cells and human pathologies and

communication and human pathologies or treating and diagnosing human cancers and other

PT

diseases has compelled researchers to focus on finding techniques and modalities help isolate

CE

and enrich exosomes, such as ultracentrifugation, size based isolation like ultra-filtration, immune-affinity capture, precipitation and micro-fluid based isolation each with their specific

(73).

AC

shortcomings (72). Various commercial kits are available for exosome extraction in this regard

Exosomes formation As a subgroup of extracellular vesicles, exosomes have biogenesis similarities to microvesicles, and to some extend to apoptotic bodies. The formation of exosomes is hallmarked by the fusion of the endosome to the cellular membrane. But multiple steps are needed to be taken beforehand;

ACCEPTED MANUSCRIPT Genesis of exosomes starts with the emergence of intra-luminal vesicles dependent on a subset of molecules called endosomal sorting complex required for transport (ESCRT). Four major complexes of ESCRT associated with intra-luminal vesicles formation include ESCRT-0, which is responsible for clustering ubiquitinated proteins on the endosome, ESCRT-I with bridging

T

roles between ESCRT 0 and 2, ESCRT2 with roles in budding, and ESCRT3 which recruits

IP

other proteins to the site of endosome formation and vesicle scission (74). The next major step in

CR

the formation of exosomes comprises the transport and fusion of multi-vesicle bodies with the cell membrane. This step is mainly mediated by RAB family of proteins, which control functions

US

such as budding, mobility and interactions of the cytoskeleton (75, 76). The final step in the

AN

formation of the exosome is the fusion of the membranes of the multi-vesicle bodies and the cell membrane. This is regulated by soluble NSF-attachment protein receptor (SNARE) complexes,

M

with considerable heterogeneity among various cell lines and conditions (77). This step results in

PT

Composition of exosomes

ED

the releasing of the exosomes into extracellular space.

Since the discovery of exosomes, the knowledge of their composition has grown significantly.

CE

Early studies of exosomes revealed that these entities were indeed a small space covered by a

AC

lipid membrane (4). More recent studies have established that exosomes are a complicated mixture of various lipids, proteins and RNAs. The abundance is so much that specific databases such as EXOCARTA have been created to list the latest molecules found in the structure of exosomes. Based on this data, over 1010 kinds of lipids, over 9690 kinds of proteins, more than 3300 kinds of mRNAs, over 1400 kinds of mi-RNAs, 18 kinds of r-RNAs, 60 kinds of t-RNAs, 110 kinds of sno-RNAs, 27 kinds of snRNAs, 6 kinds of Inc-RNAs, 3 kinds of Linc-RNAs, 5 kinds of nc-RNAs are present in the structure of the exosomes being made in different cell lines

ACCEPTED MANUSCRIPT in different species and under different conditions (78, 79). The components of exosomes significantly dictate the function and application that these entities have in multiple contexts. Probably the most important functional compartment consists of proteins (tetraspanins, Rab family proteins etc.) with vital roles in the creation of exosomes.); they can have enzymatic

T

activities (such as peroxidase, dehydrogenase) or can induce functional overlaps between

IP

exosomes and some signaling pathways (such as NOTCH and WNT-b catenin pathway) (80). Of

CR

clinical interest, however, are some proteins of exosomes proposed to be tumor markers, such as CEA in colorectal cancer, or Survivin in breast cancer (81). Furthermore, exosomes have shown

US

to have major histocompatibility complex class I and II, shown in studies focusing on

AN

bronchoalveolar lavage fluid showing that exosomes could have roles in the local immune

exosomes in the bone marrow (83).

M

system of the respiratory system (82), and in a study which found the same function for

ED

In addition to proteins, exosomes have multiple RNA molecules, both functional m-RNAs with

PT

enriched 3′-untranslated regions and a wide array of non-coding RNAs (84). The diversity of various m-RNAs existent in exosomes led to the creation of a specific data base for this purpose,

CE

called the exoRBase (85). The study that found the abundance of m-RNAs in exosomes also

AC

noted that these m-RNAs could have regulatory functions in the host cells, because of their enriched 3′-untranslated regions (84). Exosome microRNAs are also important from a functional point of view as their role has been stablished in numerous human pathologies, including cancer. It has been shown that these micro-RNAs have key mediatory roles in promoting angiogenesis, facilitation of migration and metastasis as well as changing the tumor micro-environment (86).

ACCEPTED MANUSCRIPT Cellular uptake, and intracellular trafficking Though we are well on our way to understanding exosomes’ function and structure, our knowledge of how these entities are incorporated into cells is still young, only recently research initiatives have been able to describe to some extend how exosomes are taken by cells. After

T

release, exosomes are freely, but temporarily, available in body fluids and inter-cellular spaces

IP

before being relocated in the reticolo-endothelial system. When an exosome comes into contact

CR

with a cell membrane, the interaction between receptor molecules on the two membranes is necessary to establish the function of exosomes. It is believed that exosomes tend to attach to

US

cells with a suitable receptor profile for MHC classes 1 and 2 already present externally on

AN

exosomes, which is determined by the parent cell (87). Exosomes can internalize into cells in different methods, including fusion, probably mediated by integrins and tetraspanins (88, 89),

M

phagocytosis which is dependent on the action of opsonins, actin cytoskeleton and dynamin2

ED

(90), Macropinocytosis dependent on phosphatidylinositol 3-kinase and Na+−H+ ion exchange (91, 92), Receptor- and Raft-mediated Endocytosis (Clathrin-mediated Endocytosis) which relies

PT

on a ligand-receptor interaction between the exosome and the cell or the presence of rich

CE

cholesterol- and sphingolipid domains on the cell membrane (91, 93), Heparan sulfate proteoglycan-dependent endocytosis which is dependent on Heparan sulfate proteoglycan, a

AC

macromolecule receptor (94). A study by Tian et al. found that four related steps distinguishable in the trafficking of exosomes. In the first step, which is probably dependent on actins, exosomes move in the periphery of cells, in the second step, exosomes moved from the periphery to the perinuclear region, the third step, is recognized by exosomes entering a random movement and in the last step, exosomes made a reversely move away from the central parts of the cell towards the periphery, mediated by kinesins (95).

ACCEPTED MANUSCRIPT Isolation and characterization of exosomes As tiny particles smaller than 100 nm, isolation and purification of exosomes may by elusive (96), as they vary greatly in size, shape, density, surface potential and surface molecules. However, advances in laboratory methods, have enabled researchers to utilize the unique

isolation,

ultrafiltration

techniques

plus

sieving

IP

ultracentrifugation-based

T

characteristics of exosomes in order to isolate them. A few of these methods include and

exclusion

CR

chromatography methods, which sort out exosomes based on size; immune-affinity capture-

US

based techniques, which utilize the existence of specific molecules on the membrane of the exosomes; precipitation, which uses solubility or dispensability of exosomes; and finally

AN

microfluidics-based isolation techniques, which separate exosomes according to their multiple physical and biologic characteristics (72). These methods have their own advantages and

M

disadvantages with many factors affecting their efficiency based on the specimen, technical

ED

competency, number of specimens and the given time, so researchers should decide which

PT

method suits their needs better. The differential centrifugation is probably the most frequently used method used for exosome isolation (97). In addition to exosome isolation, various

CE

techniques are made available for the evaluation of different, which vary likewise based on

AC

characteristics such as shape, size, size distribution, quantity, surface charge, and biochemical composition of these nanoparticles. Some of techniques use super-high resolution electron microscopy (EM), cryo-EM and immuno-cryoEM to evaluate structure, size and surface molecules; low resolution nanoparticle tracking analysis (NTA) are used in the assessment of size and physical properties such as zeta potential; some of other important techniques include protein analysis via mass spectrometry, western blotting, immunofluorescence staining, and ELISA, RNA analysis via RNA sequencing, PCR and other platforms, and biochemical analyses

ACCEPTED MANUSCRIPT of lipids, sugar, and other components(80). Validation of the isolated exosomes can also be challenging, because of the similarities between exosomes and other cellular components and debris. Since exosomes are of endosomal origin, it would be practical to use molecules such as tetraspannins like CD9, 63, 81 and 82, Hsc70 and Hsp90, MVB proteins such as Alix and

IP

T

TSG101 as markers of detection (98).

CR

Labeling of Exosomes

The recently gained importance of exosomes has made their visualization and detection a

US

necessity. Multiple protocols have been proposed for exosome labeling, with fluorescent staining paying a major role in most of them. A recent protocol suggested by Morales-Kastresana et al.

AN

recommended the use of 5-(and-6)-carboxyfluorescein diacetate succinimidyl ester as a

M

fluorescent dye, coupled with nanoFACS a high end flow cytometric method, and a

ED

complementary size exclusion chromatography to undermine the unconjugated labels (99). Other labeling agents include such as PKH26 (lipophilic fluorescent dyes), 1, 1'-Dioctadecyl-3, 3, 3', (100),

PKH67

(101),

octadecyl rhodamine B

PT

3'-tetramethylindocarbocyanine perchlorate

chloride (102), SYTO RNA for exosomes with RNAs (103), BODIPY TR ceramide (104) and

CE

possibly more agents to be utilized in the future. Other methods of staining have also been

AC

suggested for exosome labeling such as immunostaining, negative staining and immuno-gold labelling (105) as well as radiolabeling of extracellular vesicles, a well-studied method which could be done with agents such as (99m)Tc used in in vivo studies (106), 111In-oxine (107) and Iodine 125 (108).

