Next Article in Journal
Pancreatic Cancer Cells Undergo Immunogenic Cell Death upon Exposure to Gas Plasma-Oxidized Ringers Lactate
Next Article in Special Issue
PIWI-RNAs Small Noncoding RNAs with Smart Functions: Potential Theranostic Applications in Cancer
Previous Article in Journal
Epidemiology and Characteristics of Gastric Carcinoma in Childhood—An Analysis of Data from Population-Based and Clinical Cancer Registries
Previous Article in Special Issue
Non-Coding RNAs in Airway Diseases: A Brief Overview of Recent Data
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Modulating Effects of Cancer-Derived Exosomal miRNAs and Exosomal Processing by Natural Products

1
Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
2
School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
3
Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaoshiung Medical University, Kaohsiung 80708, Taiwan
4
Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
5
School of Dentistry, Taipei Medical University, Taipei 11031, Taiwan
6
Department of Oral and Maxillofacial Surgery, Chi-Mei Medical Center, Tainan 71004, Taiwan
7
Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
8
Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
9
Institute of Biomedical and Genetic Engineering (IBGE), Islamabad 54000, Pakistan
10
Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
11
Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2023, 15(1), 318; https://doi.org/10.3390/cancers15010318
Submission received: 23 November 2022 / Revised: 28 December 2022 / Accepted: 29 December 2022 / Published: 3 January 2023

Abstract

:

Simple Summary

Cancer cells generate exosomes (extracellular vesicles) to regulate many cell functions for tumor progression. Many exosome-modulating clinical drugs have been developed for effective cancer therapy, but the functions and exosome processing (secretion and assembly) modulation by natural products are not well understood. In this review, we fill the gaps between natural products-modulated miRNAs and exosome-processing by the target gene prediction of the bioinformatics database. The cancer-derived exosomal miRNAs and their exosome processing and modulated cell functions by natural products are well organized. Consequently, this review provides a comprehensive and potential modulating mechanism and targets for exosome processing and cancer cell functions for natural products.

Abstract

Cancer-derived exosomes exhibit sophisticated functions, such as proliferation, apoptosis, migration, resistance, and tumor microenvironment changes. Several clinical drugs modulate these exosome functions, but the impacts of natural products are not well understood. Exosome functions are regulated by exosome processing, such as secretion and assembly. The modulation of these exosome-processing genes can exert the anticancer and precancer effects of cancer-derived exosomes. This review focuses on the cancer-derived exosomal miRNAs that regulate exosome processing, acting on the natural-product-modulating cell functions of cancer cells. However, the role of exosomal processing has been overlooked in several studies of exosomal miRNAs and natural products. In this study, utilizing the bioinformatics database (miRDB), the exosome-processing genes of natural-product-modulated exosomal miRNAs were predicted. Consequently, several natural drugs that modulate exosome processing and exosomal miRNAs and regulate cancer cell functions are described here. This review sheds light on and improves our understanding of the modulating effects of exosomal miRNAs and their potential exosomal processing targets on anticancer treatments based on the use of natural products.

1. Introduction

Exosomes are extracellular vesicles of 30–100 nm in size that are secreted by both cancer and normal cells [1,2]. Cancer cells secrete more abundant and complex compositions in exosomes than normal cells. Cancer-derived exosomes exhibit diverse functions in regulating proliferation, migration, invasion, metastasis, drug resistance, inflammation, and immune responses [1,3,4].
The general structure and biogenesis of exosomes are shown in Figure 1 [5,6,7,8,9,10,11,12,13]. Many proteins, lipids, DNAs, mRNAs, and non-coding RNAs (circular RNAs, long-noncoding RNAs, and microRNAs (miRNAs)) exist in exosomes [1,2,3]. In general, the membrane proteins of exosomes include major histocompatibility complex (MHC)-I, MHC-II, flotillin, tetraspanins (CD9, CD82, CD81, and CD63), cell adhesion molecules (CAMs), integrins, and transmembrane proteins. The soluble proteins of exosomes include TSG101, heat shock protein 70 (Hsp70), Hsp90, and protein kinase B (AKT). The lipid rafts of exosomes include ceramides, sphingolipids, and cholesterol [5,6,7,8,9,10,11,12,13]. The process of exosome biogenesis starts with the initiation of endocytosis, early endosome, late endosome, and multivesicular body (MVB) formation, plasma membrane fusion, and release by exocytosis [5,6,7,8,9,10,11,12,13]. Exosome biogenesis consists of two stages, namely exosomal assembly and secretion. Many exosomal components (DNA, RNA, and proteins) are uploaded during exosome biogenesis.
miRNAs are a group of small non-coding RNAs of 21–25 nucleotides in size. miRNAs can modulate gene expression by inhibiting mRNA translation or improving the mRNA degradation of target genes [14]. By binding to the 3′-untranslated regions (UTR) of target genes, miRNAs can knock down target gene expressions to assess their diverse functions.
Among the non-coding RNAs, this review focuses only on miRNAs, particularly exosomal miRNAs. miRNA uptake into exosomes is not a random but selective process involving secretion and transportation between exosome donors and receptors [15]. Exosomes play a vital role in regulating the development of oral [16,17], head/neck [18], breast [19], prostate [20], pancreatic [21], colon [22], gynecologic [23], liver [24], and myeloma cancer cells. In preclinical applications, exosomes are applied in diagnosis as several cancer biomarkers [25,26] and in cancer therapy using animal models [27,28,29,30]. Moreover, the exosomal miRNAs also function as modulators of drug resistance and cancer metastasis [25].
As mentioned above, exosomes and miRNAs have a close relationship in regulating cell functions. Recently, anticancer studies using natural products have shown progression in research involving exosomes and miRNAs. However, the potential impacts of exosomes and miRNAs on natural-product-regulating cancer cell functions lack systemic organization. The modulating effects of natural products on exosome biogenesis and exosomal miRNAs are discussed later, particularly in regard to their capacity for regulating exosomal processing (secretion and assembly). Moreover, some natural products and exosomal miRNAs show anticancer effects but lack investigation regarding their impacts on exosomal processing. This gap can be filled by utilizing the miRDB database [31], a bioinformatic tool which can predict the target genes of exosome processing by inputting natural-product-modulated exosomal miRNAs.
In the following review, we first explore the relationship between exosome processing (secretion and assembly) and natural products (Section 2), because the impact of exosome processing is rarely discussed in detail in the literature. Next, the prediction of the targeting of exosome-processing and AKT-signaling genes of exosome miRNAs is assessed (Section 3), because the contribution of exosome processing is rarely emphasized in the literature. The modulating effects of exosome production by natural products and their exosome delivery potential for cancer treatment (Section 4) are explored. Finally, the regulation of the cancer cell functions of natural-product-modulating miRNAs and exosomes (Section 5) is summarized (Figure 2). Consequently, this review sheds light on the organization of the relationship between exosomal processing and its related genes, exosomal miRNAs, cell functions, and natural products.

2. Exosome Processing (Secretion and Assembly) and Natural-Product-Modulated Cell Functions

2.1. Exosome Processing (Secretion and Assembly) Genes

Several extracellular biogenesis genes, including exosomal secretion and assembly, were summarized using the Mouse Genome Database in Gene Oncology (GO) functions http://www.informatics.jax.org/vocab/gene_ontology/GO:1990182 (retrieval date: 11 November 2022) [32]. Exosomal secretion begins with the fusion of the partial endosomal membrane of a multivesicular body (MVB) with the plasma membrane, and it ends with the release of membrane-bounded vesicles into the extracellular space (Figure 1). Three main functions of exosomal secretion are classified: multiple vesicular body fusion into the apical plasma membrane, the negative regulation of exosomal secretion, and the positive regulation of exosomal secretion. Furthermore, exosomal assembly is the process in which a set of components are incorporated, aggregated, and bonded to generate an extracellular vesicular exosome. Three main functions of exosomal assembly are classified: extracellular exosome assembly, the positive regulation of extracellular exosome assembly, and the negative regulation of extracellular exosome assembly.
For exosomal secretion, “multiple vesicular body fusion into the apical plasma membrane” includes four genes, including COP9 signalosome subunit 5 (COPS5), RAB11A, a member of the RAS oncogene family (RAB11A), RAB27A, a member of the RAS oncogene family (RAB27A), and STEAP family member 3 (STEAP3). Five genes are listed as modulators of “negative regulation of exosomal secretion”, including ATPase class II, type 9A (ATP9A), parkin RBR E3 ubiquitin protein ligase (PRKN), RAB7A, a member of the RAS oncogene family (RAB7A), RAB7B, a member of the RAS oncogene family (RAB7B), and vacuolar protein sorting 4B (VPS4B).
For exosomal secretion, 14 genes are listed as modulators of the “positive regulation of exosomal secretion”, including ATPase type 13A2 (ATP13A2), charged multivesicular body protein 2A (CHMP2A), HGF-regulated tyrosine kinase substrate (HGS), myosin VB (MYO5B), programmed cell death 6 interacting protein (PDCD6IP), RAB7A, RAB7B, syndecan 1 (SDC1), syndecan 4 (SDC4), syndecan binding protein (SDCBP), sphingomyelin phosphodiesterase 3, neutral (SMPD3), SNF8, the endosomal sorting complexes required for transport (ESCRT)-II complex subunit, homolog (S. cerevisiae) (SNF8), signal-transducing adaptor molecule (SH3 domain and ITAM motif) 1 (STAM), tumor susceptibility gene 101 (TSG101), vacuolar protein sorting 4A (VPS4A), and VPS4B. RAB7A, RAB7B, and VPS4B also belong to the genes with a “negative regulation of exosomal secretion” function.
For exosomal assembly, the CD34 antigen (CD34) gene is listed among the cells that affect “extracellular exosome assembly”. Four genes are listed as modulators of “positive regulation of extracellular exosome assembly”, including PDCD6IP, SDC1, SDC4, and SDCBP, which also belong to the group of genes with an exosomal secretion function. Three genes are listed among the cells that affect the “regulation of extracellular exosome assembly”, including PDCD6IP, STAM, and TSG101, which also belong to the group of cells with an exosomal secretion function mentioned above.

2.2. Exosomal Secretion and Assembly Effects of Natural Products in Regulating Cell Functions

Some of the exosomal secretion (Section 2.2.1) and assembly (Section 2.2.2) genes have been reported to be regulated by natural products (Table 1).

2.2.1. Exosomal Secretion Effects of Natural Products in Regulating Cell Functions

Natural-product-derived exosomes exhibit a capacity for the sophisticated regulation of cell functions in non-cancer and cancer cells. The potential roles of exosome-processing genes in natural product treatments can be predicted by target gene retrieval using the miRDB database (Table 1).
For non-cancer cells, several natural products exhibit modulating effects on cell functions in regard to exosome processing. Drug-triggered hepatic precancerous lesions upregulate exosomal RAB11A mRNA. Hesperidin downregulates exosomal RAB11A mRNA and upregulates exosomal miR-1298, resulting in hepatoprotective effects on rats (Table 1) [33]. Several natural products, such as tenuifolin, schisandrin A, celastrol, salidroside, and carnosic acid, were demonstrated to exhibit neuroprotection effects through PINK1 modulation [34]. Moreover, PINK1 and PRKN cooperate to regulate the mitophagy of renal proximal tubular cells [35]. Hence, these natural products (tenuifolin, schisandrin A, celastrol, salidroside, and carnosic acid) may modulate PRKN expression. Salvianolic acid B, a Salvia miltiorrhiza Bge-derived bioactive compound, suppresses renal interstitial fibrosis by inducing SDC1/E-cadherin in angiotensin II-treated proximal tubular cells [36]. Combined, Echinacea angustifolia DC. and Zingiber officinale lipophilic extracts show immunomodulatory effects by downregulating SDCBP expression based on human studies [37]. Bavachinin, a Fructus psoraleae-derived natural product, provides protection against the palmitic-acid-induced death of hepatocytes by upregulating VPS4B expression (Table 1) [38].
Table 1. Connecting natural products to exosomal secretion and assembly in the regulation of cell functions.
Table 1. Connecting natural products to exosomal secretion and assembly in the regulation of cell functions.
Natural ProductsGenesCell FunctionsCancerReferences
Exosomal secretion
Methanolic extract of ↓COPS5apoptosiscervical[39]
Moringa oleifera
Rutin↓COPS5apoptosiscervical[40]
Hesperidin↓RAB11Ahepatoprotective(rat)[33]
Heteronemin↑STEAP3ferroptosispancreatic[41]
Dihydroartemisinin↓STEAP3anti-iron uptakeliver[42]
Robustaflavone A↓STEAP3ferroptosisbreast[43]
Tenuifolin, Schisandrin A, ↑PRKNmitophagy(renal tubular cells)[35]
 Celastrol, Salidroside,
 Carnosic acid
Liensinine↑RAB7Aanti-autophagybreast[44]
Sulfisoxazole↓VPS4Bantimetastaticbreast[45]
Bavachinin↑VPS4Bpro-survival(hepatocyte)[38]
Squalamine↑ATP13A2α-synuclein aggregationneuroblastoma[46]
7-α-Hydroxyfrullanolide↑PDCD6IPapoptosisbreast[47]
Salvianolic acid B↓SDC1renal interstitial fibrosis(proximal tubular cells)[36]
Rutaecarpine↓SDC1antimigrationglioblastoma[48]
Echinacea angustifolia/↓SDCBPimmunomodulation(human study)[37]
Zingiber officinale
extracts
Dioscin↑SDCBPapoptosis, autophagy, liver ca[49]
DNA damage
Sulforaphane↑SDCBPapoptosisleukemia[50]
Acetyl-11-keto-b-boswellic ↑SMPD3antiproliferationcolon[51]
 acid
Withanolide D↑/↓SMPD3apoptosisleukemia[52]
Resveratrol↑TSG101antiproliferationintestinal tumor[53]
Exosomal assembly
Astragalus membranaceus↑CD34angiogenesis(myocardial infarction)[54]
 extract
D Rhamnose bhederin↓STAMchemoresistancebreast[55]
, enhance or activate; , inhibit or inactivate. Some are non-cancer studies, shown in parentheses. PDCD6IP, SDC1, SDC4, SDCBP, STAM, and TSG101 exhibit the regulating functions of both exosomal secretion and assembly, as described in Section 2.1.
For cancer cells, several natural products exhibit modulating effects on cell functions in regard to exosome processing. The methanolic extract of Moringa oleifera leaves suppresses the proliferation and causes the G1 arrest and apoptosis of cervical cancer cells by downregulating COPS5 (Table 1) [39]. Rutin, a bioflavonoid, induces the apoptosis of cervical cancer cells by downregulating COPS5 [40]. Heteronemin, a marine sesterterpenoid, induces ferroptosis by upregulating the protein expression of divalent metal transporter-1 (DMT1) and STEAP3 in pancreatic cancer cells [41]. Dihydroartemisinin, a metabolite of artemisinin, downregulates the DMT1 and STEAP3 genes controlling iron uptake in liver cancer HepG2 cells (Table 1) [42].
Selaginella trichoclada-derived robustaflavone A induces ferroptosis by downregulating the expressions of acyl-CoA synthetase long-chain family member 4 (ACSL4), ACSL5, STEAP3, lysophosphatidylcholine acyltransferase (LPCAT3), and autophagy-related 7 (ATG7) genes in breast cancer cells (Table 1) [43]. Liensinine, a Nelumbo nucifera-derived isoquinoline alkaloid, induces the expression of the small GTP-binding protein RAB7A and suppresses autophagosome–lysosome fusion for the degradation of breast cancer cells [44]. Squalamine inhibits the α-synuclein aggregation of neuroblastoma cells [56]. The inhibition of ATP13A2 destroys lysosomal membrane integrity and induces the α-synuclein accumulation of neuroblastoma cells (Table 1) [46]. All this warrants a detailed assessment of the role of ATP13A2 in squalamine treatment.
Sulfisoxazole shows antimetastatic effects on breast cancer cells by downregulating VPS4B mRNA expression (Table 1) [45]. 7-α-Hydroxyfrullanolide, an Asteraceae-plant-derived natural product, upregulates PDCD6IP expression in breast cancer cells [47]. Rutaecarpine downregulates the mRNA expression of the SDC1 gene to suppress glioblastoma cell migration [48]. Dioscin, a steroid saponin, causes the apoptosis, autophagy, and DNA damage of liver cancer cells by upregulating SDCBP expression [49]. Sulforaphane, a cruciferous-vegetable-derived compound, triggers apoptosis by upregulating SDCBP expression in leukemia HL-60 cells [50]. Acetyl-11-keto-b-boswellic acid (AKBA), a Boswellia serrata-derived natural product, inhibits the proliferation of colon cancer cells by upregulating SMPD3 [51]. Withanolide D triggers the apoptosis of leukemia MOLT-4 cells by upregulating SMPD3 expression after 15 min and downregulating after 45 min [52]. The oral administration of resveratrol suppresses intestinal tumorigenesis in mice and upregulates TSG101 mRNA expression (Table 1) [53]. All this warrants a detailed assessment exploring the roles of more natural-product-regulated cell functions in exosomal secretion in the future.

