Next Article in Journal
Flavonoid Metabolites in Serum and Urine after the Ingestion of Selected Tropical Fruits
Next Article in Special Issue
Different Dimensions of the Home Food Environment May Be Associated with the Body Mass Index of Older Adults: A Cross-Sectional Survey Conducted in Beijing, China
Previous Article in Journal
Intermittent Fasting Reduces Neuroinflammation and Cognitive Impairment in High-Fat Diet-Fed Mice by Downregulating Lipocalin-2 and Galectin-3
Previous Article in Special Issue
BMI and the Food Retail Environment in Melbourne, Australia: Associations and Temporal Trends
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Dietary Epigenetic Modulators: Unravelling the Still-Controversial Benefits of miRNAs in Nutrition and Disease

by
Elisa Martino
1,
Nunzia D’Onofrio
1,*,
Anna Balestrieri
2,
Antonino Colloca
1,
Camilla Anastasio
1,
Celestino Sardu
3,
Raffaele Marfella
3,
Giuseppe Campanile
4,† and
Maria Luisa Balestrieri
1,†
1
Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
2
Food Safety Department, Istituto Zooprofilattico Sperimentale del Mezzogiorno, 80055 Portici, Italy
3
Department of Advanced Clinical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
4
Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Nutrients 2024, 16(1), 160; https://doi.org/10.3390/nu16010160
Submission received: 7 December 2023 / Revised: 29 December 2023 / Accepted: 30 December 2023 / Published: 3 January 2024
(This article belongs to the Special Issue Food Environment and Its Effects on Human Nutrition and Health)

Abstract

:
In the context of nutrient-driven epigenetic alterations, food-derived miRNAs can be absorbed into the circulatory system and organs of recipients, especially humans, and potentially contribute to modulating health and diseases. Evidence suggests that food uptake, by carrying exogenous miRNAs (xenomiRNAs), regulates the individual miRNA profile, modifying the redox homeostasis and inflammatory conditions underlying pathological processes, such as type 2 diabetes mellitus, insulin resistance, metabolic syndrome, and cancer. The capacity of diet to control miRNA levels and the comprehension of the unique characteristics of dietary miRNAs in terms of gene expression regulation show important perspectives as a strategy to control disease susceptibility via epigenetic modifications and refine the clinical outcomes. However, the absorption, stability, availability, and epigenetic roles of dietary miRNAs are intriguing and currently the subject of intense debate; additionally, there is restricted knowledge of their physiological and potential side effects. Within this framework, we provided up-to-date and comprehensive knowledge on dietary miRNAs’ potential, discussing the latest advances and controversial issues related to the role of miRNAs in human health and disease as modulators of chronic syndromes.

1. Introduction

Among lifestyle factors, diet displays a strong impact on human health. Over decades, unhealthy dietary habits have been responsible for the spread of many severe chronic degenerative diseases such as obesity, type 2 diabetes mellitus (T2DM), cardiovascular diseases, and cancer [1]. Food intake may modify gene expression as well as disease susceptibility through the regulation of several epigenetic modulators [1]. MicroRNAs (miRNAs) play critical roles in gene regulation and biological processes [2,3]. They are transcribed by RNA polymerase II, leading to the generation of a hairpin structure called primary microRNA or pri-miRNA, which is further processed by the RNA endonucleases located in the nucleus (Drosha) and in the cytoplasm (Dicer) to generate a duplex of 22–25 nucleotides [2]. This latter is then divided into two mature sequences associated with the Argonaute protein (AGO), resulting in the formation of the RNA-induced silencing complex (RISC), which can ultimately act as a translation repressor on target genes [3]. Gene expression regulation at intracellular and extracellular levels is improved by mature transcript encapsulation into extracellular vesicles, such as exosomes, secreted by multiple cell types as mediators of cell-to-cell communication [4]. Because of their biocompatibility and biostability, extracellular vesicles are considered regulatory cargos in a wide range of intercellular communication and crosstalk systems [5,6]. MiRNAs have been described in body fluids as blood, serum, plasma, urine, saliva, and breast milk, and changes in their circulating levels are related to a plethora of different chronic syndromes, including obesity, cardiovascular and neurodegenerative diseases, T2DM, and cancer [7,8]. Preserving the human miRNA profile could contribute to the prevention of diseases and the maintenance of good health. MiRNAs have been widely reported in plants, animals, and humans [9]. Plant-derived miRNAs bind to recipient target transcripts with quite perfect complementarity, acting as small interfering RNAs (siRNAs), while animal miRNAs interact with the host mRNA targets with imperfect complementarity, thus inducing their translational repression [10,11]. Given this imperfect complementarity, a single exogenous miRNA (xenomiRNA) is able to recognize multiple target sites and modulate different target genes in the host [10,11]. After ingestion, xenomiRNAs could modulate the gene expression by a cross-kingdom pattern or horizontal transfer of genetic information to the host [10,11]. Bioactive dietary compounds, by affecting directly and indirectly gene activity, have been related to epigenetic changes, such as DNA methylation, histone acetylation, and the modulation of miRNA expression in both physiological and pathological conditions [12,13]. However, the clinical relevance of food-derived xenomiRNAs in human diseases is still undefined. Several studies have demonstrated that the intricate interplay between miRNAs and nutrients could regulate health and chronic diseases, hence pointing to food modification as a pivotal tool in different diseases [14,15]. However, further studies to reveal the precise mode of action of dietary active compounds and how nutrients and bioactive molecules affect miRNA expression are still required. This review aims to provide an update on the role of dietary miRNAs in health and diseases by underlining the benefits, obstacles, and controversies of the relationship between food-derived miRNAs and the pathogenesis of chronic diseases.

2. Dietary XenomiRNAs in Health and Disease

XenomiRNAs represent a family of exogenous miRNAs characterized by several dietary sources, animals and vegetables, and are able to integrate into the total miRNA profile of a recipient [16]. Once in the host, these small molecules can be absorbed by the gastrointestinal tract, packaged into vesicles, released into the bloodstream, and delivered to multiple cells and tissues, thus promoting healthy state or affecting the development of chronic diseases, including cancer [17,18,19]. In the following sections, the role of xenomiRNAs from both animals and vegetables as regulators of several chronic conditions will be extensively discussed (Figure 1).

2.1. XenomiRNAs from Animal Sources

The following subsection is dedicated to the most studied and characterized xenomiRNAs derived from animal sources and their involvement in chronic syndromes and cancer.

2.1.1. Eggs

Analysis of RNA sequencing revealed the content of several miRNAs, such as gga-mir-2188, gga-mir-30c-5p and gga-mir-92-3p, in the edible parts of chicken eggs, suggesting these noncoding RNAs as interesting tools to take into account for the improvement of egg nutritional value [20]. More recently, Fratantonio et al. described the availability of miRNA-related exosomal vesicles from chicken eggs in mice and humans [21]. Accumulated in the brain, intestine, and lung, miRNA-exosomes regulated spatial learning and memory function in C57BL/6J mice, while egg-derived miRNA levels increased in human peripheral blood following exosomal oral administration (Figure 1) [21].

2.1.2. Meat

The effects of the chronic administration of cooked pork-derived exosomal vesicles were evaluated in mouse model [22]. The upregulated miR-1, miR-133a-3p, miR-206 and miR-99a plasma levels resulted in glucose and insulin metabolism impairment, as well as in lipid droplet accrual in mice liver, supporting the role of pork-derived miRNAs in the development of metabolic disorders (Figure 1) [22]. The bta-miR-154c has been recently characterized by comestible parts of beef. This xenomiRNA is able to contrast the human digestion processes being absorbed from Caco-2, SW480, and SW620 colorectal cancer cells [23]. However, the comprehension of the specific role of miR-154c in colorectal carcinoma requires further investigation [23].

2.1.3. Milk

The classification of milk as an “epi-nutrient”, rich in bioactive compounds and functional molecules, as well as its involvement in counteracting chronic syndromes and cancer, have been recently investigated [24,25,26,27,28]. A total of 678 miRNAs were identified in bovine milk-derived exosomal vesicles [29,30]. These miRNAs are involved in a wide range of cell metabolic pathways, such as miR-181 and miR-155, related to the normal function and differentiation of T and B cells, miR-let-7c, miR-17, miR-92, miR-223 implied in the regulation of immunity and inflammatory cells, and miR-30a, a regulator of autophagy in post-acute myocardial infarction (Figure 2) [31,32,33].
The most abundant miRNA in milk exosomes is miR-148a, able to modulate oral cavity homeostasis as well as to inhibit 5′ AMP-activated protein kinase (AMPK) and phosphatase and tensin homolog (PTEN), both suppressors of mammalian target of rapamycin complex 1 (mTORC1), a pivotal regulator of multiple cell metabolic pathways [31,32,33]. Moreover, milk-derived miR-148a impairs insulin secretion with diabetogenic action through its ability to promote pancreatic β-cell de-differentiation via mTORC1-high/AMPK-low phenotype [33]. It has been reported that miR-148a and miR-21 alter the α-synuclein homeostasis, causing its overexpression and aggregation, with toxic effects on dopaminergic neurons and pancreatic β-cells, thus exerting a possible role in the pathogenesis of Parkinson disease and T2DM [34]. MiRNAs belonging to the miR-148 family affect the immune response, suppressing calcium/calmodulin-dependent protein kinase IIα (CaMKIIα) and the subsequent toll-like receptor (TLR)-mediated expression of major histocompatibility complex II (MHC II) in dendritic cells [31]. Among miRNAs derived from milk exosomes, miR-125a is involved in the modulation of immune response to bacterial and viral aggressions, while the human homolog miR-718, still involved in immune response regulating p53, also regulates vascular endothelial growth factor (VEGF) and insulin growth factor (IGF) pathways [35], and miR-146 exerts a regulatory function in TLR signaling and in the resolution of bacterial infections [36]. As bovine milk, human breast milk-derived miRNAs play a crucial role in modulating development and differentiation of immune system cells and counteracting the onset of metabolic disorders [37,38]. In the first six months of lactation, miR-181a and b, miR-155, miR-125b, and the cluster miR-17-92 actively regulate the T- and B-cell maturation and the tumor necrosis factor α (TNF-α) activation, modulating the immune response of the baby [38], while miR-22-3p counteracts the development of insulin resistance and the onset of T2DM, attenuating the Wnt pathway [37]. Milk-derived miRNAs are also involved in different pivotal metabolic pathways. The presence in milk exosomes of miR-181a-5p has been related to anti-atherogenic effects and reduced vascular inflammation, due to its ability to downregulate nuclear factor kappaB (NF-κB) levels [39]. Another miRNA characterized from milk exosomal vesicles is miR-29, capable of targeting IL-23, a cytokine involved in intestinal damage. To this aim, treatment with milk vesicles containing miR-29 stimulated intestinal stem cell proliferation and gut recovery under several pathological conditions [40], while incubation with miR-31-5p from milk-derived exosomes improved in vitro endothelial function and promoted angiogenesis and diabetic wound healing in vivo [41]. Recent studies reported that oral administration of exosomal vesicles prevented colon shortening, intestinal epithelium disruption, infiltration of inflammatory cells, and tissue fibrosis in a mouse ulcerative colitis model via inhibition of the TLR4-NF-κB signaling pathway and nucleotide-binding oligomerization domain, the NLR family pyrin domain containing 3 (NLRP3) inflammasome activation [30]. Transfection with the milk-derived miR-22 promoted cell proliferation by inhibition of CCAAT/enhancer-binding protein δ (C/EBPδ) expression and promotion of intestinal development in human intestinal epithelial cells [42]. On the other hand, the milk-exosomal miR-148a and miR-30b have been correlated to adipogenic effects, supporting a correlation between milk consumption and obesity incidence [43]. In vitro and in vivo studies reported that milk exosome-derived miRNAs also exert oncogenic or oncosuppressor properties (Figure 2). An association has been reported between cow milk consumption and large B-cell lymphoma development, sustained by miR-148a-3p and miR-155-5p/miR-29b-5p increase via let-7-5p/miR-125b-5p [44]. As oncomir, miR-21 promotes cell growth and anabolism, encouraging cell proliferation and cancer development by activating mTORC1 [45]. MiR-21 and miR-155 have been related to the most advanced stages of breast cancer progression, being involved in the development of tamoxifen resistance, metastasis formation, and worst prognosis [46]. Commercial milk consumption has been correlated to an increased risk of estrogen receptor-positive breast cancer development, due to the content of multiple oncogenic factors, as miR-148a-3p and miR-21-5p [47]. Milk-derived miRNAs have been associated with prostate cancer tumorigenesis promotion. In vitro studies have shown that milk-derived miR-148 increased prostate cancer proliferation by inhibiting cyclin-dependent kinase inhibitor 1B (CDKN1B) and promoted DNA methyltransferase 1 (DNMT1)-dependent epithelial-mesenchymal transition (EMT) [45]. Another significant milk-derived miRNA, miR-125b, increases the development of prostate xenograft cancer targeting proapoptotic genes, as p53, p53-upregulated modulator of apoptosis (PUMA), and BCL2 Antagonist/Killer 1 (BAK1) [48], and regulates several tumorigenic pathways, including NF-κB, p53, phosphatidyl inositol 3-kinase (PI3K)/protein kinase B (AKT)/mTORC1, erb-b2 receptor tyrosine kinase 2 (ERBB2), and Wnt [45]. Epidemiological studies highlighted the association between milk consumption and reduced risk of colorectal cancer [49]. The expression of miR-148a is downregulated in colorectal cancer (CRC), where it exerts a tumor-suppressor activity interfering in NF-κB and signal transducer and activator of transcription 3 (STAT3) pathways and modulating cancer related immune response via inhibition of the programmed death ligand-1 (PD-L1) levels [45]. The in vitro antineoplastic effect of miR-148a-3p overexpression also occurs through mitochondrial impairment, lipid peroxidation, and ferroptosis sustained by the Acyl CoA synthetase long-chain family member 4 (ACSL4)/transferrin receptor (TFRC)/Ferritin axis and direct solute carrier family seven-member 11 (SLC7A11) downregulation in the CRC model [50]. Another buffalo milk-derived miRNA, miR-27b, exerted antineoplastic effects on HCT116 and HT-29 CRC cells by inducing mitochondrial oxidative stress, lysosome accumulation, and apoptotic cell death mediated by endoplasmic reticulum (ER) stress [51].

