Journal of Clinical Epigenetics Open Access

  • ISSN: 2472-1158
  • Journal h-index: 10
  • Average acceptance to publication time (5-7 days)
  • Average article processing time (30-45 days) Less than 5 volumes 30 days
    8 - 9 volumes 40 days
    10 and more volumes 45 days

Review Article - (2016) Volume 2, Issue 1

The Emerging Role of Extracellular Vesicle-Derived miRNAs: Implication in Cancer Progression and Stem Cell Related Diseases

Qiwei Yang*, Michael P Diamond and Ayman Al-Hendy

Division of Translation Research, Department of Obstetrics and Gynecology, Augusta University, Medical College of Georgia, Augusta, GA, USA

*Corresponding Author:

Qiwei Yang
Division of Translation Research, Department of Obstetrics and Gynecology
Augusta University, Medical College of Georgia, Augusta, GA, USA
Tel: 706-721-8801
E-mail: QYANG@gru.edu

Received date: January 18, 2016; Accepted date: January 27, 2016; Published date: January 31, 2016

Citation: Yang Q, Diamond MP, Al-Hendy A. The Emerging Role of Extracellular Vesicle- Derived miRNAs: Implication in Cancer Progression and Stem Cell Related Diseases. J Clin Epigenet. 2016, 2:1. DOI: 10.21767/2472-1158.100013

Copyright: © 2016 Yang Q, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Visit for more related articles at Journal of Clinical Epigenetics

Abstract

Cells release into the extracellular environment, diverse types of membrane vesicles of endosomal and plasma membrane origin called exosomes and microvesicles. A number of studies indicate that these extracellular vehicles (EVs) mediate the interaction between cancer cells and their microenvironment; and thereby, play a critical role in the development of cancers. EVs contain cargo which consist of proteins, lipids, mRNAs, and miRNAs that can be delivered to different types of cells in nascent as well as distal locations. Discovery of this latter cargo has drawn an increasing amount of attention, due to their altering effects on the transcriptome, proteins, and subsequent cellular characteristics in recipient cells. Cancer cell derived exosomes (CCEs) have been identified in body fluids of cancer patients including urine, plasma and saliva. Because CCE content largely depends on tumor type and stage, they invariably lend great potential in serving as prognostic and diagnostic markers. Notably, accumulating evidence demonstrates that EV-derived miRNAs have key roles in regulating various aspects of cellular homeostasis, including proliferation, survival, migration, metastasis, and the immune system etc. More recently, diagnostic and therapeutic exploitation of stem cells derived EVs are under investigation. This review aims to summarize recent advances in EV-derived miRNAs in a variety of tumor types, and suggests that these cancer-derived exosomal miRNAs play a critical role in regulating cellular functions in surrounding and distant locations. It also discusses the role of adverse environmental exposure in altering stem cell exosomal miRNA profiling, which we believe leads to changes in the extracellular environment as well as a diverse range of biological processes.

Keywords

Extracellular vesicles; Exosomes; miRNA, Cancer cells; Stem cells

Introduction

miRNA and their functions

MicroRNAs (miRNAs) are small, non-coding RNA molecules (21-23 nt) found in various species that play an important role in transcriptional and post-transcriptional regulation of gene expression [1,2]. For most miRNAs, primary miRNA transcripts are generated by RNA polymerase II, either as separate transcriptional units or embedded within the introns of protein coding genes [3]. MiRNAs are generated from a long primary transcript (pri-miRNA) which is cleaved in the nucleus by a complex formed by the protein DGCR8/Pasha and the RNAse III Drosha to a shorter hair-pin structure (50–120 nt) which constitutes the miRNA precursor (pre-miRNA) [4]. The pre-miRNA is exported to the cytoplasm, where it is processed by the RNAse III enzyme Dicer to a 22-nucleotide duplex and then loaded onto the miRNA-containing RNA-induced silencing complex (miRISC) where miRNA duplex is unwound and the single stranded 22 nt mature miRNA, which originates from one arm of the pre-miRNA hairpin, is bound to an Argonaute protein [5]. Only one strand of the duplex remains in the RISC-Loading complex as a mature miRNA (guide strand or miRNA), whereas the other (passenger strand) is rapidly degraded. Mature miRNAs function by binding to complementary sequences within mRNA molecules, usually resulting in gene silencing via translational repression or target degradation [5,6]. The presence in the target mRNA 3′-UTR of multiple sites for the same or different miRNAs generally confers repression, more effectively. To date, approximately 2,000 miRNAs have been identified in the human genome [7-9] which target over 60% of mammalian genes [10,11]. Sharing of seed sequences and the presence of overlapping targets indicate that a single miRNA can target many different mRNAs and that different miRNAs can target the same mRNA as well. This suggests that miRNAs are involved in integrating gene regulatory networks, in both physiological and pathological conditions. miRNA was first discovered in the early 1990s [12]. Subsequently, many short regulatory RNAs were identified in almost all multicellular organisms [13-17], in single cellular algae, and DNA viruses [18,19]. miRNAs have been found to be involved in many biological events including normal development, differentiation, growth control, and numerous diseases including cancer [20-25]. Importantly, microRNAs are already considered diagnostic and prognostic biomarkers for patient stratification as well as therapeutic targets and agents.

miRNA in cancers/tumors

miRNA negatively regulates gene expression by binding to the 3′ untranslated region (3′UTR) of mRNA, leading to degradation or translation blockade. Deregulation of miRNA is tightly linked to cancer initiation and progression, and circulating miRNAs have emerged as potential biomarkers for cancer diagnosis and prognosis. In 2002, the first study to demonstrate the link between deregulation of mir-15 and 16 and the pathogenesis of chronic lymphocytic leukemia was reported [26]. Subsequently, miRNAs have been suggested as biomarkers in cancer, because miRNAexpression arrays are found to be more efficient in classifying cancer than mRNA-expression arrays [27]. miRNAs have been found not only in tissues, but also in body fluids including serum, plasma, and urine in a stable form that is protected from RNase activity in association with RISC, either free in blood or in membrane-enclosed vesicles (such as exosomes) [28-31]. In other words, miRNAs can be secreted into the extracellular environment through exosomes or in complexes with protein or lipid-based carriers. Accumulating evidence demonstrates that miRNAs as well as proteins can be transferred to neighboring or distant cells in these secretory forms to modulate cell function. Extracellular miRNAs are therefore emerging as a new group of messengers and effectors in intercellular communication.