2.2.2. Exosomal Assembly Effects of Natural Products in Regulating Cell Functions

Natural products also regulate exosomal assembly. Astragalus membranaceus extract enhances angiogenesis in myocardial infarction rats by upregulating vascular endothelial growth factor (VEGF), CD34, and endothelial nitric oxide synthase (eNOS) expression [54]. Moreover, D Rhamnose bhederin, a Clematis ganpiniana-derived bioactive component, inhibits exosome secretion from docetaxel-resistant breast cancer cells and alleviates the transmission of resistance [55]. D Rhamnose bhederin downregulates several miRNAs (miR-16-5p, miR-23a-3p, miR-24-3p, miR-26a-5p, and miR-27a-3p), in which miR-24-3p is predicted to target exosomal-assembly related genes, such as STAM, according to the miRDB database [31]. This warrants a detailed assessment exploring the roles of more natural-product-regulated cell functions in exosomal assembly in the future.

3. Prediction of the Targeting of Exosome-Processing and AKT-Signaling Genes of Certain Exosome miRNAs

Several exosomal miRNAs have been reviewed previously [4,16,57]. However, their potential impacts on exosomal processing (secretion and assembly) have rarely been investigated. In this study, utilizing the miRDB database [31], these exosomal miRNAs targeting exosome-processing genes (Section 2) were retrieved (Table 2). miR-29a-3p is predicted to target SMPD3. miR-101-3p and miR-21-5p/miR-30a-5p are predicted to target RAB27A and RAB11A, respectively. miR-6887-5p is predicted to target RAB7A and RAB7B. miR-142-3p and miR-24-3p are predicted to target HGS and ATP13A2/STAM, respectively. miR-106a-5p, miR-106b-5p, miR-21-5p, miR-223-3p, miR-365a-3p, and miR-374a-5p are predicted to target MYO5B. miR-522-3p and miR-8485 are predicted to target PDCD6IP (Table 2).
Additionally, miR-8485 is also predicted to target SDC1 and SDCBP. miR-128-3p, miR-142-3p, miR-200c-3p, miR-223-3p, and miR-8485 are predicted to target STAM. miR-106a-5p and miR-106b-5p are predicted to target TSG101. Finally, miR-128-3p, miR-32-5p, and miR-92a-3p are predicted to target VPS4B (Table 2). Consequently, these examples demonstrate that many of the reported exosomal miRNAs have the potential to target exosomal processing genes. This warrants an advanced examination exploring the roles of exosomal processing genes in several reported exosomal miRNA studies in the future.
Moreover, some exosomal miRNAs are reported to regulate AKT signaling [58,59,60,61]. Among the examples of exosomal miRNAs listed in Table 2, miR-29a-3p is predicted to target AKT2 and AKT3 (Table 2). miR-374a-5p is predicted to target AKT1. miR-101-3p, miR-106a-5p, miR-106b-5p, miR-365a-3p, miR-6887-5p, and miR-30a-3p are predicted to target AKT1, ATK2, or AKT3. Those AKT1-, AKT2-, and AKT3-targeting exosomal miRNAs (miR-29a-3p, miR-374a-5p, miR-101-3p, miR-106a-5p, miR-106b-5p, miR-365a-3p, miR-6887-5p, and miR-30a-3p) also target some exosomal processing genes (Table 2).
Exosomal proteins can activate AKT signaling in the regulation of metastasis. Annexin A5, one of the exosome proteins in prostate cancer tissues, activates AKT signaling to stimulate the epithelial–mesenchymal transition (EMT) and upregulate matrix metalloproteinase-2 (MMP2) and MMP9 expression [5]. The exosomal miRNAs involved in AKT signaling have been applied in animal [59] and preclinical experiments [60]. Bone-marrow—mesenchymal-stem-cell-derived exosomes, which are rich in miR-126-3p (miR-126), enhance the migration and angiogenesis of human umbilical vein endothelial cells (HUVECs) [59]. This exosomal miR-126-3p stimulates vascularization at wound sites and improves cutaneous wound healing in mice models. Plasma exosomes isolated from Graves ophthalmopathy with an effective response to intravenous glucocorticoid therapy contain a high level of miR-885-3p, showing AKT inhibition and improving glucocorticoid sensitivity [60]. Consequently, the exosomal miRNAs with AKT modulating ability are potential tools for preclinical applications.
Table 2. Connecting some exosomal miRNAs to the predicted targets of exosome processing and AKT genes.
Table 2. Connecting some exosomal miRNAs to the predicted targets of exosome processing and AKT genes.
ATP9AATP13A2HGSMYO5BRAB27ARAB11ARAB7ARAB7BPDCD6IPSDC1SDCBPSMPD3STAMTSG101VPS4BAKT
miR-29a-3p [4,16] SMPD3 AKT2/3
miR-101-3p [4] RAB27A AKT3
miR-106a-5p [16,57] MYO5B TSG101 AKT3
miR-106b-5p [57] MYO5B TSG101 AKT3
miR-128-3p [57] STAM VPS4B
miR-142-3p [4,16] HGS STAM
miR-200c-3p [4,16] STAM
miR-21-5p [4,57] MYO5B RAB11A
miR-223-3p [16] MYO5B STAM
miR-24-3p [4,16] ATP13A2 STAM
miR-32-5p [57] VPS4B
miR-365a-3p [57] MYO5B AKT3
miR-374a-5p [57] MYO5B AKT1
miR-522-3p [57] PDCD6IP
miR-6887-5p [4] RAB7ARAB7B AKT3
miR-8485 [4]ATP9A PDCD6IPSDC1SDCBP STAM
miR-92a-3p [57] VPS4B
miR-30a-3p [62] RAB11A AKT3
The predicted targets for exosomal processing and AKT genes of the exosomal miRNAs were retrieved from the miRDB database (retrieval date: 12 November 2022).

4. Natural Products Modulate Exosome Production and Their Exosome Delivery for the Purpose of Cancer Treatment

Many natural-product-derived exosomes have been demonstrated to exhibit theranostic effects in cancer therapy [63,64,65,66]. The modulating results of exosome biogenesis and delivery by natural products are discussed as follows below.
Some natural products improve exosome biogenesis. Sulforaphane suppresses the fusion of early and late endosomes (GFP-Rab5a and GFP-Rab7a) with the lysosome, blocks the autophagy flux, promotes exosome production, and triggers exosome-dependent paracrine senescence by downregulating mTOR and transcription factor binding to IGHM enhancer 3 (TFE3) [67]. Sulforaphane induces a high protein concentration of exosomes and causes the accumulation of exosome marker CD63 in esophageal cancer cells. Moreover, supernatants from sulforaphane-treated cancer cells show high CD63 expression [67]. Consequently, sulforaphane triggers exosome biogenesis and secretion in esophageal cancer cells.
In contrast, some natural products suppress exosome biogenesis. Autophagy and lysosome dysfunction enhance exosome secretion [68,69] and vice versa. Asteltoxin inhibits mitochondrial ATP synthase and exosome generation by upregulating AMPK-dependent mTORC1 inactivation and lysosome activation [70]. Transmission electron microscopy analysis shows that asteltoxin induces lysosome–MVB fusion, causing the downregulation of exosome generation. Berberine suppresses the proliferation of colon cancer cells by downregulating acetyl-CoA carboxylase (ACC) for fatty acid synthesis and reducing exosome biogenesis and the secretion of colon and cervical cancer cells [71], an observation which is supported by the finding that berberine downregulates syntenin and TSG101, as intracellular vesicle markers.
Some natural-product-derived exosomes exhibit modulating effects on cell functions. Exosomes used in phytoagent deoxyelephantopin treatment, a plant deoxyelephantopin derivative, suppress the ROS-mediated proliferation of breast cancer cells, reversed by N-acetylcysteine [72]. Phytoagent deoxyelephantopin also enhances calcium-dependent exosome secretion from breast cancer cells. Momordica charantia-derived exosome-like nanovesicles suppress the proliferation and migration of glioma cells by downregulating phosphorylated PI3K/AKT [73]. Fusobacterium nucleatum is rich in colon cancer lesions associated with colon cancer carcinogenesis and metastases. Exosomes from Fusobacterium nucleatum enhance the invasion of colon cancer cells. This invasion is prevented by the bioactive compounds of Paris polyphylla, such as pennogenin 3-O-beta-chacotrioside and polyphyllin VI, which exhibit cell-killing effects on Fusobacterium nucleatum [74]. All this warrants an advanced examination exploring the impacts of exosomal biogenesis by natural products and natural-product-derived exosomes on cell functions in the future.
Moreover, exosomes are naturally generated, showing lower cytotoxicity and immunogenicity and higher biocompatibility than lipid-based nanoparticles [6,65,75]. Exosomes were reported to effectively deliver several natural products that can be exploited for preclinical anticancer therapy in vitro and in vivo [65]. The oral delivery of paclitaxel using milk-derived exosomes results in less side effects of immunologic toxicity and higher antitumor effects than i.v. in lung-tumor-xenograft nude mice [76]. Exosome-delivered curcumin exhibits a high in vitro stability and in vivo bioavailability [77]. Celastrol-loaded milk exosomes show a high degree of anti-lung tumor growth with in vivo biosafety [78]. This warrants the advanced testing of more natural products based on exosome delivery strategies in the future.

5. The Role of Natural-Product-Modulating miRNAs and Exosomes in Regulating Cancer Cell Functions

A mounting array of literature reports that natural products modulate many miRNAs that regulate their target genes to affect several of the cell functions of cancer cells [79]. However, most of these studies did not investigate the impacts of exosomal miRNAs on anticancer effects using natural products.
Recently, several natural-product-induced exosomal miRNA studies have been reported. Aurea helianthus extract inhibits the migration and induces the senescence and autophagy of endometrial cancer cells [80]. Several miRNAs derived from the induced exosomes in these extract-treated endometrial cancer cells were upregulated or downregulated. However, there is a lack of systemic information on the modulating effects of drug-induced exosomal miRNAs based on natural products. Most natural-product-modulating miRNA studies have focused on impacts on cancer cell functions without considering the contribution of exosomes. Consequently, there are gaps between exosomal miRNAs and natural products in terms of their anticancer effects.
A total of 26 natural-product-modulated exosomal miRNAs that regulate cancer cell functions, such as antiproliferation (Section 5.1), apoptosis (Section 5.2), antimigration/anti-invasion/anti-EMT/anti-angiogenesis (Section 5.3), the modulation of chemo- and radio-resistance (Section 5.4), and others (Section 5.5), are summarized in Table 3. Many non-exosomal miRNA studies have assessed the impacts of the anticancer effects of natural products. In the future, a detailed examination of the roles of exosomes and investigations of their anticancer effects related to miRNAs and natural products should be carried out.

5.1. Antiproliferation by Natural-Product-Modulated Exosomal miRNAs

Natural products may regulate cell proliferation by modulating miR-424-5p, miR-21-5p (miR-21), miR-101-3p, miR-1246, miR-155-5p (miR-155), miR-30a-5p, miR-34a-5p, and miR-30d-5p (Table 3), as described in the following section.
miR-424-5p is downregulated in breast cancer tissues. Resveratrol inhibits the proliferation of breast cancer cells by upregulating miR-424-5p and downregulating heterogeneous nuclear ribonucleoprotein A1 (HNRNPA1) [85] (Table 3).
Berberine, a natural alkaloid, inhibits the proliferation of multiple myeloma cells by downregulating miR-21-5p and upregulating its target, programmed cell death 4 (PDCD4) [86] (Table 3). The natural product butylcycloheptyl prodiginine promotes the antiproliferation of colon cancer cells by binding pre-miR-21-5p to inhibit the function of miR-21-5p [89]. Honokiol, a Magnolia officinalis-derived natural product, promotes the antiproliferation of osteosarcoma cells by downregulating miR-21-5p/AKT signaling [90]. Sophocarpine, a Sophora flavescens-derived bioactive compound, suppresses the proliferation of head and neck cancer cells by targeting miR-21-5p [92]. Dihydromyricetin, a natural flavonoid, suppresses the proliferation of cholangiocarcinoma cells by downregulating miR-21-5p [94]. Curcumin decreases the proliferation of liver cancer cells by downregulating miR-21-5p and upregulating its target, SRY-box transcription factor 6 (SOX6) [95] (Table 3).
Exosomal miR-101-3p displays tumor-suppressive and oncogenic functions. Oral cancer cells express low levels of miR-101-3p by targeting the collagen type X alpha 1 chain (COL10A1). In contrast, exosomes derived from bone marrow mesenchymal stem cells overexpress miR-101-3p to suppress oral cancer proliferation and migration [131]. miR-101-3p mimics suppress the proliferation and migration and trigger the apoptosis of medulloblastoma cells by targeting the enhancer of zeste homolog 2 (EZH2), a histone methyltransferase [132]. In contrast, exosomal miR-101-3p exhibits an oncogenic function to improve the proliferation and migration of colon cancer cells by downregulating its target, the homeodomain-interacting protein kinase (HIPK3) [133]. Berberine, a plant-bark-derived alkaloid, suppresses the proliferation of endometrial cancer cells by upregulating miR-101-3p to downregulate cyclo-oxygenase-2 (COX-2) [88] (Table 3).
Exosomal miR-1246 is reported to regulate cell migration. Exosomal miR-1246 from highly metastatic oral cancer cells promotes the migration and invasion of poorly metastatic oral cancer cells by downregulating the DENN/MADD-domain-containing 2D (DENND2D) [134]. In addition to antimigration, miR-1246 was reported to modulate proliferation (Table 3). Bladder cancer T24 cells highly express miR-1246. Curcumin inhibits the proliferation of bladder cancer cells by downregulating miR-1246 [98]. Combined treatment (curcumin and X-ray) synergistically suppresses its proliferation to a greater extent than individual treatments by decreasing miR-1246 expression [98].
Moreover, natural products were reported to modulate miR-155-5p, miR-30a-5p, and miR-34a-5p expression, regulating cancer cell proliferation (Table 3). Genistein, a soy isoflavone phytoestrogen, suppresses the proliferation of breast cancer cells by downregulating miR-155-5p [105]. Thymoquinone, a black-seed-oil-derived compound, suppresses liver fibrosis by upregulating miR-30a-5p to inhibit its target, such as snail family transcriptional repressor 1 (SNAI1), inhibiting EMT [111]. Norcantharidin, a cantharidin derivative, inhibits the proliferation of giant-cell tumors of the bone by upregulating miR-30a-5p and downregulating AKT, reversed by inhibiting miR-30a-5p [62]. Emodin, a natural anthraquinone derivative, suppresses liver cancer cell proliferation by upregulating miR-34a-5p [122].
Exosomal miR-30d-5p appears in higher levels in cervical cancer tissues than in normal controls [135]. However, the miR-30d-5p-modulating cell function has rarely been reported, particularly in regard to the antiproliferation of cancer cells. A recent study of natural products reported the antiproliferation effect achieved by the modulation of miR-30d-5p (Table 3). Piperlongumine, a long-pepper-derived amide alkaloid, suppresses the proliferation of osteosarcoma cells by downregulating miR-30d-5p and upregulating its target, the suppressor of cytokine signaling 3 (SOCS3) [130].
Furthermore, natural products may regulate cancer cell proliferation by modulating miR-200c-3p (Table 3). (−)-Sativan, a Spatholobus suberectus-derived isoflavane, suppresses the proliferation of breast cancer cells by upregulating miR-200c-3p to downregulate its direct target, such as prickle planar cell polarity protein 2 (PRICKLE2; EPM5) [109].