2.2. XenomiRNAs from Vegetable Sources

Several miRNAs characterized from different vegetables and involved in chronic diseases, including cancer, have been evaluated (Figure 1). It has been described the ability of miR-156a, contained in different vegetables, such as cabbage, spinach, and lettuce, to target junctional adhesion molecule A (JAM-A), thus suppressing the development of atherosclerosis in human aortic endothelial cells through the inhibition of monocyte adhesion, occurring under inflammatory stress [52]. Similarly, the miR-167e-5p, characterized by Moringa oleifera, exerted a time- and dose-dependent anticancer action in Caco-2 cells acting on the β-catenin pathway [53]. The miR-159, particularly abundant in broccoli, is able to suppress breast tumor development, both in vitro and in vivo models [54,55]. In silico prediction identified two miRNA sequences, bra-miR156g-5p and Myseq-330, in broccoletti Brassica rapa sylvestris, as targeting apoptosis-related human genes, although further in vitro studies on pancreatic cancer cells did not support a miR-based modulating role in cancer growth [56].

2.2.1. Rice

An extensive study on miRNAs derived from Oryza sativa rice revealed their binding affinity for multiple human genes involved in cardiovascular and neurological diseases and cancer [57]. The Oryza sativa-derived osa-miR-172d-5p exerted in vitro beneficial effects in human lung fibroblasts, suppressing transforming growth factor (TGF)-β activated kinase 1 (MAP3K7) binding protein 1 (Tab1) and TGF-β-induced fibrotic gene expression in a bleomycin-induced lung fibrosis model [58]. Analyses on mice and human serum after rice consumption revealed miR-168 circulating levels associated with reduced clearance of plasmatic low-density lipoprotein (LDL) [59,60], while the rice aleurone-derived hvu-miR-168-3p increased the glucose transporter 1 (GLUT1) expression and reduced blood glucose levels by specifically inhibiting the electron transport chain complex I in both in vitro and in vivo models [61].

2.2.2. Ginger

The action of different xenomiRNAs from vegetable sources in the modulation of oncological pathways has been reported. A report on stomach tissues of patients with different gastric conditions, such as gastritis, metaplasia, and cancer, unveiled higher plant-derived miR-168 levels in tissues of patients affected by intestinal metaplasia [62]. In particular, the ginger-derived miR-1078, by regulating leptin, is related to lipopolysaccharide (LPS)-induced interleukin (IL)-6 expression, whereas miR-167a acts as a gut microbiota modulator targeting Lactobacillus rhamnosus GG SpaC pilus adhesin [63,64]. Among the different miRNAs contained in the ginger-derived exosomes-like nanoparticles (GELNs), mdo-miR-7267-3p increased the production of IL-22, thus inducing mice colitis [65], while the plant-derived miR-34a, miR-143, and miR-145 act as tumor suppressors in the mouse CRC model [66].

2.2.3. Soybean

In vitro studies showed that the soybean-derived miR-159a exerted an antitumor activity, suppressing Caco-2 proliferation and triggering apoptotic cell death [67], while gma-miR-4995 targets the transcripts of two long noncoding RNAs, metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) and nuclear paraspeckle assembly transcript 1 (NEAT1), strongly expressed during the metastasis formation of several cancer types. The inhibitory action of gma-miR-4995 on MALAT1 and NEAT1 results in the attenuation of cell proliferation and apoptosis induction in CRC cell lines [68]. The soybean exosome-like nanoparticles (ELNs)-derived miR-5781 is able to modulate the inflammatory response by targeting IL-17A [62], while miR-159a might prevent hepatic fibrosis suppressing glycogen synthase kinase-3β (GSK-3β)-mediated NF-κB and TGF-β1 pathways, thus impairing TGF-β1- and platelet-derived growth factor (PDGF)-related hepatic stellate cell activation [69].

2.2.4. Fruits

The ELNs from 11 different edible fruits (blueberry, coconut, grapefruit, Hami melon, kiwifruit, orange, pea, pear, and tomato) contain several miRNAs capable of specifically targeting genes encoding inflammatory mediators [63]. The apple-derived mdm-miR-7121d-h can downregulate the intestinal solute carrier organic anion transporter family member 2B1 (SLCO2B1), thus influencing enteric macromolecules absorption in the human intestine [70].

3. Food-Derived Nutrients as miRNA Regulators in Chronic Diseases

Nutrients affect the miRNA profile through the direct or indirect modulation of gene expression. Multiple macro- and micronutrients, such as fatty acids, carbohydrates, vitamins and phytochemicals, are able to regulate miRNA levels [31], thus rendering food epigenetics more intriguing since several pathological patterns and their related pathways are still unravelled. Herein, we report the capability of many phytochemicals, belonging to the macrocategory of polyphenols (resveratrol, curcumin, quercetin, genistein, epigallocatechin gallate), and nonpolyphenols (fatty acids and vitamins), to affect miRNA expression and the involvement of these noncoding RNAs in chronic diseases and cancer (Figure 3).

3.1. Inflammatory and Degenerative Diseases

Several bioactive compounds, such as resveratrol, curcumin, polyunsatured fatty acids (PUFA), and quercetin act as antioxidants by modulating the expression of miRNA involved in crucial inflammatory pathways (Figure 3). Resveratrol displayed anti-inflammatory actions by inducing miR-663 and miR Let7A upregulation and a decrease of miR-155 in human monocytes stressed with LPS [71,72,73]. This polyphenolic molecule ameliorated liver fibrosis and reduced hepatocyte apoptosis by inhibiting miR-190a-5p expression, upregulated by profibrogenetic factors such as TGF-β1 and CCl4 [74]. The reduction of miR-155 levels to counteract the LPS-induced inflammation has also been reported in macrophages treated with curcumin [75] as well as in mouse macrophages and human microvascular endothelial cells. The treatment with PUFA induced the negative regulation of inflammatory-related miRNAs levels, such as miR-146a, miR-146b, miR-21, miR-125a, and miR-155 [76]. In vitro treatment with apple-derived exosomes displayed anti-inflammatory activity via the promotion of miR-146 expression in human macrophages [77]. Recently, the potential therapeutic role of quercetin, resveratrol, curcumin, and vitamin D against the psoriasis-related inflammatory and proliferative pathogenetic pathways via miR-155, miR-146, and miR-125b regulation has been described [78]. The protective action of quercetin in endometriosis has been reported both in vitro and in vivo. Treatment with quercetin led to miR-145 upregulation, thus negatively regulating the TGF-β1/small mother against the decapentaplegic (SMAD)2/SMAD3 pathway and modulating the pathological process of endometrial fibrosis [79]. Additionally, quercetin downregulated cyclin D1 levels, opposing cell proliferation and inducing apoptosis, by enhancing miR-503-5p, miR-1283, miR-3714, and miR-6867-5p levels [80]. In Alzheimer disease, quercetin exhibited a neuroprotective role in maintaining miRNA homeostasis in neuronal cells by preventing miR-125b, miR-26a, and miR-2218 altered expression [81]. Treatment with resveratrol and selenium nanoparticles reduced metabolic dysfunction and neuroinflammation via increased amyloid-β clearance, sirtuin (SIRT)1 upregulation, and STAT3, IL-1β, and miR-143 downregulation in a rat model of Alzheimer disease [82].

3.2. Metabolic Diseases

Many phytochemicals are able to counteract the pathogenesis of metabolic conditions by acting as miRNA regulators (Figure 3). Resveratrol exerted beneficial action on age-related alterations, as senile sarcopenia, by promoting peroxisome proliferator-activated receptor-gamma coactivator 1α (PGC1α) expression and myocyte differentiation by miR-21 and miR-27b upregulation and a decrease of miR-133b, miR-30b, and miR-149 levels [83]. In addition, the resveratrol-induced miR-21 upregulation alleviated cognitive impairment due to insulin resistance and diabetes in mice [84]. The link between saturated fatty acids (SFA) and poor health outcomes and metabolic disorders [85] is already well established; additionally, it is reported that SFA effects on human health could also depend on their influence on miRNAs [86]. Rat myoblast cells treated with palmitic acid (PA) developed insulin resistance and T2DM triggered by miR-29a increase [86]. In addition, PA decreased insulin-induced activation of the PI3K-AKT pathway, enhancing the expression of miR-3180-3p and miR-4632-5p, thus favoring insulin resistance development in HepG2 cells [87]. Stimulation with PA treatment induced mouse cardiomyocyte injuries derived from atrial arrhythmia by targeting miR-27b [88]. High fat and hypercaloric diets can modulate the levels of miRNAs involved in lipid metabolism, cell homeostasis, and fibrogenesis. Evidence demonstrates that fat-rich dietary regimens are associated with downregulation of miR-122 and upregulation of miR-200a, miR-200b, and miR-429 in liver, determining the onset of nonalcoholic fatty liver disease (NAFLD) [89,90]. Similarly, treatment with Moringa oleifera prevented liver damage and nonalcoholic steatohepatitis (NASH) progression via SIRT1 upregulation and miR-21a-5p, miR-103-3p, miR-122-5p, and miR-34a-5p downregulation [91]. The role of vitamins in many metabolic processes, as well as in modulating the immune system and disease prevention, has been extensively reported [92]. To this aim, recent reports revealed that treatment with the carotenoid astaxanthin has been able to promote miR-382-5p expression in hepatic stellate cells, thus opposing liver dysfunctional fibrosis [93]. The potential of vitamin D3 and all trans retinoic acid formulations in the prevention of diabetic cardiovascular complications via miR-126 upregulation has been revealed in diabetic mice [94]. Evidence described the ability of quercetin to ameliorate diabetic nephropathy damage by miR-485-5p upregulation and yes-associated protein 1 (YAP1) suppression in human mesangial cells [95] and to attenuate testosterone secretion dysfunction in diabetic rats by reducing ER stress through miR-1306-5p/hydroxysteroid 17-β dehydrogenase 7 (HSD17B7) axis modulation [96]. The combined treatment of catechin epigallocatechin gallate (EGCG) and quercetin prevented insulin resistance by increasing the expression of miR-27a-3p and miR-96-5p, which directly target Forkhead Box O1 (FOXO1), reducing the production of glucose and the transcription of gluconeogenic enzymes [97]. In addition, ECGC showed therapeutic potential in obesity inhibiting white and beige 3T3-L1 and D12 preadipocyte growth by upregulated miR-let-7a and subsequent high-mobility group AT-hook 2 (HMGA2) suppression [98] and inhibiting the MAPK7 pathway by increased miR-143 levels [99]. Clinical data from a cohort of women affected by overweight and insulin resistance revealed that blood orange juice consumption (500 mL per day) for four weeks resulted in upregulated miR-144-3p, miR-424-5p, miR-144-3p, and miR-130b-3p and decreased let-7f-5p and miR-126-3p levels in peripheral blood mononuclear cells, leading to attenuated IL-6 and NF-κB mRNA expression [100].

3.3. Cancer

Given the beneficial effects mediated by nutrients on miRNA profile, several studies dissected their use as an oncological approach in different neoplastic contexts (Figure 3).

3.3.1. Digestive System Cancers

Resveratrol exerted anti-inflammatory effects and attenuated colitis-induced tumorigenesis by increasing miR-101b and miR-455 levels [101]. In colorectal cancer, resveratrol decreased the expression levels of characterized oncomiRs, as miR-17, miR-21, miR-25, and miR-92a-2 [102], and exerted a pivotal regulation on many tumor suppressors, such as PTEN and SMAD3, as well as on the oncogenic TGF-β pathway related to progression and metastasis of colorectal cancer by targeting the expression levels of miR-1 and miR-146b-5p [103]. The resveratrol-related modulation of the tumor promoter TGF-β1 was further associated with upregulated miR-663 levels as reported in the SW480 cell line [104]. Genistein exhibited in vitro and in vivo anticancer effect inhibiting miR-95 and its targets AKT and serum and glucocorticoid-regulated kinase 1 (SGK1) on HCT116 cells [105,106] and enhanced miR-1275 levels thus suppressing Eukaryotic Translation Initiation Factor 5A2 (EIF5A2)/PI3K/AKT pathway in hepatocellular carcinoma [107]. In mice colorectal cancer tissues, the walnut-based diet PUFAs have been associated with reduced levels of miR-1903, miR-467c, and miR-3068, along with augmented miR-297a expression levels, resulting in anti-inflammatory, antiangiogenetic, antiproliferative and pro-apoptotic effects [108,109]. In vivo studies showed that vitamin D suppressed colorectal cancer cell proliferation, downregulating histone demethylase 1A (JMJD1A) by increasing miR-627 levels [110,111]. The EGCG treatment was able to suppress miR-483 levels via hypermethylation of its promoter region, modulating the expression of metastatic markers as E-cadherin and vimentin in a mouse model of hepatocellular carcinoma [112], while resveratrol opposed the tumor progression via downregulation of miR-155-5p in gastric cancer cells [113].

3.3.2. Hormone-Dependent Cancers

Resveratrol opposed the overexpressed levels of miR-17, miR-18b, miR-20a, miR-20b, miR-92b, miR-106a, and miR-106b in prostate cancer [114,115], whilst genistein was able to restore in vivo and in vitro miR-574-3p expression levels, generally downregulated in prostate cancer cells [116,117]. Treatment with vitamin D positively regulated the levels of miR-100 and miR-125b in primary prostate cancer tissues, opposing tumor progression [118,119], while in breast cancer T47D and SK-BR-3, cell line treatment with retinoic acid strongly increased miR-10a expression and retinoic acid receptor β (RARβ), whose loss is associated with breast carcinogenesis [120]. Vitamin D was also shown to decrease breast cancer cell capacity to elude natural killer lymphocyte attacks through the reduction of miR-302c and miR-520c expression [121,122]. The effect of curcumin treatment on miR-21 was effective in inhibiting the growth of MCF-7 breast cancer cells [123]. Curcumin treatment also positively regulated miR-181b, miR-34a, miR-16, miR-15a, and miR-146b-5p and down-regulated miR-19a, and miR-19b expression in estrogen receptor-positive primary cells and several breast cancer cell lines with effects on inflammatory cascade, cell cycle progression, survival, and invasiveness [124,125]. The curcumin-dependent miR-146b-5p upregulation suppressed the transactivation of the breast stromal fibroblasts, responsible for the epithelial-to-mesenchymal transition in breast cancer stromal fibroblasts [126]. In vitro studies proved that EGCG counteracted breast cancer progression, attenuating the expression of miR-25 [127], while genistein reduced the expression levels of onco-miR-155, the regulator of several tumor suppressors as PTEN and p27, and impaired cell mobility via p21 activated kinase 2 (PAK2) and miR-23b upregulation in breast cancer cells [128,129]. Resveratrol modulated the oncosuppressor ARH-I expression and limited the “awakening” of ovarian cancer cells, reducing the oncomiR-1305 [130].