The major causes of altered miRNA expression in cancer include genetic and epigenetic alterations, aberrant transcription factor activity, mutation in miRNA biogenesis pathways, and other mechanisms which seek to elude miRNA repression [3,32].

miRNAs in extracellular vesicles

Exosomes: Exosomes are cell-derived extracellular vesicles (EVs) present in many and perhaps all biological fluids, including blood, urine, and cultured media of cell cultures [33,34]. Exosomes range in diameter, between 40 and 150 nm. In mammals, exosomes are either released from the cells when multivesicular bodies fuse with plasma membrane or they are released directly from the plasma membrane [35]. It is becoming increasingly clear that exosomes have specialized functions and play a key role in many biological events including tumorigenesis, metabolism, coagulation, intercellular signaling, and the immune system. Consequently, there is growing interest in the clinical applications of exosomes. Exosomes could potentially be utilized in the prognosis, therapy, and as biomarkers for health and disease.

Exosomes contain various molecular constituents, including proteins, nucleic acids, and lipids [36]. According to ExoCarta (https://www.exocarta.org), an exosome database, 4563 proteins, 194 lipids, 1639 mRNAs, and 764 miRNAs have been identified in exosomes from multiple organisms [37 38]. Although the exosomal protein composition varies with cell and tissue of origin, most exosomes contain an evolutionary-conserved, common set of protein molecules such as chaperones, subunits of the trimeric G proteins, cytoskeletal proteins, tetraspanin proteins, and other proteins [39,40]. The 2007 hallmark study, which demonstrated that exosomes contain RNA cargo [41] has garnered great interest in understanding the role of exosomes in cell-to-cell interaction leading to an altered expression pattern in recipient cells. That study also revealed the differences in cellular and exosomal mRNA – while for the first time, miRNA content was described, as well as the functionality of the exosomal mRNA cargo [41]. Soon after this discovery, exosomal miRNA, and exosome derived miRNAs were evidenced to be surrogate diagnostic markers for biopsy profiling [42,43].

Many advances have been made in the field of exosome isolation with improving knowledge and emerging novel technologies. To date, several strategies including ultracentrifugation, size-based isolation, precipitation, and affinity-based capture have been utilized to isolate exosomes [44].

Several mechanisms have been hypothesized in describing the interactions of exosomes and recipient cells. Exosomes can bind to cells through receptor–ligand interactions. Alternatively, exosomes can putatively attach or fuse with the target-cell membrane, delivering exosomal surface proteins and perhaps cytoplasm to the recipient cell. Finally, exosomes may also be internalized by the recipient cells via endocytosis [41]. Therefore, more research studies are focused on investigating the role that exosomes may play in cell-to-cell signaling; often hypothesizing that delivery of their RNA cargo molecules will explain biological effects. Accumulating evidence demonstrates that these cellto- cell communications influence both physiological and pathological processes [45-47].

Role of miRNAs from cancer cell-derived exosomes: Evidence indicates that both normal and cancer cells communicate via the release and delivery of molecules. These molecules include miRNAs which are packed into exosomes and significantly influence the physiological and pathological conditions of cells [48]. Tumor-derived exosomes can modify normal, healthy cells by altering their translational profile to promote tumor progression. Exosomes are quickly emerging as powerful sources for molecule transfer between cells in the mediation of both beneficial and pathological processes. The oncogenic profiling of miRNA in exosomes varies with cancer type and exhibits a variety of functions depending on the recipient cell [49-51] (Table1).

miRNAs Tissues/cells/diseases Functions References Year
miR-146b, miR-222 indolent papillary thyroid cancer Proliferation Lee JC, et al. 2015
miR-24-3p, miR-891a, miR106a-5p, miR-20a-5p, miR-1908 nasopharyngeal carcinoma proliferation Ye SB, et al. 2014
miR-30a, miR-138, miR-146, miR-203 chronic myeloid leukemia chemoresistance Taganov; Duncan; Yu et al. 2006, 2008, 2012
miR-21, miR-155 neuroblastoma chemoresistance Chal.lagundla et al. 2015
miR-122 breast cancer metabolism Fong et al. 2015
miR-135 multiple myeloma angiogenesis Umezu et al. 2014
miR-19a astrocyte metastasis Zhang et al. 2015
miR-21, miR-141, miR-200, miR-203, miR-205, miR-214 ovarian cancer diagnostic signatures Taylor et al. 2008
miR-17, miR-19a, mir-21, miR-126, miR-149 melanoma diagnostic signatures Pfeffer et al. 2015
miR-21, miR-34a MSCs tumor supportive miRNAs Vallabhaneni et al. 2015
miR-221 GC-MSCs proliferation, migration Wang et al. 2015
miR-23b BM-MSCs metastasis Ono et al. 2014
miR-200 breast cancer stem cells metastasis Shimono et al. 2009
miR-19b, miR-26b, miR-203 colonic stem cells dietary response miRNAs Shah MS, et al. 2016

Table 1 Extracellular vesicle-derived miRNAs in cancers and stem cells .

Diagnostic signatures: An ever-increasing body of literature demonstrates that molecular constituents of exosomes, especially exosomal miRNAs hold great promise as novel biomarkers for clinical diagnosis. Among ovarian cancer patients, cancer-derived exosomal miRNAs included miR-21, miR-141, miR-200a, miR- 200c, miR-200b, miR-203, miR-205, and miR-214 which exhibited significantly elevated levels as compared to benign disease. These results suggest that microRNA profiling of circulating tumor exosomes could potentially be used as surrogate diagnostic markers for ovarian cancer [42]. In melanoma, exosomal miRNAs included miR-17, miR-19a, miR-21, miR-126, and miR-149 and expressed higher levels in patients with metastatic sporadic melanoma as compared with familial melanoma patients or unaffected control subjects This indicates that distinct exosomal miRNAs may play important roles in tumor progression and metastasis, and may be used as predictive biomarkers to monitor remission as well as relapse following therapeutic intervention [52]. In addition, based on high-throughput next generation sequencing data, the exosomal miRNA signature is potentially well-suited to serve as a peripheral screening tool for Alzheimer’s disease [37,53].