ACCEPTED MANUSCRIPT Drug Loading into Exosomes We previously noted that exosomes are composed of both hydrophilic and hydrophobic compounds, a fact that makes loading therapeutic agents into exosomes challenging, especially when the trend is to use exosomes to deliver various agents ranging from RNAs to proteins of

T

various size. Various methods have been introduced in order to tackle the problem efficiently,

IP

which are classified into two major groups of active and passive. The passive methods refer to

CR

techniques in which mostly hydrophobic agents are incubated with exosomes or cells with the

US

capacity of forming and excreting exosomes (109). These methods have gained significance as it was proposed that exosomes that were made by cancer contributed to tumor progression by

AN

effecting the tumor micro-environment, and that the same functions could be used to introduce miRNAs with anti-tumor effects (110, 111). The active ones, consisted of sonication, extrusion,

M

freeze and thaw cycles, electroporation, incubation with membrane permeabilizers and click

ED

chemistry method for direct conjugation. In sonication, a mechanical force, for example sound

PT

energy, is used to temporarily disrupt the membrane of the exosome so that the therapeutic compound can enter the exosome. In a study by Haney et al. with the aim of introducing

CE

constructs to be used for the treatment of Parkinson’s disease, researchers sonicated the

AC

exosomes in a mixture containing catalase(112). Another study also used sonication to produce paclitaxel loaded exosomes, where researchers found that the use of sonication resulted in an optimal loading efficacy and sustainable release of paclitaxel (113). The next method of active loading which also disrupts the membrane of the exosome is extrusion. In this method sheer mechanical force is used to combine exosomes and drugs using a lipid extruder under controlled temperatures. One example is the hot melt extrusion process which uses high temperatures and pressures (114). This method was used in a study by Yang et al. where extrusion of MCF-10A

ACCEPTED MANUSCRIPT cells was done in order to deliver si-RNAs to cancerous tissues (115). A similar study by Jang et al. further evaluated the efficacy of using exclusion in creating exosome like structures to target cancer cells, and found that the exosome mimetics resulted from the extrusion process were able to induce TNF-α-stimulated cell death (116).

IP

T

Freeze and thaw cycles differs from the previously mentioned methods in that it does not require

CR

physical force for drug encapsulation. Instead, a mixture of exosomes and the drug is incubated and rapidly frozen, and then let to thaw, multiple times (117). This method tends to better

US

preserve the integrity of the membrane of the exosome, but it may produce un-even sized

AN

exosomes due to the fusion of exosomes together (118).

Electroporation, or electropermeabilization, is a process in which an electrical field is used to

M

create pores in the membranes of exosomes in order to facilitate the influx of multiple agents into

ED

the exosome (119). This method of introduction has been mainly used to deliver RNA products, such as miRNAs (120). A study by Liang et al. used electroporation- created exosomes

PT

containing miR-26ato suppress cell division and migration in HepG2 cells (121). Another study

CE

loaded exosomes with RNAs targeting c-MYC using this method (122). An alternative pathway to induce pore formation in cells is to use special compounds such as saponin, a toxic agent

AC

found in marine creatures and plants which tends to act as a soap (123). The study done by Haney et al. used this method to introduce catalase to in vivo and in vitro models of Parkinson’s disease (112). The theoretical limitation of saponin use is that it is a toxic agent and has the potential to induce autophagy via multiple apoptotic pathways (124). Two additional pathways exist for loading agents onto exosomes by cross linking exosomes to them. Chemistry click and antibody use are examples of these methods. These two methods have

ACCEPTED MANUSCRIPT not been studied as much as the others, but seem promising as being quick, specific and less invasive towards exosome structural integrity (125, 126). Bio-distribution of Exosomes Bio-distribution of exosomes is an important matter to consider in using them as a therapeutic

IP

T

strategy. In vivo studies have shown that it is dependent on the source cell, route of

CR

administration and various methods of targeting. A study by Morishita et al. looked into various studies on the topic of exosome pharmacokinetics and found that with regard to characteristics of

US

exosomes, most of them would have the most concentration in the liver, spleen and the pancreas, alongside other organs of the abdomen and the lungs (127). In a study by Wiklander et al.

AN

multiple kinds of extracellular vesicles from cell lines of HEK293T, C2C12, B16-F10 and OLN-

M

93 were extracted and administered in routes of I.V, I.P and S.C. It was shown that I.V administration caused a significant increase in levels of exosomes in the liver compared to the

ED

other two methods, but it led to a decreased accumulation in the pancreas and the gastrointestinal

PT

system as well. Cell lines also made different exosomes with different affinities, as ones made by the C2C12 cell line had increased accumulation in the liver, compared to ones from dendritic

CE

cells with the least accumulation (128). Distributions of exosomes has been shown to be

AC

dependent on tissue structure and the vasculature of the environment, as it was shown that I.V administrated exosomes were able to concentrate in xenograft tumors with leaky vasculature in less than an hour (129). As we already noted, swift clearance of natural exosomes by liver and spleen is one challenge of exosome use . Many efforts have been made to increase the stability of exosomes, either by modifying the surface molecules of exosomes or using polyethylene glycol to cover the exosomes. Kooijmans et al. even suggested using polyethylene glycol combined with nanobodies to confer specificity to exosomes. In their study, polyethylene glycol was

ACCEPTED MANUSCRIPT conjugated to nanobodies specific for epidermal growth factor receptor, which resulted in a significant increase in binding of exosomes to tumor cells over-expressing EGFR (130).

The exosome function in physiological and pathological conditions

T

Exosomes released from the membrane are biologically active entities that mediate different and

IP

absolutely important physiological and biological processes attributed to them since their first

CR

introduction in reticulocytes (131, 132). Participating in immune response and facilitating antigen presenting are two most documented functions of exosomes (61). For example, it was reported

US

that CD4+ T cells can be potently stimulated by exosomes released from Eppstein- Barr virus-

AN

transformed B cells (133). Therefore, activation of T cells is a part of exosomes-mediated stimulation of immune responses (133). Furthermore, tumor cells- derived exosomes have also

M

demonstrated to modulate immune reactions (134).

ED

In cancer, exosomes are major players of cell–cell communication between tumor cells through exchanging efficient information in order to induce a pro-tumoral milieu for tumorigenesis and

PT

control the immune system promoting tumor initiation/progression (68, 134). A cumulative body

CE

of studies have attributed a critical function to exosomes in angiogenesis, apoptosis, inflammation and coagulation (135). Promotion of pro-tumor micro-environment and harboring metastatic

AC

niches is another important function of exosomes (136). Additionally Exosomes may suppress the immune system and, thus mediate tumor escape from the immune attack. Exosomes also deliver mutant proteins such as KRAS and MET oncoprotein into tumor cells through endocytosis or other internalization strategies (81). In addition to proteins, onco-miRNA are also carried by exosomes to modulate the tumor cells function at post-transcriptional level (137-139). Besides the critical function of exosomes in the cancer progression, these carriers also play major function in the normal physiology and development of the nervous system, as well as regeneration of normal

ACCEPTED MANUSCRIPT neurons. In a study by Lachenal et al (140) it was reported that exosomes released from somatodendritic compartments are involved in the trans-synaptic diffusion of pathogenic molecules throughout the tissue. Additionally, the authors observed that glutamatergic synapses are also involved in the regulation of exosomal release as a part of normal synaptic physiology. More

T

interestingly, Frühbeis et al (141) reported that exosomes are key players of local cell–cell

IP

communication between neurons and oligodendrocytes. Neuronal stress-induced exosomes

CR

derived from oligodendrocytes deliver protective cargos such as mRNA, miRNA, proteins, and glycolytic enzymes to axons, and exert neuroprotection (142). In addition to delivering

US

neuroprotective cargos, exosomes are also associated with the transportation of misfolded

AN

proteins, hence initiation and progression of various neurodegenerative diseases (143). For example, in Parkinson’s disease, α-synuclein mutated proteins are transported from producing

M

cells to the nearby cells via exosomes, whereby the disease spreads from cell to cell within the

ED

brain (144). In another neurodegenerative disorder, Alzheimer’s disease, exosomal surface was reported to function as a seed for the β-amyloid aggregation after protein conformational

and

Gerstmann–Sträussler–Scheinker

syndrome,

exosomes

mediate

the

CE

Creutzfeldt–Jakob

PT

modifications, and promotion of neurodegeneration (145-147). In the prion diseases such as

propagation of the disease-related infectious agent, PrPSc (148-150). Taking together, these

AC

findings implicate a role for exosomes in neurodegenerative diseases, making them an interesting biomarker for diagnostic and prognostic purposes thereof. Exosomes function is also frequently investigated in cardiovascular diseases because of their importance in the regulation of angiogenesis, coagulation, and inflammation (151, 152). The molecular components of exosomes may have a significant function in cell–cell communication and exerting cardioprotective effects in pathological conditions such as myocardial infarction, and ischemic reperfusion (153-155).

ACCEPTED MANUSCRIPT Exosomes as bio shuttle for gene therapy Observation of different contents of exosomes with different source, particular RNA and protein profiles, as well as exosome-mediated effects on other cells led to the hypothesis that exosomes might be good candidates for transferring gene in the gene therapy process (156). In an early

IP

T

study in this context, Eldh et al (157) demonstrated that the plausible mechanism that was

CR

involved in the cell- cell communication were actually RNA-contained exosomes. Individual cells exposed to oxidative stress release exosomes, which can affect target cells and mediate resistance

US

to oxidative stress (157). Additionally, exosomes originated from EBV-infected nasopharyngeal carcinoma (NPC) cells carried signaling pathways-related particular molecules, as well as virus-

AN

coded microRNSs (miRNAs), which mediated intracellular communication and exerted a potent

M

impact on the proliferation of other neighboring cells (158). Therefore, transferring of important stimulatory or inhibitory information between donor and recipient cells, as a key function of

ED

exosomes contributes to the various aspects of development, as well as pathogenesis of various

PT

diseases (159). Being a natural transporters, low homing in liver, low toxicity and immune reaction, easily crossing biological barriers, especially blood brain barrier, art among some

CE

important features rendering exosomes advantageous over other vectors (160).

AC

In gene therapy process, exosomes are used as carriers of a basic amount of therapeutic agents, such as small interfering RNAs (siRNAs) and miRNAs, in order to deliver them in more efficient and specific manner to the target tissues (161). Several recent studies have delivered therapeutic siRNA into the target tissues by using exosomes (table 1) (119, 162-164). SiRNAs-loaded exosomes are frequently shown to selectively silence gene expression. Low stability of siRNAs and rapid degradation in the systemic circulation are the two most important problems facing siRNA-based gene therapy, and are now resolved by the advent of exosomes delivering (165,

ACCEPTED MANUSCRIPT 166). Exosomes have amply shown to be good candidates for the efficient delivery of nucleotide sequences into recipient cells, some studies have evaluated shuttling potential of these natural nano-carriers as a therapeutic strategies in combating cancer, neurodegenerative disease, cardiovascular disorders and other diseases.