3.3.3. Respiratory Tract Cancers

In lung cancer cells, curcumin treatment was able to inhibit cell invasion via upregulation of miR-874, which directly targets matrix metalloproteinase-2 (MMP-2), and miR-98, which suppresses MMP-2 and MMP-9 pathways by targeting LIN28A [131]. In human non-small cell lung carcinoma (NSCLC) A549 cell line, curcumin-induced apoptosis and reduced migration and invasion through miR-206 upregulation and suppression of the PI3K/AKT/mTOR signaling pathway [132]. In addition, treatment with curcumin suppressed tumor progression by increasing miR-192-5p expression and c-Myc reduction in NSCLC cells A427 and A549 [133]. EGCG repressed c-myc expression via upregulation of tumor suppressors miR-let-7a-1 and miR-let-7g in lung cancer cells [134,135]. Quercetin displayed anti-cancer properties by opposing cell survival and enhancing apoptosis in NSCLC cells by increased miR-34a-5p and downregulation of a long noncoding RNA small nucleolar RNA host gene 7 (SNHG7) [136], while in oral squamous cell carcinoma Cal-27 cells quercetin inhibited cell proliferation and metastasis via accumulation of miR-1254 and subsequent downregulation of CD36 [137], and suppressed proliferation and MMP-9 and -2 levels via a miR-16/homeobox A10 (HOXA10) axis increase [138]. In vitro treatment with quercetin promoted apoptosis and repressed metastatic feature in esophageal cancer cells by upregulating miR-1-3p and suppression of Transgelin 2 (TAGNL2) expression [139], while an increase in genistein-induced mir-34a led to production of reactive oxygen species (ROS) and apoptosis in head and neck cancer [140].

3.3.4. Neuroectodermal Cancers

Recent reports described the curcumin-induced miR-222-3p upregulation, thus negatively modulating the expression levels of its target SOX10, causing the inactivation of the SOX10/Notch pathway and limiting proliferation, migration, and invasion of melanoma cells [141]. Similarly, treatment with EGCG upregulated miR-let-7b, thus suppressing growth and development in melanoma cells [142]. Resveratrol induced apoptosis via miR-21 in human glioma cells [143] and genistein inhibited the retinoblastoma cell viability by miR-145 upregulation [144]. The glycoside flavonoid purple sweet potato delphinidin-3-rutin (PSPD3R) suppressed in vitro and in vivo glioma proliferation regulating autophagy by inducing the AKT/cAMP response element-binding protein (CREB)/miR-20b-5p/autophagy related 7 (Atg7) pathway [145].

4. Latest Dietary miRNA-Based Animal Models and Clinical Trials

Most of the evidence on the effects of food-borne bioactive compounds on the regulation of human miRNAs in various diseases comes from preliminary in vitro studies, recently validated in animal models. The antidiabetic effects of quercetin and ECGC were proved in mice fed with an enriched-polyphenol diet (0.05% w/w) ad libitum for 10 weeks [97]. Results revealed the capacity of quercetin and EGCG enrichment to increase miR-27a-3p and miR-96-5p and inhibit gluconeogenesis and FOXO1 pathways, with enhanced effects when combined (Table 1) [97]. BALB/c mice overexpressing or not miR-483-3p were treated for two weeks with ad libitum 0%, 0.1%, or 0.5% EGCG solution, then HepG2 cells were inoculated, and the diet prolonged for two months [112]. Supplementation with EGCG reduced hepatocarcinoma lung metastasis and opposed EMT via miR-483-3p inhibition (Table 1) [112]. Although few clinical trials evaluating the relationship between food-borne miRNAs and human health have been conducted, the number of clinical trials is continuously increasing (www.clinicaltrials.gov) (Table 1). New insights into alterations of skeletal muscle miRNA expression when consuming 1 g/kg/h carbohydrate during the first 3 h of recovery from aerobic exercise (NCT03250234) have been recently described [146]. Authors reported that carbohydrate consumption during an 80 min bout of steady-state treadmill exercise led to increased expression of miR-19b-3p, miR-99a-5p, miR-100-5p, miR-222-3p, miR-324-3p, and miR486-5p immediately following and/or within 3 h from recovery compared with a nutrient-free control, thus resulting in downregulation of breakdown protein gene expression and better muscle recovery (Table 1) [146].
Results from the RESMENA study (NCT01087086) evaluated the effect of the Mediterranean dietary pattern on miRNA levels in white blood cells of 40 patients with metabolic syndrome. Data showed that 8-week hypocaloric diet (30% caloric deficiency) based on Mediterranean diet reduced the expression of miR-155, miR-125, miR-130, miR-132, and miR-422, associated with cancer, atherogenic and adipogenic processes, and other inflammatory conditions (Table 1) [147]. The effects of 60 months with a typical Mediterranean regimen (Med diet) versus a low-fat high complex carbohydrate (LFHCC) diet were correlated to T2DM development and differences in miRNA plasma levels in CORDIOPREV study (NCT00924937) [148]. Following LFHCC consumption, patients with low plasma miR-145 levels showed a higher risk of developing T2DM, as well as the subjects assuming a Med diet with low levels of miR-29a, miR-28-3p, and miR-126 and high miR-150 expression (Table 1) [148]. Current therapeutic options for the treatment of antiphospholipid syndrome showed the efficacy of daily ubiquinol treatment (200 mg for 1 month) in the regulation of a profile of monocyte miRNAs and identified novel and specific miRNA-mRNA regulatory networks associated with atherothrombosis development (NCT02218476) [149]. A completed clinical trial (NCT01634841) provided a systematic investigation of the role of walnuts in preventing or slowing age-related cognitive decline and macular degeneration [150]. Authors screened miRNA profiles in serum samples (before and after intervention) of eight randomly selected participants from the walnut arm, showing 53 miRNAs modulated after one year assuming 15% of daily energy as walnuts, 36 of them being upregulated and 17 downregulated [150]. Compared with participants in the control diet, walnuts consumed for 1 year significantly increased the serum concentration of miR-551 related to the inhibited progression of several cancers [150]. The antineoplastic effects of vitamin D were evaluated in prostate cancer [118]. A cohort of prostate cancer patients was treated with three different vitamin D3 doses (400, 10,000 or 40,000 IU/day) in the time between randomization and prostatectomy (approximately 3–8 weeks). Vitamin D3 assumption determined an increase in tumor suppressors miR-100 and miR-125b in both pathological and healthy prostatic tissue [118]. The HYPODD study correlated the vitamin D supplementation to miR-21 circulating levels in the pathogenesis of cardiometabolic disorders [151]. Hypovitaminosis D correction (50,000 UI/week for 8 weeks and then 50,000 UI/month for 10 months) ameliorated the cardiovascular risk profiles in hypertensive patients without affecting the miR-21 circulating expression (Table 1) [151]. The effects of 4-week time-restricted eating (TRE), a popular form of intermittent fasting, determined downregulation of a miRNA panel (miR-4649-5p, miR-2467-3p, miR-543, miR-301a-3p, miR-3132, miR-19a-5p, miR-495-3p, and miR-4761-3p), which, in turn, could inhibit cell growth pathways while activating cell survival and promoting healthy aging (NCT03590847) [152]. A study conducted on 125 subjects with hypercholesterolemia (ACTRN12619000170123) evaluated the effect of daily assumption of a nutraceutical combination containing 400 mg phytosterols, 100 mg bergamot extract, 20 mg olive extract, and 52 µM vitamin K2 up to 12 weeks of treatment [153]. The evaluated nutraceutical combination did not change the serum lipid profile and inflammation-related miRNAs and biomarkers [153].
Table 1. Dose effect sizes, in vivo models, target miRNAs, and biological relevance to foods, constituents, and dietary regimen or supplementation.
Table 1. Dose effect sizes, in vivo models, target miRNAs, and biological relevance to foods, constituents, and dietary regimen or supplementation.
Food, Constituent or RegimenDose Effect SizeModelTarget
miRNA
Biological
Relevance
Refs.
Quercetin0.05% w/wMousemiR-27a-3p,
miR-96-5p
Antidiabetic[97]
EGCG0.05% w/wMousemiR-27a-3p, miR-96-5p, miR-483Antidiabetic[97]
0%, 0.1%, 0.5% solution ad libitumMousemiR-483Anticancer properties in hepatocellular carcinoma[112]
Walnut30–60 g/dHumanmiR-551Anticancer properties[150]
Vitamin D400, 10,000 or 40,000 IU/dayHumanmiR-100,
miR-125b
Anticancer properties in prostate cancer[118]
50,000 UI/week for 8 weeks, 50,000 UI/month for 10 monthsHumanmiR-21Lower cardiovascular risk in hypertensive patients[151]
Carbohydrates1 g/kg/h in 3 h after 2 cycle ergometry glycogen depletion HumanmiR-19b-3p, miR-99a-5p, miR-100-5p, miR-222-3p, miR-324-3p, miR-486-5pEnhanced recovery after exercise [146]
Ubiquitinol200 mg/dHuman20 different miRNAsAnti-phospholipids syndrome[149]
Mediterranean diet8-week hypocaloric dietHumanmiR-155, miR-125, miR-130, miR-132, miR-422Anti-inflammatory, anticancer,
anti-atherogenic
[147]
low-fat high complex carbohydrate vs. Mediterranean diet for 60 monthsHumanmiR-145, miR-29a, miR-28-3p, miR-126, miR-150Diabetic risk[148]
Nutraceutical combination400 mg phytosterols, 100 mg bergamot extract, 20 mg olive extract, 52 µM vitamin K2HumanmiR-21, miR-146a miR-126No significant effects on lipid and inflammatory profile, as on miRNA levels[153]

5. Questions Opening on the Potential Impact of Dietary miRNAs on Health and Disease

Increasing evidence highlight the potential of dietary miRNAs to modulate human pathophysiology, suggesting new dietary-based intervention approaches [154]. However, a preventive strategy based on xenomiRNAs denotes multiple limitations, due to their poor bioavailability, the restricted knowledge of the potential side effects related to high consumption, and the appropriate dietary intake according to their stability due to manufacturing processes and food storage [155]. The bioavailability of food-targeted miRNAs after ingestion is still a controversial issue. In vitro and in vivo evidences reported upregulated miR-29b, miR-200c, miR-21-5p, and miR-30a-5p plasma levels after milk consumption, supporting the availability of these molecules [17,156]. On the other hand, given the base sequence complementarity between miR-21-5p and miR-30a-5p in humans and animals, the precise assessment of xenomiRNA uptaken by diet is quite uncertain [157]. Moreover, the increase in miRNA plasmatic concentration could be ascribed to endogenous responses to other milk-derived compounds [158]. Analyses by real time quantitative PCR (RT-qPCR) and RNA-sequencing on human blood did not provide supporting evidences about the absorption of specific bovine miRNAs after milk consumption [159], as well as results on milk-fed mice highlighted the absence of miRNA absorption and revealed their rapid degradation in intestinal fluids [160]. Analyses by RT-qPCR performed on post-prandial plasma of pigtail macaques reported absent or low-levels of nonspecific amplified miRNAs, thus supporting the scarce intestinal absorption of plant xenomiRNAs [161]. Likewise, the plant-derived miR-168 has been detected in feces and gastrointestinal mucosa while being undetectable in blood, confirming that the potential availability of gastrointestinal tract is not accompanied by systemic absorption [162]. On the other hand, the detection of the plant miR-172 in gastrointestinal tract, serum and feces of mice fed with plant RNA extracts, confirmed the possibility of a dietary-derived miRNA amount absorbed by intestinal tract [163]. Growing evidence reported xenomiRNAs as food contaminants, describing some plant-derived miRNAs as a consequence of experimental contamination and artifacts [19,164]. In addition, the biological and functional role of dietary miRNAs from different sources in human health is still controversial. Different milk-derived miRNAs have been reported to modulate oncogenic and adipogenic pathways, promoting the development of B-cell lymphoma, hormone-dependent breast and prostate cancer [43,44,45,47], as well as the ginger-related miRNA has been associated with colitis development [64] and the pork-derived miRNAs to the development of metabolic disorders in mice [22]. The evaluation of food-derived xenomiRNA effects cannot refrain from considering the co-existence of a great variety of dietary bioactive compounds. In the absence of supplementary intake of dietary exogenous miRNAs, many phytochemicals or animal-based food derivatives may modify the risk of diseases, targeting the reported biological effects [29,165]. Indeed, several food-derived compounds exert their regulatory effects on different diseases either by upregulating or downregulating different signaling pathways, as well as acting on many epigenetic patterns [166]. Dietary constituents are able to affect epigenetic mechanisms, including DNA status modulation, histone methylation, and acetylation, by aiming key epigenetic modulators such as DNA methyltransferases and histone deacetylases, thus resulting in local or global changes in epigenetics and subsequent gene transcription and expression levels [29,165,166]. Attractive strategies from a better knowledge of nutrimiromics are undeniable [167], although further studies are needed to determine the pharmacokinetics and the possible side effects of dietary miRNA-based intervention in human health. Based on current evidence here summarized, it has to take into account both the promising results of food-derived miRNAs and negative effects exerted by the dietary molecules on human health, thus their dual role in chronic diseases still represents an open door in nutrition-based strategies.