Cell proliferation

Increasing evidence shows that cancer-derived exosomes contain miRNAs, which regulate the proliferation of recipient cells. In indolent papillary thyroid cancer (PTC), PTC-derived exosomes contain miR-146b and miR-222, which alter proliferation of other cells in a complex manner [54]. In nasopharyngeal carcinoma (NPC), NPC sera and NPC-cell derived exosomes commonly contain miR-24-3p, miR-891a, miR106a-5p, miR-20a-5p, and miR-1908. These over-expressed miRNA clusters down-regulate the MARK1 signaling pathway to alter cell proliferation and differentiation [55]. In pediatric cancer neuroblastoma (NB), using ultracentrifugation and exosome precipitation procedure, isolated exosomes from MYCN-amplified NB exhibit highly expressed miRNAs which are associated with a range of cellular and molecular functions related to cell growth and cell death [56].

Resistance to chemotherapy

Development of chemoresistance is a persistent problem during the treatment of local and disseminated disease. In chronic myeloid leukemia (CML), miRNAs can act as oncogenes or tumor suppressor genes which then contribute to the pathogenesis, disease progression, and resistance to therapy [57]. For example, several miRNAs including miR-146, miR-138 and mir-30a, miR- 203 are involved in imatinib treatment related resistance, [58-61] suggesting that the potential use of these small RNAs as therapeutic targets holds new opportunities in the treatment of CML . Another recent experiment showed that cross-talk between NB cells and human monocytes by exosomal miR-21 and miR-155 plays a crucial role in chemotherapy resistance [61]. This study demonstrated that NB cells secrete exosomic miR-21 transferred to human monocytes. The miR-21 is able to bind to the Toll-Like Receptor 8 in the recipient cells of human monocytes leading to increased expression of miR-155. Then, the monocytes secrete exosomal miR-155 transferred to NB cells leading to decreased Telomeric Repeat Binding Factor (NIMA-Interacting) 1 (TERF1, an inhibitor of telomerase) which is associated with chemoresistance in NB [62]. Exosomic miR-155-mediated crosstalk between human monocytes and NB cells increases the telomerase activity and confers the drug resistance to NB cells in response to cisplatin treatment. These data demonstrate that the miR-21/Mir-155/TERF1 circuitry contributes to chemoresistance in NB [61].

Metabolism

In breast cancer, miRNA from exosomes have been implicated in metabolism and metabolic disorders [63]. Breast cancer-secreted miR-122 via exosomes reprograms glucose metabolism in the premetastatic niche to promote metastasis. In vivo inhibition of miR- 122 restores glucose uptake in distant organs such as brain and lungs, and decreases the incidence of metastasis. These results demonstrate that miR-122 from CCEs are able to reprogram systemic metabolism in the facilitation of disease progression [63].

Angiogenesis

Exosomal miRNA transfer is believed to be involved in angiogenesis. In blood vessels, EV transfer of miRNAs modulates atherosclerosis and angiogenesis [64]. Several studies demonstrate the roles of miRNAs in activating cellular changes and modulating angiogenesis via the shuttling of miRNAs from other cells into endothelial cells (ECs). The human monocytic cell line, THP-1 is known to have abundant levels of miR-150, whereas miR-150 is low to absent in ECs. miR-150 transfers from THP-1 monocytes via EVs into ECs resulting in significantly elevated miR- 150 levels in ECs. Subsequently, protein levels of miR-150 target c-Myb are decreased in ECs resulting in enhanced cell migration [65,66]. One of the major hallmarks of cancerous cells lies in their ability to grow tumors and generate their own vasculature; an essential element in disease progression. It becomes clear that cancer derived EV can exert complex effects on ECs, their progenitors and on supporting cells; thereby, contributing to vessel formation within tumors. For example, Tspan8 is expressed in pancreatic cancer cells, and exhibits characteristics of promoting angiogenesis [67]. Tspan8 is involved in ECs and cancer cell EV interaction [68]. Subsequently, EV uptake by ECs elevated expression levels of pro-angiogenesis related factors to enhance angiogenesis [68]. In multiple myeloma (MM), the massive proliferation of plasma cells causes hypoxia. The hypoxia-resistant MM cells (HR-MM) produced more exosomes than the parental cells under normoxia or acute hypoxia conditions. Furthermore, HR-MM derived exosomes exhibit high levels of miR-135, which directly suppressed its target factor-inhibiting hypoxia-inducible factor 1 (FIH-1) in ECs, leading to enhanced endothelial tube formation under hypoxia via the HIF-FIH signaling pathway [69]. These experiments indicate that exosome-derived miRNAs from various tumors/cancers target surrounding or distant cells, ultimately changing the recipient cell’s function.

Invasion/Metastasis

Cancer biology is tightly regulated by cell-to-cell interaction. It is believed that initiation and progression of cancer is tightly regulated by tumor-associated stroma, which consists of extracellular matrix components and several cell types, including cancer-associated fibroblasts (CAF), immune cells, vascular cells, and bone marrow–derived cells [70]. It has been shown that fibroblasts secrete exosomes that promote breast cancer cells (BCCs) protrusive activity, motility, and metastasis by activating autocrine Wnt-PCP signaling in BCCs [71].

Epithelial-to-mesenchymal transition (EMT) is a process by which epithelial cells lose their cell polarity and cell-cell adhesion, and gain migratory and invasive properties. EMT has been implicated in the initiation of metastasis for cancer progression. In bladder cancer, the cancer-derived exosomes are capable of decreasing expression of epithelial markers E-cadherin and β-catenin, and increasing the migration and invasion of urothelial cells [72]. These studies suggest the important role of exosomes in the invasiveness and metastasis of disease.

The dynamic and reciprocal cross-talk between metastatic cells and their microenvironment during the adaptive metastatic outgrowth has recently been demonstrated via EV-derived miRNA [73]. In brain, astrocyte-derived exosomes mediate an intercellular transfer of PTEN-targeting microRNAs (miR-19a in miR-17~92 cluster play a major role in the down regulation of PTEN) to metastatic tumor cells, while astrocyte-specific depletion of PTEN-targeting microRNAs or blockade of astrocyte exosome secretion rescues PTEN loss and suppresses brain metastasis. In addition, this adaptive PTEN loss in brain metastatic tumor cells leads to an increased secretion of the chemokine CCL2, which recruits IBA1-expressing myeloid cells that reciprocally enhance the outgrowth of brain metastatic tumor cells via increased proliferation and decreased apoptosis [73]. These data suggest that exosomal miRNAs may play an essential role in the dynamic interaction between metastatic tumor cells and extrinsic signals at individual metastatic organ sites which significantly contribute to subsequent outgrowth.