T

Application in cancer

IP

Cancer is probably the single biggest challenge facing medicine and healing from cancer is

CR

regarded as the Holy Grail of medicine (167-170). Recent attention in cancer therapy has been

US

directed to the use of exosomes as natural carriers for delivering small nucleotides, specifically transferring of multiple miRNA molecules such as let-7a, miR-143, miR-146b, and miR-223, into

AN

different cancer cell lines in order to halt the tumor growth, as well as suppressing the target

M

genes (171-174). Banizs et al (175) showed that endothelial cells- derived exosomes have delivered siRNA against luciferase into primary endothelial cells. In addition, exosomes

ED

containing siRNA against GFP, or a c-Myc shRNA were efficiently transfected into recipient

PT

cells, and consequently resulted in suppression of target gene expression (122). Munoz et al (176)showed that exosome base delivery of anti-miR- 9 overcame the chemo

CE

resistance in glioblastoma. Zhang et al (177) demonstrated that exosomes derived from mouse fibroblast L929 cell line could be used as an ideal siRNA delivery tool for the transportation of

AC

siRNA against transforming growth factor (TGF) 1, thereby treatment of murine sarcomas tumor cells. Cancer cells treatment with exosomes containing TGF-1 siRNA significantly reduced the proliferation and invasion of cancer cells, as well as resulting in considerable increase in the apoptosis of tumor cells. In animal model of cancer, it was reported that TGF- 1 levels and more importantly the TGF- 1 signaling downstream were suppressed due to intravenous injection of designed exosome into tumor cells. Therefore, exosomes- mediated gene silencing effectively

ACCEPTED MANUSCRIPT inhibited the growth of cancer, as well as their metastases in lung (177). Shtam et al (178) transfected HeLa and HT1080 cell lines with exosomes containing siRNAs against RAD51 and RAD52 to evaluate the possible therapeutic application, where siRNAs caused an effective gene silencing at post-transcription level in receipt cells. In addition, S/G2 cell cycle arrest, as well as

T

massive cell death was the significant result of the down-regulation of RAD51 but not RAD52

IP

protein by recipient cell treatment with exosomes containing the specific siRNA. siRNA

CR

mediated- suppression of RAD51 decreased the recruitment of RAD51 as a key player of homologous recombination at double-stranded breaks induced in HeLa cells by ionizing radiation

US

(178). In another study, Yang et al (179) used a modified exosomes to deliver siRNA against

AN

Survivin into bladder cancer in vitro an in vivo. A tumor-penetrating peptide (iRGD peptide) was expressed on the membrane surface of the designed exosome, which facilitated the delivery of

M

exosome cargo into tumor cells. The expression levels of Survivin was significantly suppressed in

ED

exosome-recipient bladder cancer cells. Likewise, these exosomes significantly suppressed the growth rate of tumor in animal models (179). In another study done also on the bladder cancer

PT

cell lines, the efficacy of the exosomes derived from human embryonic kidney 293 (HEK293)

CE

cell and mesenchymal stem cell (MSC) for the delivery of siRNA against polo-like kinase-1 (PLK-1) gene into recipient cells was evaluated (180). Successful suppression of the PLK-1

AC

expression mediated by exosome resulted in a significant decrease in cancer cells proliferation (180). Yang et al (115) designed a nanoscale exosome-mimics containing CDK4 siRNA and transfected it into breast cancer cell lines. The expression levels of CDK4 plummeted in exosome transfected cells and this inhibitory effect was accompanied by the induction of cell cycle arrest at G1. The authors also examined the anti-tumor impacts of exosomes in BALB/C-nu mice, a mouse model of breast cancer, and showed significant suppression of tumor growth (115). In addition to

ACCEPTED MANUSCRIPT application of exosomes as a delivery system for suppressing tumor growth, some recent studies have investigated the potential of exosomes to deliver nucleotide sequences for the inhibition of angiogenesis, metastasis, as well as overcoming multidrug resistance. For example, Gong et al (181) demonstrated that mesenchymal stem cells (MSCs)-derived exosomes containing pro-

T

angiogenic miRNAs, such as miR-424, -30b, -30c, and Let-7, promoted tube-like structure

IP

formation and angiogenesis capacity of human umbilical vein endothelial cells (HUVECs). Cells

CR

treatment with the exosome secretion blocker GW4869 reduced the pro-angiomiRs containing exosomes and hence blocking their stimulatory effects (181). Liao et al (182). showed that

US

transfection of esophageal cancer cell line with exosomes shuttling miR-21 resulted in significant

AN

promotion of the invasion and migration of cancer cells. It was also demonstrated that these exosomes targeted programmed cell death 4 (PDCD4) and activated c-Jun N-terminal kinase

M

(JNK) signaling pathway downstream. Therefore, the authors reported exosome-containing miR-

ED

21a has a close association with esophageal cancer recurrence and distant metastasis (182). On the other hand, delivery of MSC derived-exosomes containing anti-mir-9 to glioblastoma

PT

multiform cells was shown to reverse multidrug resistance to temozolomide. The underlying

CE

mechanisms included the downregulation of multidrug resistance transporters such as Pglycoprotein and induction of apoptosis (176). Taking together, it has been indicated that

AC

exosomes could be an appropriate vehicle for the delivery of miRNAs and siRNAs to inhibit the various aspects of tumor progression (174). More importantly, delivery of tumor-suppressive miRNAs via exosomes into cancer cells could successfully inhibit cancer proliferation through the silencing of target genes (164). In addition to direct targeting of tumor cells by exosomes, it was reported that stimulation of dendritic cells by exosomes containing trans-activator gene enabled the immune system to interact and perish cancer cells (183).

ACCEPTED MANUSCRIPT Application in neurodegenerative and brain- related diseases In addition to human malignancies, exosomes have also been used as shuttling system todeliver siRNAs into neurons in order to affect the production rate of particular proteins implicated in neural diseases characterized by the accumulation of misfolded proteins or protein deficiencies

T

(184). This application of exosomes is related to their capability in crossing the blood brain

IP

barrier (BBB) (137). Alvarez-Erviti et al (163). dendritic cells-derived exosomes could

CR

efficiently deliver siRNA against BACE1, as an important gene contributed in pathogenesis of

US

Alzheimer’s disease to the brain in mice. Exosomes-containing siRNA are shown to specifically deliver their cargo to neurons, microglia, oligodendrocytes in the brain, and consequently

AN

knockdown target gene. Exosomes with some important features such as broad distribution and high efficacy for delivering siRNA to brain contribute to designing novel, specific and potent

M

therapeutic strategies to combat Huntington’s and other neurodegenerative diseases. In a study

ED

by Didiot et al (185), exosomes containing hydrophobically modified siRNAs (hsiRNAs) that

PT

target Huntingtin mRNA were shown to efficiently become internalized by mouse primary cortical neurons, which resulted in downregulation of Huntingtin gene. Additionally, unilateral

CE

infusion of exosomes containing hsiRNA into mouse striatum promoted bilateral distribution of oligonucleotide and statistically significant bilateral downregulation of Huntingtin mRNA, in

AC

comparison to hsiRNAs alone. Suppressing the expression levels of Alpha-synuclein ( -Syn), whose aggregation characterizes Parkinson’s disease, is said to help attenuate the progression of the disease. Copper et al (186) designed an exosome containing siRNA against  -Syn to induce a significant reduction in the expression as well as the aggregated levels of -Syn in mouse brains. The expression levels of  -Syn was significantly decreased in the brain of treated mice. As a result, protein aggregates levels dropped in dopaminergic neurons of the substantia nigra.

ACCEPTED MANUSCRIPT Furthermore, it was reported that communication between mesenchymal stem cells (MSCs) and brain parenchymal cells, obtained by transferring miR-133b-loaded exosomes into neural cells, influences the regulation of neurite outgrowth (187). Following treatment of stroke, this communication acts to promote neural plasticity and functional remodeling. In other words,

and synaptophysin, hencestimulating neurite

IP

regulating the expression levels of NF-200

T

MSCs-derived exosomes mediate the miR-133b transferring into astrocytes and neurons, hence

CR

remodeling and recovery after stroke (188). The beneficial effects of neurons transfection with exosomes carrying miRNA in functional recovery after stroke was also reported in a study by

US

Xin et al (189). The authors observed that mice treatment with exosome containing miR-17–92

AN

cluster resulted in the significant recovery of neurological functions and increase in the proliferation of oligodendrocytes, as well as neurogenesis and neurite repairing/ neuronal

M

dendrite plasticity in the ischemic sites. A recent study used exosomes-based gene delivery for

ED

the treatment of morphine relapse in vitro and in vivo (190). The authors indicated that siRNA against opioid receptor mu delivered by exosomes potently suppressed morphine relapse via

PT

decreasing expression levels of the opioid receptor mu (190).

CE

Application in cardiovascular and other diseases

AC

In recent years, a great progress has been made in the application of gene therapy for cardiovascular diseases (191, 192). Accumulating body of studies have demonstrated the critical role of exosomes and exosomes-mediated cross-talk between multiple cell types in the perpetuation of heart homeostasis, as well as development of cardiac disorders (193-196). Exosomes derived from cardiac progenitor cells (CPC) containing miR-210 and miR-132 could deliver their content into HL-1 cells (193). Under normal conditions, plasma-derived exosomes are reported to be involved in the protection of healthy cardiomyocytes from ischemia/

ACCEPTED MANUSCRIPT reperfusion

injury

(154,

197).

Following

myocardial infarction,

CPC-derived

exosomes

containing some protective factors such as clusterin, miR-21, and miR-210 were released and exerted potent protective effects as inhibition of apoptosis in cardiomyocytes, stimulation of angiogenesis, promotion of repair mechanisms, and, subsequently, improvement of cardiac

T

function (155, 198, 199). Therefore, the exosome application as a gene delivery shuttle seems

IP

rational for cardiovascular diseases. Additionally, the beneficial effects of exosome-base gene

CR

therapy was reported in ischemic kidney injury (200). Endothelial colony forming cells (ECFCs)derived exosomes enriched with miR-486-5p caused significant upregulation and downregulation

US

of miR-486-5p and PTEN, respectively, as well as stimulation of Akt phosphorylation in

AN

hypoxia-exposed endothelial cells. Injection of ECFC-derived exosomes into animal model of ischemic kidney injury strongly resulted in the induction of functional and histologic protection,

M

such as reduced PTEN levels and activated Akt, all related to overexpression of miR-486-5p in

ED

the kidney. Thus, it was suggested that delivery of ECFC-derived exosomes can decrease ischemic kidney injury via transferring miR-486-5p targeting PTEN (200). plays

a

critical

role

PT

MiRNA-122

in

the

hepatocyte

damage

and

inflammation

in

CE

monocytes/macrophages, hence pathogenesis of alcoholic hepatitis (AH). Momen-Heravi et al (201) demonstrated that delivery of exosome-loaded miRNA-122 inhibitor suppress the

AC

inflammatory effects of exosomes from ethanol-treated hepatocytes. Alcohol binge drinking resulted in significant enhancement of the number of exosomes containing miRNA-122 in the healthy groups. In monocytes, exosome- mediated shuttling of miRNA-122 resulted in the significant suppression of heme oxygenase -1 (HO-1) pathway, sensitization of cells to liposaccharide stimulation, and overexpression of pro-inflammatory cytokines. Table 1: exosomes application for gene delivery in various pathological conditions

ACCEPTED MANUSCRIPT Exosome cargo

Exosome source

Results

Ref.