6. Conclusions

Herein, we provided an up-to-date comprehensive review of food-derived miRNA activities in human health and disease, reporting both beneficial and controversial issues of dietary miRNAs as regulators of chronic conditions. Recent reports aim to elucidate the association between altered miRNA expression and pathological processes contributing to the onset of several chronic diseases. The crucial roles of different miRNAs in the development of T2DM, insulin resistance and obesity [168,169,170,171] have been described, as well as in the progression of other chronic syndromes, such as asthma, allergy, and chronic kidney disease (CKD) [172,173,174,175,176]. This evidence points out nutrient-regulated miRNAs and food-derived miRNAs as intriguing players in food-related health and disease, although the knowledge on miRNA specific role is partially known and still debatable, requiring future studies to broaden knowledge on possible molecular targets and their modulation. In this regard, lifestyle habits, affecting the expression levels of miRNAs, represent a crucial element in the perspective of preventive and personalized medicine. A healthy diet with a personalized choice of nutrients based on individual needs related to age and state of health, i.e., ranging from health to disease or disability/rehabilitative conditions, is essential for maintaining the state of health and to slow down the aging clock. To this end, dietary miRNAs could represent intervention tools in precision nutrition, although with several limitations to overcome, including bioinstability, poor availability, unknown side effects, and multiple molecular targets. Within this framework, future preclinical and clinical studies will benefit from multidisciplinary and translational design taking into consideration some host-related aspects such as microbiome, circadian rhythm and self-susceptibility, and miRNA-derived limitations, as scarce bioavailability and undefined content in body fluids, which undoubtedly add complexity in dissecting the still controversial role of dietary miRNAs in human health and diseases.

Author Contributions

Conceptualization, E.M., N.D. and M.L.B.; methodology, E.M., N.D. and M.L.B.; visualization, A.C. and C.A.; writing- original draft, E.M., N.D. and M.L.B.; supervision, A.B., C.S., R.M., G.C. and M.L.B.; funding acquisition, N.D. and M.L.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by STUPORANIMA Project, Funder: Italian Ministry of Health-RC, Number: IZS ME 09/22; ENDO-CARE Project, Funder: Ministero dell’Università e della Ricerca, Next Generation EU Number: P2022SZE5Y; Fuelmilk Project, Funder: Ministero dell’Università e della Ricerca, Number: 2022KH87XS; FORMULAMI Project, Funder: Ministero dello Sviluppo Economico, Number: F/310110/04/X56.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Di Renzo, L.; Gualtieri, P.; Romano, L.; Marrone, G.; Noce, A.; Pujia, A.; Perrone, M.A.; Aiello, V.; Colica, C.; De Lorenzo, A. Role of Personalized Nutrition in Chronic-Degenerative Diseases. Nutrients 2019, 11, 1707. [Google Scholar] [CrossRef]
  2. Shi, Y.; Liu, Z.; Lin, Q.; Luo, Q.; Cen, Y.; Li, J.; Fang, X.; Gong, C. MiRNAs and Cancer: Key Link in Diagnosis and Therapy. Genes 2021, 12, 1289. [Google Scholar] [CrossRef]
  3. Leitão, A.L.; Enguita, F.J. A Structural View of miRNA Biogenesis and Function. Noncoding RNA 2022, 8, 10. [Google Scholar] [CrossRef]
  4. Sadri, M.; Shu, J.; Kachman, S.D.; Cui, J.; Zempleni, J. Milk exosomes and miRNA cross the placenta and promote embryo survival in mice. Reproduction 2020, 160, 501–509. [Google Scholar] [CrossRef]
  5. Mori, M.A.; Ludwig, R.G.; Garcia-Martin, R.; Brandão, B.B.; Kahn, C.R. Extracellular miRNAs: From Biomarkers to Mediators of Physiology and Disease. Cell Metab. 2019, 30, 656–673. [Google Scholar] [CrossRef]
  6. Kučuk, N.; Primožič, M.; Knez, Ž.; Leitgeb, M. Exosomes Engineering and Their Roles as Therapy Delivery Tools, Therapeutic Targets, and Biomarkers. Int. J. Mol. Sci. 2021, 22, 9543. [Google Scholar] [CrossRef]
  7. Abozaid, Y.J.; Zhang, X.; Mens, M.M.J.; Ahmadizar, F.; Limpens, M.; Ikram, M.A.; Rivadeneira, F.; Voortman, T.; Kavousi, M.; Ghanbari, M. Plasma circulating microRNAs associated with obesity, body fat distribution, and fat mass: The Rotterdam Study. Int. J. Obes. 2022, 46, 2137–2144. [Google Scholar] [CrossRef]
  8. McNeill, E.M.; Hirschi, K.D. Roles of Regulatory RNAs in Nutritional Control. Annu. Rev. Nutr. 2020, 40, 77–104. [Google Scholar] [CrossRef]
  9. Quintanilha, B.J.; Reis, B.Z.; Duarte, G.B.S.; Cozzolino, S.M.F.; Rogero, M.M. Nutrimiromics: Role of microRNAs and Nutrition in Modulating Inflammation and Chronic Diseases. Nutrients 2017, 9, 1168. [Google Scholar] [CrossRef]
  10. Sanchita; Trivedi, R.; Asif, M.H.; Trivedi, P.K. Dietary plant miRNAs as an augmented therapy: Cross-kingdom gene regulation. RNA Biol. 2018, 15, 1433–1439. [Google Scholar] [CrossRef]
  11. Lukasik, A.; Brzozowska, I.; Zielenkiewicz, U.; Zielenkiewicz, P. Detection of Plant miRNAs Abundance in Human Breast Milk. Int. J. Mol. Sci. 2017, 19, 37. [Google Scholar] [CrossRef]
  12. Cannataro, R.; Cione, E. Diet and miRNA: Epigenetic Regulator or a New Class of Supplements? Microrna 2022, 11, 89–90. [Google Scholar] [CrossRef]
  13. Otsuka, K.; Ochiya, T. Possible connection between diet and microRNA in cancer scenario. Semin. Cancer Biol. 2021, 73, 4–18. [Google Scholar] [CrossRef]
  14. Rasheed, Z.; Rasheed, N.; Al-Shaya, O. Epigallocatechin-3-O-gallate modulates global microRNA expression in interleukin-1β-stimulated human osteoarthritis chondrocytes: Potential role of EGCG on negative co-regulation of microRNA-140-3p and ADAMTS5. Eur. J. Nutr. 2018, 57, 917–928. [Google Scholar] [CrossRef]
  15. Preethi, K.A.; Sekar, D. Dietary microRNAs: Current status and perspective in food science. J. Food Biochem. 2021, 45, e13827. [Google Scholar] [CrossRef]
  16. Fan, Y.; Habib, M.; Xia, J. Xeno-miRNet: A comprehensive database and analytics platform to explore xeno-miRNAs and their potential targets. PeerJ 2018, 6, e5650. [Google Scholar] [CrossRef]
  17. Baier, S.R.; Nguyen, C.; Xie, F.; Wood, J.R.; Zempleni, J. MicroRNAs are absorbed in biologically meaningful amounts from nutritionally relevant doses of cow milk and affect gene expression in peripheral blood mononuclear cells, HEK-293 kidney cell cultures, and mouse livers. J. Nutr. 2014, 144, 1495–1500. [Google Scholar] [CrossRef]
  18. Zhang, L.; Chen, T.; Yin, Y.; Zhang, C.Y.; Zhang, Y.L. Dietary microRNA-A Novel Functional Component of Food. Adv. Nutr. 2019, 10, 711–721. [Google Scholar] [CrossRef]
  19. Fromm, B.; Kang, W.; Rovira, C.; Cayota, A.; Witwer, K.; Friedländer, M.R.; Tosar, J.P. Plant microRNAs in human sera are likely contaminants. J. Nutr. Biochem. 2019, 65, 139–140. [Google Scholar] [CrossRef]
  20. Wade, B.; Cummins, M.; Keyburn, A.; Crowley, T.M. Isolation and detection of microRNA from the egg of chickens. BMC Res. Notes 2016, 9, 283. [Google Scholar] [CrossRef]
  21. Fratantonio, D.; Munir, J.; Shu, J.; Howard, K.; Baier, S.R.; Cui, J.; Zempleni, J. The RNA cargo in small extracellular vesicles from chicken eggs is bioactive in C57BL/6 J mice and human peripheral blood mononuclear cells ex vivo. Front. Nutr. 2023, 10, 1162679. [Google Scholar] [CrossRef]
  22. Shen, L.; Ma, J.; Yang, Y.; Liao, T.; Wang, J.; Chen, L.; Zhang, S.; Zhao, Y.; Niu, L.; Hao, X.; et al. Cooked pork-derived exosome nanovesicles mediate metabolic disorder-microRNA could be the culprit. J. Nanobiotechnol. 2023, 21, 83. [Google Scholar] [CrossRef]
  23. Pieri, M.; Theori, E.; Dweep, H.; Flourentzou, M.; Kalampalika, F.; Maniori, M.A.; Papagregoriou, G.; Papaneophytou, C.; Felekkis, K. A bovine miRNA, bta-miR-154c, withstands in vitro human digestion but does not affect cell viability of colorectal human cell lines after transfection. FEBS Open Bio 2022, 12, 925–936. [Google Scholar] [CrossRef]
  24. Servillo, L.; D’Onofrio, N.; Giovane, A.; Casale, R.; Cautela, D.; Castaldo, D.; Iannaccone, F.; Neglia, G.; Campanile, G.; Balestrieri, M.L. Ruminant meat and milk contain δ-valerobetaine, another precursor of trimethylamine N-oxide (TMAO) like γ-butyrobetaine. Food Chem. 2018, 260, 193–199. [Google Scholar] [CrossRef]
  25. D’Onofrio, N.; Balestrieri, A.; Neglia, G.; Monaco, A.; Tatullo, M.; Casale, R.; Limone, A.; Balestrieri, M.L.; Campanile, G. Antioxidant and Anti-Inflammatory Activities of Buffalo Milk δ-Valerobetaine. J. Agric. Food Chem. 2019, 67, 1702–1710. [Google Scholar] [CrossRef]
  26. D’Onofrio, N.; Cacciola, N.A.; Martino, E.; Borrelli, F.; Fiorino, F.; Lombardi, A.; Neglia, G.; Balestrieri, M.L.; Campanile, G. ROS-Mediated Apoptotic Cell Death of Human Colon Cancer LoVo Cells by Milk δ-Valerobetaine. Sci. Rep. 2020, 10, 8978. [Google Scholar] [CrossRef]
  27. D’Onofrio, N.; Mele, L.; Martino, E.; Salzano, A.; Restucci, B.; Cautela, D.; Tatullo, M.; Balestrieri, M.L.; Campanile, G. Synergistic Effect of Dietary Betaines on SIRT1-Mediated Apoptosis in Human Oral Squamous Cell Carcinoma Cal 27. Cancers 2020, 12, 2468. [Google Scholar] [CrossRef]
  28. D’Onofrio, N.; Martino, E.; Mele, L.; Colloca, A.; Maione, M.; Cautela, D.; Castaldo, D.; Balestrieri, M.L. Colorectal Cancer Apoptosis Induced by Dietary δ-Valerobetaine Involves PINK1/Parkin Dependent-Mitophagy and SIRT3. Int. J. Mol. Sci. 2021, 22, 8117. [Google Scholar] [CrossRef]
  29. López de Las Hazas, M.C.; Del Pozo-Acebo, L.; Hansen, M.S.; Gil-Zamorano, J.; Mantilla-Escalante, D.C.; Gómez-Coronado, D.; Marín, F.; Garcia-Ruiz, A.; Rasmussen, J.T.; Dávalos, A. Dietary bovine milk miRNAs transported in extracellular vesicles are partially stable during GI digestion, are bioavailable and reach target tissues but need a minimum dose to impact on gene expression. Eur. J. Nutr. 2022, 61, 1043–1056. [Google Scholar] [CrossRef]
  30. Tong, L.; Hao, H.; Zhang, Z.; Lv, Y.; Liang, X.; Liu, Q.; Liu, T.; Gong, P.; Zhang, L.; Cao, F.; et al. Milk-derived extracellular vesicles alleviate ulcerative colitis by regulating the gut immunity and reshaping the gut microbiota. Theranostics 2021, 11, 8570–8586. [Google Scholar] [CrossRef]
  31. García-Martínez, J.; Pérez-Castillo, Í.M.; Salto, R.; López-Pedrosa, J.M.; Rueda, R.; Girón, M.D. Beneficial Effects of Bovine Milk Exosomes in Metabolic Interorgan Cross-Talk. Nutrients 2022, 14, 1442. [Google Scholar] [CrossRef] [PubMed]
  32. Chen, Z.; Xie, Y.; Luo, J.; Chen, T.; Xi, Q.; Zhang, Y.; Sun, J. Milk exosome-derived miRNAs from water buffalo are implicated in immune response and metabolism process. BMC Vet. Res. 2020, 16, 123. [Google Scholar] [CrossRef] [PubMed]
  33. Melnik, B.C. Milk exosomal miRNAs: Potential drivers of AMPK-to-mTORC1 switching in β-cell de-differentiation of type 2 diabetes mellitus. Nutr. Metab. 2019, 16, 85. [Google Scholar] [CrossRef] [PubMed]
  34. Melnik, B.C. Synergistic Effects of Milk-Derived Exosomes and Galactose on α-Synuclein Pathology in Parkinson’s Disease and Type 2 Diabetes Mellitus. Int. J. Mol. Sci. 2021, 22, 1059. [Google Scholar] [CrossRef] [PubMed]
  35. Shome, S.; Jernigan, R.L.; Beitz, D.C.; Clark, S.; Testroet, E.D. Non-coding RNA in raw and commercially processed milk and putative targets related to growth and immune-response. BMC Genom. 2021, 22, 749. [Google Scholar] [CrossRef] [PubMed]
  36. Aarts, J.; Boleij, A.; Pieters, B.C.H.; Feitsma, A.L.; van Neerven, R.J.J.; Ten Klooster, J.P.; M’Rabet, L.; Arntz, O.J.; Koenders, M.I.; van de Loo, F.A.J. Flood Control: How Milk-Derived Extracellular Vesicles Can Help to Improve the Intestinal Barrier Function and Break the Gut-Joint Axis in Rheumatoid Arthritis. Front. Immunol. 2021, 12, 703277. [Google Scholar] [CrossRef] [PubMed]
  37. Kaur, K.; Vig, S.; Srivastava, R.; Mishra, A.; Singh, V.P.; Srivastava, A.K.; Datta, M. Elevated Hepatic miR-22-3p Expression Impairs Gluconeogenesis by Silencing the Wnt-Responsive Transcription Factor Tcf7. Diabetes 2015, 64, 3659–3669. [Google Scholar] [CrossRef] [PubMed]
  38. Lönnerdal, B. Human Milk MicroRNAs/Exosomes: Composition and Biological Effects; Nestlé Nutrition Institute Workshop Series; Nestlé Nutrition Institute: La Tour-de-Peilz, Switzerland; S. Karger AG: Basel, Switzerland, 2019; Volume 90, pp. 83–92. [Google Scholar] [CrossRef]
  39. Su, Y.; Yuan, J.; Zhang, F.; Lei, Q.; Zhang, T.; Li, K.; Guo, J.; Hong, Y.; Bu, G.; Lv, X.; et al. MicroRNA-181a-5p and microRNA-181a-3p cooperatively restrict vascular inflammation and atherosclerosis. Cell Death Dis. 2019, 10, 365. [Google Scholar] [CrossRef]
  40. Lin, Y.; Lu, Y.; Huang, Z.; Wang, Y.; Song, S.; Luo, Y.; Ren, F.; Guo, H. Milk-Derived Small Extracellular Vesicles Promote Recovery of Intestinal Damage by Accelerating Intestinal Stem Cell-Mediated Epithelial Regeneration. Mol. Nutr. Food Res. 2022, 66, e2100551. [Google Scholar] [CrossRef]
  41. Yan, C.; Chen, J.; Wang, C.; Yuan, M.; Kang, Y.; Wu, Z.; Li, W.; Zhang, G.; Machens, H.G.; Rinkevich, Y.; et al. Milk exosomes-mediated miR-31-5p delivery accelerates diabetic wound healing through promoting angiogenesis. Drug Deliv. 2022, 29, 214–228. [Google Scholar] [CrossRef]
  42. Jiang, R.; Lönnerdal, B. Milk-Derived miR-22-3p Promotes Proliferation of Human Intestinal Epithelial Cells (HIECs) by Regulating Gene Expression. Nutrients 2022, 14, 4901. [Google Scholar] [CrossRef] [PubMed]
  43. Abbas, M.A.; Al-Saigh, N.N.; Saqallah, F.G. Regulation of adipogenesis by exosomal milk miRNA. Rev. Endocr. Metab. Disord. 2023, 24, 297–316. [Google Scholar] [CrossRef] [PubMed]
  44. Melnik, B.C.; Stadler, R.; Weiskirchen, R.; Leitzmann, C.; Schmitz, G. Potential Pathogenic Impact of Cow’s Milk Consumption and Bovine Milk-Derived Exosomal MicroRNAs in Diffuse Large B-Cell Lymphoma. Int. J. Mol. Sci. 2023, 24, 6102. [Google Scholar] [CrossRef] [PubMed]
  45. Melnik, B.C. Lifetime Impact of Cow’s Milk on Overactivation of mTORC1: From Fetal to Childhood Overgrowth, Acne, Diabetes, Cancers, and Neurodegeneration. Biomolecules 2021, 11, 404. [Google Scholar] [CrossRef] [PubMed]
  46. Melnik, B.C.; Schmitz, G. Exosomes of pasteurized milk: Potential pathogens of Western diseases. J. Transl. Med. 2019, 17, 3. [Google Scholar] [CrossRef] [PubMed]
  47. Melnik, B.C.; John, S.M.; Carrera-Bastos, P.; Cordain, L.; Leitzmann, C.; Weiskirchen, R.; Schmitz, G. The Role of Cow’s Milk Consumption in Breast Cancer Initiation and Progression. Curr. Nutr. Rep. 2023, 12, 122–140. [Google Scholar] [CrossRef] [PubMed]
  48. Shi, X.B.; Xue, L.; Ma, A.H.; Tepper, C.G.; Kung, H.J.; White, R.W. miR-125b promotes growth of prostate cancer xenograft tumor through targeting pro-apoptotic genes. Prostate 2011, 71, 538–549. [Google Scholar] [CrossRef]
  49. Zhang, X.; Chen, X.; Xu, Y.; Yang, J.; Du, L.; Li, K.; Zhou, Y. Milk consumption and multiple health outcomes: Umbrella review of systematic reviews and meta-analyses in humans. Nutr. Metab. 2021, 18, 7. [Google Scholar] [CrossRef]
  50. Martino, E.; Balestrieri, A.; Aragona, F.; Bifulco, G.; Mele, L.; Campanile, G.; Balestrieri, M.L.; D’Onofrio, N. MiR-148a-3p Promotes Colorectal Cancer Cell Ferroptosis by Targeting SLC7A11. Cancers 2023, 15, 4342. [Google Scholar] [CrossRef]
  51. Martino, E.; Balestrieri, A.; Mele, L.; Sardu, C.; Marfella, R.; D’Onofrio, N.; Campanile, G.; Balestrieri, M.L. Milk Exosomal miR-27b Worsen Endoplasmic Reticulum Stress Mediated Colorectal Cancer Cell Death. Nutrients 2022, 14, 5081. [Google Scholar] [CrossRef]
  52. Hou, D.; He, F.; Ma, L.; Cao, M.; Zhou, Z.; Wei, Z.; Xue, Y.; Sang, X.; Chong, H.; Tian, C.; et al. The potential atheroprotective role of plant MIR156a as a repressor of monocyte recruitment on inflamed human endothelial cells. J. Nutr. Biochem. 2018, 57, 197–205. [Google Scholar] [CrossRef] [PubMed]
  53. Li, M.; Chen, T.; He, J.J.; Wu, J.H.; Luo, J.Y.; Ye, R.S.; Xie, M.Y.; Zhang, H.J.; Zeng, B.; Liu, J.; et al. Plant MIR167e-5p Inhibits Enterocyte Proliferation by Targeting β-Catenin. Cells 2019, 8, 1385. [Google Scholar] [CrossRef] [PubMed]
  54. Alshehri, B. Plant-derived xenomiRs and cancer: Cross-kingdom gene regulation. Saudi J. Biol. Sci. 2021, 28, 2408–2422. [Google Scholar] [CrossRef] [PubMed]
  55. Chin, A.R.; Fong, M.Y.; Somlo, G.; Wu, J.; Swiderski, P.; Wu, X.; Wang, S.E. Cross-kingdom inhibition of breast cancer growth by plant miR159. Cell Res. 2016, 26, 217–228. [Google Scholar] [CrossRef] [PubMed]
  56. Xiao, X.; Sticht, C.; Yin, L.; Liu, L.; Karakhanova, S.; Yin, Y.; Georgikou, C.; Gladkich, J.; Gross, W.; Gretz, N.; et al. Novel plant microRNAs from broccoletti sprouts do not show cross-kingdom regulation of pancreatic cancer. Oncotarget 2020, 11, 1203–1217. [Google Scholar] [CrossRef] [PubMed]
  57. Rakhmetullina, A.; Pyrkova, A.; Aisina, D.; Ivashchenko, A. In silico prediction of human genes as potential targets for rice miRNAs. Comput. Biol. Chem. 2020, 87, 107305. [Google Scholar] [CrossRef] [PubMed]
  58. Kumazoe, M.; Ogawa, F.; Hikida, A.; Shimada, Y.; Yoshitomi, R.; Watanabe, R.; Onda, H.; Fujimura, Y.; Tachibana, H. Plant miRNA osa-miR172d-5p suppressed lung fibrosis by targeting Tab1. Sci. Rep. 2023, 13, 2128. [Google Scholar] [CrossRef]
  59. Zhang, L.; Hou, D.; Chen, X.; Li, D.; Zhu, L.; Zhang, Y.; Li, J.; Bian, Z.; Liang, X.; Cai, X.; et al. Exogenous plant MIR168a specifically targets mammalian LDLRAP1: Evidence of cross-kingdom regulation by microRNA. Cell Res. 2012, 22, 107–126. [Google Scholar] [CrossRef]
  60. Rabuma, T.; Gupta, O.P.; Chhokar, V. Recent advances and potential applications of cross-kingdom movement of miRNAs in modulating plant’s disease response. RNA Biol. 2022, 19, 519–532. [Google Scholar] [CrossRef]
  61. Akao, Y.; Kuranaga, Y.; Heishima, K.; Sugito, N.; Morikawa, K.; Ito, Y.; Soga, T.; Ito, T. Plant hvu-MIR168-3p enhances expression of glucose transporter 1 (SLC2A1) in human cells by silencing genes related to mitochondrial electron transport chain complex I. J. Nutr. Biochem. 2022, 101, 108922. [Google Scholar] [CrossRef]
  62. Link, J.; Thon, C.; Petkevicius, V.; Steponaitiene, R.; Malfertheiner, P.; Kupcinskas, J.; Link, A. The Translational Impact of Plant-Derived Xeno-miRNA miR-168 in Gastrointestinal Cancers and Preneoplastic Conditions. Diagnostics 2023, 13, 2701. [Google Scholar] [CrossRef] [PubMed]
  63. Xiao, J.; Feng, S.; Wang, X.; Long, K.; Luo, Y.; Wang, Y.; Ma, J.; Tang, Q.; Jin, L.; Li, X.; et al. Identification of exosome-like nanoparticle-derived microRNAs from 11 edible fruits and vegetables. PeerJ 2018, 6, e5186. [Google Scholar] [CrossRef] [PubMed]
  64. Manzaneque-López, M.C.; Sánchez-López, C.M.; Pérez-Bermúdez, P.; Soler, C.; Marcilla, A. Dietary-Derived Exosome-like Nanoparticles as Bacterial Modulators: Beyond MicroRNAs. Nutrients 2023, 15, 1265. [Google Scholar] [CrossRef] [PubMed]
  65. Teng, Y.; Ren, Y.; Sayed, M.; Hu, X.; Lei, C.; Kumar, A.; Hutchins, E.; Mu, J.; Deng, Z.; Luo, C.; et al. Plant-Derived Exosomal MicroRNAs Shape the Gut Microbiota. Cell Host Microbe 2018, 24, 637–652.e8. [Google Scholar] [CrossRef] [PubMed]
  66. Mlotshwa, S.; Pruss, G.J.; MacArthur, J.L.; Endres, M.W.; Davis, C.; Hofseth, L.J.; Peña, M.M.; Vance, V. A novel chemopreventive strategy based on therapeutic microRNAs produced in plants. Cell Res. 2015, 25, 521–524. [Google Scholar] [CrossRef] [PubMed]
  67. Liu, J.; Wang, F.; Weng, Z.; Sui, X.; Fang, Y.; Tang, X.; Shen, X. Soybean-derived miRNAs specifically inhibit proliferation and stimulate apoptosis of human colonic Caco-2 cancer cells but not normal mucosal cells in culture. Genomics 2020, 112, 2949–2958. [Google Scholar] [CrossRef]
  68. Marzano, F.; Caratozzolo, M.F.; Consiglio, A.; Licciulli, F.; Liuni, S.; Sbisà, E.; D’Elia, D.; Tullo, A.; Catalano, D. Plant miRNAs Reduce Cancer Cell Proliferation by Targeting MALAT1 and NEAT1: A Beneficial Cross-Kingdom Interaction. Front Genet. 2020, 11, 552490. [Google Scholar] [CrossRef]
  69. Yu, W.Y.; Cai, W.; Ying, H.Z.; Zhang, W.Y.; Zhang, H.H.; Yu, C.H. Exogenous Plant gma-miR-159a, Identified by miRNA Library Functional Screening, Ameliorated Hepatic Stellate Cell Activation and Inflammation via Inhibiting GSK-3β-Mediated Pathways. J. Inflamm. Res. 2021, 14, 2157–2172. [Google Scholar] [CrossRef]
  70. Komori, H.; Fujita, D.; Shirasaki, Y.; Zhu, Q.; Iwamoto, Y.; Nakanishi, T.; Nakajima, M.; Tamai, I. MicroRNAs in Apple-Derived Nanoparticles Modulate Intestinal Expression of Organic Anion-Transporting Peptide 2B1/SLCO2B1 in Caco-2 Cells. Drug Metab. Dispos. 2021, 49, 803–809. [Google Scholar] [CrossRef]
  71. Javaid, A.; Zahra, D.; Rashid, F.; Mashraqi, M.; Alzamami, A.; Khurshid, M.; Ali Ashfaq, U. Regulation of micro-RNA, epigenetic factor by natural products for the treatment of cancers: Mechanistic insight and translational association. Saudi J. Biol. Sci. 2022, 29, 103255. [Google Scholar] [CrossRef]
  72. Song, J.; Jun, M.; Ahn, M.R.; Kim, O.Y. Involvement of miR-Let7A in inflammatory response and cell survival/apoptosis regulated by resveratrol in THP-1 macrophage. Nutr. Res. Pract. 2016, 10, 377–384. [Google Scholar] [CrossRef] [PubMed]
  73. Tili, E.; Michaille, J.J.; Adair, B.; Alder, H.; Limagne, E.; Taccioli, C.; Ferracin, M.; Delmas, D.; Latruffe, N.; Croce, C.M. Resveratrol decreases the levels of miR-155 by upregulating miR-663, a microRNA targeting JunB and JunD. Carcinogenesis 2010, 31, 1561–1566. [Google Scholar] [CrossRef] [PubMed]
  74. Liang, F.; Xu, X.; Tu, Y. Resveratrol inhibited hepatocyte apoptosis and alleviated liver fibrosis through miR-190a-5p/HGF axis. Bioorganic Med. Chem. 2022, 57, 116593. [Google Scholar] [CrossRef] [PubMed]
  75. Ma, F.; Liu, F.; Ding, L.; You, M.; Yue, H.; Zhou, Y.; Hou, Y. Anti-inflammatory effects of curcumin are associated with down regulating microRNA-155 in LPS-treated macrophages and mice. Pharm. Biol. 2017, 55, 1263–1273. [Google Scholar] [CrossRef] [PubMed]
  76. Roessler, C.; Kuhlmann, K.; Hellwing, C.; Leimert, A.; Schumann, J. Impact of Polyunsaturated Fatty Acids on miRNA Profiles of Monocytes/Macrophages and Endothelial Cells-A Pilot Study. Int. J. Mol. Sci. 2017, 18, 284. [Google Scholar] [CrossRef] [PubMed]
  77. Trentini, M.; Zanotti, F.; Tiengo, E.; Camponogara, F.; Degasperi, M.; Licastro, D.; Lovatti, L.; Zavan, B. An Apple a Day Keeps the Doctor Away: Potential Role of miRNA 146 on Macrophages Treated with Exosomes Derived from Apples. Biomedicines 2022, 10, 415. [Google Scholar] [CrossRef] [PubMed]
  78. Duchnik, E.; Kruk, J.; Tuchowska, A.; Marchlewicz, M. The Impact of Diet and Physical Activity on Psoriasis: A Narrative Review of the Current Evidence. Nutrients 2023, 15, 840. [Google Scholar] [CrossRef]
  79. Xu, J.; Tan, Y.L.; Liu, Q.Y.; Huang, Z.C.; Qiao, Z.H.; Li, T.; Hu, Z.Q.; Lei, L. Quercetin regulates fibrogenic responses of endometrial stromal cell by upregulating miR-145 and inhibiting the TGF-β1/Smad2/Smad3 pathway. Acta Histochem. 2020, 122, 151600. [Google Scholar] [CrossRef]
  80. Park, S.; Lim, W.; Bazer, F.W.; Whang, K.Y.; Song, G. Quercetin inhibits proliferation of endometriosis regulating cyclin D1 and its target microRNAs in vitro and in vivo. J. Nutr. Biochem. 2019, 63, 87–100. [Google Scholar] [CrossRef]
  81. Benameur, T.; Soleti, R.; Porro, C. The Potential Neuroprotective Role of Free and Encapsulated Quercetin Mediated by miRNA against Neurological Diseases. Nutrients 2021, 13, 1318. [Google Scholar] [CrossRef]
  82. Abozaid, O.A.R.; Sallam, M.W.; El-Sonbaty, S.; Aziza, S.; Emad, B.; Ahmed, E.S.A. Resveratrol-Selenium Nanoparticles Alleviate Neuroinflammation and Neurotoxicity in a Rat Model of Alzheimer’s Disease by Regulating Sirt1/miRNA-134/GSK3β Expression. Biol. Trace Elem. Res. 2022, 200, 5104–5114. [Google Scholar] [CrossRef] [PubMed]
  83. Cannataro, R.; Carbone, L.; Petro, J.L.; Cione, E.; Vargas, S.; Angulo, H.; Forero, D.A.; Odriozola-Martínez, A.; Kreider, R.B.; Bonilla, D.A. Sarcopenia: Etiology, Nutritional Approaches, and miRNAs. Int. J. Mol. Sci. 2021, 22, 9724. [Google Scholar] [CrossRef] [PubMed]
  84. El-Sayed, N.S.; Elatrebi, S.; Said, R.; Ibrahim, H.F.; Omar, E.M. Potential mechanisms underlying the association between type II diabetes mellitus and cognitive dysfunction in rats: A link between miRNA-21 and Resveratrol’s neuroprotective action. Metab. Brain Dis. 2022, 37, 2375–2388. [Google Scholar] [CrossRef] [PubMed]
  85. Unger, A.L.; Torres-Gonzalez, M.; Kraft, J. Dairy Fat Consumption and the Risk of Metabolic Syndrome: An Examination of the Saturated Fatty Acids in Dairy. Nutrients 2019, 11, 2200. [Google Scholar] [CrossRef] [PubMed]
  86. Yang, W.M.; Jeong, H.J.; Park, S.Y.; Lee, W. Induction of miR-29a by saturated fatty acids impairs insulin signaling and glucose uptake through translational repression of IRS-1 in myocytes. FEBS Lett. 2014, 588, 2170–2176. [Google Scholar] [CrossRef] [PubMed]
  87. Tashiro, E.; Nagasawa, Y.; Itoh, S.; Imoto, M. Involvement of miR-3180-3p and miR-4632-5p in palmitic acid-induced insulin resistance. Mol. Cell Endocrinol. 2021, 534, 111371. [Google Scholar] [CrossRef]
  88. Takahashi, K.; Sasano, T.; Sugiyama, K.; Kurokawa, J.; Tamura, N.; Soejima, Y.; Sawabe, M.; Isobe, M.; Furukawa, T. High-fat diet increases vulnerability to atrial arrhythmia by conduction disturbance via miR-27b. J. Mol. Cell Cardiol. 2016, 90, 38–46. [Google Scholar] [CrossRef]
  89. Alisi, A.; Da Sacco, L.; Bruscalupi, G.; Piemonte, F.; Panera, N.; De Vito, R.; Leoni, S.; Bottazzo, G.F.; Masotti, A.; Nobili, V. Mirnome analysis reveals novel molecular determinants in the pathogenesis of diet-induced nonalcoholic fatty liver disease. Lab Investig. 2011, 91, 283–293. [Google Scholar] [CrossRef]
  90. Deng, P.; Li, M.; Wu, Y. The Predictive Efficacy of Serum Exosomal microRNA-122 and microRNA-148a for Hepatocellular Carcinoma Based on Smart Healthcare. J. Healthc. Eng. 2022, 2022, 5914541. [Google Scholar] [CrossRef]
  91. Monraz-Méndez, C.A.; Escutia-Gutiérrez, R.; Rodriguez-Sanabria, J.S.; Galicia-Moreno, M.; Monroy-Ramírez, H.C.; Sánchez-Orozco, L.; García-Bañuelos, J.; De la Rosa-Bibiano, R.; Santos, A.; Armendáriz-Borunda, J.; et al. Moringa oleifera Improves MAFLD by Inducing Epigenetic Modifications. Nutrients 2022, 14, 4225. [Google Scholar] [CrossRef]
  92. Nur, S.M.; Rath, S.; Ahmad, V.; Ahmad, A.; Ateeq, B.; Khan, M.I. Nutritive vitamins as epidrugs. Crit. Rev. Food Sci. Nutr. 2021, 61, 1–13. [Google Scholar] [CrossRef] [PubMed]
  93. 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] [PubMed]
  94. Sharifzadeh, M.; Esmaeili-Bandboni, A.; Emami, M.R.; Naeini, F.; Zarezadeh, M.; Javanbakht, M.H. The effects of all trans retinoic acid, vitamin D3 and their combination on plasma levels of miRNA-125a-5p, miRNA-34a, and miRNA-126 in an experimental model of diabetes. Avicenna J. Phytomed. 2022, 12, 67–76. [Google Scholar] [CrossRef] [PubMed]
  95. Wan, H.; Wang, Y.; Pan, Q.; Chen, X.; Chen, S.; Li, X.; Yao, W. Quercetin attenuates the proliferation, inflammation, and oxidative stress of high glucose-induced human mesangial cells by regulating the miR-485-5p/YAP1 pathway. Int. J. Immunopathol. Pharmacol. 2022, 36, 20587384211066440. [Google Scholar] [CrossRef] [PubMed]
  96. Wang, D.; Li, Y.; Zhai, Q.Q.; Zhu, Y.F.; Liu, B.Y.; Xu, Y. Quercetin ameliorates testosterone secretion disorder by inhibiting endoplasmic reticulum stress through the miR-1306-5p/HSD17B7 axis in diabetic rats. Bosn. J. Basic Med. Sci. 2022, 22, 191–204. [Google Scholar] [CrossRef] [PubMed]
  97. Liu, H.; Guan, H.; Tan, X.; Jiang, Y.; Li, F.; Sun-Waterhouse, D.; Li, D. Enhanced alleviation of insulin resistance via the IRS-1/Akt/FOXO1 pathway by combining quercetin and EGCG and involving miR-27a-3p and miR-96-5p. Free Radic. Biol. Med. 2022, 181, 105–117. [Google Scholar] [CrossRef] [PubMed]
  98. Chen, W.T.; Yang, M.J.; Tsuei, Y.W.; Su, T.C.; Siao, A.C.; Kuo, Y.C.; Huang, L.R.; Chen, Y.; Chen, S.J.; Chen, P.C.; et al. Green Tea Epigallocatechin Gallate Inhibits Preadipocyte Growth via the microRNA-let-7a/HMGA2 Signaling Pathway. Mol. Nutr. Food Res. 2023, 67, e2200336. [Google Scholar] [CrossRef]
  99. Chen, C.P.; Su, T.C.; Yang, M.J.; Chen, W.T.; Siao, A.C.; Huang, L.R.; Lin, Y.Y.; Kuo, Y.C.; Chung, J.F.; Cheng, C.F.; et al. Green tea epigallocatechin gallate suppresses 3T3-L1 cell growth via microRNA-143/MAPK7 pathways. Exp. Biol. Med. 2022, 247, 1670–1679. [Google Scholar] [CrossRef]
  100. Capetini, V.C.; Quintanilha, B.J.; de Oliveira, D.C.; Nishioka, A.H.; de Matos, L.A.; Ferreira, L.R.P.; Ferreira, F.M.; Sampaio, G.R.; Hassimotto, N.M.A.; Lajolo, F.M.; et al. Blood orange juice intake modulates plasma and PBMC microRNA expression in overweight and insulin-resistant women: Impact on MAPK and NFκB signaling pathways. J. Nutr. Biochem. 2023, 112, 109240. [Google Scholar] [CrossRef]
  101. Gowd, V.; Kanika; Jori, C.; Chaudhary, A.A.; Rudayni, H.A.; Rashid, S.; Khan, R. Resveratrol and resveratrol nano-delivery systems in the treatment of inflammatory bowel disease. J. Nutr. Biochem. 2022, 109, 109101. [Google Scholar] [CrossRef]
  102. Cadieux, Z.; Lewis, H.; Esquela-Kerscher, A. Role of Nutrition, the Epigenome, and microRNAs in Cancer Pathogenesis; The Royal Society of Chemistry: London, UK, 2019; Volume 1, pp. 1–35. [Google Scholar] [CrossRef]
  103. Farooqi, A.A.; Khalid, S.; Ahmad, A. Regulation of Cell Signaling Pathways and miRNAs by Resveratrol in Different Cancers. Int. J. Mol. Sci. 2018, 19, 652. [Google Scholar] [CrossRef] [PubMed]
  104. Huang, L.; Zhang, S.; Zhou, J.; Li, X. Effect of resveratrol on drug resistance in colon cancer chemotherapy. RSC Adv. 2019, 9, 2572–2580. [Google Scholar] [CrossRef] [PubMed]
  105. Khan, A.W.; Farooq, M.; Haseeb, M.; Choi, S. Role of Plant-Derived Active Constituents in Cancer Treatment and Their Mechanisms of Action. Cells 2022, 11, 1326. [Google Scholar] [CrossRef] [PubMed]
  106. Qin, J.; Chen, J.X.; Zhu, Z.; Teng, J.A. Genistein inhibits human colorectal cancer growth and suppresses miR-95, Akt and SGK1. Cell Physiol. Biochem. 2015, 35, 2069–2077. [Google Scholar] [CrossRef] [PubMed]
  107. Yang, X.; Jiang, W.; Kong, X.; Zhou, X.; Zhu, D.; Kong, L. Genistein Restricts the Epithelial Mesenchymal Transformation (EMT) and Stemness of Hepatocellular Carcinoma via Upregulating miR-1275 to Inhibit the EIF5A2/PI3K/Akt Pathway. Biology 2022, 11, 1383. [Google Scholar] [CrossRef]
  108. Tsoukas, M.A.; Ko, B.J.; Witte, T.R.; Dincer, F.; Hardman, W.E.; Mantzoros, C.S. Dietary walnut suppression of colorectal cancer in mice: Mediation by miRNA patterns and fatty acid incorporation. J. Nutr. Biochem. 2015, 26, 776–783. [Google Scholar] [CrossRef]
  109. Tu, M.; Wang, W.; Zhang, G.; Hammock, B.D. ω-3 Polyunsaturated Fatty Acids on Colonic Inflammation and Colon Cancer: Roles of Lipid-Metabolizing Enzymes Involved. Nutrients 2020, 12, 3301. [Google Scholar] [CrossRef]
  110. Negri, M.; Gentile, A.; de Angelis, C.; Montò, T.; Patalano, R.; Colao, A.; Pivonello, R.; Pivonello, C. Vitamin D-Induced Molecular Mechanisms to Potentiate Cancer Therapy and to Reverse Drug-Resistance in Cancer Cells. Nutrients 2020, 12, 1798. [Google Scholar] [CrossRef]
  111. Padi, S.K.; Zhang, Q.; Rustum, Y.M.; Morrison, C.; Guo, B. MicroRNA-627 mediates the epigenetic mechanisms of vitamin D to suppress proliferation of human colorectal cancer cells and growth of xenograft tumors in mice. Gastroenterology 2013, 145, 437–446. [Google Scholar] [CrossRef]
  112. Kang, Q.; Tong, Y.; Gowd, V.; Wang, M.; Chen, F.; Cheng, K.W. Oral administration of EGCG solution equivalent to daily achievable dosages of regular tea drinkers effectively suppresses miR483-3p induced metastasis of hepatocellular carcinoma cells in mice. Food Funct. 2021, 12, 3381–3392. [Google Scholar] [CrossRef]
  113. Su, N.; Li, L.; Zhou, E.; Li, H.; Wu, S.; Cao, Z. Resveratrol Downregulates miR-155-5p to Block the Malignant Behavior of Gastric Cancer Cells. BioMed Res. Int. 2022, 2022, 6968641. [Google Scholar] [CrossRef] [PubMed]
  114. Dhar, S.; Hicks, C.; Levenson, A.S. Resveratrol and prostate cancer: Promising role for microRNAs. Mol. Nutr. Food Res. 2011, 55, 1219–1229. [Google Scholar] [CrossRef] [PubMed]
  115. Maleki Dana, P.; Sadoughi, F.; Asemi, Z.; Yousefi, B. The role of polyphenols in overcoming cancer drug resistance: A comprehensive review. Cell Mol. Biol. Lett. 2022, 27, 1. [Google Scholar] [CrossRef] [PubMed]
  116. Chiyomaru, T.; Yamamura, S.; Fukuhara, S.; Hidaka, H.; Majid, S.; Saini, S.; Arora, S.; Deng, G.; Shahryari, V.; Chang, I.; et al. Genistein up-regulates tumor suppressor microRNA-574-3p in prostate cancer. PLoS ONE 2013, 8, e58929. [Google Scholar] [CrossRef] [PubMed]
  117. Ji, X.; Liu, K.; Li, Q.; Shen, Q.; Han, F.; Ye, Q.; Zheng, C. A Mini-Review of Flavone Isomers Apigenin and Genistein in Prostate Cancer Treatment. Front. Pharmacol. 2022, 13, 851589. [Google Scholar] [CrossRef] [PubMed]
  118. Giangreco, A.A.; Vaishnav, A.; Wagner, D.; Finelli, A.; Fleshner, N.; Van der Kwast, T.; Vieth, R.; Nonn, L. Tumor suppressor microRNAs, miR-100 and -125b, are regulated by 1,25-dihydroxyvitamin D in primary prostate cells and in patient tissue. Cancer Prev. Res. 2013, 6, 483–494. [Google Scholar] [CrossRef] [PubMed]
  119. Stephan, C.; Ralla, B.; Bonn, F.; Diesner, M.; Lein, M.; Jung, K. Vitamin D Metabolites in Nonmetastatic High-Risk Prostate Cancer Patients with and without Zoledronic Acid Treatment after Prostatectomy. Cancers 2022, 14, 1560. [Google Scholar] [CrossRef] [PubMed]
  120. Khan, S.; Wall, D.; Curran, C.; Newell, J.; Kerin, M.J.; Dwyer, R.M. MicroRNA-10a is reduced in breast cancer and regulated in part through retinoic acid. BMC Cancer 2015, 15, 345. [Google Scholar] [CrossRef]
  121. Min, D.; Lv, X.B.; Wang, X.; Zhang, B.; Meng, W.; Yu, F.; Hu, H. Downregulation of miR-302c and miR-520c by 1,25(OH)2D3 treatment enhances the susceptibility of tumour cells to natural killer cell-mediated cytotoxicity. Br. J. Cancer 2013, 109, 723–730. [Google Scholar] [CrossRef]
  122. Muñoz, A.; Grant, W.B. Vitamin D and Cancer: An Historical Overview of the Epidemiology and Mechanisms. Nutrients 2022, 14, 1448. [Google Scholar] [CrossRef]
  123. Wang, X.; Hang, Y.; Liu, J.; Hou, Y.; Wang, N.; Wang, M. Anticancer effect of curcumin inhibits cell growth through miR-21/PTEN/Akt pathway in breast cancer cell. Oncol. Lett. 2017, 13, 4825–4831. [Google Scholar] [CrossRef] [PubMed]
  124. Kronski, E.; Fiori, M.E.; Barbieri, O.; Astigiano, S.; Mirisola, V.; Killian, P.H.; Bruno, A.; Pagani, A.; Rovera, F.; Pfeffer, U.; et al. miR181b is induced by the chemopreventive polyphenol curcumin and inhibits breast cancer metastasis via down-regulation of the inflammatory cytokines CXCL1 and -2. Mol. Oncol. 2014, 8, 581–595. [Google Scholar] [CrossRef] [PubMed]
  125. Norouzi, S.; Majeed, M.; Pirro, M.; Generali, D.; Sahebkar, A. Curcumin as an Adjunct Therapy and microRNA Modulator in Breast Cancer. Curr. Pharm. Des. 2018, 24, 171–177. [Google Scholar] [CrossRef] [PubMed]
  126. Al-Ansari, M.M.; Aboussekhra, A. miR-146b-5p mediates p16-dependent repression of IL-6 and suppresses paracrine procarcinogenic effects of breast stromal fibroblasts. Oncotarget 2015, 6, 30006–30016. [Google Scholar] [CrossRef] [PubMed]
  127. Zan, L.; Chen, Q.; Zhang, L.; Li, X. Epigallocatechin gallate (EGCG) suppresses growth and tumorigenicity in breast cancer cells by downregulation of miR-25. Bioengineered 2019, 10, 374–382. [Google Scholar] [CrossRef]
  128. 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]
  129. Javed, Z.; Khan, K.; Herrera-Bravo, J.; Naeem, S.; Iqbal, M.J.; Sadia, H.; Qadri, Q.R.; Raza, S.; Irshad, A.; Akbar, A.; et al. Genistein as a regulator of signaling pathways and microRNAs in different types of cancers. Cancer Cell Int. 2021, 21, 388. [Google Scholar] [CrossRef] [PubMed]
  130. Esposito, A.; Ferraresi, A.; Salwa, A.; Vidoni, C.; Dhanasekaran, D.N.; Isidoro, C. Resveratrol Contrasts IL-6 Pro-Growth Effects and Promotes Autophagy-Mediated Cancer Cell Dormancy in 3D Ovarian Cancer: Role of miR-1305 and of Its Target ARH-I. Cancers 2022, 14, 2142. [Google Scholar] [CrossRef]
  131. Wan Mohd Tajuddin, W.N.B.; Lajis, N.H.; Abas, F.; Othman, I.; Naidu, R. Mechanistic Understanding of Curcumin’s Therapeutic Effects in Lung Cancer. Nutrients 2019, 11, 2989. [Google Scholar] [CrossRef]
  132. Wang, N.; Feng, T.; Liu, X.; Liu, Q. Curcumin inhibits migration and invasion of non-small cell lung cancer cells through up-regulation of miR-206 and suppression of PI3K/AKT/mTOR signaling pathway. Acta Pharm. 2020, 70, 399–409. [Google Scholar] [CrossRef]
  133. Pan, Y.; Sun, Y.; Liu, Z.; Zhang, C. miR-192-5p upregulation mediates the suppression of curcumin in human NSCLC cell proliferation, migration and invasion by targeting c-Myc and inactivating the Wnt/β-catenin signaling pathway. Mol. Med. Rep. 2020, 22, 1594–1604. [Google Scholar] [CrossRef] [PubMed]
  134. Zhong, Z.; Dong, Z.; Yang, L.; Chen, X.; Gong, Z. Inhibition of proliferation of human lung cancer cells by green tea catechins is mediated by upregulation of let-7. Exp. Ther. Med. 2012, 4, 267–272. [Google Scholar] [CrossRef] [PubMed]
  135. Qamar, M.; Akhtar, S.; Ismail, T.; Wahid, M.; Barnard, R.T.; Esatbeyoglu, T.; Ziora, Z.M. The Chemical Composition and Health-Promoting Effects of the Grewia Species—A Systematic Review and Meta-Analysis. Nutrients 2021, 13, 4565. [Google Scholar] [CrossRef] [PubMed]
  136. Chai, R.; Xu, C.; Lu, L.; Liu, X.; Ma, Z. Quercetin inhibits proliferation of and induces apoptosis in non-small-cell lung carcinoma via the lncRNA SNHG7/miR-34a-5p pathway. Immunopharmacol. Immunotoxicol. 2021, 43, 693–703. [Google Scholar] [CrossRef] [PubMed]
  137. Chen, L.; Xia, J.S.; Wu, J.H.; Chen, Y.G.; Qiu, C.J. Quercetin suppresses cell survival and invasion in oral squamous cell carcinoma via the miR-1254/CD36 cascade in vitro. Hum. Exp. Toxicol. 2021, 40, 1413–1421. [Google Scholar] [CrossRef] [PubMed]
  138. Zhao, J.; Fang, Z.; Zha, Z.; Sun, Q.; Wang, H.; Sun, M.; Qiao, B. Quercetin inhibits cell viability, migration and invasion by regulating miR-16/HOXA10 axis in oral cancer. Eur. J. Pharmacol. 2019, 847, 11–18. [Google Scholar] [CrossRef] [PubMed]
  139. Wang, Y.; Chen, X.; Li, J.; Xia, C. Quercetin Antagonizes Esophagus Cancer by Modulating miR-1-3p/TAGLN2 Pathway-Dependent Growth and Metastasis. Nutr. Cancer 2022, 74, 1872–1881. [Google Scholar] [CrossRef] [PubMed]
  140. Hsieh, P.L.; Liao, Y.W.; Hsieh, C.W.; Chen, P.N.; Yu, C.C. Soy Isoflavone Genistein Impedes Cancer Stemness and Mesenchymal Transition in Head and Neck Cancer through Activating miR-34a/RTCB Axis. Nutrients 2020, 12, 1924. [Google Scholar] [CrossRef]
  141. Tang, Y.; Cao, Y. Curcumin Inhibits the Growth and Metastasis of Melanoma via miR-222-3p/SOX10/Notch Axis. Dis. Markers 2022, 2022, 3129781. [Google Scholar] [CrossRef]
  142. Yamada, S.; Tsukamoto, S.; Huang, Y.; Makio, A.; Kumazoe, M.; Yamashita, S.; Tachibana, H. Epigallocatechin-3-O-gallate up-regulates microRNA-let-7b expression by activating 67-kDa laminin receptor signaling in melanoma cells. Sci. Rep. 2016, 6, 19225. [Google Scholar] [CrossRef]
  143. Li, H.; Jia, Z.; Li, A.; Jenkins, G.; Yang, X.; Hu, J.; Guo, W. Resveratrol repressed viability of U251 cells by miR-21 inhibiting of NF-κB pathway. Mol. Cell Biochem. 2013, 382, 137–143. [Google Scholar] [CrossRef]
  144. Wei, D.; Yang, L.; Lv, B.; Chen, L. Genistein suppresses retinoblastoma cell viability and growth and induces apoptosis by upregulating miR-145 and inhibiting its target ABCE1. Mol. Vis. 2017, 23, 385–394. [Google Scholar]
  145. Wang, M.; Liu, K.; Bu, H.; Cong, H.; Dong, G.; Xu, N.; Li, C.; Zhao, Y.; Jiang, F.; Zhang, Y.; et al. Purple sweet potato delphinidin-3-rutin represses glioma proliferation by inducing miR-20b-5p/Atg7-dependent cytostatic autophagy. Mol. Ther. Oncolytics 2022, 26, 314–329. [Google Scholar] [CrossRef]
  146. Margolis, L.M.; Carrigan, C.T.; Murphy, N.E.; DiBella, M.N.; Wilson, M.A.; Whitney, C.C.; Howard, E.E.; Pasiakos, S.M.; Rivas, D.A. Carbohydrate intake in recovery from aerobic exercise differentiates skeletal muscle microRNA expression. Am. J. Physiol Endocrinol. Metab. 2022, 323, E435–E447. [Google Scholar] [CrossRef]
  147. Marques-Rocha, J.L.; Milagro, F.I.; Mansego, M.L.; Zulet, M.A.; Bressan, J.; Martínez, J.A. Expression of inflammation-related miRNAs in white blood cells from subjects with metabolic syndrome after 8 wk of following a Mediterranean diet-based weight loss program. Nutrition 2016, 32, 48–55. [Google Scholar] [CrossRef]
  148. Jimenez-Lucena, R.; Alcala-Diaz, J.F.; Roncero-Ramos, I.; Lopez-Moreno, J.; Camargo, A.; Gomez-Delgado, F.; Quintana-Navarro, G.M.; Vals-Delgado, C.; Rodriguez-Cantalejo, F.; Luque, R.M.; et al. MiRNAs profile as biomarkers of nutritional therapy for the prevention of type 2 diabetes mellitus: From the CORDIOPREV study. Clin. Nutr. 2021, 40, 1028–1038. [Google Scholar] [CrossRef]
  149. Pérez-Sánchez, C.; Aguirre, M.Á.; Ruiz-Limón, P.; Ábalos-Aguilera, M.C.; Jiménez-Gómez, Y.; Arias-de la Rosa, I.; Rodriguez-Ariza, A.; Fernández-Del Río, L.; González-Reyes, J.A.; Segui, P.; et al. Ubiquinol Effects on Antiphospholipid Syndrome Prothrombotic Profile: A Randomized, Placebo-Controlled Trial. Arter. Thromb. Vasc. Biol. 2017, 37, 1923–1932. [Google Scholar] [CrossRef]
  150. Gil-Zamorano, J.; Cofán, M.; López de Las Hazas, M.C.; García-Blanco, T.; García-Ruiz, A.; Doménech, M.; Serra-Mir, M.; Roth, I.; Valls-Pedret, C.; Rajaram, S.; et al. Interplay of Walnut Consumption, Changes in Circulating miRNAs and Reduction in LDL-Cholesterol in Elders. Nutrients 2022, 14, 1473. [Google Scholar] [CrossRef] [PubMed]