Role of miRNAs in stem cell derived exosomes

Stem cells in cancer/tumor: Stem cells are undifferentiated biological cells that can differentiate into specialized cells, and divide to produce more stem cells. Normal stem cells (NSCs) have two main defining properties. First, they can renew themselves, which allows self-perpetuation and maintenance of a pool of totipotent stem cells. Self-renewal can occur by means of symmetric mitosis in which a stem cell produces two daughter stem cells, or asymmetric division in which a stem cell produces a daughter stem cell and another cell that is committed to a certain line of differentiation [74]. Second, NSCs can differentiate into multiple lineages, thus replacing and maintaining major functional elements that characterize surrounding tissue [75] (Table1).

Cancer stem cells (CSCs) are cancerous cells (found within tumors or hematological cancers) that possess characteristics associated with normal stem cells, specifically the ability to give rise to all cell types found in a particular cancer sample. CSCs are tumorigenic and may generate tumors through stem cell processes of self-renewal and differentiation into multiple cell types. CSCs are small cancerous cell populations with stem-like properties including cell proliferation, multiple differentiation and tumor initiation capacities. CSCs are therapy-resistant and cause cancer metastasis and recurrence. One recurrent key issue in cancer therapy lies in the targeting and elimination of CSCs, in order to cure cancer completely without relapse and metastasis [76]. To target CSCs, miRNAs are considered CSC markers.

Conclusive evidence for CSCs was first discovered by isolating a subpopulation of leukaemic cells that expressed the specific surface marker CD34, but lacked the CD38 marker [77]. The CD34+/CD38 subpopulation is capable of initiating tumors in NOD/SCID mice that are histologically similar to the donor. Later on, CSCs and tumor stem cells (TSCs) from various types of cancer/tumor were identified including breast cancer, ovarian cancer, prostate adenocarcinomas, brain gliomas, lung cancer, colorectal carcinomas, melanoma, and fibroids [78-86]. Recently, our group isolated and characterized myometrial and fibroid stem cells using dual Stro-1/CD44 surface markers and demonstrated the role of these stem cells in the pathogenesis of uterine fibroids [87].

Exosomal miRNAs in cell proliferation, migration and apoptosis: Tumor-stroma interaction is critical for carcinogenesis and cancer progression. Recently published studies have demonstrated that exosomes from stem cells induce proliferation and migration in several types of cells [88,89]. Cancer-derived MSCs exhibit aberrant miRNA expression patterns. For instance, gastric cancerassociated MSCs (GC-MSCs) and their non-cancerous tissue MSCs (GCN-MSCs) exhibit a different miRNA profile. Isolated exosomes from GC-MSCs exhibited a higher content of miR-221, were instantly internalized by gastric cancer cells, and significantly promoted cell proliferation and migration [90].

Human mesenchymal stem cells (hMSCs) can act as stromal cells for solid tumors. For example, when hMSCs were localized, and integrated into tumor associated stroma, they were shown to promote tumor growth and angiogenesis through juxtacrine, paracrine and endocrine mechanisms [91-93]. A recent study on hMSCs [94] demonstrated that exosomes/EVs from serum deprived hMSCs act as carriers that transport tumor supportive miRNAs. Through a series of ultracentrifugation steps, isolated exosomes containing anti-apoptotic miR–21 and miR-34a [94] have been demonstrated to be involved in cell survival and proliferation [95,96]. In addition, these hMSC-derived EVs increase the overall survival of cancer cells under serum-deprived conditions.

Exosomal miRNAs in invasion/metastasis: MicroRNAs (miRNAs) have recently been recognized as targets for anti-metastatic therapy against cancer malignancy [97]. Mesenchymal-epithelial transitions (MET), are integral steps of cell fate specification during gastrulation and organogenesis. MET occur during normal development, cancer metastasis, and induced pluripotent stem cell reprogramming. In cancer progression, reactivation of the MET program promotes tumor metastasis by driving tumor cell invasion and enhancing tumor cell survival during the metastatic cascade. MicroRNAs have emerged as key regulators of CSCs and MET. Among them, the miR-200 family plays a particularly important role in integrating the MET program and core stem cell pathways [98]. The overexpression of miR-200 suppresses the clonogenicity of breast cancer stem cells (BCSCs) and the ability of multipotent mammary stem cells (MaSCs) to regenerate mammary ductal trees. On the other hand, inhibition of miR-200 increases the number of CSCs in breast cancers.

Another study focused on the role of miRNAs in metastasis used the co-culturing system of bone marrow-metastatic human breast cancer cell line (BM2) with human bone marrow mesenchymal stem cells (BM-MSCs). This study revealed that BM-MSCs suppressed the proliferation and invasion of BM2 cells [99]. Acquisition of these dormant phenotypes in BM2 cells was also observed by culturing the cells in BM-MSC-conditioned media or with exosomes from BM-MSC cultures. Although various miRNAs exhibited increased expression in BM-MSC-derived exosomes as compared with those from adult fibroblasts, overexpression of miR-23b in BM2 cells induced dormant phenotypes through the suppression of the target gene, MARCKS. This latter target gene encodes a protein that promotes cell cycling and motility [99]. These studies suggest that exosomal miRNAs from BM-MSCs may promote breast cancer cell dormancy in a metastatic niche, and EV-mediated communication has a major influence on key aspects of cancer progression. Future directions could include studies which focus on developing a thorough understanding as to whether EV-derived miRNA profiling from metastatic CSCs differs from those of CSCs in a primary site.

miRNAs from stem cells in response to environmental exposure Exosome production and content may be influenced by molecular signals, depending on the origin of the cells. The origin of CSCs is still an area of ongoing investigation. In brief, CSCs can be generated as mutants of developing stem cells, adult stem cells, or differentiated cells that require stem-like attributes.

Alternately, exosome production and content may be altered in response to adverse environmental exposure. For example, tumor cells exposed to hypoxia secrete exosomes with enhanced angiogenic and metastatic potential, suggesting that tumor cells adapt to a hypoxic microenvironment by secreting exosomes to stimulate angiogenesis or facilitate metastasis in a more favorable environment [100,101].