5.2. Apoptosis by Natural-Product-Modulated Exosomal miRNAs

Several natural products modulate apoptosis in cancer cells by downregulating miR-21-5p, miR-196a-5p (miR-196a), miR-210-3p, miR-365a-3p (miR-365), miR-34a-5p (miR-34a), miR-144-3p (miR-144), miR-23b-3p, and miR-382-5p (Table 3), as described in the following section.
Several natural products promote apoptosis through miR-21-5p in cancer cells. Ursolic acid triggers the apoptosis of glioblastoma cells by downregulating miR-21-5p [81] (Table 3). Resveratrol triggers the apoptosis of bladder cancer cells by downregulating miR-21-5p and AKT phosphorylation, reversed by miR-21-5p overexpression [83]. The downregulation of miR-196a-5p enhances cisplatin resistance [102]. Psoralen, a natural photosensitizing drug, triggers the apoptosis of gastric cancer cells to alleviate cisplatin resistance by upregulating miR-196a-5p and downregulating homeobox B7 (HOXB7) and HER2 expression [102]. Honokiol enhances the apoptosis of osteosarcoma cells by downregulating miR-21-5p/AKT signaling [90]. Dihydromyricetin triggers the apoptosis of cholangiocarcinoma cells by downregulating miR-21-5p [94]. PRP1, a Platycodonis-radix-derived polysaccharide, promotes the apoptosis of liver cancer cells by reducing miR-21-5p expression and inactivating AKT [100].
Exosomal miR-210-3p promotes the angiogenesis and tubulogenesis of endothelial cells [136] and enhances the metastasis of lung cancer cells [137]. A recent study demonstrated the novel function of the apoptosis-modulating effect of miR-210-3p, regulated by natural products. In hypoxic conditions, colon cancer cells enhance tumor progression. The transmission of hypoxic colon-cancer-cell-derived exosomal miR-210-3p to normoxic tumor cells prevents apoptosis and induces a protumoral effect [138]. 1΄S-1΄-Acetoxychavicol acetate (ACA), a wild ginger Alpinia conchigera-derived natural product, triggers the apoptosis of cervical cancer cells by downregulating miR-210-3p to upregulate its target, SMAD family member 4 (SMAD4) [113] (Table 3).
Exosomal miR-365a-3p regulates the chemoresistance of cancer cells involved in apoptosis. Exosomal miR-365a-3p derived from imatinib-resistant chronic myeloid leukemia (CML) cells provides drug resistance to, and prevents apoptosis in, sensitive CML cells [139].
Natural product studies showed that miR-365a-3p exhibits an apoptosis-modulating effect (Table 3). Crocin, a carotenoid pigment of saffron, induces cervical cancer cell apoptosis by upregulating Bax and downregulating BCL2 and miR-365a-3p [114]. The combination treatment of cervical cancer cells with crocin and cisplatin promotes antiproliferation and apoptosis by downregulating miR-365a-3p, an upregulator of BAX and BCL2 [140].
Exosomal miR-421 regulates the chemoresistance of cancer cells. Exosomes from cisplatin-resistant oral cancer patients enhance the proliferation and reduce the cisplatin sensitivity of cisplatin-resistant cells by downregulating miR-421 expression [141]. The hypermethylation of transcription-factor-activating-enhancer-binding protein 2e (TFAP2E) enhances 5-fluorouracil chemoresistance in gastric cancer cells by upregulating exosomal miR-421 [142]. However, the impact of apoptosis by exosomal miR-421 is unclear. A recent natural product study reported the apoptosis function through the modulation of miR-421 (Table 3). Isoliquiritigenin induces the apoptosis and DNA damage of oral cancer cells by downregulating miR-421 expression [116].
Exosomal miR-34a-5p regulates the proliferation of cancer cells. Normal fibroblasts exhibit higher miR-34a-5p levels than cancer-associated fibroblasts from oral cancer patients. miR-34a-5p overexpression in cancer-associated fibroblasts suppresses cancer cell proliferation and migration [143]. Exosomal miR-34a-5p induces the antiproliferation and apoptosis of pancreatic cancer cells [144]. Several natural product studies have demonstrated the apoptosis function of cancer cells through the modulation of miR-34a-5p (Table 3). Dihydroartemisinin triggers the apoptosis of prostate cancer cells by upregulating miR-34a-5p [120]. Isovitexin, a flavonoid, triggers the apoptosis of osteosarcoma cells by upregulating miR-34a-5p and downregulating BCL2 [121]. Crocin, a saffron-derived pigment, triggers the ROS-dependent apoptosis of papillary thyroid cancer cells by downregulating the miR-34a-5p and upregulating its target, protein tyrosine phosphatase non-receptor type 4 (PTPN4) [115].
miR-144-3p exhibits differential expressions in different cancer cells. miR-144-3p is downregulated in lung cancer cells. Exosomal miR-144-3p from bone marrow-derived mesenchymal stem cells suppresses the proliferation of lung cancer cells by downregulating cyclin E1 (CCNE1) and CCNE2 [145]. In contrast, miR-144-3p is upregulated in nasopharyngeal cancer cells. Exosomal miR-144-3p from nasopharyngeal cancer cells promotes angiogenesis [146]. However, the apoptosis-inducing effect of exosomal miR-421 has rarely been examined. Recent natural product studies demonstrated apoptosis induction through the modulation of miR-144-3p (Table 3). Berberine induces the apoptosis of lung cancer cells by upregulating miR-144-3p [87]. Licochalcone A, a Glycyrrhiza inflata-derived natural product, induces ER stress and the apoptosis of lung cancer cells by upregulating miR-144-3p [126].
miR-23b-3p suppresses the proliferation and migration of prostate [147] cancer cells, while it enhances pancreatic cell migration [148] and salivary cancer cell angiogenesis and metastasis [149]. However, the apoptosis function of exosomal miR-421 has rarely been reported. A natural product investigation validated the fact that apoptosis induction results from the modulation of miR-23b-3p (Table 3). 10-Hydroxycamptothecin, a Nothapodytes nimmoniana-derived natural product, causes the apoptosis of fibroblasts by upregulating miR-23b-3p [127].
Exosomal miR-382-5p from cancer-associated fibroblasts enhances the migration of oral cancer cells [150]. A recent natural product study demonstrated the novel function of apoptosis induction through the modulation of miR-382-5p (Table 3). Polydatin, a metabolite of trans-resveratrol, inhibits the proliferation and causes the apoptosis of colon cancer cells by upregulating miR-382-5p and downregulating its target, programmed cell death ligand 1 (PD-L1) [129].

5.3. Antimigration/Anti-Invasion/Anti-EMT/Anti-Angiogenesis by Natural-Product-Modulated Exosomal miRNAs

Several natural products regulate the migration, invasion, and angiogenesis of cancer cells by modulating miR-101-3p, miR-30a-5p, miR-34a-5p, miR-200c-3p, miR-21-5p, and miR-421 (Table 3), as described in the following section.
Some miRNAs showing migration-suppressing effects are upregulated by several natural products. Berberine suppresses the migration of endometrial cancer cells by upregulating miR-101-3p to downregulate cyclo-oxygenase-2 (COX-2) [88] (Table 3). Norcantharidin inhibits the migration of giant-cell tumors of the bone by upregulating miR-30a-5p and downregulating AKT, reversed by inhibiting miR-30a-5p [62]. Dihydroartemisinin suppresses the migration of prostate cancer cells by upregulating miR-34a-5p [120]. Honokiol, a Magnolia grandiflora-derived polyphenol, suppresses the leptin-promoted EMT of breast cancer cells by upregulating miR-34a-5p [91]. Anisomycin suppresses angiogenesis in ovarian cancer stem cells by upregulating miR-421 [117].
Similarly, exosomal miR-200c-3p suppresses the migration and invasion of lipopolysaccharide (LPS)-treated colon cancer cells by targeting zinc finger E-box-binding homeobox-1 (ZEB-1) [151]. The natural compounds enoxolone, magnolol, and palmatine chloride suppress the invasion of breast cancer cells by upregulating miR-200c-3p [108]. (−)-Sativan inhibits the migration of breast cancer cells by upregulating miR-200c-3p [109]. Similarly, curcumin, acting on colon cancer cells, exhibits the downregulation of EMT-related gene expression by upregulating miR-200c-3p and downregulating its target, PRICKLE2 [96]. Isoliquiritigenin, a Glycyrrhizae Rhizoma-derived bioactive component, inhibits migration, metastasis, and breast tumor growth by inhibiting EMT and upregulating miR-200c-3p, which is downregulated in breast cancer tissues [110]. Ursolic acid, a pentacyclic triterpenoid, induces the apoptosis and inhibits the invasive ability of colon cancer cells by upregulating miR-200c-3p [82].
In contrast, some miRNAs showing migration-promoting effects are suppressed by natural product treatments. The transfer of hypoxic oral cancer exosomes containing miR-21-5p to normal cells improves their pro-metastatic effects [152]. Sophocarpine suppresses the epithelial–mesenchymal transition (EMT) of head and neck cancer cells by targeting miR-21-5p [92]. Dihydromyricetin inhibits the migration of cholangiocarcinoma cells by downregulating miR-21-5p [94]. Curcumin decreases the migration of liver cancer cells by downregulating miR-21-5p [95]. Sinomenine, a Sinomenium acutum-derived alkaloid, shows antimigration effects on lung cancer cells by suppressing miR-21-5p and MMP2/9 [101]. Asparanin A, a vegetable- and Asparagus officinalis-derived natural product, suppresses the migration of endometrial cancer cells by downregulating miR-421 [118].

5.4. Modulation of Chemo- and Radio-Resistance by Natural-Product-Modulated Exosomal miRNAs

Several natural products regulate migration, invasion, and angiogenesis in cancer cells by modulating miR-21-5p, miR-155-5p, miR-34a-5p, and miR-31-5p, as described in the following section.
Some miRNAs showing resistance-promoting effects are downregulated by several natural products. miR-21-5p has been identified in exosomes from hypoxic oral cancer cells. miR-21-5p-containing hypoxic oral cancer exosomes also exhibit cisplatin resistance in oral cancer cells, as evidenced by exosome transfer experiments [153]. Natural products may inhibit drug resistance in cancer cells by downregulating miR-21-5p. Tricin, an Allium atroviolaceum-derived compound, sensitizes the docetaxel response to prostate cancer cells by downregulating miR-21-5p [93] (Table 3).
Similarly, exosomal miR-155-5p enhances the migration or metastasis of gastric [154], lung [155], renal [156], and colon [157] cancer cells. Recently, a resistance-modulating function of exosome miR-155-5p was reported. Exosome miR-155-5p from oral cancer cells improves cisplatin resistance to cisplatin-sensitive cells by upregulating EMT [158]. A natural product study showed the resistance-modulating function of miR-155-5p (Table 3). (−)-Epigallocatechin gallate (EGCG), a green- or red-tea-derived bioactive compound, improves 5-fluorouracil (5-FU) sensitivity in colon cancer cells by suppressing miR-155-5p expression [106].
In contrast, some miRNAs showing resistance-suppressing effects are upregulated by several natural products. Rhamnetin and cirsiliol, the quercetin and flavonoid derivatives, enhance radiosensitization and suppress lung cancer cell EMT by upregulating miR-34a-5p [119]. miR-34a-5p is downregulated in liver cancer tissues. EGCG improves the radiosensitization of liver cancer cells by upregulating miR-34a-5p [107]. Similarly, bladder cancer tissues exhibit a low level of miR-31-5p. Mitomycin C sensitivity is enhanced in bladder cancer cells by upregulating miR-31-5p to target integrin α5 (ITGA5) [125].

5.5. Potential Modulation Effects of Target Immunotherapy of Cancer by Natural-Product-Modulated Exosomal miRNAs

The tumor immune microenvironment (TIME) comprises several types of immune cells. Some miRNAs were identified in the tumor-associated macrophages (TAM), natural killer (NK) cells, and myeloid-derived suppressor cells (MDSC) of TIME [159]. A comparison illustrated that some of them overlapped with the exosomal miRNAs modulated by natural products (Table 3). Upon inspection, some of the exosomal miRNAs (miR-21-5p, miR-200c-3p, miR-155-5p, miR-30a-5p, miR-34a-5p, miR-130a-3p, miR-101-3p, miR-142-3p, and miR-24-3p) listed in Table 3 were reported in certain immune cells of TIME [159], such as TAM, NK, and MDSC. However, the review in question [159] rarely mentioned the impacts of natural products.
Here, we discuss the indirect connections of these exosomal miRNAs to natural products (Table 4). TAM upregulates several exosomal miRNAs (miR-21-5p, miR-155-5p, miR-30a-5p, miR-101-3p, and miR-142-3p) but downregulates miR-34a-5p [159]. NK upregulates miR-155-5p, miR-130a-3p, miR-101-3p, and miR-24-3p. MDSC upregulates miR-21-5p, miR-200c-3p, miR-155-5p, and miR-30a-5p. Since some of the miRNAs listed in Table 4 are downregulated or upregulated by several natural products, their potential impacts in modulating the expressions of the TAM, NK, and MDSC of TIME are worthy of attention. Notably, TIME miRNAs that are not included in Table 3 and Table 4 may be modulated by natural products and, thus, require detailed investigation.
All this warrants a detailed assessment of all the miRNA-modulating effects of these natural products that are employed in cancer studies in the future.