Breast cancer cell lines

let-7a miRNA

Human embryonic kidney cell line 293 (HEK293) cells HeLa and HT1080 human fibosarcoma cells non-cancerous cells

Suppression of tumor growth

(171)

M assive reproductive cell death Selective gene silencing

(162)

Inhibition of cell proliferation

(202)

M acrophages PLC-luc cells,

Invasion of breast cancer cells Protein expression

(173) (203)

miRNA released via ceramide-dependent (164) secretory mechanism

Promotion of angiogenesis Horizontal transfer of drug-resistant trait

Tumor-suppressive microRNAs miR-223 mRNA

Breast cancer cells Hepatocellular carcinoma cells M etastatic prostate siRNA cancer cell line

siRNA

non-small cell lung cancer cell lines M esenchymal stem cells chronic myeloid leukemia cells Immortalized hepatocyte

miRNA - 222-3p miR-30b miR-365

Endothelial cells Sensitive cancer cells

Esophageal cancer cells

miR-21

Breast cancer cells Bladder cancer cells M ouse xenograft tumor Xenografted nude mice

Non-coding RNAs siRNA

AN

M

(174)

Liver tissue

Promoting colon cancer progression

(209)

Regulation of gene expression, Promoting neurite remodeling, Improving functional recovery Specific gene knockdown

(188)

Enhancing neuroplasticity and functional recovery after stroke Reducing alpha-synuclein aggregates in brains

(189)

siRNA

M ultipotent mesenchymal stromal cells Self-derived dendritic cells M esenchymal stromal cell M urine dendritic cells harvested from bone marrow Peripheral blood

Selective gene silencing

(119)

siRNA

M odified exosomes

Treatment of morphine relapse

(190)

hsiRNAs

Glioblastoma cells

Huntingtin mRNA silencing

(185)

Anti-miR-9

M esenchymal Cell

AC

CE

Rat model of primary miR-146b brain tumor Colon carcinoma cell miR-193a lines Brain cells subjected to miR-133b cerebral artery occlusion

Brain cells in siRNA Alzheimer’s disease Rat brain cells subjected miR-17–92 Cluster to stoke Brain of transgenic mice siRNA

M onocytes and lymphocytes M ouse skeletal muscle cell lines M ouse primary cortical neurons Glioblastoma multiforme cells

Promotion of cell motility (207) Activation of PI3K/AKT and M APK signal transductions Enhanced secretion of M M P-2 and M MP-9 Esophageal cancer Promotion of cell migration and invasion (182) cell line EC9706 Stromal cells Regulation of therapy resistance pathways (208) M odified exosomes Inhibition of tumor growth (179) Human monocytic Inhibition of tumor growth leukemia THP-1 cells M arrow stromal cells Inhibition of glioma growth

PT

miR-143

(205) (206)

proteins Hepatocellular carcinoma cells

ED

Oncogenic and RNAs

Successful knockdown of target mRNA (180) and protein Transferring malignant phenotypic traits (204)

US

Bladder cancer cell lines

HEK293 human embryonic kidney cell line COS-7 African green monkey kidney fibroblast like cell line HEK293 and M SC cell Sensitive cancer cells

IP

Cancerous cells

CR

HeLa and HT1080 siRNA human fibosarcoma cells

T

Target tissue

Stem Reversing the chemoresistance

(172)

(163)

(186)

(176)

ACCEPTED MANUSCRIPT Neonatal cardiomyoctes Astrocytes

rat miR-146a Neuronal miR-124a

Effector T cells

miR-150

HSC or hepatocytes

miR -214

M ast cells

Exosomal RNA miRNAs

Cardiosphere-derived cells Neuron-conditioned medium Lymph node and spleen cells M ouse or human hepatic stellate cells

Cardiac regeneration

(210)

Astroglial GLT1 expression

(211)

Inhibition contact sensitivity

(212)

Suppression of connective tissue growth (213) factor (CCN2) 3′-UTR activity Suppression αSM A or collagen synthesis

CR

IP

M onocyte-derived dendritic cells Endothelial cell

T

shuttle M ouse and a human Protein synthesis (214) mast cell line EBV-infected B cells Repression of (215) EBV Target Genes mRNA Adenocarcinoma Proliferation, migration, sprouting, and (216) model (AS-Tspan8) maturation of EC progenitors. cells miRNA T, B and dendritic M odulation of gene expression (217) immune cells Class II transactivator B16F1 murine anti-tumor effects in recipient cells (183) gene (CIITA) melanoma cell line

Antigen-presenting cells

AN

US

Dendritic cells

M

Conclusion

Because of low toxicity and high functional efficiency, exosomes are new promising vectors for

ED

the gene therapy of various diseases. Nearly four decades after introduction of exosomes, major

PT

efforts have been made in order to determine their biological and functional features. Exosomes have opened a completely new avenue for the therapeutic delivery of small nucleic acids to

CE

specific tissues. Various studies have explored the potential clinical applications of exosome in

AC

various pathological conditions. However, this field is still in its infancy and needs further researches. Tests of therapeutic efficiency should be designed and conducted before entering the clinical application.

ACCEPTED MANUSCRIPT References

AC

CE

PT

ED

M

AN

US

CR

IP

T

1. Yabroff KR, McNeel TS, Waldron WR, Davis WW, Brown ML, Clauser S, et al. Health limitations and quality of life associated with cancer and other chronic diseases by phase of care. Medical care . 2007;45(7):629-37. 2. Friedmann T. A brief history of gene therapy. Nature genetics. 1992;2(2):93. 3. Phillips AJ. The challenge of gene therapy and DNA delivery. Journal of Pharmacy and Pharmacology. 2001;53(9):1169-74. 4. Harding CV, Heuser JE, Stahl PD. Exosomes: looking back three decades and into the future. J Cell Biol. 2013;200(4):367-71. 5. Lee Y, El Andaloussi S, Wood MJ. Exosomes and microvesicles: extracellular vesicles for genetic information transfer and gene therapy. Human molecular genetics. 2012;21(R1):R125-R34. 6. Food, Administration D. FDA guidance for industry: Gene therapy clinical trials—Observing subjects for delayed adverse events. 2006. 7. Blaese RM, Culver KW, Miller AD, Carter CS, Fleisher T, Clerici M, et al. T lymphocyte -directed gene therapy for ADA− SCID: initial trial results after 4 years. Science. 1995;270(5235):475-80. 8. Bailey CJ, Davies EL, Docherty K. Prospects for insulin delivery by ex -vivo somatic cell gene therapy. Journal of molecular medicine. 1999;77(1):244-9. 9. Yang N-S, Burkholder J, Roberts B, Martinell B, McCabe D. In vivo and in vitro gene transfer to mammalian somatic cells by particle bombardment. Proceedings of the National Academy of Sciences. 1990;87(24):9568-72. 10. Hu W, Wang Z, Hollister S, Krebsbach P. Localized viral vector delivery to enhance in situ regenerative gene therapy. Gene therapy. 2007;14(11):891. 11. J O'Loughlin A, A Woffindale C, JA Wood M. Exosomes and the emerging field of exosome -based gene therapy. Current gene therapy. 2012;12(4):262-74. 12. Nayerossadat N, Maedeh T, Ali PA. Viral and nonviral delivery systems for gene delivery. Advanced biomedical research. 2012;1. 13. Aalto-setälä K, Vuorio E. Gene Therapy of Single-gene Disorders: Preface to the. Annals of medicine. 1997;29(6):549-51. 14. Burney TJ, Davies JC. Gene therapy for the treatment of cystic fibrosis. The application of clinical genetics. 2012;5:29. 15. Simonato M, Bennett J, Boulis NM, Castro MG, Fink DJ, Goins WF, et al. Progress in gene therapy for neurological disorders. Nature Reviews Neurology. 2013;9(5):277. 16. Scheller E, Krebsbach P. Gene therapy: design and prospects for craniofacial regeneration. Journal of dental research. 2009;88(7):585-96. 17. Chen Y. Orthopedic applications of gene therapy. Journal of orthopaedic science. 2001;6(2):199207. 18. Grozescu T, Popa F. Immunotherapy and gene therapy in prostate cancer treatment. Journal of medicine and life. 2017;10(1):54. 19. Shariat SF, Desai S, Song W, Khan T, Zhao J, Nguyen C, et al. Adenovirus-mediated transfer of inducible caspases: a novel “death switch” gene therapeutic approach to prostate cancer. Cancer research. 2001;61(6):2562-71. 20. Sotomayor MG, Yu H, Antonia S, Sotomayor EM, Pardoll DM. Advances in gene therapy for malignant melanoma. Cancer Control. 2002;9(1):39-48.