Abnormal environmental exposures have been shown to be involved in many diseases [102,103]. Since the involvement of miRNAs and tumor-initiating cells/progenitor cells play a crucial role in the development of tumors and many other diseases, the environmental exposure can impact stem cell regulatory networks by modulating the steady-state levels of miRNAs. Therefore, miRNA changes may be sensitive indicators of the effects of acute and chronic environmental exposure. To understand how colonic stem cell populations respond to environmental factors such as diet and carcinogens, Shah et al, determined the effects of the chemoprotective fish oil/pectin diet on miRNAs and mRNAs in colonic stem cells obtained from Lgr5-EGFP-IRES-creER knock-in mice [104]. Following global miRNA profiling, 26 miRNAs (P<0.05) were differentially expressed in Lgr5 (high) stem cells as compared to Lgr5 (negative) differentiated cells. Fish oil/pectin treatment up-regulated miR-19b, miR-26b and miR-203 expression as compared to corn oil plus cellulose (CCA) specifically in Lgr5 (high) cells. They further demonstrated that only miR-19b and its indirect target PTK2B were modulated by the fish oil/pectin diet in Lgr5 (negative) cells [104].

In a separate observation from another animal diet model, 8-week-old mothers of 1-day old rat pups were fed diets containing deficient or enriched amounts of n-3 polyunsaturated fatty acids (n-3 PUFAs) from two weeks before breeding up until delivery. The results indicated that rat neural stem cells/neural progenitors (NSC) proliferation and differentiation were dually altered by the in utero polyunsaturated fatty acid supply, along with marked alterations in mRNA and miRNA expression [105]. Therefore, fetal exposure to n-3 PUFA deficient diet altered NSC characteristics, and reprogramed expression patterns of mRNAs and miRNAs. However the link between miRNA and exosomes in stem cells in response to adverse environmental exposure requires further investigation.

Conclusion

Exosomes naturally carry miRNAs; therefore, they are used as carriers to deliver miRNAs in several therapeutic applications. A number of studies have shown that exosomes can deliver tissue-targeted siRNA and miRNAs to modulate gene expression pattern within target cells [106-108]. Moreover, several clinical trials on exosome-based therapies for cancer treatment are being conducted [109-111].

Although great strides have been made in understanding the role of miRNAs, and we now know that their associated networks play in a variety of biological progress and diseases, there still remains limited information regarding how the interaction between stem cells and their surrounding differentiated cells happens through EV derived miRNAs, therefore alters the transcriptome in recipient cells leading to cancer/tumor progression. Further studies focused on the interplay between gene expression reprogramming and stem cell features via exosomes will augment our understanding and identification of critical exosomal miRNA targets and related events. Considering the reversibility of epigenetic alterations as well as the pivotal role they play in early carcinogenesis and other diseases, reversion of these alterations could be a promising approach for providing novel therapeutic targets for treatment of a variety of diseases.

Acknowledgement

This work was supported in part by an Augusta University Startup package, the National Institutes of Health grant HD04622811 (to AA), and the Augusta University Intramural Grants Program (QY). We would like to thank Walidah Walker, MPH for editing this manuscript.

Conflict of Interest

None of the authors have a financial relationship with a commercial entity that has an interest in the subject of this manuscript.