5.6. Other Cell Functions Influenced by Natural-Product-Modulated Exosomal miRNAs

Several miRNAs, such as miR-31-5p, miR-3188, miR-24-3p, miR-30a-5p, miR-130a-3p, miR-142-3p, miR-30a-5p, and miR-382-5p, exhibit diverse effects other than the modulation of proliferation, apoptosis, migration, and resistance. Although these miRNAs modulate many functions, only a few natural products were retrieved from Pubmed and Google scholar.
Exosomal miR-31-5p regulates the proliferation and drug resistance of cancer cells. Macrophage-derived exosomal miR-31-5p enhances oral cancer cell proliferation by downregulating large tumor suppressor 2 (LATS2) [160]. Exosomal miR-31-5p from hypoxic lung cancer cells promotes metastasis [161]. Moreover, exosomal miR-31-5p is also involved in the regulation of drug resistance. Exosomal miR-31-5p enhances sorafenib resistance in renal cancer cells by targeting mutL homolog 1 (MLH1) [162]. Forkhead box C1 (FOXC1) functions as a transcriptional factor to promote the transcription of miR-31-5p and downregulate LATS2, leading to oxaliplatin resistance in colon cancer cells [163]. A natural product study showed that resveratrol alleviates 2,4,6-trinitrobenzenesulfonic-acid-solution (TNBS)-induced colitis by suppressing miR-31-5p expression to increase the number of regulatory T-cells [84] (Table 3).
Cancer-associated fibroblasts enhance the progression of head and neck cancer cells by downregulating exosomal miR-3188 [164]. Without considering exosomes, other cancer studies also reported the tumor suppressive function of miR-3188. miR-3188 inhibits the proliferation of nasopharyngeal [165] and lung [166] cancer cells by targeting mTOR. A natural product study showed that pinolenic acid, a Pinus-species-derived natural product, upregulates miR-3188 to target the pyruvate dehydrogenase Kinase 4 (PDK4) and the mitochondrially encoded ATP synthase membrane subunit 6 (MT-ATP6) genes, showing anti-inflammatory effects in rheumatoid arthritis patients [103] (Table 3).
Exosomal miR-24-3p modulates proliferation and drug resistance in cancer cells. Salivary exosomal miR-24-3p promotes the proliferation of oral cancer cells by targeting period circadian regulator 1 (PER1) [167]. Exosomal miR-24-3p from cancer-associated fibroblasts enhances methotrexate resistance and inhibits the apoptosis of colon cancer cells by suppressing caudal type homeobox 2 (CDX2) or hephaestin (HEPH) expression [168]. A natural product study showed that doxorubicin causes the miR-24-3p overexpression of the left ventricle [104]. Pachymic acid, a Poria cocos-derived natural product, alleviates doxorubicin-induced heart failure in rats by downregulating miR-24-3p [104] (Table 3).
Exosomal miR-30a-5p (miR-30a) regulates several cell functions, such as chemoresistance and migration. Cisplatin-sensitive oral cancer cells exhibit higher miR-30a and lower Beclin 1 (BECN1) expression levels than cisplatin-resistant cells [169]. Exosomes from miR-30a-mimic-transfected cisplatin-resistant cells downregulate BECN1 and BCL2 expression to sensitize the cells to cisplatin. Vascular endothelial cells express exosomes containing higher miR-30a-5p levels than lung cancer cells. Exosomal miR-30a-5p derived from vascular endothelial cells suppresses the proliferation and migration of lung cancer cells by targeting cyclin E2 (CCNE2) [170]. Colon cancer mesenchymal stem cells are abundant in exosomal miR-30a-5p. This stem cell exosomal miR-30a-5p improves the proliferation and migration of colon cancer cells by targeting the MIA SH3 domain ER export factor 3 (MIA3) [171]. A natural product study showed that Nicotine causes the G1 arrest of periodontal ligament cells by upregulating miR-30a-5p to target CCNE2 [95] (Table 3).
Exosomal miR-130a-3p (miR-130a) regulates the proliferation and migration of cancer cells. Breast cancer tissues and plasma exosomes exhibit low miR-130a-3p levels. The overexpression of miR-130a-3p in breast cancer stem-cell-like cells suppresses proliferation and migration by targeting RAB5B, member of the RAS oncogene family (RAB5B) [172]. The serum of differentiated thyroid cancer patients shows low levels of exosomal miR-130a-3p, which upregulates its target, insulin-like growth factor 1 (IGF-1) [173]. Some natural product studies showed that miR-130a-3p possessed inflammation-related functions (Table 3). Kaempferol, a dietary flavonoid, suppresses the cytokine production of chondrocytes by upregulating miR-130a-3p and downregulating its targets, such as the signal transducer and activator of transcription 3 (STAT3) [123]. Chicoric acid, an Echinacea-derived natural product, reduces the LPS-induced inflammation of lung cancer cells by downregulating miR-130a-3p and upregulating IGF-1 [124] (Table 3).
miR-142-3p exhibits differential patterns in regulating proliferation in different cancer cells. An increase in miR-142-3p in oral cancer cells suppresses tumor-promoting changes in the recipient endothelial cells [174]. Exosomal miR-142-3p from monocytes can be transferred to retinoblastoma cells, inhibiting their proliferation [175]. In contrast, exosomal miR-142-3p may exhibit a proliferation-promoting effect. Exosomal miR-142-3p from HBV-infected liver cancer cells induces the ferroptosis of M1 macrophages to improve the proliferation of liver cancer cells [176]. Some natural product studies showed that miR-144-3p can modulate several cell functions, such as the regulation of proteasome, ER stress, and autophagy. Curcumin suppresses 20S proteasome activity in breast cancer cells by upregulating miR-142-3p and downregulating its target, such as the proteasome 20S subunit beta 5 (PSMB5) [97]. Licochalcone A, a Glycyrrhiza inflata-derived natural product, induces ER stress in lung cancer cells by upregulating miR-144-3p [126]. Berberine induces the autophagy of lung cancer cells by upregulating miR-144-3p [87] (Table 3).
Some natural product studies showed that miRNAs can modulate liver fibrosis. Thymoquinone, a black-seed-oil-derived compound, suppresses liver fibrosis by upregulating miR-30a-5p to inhibit its target, such as snail family transcriptional repressor 1 (SNAI1), suppressing EMT [111]. Astaxanthin, a xanthophyll carotenoid, inactivates liver-fibrosis-associated hepatic stellate cells by downregulating miR-382-5p [128]. This warrants a detailed evaluation of all the miRNA-modulating effects of the aforementioned natural products employed in cancer studies.
Furthermore, several exosomal miRNAs have been identified in a number of cancer cells, but no natural product studies have been reported to date. Some exosomal miRNAs show proliferation-/invasion-promoting effects. Exosomal miR-626 enhances the proliferation and migration of oral cancer cells by targeting nuclear factor I/B (NFIB) [177]. miR-10b-5p shows a higher expression in metastatic breast cancer cells than in non-metastatic breast cancer or normal cells. Exosomal miR-10b-5p transmission enhances the invasion capacity of normal breast cancer [178]. The delivery of exosomal miR-10b-5p from gastric cancer cells also improves the proliferation of fibroblasts [179].
In contrast, some exosomal miRNAs show proliferation-/invasion-suppressing effects. A vitamin D analog, eldecalcitol (ED-71)-induced exosomal miR-6887-5p, suppresses oral cancer cell proliferation by targeting the 3′-UTR of heparin-binding protein 17/fibroblast growth-factor-binding protein-1 (HBp17/FGFBP-1) [180]. Exosomal miR-3180-3p suppresses lung cancer proliferation and metastasis by targeting forkhead box P4 (FOXP4) [181].

5.7. Overview of the Natural Products and Their Modulating Exosomal miRNAs That Regulate Exosomal Processing

The connections of natural products with their exosomal miRNA-regulated cell functions are summarized in Table 3. However, the impacts of exosomal processing, their genes related to these natural products, and their modulated exosomal miRNAs remain unclear. Utilizing the miRDB database [31], the target prediction of the exosome-processing genes for these natural-product-modulated exosomal miRNAs (Table 3) was performed. From exosomal assembly to secretion, the exosomal processing genes targeted by natural-product-modulated exosomal miRNAs were plotted (Figure 3). This warrants a careful investigation of the predicted targets of these natural-product-modulated exosomal miRNAs based on experiments in the future.

6. Conclusions

Tumor-derived exosomes containing many biomolecules can regulate sophisticated cell functions. This review focused on our understanding of the roles of exosomal miRNAs in controlling cancer cell functions. The impacts of the modulating effects of natural products in regulating exosome processing and exosomal miRNAs were also summarized.
Many natural products exhibit diverse functions and affect the expression of many genes, but they the impacts of natural products on exosome biogenesis have been overlooked. By examining exosomal processing information derived from the GO database and PubMed/Google scholar searches, we noted that some of the altered genes belong to the classification of exosomal processing. Accordingly, this work represents a novel contribution to the study of the exosomal processing of natural products.
Similarly, many exosomal miRNAs have been reported but lack detailed investigations of their regulation of exosomal processing. By utilizing the miRDB database, the potential impacts of exosomal processing genes were predicted to be targeted by exosomal miRNAs. This prediction further provides a direction for future research, which should aim to assess the detailed mechanisms of exosomal miRNAs, although further experiments are still required to confirm them.
Finally, we collected and organized several natural products and their associated modulations of exosomal miRNAs and cell functions, such as proliferation, apoptosis, migration, the tumor immune microenvironment, and other diverse effects. The potential roles of exosomal processing in these natural product investigations were further assessed using information retrieved from the miRDB database. Similarly, we demonstrated that some natural-product-modulated exosomal miRNAs overlap with tumor-immune-microenvironment-associated miRNAs. Although they are indirectly connected, this information provides a future direction for research, which should aim to validate whether these natural products can modulate exosomal miRNAs to regulate the tumor immune microenvironment.
Consequently, we offer a clear conclusion that several exosome-processing genes involved in exosomal secretion and assembly are organized in connection to natural products based on our utilization of the miRDB database to retrieve the target predictions of exosomal miRNAs. Accordingly, we filled the gaps in current knowledge between the exosomal processing of exosomal miRNAs and natural products.
Notably, the miRDB-database-predicted targets of exosomal processing genes were collected based on different cell types. Different cell types may show various miRNAs and targeting responses. This warrants careful examination based on wet experiments to validate the relationship between exosomal miRNAs and natural products in order to explore their impacts on the modulation of cancer cell functions.
This review sheds light on the connections between exosomes, exosomal miRNAs, natural products, and cancer cell functions, providing a clear direction for future research on the modulation of exosomal miRNAs by natural products.

Author Contributions

Conceptualization, Y.-T.C., J.-Y.T., A.A.F. and H.-W.C.; methodology, Y.-T.C., J.-P.S., C.-Y.Y., F.-R.C., K.-H.Y. and M.-F.H.; supervision, A.A.F. and H.-W.C.; writing—original draft, Y.-T.C., J.-Y.T. and H.-W.C.; writing—review and editing, A.A.F. and H.-W.C. All authors have read and agreed to the published version of the manuscript.

Funding

This study was partly supported by funds from the Ministry of Science and Technology (MOST 111-2320-B-037-015-MY3 and MOST 110-2314-B-037-074-MY3), the Kaohsiung Medical University (KMU-DK(A)111008), and the Kaohsiung Medical University Research Center (KMU-TC108A04).

Acknowledgments

We: the authors, thank our colleague Hans-Uwe Dahms for editing the manuscript.

Conflicts of Interest

The authors declare that there are no conflict of interest.