  151. Rendina, D.; D Elia, L.; Abate, V.; Rebellato, A.; Buondonno, I.; Succoio, M.; Martinelli, F.; Muscariello, R.; De Filippo, G.; D Amelio, P.; et al. Vitamin D Status, Cardiovascular Risk Profile, and miRNA-21 Levels in Hypertensive Patients: Results of the HYPODD Study. Nutrients 2022, 14, 2683. [Google Scholar] [CrossRef] [PubMed]
  152. Saini, S.K.; Singh, A.; Saini, M.; Gonzalez-Freire, M.; Leeuwenburgh, C.; Anton, S.D. Time-Restricted Eating Regimen Differentially Affects Circulatory miRNA Expression in Older Overweight Adults. Nutrients 2022, 14, 1843. [Google Scholar] [CrossRef] [PubMed]
  153. Protic, O.; Di Pillo, R.; Montesanto, A.; Galeazzi, R.; Matacchione, G.; Giuliani, A.; Sabbatinelli, J.; Gurău, F.; Silvestrini, A.; Olivieri, F.; et al. Randomized, Double-Blind, Placebo-Controlled Trial to Test the Effects of a Nutraceutical Combination Monacolin K-Free on the Lipid and Inflammatory Profile of Subjects with Hypercholesterolemia. Nutrients 2022, 14, 2812. [Google Scholar] [CrossRef]
  154. Li, Y.; Teng, Z.; Zhao, D. Plant-derived cross-kingdom gene regulation benefits human health. Trends Plant Sci. 2023, 28, 626–629. [Google Scholar] [CrossRef]
  155. Mar-Aguilar, F.; Arreola-Triana, A.; Mata-Cardona, D.; Gonzalez-Villasana, V.; Rodríguez-Padilla, C.; Reséndez-Pérez, D. Evidence of transfer of miRNAs from the diet to the blood still inconclusive. PeerJ 2020, 8, e9567. [Google Scholar] [CrossRef]
  156. Wang, L.; Sadri, M.; Giraud, D.; Zempleni, J. RNase H2-Dependent Polymerase Chain Reaction and Elimination of Confounders in Sample Collection, Storage, and Analysis Strengthen Evidence That microRNAs in Bovine Milk Are Bioavailable in Humans. J. Nutr. 2018, 148, 153–159. [Google Scholar] [CrossRef]
  157. Fromm, B.; Tosar, J.P.; Lu, Y.; Halushka, M.K.; Witwer, K.W. Human and Cow Have Identical miR-21-5p and miR-30a-5p Sequences, Which Are Likely Unsuited to Study Dietary Uptake from Cow Milk. J. Nutr. 2018, 148, 1506–1507. [Google Scholar] [CrossRef]
  158. Witwer, K.W. Diet-responsive mammalian miRNAs are likely endogenous. J. Nutr. 2014, 144, 1880–1881. [Google Scholar] [CrossRef]
  159. Auerbach, A.; Vyas, G.; Li, A.; Halushka, M.; Witwer, K. Uptake of dietary milk miRNAs by adult humans: A validation study. F1000Res 2016, 5, 721. [Google Scholar] [CrossRef]
  160. Title, A.C.; Denzler, R.; Stoffel, M. Uptake and Function Studies of Maternal Milk-derived MicroRNAs. J. Biol. Chem. 2015, 290, 23680–23691. [Google Scholar] [CrossRef]
  161. Witwer, K.W.; McAlexander, M.A.; Queen, S.E.; Adams, R.J. Real-time quantitative PCR and droplet digital PCR for plant miRNAs in mammalian blood provide little evidence for general uptake of dietary miRNAs: Limited evidence for general uptake of dietary plant xenomiRs. RNA Biol. 2013, 10, 1080–1086. [Google Scholar] [CrossRef] [PubMed]
  162. Link, J.; Thon, C.; Schanze, D.; Steponaitiene, R.; Kupcinskas, J.; Zenker, M.; Canbay, A.; Malfertheiner, P.; Link, A. Food-Derived Xeno-microRNAs: Influence of Diet and Detectability in Gastrointestinal Tract-Proof-of-Principle Study. Mol. Nutr. Food Res. 2019, 63, e1800076. [Google Scholar] [CrossRef] [PubMed]
  163. Liang, G.; Zhu, Y.; Sun, B.; Shao, Y.; Jing, A.; Wang, J.; Xiao, Z. Assessing the survival of exogenous plant microRNA in mice. Food Sci. Nutr. 2014, 2, 380–388. [Google Scholar] [CrossRef]
  164. Witwer, K.W. Alternative miRNAs? Human sequences misidentified as plant miRNAs in plant studies and in human plasma. F1000Research 2018, 7, 244. [Google Scholar] [CrossRef]
  165. Jia, M.; He, J.; Bai, W.; Lin, Q.; Deng, J.; Li, W.; Bai, J.; Fu, D.; Ma, Y.; Ren, J.; et al. Cross-kingdom regulation by dietary plant miRNAs: An evidence-based review with recent updates. Food Funct. 2021, 12, 9549–9562. [Google Scholar] [CrossRef]
  166. Agrawal, P.; Kaur, J.; Singh, J.; Rasane, P.; Sharma, K.; Bhadariya, V.; Kaur, S.; Kumar, V. Genetics, Nutrition, and Health: A New Frontier in Disease Prevention. J. Am. Nutr. Assoc. 2023, 28, 1–13. [Google Scholar] [CrossRef]
  167. 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. 2018, 62, 1700080. [Google Scholar] [CrossRef]
  168. Dalgaard, L.T.; Sørensen, A.E.; Hardikar, A.A.; Joglekar, M.V. The microRNA-29 family: Role in metabolism and metabolic disease. Am. J. Physiol. Cell Physiol. 2022, 323, C367–C377. [Google Scholar] [CrossRef]
  169. Hung, Y.H.; Kanke, M.; Kurtz, C.L.; Cubitt, R.; Bunaciu, R.P.; Miao, J.; Zhou, L.; Graham, J.L.; Hussain, M.M.; Havel, P.; et al. Acute suppression of insulin resistance-associated hepatic miR-29 in vivo improves glycemic control in adult mice. Physiol. Genom. 2019, 51, 379–389. [Google Scholar] [CrossRef]
  170. Ma, E.; Fu, Y.; Garvey, W.T. Relationship of Circulating miRNAs with Insulin Sensitivity and Associated Metabolic Risk Factors in Humans. Metab. Syndr. Relat. Disord. 2018, 16, 82–89. [Google Scholar] [CrossRef] [PubMed]
  171. Pordzik, J.; Eyileten-Postuła, C.; Jakubik, D.; Czajka, P.; Nowak, A.; De Rosa, S.; Gąsecka, A.; Cieślicka-Kapłon, A.; Sulikowski, P.; Filipiak, K.J.; et al. MiR-126 Is an Independent Predictor of Long-Term All-Cause Mortality in Patients with Type 2 Diabetes Mellitus. J. Clin. Med. 2021, 10, 2371. [Google Scholar] [CrossRef] [PubMed]
  172. Fourdinier, O.; Schepers, E.; Metzinger-Le Meuth, V.; Glorieux, G.; Liabeuf, S.; Verbeke, F.; Vanholder, R.; Brigant, B.; Pletinck, A.; Diouf, M.; et al. Serum levels of miR-126 and miR-223 and outcomes in chronic kidney disease patients. Sci. Rep. 2019, 9, 4477. [Google Scholar] [CrossRef] [PubMed]
  173. Franczyk, B.; Gluba-Brzózka, A.; Olszewski, R.; Parolczyk, M.; Rysz-Górzyńska, M.; Rysz, J. miRNA biomarkers in renal disease. Int. Urol. Nephrol. 2022, 54, 575–588. [Google Scholar] [CrossRef] [PubMed]
  174. Kim, J.Y.; Stevens, P.; Karpurapu, M.; Lee, H.; Englert, J.A.; Yan, P.; Lee, T.J.; Pabla, N.; Pietrzak, M.; Park, G.Y.; et al. Targeting ETosis by miR-155 inhibition mitigates mixed granulocytic asthmatic lung inflammation. Front. Immunol. 2022, 13, 943554. [Google Scholar] [CrossRef]
  175. Weidner, J.; Bartel, S.; Kılıç, A.; Zissler, U.M.; Renz, H.; Schwarze, J.; Schmidt-Weber, C.B.; Maes, T.; Rebane, A.; Krauss-Etschmann, S.; et al. Spotlight on microRNAs in allergy and asthma. Allergy 2021, 76, 1661–1678. [Google Scholar] [CrossRef]
  176. Zhang, K.; Liang, Y.; Feng, Y.; Wu, W.; Zhang, H.; He, J.; Hu, Q.; Zhao, J.; Xu, Y.; Liu, Z.; et al. Decreased epithelial and sputum miR-221-3p associates with airway eosinophilic inflammation and CXCL17 expression in asthma. Am. J. Physiol. Lung Cell Mol. Physiol. 2018, 315, L253–L264. [Google Scholar] [CrossRef]
Figure 1. XenomiRNAs from animal and vegetable sources. Exogenous miRNAs from animal and vegetable sources are absorbed by the gastrointestinal tract, packaged into vesicles, and delivered via blood stream to cells and tissues. Here, exogenous miRNAs are able to regulate the development of chronic diseases, such as inflammation, fibrosis, metabolic disorders, neurological conditions, cardiovascular diseases, and cancer, by modulating different cellular pathways. The up arrows stand for upregulation, while the down ones indicate downregulation. GSK-3β, glycogen synthase kinase-3β; IL, interleukin; JAM-A, junctional adhesion molecule A; LDL, low-density lipoprotein; LPS, lipopolysaccharide; MALAT1, metastasis-associated lung adenocarcinoma transcript 1; miR/miRNA, microRNA; NEAT1, nuclear paraspeckle assembly transcript 1; NF-κB, nuclear factor kappaB; RISC, RNA-induced silencing complex; SLCO2B1, Solute Carrier Organic Anion Transporter Family Member 2B1; SLM, spatial learning and memory; Tab1, TGF-β activated kinase 1 (MAP3K7) binding protein 1; TCF7, transcription factor 7; TGF-β1, transforming growth factor-β1.
Figure 1. XenomiRNAs from animal and vegetable sources. Exogenous miRNAs from animal and vegetable sources are absorbed by the gastrointestinal tract, packaged into vesicles, and delivered via blood stream to cells and tissues. Here, exogenous miRNAs are able to regulate the development of chronic diseases, such as inflammation, fibrosis, metabolic disorders, neurological conditions, cardiovascular diseases, and cancer, by modulating different cellular pathways. The up arrows stand for upregulation, while the down ones indicate downregulation. GSK-3β, glycogen synthase kinase-3β; IL, interleukin; JAM-A, junctional adhesion molecule A; LDL, low-density lipoprotein; LPS, lipopolysaccharide; MALAT1, metastasis-associated lung adenocarcinoma transcript 1; miR/miRNA, microRNA; NEAT1, nuclear paraspeckle assembly transcript 1; NF-κB, nuclear factor kappaB; RISC, RNA-induced silencing complex; SLCO2B1, Solute Carrier Organic Anion Transporter Family Member 2B1; SLM, spatial learning and memory; Tab1, TGF-β activated kinase 1 (MAP3K7) binding protein 1; TCF7, transcription factor 7; TGF-β1, transforming growth factor-β1.
Nutrients 16 00160 g001
Figure 2. Milk-derived miRNAs and human health. Identified in exosomal vesicles, miRNAs contained in milk are involved in a wide range of metabolic pathways, as the normal function and differentiation of T and B cells, homeostasis of oral cavity, pathogenesis of Parkinson disease, anti-atherogenic action, promotion of angiogenesis, and breast and prostate cancer. MiRNAs contained in milk are also able to counteract diabetes and colorectal cancer development, as well as regulate autophagy in post-acute myocardial infarction. The green arrows stand for promoting activity, while the red ones indicate an opposing role.
Figure 2. Milk-derived miRNAs and human health. Identified in exosomal vesicles, miRNAs contained in milk are involved in a wide range of metabolic pathways, as the normal function and differentiation of T and B cells, homeostasis of oral cavity, pathogenesis of Parkinson disease, anti-atherogenic action, promotion of angiogenesis, and breast and prostate cancer. MiRNAs contained in milk are also able to counteract diabetes and colorectal cancer development, as well as regulate autophagy in post-acute myocardial infarction. The green arrows stand for promoting activity, while the red ones indicate an opposing role.
Nutrients 16 00160 g002
Figure 3. Diet-derived nutrients and miRNA expression. Bioactive compounds, such as curcumin, quercetin, vitamins, fatty acids, EGCG, resveratrol, and genistein, affect human miRNA expression and regulate several pathways, thus modulating the development of pathological chronic states, including inflammatory diseases, metabolic disorders, and cancer. The up arrows stand for upregulation, while the down ones indicate downregulation. AKT, protein kinase B; EGCG, epigallocatechin gallate; FOXO1, Forkhead Box O1; JMJD1A, Jumonji domain-containing 1A; LIN28A, Lin-28 Homolog A; LPS, lipopolysaccharide; miR, microRNA; PAK2, p21 activated kinase 2; MMP, matrix metalloproteinase; PGC1α, peroxisome proliferator-activated receptor gamma coactivator 1α; PI3K, phosphatidyl inositol 3-kinase; PTEN, phosphatase and tensin homolog; SMAD, small mother against decapentaplegic; TGF, transforming growth factor.
Figure 3. Diet-derived nutrients and miRNA expression. Bioactive compounds, such as curcumin, quercetin, vitamins, fatty acids, EGCG, resveratrol, and genistein, affect human miRNA expression and regulate several pathways, thus modulating the development of pathological chronic states, including inflammatory diseases, metabolic disorders, and cancer. The up arrows stand for upregulation, while the down ones indicate downregulation. AKT, protein kinase B; EGCG, epigallocatechin gallate; FOXO1, Forkhead Box O1; JMJD1A, Jumonji domain-containing 1A; LIN28A, Lin-28 Homolog A; LPS, lipopolysaccharide; miR, microRNA; PAK2, p21 activated kinase 2; MMP, matrix metalloproteinase; PGC1α, peroxisome proliferator-activated receptor gamma coactivator 1α; PI3K, phosphatidyl inositol 3-kinase; PTEN, phosphatase and tensin homolog; SMAD, small mother against decapentaplegic; TGF, transforming growth factor.
Nutrients 16 00160 g003
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

Martino, E.; D’Onofrio, N.; Balestrieri, A.; Colloca, A.; Anastasio, C.; Sardu, C.; Marfella, R.; Campanile, G.; Balestrieri, M.L. Dietary Epigenetic Modulators: Unravelling the Still-Controversial Benefits of miRNAs in Nutrition and Disease. Nutrients 2024, 16, 160. https://doi.org/10.3390/nu16010160

AMA Style

Martino E, D’Onofrio N, Balestrieri A, Colloca A, Anastasio C, Sardu C, Marfella R, Campanile G, Balestrieri ML. Dietary Epigenetic Modulators: Unravelling the Still-Controversial Benefits of miRNAs in Nutrition and Disease. Nutrients. 2024; 16(1):160. https://doi.org/10.3390/nu16010160

Chicago/Turabian Style

Martino, Elisa, Nunzia D’Onofrio, Anna Balestrieri, Antonino Colloca, Camilla Anastasio, Celestino Sardu, Raffaele Marfella, Giuseppe Campanile, and Maria Luisa Balestrieri. 2024. "Dietary Epigenetic Modulators: Unravelling the Still-Controversial Benefits of miRNAs in Nutrition and Disease" Nutrients 16, no. 1: 160. https://doi.org/10.3390/nu16010160

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