References

  1. Chen K, Rajewsky N (2007) The evolution of gene regulation by transcription factors and microRNAs. Nature reviews Genetics 8:93-103.
  2. Yang Q, Sun M (2014) Role of MicroRNAs in Hypoxia-Induced Pulmonary Hypertension. Cardiol Pharmacol 3:e124.
  3. Jansson MD, Lund AH (2012) MicroRNA and cancer. Molecular oncology 6:590-610.
  4. Winter J, Jung S, Keller S, Gregory RI, Diederichs S (2009) Many roads to maturity: microRNA biogenesis pathways and their regulation. Nature cell biology 11:228-234.
  5. Bartel DP (2009) MicroRNAs: target recognition and regulatory functions. Cell 136:215-233.
  6. Kusenda B, Mraz M, Mayer J, Pospisilova S (2006) MicroRNA biogenesis, functionality and cancer relevance. Biomedical papers of the Medical Faculty of the University Palacky, Olomouc, Czechoslovakia 150:205-215.
  7. Bentwich I, Avniel A, Karov Y, Aharonov R, Gilad S,et al. (2005)Identification of hundreds of conserved and nonconserved human microRNAs. Nature genetics 37:766-770.
  8. Bentwich I (2005) Prediction and validation of microRNAs and their targets. FEBS letters 579:5904-5910.
  9. Kozomara A, Griffiths-Jones S (2014) miRBase: annotating high confidence microRNAs using deep sequencing data. Nucleic acids research 42:D68-73.
  10. Lewis BP, Burge CB, Bartel DP (2005) Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 120:15-20.
  11. Friedman RC, Farh KK, Burge CB, Bartel DP (2009) Most mammalian mRNAs are conserved targets of microRNAs. Genome research 19:92-105.
  12. Lee RC, Feinbaum RL, Ambros V (1993) The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 75:843-854.
  13. Lagos-Quintana M, Rauhut R, Lendeckel W, Tuschl T (2001) Identification of novel genes coding for small expressed RNAs. Science 294:853-858.
  14. Lau NC, Lim LP, Weinstein EG, Bartel DP (2001) An abundant class of tiny RNAs with probable regulatory roles in Caenorhabditis elegans. Science 294:858-862.
  15. Lee RC, Ambros V (2001) An extensive class of small RNAs in Caenorhabditis elegans. Science 294:862-864.
  16. Lim LP, Glasner ME, Yekta S, Burge CB, Bartel DP (2003) Vertebrate microRNA genes. Science 299:1540.
  17. Reinhart BJ, Weinstein EG, Rhoades MW, Bartel B, Bartel DP (2002) MicroRNAs in plants. Genes & development 16:1616-1626.
  18. Pfeffer S, Zavolan M, Grasser FA, Chien M, Russo JJ et al. (2004). Identification of virus-encoded microRNAs. Science 304:734-736.
  19. Sullivan CS, Ganem D (2005) MicroRNAs and viral infection. Molecular cell 20:3-7.
  20. Lim LP, Lau NC, Garrett-Engele P, Grimson A, Schelter JM et al. (2005). Microarray analysis shows that some microRNAs downregulate large numbers of target mRNAs. Nature 433:769-773.
  21. Chen CZ, Li L, Lodish HF, Bartel DP (2004) MicroRNAs modulate hematopoietic lineage differentiation. Science 303:83-86.
  22. Brennecke J, Hipfner DR, Stark A, Russell RB, Cohen SM (2003) bantam encodes a developmentally regulated microRNA that controls cell proliferation and regulates the proapoptotic gene hid in Drosophila. Cell 113:25-36.
  23. Han M, Wang F, Gu Y, Pei X, Guo G, Yu C et al.(2016) MicroRNA-21 induces breast cancer cell invasion and migration by suppressing smad7 via EGF and TGF-beta pathways. Oncology reports 35:73-80.
  24. Huang K, Tang Y, He L, Dai Y (2016) MicroRNA-340 inhibits prostate cancer cell proliferation and metastasis by targeting the MDM2-p53 pathway. Oncology reports 35:887-895.
  25. Saha B, Momen-Heravi F, Kodys K, Szabo G (2016) MicroRNA Cargo of Extracellular Vesicles from Alcohol-exposed Monocytes Signals Naive Monocytes to Differentiate into M2 Macrophages. The Journal of biological chemistry 291:149-159.
  26. Calin GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, Noch E, Aldler H et al. (2002) Frequent deletions and down-regulation of micro- RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proceedings of the National Academy of Sciences of the United States of America 99:15524-15529.
  27. Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, et al. (2005) MicroRNA expression profiles classify human cancers. Nature 435:834-838.
  28. De Smaele E, Ferretti E, Gulino A (2010) MicroRNAs as biomarkers for CNS cancer and other disorders. Brain research 1338:100-111.
  29. Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK et al. (2008) Circulating microRNAs as stable blood-based markers for cancer detection. Proceedings of the National Academy of Sciences of the United States of America 105:10513-10518.
  30. Simpson RJ, Lim JW, Moritz RL, Mathivanan S (2009) Exosomes: proteomic insights and diagnostic potential. Expert review of proteomics 6:267-283.
  31. Matullo G, Naccarati A, Pardini B (2015) microRNA expression profiling in bladder cancer: The challenge of Next Generation Sequencing in tissues and biofluids. International journal of cancer Journal international du cancer.
  32. Yang Q, Mas A, Diamond MP, Al-Hendy A (2015) The Mechanism and Function of Epigenetics in Uterine Leiomyoma Development. Reprod Sci.
  33. van der Pol E, Boing AN, Harrison P, Sturk A, Nieuwland R (2012) Classification, functions, and clinical relevance of extracellular vesicles. Pharmacological reviews 64:676-705.
  34. Keller S, Sanderson MP, Stoeck A, Altevogt P (2006) Exosomes: from biogenesis and secretion to biological function. Immunology letters 107:102-108.
  35. Booth AM, Fang Y, Fallon JK, Yang JM, Hildreth JE, Gould SJ (2006) Exosomes and HIV Gag bud from endosome-like domains of the T cell plasma membrane. The Journal of cell biology 172:923-935.
  36. Penfornis P, Vallabhaneni KC, Whitt J, Pochampally R (2016) Extracellular vesicles as carriers of microRNA, proteins and lipids in tumor microenvironment. International journal of cancer Journal international du cancer 138:14-21.
  37. Van Giau V, An SS (2016) Emergence of exosomal miRNAs as a diagnostic biomarker for Alzheimer's disease. Journal of the neurological sciences 360:141-152.
  38. Mathivanan S, Fahner CJ, Reid GE, Simpson RJ (2012) ExoCarta 2012: database of exosomal proteins, RNA and lipids. Nucleic acids research 40:D1241-1244.
  39. Rauschenberger L, Staar D, Thom K, Scharf C, Venz S et al. (2015) Exosomal particles secreted by prostate cancer cells are potent mRNA and protein vehicles for the interference of tumor and tumor environment. The Prostate 76: 409-24.
  40. Kumari N, Saxena S, Agrawal U (2015) Exosomal protein interactors as emerging therapeutic targets in urothelial bladder cancer. Journal of the Egyptian National Cancer Institute 27:51-58.
  41. Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ,et al. (2007) Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nature cell biology 9:654-659.
  42. Taylor DD, Gercel-Taylor C (2008) MicroRNA signatures of tumor-derived exosomes as diagnostic biomarkers of ovarian cancer. Gynecologic oncology 110:13-21.