References

  1. Zhang, C.; Ji, Q.; Yang, Y.; Li, Q.; Wang, Z. Exosome: Function and role in cancer metastasis and drug resistance. Technol. Cancer Res. Treat. 2018, 17, 1533033818763450. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Dilsiz, N. Role of exosomes and exosomal microRNAs in cancer. Future Sci. OA 2020, 6, FSO465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Thery, C.; Zitvogel, L.; Amigorena, S. Exosomes: Composition, biogenesis and function. Nat. Rev. Immunol. 2002, 2, 569–579. [Google Scholar] [CrossRef] [PubMed]
  4. Zhao, C.; Zhang, G.; Liu, J.; Zhang, C.; Yao, Y.; Liao, W. Exosomal cargoes in OSCC: Current findings and potential functions. PeerJ 2020, 8, e10062. [Google Scholar] [CrossRef] [PubMed]
  5. Feng, S.; Lou, K.; Zou, X.; Zou, J.; Zhang, G. The potential role of exosomal proteins in prostate cancer. Front. Oncol. 2022, 12, 873296. [Google Scholar] [CrossRef] [PubMed]
  6. Liang, Y.; Duan, L.; Lu, J.; Xia, J. Engineering exosomes for targeted drug delivery. Theranostics 2021, 11, 3183–3195. [Google Scholar] [CrossRef]
  7. Jan, A.T.; Rahman, S.; Badierah, R.; Lee, E.J.; Mattar, E.H.; Redwan, E.M.; Choi, I. Expedition into exosome biology: A perspective of progress from discovery to therapeutic development. Cancers 2021, 13, 1157. [Google Scholar] [CrossRef]
  8. Kalluri, R.; LeBleu, V.S. The biology, function, and biomedical applications of exosomes. Science 2020, 367, eaau6977. [Google Scholar] [CrossRef]
  9. Gurunathan, S.; Kang, M.H.; Jeyaraj, M.; Qasim, M.; Kim, J.H. Review of the isolation, characterization, biological function, and multifarious therapeutic approaches of exosomes. Cells 2019, 8, 307. [Google Scholar] [CrossRef] [Green Version]
  10. Pegtel, D.M.; Gould, S.J. Exosomes. Annu. Rev. Biochem. 2019, 88, 487–514. [Google Scholar] [CrossRef]
  11. Farooqi, A.A.; Desai, N.N.; Qureshi, M.Z.; Librelotto, D.R.N.; Gasparri, M.L.; Bishayee, A.; Nabavi, S.M.; Curti, V.; Daglia, M. Exosome biogenesis, bioactivities and functions as new delivery systems of natural compounds. Biotechnol. Adv. 2018, 36, 328–334. [Google Scholar] [CrossRef] [PubMed]
  12. Teng, F.; Fussenegger, M. Shedding light on extracellular vesicle biogenesis and bioengineering. Adv. Sci. 2020, 8, 2003505. [Google Scholar] [CrossRef] [PubMed]
  13. Hu, S.; Liu, Y.; Guan, S.; Qiu, Z.; Liu, D. Natural products exert anti-tumor effects by regulating exosomal ncRNA. Front. Oncol. 2022, 12, 1006114. [Google Scholar] [CrossRef] [PubMed]
  14. Cai, Y.; Yu, X.; Hu, S.; Yu, J. A brief review on the mechanisms of miRNA regulation. Genom. Proteom. Bioinform. 2009, 7, 147–154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Bhome, R.; Del Vecchio, F.; Lee, G.H.; Bullock, M.D.; Primrose, J.N.; Sayan, A.E.; Mirnezami, A.H. Exosomal microRNAs (exomiRs): Small molecules with a big role in cancer. Cancer Lett. 2018, 420, 228–235. [Google Scholar] [CrossRef]
  16. Lu, Y.; Zheng, Z.; Yuan, Y.; Pathak, J.L.; Yang, X.; Wang, L.; Ye, Z.; Cho, W.C.; Zeng, M.; Wu, L. The emerging role of exosomes in oral squamous cell carcinoma. Front. Cell Dev. Biol. 2021, 9, 628103. [Google Scholar] [CrossRef]
  17. Dhar, R.; Mallik, S.; Devi, A. Exosomal microRNAs (exoMIRs): Micromolecules with macro impact in oral cancer. 3 Biotech 2022, 12, 155. [Google Scholar] [CrossRef]
  18. Li, Y.; Gao, S.; Hu, Q.; Wu, F. Functional properties of cancer epithelium and stroma-derived exosomes in head and neck squamous cell carcinoma. Life 2022, 12, 757. [Google Scholar] [CrossRef]
  19. St-Denis-Bissonnette, F.; Khoury, R.; Mediratta, K.; El-Sahli, S.; Wang, L.; Lavoie, J.R. Applications of extracellular vesicles in triple-negative breast cancer. Cancers 2022, 14, 451. [Google Scholar] [CrossRef]
  20. Lorenc, T.; Klimczyk, K.; Michalczewska, I.; Slomka, M.; Kubiak-Tomaszewska, G.; Olejarz, W. Exosomes in prostate cancer diagnosis, prognosis and therapy. Int. J. Mol. Sci. 2020, 21, 2118. [Google Scholar] [CrossRef]
  21. Li, X.; Jiang, W.; Gan, Y.; Zhou, W. The application of exosomal microRNAs in the treatment of pancreatic cancer and its research progress. Pancreas 2021, 50, 12–16. [Google Scholar] [CrossRef] [PubMed]
  22. Babaker, M.A.; Aljoud, F.A.; Alkhilaiwi, F.; Algarni, A.; Ahmed, A.; Khan, M.I.; Saadeldin, I.M.; Alzahrani, F.A. The Therapeutic Potential of Milk Extracellular Vesicles on Colorectal Cancer. Int. J. Mol. Sci. 2022, 23, 6812. [Google Scholar] [CrossRef] [PubMed]
  23. Hashemipour, M.; Boroumand, H.; Mollazadeh, S.; Tajiknia, V.; Nourollahzadeh, Z.; Rohani Borj, M.; Pourghadamyari, H.; Rahimian, N.; Hamblin, M.R.; Mirzaei, H. Exosomal microRNAs and exosomal long non-coding RNAs in gynecologic cancers. Gynecol. Oncol. 2021, 161, 314–327. [Google Scholar] [CrossRef] [PubMed]
  24. Sun, W.; Fu, S.; Wu, S.; Tu, R. Growing evidence of exosomal microRNA-related metastasis of hepatocellular carcinoma. BioMed Res. Int. 2020, 2020, 4501454. [Google Scholar] [CrossRef] [PubMed]
  25. Bhattacharjee, R.; Mitra, P.; Gupta, N.; Sharma, S.; Singh, V.K.; Mukerjee, N.; Dhasmana, A.; Gundamaraju, R. Cellular landscaping of exosomal miRNAs in cancer metastasis: From chemoresistance to prognostic markers. Adv. Cancer Biol.-Metastasis 2022, 5, 100050. [Google Scholar] [CrossRef]
  26. Sorop, A.; Constantinescu, D.; Cojocaru, F.; Dinischiotu, A.; Cucu, D.; Dima, S.O. Exosomal microRNAs as biomarkers and therapeutic targets for hepatocellular carcinoma. Int. J. Mol. Sci. 2021, 22, 4997. [Google Scholar] [CrossRef] [PubMed]
  27. Hu, M.; Cao, Z.; Jiang, D. The effect of miRNA-modified exosomes in animal models of spinal cord injury: A meta-analysis. Front. Bioeng Biotechnol 2021, 9, 819651. [Google Scholar] [CrossRef]
  28. Cetin, Z.; Saygili, E.I.; Gorgisen, G.; Sokullu, E. Preclinical experimental applications of miRNA loaded BMSC extracellular vesicles. Stem Cell Rev. Rep. 2021, 17, 471–501. [Google Scholar] [CrossRef]
  29. Rezaei, R.; Baghaei, K.; Hashemi, S.M.; Zali, M.R.; Ghanbarian, H.; Amani, D. Tumor-derived exosomes enriched by miRNA-124 promote anti-tumor immune response in CT-26 tumor-bearing mice. Front. Med. 2021, 8, 619939. [Google Scholar] [CrossRef]
  30. Guglielmi, L.; Nardella, M.; Musa, C.; Cifola, I.; Porru, M.; Cardinali, B.; Iannetti, I.; Di Pietro, C.; Bolasco, G.; Palmieri, V.; et al. Circulating miRNAs in small extracellular vesicles secreted by a human melanoma xenograft in mouse brains. Cancers 2020, 12, 1635. [Google Scholar] [CrossRef]
  31. Chen, Y.; Wang, X. miRDB: An online database for prediction of functional microRNA targets. Nucleic Acids Res. 2020, 48, D127–D131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Bult, C.J.; Blake, J.A.; Smith, C.L.; Kadin, J.A.; Richardson, J.E.; Mouse Genome Database, G. Mouse Genome Database (MGD) 2019. Nucleic Acids Res. 2019, 47, D801–D806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Hasanin, A.H.; Matboli, M.; Seleem, H.S. Hesperidin suppressed hepatic precancerous lesions via modulation of exophagy in rats. J. Cell. Biochem. 2020, 121, 1295–1306. [Google Scholar] [CrossRef] [PubMed]
  34. Li, Y.Q.; Zhang, F.; Yu, L.P.; Mu, J.K.; Yang, Y.Q.; Yu, J.; Yang, X.X. Targeting PINK1 using natural products for the treatment of human diseases. BioMed Res. Int. 2021, 2021, 4045819. [Google Scholar] [CrossRef] [PubMed]
  35. Tang, C.; Han, H.; Yan, M.; Zhu, S.; Liu, J.; Liu, Z.; He, L.; Tan, J.; Liu, Y.; Liu, H.; et al. PINK1-PRKN/PARK2 pathway of mitophagy is activated to protect against renal ischemia-reperfusion injury. Autophagy 2018, 14, 880–897. [Google Scholar] [CrossRef] [Green Version]
  36. Hu, Y.; Wang, M.; Pan, Y.; Li, Q.; Xu, L. Salvianolic acid B attenuates renal interstitial fibrosis by regulating the HPSE/SDC1 axis. Mol. Med. Rep. 2020, 22, 1325–1334. [Google Scholar] [CrossRef] [PubMed]
  37. Dall’Acqua, S.; Grabnar, I.; Verardo, R.; Klaric, E.; Marchionni, L.; Luidy-Imada, E.; Sut, S.; Agostinis, C.; Bulla, R.; Perissutti, B.; et al. Combined extracts of Echinacea angustifolia DC. and Zingiber officinale Roscoe in softgel capsules: Pharmacokinetics and immunomodulatory effects assessed by gene expression profiling. Phytomedicine Int. J. Phytother. Phytopharm. 2019, 65, 153090. [Google Scholar] [CrossRef] [PubMed]
  38. Dong, X.; Zhu, Y.; Wang, S.; Luo, Y.; Lu, S.; Nan, F.; Sun, G.; Sun, X. Bavachinin inhibits cholesterol synthesis enzyme FDFT1 expression via AKT/mTOR/SREBP-2 pathway. Int. Immunopharmacol. 2020, 88, 106865. [Google Scholar] [CrossRef]
  39. Pandey, P.; Khan, F. Jab1 inhibition by methanolic extract of Moringa oleifera leaves in cervical cancer cells: A potent targeted therapeutic approach. Nutr. Cancer 2021, 73, 2411–2419. [Google Scholar] [CrossRef]
  40. Pandey, P.; Khan, F.; Alzahrani, F.A.; Qari, H.A.; Oves, M. A novel approach to unraveling the apoptotic potential of rutin (bioflavonoid) via targeting Jab1 in cervical cancer cells. Molecules 2021, 26, 5529. [Google Scholar] [CrossRef]
  41. Kaftan, G.; Erdoğan, M.; El-Shazly, M.; Lu, M.-C.; Shih, S.-P.; Lin, H.-Y.; Saso, L.; Armagan, G. Heteronemin promotes iron-dependent cell death in pancreatic cancer. Res. Sq. 2022. [Google Scholar] [CrossRef]
  42. Ba, Q.; Zhou, N.; Duan, J.; Chen, T.; Hao, M.; Yang, X.; Li, J.; Yin, J.; Chu, R.; Wang, H. Dihydroartemisinin exerts its anticancer activity through depleting cellular iron via transferrin receptor-1. PloS ONE 2012, 7, e42703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Xie, Y.; Zhou, X.; Li, J.; Yao, X.C.; Liu, W.L.; Kang, F.H.; Zou, Z.X.; Xu, K.P.; Xu, P.S.; Tan, G.S. Identification of a new natural biflavonoids against breast cancer cells induced ferroptosis via the mitochondrial pathway. Bioorganic Chem. 2021, 109, 104744. [Google Scholar] [CrossRef] [PubMed]
  44. Zhou, J.; Li, G.; Zheng, Y.; Shen, H.M.; Hu, X.; Ming, Q.L.; Huang, C.; Li, P.; Gao, N. A novel autophagy/mitophagy inhibitor liensinine sensitizes breast cancer cells to chemotherapy through DNM1L-mediated mitochondrial fission. Autophagy 2015, 11, 1259–1279. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Im, E.J.; Lee, C.H.; Moon, P.G.; Rangaswamy, G.G.; Lee, B.; Lee, J.M.; Lee, J.C.; Jee, J.G.; Bae, J.S.; Kwon, T.K.; et al. Sulfisoxazole inhibits the secretion of small extracellular vesicles by targeting the endothelin receptor A. Nat. Commun. 2019, 10, 1387. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Si, J.; Van den Haute, C.; Lobbestael, E.; Martin, S.; van Veen, S.; Vangheluwe, P.; Baekelandt, V. ATP13A2 regulates cellular alpha-synuclein multimerization, membrane association, and externalization. Int. J. Mol. Sci. 2021, 22, 2689. [Google Scholar] [CrossRef]
  47. Chimplee, S.; Roytrakul, S.; Sukrong, S.; Srisawat, T.; Graidist, P.; Kanokwiroon, K. Anticancer effects and molecular action of 7-alpha-hydroxyfrullanolide in G2/M-phase arrest and apoptosis in triple negative breast cancer cells. Molecules 2022, 27, 407. [Google Scholar] [CrossRef]
  48. Liu, Y.; Chen, Y.; Zhu, R.; Xu, L.; Xie, H.Q.; Zhao, B. Rutaecarpine inhibits U87 glioblastoma cell migration by activating the aryl hydrocarbon receptor signaling pathway. Front. Mol. Neurosci. 2021, 14, 765712. [Google Scholar] [CrossRef]
  49. Mao, Z.; Han, X.; Chen, D.; Xu, Y.; Xu, L.; Yin, L.; Sun, H.; Qi, Y.; Fang, L.; Liu, K.; et al. Potent effects of dioscin against hepatocellular carcinoma through regulating TP53-induced glycolysis and apoptosis regulator (TIGAR)-mediated apoptosis, autophagy, and DNA damage. Br. J. Pharmacol. 2019, 176, 919–937. [Google Scholar] [CrossRef] [Green Version]
  50. Shang, H.S.; Shih, Y.L.; Lee, C.H.; Hsueh, S.C.; Liu, J.Y.; Liao, N.C.; Chen, Y.L.; Huang, Y.P.; Lu, H.F.; Chung, J.G. Sulforaphane-induced apoptosis in human leukemia HL-60 cells through extrinsic and intrinsic signal pathways and altering associated genes expression assayed by cDNA microarray. Environ. Toxicol. 2017, 32, 311–328. [Google Scholar] [CrossRef]
  51. Shen, Y.; Takahashi, M.; Byun, H.M.; Link, A.; Sharma, N.; Balaguer, F.; Leung, H.C.; Boland, C.R.; Goel, A. Boswellic acid induces epigenetic alterations by modulating DNA methylation in colorectal cancer cells. Cancer Biol. Ther. 2012, 13, 542–552. [Google Scholar] [CrossRef] [Green Version]
  52. Mondal, S.; Mandal, C.; Sangwan, R.; Chandra, S.; Mandal, C. Withanolide D induces apoptosis in leukemia by targeting the activation of neutral sphingomyelinase-ceramide cascade mediated by synergistic activation of c-Jun N-terminal kinase and p38 mitogen-activated protein kinase. Mol. Cancer 2010, 9, 239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Schneider, Y.; Duranton, B.; Gosse, F.; Schleiffer, R.; Seiler, N.; Raul, F. Resveratrol inhibits intestinal tumorigenesis and modulates host-defense-related gene expression in an animal model of human familial adenomatous polyposis. Nutr. Cancer 2001, 39, 102–107. [Google Scholar] [CrossRef] [PubMed]
  54. Han, L.; Liu, N.; Yang, L.; Mao, Y.; Ye, S. Astragalus membranaceus extract promotes angiogenesis by inducing VEGF, CD34 and eNOS expression in rats subjected to myocardial infarction. Int. J. Clin. Exp. Med. 2016, 9, 5709–5718. [Google Scholar]
  55. Chen, W.X.; Xu, L.Y.; Qian, Q.; He, X.; Peng, W.T.; Fan, W.Q.; Zhu, Y.L.; Tang, J.H.; Cheng, L. d Rhamnose beta-hederin reverses chemoresistance of breast cancer cells by regulating exosome-mediated resistance transmission. Biosci. Rep. 2018, 38, BSR20180110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Perni, M.; Galvagnion, C.; Maltsev, A.; Meisl, G.; Muller, M.B.; Challa, P.K.; Kirkegaard, J.B.; Flagmeier, P.; Cohen, S.I.; Cascella, R.; et al. A natural product inhibits the initiation of alpha-synuclein aggregation and suppresses its toxicity. Proc. Natl. Acad. Sci. USA 2017, 114, E1009–E1017. [Google Scholar] [CrossRef] [Green Version]