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

M

AN

US

CR

IP

T

21. Kwiatkowska A, Nandhu MS, Behera P, Chiocca EA, Viapiano MS. Strategies in gene therapy for glioblastoma. Cancers. 2013;5(4):1271-305. 22. Cross D, Burmester JK. Gene therapy for cancer treatment: past, present and future. Clin ical medicine & research. 2006;4(3):218-27. 23. Kesmodel SB, Spitz FR. Gene therapy for cancer and metastatic disease. Expert reviews in molecular medicine. 2003;5(17):1-18. 24. Hanna E, Rémuzat C, Auquier P, Toumi M. Gene therapies development: slow progress and promising prospect. Journal of Market Access & Health Policy. 2017;5(1):1265293. 25. De Wilde S, Guchelaar H-J, Zandvliet ML, Meij P. Clinical development of gene-and cell-based therapies: overview of the European landscape. Molecular Therapy-Methods & Clinical Development. 2016;3. 26. Baban CK, Cronin M, O’Hanlon D, O’Sullivan GC, Tangney M. Bacteria as vectors for gene therapy of cancer. Bioengineered bugs. 2010;1(6):385-94. 27. Nguyen VH, Kim H-S, Ha J-M, Hong Y, Choy HE, Min J-J. Genetically engineered Salmonella typhimurium as an imageable therapeutic probe for cancer. Cancer research. 2010;70(1):18-23. 28. Cronin M, Stanton R, Francis K, Tangney M. Bacterial vectors for imaging and cancer gene therapy: a review. Cancer gene therapy. 2012;19(11):731. 29. Jeong J-O, Han JW, Kim J-M, Cho H-J, Park C, Lee N, et al. Malignant Tumor Formation After Transplantation of Short-Term Cultured Bone Marrow Mesenchymal Stem Cells in Experimental Myocardial Infarction and Diabetic NeuropathyNovelty and Significance. Circulation research. 2011;108(11):1340-7. 30. Dizaj SM, Jafari S, Khosroushahi AY. A sight on the current nanoparticle -based gene delivery vectors. Nanoscale research letters. 2014;9(1):252. 31. Wang K, Kievit FM, Zhang M. Nanoparticles for cancer gene therapy: Recent advances, challenges, and strategies. Pharmacological research. 2016;114:56-66. 32. Liu C, Zhang N. Nanoparticles in gene therapy: principles, prospects, and challenges. Progress in molecular biology and translational science. 104: Elsevier; 2011. p. 509-62. 33. Bushman FD. Retroviral integration and human gene therapy. The Journal of clinical investigation. 2007;117(8):2083-6. 34. Lukashev A, Zamyatnin A. Viral vectors for gene therapy: current state and clinical perspectives. Biochemistry (Moscow). 2016;81(7):700-8. 35. Hukkanen V. Herpesvirus vectors in gene therapy. Bentham Science Publishers; 2010. 36. Doerfler W. Adenoviruses. 1996. 37. Raper SE, Chirmule N, Lee FS, Wivel NA, Bagg A, Gao G-p, et al. Fatal systemic inflammatory response syndrome in a ornithine transcarbamylase deficient patient following adenoviral gene transfer. Molecular genetics and metabolism. 2003;80(1):148-58. 38. Vandendriessche T, Thorrez L, Acosta‐Sanchez A, Petrus I, Wang L, Ma L, et al. Efficacy and safety of adeno‐associated viral vectors based on serotype 8 and 9 vs. lentiviral vectors for hemophilia B gene therapy. Journal of Thrombosis and Haemostasis. 2007;5(1):16-24. 39. Stein CS, Kang Y, Sauter SL, Townsend K, Staber P, Derksen TA, et al. In vivo treatment of hemophilia A and mucopolysaccharidosis type VII using nonprimate lentiviral vectors. Molecular Therapy. 2001;3(6):850-6. 40. Daya S, Berns KI. Gene therapy using adeno-associated virus vectors. Clinical microbiology reviews. 2008;21(4):583-93. 41. Kotterman MA, Chalberg TW, Schaffer DV. Viral vectors for gene therapy: translational and clinical outlook. Annual review of biomedical engineering. 2015;17:63-89. 42. Moroziewicz D, Kaufman HL. Gene therapy with poxvirus vectors. Current opinion in molecular therapeutics. 2005;7(4):317-25.

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

M

AN

US

CR

IP

T

43. Jacobs A, Breakefield XO, Fraefel C. HSV-1-based vectors for gene therapy of neurological diseases and brain tumors: part II. Vector systems and applications. Neoplasia. 1999;1(5):402-16. 44. Manservigi R, Argnani R, Marconi P. HSV recombinant vectors for gene therapy. The open virology journal. 2010;4:123. 45. Ramamoorth M, Narvekar A. Non viral vectors in gene therapy-an overview. Journal of clinical and diagnostic research: JCDR. 2015;9(1):GE01. 46. Yin H, Kanasty RL, Eltoukhy AA, Vegas AJ, Dorkin JR, Anderson DG. Non-viral vectors for genebased therapy. Nature Reviews Genetics. 2014;15(8):541. 47. Seow Y, Wood MJ. Biological gene delivery vehicles: beyond viral vectors. Molecular therapy. 2009;17(5):767-77. 48. Bessis N, GarciaCozar F, Boissier M. Immune responses to gene therapy vectors: influence on vector function and effector mechanisms. Gene therapy. 2004;11(S1):S10. 49. Mingozzi F, High KA. Immune responses to AAV vectors: overcoming barriers to successful gene therapy. Blood. 2013:blood-2013-01-306647. 50. Hacein-Bey-Abina S, Garrigue A, Wang GP, Soulier J, Lim A, Morillon E, et al. Insertional oncogenesis in 4 patients after retrovirus-mediated gene therapy of SCID-X1. The Journal of clinical investigation. 2008;118(9):3132-42. 51. Gonin P, Buchholz C, Pallardy M, Mezzina M. Gene therapy bio-safety: scientific and regulatory issues. Gene Therapy. 2005;12(S1):S146. 52. Mali S. Delivery systems for gene therapy. Indian journal of human genetics. 2013;19(1):3. 53. Thomas CE, Ehrhardt A, Kay MA. Progress and problems with the use of viral vectors for gene therapy. Nature Reviews Genetics. 2003;4(5):346. 54. Tiffany M, Kay MA. 253. Expanded Packaging Capacity of AAV by Lumenal Charge Alteration. Molecular Therapy. 2016;24:S99-S100. 55. Counsell JR, Asgarian Z, Meng J, Ferrer V, Vink CA, Howe SJ, et al. Lentiviral vectors can be used for full-length dystrophin gene therapy. Scientific reports. 2017;7(1):79. 56. O'Neill SM, Hinkle C, Chen S-J, Sandhu A, Hovhannisyan R, Stephan S, et al. Targeting adipose tissue via systemic gene therapy. Gene therapy. 2014;21(7):653. 57. Kawakami Y, Curiel DT. Vector Targeting in Gene Therapy. Reviews in Cell Biology and Molecular Medicine. 2006. 58. Edgar JR. Q&A: What are exosomes, exactly? BMC biology. 2016;14(1):46. 59. Colombo M, Moita C, van Niel G, Kowal J, Vigneron J, Benaroch P, et al. Analysis of ESCRT functions in exosome biogenesis, composition and secretion highlights the heterogeneity of extracellular vesicles. J Cell Sci. 2013;126(24):5553-65. 60. Stuffers S, Sem Wegner C, Stenmark H, Brech A. Multivesicular endosome biogenesis in the absence of ESCRTs. Traffic. 2009;10(7):925-37. 61. Théry C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function. Nature Reviews Immunology. 2002;2(8):569. 62. Johnstone RM, Adam M, Hammond J, Orr L, Turbide C. Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). Journal of Biological Chemistry. 1987;262(19):9412-20. 63. Kriebel PW, Barr VA, Rericha EC, Zhang G, Parent CA. Collective cell migration requires vesicular trafficking for chemoattractant delivery at the trailing edge. The Journal of cell biology. 2008;183(5):94961. 64. Wu X, Zheng T, Zhang B. Exosomes in Parkinson’s disease. Neuroscience bulletin. 2017;33(3):331-8. 65. Selmaj I, Mycko MP, Raine CS, Selmaj KW. The role of exosomes in CNS inflammation and their involvement in multiple sclerosis. Journal of neuroimmunology. 2017;306:1-10.

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

M

AN

US

CR

IP

T

66. Malm T, Loppi S, Kanninen KM. Exosomes in Alzheimer's disease. Neurochemistry international. 2016;97:193-9. 67. Properzi F, Logozzi M, Fais S. Exosomes: the future of biomarkers in medicine. Biomarkers in medicine. 2013;7(5):769-78. 68. Kalluri R. The biology and function of exosomes in cancer. The Journal of clinical investigation. 2016;126(4):1208-15. 69. Greening DW, Gopal SK, Xu R, Simpson RJ, Chen W, editors. Exosomes and their roles in immune regulation and cancer. Seminars in cell & developmental biology; 2015: Elsevier. 70. Todorova D, Simoncini S, Lacroix R, Sabatier F, Dignat-George F. Extracellular vesicles in angiogenesis. Circulation research. 2017;120(10):1658-73. 71. Hu Y, Yan C, Mu L, Huang K, Li X, Tao D, et al. Fibroblast-derived exosomes contribute to chemoresistance through priming cancer stem cells in colorectal cancer. PloS one. 2015;10(5):e0125625. 72. Li P, Kaslan M, Lee SH, Yao J, Gao Z. Progress in exosome isolation techniques. Theranostics. 2017;7(3):789. 73. Tang Y-T, Huang Y-Y, Zheng L, Qin S-H, Xu X-P, An T-X, et al. Comparison of isolation methods of exosomes and exosomal RNA from cell culture medium and serum. International journal of molecular medicine. 2017;40(3):834-44. 74. Schmidt O, Teis D. The ESCRT machinery. Current Biology. 2012;22(4):R116-R20. 75. Kelly EE, Horgan CP, Goud B, McCaffrey MW. The Rab family of proteins: 25 years on. Portland Press Limited; 2012. 76. Hutagalung AH, Novick PJ. Role of Rab GTPases in membrane traffic and cell physiology. Physiological reviews. 2011;91(1):119-49. 77. Kowal J, Tkach M, Théry C. Biogenesis and secretion of exosomes. Current opinion in cell biology. 2014;29:116-25. 78. Keerthikumar S, Chisanga D, Ariyaratne D, Al Saffar H, Anand S, Zhao K, et al. ExoCarta: a webbased compendium of exosomal cargo. Journal of molecular biology. 2016;428(4):688-92. 79. Mathivanan S, Fahner CJ, Reid GE, Simpson RJ. ExoCarta 2012: database of exosomal proteins, RNA and lipids. Nucleic acids research. 2011;40(D1):D1241-D4. 80. Kibria G, Ramos EK, Gius DR, Wan Y, Liu H. Exosomes as a Drug Delivery System in Cancer Therapy: Potential and Challenges. Molecular pharmaceutics. 2018. 81. Li W, Li C, Zhou T, Liu X, Liu X, Li X, et al. Role of exosomal proteins in cancer diagnosis. Molecular cancer. 2017;16(1):145. 82. Admyre C, Grunewald J, Thyberg J, Gripenbäck S, Tornling G, Eklund A, et al. Exosomes with major histocompatibility complex class II and co-stimulatory molecules are present in human BAL fluid. European Respiratory Journal. 2003;22(4):578-83. 83. Utsugi-Kobukai S, Fujimaki H, Hotta C, Nakazawa M, Minami M. MHC class I-mediated exogenous antigen presentation by exosomes secreted from immature and mature bone marrow derived dendritic cells. Immunology letters. 2003;89(2-3):125-31. 84. Batagov AO, Kurochkin IV. Exosomes secreted by human cells transport largely mRNA fragments that are enriched in the 3′-untranslated regions. Biology direct. 2013;8(1):12. 85. Li S, Li Y, Chen B, Zhao J, Yu S, Tang Y, et al. exoRBase: a database of circRNA, lncRNA and mRNA in human blood exosomes. Nucleic acids research. 2017;46(D1):D106-D12. 86. Zhang J, Li S, Li L, Li M, Guo C, Yao J, et al. Exosome and exosomal microRNA: trafficking, sorting, and function. Genomics, proteomics & bioinformatics. 2015;13(1):17-24. 87. McKelvey KJ, Powell KL, Ashton AW, Morris JM, McCracken SA. Exosomes: mechanisms of uptake. Journal of circulating biomarkers. 2015;4:7. 88. Parolini I, Federici C, Raggi C, Lugini L, Palleschi S, De Milito A, et al. Microenvironmental pH is a key factor for exosome traffic in tumor cells. Journal of Biological Chemistry. 2009:jbc. M109. 041152.

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

M

AN

US

CR

IP

T

89. Hemler ME. Tetraspanin proteins mediate cellular penetration, invasion, and fusion events and define a novel type of membrane microdomain. Annual review of cell and developmental biology. 2003;19(1):397-422. 90. Feng D, Zhao WL, Ye YY, Bai XC, Liu RQ, Chang LF, et al. Cellular internalization of exosomes occurs through phagocytosis. Traffic. 2010;11(5):675-87. 91. Tian T, Zhu Y-L, Zhou Y-Y, Liang G-F, Wang Y-Y, Hu F-H, et al. Exosome uptake through clathrinmediated endocytosis and macropinocytosis and mediating miR-21 delivery. Journal of Biological Chemistry. 2014:jbc. M114. 588046. 92. Fitzner D, Schnaars M, van Rossum D, Krishnamoorthy G, Dibaj P, Bakhti M, et al. Selective transfer of exosomes from oligodendrocytes to microglia by macropinocytosis. J Cell Sci. 2011;124(3):447-58. 93. Mayor S, Pagano RE. Pathways of clathrin-independent endocytosis. Nature reviews Molecular cell biology. 2007;8(8):603. 94. Christianson HC, Belting M. Heparan sulfate proteoglycan as a cell-surface endocytosis receptor. Matrix Biology. 2014;35:51-5. 95. Tian T, Wang Y, Wang H, Zhu Z, Xiao Z. Visualizing of the cellular uptake and intracellular trafficking of exosomes by live‐cell microscopy. Journal of cellular biochemistry. 2010;111(2):488-96. 96. Whiteside TL. Extracellular vesicles isolation and their biomarker potential: are we ready for testing? Annals of translational medicine. 2017;5(3). 97. Yakimchuk K. Exosomes: isolation and characterization methods and specific markers. Mater Methods. 2015;5:1450-3. 98. Szatanek R, Baran J, Siedlar M, Baj-Krzyworzeka M. Isolation of extracellular vesicles: determining the correct approach. International journal of molecular medicine. 2015;36(1):11-7. 99. Morales-Kastresana A, Telford B, Musich TA, McKinnon K, Clayborne C, Braig Z, et al. Labeling extracellular vesicles for nanoscale flow cytometry. Scientific reports. 2017;7(1):1878. 100. Nanbo A, Kawanishi E, Yoshida R, Yoshiyama H. Purification and Fluorescent Labeling of Exosomes. Journal of Virology. 2013. 101. Takov K, Yellon DM, Davidson SM. Confounding factors in vesicle uptake studies using fluorescent lipophilic membrane dyes. Journal of extracellular vesicles. 2017;6(1):1388731. 102. Tian T, Zhu YL, Hu FH, Wang YY, Huang NP, Xiao ZD. Dynamics of exosome internalization and trafficking. Journal of cellular physiology. 2013;228(7):1487-95. 103. Li Q, Kim Y, Namm J, Kulkarni A, Rosania GR, Ahn Y-H, et al. RNA-selective, live cell imaging probes for studying nuclear structure and function. Chemistry & biology. 2006;13(6):615-23. 104. Harp D, Driss A, Mehrabi S, Chowdhury I, Xu W, Liu D, et al. Exosomes derived from endometriotic stromal cells have enhanced angiogenic effects in vitro. Cell and tissue research. 2016;365(1):187-96. 105. Jung MK, Mun JY. Sample Preparation and Imaging of Exosomes by Transmission Electron Microscopy. Journal of visualized experiments: JoVE. 2018(131). 106. Choi H, Jang SC, Yoo MY, Park JY, Choi NE, Oh HJ, et al. Noninvasive imaging of radiolabeled exosome-mimetic nanovesicle using 99m Tc-HMPAO. Scientific reports. 2015;5:15636. 107. Gangadaran P, Hong CM, Ahn B-C. An Update on in Vivo Imaging of Extracellular Vesicles as Drug Delivery Vehicles. Frontiers in pharmacology. 2018;9:169. 108. Morishita M, Takahashi Y, Nishikawa M, Sano K, Kato K, Yamashita T, et al. 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. Journal of pharmaceutical sciences. 2015;104(2):705-13. 109. Luan X, Sansanaphongpricha K, Myers I, Chen H, Yuan H, Sun D. Engineering exosomes as refined biological nanoplatforms for drug delivery. Acta Pharmacologica Sinica. 2017;38(6):754.

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

M

AN

US

CR

IP

T

110. Bell E, Taylor MA. Functional roles for exosomal microRNAs in the tumour microenvironment. Computational and structural biotechnology journal. 2017;15:8-13. 111. Silva M, A Melo S. Non-coding RNAs in exosomes: new players in cancer biology. Current genomics. 2015;16(5):295-303. 112. Haney MJ, Klyachko NL, Zhao Y, Gupta R, Plotnikova EG, He Z, et al. Exosomes as drug delivery vehicles for Parkinson's disease therapy. Journal of Controlled Release. 2015;207:18-30. 113. Kim MS, Haney MJ, Zhao Y, Mahajan V, Deygen I, Klyachko NL, et al. Development of exosomeencapsulated paclitaxel to overcome MDR in cancer cells. Nanomedicine: Nanotechnology, Biology and Medicine. 2016;12(3):655-64. 114. Repka MA, Majumdar S, Kumar Battu S, Srirangam R, Upadhye SB. Applications of hot-melt extrusion for drug delivery. Expert opinion on drug delivery. 2008;5(12):1357-76. 115. Yang Z, Xie J, Zhu J, Kang C, Chiang C, Wang X, et al. Functional exosome-mimic for delivery of siRNA to cancer: in vitro and in vivo evaluation. Journal of Controlled Release. 2016;243:160-71. 116. Jang SC, Kim OY, Yoon CM, Choi D-S, Roh T-Y, Park J, et al. Bioinspired exosome-mimetic nanovesicles for targeted delivery of chemotherapeutics to malignant tumors. ACS nano. 2013;7(9):7698-710. 117. Cheng Y, Zeng Q, Han Q, Xia W. Effect of pH, temperature and freezing-thawing on quantity changes and cellular uptake of exosomes. Protein & cell. 2018:1-5. 118. Sato YT, Umezaki K, Sawada S, Mukai S-a, Sasaki Y, Harada N, et al. Engineering hybrid exosomes by membrane fusion with liposomes. Scientific reports. 2016;6:21933. 119. Wahlgren J, Karlson TDL, Brisslert M, Vaziri Sani F, Telemo E, Sunnerhagen P, et al. Plasma exosomes can deliver exogenous short interfering RNA to monocytes and lymphocytes. Nucleic acids research. 2012;40(17):e130-e. 120. Zhang D, Lee H, Zhu Z, Minhas JK, Jin Y. Enrichment of selective miRNAs in exosomes and delivery of exosomal miRNAs in vitro and in vivo. American Journal of Physiology -Lung Cellular and Molecular Physiology. 2016;312(1):L110-L21. 121. Liang G, Kan S, Zhu Y, Feng S, Feng W, Gao S. Engineered exosome-mediated delivery of functionally active miR-26a and its enhanced suppression effect in HepG2 cells. International journal of nanomedicine. 2018;13:585. 122. Lunavat TR, Jang SC, Nilsson L, Park HT, Repiska G, Lässer C, et al. RNAi delivery by exosomemimetic nanovesicles–Implications for targeting c-Myc in cancer. Biomaterials. 2016;102:231-8. 123. Rao A, Sung M-K. Saponins as anticarcinogens. The Journal of nutrition. 1995;125(suppl_3):717S-24S. 124. Podolak I, Galanty A, Sobolewska D. Saponins as cytotoxic agents: a review. Phytochemistry Reviews. 2010;9(3):425-74. 125. Higginbotham JN, Zhang Q, Jeppesen DK, Scott AM, Manning HC, Ochieng J, et al. Identification and characterization of EGF receptor in individual exosomes by fluorescence -activated vesicle sorting. Journal of extracellular vesicles. 2016;5(1):29254. 126. Smyth T, Petrova K, Payton NM, Persaud I, Redzic JS, Graner MW, e t al. Surface functionalization of exosomes using click chemistry. Bioconjugate chemistry. 2014;25(10):1777-84. 127. Morishita M, Takahashi Y, Nishikawa M, Takakura Y. Pharmacokinetics of exosomes—an important factor for elucidating the biological roles of exosomes and for the development of exosomebased therapeutics. Journal of pharmaceutical sciences. 2017;106(9):2265-9. 128. Wiklander OP, Nordin JZ, O'Loughlin A, Gustafsson Y, Corso G, Mäger I, et al. Extracellular vesicle in vivo biodistribution is determined by cell source, route of administration and targeting. Journal of extracellular vesicles. 2015;4(1):26316. 129. Lai CP, Mardini O, Ericsson M, Prabhakar S, Maguire CA, Chen JW, et al. Dynamic biodistribution of extracellular vesicles in vivo using a multimodal imaging reporter. ACS nano. 2014;8(1):483-94.

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

M

AN

US

CR

IP

T

130. Kooijmans S, Fliervoet L, Van Der Meel R, Fens M, Heijnen H, en Henegouwen PvB, et al. PEGylated and targeted extracellular vesicles display enhanced cell specificity and circulation time. Journal of Controlled Release. 2016;224:77-85. 131. Viaud S, Ploix S, Lapierre V, Théry C, Commere P-H, Tramalloni D, et al. Updated technology to produce highly immunogenic dendritic cell-derived exosomes of clinical grade: a critical role of interferon-γ. Journal of immunotherapy. 2011;34(1):65-75. 132. Johnstone R, Bianchini A, Teng K. Reticulocyte maturation and exosome release: transferrin receptor containing exosomes shows multiple plasma membrane functions. Blood. 1989;74(5):1844-51. 133. Raposo G, Nijman HW, Stoorvogel W, Liejendekker R, Harding CV, Melief CJ, et al. B lymphocytes secrete antigen-presenting vesicles. Journal of Experimental Medicine. 1996;183(3):1161-72. 134. Whiteside TL. Tumor-Derived Exosomes and Their Role in Tumor-Induced Immune Suppression. Vaccines. 2016;4(4):35. 135. Janowska‐Wieczorek A, Wysoczynski M, Kijowski J, Marquez‐Curtis L, Machalinski B, Ratajczak J, et al. Microvesicles derived from activated platelets induce metastasis and angiogenesis in lung cancer. International journal of cancer. 2005;113(5):752-60. 136. Rana S, Malinowska K, Zöller M. Exosomal tumor microRNA modulates premetastatic organ cells. Neoplasia. 2013;15(3):281IN14-95IN31. 137. Montecalvo A, Larregina AT, Shufesky WJ, Stolz DB, Sullivan ML, Karlsson JM, et al. Mechanism of transfer of functional microRNAs between mouse dendritic cells via exosomes. Blood. 2012;119(3):756-66. 138. Kosaka N, Takeshita F, Yoshioka Y, Hagiwara K, Katsuda T, Ono M, et al. Exosomal tumorsuppressive microRNAs as novel cancer therapy:“exocure” is another choice for cancer treatment. Advanced drug delivery reviews. 2013;65(3):376-82. 139. Peinado H, Alečković M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G, et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nature medicine. 2012;18(6):883-91. 140. Lachenal G, Pernet-Gallay K, Chivet M, Hemming FJ, Belly A, Bodon G, et al. Release of exosomes from differentiated neurons and its regulation by synaptic glutamatergic activity. Molecular and Cellular Neuroscience. 2011;46(2):409-18. 141. Frühbeis C, Fröhlich D, Kuo WP, Amphornrat J, Thilemann S, Saab AS, et al. Neurotransmittertriggered transfer of exosomes mediates oligodendrocyte–neuron communication. PLoS biology. 2013;11(7):e1001604. 142. Frühbeis C, Fröhlich D, Kuo WP, Krämer-Albers E-M. Extracellular vesicles as mediators of neuron-glia communication. Frontiers in cellular neuroscience. 2013;7. 143. Schneider A, Simons M. Exosomes: vesicular carriers for intercellular communication in neurodegenerative disorders. Cell and tissue research. 2013;352(1):33-47. 144. Danzer KM, Kranich LR, Ruf WP, Cagsal-Getkin O, Winslow AR, Zhu L, et al. Exosomal cell-to-cell transmission of alpha synuclein oligomers. Molecular neurodegeneration. 2012;7(1):42. 145. Götz J, Chen F, Van Dorpe J, Nitsch R. Formation of neurofibrillary tangles in P301L tau transgenic mice induced by Aβ42 fibrils. Science. 2001;293(5534):1491-5. 146. Bolmont T, Clavaguera F, Meyer-Luehmann M, Herzig MC, Radde R, Staufenbiel M, et al. Induction of tau pathology by intracerebral infusion of amyloid-β-containing brain extract and by amyloid-β deposition in APP× Tau transgenic mice. The American journal of pathology. 2007;171(6):2012-20. 147. Simons M, Raposo G. Exosomes–vesicular carriers for intercellular communication. Current opinion in cell biology. 2009;21(4):575-81.

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

M

AN

US

CR

IP

T

148. Fevrier B, Vilette D, Archer F, Loew D, Faigle W, Vidal M, et al. Cells release prions in association with exosomes. Proceedings of the National Academy of Sciences of the United States of America. 2004;101(26):9683-8. 149. Vella L, Sharples R, Lawson V, Masters C, Cappai R, Hill A. Packaging of prions into exosomes is associated with a novel pathway of PrP processing. The Journal of pathology. 2007;211(5):582-90. 150. Coleman BM, Hanssen E, Lawson VA, Hill AF. Prion-infected cells regulate the release of exosomes with distinct ultrastructural features. The FASEB Journal. 2012;26(10):4160-73. 151. Amabile N, Rautou P-E, Tedgui A, Boulanger CM, editors. Microparticles: key protagonists in cardiovascular disorders. Seminars in thrombosis and hemostasis; 2010: © Thieme Medical Publishers. 152. Lawson C, Vicencio JM, Yellon DM, Davidson SM. Microvesicles and exosomes: new players in metabolic and cardiovascular disease. Journal of Endocrinology. 2016;228(2):R57-R71. 153. de Jong OG, Verhaar MC, Chen Y, Vader P, Gremmels H, Posthuma G, et al. Cellular stress conditions are reflected in the protein and RNA content of endothelial cell -derived exosomes. Journal of extracellular vesicles. 2012;1(1):18396. 154. Vicencio JM, Yellon DM, Sivaraman V, Das D, Boi-Doku C, Arjun S, et al. Plasma exosomes protect the myocardium from ischemia-reperfusion injury. Journal of the American College of Cardiology. 2015;65(15):1525-36. 155. Chen L, Wang Y, Pan Y, Zhang L, Shen C, Qin G, et al. Cardiac progenitor-derived exosomes protect ischemic myocardium from acute ischemia/reperfusion injury. Biochemical and biophysical research communications. 2013;431(3):566-71. 156. Yeo RWY, Lai RC, Zhang B, Tan SS, Yin Y, Teh BJ, et al. Mesenchymal stem cell: an efficient mass producer of exosomes for drug delivery. Advanced drug delivery reviews. 2013;65(3):336-41. 157. Eldh M, Ekström K, Valadi H, Sjöstrand M, Olsson B, Jernås M, et al. Exosomes communicate protective messages during oxidative stress; possible role of exosomal shuttle RNA. PloS one. 2010;5(12):e15353. 158. Meckes DG, Shair KH, Marquitz AR, Kung C-P, Edwards RH, Raab-Traub N. Human tumor virus utilizes exosomes for intercellular communication. Proceedings of the National Academy of Sciences. 2010;107(47):20370-5. 159. Andaloussi SE, Lakhal S, Mäger I, Wood MJ. Exosomes for targeted siRNA delivery across biological barriers. Advanced drug delivery reviews. 2013;65(3):391-7. 160. Taylor DD, Gercel-Taylor C. MicroRNA signatures of tumor-derived exosomes as diagnostic biomarkers of ovarian cancer. Gynecologic oncology. 2008;110(1):13-21. 161. Marcus ME, Leonard JN. FedExosomes: engineering therapeutic biological nanoparticles that truly deliver. Pharmaceuticals. 2013;6(5):659-80. 162. Shtam TA, Kovalev RA, Varfolomeeva EY, Makarov EM, Kil YV, Filatov MV. Exosomes are natural carriers of exogenous siRNA to human cells in vitro. 2013. 163. Alvarez-Erviti L, Seow Y, Yin H, Betts C, Lakhal S, Wood MJ. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nature biotechnology. 2011;29(4):341-5. 164. Kosaka N, Iguchi H, Yoshioka Y, Takeshita F, Matsuki Y, Ochiya T. Secretory mechanisms and intercellular transfer of microRNAs in living cells. Journal of Biological Chemistry. 2010;285(23):1744252. 165. Ozpolat B, Sood AK, Lopez-Berestein G. Liposomal siRNA nanocarriers for cancer therapy. Advanced drug delivery reviews. 2014;66:110-6. 166. Tabernero J, Shapiro GI, LoRusso PM, Cervantes A, Schwartz GK, Weiss GJ, et al. First-in-humans trial of an RNA interference therapeutic targeting VEGF and KSP in cancer patients with liver involvement. Cancer discovery. 2013. 167. Yousefi B, Samadi N, Baradaran B, Rameshknia V, Shafiei-Irannejad V, Majidinia M, et al. Differential effects of peroxisome proliferator-activated receptor agonists on doxorubicin-resistant

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

M

AN

US

CR

IP

T

human myelogenous leukemia (K562/DOX) cells. Cellular and molecular biology (Noisy -le-Grand, France). 2015;61(8):118-22. 168. Tehrani SS, Karimian A, Parsian H, Majidinia M, Yousefi B. Multiple functions of long non‐coding RNAs in oxidative stress, DNA damage response and cancer progression. Journal of cellular biochemistry. 2018;119(1):223-36. 169. Majidinia M, Alizadeh E, Yousefi B, Akbarzadeh M, Zarghami N. Downregulation of Notch signaling pathway as an effective chemosensitizer for cancer treatment. Drug research. 2016;66(11):571-9. 170. Majidinia M, Yousefi B. DNA repair and damage pathways in breast cancer development and therapy. DNA repair. 2017;54:22-9. 171. Ohno S-i, Takanashi M, Sudo K, Ueda S, Ishikawa A, Matsuyama N, et al. Systemically injected exosomes targeted to EGFR deliver antitumor microRNA to breast cancer cells. Molecular Therapy. 2013;21(1):185-91. 172. Katakowski M, Buller B, Zheng X, Lu Y, Rogers T, Osobamiro O, et al. Exosomes from marrow stromal cells expressing miR-146b inhibit glioma growth. Cancer letters. 2013;335(1):201-4. 173. Yang M, Chen J, Su F, Yu B, Su F, Lin L, et al. Microvesicles secreted by macrophages shuttle invasion-potentiating microRNAs into breast cancer cells. Molecular cancer. 2011;10(1):117. 174. Akao Y, Iio A, Itoh T, Noguchi S, Itoh Y, Ohtsuki Y, et al. Microvesicle -mediated RNA molecule delivery system using monocytes/macrophages. Molecular therapy. 2011;19(2):395-9. 175. Banizs AB, Huang T, Dryden K, Berr SS, Stone JR, Nakamoto RK, et al. In vitro evaluation of endothelial exosomes as carriers for small interfering ribonucleic acid delivery. International jou rnal of nanomedicine. 2014;9:4223-30. 176. Munoz JL, Bliss SA, Greco SJ, Ramkissoon SH, Ligon KL, Rameshwar P. Delivery of functional anti miR-9 by mesenchymal stem cell–derived exosomes to glioblastoma multiforme cells conferred chemosensitivity. Molecular Therapy—Nucleic Acids. 2013;2(10):e126. 177. Zhang Y, Li L, Yu J, Zhu D, Zhang Y, Li X, et al. Microvesicle-mediated delivery of transforming growth factor β1 siRNA for the suppression of tumor growth in mice. Biomaterials. 2014;35(14):4390400. 178. Shtam TA, Kovalev RA, Varfolomeeva EY, Makarov EM, Kil YV, Filatov MV. Exosomes are natural carriers of exogenous siRNA to human cells in vitro. Cell Communication and Signaling. 2013;11(1):88. 179. Yang R, Yan X, Zhang S, Guo H. MP88-16 TARGETED EXOSOME-MEDIATED DELIVERY OF SURVIVIN SIRNA FOR THE TREATMENT OF BLADDER CANCER. The Journal of Urology. 2017;197(4):e1180. 180. Greco KA, Franzen CA, Foreman KE, Flanigan RC, Kuo PC, Gupta GN. PLK-1 silencing in bladder cancer by siRNA delivered with exosomes. Urology. 2016;91:241. e1-. e7. 181. Gong M, Yu B, Wang J, Wang Y, Liu M, Paul C, et al. Mesenchymal stem cells release exosomes that transfer miRNAs to endothelial cells and promote angiogenesis. Oncotarget. 2017;5. 182. Liao J, Liu R, Shi Y-J, Yin L-H, Pu Y-P. Exosome-shuttling microRNA-21 promotes cell migration and invasion-targeting PDCD4 in esophageal cancer. International journal of oncology. 2016;48(6):256779. 183. Lee YS, Kim SH, Cho JA, Kim CW. Introduction of the CIITA gene into tumor cells produces exosomes with enhanced anti-tumor effects. Experimental & molecular medicine. 2011;43(5):281-90. 184. Liu R, Liu J, Ji X, Liu Y. Synthetic nucleic acids delivered by exosomes: a potential therapeutic for generelated metabolic brain diseases. Metabolic brain disease. 2013;28(4):551-62. 185. Didiot M-C, Hall LM, Coles AH, Haraszti RA, Godinho BM, Chase K, et al. Exosome -mediated delivery of hydrophobically modified siRNA for Huntingtin mRNA silencing. Molecular Therapy. 2016;24(10):1836-47.

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

M

AN

US

CR

IP

T

186. Cooper JM, Wiklander P, Nordin JZ, Al‐Shawi R, Wood MJ, Vithlani M, et al. Systemic exosomal siRNA delivery reduced alpha‐synuclein aggregates in brains of transgenic mice. Movement Disorders. 2014;29(12):1476-85. 187. Xin H, Li Y, Buller B, Katakowski M, Zhang Y, Wang X, et al. Exosome‐mediated transfer of miR‐ 133b from multipotent mesenchymal stromal cells to neural cells contributes to neurite outgrowth. Stem cells. 2012;30(7):1556-64. 188. Xin H, Li Y, Liu Z, Wang X, Shang X, Cui Y, et al. MiR‐133b promotes neural plasticity and functional recovery after treatment of stroke with multipotent mesenchymal stromal cells in rats via transfer of exosome‐enriched extracellular particles. Stem cells. 2013;31(12):2737-46. 189. Xin H, Katakowski M, Wang F, Qian J-Y, Liu XS, Ali MM, et al. Microrna cluster miR-17–92 cluster in exosomes enhance neuroplasticity and functional recovery after stroke in rats. Stroke. 2017:STROKEAHA. 116.015204. 190. Liu Y, Li D, Liu Z, Zhou Y, Chu D, Li X, et al. Targeted exosome-mediated delivery of opioid receptor Mu siRNA for the treatment of morphine relapse. Scientific reports. 2015;5. 191. Hulot J-S, Ishikawa K, Hajjar RJ. Gene therapy for the treatment of heart failure: promise postponed. European heart journal. 2016;37(21):1651-8. 192. Hammoudi N, Ishikawa K, Hajjar RJ. Adeno-associated virus-mediated gene therapy in cardiovascular disease. Current opinion in cardiology. 2015;30(3):228. 193. Cervio E, Barile L, Moccetti T, Vassalli G. Exosomes for intramyocardial intercellular communication. Stem cells international. 2015;2015. 194. Das S, Halushka MK. Extracellular vesicle microRNA transfer in cardiovascular disease. Cardiovascular pathology. 2015;24(4):199-206. 195. Emanueli C, Shearn AI, Angelini GD, Sahoo S. Exosomes and exosomal miRNAs in cardiovascular protection and repair. Vascular pharmacology. 2015;71:24-30. 196. Bang C, Batkai S, Dangwal S, Gupta SK, Foinquinos A, Holzmann A, et al. Cardiac fibroblast– derived microRNA passenger strand-enriched exosomes mediate cardiomyocyte hypertrophy. The Journal of clinical investigation. 2014;124(5):2136. 197. Hall JL. Exosomes decrease infarct size. Journal of the American College of Cardiology; 2015. 198. Khan M, Nickoloff E, Abramova T, Johnson J, Verma SK, Krishnamurthy P, et al. Embryonic stem cell-derived exosomes promote endogenous repair mechanisms and enhance cardiac function following myocardial infarction. Circulation research. 2015:CIRCRESAHA. 115.305990. 199. Foglio E, Puddighinu G, Fasanaro P, D'Arcangelo D, Perrone GA, Mocini D, et al. Exosomal clusterin, identified in the pericardial fluid, improves myocardial performance following MI through epicardial activation, enhanced arteriogenesis and reduced apoptosis. International journal of cardiology. 2015;197:333-47. 200. Viñas JL, Burger D, Zimpelmann J, Haneef R, Knoll W, Campbell P, et al. Transfer of microRNA486-5p from human endothelial colony forming cell–derived exosomes reduces ischemic kidney injury. Kidney international. 2016;90(6):1238-50. 201. Momen-Heravi F, Bala S, Kodys K, Szabo G. Exosomes derived from alcohol-treated hepatocytes horizontally transfer liver specific miRNA-122 and sensitize monocytes to LPS. Scientific reports. 2015;5. 202. Kosaka N, Iguchi H, Yoshioka Y, Hagiwara K, Takeshita F, Ochiya T. Competitive interactions of cancer cells and normal cells via secretory microRNAs. Journal of Biological Chemistry. 2012;287(2):1397-405. 203. Kogure T, Lin WL, Yan IK, Braconi C, Patel T. Intercellular nanovesicle‐mediated microRNA transfer: A mechanism of environmental modulation of hepatocellular cancer cell growth. Hepatology. 2011;54(4):1237-48.

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

M

AN

US

CR

IP

T

204. Wei F, Ma C, Zhou T, Dong X, Luo Q, Geng L, et al. Exosomes derived from gemcitabine -resistant cells transfer malignant phenotypic traits via delivery of miRNA-222-3p. Molecular cancer. 2017;16(1):132. 205. Gong M, Yu B, Wang J, Wang Y, Liu M, Paul C, et al. Mesenchymal stem cells release exosomes that transfer miRNAs to endothelial cells and promote angiogenesis. Oncotarget. 2017;8(28):45200. 206. Min Q-h, Wang X-z, Zhang J, Chen Q-G, Li S-q, Liu X-q, et al. Exosomes derived from imatinibresistant chronic myeloid leukemia cells mediate a horizontal transfer of drug-resistant trait by delivering mir-365. Experimental cell research. 2018;362(2):386-93. 207. He M, Qin H, Poon TC, Sze S-C, Ding X, Co NN, et al. Hepatocellular carcinoma-derived exosomes promote motility of immortalized hepatocyte through transfer of oncogenic proteins and RNAs. Carcinogenesis. 2015;36(9):1008-18. 208. Boelens MC, Wu TJ, Nabet BY, Xu B, Qiu Y, Yoon T, et al. Exosome transfer from stromal to breast cancer cells regulates therapy resistance pathways. Cell. 2014;159(3):499-513. 209. Teng Y, Ren Y, Hu X, Mu J, Samykutty A, Zhuang X, et al. MVP-mediated exosomal sorting of miR193a promotes colon cancer progression. Nature communications. 2017;8:14448. 210. Ibrahim AG-E, Cheng K, Marbán E. Exosomes as critical agents of cardiac regeneration triggered by cell therapy. Stem cell reports. 2014;2(5):606-19. 211. Morel L, Regan M, Higashimori H, Ng SK, Esau C, Vidensky S, et al. Neuronal exosomal miRNAdependent translational regulation of astroglial glutamate transporter GLT1. Journal of Biological Chemistry. 2013;288(10):7105-16. 212. Bryniarski K, Ptak W, Jayakumar A, Püllmann K, Caplan MJ, Chairoungdua A, et al. Antigenspecific, antibody-coated, exosome-like nanovesicles deliver suppressor T-cell microRNA-150 to effector T cells to inhibit contact sensitivity. Journal of Allergy and Clinical Immunology. 2013;132(1):170-81. e9. 213. Chen L, Charrier A, Zhou Y, Chen R, Yu B, Agarwal K, et al. Epigenetic regulation of connective tissue growth factor by MicroRNA‐214 delivery in exosomes from mouse or human hepatic stellate cells. Hepatology. 2014;59(3):1118-29. 214. Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, Lötvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nature cell biology. 2007;9(6):654-9. 215. Pegtel DM, Cosmopoulos K, Thorley-Lawson DA, van Eijndhoven MA, Hopmans ES, Lindenberg JL, et al. Functional delivery of viral miRNAs via exosomes. Proceedings of the National Academy of Sciences. 2010;107(14):6328-33. 216. Nazarenko I, Rana S, Baumann A, McAlear J, Hellwig A, Trendelenburg M, et al. Cell surface tetraspanin Tspan8 contributes to molecular pathways of exosome-induced endothelial cell activation. Cancer research. 2010;70(4):1668-78. 217. Mittelbrunn M, Gutiérrez-Vázquez C, Villarroya-Beltri C, González S, Sánchez-Cabo F, González MÁ, et al. Unidirectional transfer of microRNA-loaded exosomes from T cells to antigen-presenting cells. Nature communications. 2011;2:282.

AN

US

CR

IP

T

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

M

Figure 1: Key components of a typical exosome.

AC

CE

PT

ED

M

AN

US

CR

IP

T

ACCEPTED MANUSCRIPT

Figure 1