  43. Rabinowits G, Gercel-Taylor C, Day JM, Taylor DD, Kloecker GH (2009) Exosomal microRNA: a diagnostic marker for lung cancer. Clinical lung cancer 10:42-46.
  44. Zeringer E, Barta T, Li M, Vlassov AV (2015) Strategies for isolation of exosomes. Cold Spring Harbor protocols 2015:319-323.
  45. Khan M, Nickoloff E, Abramova T, Johnson J, Verma SK,et al. (2015) Embryonic stem cell-derived exosomes promote endogenous repair mechanisms and enhance cardiac function following myocardial infarction. Circulation research 117:52-64.
  46. Ailawadi S, Wang X, Gu H, Fan GC (2015) Pathologic function and therapeutic potential of exosomes in cardiovascular disease. Biochimica et biophysica acta 1852:1-11.
  47. Pulliam L, Gupta A (2015) Modulation of cellular function through immune-activated exosomes. DNA and cell biology 34:459-463.
  48. Zoller M (2016) Exosomes in Cancer Disease. Methods Mol Biol 1381:111-149.
  49. Brinton LT, Sloane HS, Kester M, Kelly KA (2015) Formation and role of exosomes in cancer. Cellular and molecular life sciences : CMLS 72:659-671.
  50. Lowry MC, Gallagher WM, O'Driscoll L (2015) The Role of Exosomes in Breast Cancer. Clinical chemistry 61:1457-1465.
  51. Overbye A, Skotland T, Koehler CJ, Thiede B, Seierstad T,et al. (2015) Identification of prostate cancer biomarkers in urinary exosomes. Oncotarget 6:30357-30376.
  52. Pfeffer SR, Grossmann KF, Cassidy PB, Yang CH, Fan M, et al. (2015) Detection of Exosomal miRNAs in the Plasma of Melanoma Patients. Journal of clinical medicine 4:2012-2027.
  53. Cheng L, Quek CY, Sun X, Bellingham SA, Hill AF (2013) The detection of microRNA associated with Alzheimer's disease in biological fluids using next-generation sequencing technologies. Frontiers in genetics 4:150.
  54. Lee JC, Zhao JT, Gundara J, Serpell J, Bach LA, Sidhu S (2015) Papillary thyroid cancer-derived exosomes contain miRNA-146b and miRNA-222. The Journal of surgical research 196:39-48.
  55. Ye SB, Li ZL, Luo DH, Huang BJ, Chen YS, et al. (2014) Tumor-derived exosomes promote tumor progression and T-cell dysfunction through the regulation of enriched exosomal microRNAs in human nasopharyngeal carcinoma. Oncotarget 5:5439-5452.
  56. Haug BH, Hald OH, Utnes P, Roth SA, Lokke C, et al. (2015) Exosome-like Extracellular Vesicles from MYCN-amplified Neuroblastoma Cells Contain Oncogenic miRNAs. Anticancer research 35:2521-2530.
  57. Di Stefano C, Mirone G, Perna S, Marfe G (2016) The roles of microRNAs in the pathogenesis and drug resistance of chronic myelogenous leukemia (Review). Oncology reports 35:614-624.
  58. Taganov KD, Boldin MP, Chang KJ, Baltimore D (2006) NF-kappaB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proceedings of the National Academy of Sciences of the United States of America 103:12481-12486.
  59. Duncan EA, Goetz CA, Stein SJ, Mayo KJ, Skaggs BJ, et al. (2008) IkappaB kinase beta inhibition induces cell death in Imatinib-resistant and T315I Dasatinib-resistant BCR-ABL+ cells. Molecular cancer therapeutics 7:391-397.
  60. Yu Y, Yang L, Zhao M, Zhu S, Kang R, et al. (2012) Targeting microRNA-30a-mediated autophagy enhances imatinib activity against human chronic myeloid leukemia cells. Leukemia : official journal of the Leukemia Society of America, Leukemia Research Fund, UK 26:1752-1760.
  61. Challagundla KB, Wise PM, Neviani P, Chava H, Murtadha M, et al. (2015) Exosome-mediated transfer of microRNAs within the tumor microenvironment and neuroblastoma resistance to chemotherapy. Journal of the National Cancer Institute 107.
  62. Wesbuer S, Lanvers-Kaminsky C, Duran-Seuberth I, Bolling T, et al. (2010) Association of telomerase activity with radio- and chemosensitivity of neuroblastomas. Radiat Oncol 5:66.
  63. Fong MY, Zhou W, Liu L, Alontaga AY, Chandra M,et al. (2015) Breast-cancer-secreted miR-122 reprograms glucose metabolism in premetastatic niche to promote metastasis. Nature cell biology 17:183-194.
  64. Mocharla P, Briand S, Giannotti G, Dorries C, Jakob P, et al. (2013) AngiomiR-126 expression and secretion from circulating CD34(+) and CD14(+) PBMCs: role for proangiogenic effects and alterations in type 2 diabetics. Blood 121:226-236.
  65. Zhang Y, Liu D, Chen X, Li J, Li L, et al. (2010) Secreted monocytic miR-150 enhances targeted endothelial cell migration. Molecular cell 39:133-144.
  66. Das S, Halushka MK (2015) Extracellular vesicle microRNA transfer in cardiovascular disease. Cardiovascular pathology : the official journal of the Society for Cardiovascular Pathology 24:199-206.
  67. Gesierich S, Berezovskiy I, Ryschich E, Zoller M (2006) Systemic induction of the angiogenesis switch by the tetraspanin D6.1A/CO-029. Cancer research 66:7083-7094.
  68. Nazarenko I, Rana S, Baumann A, McAlear J, Hellwig A, et al. (2010) Cell surface tetraspanin Tspan8 contributes to molecular pathways of exosome-induced endothelial cell activation. Cancer research 70:1668-1678.
  69. Umezu T, Tadokoro H, Azuma K, Yoshizawa S, Ohyashiki K, et al. (2014) Exosomal miR-135b shed from hypoxic multiple myeloma cells enhances angiogenesis by targeting factor-inhibiting HIF-1. Blood 124:3748-3757.
  70. Bissell MJ, Hines WC (2011) Why don't we get more cancer? A proposed role of the microenvironment in restraining cancer progression. Nature medicine 17:320-329.
  71. Luga V, Wrana JL (2013) Tumor-stroma interaction: Revealing fibroblast-secreted exosomes as potent regulators of Wnt-planar cell polarity signaling in cancer metastasis. Cancer research 73:6843-6847.
  72. Franzen CA, Blackwell RH, Todorovic V, Greco KA, Foreman KE, et al. (2015) Urothelial cells undergo epithelial-to-mesenchymal transition after exposure to muscle invasive bladder cancer exosomes. Oncogenesis 4:e163.
  73. Zhang L, Zhang S, Yao J, Lowery FJ, Zhang Q,et al. (2015) Microenvironment-induced PTEN loss by exosomal microRNA primes brain metastasis outgrowth. Nature 527:100-104.
  74. Reya T, Morrison SJ, Clarke MF, Weissman IL (2001) Stem cells, cancer, and cancer stem cells. Nature 414:105-111.
  75. Kakarala M, Wicha MS (2008) Implications of the cancer stem-cell hypothesis for breast cancer prevention and therapy. Journal of clinical oncology: official journal of the American Society of Clinical Oncology 26:2813-2820.
  76. Baek SJ, Ishii H, Tamari K, Hayashi K, Nishida N, et al. (2015) Cancer stem cells: The potential of carbon ion beam radiation and new radiosensitizers (Review). Oncology reports 34:2233-2237.
  77. Bonnet D, Dick JE (1997) Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nature medicine 3:730-737.
  78. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF (2003) Prospective identification of tumorigenic breast cancer cells. Proceedings of the National Academy of Sciences of the United States of America 100:3983-3988.
  79. Dontu G, Al-Hajj M, Abdallah WM, Clarke MF, Wicha MS (2003) Stem cells in normal breast development and breast cancer. Cell proliferation 36:59-72.
  80. O'Brien CA, Pollett A, Gallinger S, Dick JE (2007) A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature 445:106-110.
  81. Piccirillo SG, Reynolds BA, Zanetti N, Lamorte G, Binda E, et al. (2006) Bone morphogenetic proteins inhibit the tumorigenic potential of human brain tumour-initiating cells. Nature 444:761-765.
  82. Fang D, Nguyen TK, Leishear K, Finko R, Kulp AN, et al. (2005) A tumorigenic subpopulation with stem cell properties in melanomas. Cancer research 65:9328-9337.
  83. Kim CF, Jackson EL, Woolfenden AE, Lawrence S, Babar I, et al. (2005) Identification of bronchioalveolar stem cells in normal lung and lung cancer. Cell 121:823-835.
  84. Ono M, Maruyama T, Masuda H, Kajitani T, Nagashima T, et al. (2007) Side population in human uterine myometrium displays phenotypic and functional characteristics of myometrial stem cells. Proceedings of the National Academy of Sciences of the United States of America 104:18700-18705.
  85. Mas A, Cervello I, Gil-Sanchis C, Faus A, Ferro J, et al. (2012) Identification and characterization of the human leiomyoma side population as putative tumor-initiating cells. Fertility and sterility 98:741-751 e746.
  86. Zhang S, Balch C, Chan MW, Lai HC, Matei D, et al. (2008) Identification and characterization of ovarian cancer-initiating cells from primary human tumors. Cancer research 68:4311-4320.
  87. Mas A, Nair S, Laknaur A, Simon C, Diamond MP, Al-Hendy A (2015) Stro-1/CD44 as putative human myometrial and fibroid stem cell markers. Fertility and sterility.
  88. Shabbir A, Cox A, Rodriguez-Menocal L, Salgado M, Van Badiavas E (2015) Mesenchymal Stem Cell Exosomes Induce Proliferation and Migration of Normal and Chronic Wound Fibroblasts, and Enhance Angiogenesis In Vitro. Stem cells and development 24:1635-1647.
  89. Shi J, Ren Y, Zhen L, Qiu X (2015) Exosomes from breast cancer cells stimulate proliferation and inhibit apoptosis of CD133+ cancer cells in vitro.Mol Med Rep 11:405-409.
  90. Wang M, Zhao C, Shi H, Zhang B, Zhang L, et al. (2014) Deregulated microRNAs in gastric cancer tissue-derived mesenchymal stem cells: novel biomarkers and a mechanism for gastric cancer. British journal of cancer 110:1199-1210.
  91. Karnoub AE, Dash AB, Vo AP, Sullivan A, Brooks MW, Bell GW, Richardson AL (2007) Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature 449:557-563.
  92. Ljujic B, Milovanovic M, Volarevic V, Murray B, Bugarski D (2013) Human mesenchymal stem cells creating an immunosuppressive environment and promote breast cancer in mice. Scientific reports 3:2298.
  93. Huang WH, Chang MC, Tsai KS, Hung MC, Chen HL, Hung SC (2013) Mesenchymal stem cells promote growth and angiogenesis of tumors in mice. Oncogene 32:4343-4354.
  94. Vallabhaneni KC, Penfornis P, Dhule S, Guillonneau F, Adams KV (2015) Extracellular vesicles from bone marrow mesenchymal stem/stromal cells transport tumor regulatory microRNA, proteins, and metabolites. Oncotarget 6:4953-4967.
  95. Ma Y, Bao-Han W, Lv X, Su Y, Zhao X, Yin Y, Zhang X, Zhou Z (2013) MicroRNA-34a mediates the autocrine signaling of PAR2-activating proteinase and its role in colonic cancer cell proliferation. PloS one 8:e72383.
  96. Frankel LB, Christoffersen NR, Jacobsen A, Lindow M, Krogh A, Lund AH (2008) Programmed cell death 4 (PDCD4) is an important functional target of the microRNA miR-21 in breast cancer cells. The Journal of biological chemistry 283:1026-1033.
  97. Wu X, Liu T, Fang O, Dong W, Zhang F, Leach L, Hu X, et al. (2015) MicroRNA-708-5p acts as a therapeutic agent against metastatic lung cancer. Oncotarget 14.
  98. Shimono Y, Zabala M, Cho RW, Lobo N, Dalerba P, Qian D, Diehn M (2009) Downregulation of miRNA-200c links breast cancer stem cells with normal stem cells. Cell 138:592-603.
  99. Ono M, Kosaka N, Tominaga N, Yoshioka Y, Takeshita F, Takahashi RU (2014) Exosomes from bone marrow mesenchymal stem cells contain a microRNA that promotes dormancy in metastatic breast cancer cells. Science signaling 7:ra63.
  100. Park JE, Tan HS, Datta A, Lai RC, Zhang H, Meng W, Lim SK, Sze SK (2010) Hypoxic tumor cell modulates its microenvironment to enhance angiogenic and metastatic potential by secretion of proteins and exosomes. Molecular & cellular proteomics: MCP 9:1085-1099.
  101. Tadokoro H, Umezu T, Ohyashiki K, Hirano T, Ohyashiki JH (2013) Exosomes derived from hypoxic leukemia cells enhance tube formation in endothelial cells. The Journal of biological chemistry 288:34343-34351.
  102. Walker CL, Ho SM (2012) Developmental reprogramming of cancer susceptibility. Nature reviews Cancer 12:479-486.
  103. Yang Q, Sun M, Ramchandran R, Raj JU (2015) IGF-1 signaling in neonatal hypoxia-induced pulmonary hypertension: Role of epigenetic regulation. Vascular pharmacology.
  104. Shah MS, Kim E, Davidson LA, Knight JM, Zoh RS, Goldsby JS (2016) Comparative effects of diet and carcinogen on microRNA expression in the stem cell niche of the mouse colonic crypt. Biochimica et biophysica acta 1862:121-134.
  105. Goustard-Langelier B, Koch M, Lavialle M, Heberden C (2013) Rat neural stem cell proliferation and differentiation are durably altered by the in utero polyunsaturated fatty acid supply. The Journal of nutritional biochemistry 24:380-387.
  106. Wahlgren J, De LKT, Brisslert M, Vaziri Sani F, Telemo E, et al. (2012) Plasma exosomes can deliver exogenous short interfering RNA to monocytes and lymphocytes. Nucleic acids research 40:e130.
  107. S ELA, Mager I, Breakefield XO, Wood MJ (2013) Extracellular vesicles: biology and emerging therapeutic opportunities. Nature reviews Drug discovery 12:347-357.
  108. Tickner JA, Urquhart AJ, Stephenson SA, Richard DJ, O'Byrne KJ (2014) Functions and therapeutic roles of exosomes in cancer. Frontiers in oncology 4:127.
  109. Dai S, Wei D, Wu Z, Zhou X, Wei X, Huang H, Li G (2008) Phase I clinical trial of autologous ascites-derived exosomes combined with GM-CSF for colorectal cancer. Molecular therapy : the journal of the American Society of Gene Therapy 16:782-790.
  110. Morse MA, Garst J, Osada T, Khan S, Hobeika A, Clay TM, Valente N, et al. (2005) A phase I study of dexosome immunotherapy in patients with advanced non-small cell lung cancer. Journal of translational medicine 3:9.
  111. Johnsen KB, Gudbergsson JM, Skov MN, Pilgaard L, Moos T, Duroux M (2014) A comprehensive overview of exosomes as drug delivery vehicles - endogenous nanocarriers for targeted cancer therapy. Biochimica et biophysica acta 1846:75-87.