  57. Salehi, M.; Vafadar, A.; Khatami, S.H.; Taheri-Anganeh, M.; Vakili, O.; Savardashtaki, A.; Negahdari, B.; Naeli, P.; Behrouj, H.; Ghasemi, H.; et al. Gastrointestinal cancer drug resistance: The role of exosomal miRNAs. Mol. Biol. Rep. 2022, 49, 2421–2432. [Google Scholar] [CrossRef]
  58. Shi, L.; Zhu, W.; Huang, Y.; Zhuo, L.; Wang, S.; Chen, S.; Zhang, B.; Ke, B. Cancer-associated fibroblast-derived exosomal microRNA-20a suppresses the PTEN/PI3K-AKT pathway to promote the progression and chemoresistance of non-small cell lung cancer. Clin. Transl. Med. 2022, 12, e989. [Google Scholar] [CrossRef]
  59. Zhang, L.; Ouyang, P.; He, G.; Wang, X.; Song, D.; Yang, Y.; He, X. Exosomes from microRNA-126 overexpressing mesenchymal stem cells promote angiogenesis by targeting the PIK3R2-mediated PI3K/Akt signalling pathway. J. Cell. Mol. Med. 2021, 25, 2148–2162. [Google Scholar] [CrossRef]
  60. Sun, J.; Wei, J.; Zhang, Y.; Li, J.; Li, J.; Yan, J.; Guo, M.; Han, J.; Qiao, H. Plasma exosomes transfer miR-885-3p targeting the AKT/NFkappaB signaling pathway to improve the sensitivity of intravenous glucocorticoid therapy against Graves ophthalmopathy. Front. Immunol. 2022, 13, 819680. [Google Scholar] [CrossRef]
  61. Bae, I.S.; Kim, S.H. Milk exosome-derived microRNA-2478 suppresses melanogenesis through the Akt-GSK3β pathway. Cells 2021, 10, 2848. [Google Scholar] [CrossRef] [PubMed]
  62. Chen, F.; Wang, S.; Wei, Y.; Wu, J.; Huang, G.; Chen, J.; Shi, J.; Xia, J. Norcantharidin modulates the miR-30a/Metadherin/AKT signaling axis to suppress proliferation and metastasis of stromal tumor cells in giant cell tumor of bone. Biomed. Pharmacother. 2018, 103, 1092–1100. [Google Scholar] [CrossRef] [PubMed]
  63. Ali, N.B.; Abdull Razis, A.F.; Ooi, J.; Chan, K.W.; Ismail, N.; Foo, J.B. Theragnostic applications of mammal and plant-derived extracellular vesicles: Latest findings, current technologies, and prospects. Molecules 2022, 27, 3941. [Google Scholar] [CrossRef] [PubMed]
  64. Otsuka, K.; Yamamoto, Y.; Matsuoka, R.; Ochiya, T. Maintaining good miRNAs in the body keeps the doctor away?: Perspectives on the relationship between food-derived natural products and microRNAs in relation to exosomes/extracellular vesicles. Mol. Nutr. Food Res. 2017. Available online: https://onlinelibrary.wiley.com/doi/10.1002/mnfr.201700080. [CrossRef]
  65. Song, H.; Liu, B.; Dong, B.; Xu, J.; Zhou, H.; Na, S.; Liu, Y.; Pan, Y.; Chen, F.; Li, L.; et al. Exosome-based delivery of natural products in cancer therapy. Front. Cell Dev. Biol. 2021, 9, 650426. [Google Scholar] [CrossRef]
  66. Xu, Y.; Feng, K.; Zhao, H.; Di, L.; Wang, L.; Wang, R. Tumor-derived extracellular vesicles as messengers of natural products in cancer treatment. Theranostics 2022, 12, 1683–1714. [Google Scholar] [CrossRef]
  67. Zheng, K.; Ma, J.; Wang, Y.; He, Z.; Deng, K. Sulforaphane inhibits autophagy and induces exosome-mediated paracrine senescence via regulating mTOR/TFE3. Mol. Nutr. Food Res. 2020, 64, e1901231. [Google Scholar] [CrossRef]
  68. Komatsu, M.; Waguri, S.; Koike, M.; Sou, Y.S.; Ueno, T.; Hara, T.; Mizushima, N.; Iwata, J.; Ezaki, J.; Murata, S.; et al. Homeostatic levels of p62 control cytoplasmic inclusion body formation in autophagy-deficient mice. Cell 2007, 131, 1149–1163. [Google Scholar] [CrossRef] [Green Version]
  69. Xu, J.; Camfield, R.; Gorski, S.M. The interplay between exosomes and autophagy-partners in crime. J. Cell Sci. 2018, 131, jcs215210. [Google Scholar] [CrossRef] [Green Version]
  70. Mitani, F.; Lin, J.; Sakamoto, T.; Uehara, R.; Hikita, T.; Yoshida, T.; Setiawan, A.; Arai, M.; Oneyama, C. Asteltoxin inhibits extracellular vesicle production through AMPK/mTOR-mediated activation of lysosome function. Sci. Rep. 2022, 12, 6674. [Google Scholar] [CrossRef]
  71. Gu, S.; Song, X.; Xie, R.; Ouyang, C.; Xie, L.; Li, Q.; Su, T.; Xu, M.; Xu, T.; Huang, D.; et al. Berberine inhibits cancer cells growth by suppressing fatty acid synthesis and biogenesis of extracellular vesicles. Life Sci. 2020, 257, 118122. [Google Scholar] [CrossRef]
  72. Shiau, J.Y.; Chang, Y.Q.; Nakagawa-Goto, K.; Lee, K.H.; Shyur, L.F. Phytoagent deoxyelephantopin and its derivative inhibit triple negative breast cancer cell activity through ROS-mediated exosomal activity and protein functions. Front. Pharmacol. 2017, 8, 398. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Wang, B.; Guo, X.J.; Cai, H.; Zhu, Y.H.; Huang, L.Y.; Wang, W.; Luo, L.; Qi, S.H. Momordica charantia-derived extracellular vesicles-like nanovesicles inhibited glioma proliferation, migration, and invasion by regulating the PI3K/AKT signaling pathway. J. Funct. Foods 2022, 90, 104968. [Google Scholar] [CrossRef]
  74. Lin, L.T.; Shi, Y.C.; Choong, C.Y.; Tai, C.J. The fruits of Paris polyphylla inhibit colorectal cancer cell migration induced by Fusobacterium nucleatum-derived extracellular vesicles. Molecules 2021, 26, 4081. [Google Scholar] [CrossRef] [PubMed]
  75. Srivastava, A.; Rathore, S.; Munshi, A.; Ramesh, R. Organically derived exosomes as carriers of anticancer drugs and imaging agents for cancer treatment. Semin. Cancer Biol. 2022, 86, 80–100. [Google Scholar] [CrossRef]
  76. Agrawal, A.K.; Aqil, F.; Jeyabalan, J.; Spencer, W.A.; Beck, J.; Gachuki, B.W.; Alhakeem, S.S.; Oben, K.; Munagala, R.; Bondada, S.; et al. Milk-derived exosomes for oral delivery of paclitaxel. Nanomed. Nanotechnol. Biol. Med. 2017, 13, 1627–1636. [Google Scholar] [CrossRef]
  77. Sun, D.; Zhuang, X.; Xiang, X.; Liu, Y.; Zhang, S.; Liu, C.; Barnes, S.; Grizzle, W.; Miller, D.; Zhang, H.G. A novel nanoparticle drug delivery system: The anti-inflammatory activity of curcumin is enhanced when encapsulated in exosomes. Mol. Ther. J. Am. Soc. Gene Ther. 2010, 18, 1606–1614. [Google Scholar] [CrossRef]
  78. Aqil, F.; Kausar, H.; Agrawal, A.K.; Jeyabalan, J.; Kyakulaga, A.H.; Munagala, R.; Gupta, R. Exosomal formulation enhances therapeutic response of celastrol against lung cancer. Exp. Mol. Pathol. 2016, 101, 12–21. [Google Scholar] [CrossRef]
  79. Zhang, B.; Tian, L.; Xie, J.; Chen, G.; Wang, F. Targeting miRNAs by natural products: A new way for cancer therapy. Biomed. Pharmacother. 2020, 130, 110546. [Google Scholar] [CrossRef]
  80. Park, Y.; Lee, K.; Kim, S.W.; Lee, M.W.; Kim, B.; Lee, S.G. Effects of induced exosomes from endometrial cancer cells on tumor activity in the presence of Aurea helianthus extract. Molecules 2021, 26, 2207. [Google Scholar] [CrossRef]
  81. Wang, J.; Li, Y.; Wang, X.; Jiang, C. Ursolic acid inhibits proliferation and induces apoptosis in human glioblastoma cell lines U251 by suppressing TGF-β1/miR-21/PDCD 4 pathway. Basic Clin. Pharmacol. Toxicol. 2012, 111, 106–112. [Google Scholar] [PubMed]
  82. Zhang, L.; Cai, Q.Y.; Liu, J.; Peng, J.; Chen, Y.Q.; Sferra, T.J.; Lin, J.M. Ursolic acid suppresses the invasive potential of colorectal cancer cells by regulating the TGF-beta1/ZEB1/miR-200c signaling pathway. Oncol. Lett. 2019, 18, 3274–3282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Zhou, C.; Ding, J.; Wu, Y. Resveratrol induces apoptosis of bladder cancer cells via miR21 regulation of the Akt/Bcl2 signaling pathway. Mol. Med. Rep. 2014, 9, 1467–1473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Alrafas, H.R.; Busbee, P.B.; Nagarkatti, M.; Nagarkatti, P.S. Resveratrol downregulates miR-31 to promote to regulatory cells during prevention of TNBS-induced colitis. Mol. Nutr. Food Res. 2020, 64, e1900633. [Google Scholar] [CrossRef]
  85. Otsuka, K.; Yamamoto, Y.; Ochiya, T. Regulatory role of resveratrol, a microRNA-controlling compound, in HNRNPA1 expression, which is associated with poor prognosis in breast cancer. Oncotarget 2018, 9, 24718–24730. [Google Scholar] [CrossRef] [Green Version]
  86. Luo, X.; Gu, J.; Zhu, R.; Feng, M.; Zhu, X.; Li, Y.; Fei, J. Integrative analysis of differential miRNA and functional study of miR-21 by seed-targeting inhibition in multiple myeloma cells in response to berberine. BMC Syst. Biol. 2014, 8, 82. [Google Scholar] [CrossRef] [Green Version]
  87. Gao, Z.; Tan, C.; Sha, R. Berberine Promotes A549 Cell Apoptosis and Autophagy via miR-144. Nat. Prod. Commun. 2022, 17, 1934578–221124752. [Google Scholar] [CrossRef]
  88. Wang, Y.; Zhang, S. Berberine suppresses growth and metastasis of endometrial cancer cells via miR-101/COX-2. Biomed. Pharmacother. Biomed. Pharmacother. 2018, 103, 1287–1293. [Google Scholar] [CrossRef]
  89. Matarlo, J.S.; Krumpe, L.R.H.; Heinz, W.F.; Oh, D.; Shenoy, S.R.; Thomas, C.L.; Goncharova, E.I.; Lockett, S.J.; O’Keefe, B.R. The natural product butylcycloheptyl prodiginine binds pre-miR-21, inhibits Dicer-mediated processing of pre-miR-21, and blocks cellular proliferation. Cell Chem. Biol. 2019, 26, 1133–1142.e1134. [Google Scholar] [CrossRef]
  90. Yang, J.; Zou, Y.; Jiang, D. Honokiol suppresses proliferation and induces apoptosis via regulation of the miR21/PTEN/PI3K/AKT signaling pathway in human osteosarcoma cells. Int. J. Mol. Med. 2018, 41, 1845–1854. [Google Scholar] [CrossRef] [Green Version]
  91. Avtanski, D.B.; Nagalingam, A.; Bonner, M.Y.; Arbiser, J.L.; Saxena, N.K.; Sharma, D. Honokiol activates LKB1-miR-34a axis and antagonizes the oncogenic actions of leptin in breast cancer. Oncotarget 2015, 6, 29947–29962. [Google Scholar] [CrossRef]
  92. Liu, W.; Zhang, B.; Chen, G.; Wu, W.; Zhou, L.; Shi, Y.; Zeng, Q.; Li, Y.; Sun, Y.; Deng, X.; et al. Targeting miR-21 with sophocarpine inhibits tumor progression and reverses epithelial-mesenchymal transition in head and neck cancer. Mol. Ther. J. Am. Soc. Gene Ther. 2017, 25, 2129–2139. [Google Scholar] [CrossRef] [Green Version]
  93. Ghasemi, S.; Lorigooini, Z.; Wibowo, J.; Amini-Khoei, H. Tricin isolated from Allium atroviolaceum potentiated the effect of docetaxel on PC3 cell proliferation: Role of miR-21. Nat. Prod. Res. 2019, 33, 1828–1831. [Google Scholar] [CrossRef] [PubMed]
  94. Chen, L.; Yang, Z.S.; Zhou, Y.Z.; Deng, Y.; Jiang, P.; Tan, S.L. Dihydromyricetin inhibits cell proliferation, migration, invasion and promotes apoptosis via regulating miR-21 in human cholangiocarcinoma cells. J. Cancer 2020, 11, 5689–5699. [Google Scholar] [CrossRef] [PubMed]
  95. Zhou, C.; Hu, C.; Wang, B.; Fan, S.; Jin, W. Curcumin suppresses cell proliferation, migration, and invasion through modulating miR-21-5p/SOX6 axis in hepatocellular carcinoma. Cancer Biother. Radiopharm. 2020. [Google Scholar] [CrossRef] [PubMed]
  96. Wang, H.; Cai, X.; Ma, L. Curcumin modifies epithelial–mesenchymal transition in colorectal cancer through regulation of miR-200c/EPM5. Cancer Manag. Res. 2020, 12, 9405–9415. [Google Scholar] [CrossRef]
  97. Liu, L.; Fu, Y.; Zheng, Y.; Ma, M.; Wang, C. Curcumin inhibits proteasome activity in triple-negative breast cancer cells through regulating p300/miR-142-3p/PSMB5 axis. Phytomedicine Int. J. Phytother. Phytopharm. 2020, 78, 153312. [Google Scholar] [CrossRef]
  98. Xu, R.; Li, H.; Wu, S.; Qu, J.; Yuan, H.; Zhou, Y.; Lu, Q. MicroRNA-1246 regulates the radio-sensitizing effect of curcumin in bladder cancer cells via activating P53. Int. Urol. Nephrol. 2019, 51, 1771–1779. [Google Scholar] [CrossRef]
  99. Liu, H.; Wang, J.; Tao, Y.; Li, X.; Qin, J.; Bai, Z.; Chi, B.; Yan, W.; Chen, X. Curcumol inhibits colorectal cancer proliferation by targeting miR-21 and modulated PTEN/PI3K/Akt pathways. Life Sci. 2019, 221, 354–361. [Google Scholar] [CrossRef]
  100. He, J.Q.; Zheng, M.X.; Ying, H.Z.; Zhong, Y.S.; Zhang, H.H.; Xu, M.; Yu, C.H. PRP1, a heteropolysaccharide from Platycodonis Radix, induced apoptosis of HepG2 cells via regulating miR-21-mediated PI3K/AKT pathway. Int. J. Biol. Macromol. 2020, 158, 542–551. [Google Scholar] [CrossRef]
  101. Shen, K.H.; Hung, J.H.; Liao, Y.C.; Tsai, S.T.; Wu, M.J.; Chen, P.S. Sinomenine inhibits migration and invasion of human lung cancer cell through downregulating expression of miR-21 and MMPs. Int. J. Mol. Sci. 2020, 21, 3080. [Google Scholar] [CrossRef] [PubMed]
  102. Jin, L.; Ma, X.M.; Wang, T.T.; Yang, Y.; Zhang, N.; Zeng, N.; Bai, Z.G.; Yin, J.; Zhang, J.; Ding, G.Q.; et al. Psoralen suppresses cisplatin-mediated resistance and induces apoptosis of gastric adenocarcinoma by disruption of the miR196a-HOXB7-HER2 axis. Cancer Manag. Res. 2020, 12, 2803–2827. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Takala, R.; Ramji, D.P.; Andrews, R.; Zhou, Y.; Farhat, M.; Elmajee, M.; Rundle, S.; Choy, E. Pinolenic acid exhibits anti-inflammatory and anti-atherogenic effects in peripheral blood-derived monocytes from patients with rheumatoid arthritis. Sci. Rep. 2022, 12, 8807. [Google Scholar] [CrossRef] [PubMed]
  104. Younis, N.N.; Salama, A.; Shaheen, M.A.; Eissa, R.G. Pachymic acid attenuated doxorubicin-induced heart failure by suppressing miR-24 and preserving cardiac junctophilin-2 in rats. Int. J. Mol. Sci. 2021, 22, 10710. [Google Scholar] [CrossRef] [PubMed]
  105. de la Parra, C.; Castillo-Pichardo, L.; Cruz-Collazo, A.; Cubano, L.; Redis, R.; Calin, G.A.; Dharmawardhane, S. Soy isoflavone genistein-mediated downregulation of miR-155 contributes to the anticancer effects of genistein. Nutr. Cancer 2016, 68, 154–164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. La, X.; Zhang, L.; Li, Z.; Li, H.; Yang, Y. (-)-Epigallocatechin gallate (EGCG) enhances the sensitivity of colorectal cancer cells to 5-FU by inhibiting GRP78/NF-kappaB/miR-155-5p/MDR1 pathway. J. Agric. Food Chem. 2019, 67, 2510–2518. [Google Scholar] [CrossRef] [PubMed]
  107. Kang, Q.; Zhang, X.; Cao, N.; Chen, C.; Yi, J.; Hao, L.; Ji, Y.; Liu, X.; Lu, J. EGCG enhances cancer cells sensitivity under (60)Cogamma radiation based on miR-34a/Sirt1/p53. Food Chem. Toxicol. 2019, 133, 110807. [Google Scholar] [CrossRef] [PubMed]
  108. Hagiwara, K.; Gailhouste, L.; Yasukawa, K.; Kosaka, N.; Ochiya, T. A robust screening method for dietary agents that activate tumour-suppressor microRNAs. Sci. Rep. 2015, 5, 14697. [Google Scholar] [CrossRef] [Green Version]
  109. Peng, F.; Xiong, L.; Peng, C. (-)-Sativan inhibits tumor development and regulates miR-200c/PD-L1 in triple negative breast cancer cells. Front. Pharmacol. 2020, 11, 251. [Google Scholar] [CrossRef]
  110. Peng, F.; Tang, H.; Du, J.; Chen, J.; Peng, C. Isoliquiritigenin suppresses EMT-induced metastasis in triple-negative breast cancer through miR-200c/c-Jun/β-Catenin. Am. J. Chin. Med. 2021, 49, 505–523. [Google Scholar] [CrossRef]
  111. Geng, W.; Li, C.; Zhan, Y.; Zhang, R.; Zheng, J. Thymoquinone alleviates liver fibrosis via miR-30a-mediated epithelial-mesenchymal transition. J. Cell. Physiol. 2020, 236, 3629–3640. [Google Scholar] [CrossRef] [PubMed]
  112. Wu, L.; Yang, K.; Gui, Y.; Wang, X. Nicotine-upregulated miR-30a arrests cell cycle in G1 phase by directly targeting CCNE2 in human periodontal ligament cells. Biochem. Cell Biol. 2020, 98, 354–361. [Google Scholar] [CrossRef] [PubMed]
  113. Phuah, N.H.; Azmi, M.N.; Awang, K.; Nagoor, N.H. Down-regulation of microRNA-210 confers sensitivity towards 1′s-1′-acetoxychavicol acetate (ACA) in cervical cancer cells by targeting SMAD4. Mol. Cells 2017, 40, 291–298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Mollaei, H.; Safaralizadeh, R.; Babaei, E.; Abedini, M.R.; Hoshyar, R. The anti-proliferative and apoptotic effects of crocin on chemosensitive and chemoresistant cervical cancer cells. Biomed. Pharmacother. Biomed. Pharmacother. 2017, 94, 307–316. [Google Scholar] [CrossRef] [PubMed]
  115. Tang, Y.; Yang, H.; Yu, J.; Li, Z.; Xu, Q.; Ding, B.; Jia, G. Crocin induces ROS-mediated papillary thyroid cancer cell apoptosis by modulating the miR-34a-5p/PTPN4 axis in vitro. Toxicol. Appl. Pharmacol. 2022, 437, 115892. [Google Scholar] [CrossRef] [PubMed]
  116. Hsia, S.M.; Yu, C.C.; Shih, Y.H.; Yuanchien Chen, M.; Wang, T.H.; Huang, Y.T.; Shieh, T.M. Isoliquiritigenin as a cause of DNA damage and inhibitor of ataxia-telangiectasia mutated expression leading to G2/M phase arrest and apoptosis in oral squamous cell carcinoma. Head Neck 2016, 38 (Suppl. 1), E360–E371. [Google Scholar] [CrossRef] [PubMed]
  117. Ye, W.; Ni, Z.; Yicheng, S.; Pan, H.; Huang, Y.; Xiong, Y.; Liu, T. Anisomycin inhibits angiogenesis in ovarian cancer by attenuating the molecular sponge effect of the lncRNAMeg3/miR421/PDGFRA axis. Int. J. Oncol. 2019, 55, 1296–1312. [Google Scholar] [CrossRef] [Green Version]
  118. Zhang, F.; Ni, Z.J.; Ye, L.; Zhang, Y.Y.; Thakur, K.; Cespedes-Acuna, C.L.; Han, J.; Zhang, J.G.; Wei, Z.J. Asparanin A inhibits cell migration and invasion in human endometrial cancer via Ras/ERK/MAPK pathway. Food Chem. Toxicol. 2021, 150, 112036. [Google Scholar] [CrossRef]
  119. Kang, J.; Kim, E.; Kim, W.; Seong, K.M.; Youn, H.; Kim, J.W.; Kim, J.; Youn, B. Rhamnetin and cirsiliol induce radiosensitization and inhibition of epithelial-mesenchymal transition (EMT) by miR-34a-mediated suppression of Notch-1 expression in non-small cell lung cancer cell lines. J. Biol. Chem. 2013, 288, 27343–27357. [Google Scholar] [CrossRef] [Green Version]
  120. Paccez, J.D.; Duncan, K.; Sekar, D.; Correa, R.G.; Wang, Y.; Gu, X.; Bashin, M.; Chibale, K.; Libermann, T.A.; Zerbini, L.F. Dihydroartemisinin inhibits prostate cancer via JARID2/miR-7/miR-34a-dependent downregulation of Axl. Oncogenesis 2019, 8, 14. [Google Scholar] [CrossRef] [Green Version]
  121. Liang, X.; Xu, C.; Cao, X.; Wang, W. Isovitexin suppresses cancer stemness property and induces apoptosis of osteosarcoma cells by disruption of the DNMT1/miR-34a/Bcl-2 axis. Cancer Manag. Res. 2019, 11, 8923–8936. [Google Scholar] [CrossRef]
  122. Bai, J.; Wu, J.; Tang, R.; Sun, C.; Ji, J.; Yin, Z.; Ma, G.; Yang, W. Emodin, a natural anthraquinone, suppresses liver cancer in vitro and in vivo by regulating VEGFR2 and miR-34a. Investig. New Drugs 2020, 38, 229–245. [Google Scholar] [CrossRef]
  123. Xiao, Y.; Liu, L.; Zheng, Y.; Liu, W.; Xu, Y. Kaempferol attenuates the effects of XIST/miR-130a/STAT3 on inflammation and extracellular matrix degradation in osteoarthritis. Future Med. Chem. 2021, 13, 1451–1464. [Google Scholar] [CrossRef]
  124. Yan, Z.; Huang, Q.-L.; Chen, J.; Liu, F.; Wei, Y.; Chen, S.-L.; Wu, C.-Y.; Li, Z.; Lin, X.-P. Chicoric acid alleviates LPS-induced inflammatory response through miR-130a-3p/IGF-1pathway in human lung A549 epithelial cells. Eur. J. Inflamm. 2021, 19, 20587392211038244. [Google Scholar] [CrossRef]
  125. Xu, T.; Qin, L.; Zhu, Z.; Wang, X.; Liu, Y.; Fan, Y.; Zhong, S.; Wang, X.; Zhang, X.; Xia, L.; et al. MicroRNA-31 functions as a tumor suppressor and increases sensitivity to mitomycin-C in urothelial bladder cancer by targeting integrin alpha5. Oncotarget 2016, 7, 27445–27457. [Google Scholar] [CrossRef]
  126. Chen, G.; Ma, Y.; Jiang, Z.; Feng, Y.; Han, Y.; Tang, Y.; Zhang, J.; Ni, H.; Li, X.; Li, N. Lico A causes ER stress and apoptosis via up-regulating miR-144-3p in human lung cancer cell line H292. Front. Pharmacol. 2018, 9, 837. [Google Scholar] [CrossRef]
  127. Zeng, L.; Sun, Y.; Li, X.; Wang, J.; Yan, L. 10Hydroxycamptothecin induces apoptosis in human fibroblasts by regulating miRNA23b3p expression. Mol. Med. Rep. 2019, 19, 2680–2686. [Google Scholar] [CrossRef] [Green Version]
  128. Bae, M.; Kim, M.B.; Lee, J.Y. Astaxanthin attenuates the changes in the expression of microRNAs involved in the activation of hepatic stellate cells. Nutrients 2022, 14, 962. [Google Scholar] [CrossRef]
  129. Jin, Y.; Zhan, X.B.; Zhang, B.; Chen, Y.; Liu, C.F.; Yu, L.L. Polydatin exerts an antitumor effect through regulating the miR-382/PD-L1 axis in colorectal cancer. Cancer Biother. Radiopharm. 2020, 35, 83–91. [Google Scholar] [CrossRef]
  130. Hu, Y.; Luo, X.; Zhou, J.; Chen, S.; Gong, M.; Deng, Y.; Zhang, H. Piperlongumine inhibits the progression of osteosarcoma by downregulating the SOCS3/JAK2/STAT3 pathway via miR-30d-5p. Life Sci. 2021, 277, 119501. [Google Scholar] [CrossRef]
  131. Xie, C.; Du, L.Y.; Guo, F.; Li, X.; Cheng, B. Exosomes derived from microRNA-101-3p-overexpressing human bone marrow mesenchymal stem cells suppress oral cancer cell proliferation, invasion, and migration. Mol. Cell. Biochem. 2019, 458, 11–26. [Google Scholar] [CrossRef]
  132. Xue, P.; Huang, S.; Han, X.; Zhang, C.; Yang, L.; Xiao, W.; Fu, J.; Li, H.; Zhou, Y. Exosomal miR-101-3p and miR-423-5p inhibit medulloblastoma tumorigenesis through targeting FOXP4 and EZH2. Cell Death Differ. 2022, 29, 82–95. [Google Scholar] [CrossRef]
  133. Tao, L.; Xu, C.; Shen, W.; Tan, J.; Li, L.; Fan, M.; Sun, D.; Lai, Y.; Cheng, H. HIPK3 inhibition by exosomal hsa-miR-101-3p is related to metabolic reprogramming in colorectal cancer. Front. Oncol. 2021, 11, 758336. [Google Scholar] [CrossRef]
  134. Sakha, S.; Muramatsu, T.; Ueda, K.; Inazawa, J. Exosomal microRNA miR-1246 induces cell motility and invasion through the regulation of DENND2D in oral squamous cell carcinoma. Sci. Rep. 2016, 6, 38750. [Google Scholar] [CrossRef] [Green Version]
  135. Zheng, M.; Hou, L.; Ma, Y.; Zhou, L.; Wang, F.; Cheng, B.; Wang, W.; Lu, B.; Liu, P.; Lu, W.; et al. Exosomal let-7d-3p and miR-30d-5p as diagnostic biomarkers for non-invasive screening of cervical cancer and its precursors. Mol Cancer 2019, 18, 76. [Google Scholar] [CrossRef] [Green Version]
  136. Lin, X.J.; Fang, J.H.; Yang, X.J.; Zhang, C.; Yuan, Y.; Zheng, L.; Zhuang, S.M. Hepatocellular carcinoma cell-secreted exosomal microRNA-210 promotes angiogenesis in vitro and in vivo. Mol. Therapy. Nucleic Acids 2018, 11, 243–252. [Google Scholar] [CrossRef] [Green Version]
  137. Wang, L.; He, J.; Hu, H.; Tu, L.; Sun, Z.; Liu, Y.; Luo, F. Lung CSC-derived exosomal miR-210-3p contributes to a pro-metastatic phenotype in lung cancer by targeting FGFRL1. J. Cell. Mol. Med. 2020, 24, 6324–6339. [Google Scholar] [CrossRef]
  138. Ge, L.; Zhou, F.; Nie, J.; Wang, X.; Zhao, Q. Hypoxic colorectal cancer-secreted exosomes deliver miR-210-3p to normoxic tumor cells to elicit a protumoral effect. Exp. Biol. Med. 2021, 246, 1895–1906. [Google Scholar] [CrossRef]
  139. Min, Q.H.; Wang, X.Z.; Zhang, J.; Chen, Q.G.; Li, S.Q.; Liu, X.Q.; Li, J.; Liu, J.; Yang, W.M.; Jiang, Y.H.; et al. Exosomes derived from imatinib-resistant chronic myeloid leukemia cells mediate a horizontal transfer of drug-resistant trait by delivering miR-365. Exp. Cell Res. 2018, 362, 386–393. [Google Scholar] [CrossRef]
  140. Mollaei, H.; Hoshyar, R.; Abedini, M.R.; Safaralizadeh, R. Crocin enhances cisplatin-induced chemosensitivity in human cervical cancer cell line. Int. J. Cancer Manag. 2019, 12, e94909. [Google Scholar]
  141. Wang, X.; Hao, R.; Wang, F.; Wang, F. ZFAS1 promotes cisplatin resistance via suppressing miR-421 expression in oral squamous cell carcinoma. Cancer Manag. Res. 2020, 12, 7251–7262. [Google Scholar] [CrossRef] [PubMed]
  142. Jingyue, S.; Xiao, W.; Juanmin, Z.; Wei, L.; Daoming, L.; Hong, X. TFAP2E methylation promotes 5fluorouracil resistance via exosomal miR106a5p and miR421 in gastric cancer MGC803 cells. Mol. Med. Rep. 2019, 20, 323–331. [Google Scholar] [CrossRef] [PubMed]
  143. Li, Y.Y.; Tao, Y.W.; Gao, S.; Li, P.; Zheng, J.M.; Zhang, S.E.; Liang, J.; Zhang, Y. Cancer-associated fibroblasts contribute to oral cancer cells proliferation and metastasis via exosome-mediated paracrine miR-34a-5p. EBioMedicine 2018, 36, 209–220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Zuo, L.; Tao, H.; Xu, H.; Li, C.; Qiao, G.; Guo, M.; Cao, S.; Liu, M.; Lin, X. Exosomes-coated miR-34a displays potent antitumor activity in pancreatic cancer both in vitro and in vivo. Drug Des. Dev. Ther. 2020, 14, 3495–3507. [Google Scholar] [CrossRef]
  145. Liang, Y.; Zhang, D.; Li, L.; Xin, T.; Zhao, Y.; Ma, R.; Du, J. Exosomal microRNA-144 from bone marrow-derived mesenchymal stem cells inhibits the progression of non-small cell lung cancer by targeting CCNE1 and CCNE2. Stem Cell Res. Ther. 2020, 11, 87. [Google Scholar] [CrossRef]
  146. Tian, X.; Liu, Y.; Wang, Z.; Wu, S. miR-144 delivered by nasopharyngeal carcinoma-derived EVs stimulates angiogenesis through the FBXW7/HIF-1alpha/VEGF-A axis. Mol. Ther. Nucleic Acids 2021, 24, 1000–1011. [Google Scholar] [CrossRef]
  147. Zhou, C.; Chen, Y.; He, X.; Zheng, Z.; Xue, D. Functional implication of exosomal miR-217 and miR-23b-3p in the progression of prostate cancer. OncoTargets Ther. 2020, 13, 11595–11606. [Google Scholar] [CrossRef]
  148. Chen, D.; Wu, X.; Xia, M.; Wu, F.; Ding, J.; Jiao, Y.; Zhan, Q.; An, F. Upregulated exosomic miR23b3p plays regulatory roles in the progression of pancreatic cancer. Oncol. Rep. 2017, 38, 2182–2188. [Google Scholar] [CrossRef] [Green Version]
  149. Hou, C.X.; Sun, N.N.; Han, W.; Meng, Y.; Wang, C.X.; Zhu, Q.H.; Tang, Y.T.; Ye, J.H. Exosomal microRNA-23b-3p promotes tumor angiogenesis and metastasis by targeting PTEN in salivary adenoid cystic carcinoma. Carcinogenesis 2022, 43, 682–692. [Google Scholar] [CrossRef]
  150. Sun, L.P.; Xu, K.; Cui, J.; Yuan, D.Y.; Zou, B.; Li, J.; Liu, J.L.; Li, K.Y.; Meng, Z.; Zhang, B. Cancer associated fibroblast derived exosomal miR3825p promotes the migration and invasion of oral squamous cell carcinoma. Oncol. Rep. 2019, 42, 1319–1328. [Google Scholar] [CrossRef]
  151. Jiang, Y.; Ji, X.; Liu, K.; Shi, Y.; Wang, C.; Li, Y.; Zhang, T.; He, Y.; Xiang, M.; Zhao, R. Exosomal miR-200c-3p negatively regulates the migraion and invasion of lipopolysaccharide (LPS)-stimulated colorectal cancer (CRC). BMC Mol. Cell. Biol. 2020, 21, 48. [Google Scholar] [CrossRef] [PubMed]
  152. Li, L.; Li, C.; Wang, S.; Wang, Z.; Jiang, J.; Wang, W.; Li, X.; Chen, J.; Liu, K.; Li, C.; et al. Exosomes derived from hypoxic oral squamous cell carcinoma cells deliver miR-21 to normoxic cells to elicit a prometastatic phenotype. Cancer Res. 2016, 76, 1770–1780. [Google Scholar] [CrossRef] [PubMed]
  153. Liu, T.; Chen, G.; Sun, D.; Lei, M.; Li, Y.; Zhou, C.; Li, X.; Xue, W.; Wang, H.; Liu, C.; et al. Exosomes containing miR-21 transfer the characteristic of cisplatin resistance by targeting PTEN and PDCD4 in oral squamous cell carcinoma. Acta Biochim. Biophys. Sin. 2017, 49, 808–816. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Shi, S.S.; Zhang, H.P.; Yang, C.Q.; Li, L.N.; Shen, Y.; Zhang, Y.Q. Exosomal miR-155-5p promotes proliferation and migration of gastric cancer cells by inhibiting TP53INP1 expression. Pathol. Res. Pract. 2020, 216, 152986. [Google Scholar] [CrossRef] [PubMed]
  155. Li, X.; Chen, Z.; Ni, Y.; Bian, C.; Huang, J.; Chen, L.; Xie, X.; Wang, J. Tumor-associated macrophages secret exosomal miR-155 and miR-196a-5p to promote metastasis of non-small-cell lung cancer. Transl. Lung Cancer Res. 2021, 10, 1338–1354. [Google Scholar] [CrossRef]
  156. Gu, W.; Gong, L.; Wu, X.; Yao, X. Hypoxic TAM-derived exosomal miR-155-5p promotes RCC progression through HuR-dependent IGF1R/AKT/PI3K pathway. Cell Death Discov. 2021, 7, 147. [Google Scholar] [CrossRef]
  157. Wang, D.; Wang, X.; Song, Y.; Si, M.; Sun, Y.; Liu, X.; Cui, S.; Qu, X.; Yu, X. Exosomal miR-146a-5p and miR-155-5p promote CXCL12/CXCR7-induced metastasis of colorectal cancer by crosstalk with cancer-associated fibroblasts. Cell Death Dis. 2022, 13, 380. [Google Scholar] [CrossRef]
  158. Kirave, P.; Gondaliya, P.; Kulkarni, B.; Rawal, R.; Garg, R.; Jain, A.; Kalia, K. Exosome mediated miR-155 delivery confers cisplatin chemoresistance in oral cancer cells via epithelial-mesenchymal transition. Oncotarget 2020, 11, 1157–1171. [Google Scholar] [CrossRef] [Green Version]
  159. Xing, Y.; Ruan, G.; Ni, H.; Qin, H.; Chen, S.; Gu, X.; Shang, J.; Zhou, Y.; Tao, X.; Zheng, L. Tumor immune microenvironment and its related miRNAs in tumor progression. Front. Immunol. 2021, 12, 624725. [Google Scholar] [CrossRef]
  160. Yuan, Y.; Wang, Z.; Chen, M.; Jing, Y.; Shu, W.; Xie, Z.; Li, Z.; Xu, J.; He, F.; Jiao, P.; et al. Macrophage-derived exosomal miR-31-5p promotes oral squamous cell carcinoma tumourigenesis through the large tumor suppressor 2-mediated Hippo signalling pathway. J. Biomed. Nanotechnol. 2021, 17, 822–837. [Google Scholar] [CrossRef]
  161. Yu, F.; Liang, M.; Huang, Y.; Wu, W.; Zheng, B.; Chen, C. Hypoxic tumor-derived exosomal miR-31-5p promotes lung adenocarcinoma metastasis by negatively regulating SATB2-reversed EMT and activating MEK/ERK signaling. J. Exp. Clin. Cancer Res. CR 2021, 40, 179. [Google Scholar] [CrossRef] [PubMed]
  162. He, J.; He, J.; Min, L.; He, Y.; Guan, H.; Wang, J.; Peng, X. Extracellular vesicles transmitted miR-31-5p promotes sorafenib resistance by targeting MLH1 in renal cell carcinoma. Int. J. Cancer. J. Int. Cancer 2020, 146, 1052–1063. [Google Scholar] [CrossRef] [PubMed]
  163. Hsu, H.H.; Kuo, W.W.; Shih, H.N.; Cheng, S.F.; Yang, C.K.; Chen, M.C.; Tu, C.C.; Viswanadha, V.P.; Liao, P.H.; Huang, C.Y. FOXC1 regulation of miR-31-5p confers oxaliplatin resistance by targeting LATS2 in colorectal cancer. Cancers 2019, 11, 1576. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Wang, X.; Qin, X.; Yan, M.; Shi, J.; Xu, Q.; Li, Z.; Yang, W.; Zhang, J.; Chen, W. Loss of exosomal miR-3188 in cancer-associated fibroblasts contributes to HNC progression. J. Exp. Clin. Cancer Res. CR 2019, 38, 151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Zhao, M.; Luo, R.; Liu, Y.; Gao, L.; Fu, Z.; Fu, Q.; Luo, X.; Chen, Y.; Deng, X.; Liang, Z.; et al. miR-3188 regulates nasopharyngeal carcinoma proliferation and chemosensitivity through a FOXO1-modulated positive feedback loop with mTOR-p-PI3K/AKT-c-JUN. Nat. Commun. 2016, 7, 11309. [Google Scholar] [CrossRef] [Green Version]
  166. Wang, C.; Liu, E.; Li, W.; Cui, J.; Li, T. MiR-3188 inhibits non-small cell lung cancer cell proliferation through FOXO1-mediated mTOR-p-PI3K/AKT-c-JUN signaling pathway. Front. Pharmacol. 2018, 9, 1362. [Google Scholar] [CrossRef] [Green Version]
  167. He, L.; Ping, F.; Fan, Z.; Zhang, C.; Deng, M.; Cheng, B.; Xia, J. Salivary exosomal miR-24-3p serves as a potential detective biomarker for oral squamous cell carcinoma screening. Biomed. Pharmacother. 2020, 121, 109553. [Google Scholar] [CrossRef]
  168. Zhang, H.W.; Shi, Y.; Liu, J.B.; Wang, H.M.; Wang, P.Y.; Wu, Z.J.; Li, L.; Gu, L.P.; Cao, P.S.; Wang, G.R.; et al. Cancer-associated fibroblast-derived exosomal microRNA-24-3p enhances colon cancer cell resistance to MTX by down-regulating CDX2/HEPH axis. J. Cell. Mol. Med. 2021, 25, 3699–3713. [Google Scholar] [CrossRef]
  169. Kulkarni, B.; Gondaliya, P.; Kirave, P.; Rawal, R.; Jain, A.; Garg, R.; Kalia, K. Exosome-mediated delivery of miR-30a sensitize cisplatin-resistant variant of oral squamous carcinoma cells via modulating Beclin1 and Bcl2. Oncotarget 2020, 11, 1832–1845. [Google Scholar] [CrossRef]
  170. Tao, K.; Liu, J.; Liang, J.; Xu, X.; Xu, L.; Mao, W. Vascular endothelial cell-derived exosomal miR-30a-5p inhibits lung adenocarcinoma malignant progression by targeting CCNE2. Carcinogenesis 2021, 42, 1056–1067. [Google Scholar] [CrossRef]
  171. Du, Q.; Ye, X.; Lu, S.R.; Li, H.; Liu, H.Y.; Zhai, Q.; Yu, B. Exosomal miR-30a and miR-222 derived from colon cancer mesenchymal stem cells promote the tumorigenicity of colon cancer through targeting MIA3. J. Gastrointest. Oncol. 2021, 12, 52–68. [Google Scholar] [CrossRef] [PubMed]
  172. Kong, X.; Zhang, J.; Li, J.; Shao, J.; Fang, L. MiR-130a-3p inhibits migration and invasion by regulating RAB5B in human breast cancer stem cell-like cells. Biochem. Biophys. Res. Commun. 2018, 501, 486–493. [Google Scholar] [CrossRef] [PubMed]
  173. Yin, G.; Kong, W.; Zheng, S.; Shan, Y.; Zhang, J.; Ying, R.; Wu, H. Exosomal miR-130a-3p promotes the progression of differentiated thyroid cancer by targeting insulin-like growth factor 1. Oncol. Lett. 2021, 21, 283. [Google Scholar] [CrossRef]
  174. Dickman, C.T.; Lawson, J.; Jabalee, J.; MacLellan, S.A.; LePard, N.E.; Bennewith, K.L.; Garnis, C. Selective extracellular vesicle exclusion of miR-142-3p by oral cancer cells promotes both internal and extracellular malignant phenotypes. Oncotarget 2017, 8, 15252–15266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Plousiou, M.; De Vita, A.; Miserocchi, G.; Bandini, E.; Vannini, I.; Melloni, M.; Masalu, N.; Fabbri, F.; Serra, P. Growth inhibition of retinoblastoma cell line by exosome-mediated transfer of miR-142-3p. Cancer Manag. Res. 2022, 14, 2119–2131. [Google Scholar] [CrossRef] [PubMed]
  176. Hu, Z.; Zhang, H.; Liu, W.; Yin, Y.; Jiang, J.; Yan, C.; Wang, Y.; Li, L. Mechanism of HBV-positive liver cancer cell exosomal miR-142-3p by inducing ferroptosis of M1 macrophages to promote liver cancer progression. Transl. Cancer Res. 2022, 11, 1173–1187. [Google Scholar] [CrossRef]
  177. Lou, C.; Shi, J.; Xu, Q. Exosomal miR-626 promotes the malignant behavior of oral cancer cells by targeting NFIB. Mol. Biol. Rep. 2022, 49, 4829–4840. [Google Scholar] [CrossRef]
  178. Singh, R.; Pochampally, R.; Watabe, K.; Lu, Z.; Mo, Y.Y. Exosome-mediated transfer of miR-10b promotes cell invasion in breast cancer. Mol. Cancer 2014, 13, 256. [Google Scholar] [CrossRef] [Green Version]
  179. Yan, T.; Wang, X.; Wei, G.; Li, H.; Hao, L.; Liu, Y.; Yu, X.; Zhu, W.; Liu, P.; Zhu, Y.; et al. Exosomal miR-10b-5p mediates cell communication of gastric cancer cells and fibroblasts and facilitates cell proliferation. J. Cancer 2021, 12, 2140–2150. [Google Scholar] [CrossRef]
  180. Higaki, M.; Shintani, T.; Hamada, A.; Rosli, S.N.Z.; Okamoto, T. Eldecalcitol (ED-71)-induced exosomal miR-6887-5p suppresses squamous cell carcinoma cell growth by targeting heparin-binding protein 17/fibroblast growth factor-binding protein-1 (HBp17/FGFBP-1). Vitr. Cell. Dev. Biol. Anim. 2020, 56, 222–233. [Google Scholar] [CrossRef]
  181. Chen, T.; Liu, Y.; Chen, J.; Zheng, H.; Chen, Q.; Zhao, J. Exosomal miR-3180-3p inhibits proliferation and metastasis of non-small cell lung cancer by downregulating FOXP4. Thorac. Cancer 2021, 12, 372–381. [Google Scholar] [CrossRef] [PubMed]
Figure 1. General structure and biogenesis of exosomes. The basic steps of exosome biogenesis, consisting of two main stages, are provided: exosome assembly and secretion. Exosomes generally contain nucleic acids, membrane and soluble proteins, and lipids. Different cells or treatments may have different compositions of exosomes. MVB, multivesicular body.
Figure 1. General structure and biogenesis of exosomes. The basic steps of exosome biogenesis, consisting of two main stages, are provided: exosome assembly and secretion. Exosomes generally contain nucleic acids, membrane and soluble proteins, and lipids. Different cells or treatments may have different compositions of exosomes. MVB, multivesicular body.
Cancers 15 00318 g001
Figure 2. Connections between different sections of this review. Connections between natural products and exosomal processing are examined in Section 2 and Section 4. The connection between exosomal processing and exosomal miRNAs is examined in Section 3. Finally, connections between cancer cell functions, exosomal miRNAs, and natural products are examined in Section 5.
Figure 2. Connections between different sections of this review. Connections between natural products and exosomal processing are examined in Section 2 and Section 4. The connection between exosomal processing and exosomal miRNAs is examined in Section 3. Finally, connections between cancer cell functions, exosomal miRNAs, and natural products are examined in Section 5.
Cancers 15 00318 g002
Figure 3. Overview of natural products modulating exosomal miRNAs through exosomal processing genes and exosomal assembly and secretion. The targets of exosomal processing genes with respect to natural-product-modulated miRNAs (Table 3) were predicted using the miRDB database. Some natural products (Table 3) are not shown here because the exosomal processing gene targets of their modulated miRNAs could not be identified in miRDB.
Figure 3. Overview of natural products modulating exosomal miRNAs through exosomal processing genes and exosomal assembly and secretion. The targets of exosomal processing genes with respect to natural-product-modulated miRNAs (Table 3) were predicted using the miRDB database. Some natural products (Table 3) are not shown here because the exosomal processing gene targets of their modulated miRNAs could not be identified in miRDB.
Cancers 15 00318 g003
Table 3. Connecting natural products to exosomal miRNA-regulated cell functions.
Table 3. Connecting natural products to exosomal miRNA-regulated cell functions.
Natural ProductsmiRNAsCell FunctionsCancerReferences
Ursolic acid↓miR-21-5papoptosisglioblastoma[81]
↑miR-200c-3papoptosis, anti-invasioncolon[82]
Resveratrol↓miR-21-5papoptosisbladder[83]
↓miR-31-5panticolitis(T cells)[84]
↑miR-424-5pantiproliferationbreast[85]
Berberine↓miR-21-5pantiproliferationmyeloma[86]
↑miR-144-3papoptosis, autophagylung[87]
↑miR-101-3pantiproliferation, antimigrationendometrial[88]
Butylcycloheptyl prodiginine↓miR-21-5pantiproliferationcolon[89]
Honokiol↓miR-21-5papoptosisosteosarcoma[90]
↑miR-34a-5panti-EMTbreast[91]
Sophocarpine↓miR-21-5pantiproliferation, anti-EMThead/neck[92]
Tricin↓miR-21-5pchemosensitizationprostate[93]
Dihydromyricetin↓miR-21-5pantiproliferation, antimigrationcholangiocarcinoma[94]
Curcumin↓miR-21-5pantiproliferation, antimigrationliver[95]
↑miR-200c-3panti-EMTcolon[96]
↑miR-142-3p20S proteasome suppressionbreast[97]
↓miR-1246antiproliferationbladder[98]
Curcumol↓miR-21-5pantiproliferationcolon[99]
PRP1↓miR-21-5papoptosisliver[100]
Sinomenine↓miR-21-5pantimigrationlung[101]
Psoralen↑miR-196a-5papoptosisgastric[102]
Pinolenic acid↑miR-3188anti-inflammation(rheumatoid arthritis)[103]
Pachymic acid↓miR-24-3panti-heart failure(left ventricle)[104]
Genistein↓miR-155-5pantiproliferation (cardiac)[105]
(−)-Epigallocatechin gallate↓miR-155-5pchemosensitizationcolon[106]
↑miR-34a-5pradiosensitizationliver[107]
Enoxolone, Magnolol, ↑miR-200c-3panti-invasionbreast[108]
Palmatine chloride
(−)-Sativan↑miR-200c-3papoptosis, antimigrationbreast[109]
Isoliquiritigenin↑miR-200c-3pantimigrationbreast[110]
Thymoquinone↑miR-30a-5panti-liver fibrosis(liver)[111]
Nicotine↑miR-30a-5pG1 arrest(periodontal ligament)[112]
Norcantharidin↑miR-30a-5pantiproliferation, antimigrationgiant cell tumor of bone[62]
1΄S-1΄-acetoxychavicol acetate↓miR-210-3papoptosiscervical[113]
Crocin↓miR-365a-3papoptosiscervical[114]
↓miR-34a-5papoptosispapillary thyroid[115]
Isoliquiritigenin↓miR-421apoptosis, DNA damageoral[116]
Anisomycin↑miR-421anti-angiogenesisovarian[117]
Asparanin A↓miR-421antimigrationendometrial[118]
Rhamnetin, Cirsiliol↑miR-34a-5pradiosensitization, anti-EMTlung[119]
Dihydroartemisinin↑miR-34a-5papoptosis, antimigrationprostate [120]
Isovitexin↑miR-34a-5papoptosisosteosarcoma[121]
Emodin↑miR-34a-5pantiproliferationliver[122]
Kaempferol↑miR-130a-3pcytokine reduction(chondrocyte)[123]
Chicoric acid↓miR-130a-3panti-inflammationlung[124]
Mitomycin C↑miR-31-5pchemosensitizationbladder[125]
Licochalcone A↑miR-144-3pER stress, apoptosislung[126]
10-Hydroxycamptothecin↑miR-23b-3papoptosis(fibroblast)[127]
Astaxanthin↓miR-382-5panti-liver fibrosis(liver)[128]
Polydatin↑miR-382-5papoptosiscolon[129]
Piperlongumine↓miR-30d-5pantiproliferationosteosarcoma[130]
, enhance or activate; , inhibit or inactivate. Non-cancer studies are shown in parentheses.
Table 4. Connecting natural-product-modulated exosomal miRNAs to TIME.
Table 4. Connecting natural-product-modulated exosomal miRNAs to TIME.
miRNAsmiRNAs Status in TIMEmiRNA Effects of Natural Products *
miR-21-5p↑TAM [159] ↑MDSC [159]   miR-21-5p (Ursolic acid [81], Resveratrol [83], Berberine [86], Butylcycloheptyl prodiginine [89], Honokiol [90], Sophocarpine [92], Tricin [93], Dihydromyricetin [94], Curcumin [95], Curcumol [99], PRP1 [100], Sinomenine [101])
miR-200c-3p↑MDSC [159]   miR-200c-3p (Urolic acid [82], Curcumin [96], Enoxolone, Magnolol, Palmatine chloride [108], (−)-Sativan [109], Isoliquiritigenin [110])
miR-155-5p↑TAM,↑MDSC,↑NK [159]   miR-155-5p (Genistein [105], (−)-Epigallocatechin gallate [106])
miR-30a-5p↑TAM,↑MDSC [159]   miR-30a-5p (Thymoquinone [111], Nicotine [112], Norcantharidin [62])
miR-34a-5p↓TAM [159]   miR-34a-5p (Honokiol [91], (−)-Epigallocatechin gallate [107], Rhamnetin, Cirsiliol [119], Dihydroartemisinin [120], Isovitexin [121], Emodin [122])
  miR-34a-5p (Crocin [115])
miR-130a-3p↑NK [159]   miR-130a-3p (Kaempferol [123])
  miR-130a-3p (Chicoric acid [124])
miR-101-3p↑TAM [159]   miR-101-3p (Berberine [88])
miR-142-3p↑TAM [159]   miR-142-3p (Curcumin [97])
miR-24-3p↑NK [159]   miR-24-3p (Pachymic acid [104])
* These natural products and their modulated exosomal miRNAs were collected from Table 3. The literature [159] did not provide information on natural products. , enhance; , inhibit. Tumor immune microenvironment (TIME); tumor-associated macrophages (TAM); natural killer (NK); myeloid-derived suppressor cells (MDSC).
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Chuang, Y.-T.; Tang, J.-Y.; Shiau, J.-P.; Yen, C.-Y.; Chang, F.-R.; Yang, K.-H.; Hou, M.-F.; Farooqi, A.A.; Chang, H.-W. Modulating Effects of Cancer-Derived Exosomal miRNAs and Exosomal Processing by Natural Products. Cancers 2023, 15, 318. https://doi.org/10.3390/cancers15010318

AMA Style

Chuang Y-T, Tang J-Y, Shiau J-P, Yen C-Y, Chang F-R, Yang K-H, Hou M-F, Farooqi AA, Chang H-W. Modulating Effects of Cancer-Derived Exosomal miRNAs and Exosomal Processing by Natural Products. Cancers. 2023; 15(1):318. https://doi.org/10.3390/cancers15010318

Chicago/Turabian Style

Chuang, Ya-Ting, Jen-Yang Tang, Jun-Ping Shiau, Ching-Yu Yen, Fang-Rong Chang, Kun-Han Yang, Ming-Feng Hou, Ammad Ahmad Farooqi, and Hsueh-Wei Chang. 2023. "Modulating Effects of Cancer-Derived Exosomal miRNAs and Exosomal Processing by Natural Products" Cancers 15, no. 1: 318. https://doi.org/10.3390/cancers15010318

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop