Next Article in Journal
TZD-Based Hybrid Molecules Act as Dual Anti-Mycobacterium tuberculosis and Anti-Toxoplasma gondii Agents
Next Article in Special Issue
Effect of Genetic Factors, Age and Sex on Levels of Circulating Extracellular Vesicles and Platelets
Previous Article in Journal
Zirconia Nanoparticles as Reinforcing Agents for Contemporary Dental Luting Cements: Physicochemical Properties and Shear Bond Strength to Monolithic Zirconia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

An Overview of Inter-Tissue and Inter-Kingdom Communication Mediated by Extracellular Vesicles in the Regulation of Mammalian Metabolism

by
Carlos Castaño
1,2,
Anna Novials
1,2 and
Marcelina Párrizas
2,*
1
Pathogenesis and Prevention of Diabetes Group, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
2
Pathogenesis and Prevention of Diabetes Group, Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), 08036 Barcelona, Spain
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(3), 2071; https://doi.org/10.3390/ijms24032071
Submission received: 1 December 2022 / Revised: 13 January 2023 / Accepted: 16 January 2023 / Published: 20 January 2023
(This article belongs to the Special Issue Understanding Metabolic Cross-Talk in Health and Disease)

Abstract

:
Obesity and type 2 diabetes are associated with defects of insulin action in different tissues or alterations in β-cell secretory capacity that may be triggered by environmental challenges, inadequate lifestyle choices, or an underlying genetic predisposition. In addition, recent data shows that obesity may also be caused by perturbations of the gut microbiota, which then affect metabolic function and energy homeostasis in the host. Maintenance of metabolic homeostasis in complex organisms such as mammals requires organismal-level communication, including between the different organs and the gut microbiota. Extracellular vesicles (EVs) have been identified in all domains of life and have emerged as crucial players in inter-organ and inter-kingdom crosstalk. Interestingly, EVs found in edible vegetables or in milk have been shown to influence gut microbiota or tissue function in mammals. Moreover, there is a multidirectional crosstalk mediated by EVs derived from gut microbiota and body organs that has implications for host health. Untangling this complex signaling network may help implement novel therapies for the treatment of metabolic disease.

1. Introduction

The prevalence of non-communicable metabolic diseases is growing worldwide. Of these, obesity is the primary culprit, and it has been one of the major public health problems for decades, requiring novel nutritional or medical solutions [1]. Obesity rates are constantly on the rise, and it has been estimated that about 60% of the adult population will be overweight or obese by 2030 [1]. The global incidence of type 2 diabetes (T2D) is increasing in parallel [2], as it is closely related to obesity. Indeed, the term “diabesity” has been coined to refer to those cases when T2D is caused by obesity [3]. Moreover, obesity and T2D are associated with an increased risk of developing cardiovascular disease (CVD), mental disorders, cancer, and even infectious diseases such as COVID-19 [4].
To maintain metabolic homeostasis in complex organisms such as mammals, multiple organs and cell types must act in concert, and for that, they need to communicate with one another. When communication fails or is somehow disturbed, metabolic disease ensues. In the case of T2D, specific defects of insulin action or alterations in β-cell secretory capacity may underlie the development of the disease in different individuals. Indeed, extensive phenotyping of patients has consistently identified abdominal obesity, hepatic steatosis, or genetic alterations associated with deficient β-cell function as the key triggering defects [5,6,7]. Maladaptation of a tissue or cell type to environmental challenges may then influence other tissues through alterations of its secretory landscape, ultimately leading to impaired global metabolic homeostasis and masking the original malfunction [8]. Obesity induced by increased energy uptake and low physical activity is often associated with enlarged, hypertrophic white adipocytes that fail to adequately store the excess fat, leading to lipid spillover and ectopic accumulation in other tissues and decreasing insulin sensitivity [9]. Alterations of hepatic physiology caused by a diet enriched in processed food or high-fructose corn syrup can trigger hepatic steatosis that may progress to metabolic dysfunction-associated fatty liver disease (MAFLD), cirrhosis, or even hepatic carcinoma [10]. Exercise, alternatively, by increasing lean mass and basal metabolic rate, improves metabolic homeostasis [11]. In all these cases, the adipose tissue, the liver, or the muscle release humoral factors or extracellular vesicles (EVs) that may affect the function of neighboring and removed cells [8].
Importantly, in addition to lifestyle and genetic factors, obesity may also result from a perturbation of the gut microbiota, which then affects metabolic function and energy homeostasis in the host [12]. The gut microbiota is the set of microbes that colonize the gastrointestinal tract of mammals. In the case of humans, the gut microbiota is mainly composed of five different families: Bacteroidetes, Firmicutes, Actinobacteria, Proteobacteria, and Verrucomicrobia [13]. Remarkably, the gut microbiota establishes a mutualistic relationship with the host, participating in essential processes such as gut permeability, metabolism, and the physiology of the immune system [14]. Moreover, the gut microbiota can communicate with the central nervous system (SNC), participating in the regulation of hunger and appetite [15]. Environmental and endogenous factors such as dietary habits, aging, or antibiotic overuse may modify gut microbiota composition [16]. Alterations of gut microbiota homeostasis, referred to as dysbiosis, can promote overgrowth of pathogenic species, thereby playing a major role in the pathology of different diseases, including inflammatory bowel disease (IBD), CVD, neurological or autoimmune disorders, and colorectal cancer [17]. Recent studies have proposed that, whereas a healthy gut microbiota exerts a positive effect on health, dysbiosis may participate in the onset and progression of obesity and T2D [18,19]. In this sense, interventions on gut microbiota, including dietary interventions, bariatric surgery, or fecal transplantation, are postulated as novel, promising approaches to treating these metabolic disorders [20]. However, a better understanding of the mechanisms underlying the interaction between the gut microbiota and the host is necessary to identify the relevant targets.
Recently, EVs have emerged as crucial players in inter-organ and inter-kingdom crosstalk, including the communication established between the gut microbiota and its mammalian hosts [21]. Remarkably, EVs found in edible fruits and vegetables or in milk have also been shown to influence the gut microbiota or tissue function in humans and mice [21]. EVs are released by most cell types and have been identified in all domains of life. In mammals, depending on their biogenesis and size, EVs have been classified into exosomes, microvesicles (MV), and apoptotic bodies [22]. Exosomes have an endocytic origin and range in size from 30 to 150 nm (Figure 1). By contrast, MV are formed from plasma membrane evaginations and have a diameter ranging from 100 to 1000 nm. Finally, apoptotic bodies originate from cellular apoptosis, may contain cellular DNA and organelles, and range from 50 to 5000 nm. Plant cells secrete EVs as well, including membrane-derived MVs and vesicles with a biogenesis, content, and morphology similar to that of mammalian exosomes, usually called exosome-like nanovesicles (Figure 1) [23]. EVs have also been identified in bacteria. Depending on their origin, bacterial EVs (bEVs) from Gram-negative bacteria, which possess a thin peptidoglycan layer sandwiched between a double lipid membrane, may be outer-membrane vesicles (OMVs) or outer-inner membrane vesicles (OIMVs). In the case of Gram-positive bacteria, which possess a lipid membrane and an external peptidoglycan wall, bEVs are cytoplasmic-membrane vesicles (CMVs) [24] (Figure 1). Eukaryotic EVs and bEVs differ in factors such as biogenesis, composition, and tolerance to high temperatures. However, bEVs have many similarities with eukaryotic EVs in size, morphology, and stability. Importantly, a common characteristic among all EVs is the ability to carry and transfer bioactive molecules among cells. EV cargo is composed of a variety of bioactive molecules, including lipids, proteins, RNA, and DNA [22]. Among those, microRNAs (miRNAs) have raised special interest, as they can modulate gene expression in unrelated organisms, including plants–mammals, plants–bacteria, and mammals–bacteria [25]. Remarkably, miRNA-like molecules identified in bacteria have a similar size to eukaryotic miRNAs and can also be encapsulated and secreted in bEVs to protect them from degradation.
In this review, we will discuss the multidirectional crosstalk mediated by EVs derived from gut microbiota and the body organs in the context of obesity and related metabolic diseases. Moreover, we will examine the effects of EVs derived from food and probiotics/prebiotics on gut microbiota and the potential implications of this interaction on host health. Untangling this complex signaling network may help implement novel therapies for the treatment of metabolic diseases.

2. Inter-Organ Communication Mediated by EVs

2.1. EVs from the Adipose Tissues

2.1.1. White Adipocytes and Macrophages

Enlarged white adipose tissue (WAT) mass in obesity is often associated with tissue dysfunction manifested as an anomalous secretion pattern. Increased release of pro-inflammatory mediators and decreased secretion of adiponectin, for instance, are hallmarks of obesity [26]. However, the adipocytes and other cell types conforming to the WAT also release EVs that can be detected circulating in the blood. Although well-characterized tissue-specific EV markers are still lacking, vesicles enriched in proteins highly expressed in the adipose tissues, such as the transporter fatty acid binding protein (FABP) 4 or the hormone adiponectin, have been identified in human blood [27]. Moreover, adipose tissue, including both white and brown varieties, has been described as the main source of exosomal miRNAs circulating in the blood [28]. The strategy used to reach this conclusion, by deleting the miRNA processing enzyme DICER1 from mature adipocytes, did not modify the number of circulating blood exosomes, although it dramatically decreased the miRNAs associated with them. Interestingly, other authors overexpressed tdTomato fluorescent protein specifically in adipocytes, resulting in the release of fluorescent exosomes. The low ratio of tdTomato to the exosomal membrane protein CD63 in blood exosomes suggested that adipose exosomes represent a small fraction of those detected in blood [29]. These observations would be in accordance with another study concluding that the ratio of miRNAs to exosome numbers in blood is so low that probably only a small fraction of exosomes carry miRNAs [30]. Hence, adipose exosomes would be particularly enriched in miRNAs, even though they represent a low percentage of the vesicles circulating in the blood. Analysis of the RNA content of circulating EVs and correlation with RNA profiles from different tissues also suggested that the vast majority of EVs in blood come from hematopoietic cells [31], although EVs from peripheral tissues such as the liver could also be detected. Importantly, the hepatic EV fraction could still differentiate healthy patients from those with liver disease [31]. Similarly, many reports describe increased release of exosomes to the circulation by the obese adipose tissue [29]. Interestingly, knocking down adipocyte histone deacetylase SIRT1 results in obesity and increased exosome release by the adipocytes due to reduced autophagy, with the secreted exosomes then participating in setting off insulin resistance in both the adipose tissue and the liver [32].
Recently, extensive characterization of the protein and miRNA content of EVs derived from different cell lines and primary cells representing white and brown adipocytes, hepatocytes, skeletal muscle, and endothelial cells identified different molecular signatures for each cell type. Interestingly, tetraspanins CD9, CD63, and CD81, considered classical markers of EVs and of exosomes in particular and frequently used to assist in their isolation or detection, are not equally enriched in EVs from all cell types. CD63, for instance, is abundant in muscle EVs but is present only at low levels in hepatic, adipose, and endothelial EVs [33,34]. Importantly, the authors identified a set of proteins found in the EVs of all cell types examined, such as the enzymes glucose−6-phosphate isomerase (GPI) and enolase (ENO) 1 and the membrane protein colony-stimulating factor 1 receptor (CSF1R), which may prove to be of use in isolating and characterizing circulating EVs relevant to metabolic disease [33].
Many reports have focused on studying the characteristics and effects of EVs released by the different cell types that constitute the WAT, particularly adipocytes and macrophages [35,36,37,38], but also other cell types (Table 1). In this regard, it has been shown that the endothelial cells (ECs) release vesicles transporting plasma components to the adipocytes [39]. In return, ECs receive adipocyte-released EVs containing long non-coding RNA small nucleolar RNA host gene (SNHG) 9. In ECs, SNHG9 interacts with and silences TNF receptor type 1-associated death domain protein (TRADD) mRNA, reducing inflammation and apoptosis (Figure 2) [40]. In addition, adipocyte-secreted adiponectin accumulates in ECs and enhances EV release by these cells, thus decreasing intracellular ceramides and improving EC function [41]. Adipocyte-released EVs also transfer miR-221-3p to vascular smooth muscle cells (VSMCs) to induce remodeling during obesity [42] and can also influence neighboring adipocytes in a paracrine manner. Hence, transmission of miR-122 from mature adipocytes promotes adipogenesis of neighboring precursor cells by increasing the expression of sterol regulatory element binding transcription factor (SREBF) 1 [43], whereas vesicles from hypoxic adipocytes of obese subjects decrease glucose uptake of neighboring cells [44].
EVs also participate in crosstalk between adipocytes and macrophages, with vesicles released by obese adipocytes transporting neutral lipids to macrophages [29], blocking M2 polarization by transferring miRNAs such as miR-34 or miR-1224 [46,47], or inducing M1 polarization and foam cell formation [48]. In vitro, THP1 monocytes treated with anti-inflammatory interleukin (IL) 4 release exosomes that induce expression of miR-21 and other miRNAs in 3T3-L1 adipocytes, resulting in enhanced insulin-stimulated glucose uptake, whereas lipopolysaccharide (LPS)-activated THP1 cells increase pro-inflammatory gene expression in adipocytes [82,83]. In vivo, M2 polarized anti-inflammatory macrophages release exosomal miRNAs such as miR-690, which have insulin sensitizing properties, whereas pro-inflammatory M1 macrophages release miR-155 or miR-29a, which decrease insulin sensitivity [35,36,63]. Similar intra-organ communication between resident macrophages and tissue cells has been observed in other locations, such as the pancreatic islets, where M1 macrophages release exosomes enriched in miR-212 that target histone deacetylase SIRT2 and decrease insulin secretion in acceptor β-cells [64]. However, exosomes from WAT macrophages can also affect the pancreatic β-cells by transferring miR-155 and impairing insulin secretion while enhancing β-cell proliferation [65]. Similarly, exosomes from adipose tissue explants of healthy subjects or 3T3-L1 adipocytes induce proliferation of INS1 β-cells, whereas those from obese subjects induce cell death [84]. Moreover, serum exosomes from obese nondiabetic subjects carry low levels of omentin and inhibit β-cell proliferation without inducing apoptosis [85]. Direct communication between the WAT and the pancreas may then participate in the β-cell expansion and increased insulin secretion observed at the beginning of metabolic disease and the ultimate β-cell failure characteristic of advanced states of obesity and T2D.
In addition to pancreatic β-cells, adipocyte EVs can also affect other cells outside of the adipose depots, such as the muscle [38,49,52] or the liver [38,53,54,86,87]. Most reports show how EVs from aging or obese adipose tissue induce inflammation, insulin resistance, and toxicity in acceptor cells, contributing to disease spread. Transfer of exosomal miR-222 or miR-27a from obese adipocytes to muscle, for instance, reduces insulin sensitivity [38,49], whereas let-7d-3p, enriched in EVs from aged adipocytes, induces sarcopenia by decreasing muscular stem cell proliferation [52]. In addition, increased miR-199a and decreased miR-141-3p in vesicles from adipose tissue of obese mice fed a high-fat diet (HFD) induce hepatic steatosis and insulin resistance [53,54].
Adipose EVs can also affect heart function. Indeed, EVs from stressed adipocytes have been shown to carry mitochondria that increase antioxidant pathways in accepting cardiomyocytes [50]. Interestingly, most communication events take place locally. Thus, EVs from epicardial adipocytes have a protective effect after myocardial infarction by attenuating cardiac remodeling through transference of adipsin to cardiomyocytes [51], whereas EVs from epicardial adipocytes of atherosclerotic heart disease patients carry miR-3604 that blocks neuronatin expression in acceptor epicardial adipose stem cells to reduce adipogenesis [45]. Along similar lines, intramyocardial injection of vesicles isolated from the serum of diabetic patients exacerbates myocardial ischemia/reperfusion injury in the nondiabetic heart by transferring miR-130b-3p [88].
Remarkably, adipocyte-derived EVs may also mediate communication between WAT and the brain, promoting cognitive impairment by transferring miR-9-3p [55] or regulating hunger by communicating with hypothalamic neurons [89]. Furthermore, in obese mothers, adipocyte exosomes may affect fetal health [90]. Injection of EVs from the visceral WAT of obese mice into pregnant female mice resulted in increased inflammation in the placenta and fetal heart dysfunction [90].
Finally, exosomes from adipocytes also provide a link between obesity and cancer as they can modulate the function of tumor cells in breast cancer, prostate cancer, melanoma, or lung cancer, for instance [91,92,93,94,95]. By activating hypoxia-inducible factor (HIF) 1α, decreasing cyclin-dependent kinase inhibitor (CDKN) 2A activity, or transferring matrix metalloproteinase (MMP) 3 or fatty acid oxidation enzymes and substrates to tumor cells, EVs from obese adipose tissue enhance their malignancy, proliferation, and invasiveness.

2.1.2. Brown Adipocytes

Brown adipocytes differ from white adipocytes in their origin from common precursors to muscle cells [96,97]. However, brown-like adipocytes can also arise in WAT by either trans-differentiation of mature white adipocytes or brown differentiation of white adipocyte precursors, in a process known as beigeing or browning [98]. In contrast to white adipocytes, brown adipocytes contain multilocular lipid droplets and are rich in mitochondria, which express uncoupling protein (UCP) 1, thus contributing to energy expenditure and thermogenesis. By this ability and through their secretory capacity, brown adipose tissue (BAT) activation has been considered a potential strategy for the treatment of obesity [99,100]. Accordingly, EVs from BAT have been described as exerting beneficial effects. EVs released by brown adipocytes carry proteins such as nucleophosmin (NPM) 3, long non-coding RNAs, or mitochondrial components that contribute to WAT browning in vitro or in vivo (Figure 2) [56,101,102]. BAT exosomes also ameliorate diabetic kidney disease by transferring miR-30b and thus blocking pro-fibrotic transcription regulators runt-related transcription factor (RUNX) 1 and snail family zinc finger (SNAIL) 1 [57]. Interestingly, BAT ablation decreases the beneficial effects of exercise. Knockdown of the small GTPase RAB27A in intrascapular BAT, which decreased EV release by the tissue, attenuated the exercise-induced cardioprotective effects [103], thus highlighting the need for tissue crosstalk to respond to environmental challenges.

2.1.3. Mesenchymal Stem Cells

Mesenchymal stem cells (MSCs) are adult stem cells with significant anti-inflammatory and regenerative properties that have been used as cellular therapy for many ailments, including obesity and diabetes. Interestingly, EVs released by MSCs seem to reproduce some of the effects of MSCs but with a lower risk of tumorigenesis and offer the opportunity for cell-free therapy [22]. WAT is a frequently used source of MSCs due to its accessibility, and the effects of adipose MSC EVs on metabolic regulation have also been thoroughly examined. Adipose MSC EVs induce M2 macrophage polarization by transferring signal transducer and activator of transcription (STAT) 3 that then transactivates arginase (ARG) 1 [58], protect from osteoporosis by carrying osteoprotegerin (OPG), which blocks osteoclast differentiation [59], and decrease liver fibrosis by suppressing stellate cell activation [104]. Furthermore, MSC eVs protect from hepatic steatosis through the transference of miR-627 [105] or miR-223-3p [60]. EVs from MSCs also induce beneficial effects upon diabetic complications, including nephropathy, by protecting podocytes from apoptosis [106] and inflammation [107]. In addition, they can accelerate diabetic wound healing by transferring miR-21-3p or miR-126, both of which activate the phosphatidyl-inositol 3 kinase (PI3K) signaling pathway [61,62]. Similarly, MSCs derived from the human umbilical cord ameliorate experimental non-alcoholic steatohepatitis (NASH) in mice by targeting the nuclear factor erythroid 2-related factor (NRF) 2 pathway [108]. Moreover, human umbilical cord MSCs EVs increase glucose uptake by adipocytes in vitro [109] and reduce blood glucose in experimental diabetic rats by increasing insulin signaling in muscle, glycogen accumulation in the liver, and decreasing β-cell apoptosis [110].
Interestingly, by comparing the proteome of EVs released by MSCs obtained from either bone marrow or adipose tissue, it was observed that molecules present exclusively in EVs from adipose MSCs were highly correlated to angiogenesis, whereas those expressed in EVs from bone marrow were preferentially involved in cellular proliferation [111]. These data suggest that the cellular source of the EVs is of high importance in determining the ultimate effects of the therapy. Further stressing this notion, EVs of adipose MSCs from subjects with type 1 diabetes (T1D) are enlarged, reduced in number, and have a higher percentage of CD9 [112], and EVs from bone marrow MSCs obtained from aged mice contain high levels of miR-29-3p as compared with those isolated from younger mice and induce insulin resistance in acceptor young mice [113].

2.2. EVs from the Muscle

Like the adipose tissues, the skeletal muscle is a source of humoral factors, in this case called myokines, that get distributed throughout the body and participate in cell communication, helping maintain metabolic homeostasis and adapting to the enhanced metabolic demands of exercise [114,115]. The muscle also releases EVs [114], and the number and cargo of these vesicles change during and after exercise or in the context of obesity [37,116,117]. In vitro, mechanical strain enhances EV release by C2C12 cells [118]. Exercise-associated vesicles often accumulate in the liver [66,117], providing a means for coordinating tissue activity in a context of high energy requirements. We recently showed that implementation of high-intensity interval training (HIIT) in mice results in the release of exosomal miR-133b and other muscle-enriched miRNAs that then contribute to the improvement of metabolic homeostasis and insulin sensitivity induced by exercise by decreasing the expression of insulin-regulated transcription factor forkhead box (FOX) O1 in the liver (Figure 2) [66]. Similarly, other reports described the effects of muscle-derived EVs released during exercise in promoting WAT lipolysis by transferring miR-1 [67] or browning by decreasing miR-191a in EVs [68]. In HFD mice, exercise decreases miR-27a-3p in EVs from skeletal muscle, also contributing to WAT browning [119]. In contrast, EVs derived from atrophic muscle fibers of aged mice carry miR-690 that inhibits satellite cell differentiation [69], and atrophic insulin-resistant muscle from obese mice releases fewer EVs, which are enriched in proteins involved in lipid oxidation and miRNAs with nuclear location signals, such as miR-224, that target genes encoding proteins with nuclear activities in recipient adipocytes and neighboring muscle cells [37]. The quadriceps muscle of obese mice fed a palmitate-rich diet releases miR-16 in EVs that are captured by pancreatic β-cells, increasing proliferation [70] EVs released by the muscle have also been shown to communicate with the bone [120,121] or ECs [122].

2.3. EVs from the Liver

The liver is also a source of EVs that participate in metabolic regulation. Indeed, hepatic EVs carry miR-130a-3p that can be captured by adipocytes in vitro, where it targets the PH domain and leucine-rich repeat protein phosphatase (PHLPP) 2 [123], whose silencing has been shown to increase insulin sensitivity by favoring peroxisome proliferator-activated receptor (PPAR) α activity in adipocytes (Figure 2) [71]. Furthermore, injection of exosomes carrying miR-130a-3p into miR-130−/− mice correlates with increased insulin sensitivity [123]. Liver EVs also enhance glucose uptake by brown adipocytes by transferring the transmembrane protein TM4SF5 and improving insulin sensitivity [72]. Even hepatocyte-derived exosomes from early-onset obese mice have a protective effect and promote insulin sensitivity by transferring miR-3075 to acceptor cells in the muscle, the adipose tissue, or the liver itself [73].
However, in frank obesity or when liver disease is present, hepatic EVs contribute to further disturbing metabolism. Hence, hepatocytes treated with palmitic acid release EVs enriched in let−7b, whose overexpression enhances fatty acid transport by hepatocytes while reducing brown adipocyte thermogenesis. Interestingly, release of let−7b was regulated by the transforming growth factor (TGF) β pathway in hepatocytes [77], a key player in the development of liver disease [124], and it was absent in mice with hepatic-specific deletion of the receptor TGFBR2. These liver-specific knockout mice were also resistant to steatosis and obesity [77]. Hepatocyte EVs also carry ARG1 and contribute to the increase of arginase activity in the plasma of HFD mice [125,126], which is considered to initiate atherosclerosis.
One of the most characteristic hepatic-enriched miRNAs, miR-122, has been repeatedly shown to be increased in obese human and mouse plasma vesicles [76,127] and we have shown that it participates in the setting of the first steps of metabolic dysfunction associated with diabetes by targeting PPARα in WAT [76]. Similarly, miR-122 released by lipid-exposed hepatocytes is captured by macrophages, inducing expression of pro-inflammatory genes in a process requiring MMP2 [128]. Hepatic exosomal miR-122 also impairs cardiomyocyte function. Human plasma exosomes transport miR-122 into mouse primary cardiomyocytes and impair mitochondrial ATP production and oxygen consumption. In obese mice, increased hepatic and circulating exosomal miR-122 also inhibited cardiac mitochondrial function [74]. Alternatively, a recent report showed that exercise induces the release of exosomal miR-122 by the liver, which is required for the ECs of the skeletal muscle to initiate angiogenesis. EV miR-122 enhanced EC fatty acid utilization by targeting 1-acyl-sn-glycerol−3-phosphate acyltransferase (AGPAT) 1, hence favoring angiogenesis [75]. These data indicate that the effects of a miRNA often depend on context and highlight the need for limiting delivery specifically to target cells when using a miRNA mimic or inhibitor as therapy to avoid unwanted secondary effects.
Hepatic nonparenchymal cells also communicate by releasing EVs. Neutrophil EVs are enriched in anti-inflammatory miR-223-3p, which is transferred to hepatocytes and limits the progression of NASH. Interestingly, the presence of apolipoprotein (APO) E on neutrophil-derived EVs facilitated direct delivery to the hepatocytes by binding to the low-density lipoprotein receptor (LDLR) [78]. Conversely, EVs from the hepatic stellate cells carry the glucose transporter GLUT1 and the glycolytic enzyme pyruvate kinase (PK) M2 that induce a metabolic switch in neighboring quiescent stellate cells, resulting in the advancement of fibrosis [129].

2.4. EVs from Pancreatic β-Cells

The pancreatic islets are crucial regulators of metabolic homeostasis through the controlled release of insulin by β-cells or glucagon by α-cells, among other hormones and bioactive peptides. However, islet cells can also exert autocrine and endocrine effects through the release of EVs [130]. Hence, EV miR-26a released by β-cells targets regulators of insulin release and cell proliferation in the β-cell itself but also modulates peripheral insulin sensitivity (Figure 2) [79]. Exosomes from the β-cell line MIN6 injected into streptozotocin-induced diabetic mice improve the survival and glucose tolerance of the treated mice, as well as pancreas architecture and insulin content [131]. Pancreatic EVs from healthy subjects reduce amyloid formation in the pancreas, a hallmark of diabetes [132], but conversely, EVs containing islet amyloid polypeptide (IAPP) can reach hippocampal cells, inducing a pro-fission status of the mitochondrial network and providing a link between diabetes and neurodegenerative disease [80]
Rodent and human T lymphocytes release EVs with pro-inflammatory miR-155 among other miRNAs that induce β-cell apoptosis and may participate in the development of T1D [81]. Alternatively, cytokine treatment of β-cell lines induces miR-21 release in EVs, and serum miR-21 is increased in NOD1 mice and T1D children as the disease progresses [133]. Furthermore, EVs from β-cells treated with cytokines promote a pro-inflammatory islet transcriptome in neighboring cells and may contribute to β-cell failure in diabetes [134].
Finally, tumor cells hijack organismal glucose and lipid metabolism to thrive; thus, it is not surprising that they can affect β-cell function by releasing exosomes carrying miR-122 that targets PKM2 and decreases glycolysis, hence decreasing ATP-stimulated insulin release and impairing glucose metabolism to favor tumor growth [135].

3. Inter-Kingdom Communication Mediated by EVs

3.1. Environmental Factors Shaping Gut Microbiota and bEVs

The gut microbiota participates in the digestion of carbohydrates, lipids, and essential amino acids and plays a role in metabolism through the production of short-chain fatty acids (SCFAs) by colonic fermentation of dietary fiber and proteins. The beneficial effects of SCFAs on body weight, the regulation of satiety, glucose and lipid homeostasis, insulin sensitivity, and the maintenance of intestinal barrier strength have been well documented [136]. Moreover, other metabolites produced by the gut microbiota also reach the circulation of the host and have been shown to regulate gene expression in distant tissues, such as tryptophan-derived metabolites that control the expression of the miR-181 family in WAT, in this way regulating energy expenditure and insulin sensitivity in mice [137]. Additionally, gut microbiota increase energy production from food, provide low-grade inflammation, and impact adipose tissue composition, hence playing a critical role in the development of obesity. Multiple factors such as diet, physical activity, medication, or bariatric surgery may affect the composition of the gut microbiota. Factors decreasing diversity lead to dysbiosis and, consequently, weight gain and obesity. Conversely, increased diversity favors a healthy metabolism (Figure 3).
Diet exerts a powerful influence on the composition, abundance, diversity, and metabolism of gut microbiota [138]. Promotion of a specific gut microbiota composition profile has been established for a wide range of diets [139]. In mice, HFD and high-sucrose (HSD) diets have different effects on gut microbiota. Some favorable bacteria are reduced in HFD, whereas more obesity-related bacteria are present after HSD feeding, thus suggesting that dietary sucrose impacts obesity to a greater extent than dietary fat [140]. In humans, vegetarian and Mediterranean diets prevent obesity by increasing diversity, particularly the abundance of beneficial bacteria, while decreasing inflammation-related bacteria [141,142]. High-quality whole grain consumption also reduces weight gain [143]. In fact, African children, who consume a high-fiber/low-fat diet, have higher microbial diversity and fewer pathogenic bacteria, with larger amounts of Bacteroidetes than European children, who have higher amounts of Firmicutes. Actually, obese subjects have a higher Firmicutes/Bacteroidetes ratio than individuals of normal weight, and this ratio correlates negatively with energy expenditure. Interestingly, there is a negative correlation between circulating miR-122, which, as mentioned, is usually increased in obese and diabetic patients, and the presence of Bacteriodes uniformis [144]. In contrast, a low-fiber/high-fat diet reduces microbial diversity and enhances gut inflammation, thus evidencing that the westernization of dietary habits induces microbial dysbiosis [145,146]. Therefore, gut microbiota shaping through dietary interventions could be an attractive, effective, and non-invasive strategy to prevent or treat obesity and diabetes.
Alternatively, dietary supplements that can prevent weight gain act at least in part by improving lipid and glucose metabolism through modifying gut microbiota composition. Both the antioxidant derivative of carotenoids astaxanthin and the medicinal mushroom Antrodia cinnamomea optimize the Firmicutes/Bacteroidetes ratio and increase Akkermansia muciniphila in obese mice [147]. Interestingly, direct transfer of B. thetaiotaomicron to mice fed a normal diet results in a reduction in total fat and also prevents weight gain in obese mice. Furthermore, the Akkermansia phylum counteracts inflammation and improves insulin sensitivity. The remarkable protective effect of A. muciniphila on metabolism is also reported in human studies, where it has been shown that it is able to reduce intestinal permeability, reinforce the immune system, and improve glucose homeostasis [148].
Remarkably, aerobic exercise also enhances bacterial diversity, mainly through SCFA producers. Obese adults following aerobic moderate-to-intense physical activity decrease the Firmicutes/Bacteroidetes ratio [149]. Furthermore, both metformin and bariatric surgery increase Bacteroides species [150,151,152,153]. Conversely, the overuse of antibiotics induces dysbiosis and correlates with an increased risk of developing inflammatory disorders and obesity [154].
Human gut microbiota continually produces bEVs that can enter the systemic circulation and elicit a variety of responses in the host, thus evidencing the role of bEVs in microbe-host cross-kingdom communication [155]. In this sense, bEVs may participate in immune system regulation, modulation of cerebral function, or the metabolism of peripheral tissues (Figure 3). Importantly, diet and other environmental factors influence the bEV profile and its effects on host physiology, thus contributing to the development or alleviation of metabolic disorders. For instance, intestinal succinate produced after administration of a high-protein diet (HPD) induces reactive oxygen species (ROS) production by the gut microbiota that enhances the release of bEVs in mice [156]. HFD mice also have an altered content of bEVs in their stool, showing a reduction in size and changes in the global protein content, marked by an increment in LPS due to the increase in bEVs derived from LPS-expressing Proteobacteria [157]. This higher percentage of LPS-containing bEVs could potentially be an important underlying mechanism orchestrating the metabolic disorders induced by HFD [158].

3.2. bEVs and Their Impact on Inflammation and Metabolism

Importantly, bEVs from beneficial species have a protective role against inflammatory responses (Table 2). A. muciniphila bEVs, in particular, dampen the secretion of pro-inflammatory cytokines induced by pathogenic Escherichia coli bEVs on a colon epithelial cell line, thus suggesting a potential role for A. muciniphila bEVs in regulating intestinal barrier permeability [159]. Similarly, Odoribacter splanchnicus 57 bEVs possess immunomodulatory properties, mitigating the production of pro-inflammatory cytokines in enterocytes [160]. E. coli Nissle (EcN) 1917 and ECOR63 bEVs also protect the barrier function in human intestinal epithelial cell cultures [161]. The role of bEVs in modulating intestinal inflammation and gut barrier integrity has been further confirmed by several in vivo studies. For instance, oral administration of A. muciniphila, EcN, or B. fragilis bEVs attenuates the severity of colitis in mice [162,163,164]. Interestingly, bEVs not only exert immunomodulatory functions through their interaction with gut barrier cells but also communicate with intestinal immune system cells, thus influencing host immunity. In this regard, Pediococcus pentosaceus bEVs trigger immuno-suppressive responses in bone marrow-derived macrophages [165]. It should be highlighted that the induction of a moderate inflammatory bowel response is critical for gut barrier protection. In fact, bEVs with mild pro-inflammatory properties could be beneficial and contribute to gut barrier integrity and intestinal homeostasis. In this sense, E. coli C25 bEVs promote interleukin secretion by intestinal epithelial cells [166], and EcN and ECOR12 bEVs enhance the secretion of pro-inflammatory cytokines in human epithelial colorectal adenocarcinoma (Caco−2) cells, thus suggesting a role for LPS-carrying bEVs in the modulation of intestinal immune responses [167]. However, in the context of dysbiosis, increased pro-inflammatory bEVs may exacerbate inflammatory responses. Hence, Faecalibacterium prausnitzii bEVs cause gut barrier permeability and alter epithelial cells’ metabolic functions through the regulation of tight junctions and the expression of PPAR genes, as observed in Caco−2 cells [168]. This stronger pro-inflammatory response may ultimately harm the host, as in the case of colitis development in genetically susceptible mice that receive B. thetaiotaomicron bEVs [169].
Notably, bEVs also help connect intestinal immune responses and gut barrier integrity with host metabolic functions. Oral administration of A. muciniphila bEVs to HFD mice increases gut barrier integrity and ameliorates intestinal and adipose tissue inflammation, but also reduces food intake, body weight, and adiposity and improves glucose tolerance [170,171,172]. These data indicate that A. muciniphila bEVs could be a therapeutic tool to treat metabolic diseases such as obesity and diabetes [183]. Alternatively, changes in bEV profile and cargo occurring in dysbiosis can contribute to the onset and progression of metabolic diseases [184]. Remarkably, the alteration of the bEV profile is more severe than changes in gut microbiota composition. In a context of obesity-induced gut barrier permeability, bEVs and other microbiota-derived products can enter host circulation and reach insulin-sensitive tissues, evidencing the potential of bEVs as biomarkers for intestinal permeability [185]. Actually, accumulation of microbial DNA carried by bEVs in diverse organs of the host may lead to disease, such as adrenomedullary abnormalities and hypertension [177], liver steatosis and fibrosis [178], and development of obesity-associated tissue inflammation and insulin resistance in mice [179]. Accumulation of DNA-containing bEVs also promotes islet inflammation and β-cell abnormalities in obese mice [180]. Pseudomonas panacis LPS-containing bEVs, which are highly increased in the stool of HFD mice, blunt glucose metabolism after administration to mice by promoting insulin resistance in both skeletal muscle and adipose tissue [157]. Moreover, Porphyromonas gingivalis bEVs decrease hepatic glycogen synthesis in response to insulin, thus eliciting changes in glucose metabolism in the liver and contributing to the progression of T2D in mice [174]. Remarkably, bEVs can also contain miRNA-like molecules that have been shown to exert trans-kingdom gene expression regulation, in this way influencing host biological functions. For instance, bEV miRNAs of periodontal pathogens can regulate host immune system response by downregulating cytokine expression in Jurkat T cells [175], and Aggregatibacter actinomycetemcomitans bEV miRNAs promote human macrophage TNFα production and can even cross the blood-brain barrier in mice, thus evidencing a potential role of bEV miRNAs in neuroinflammatory disorders [176]. Similarly, Paenalcaligenes hominis bEVs can also enter the brain and cause cognitive function-impaired disorders [173]. Further studies are needed to understand the systemic functions of circulating bEVs, to explore their potential as biomarkers, and to identify and correlate their taxonomy to the metabolic status of gut microbiota.

Properties of bEVs Derived from Probiotics

Probiotics and prebiotics improve the composition of gut microbiota and are postulated as promising tools for the prevention and treatment of obesity [186] (Figure 3). Probiotics consist of live bacteria, typically Lactobacilli and Bifidobacteria, that promote the health of the host. In particular, probiotics are immuno-modulatory and have beneficial effects such as improved skin health and alleviation of IBD. Moreover, probiotics improve gut permeability and promote fat loss. In fact, obese mice supplemented with Lactobacillus plantarum showed a reduction in adipose deposition and enhanced lipid oxidation [187]. Recently, O. laneus was proposed as a novel probiotic able to decrease the levels of systemic succinate, which correlate with inflammation and are linked to numerous complications in obese mice [188]. Probiotics also favor microbial diversity, expanding the beneficial bacteria and reducing certain harmful species and their metabolites. In this sense, probiotics increase SCFA-producers and reduce LPS-producing bacteria. Accordingly, HFD rats supplemented with probiotics decreased weight gain and displayed positive effects on insulin and fasting blood glucose, inflammatory markers, and adipokine levels [189].
Notably, probiotic bEVs also possess anti-inflammatory properties and could be potentially used to treat inflammatory disorders, including diabetes [190,191]. For instance, L. paracasei bEVs counteract LPS-induced human colorectal cell inflammation and attenuate colitis in mice [181]. Moreover, L. casei bEVs participate in the crosstalk between gut microbiota and the modulation of the immune system of the host [182]. Both L. casei and L. plantarum bEVs’ anti-inflammatory properties can be strengthened after macrophage treatment in vitro [192]. This finding provides a new avenue to analyze if specific food ingredients could enhance probiotic bEV immunomodulatory properties in a similar manner as culture conditions and be used to effectively treat metabolic diseases.
Alternatively, prebiotics are non-metabolized food ingredients that reach the intestinal lumen and can be selectively utilized by beneficial bacteria in the host. Inulin, galacto-oligosaccharides (GOS), and fructo-oligosaccharides (FOS) are the most frequently used prebiotic supplements. FOS, for instance, increases the number of Bifidobacterium and Lactobacillus species, as well as other SCFA producers, reinforcing the gut barrier against pathogens and leading to improvements in metabolic parameters and a decline in food intake [193]. Leptin resistance is also improved by a single dose of prebiotics, but clinical trials investigating their use in the treatment of obesity are still underway. Interestingly, cocoa flavanols, dark chocolate, and lycopene have a systemic effect on metabolism mediated by the gut microbiota [194]. However, the full potential of prebiotics for weight control and metabolic regulation is still unknown.

3.3. Food-Derived EVs as Potential Therapeutic Agents

As observed for the bEVs derived from probiotics, dietary EVs can participate in the interaction between diet and gut microbiota, thus suggesting that EVs might be involved in the previously described effect of diet on host physiology. Additionally, dietary EVs can directly interact with mammalian cells, such as epithelial cells, immune cells, tumor cells, or hepatocytes, thus mediating biological functions and ultimately impacting on host health (Figure 4) [195,196]. EVs from plants and food are absorbed daily through the intestine. Importantly, EVs have been shown to survive the acidic environment of the digestive tract because of the protection afforded by the phospholipid bilayer [197]. The miRNAs in milk-derived exosomes are still detected after RNase, freeze-thawing, and acidic treatments [198], and curcumin encapsulated in exosomes is four times more stable than free curcumin [199]. The uptake of milk-derived exosomes by intestinal epithelial cells (IECs) is mediated by endocytosis and depends on cell and exosome surface glycoproteins in human and rat intestinal cells [200]. Moreover, EVs from food also cross the endothelial cell barrier of blood vessels and reach distal organs, thus influencing the systemic condition of the host. Oral administration of fluorescently labeled milk exosomes in mice showed accumulation in the liver [201,202].
Recent characterizations of EVs from food show that they might contain pharmacologically active molecules. Therefore, food EVs are attracting increasing interest due to their relevance in modulating cellular processes as well as their potential as therapeutic vehicles for the treatment of a variety of diseases, such as inflammatory disorders, hepatic steatosis, and cancer. For instance, extracellular vesicles isolated from homogenized ginger rhizome roots using a sucrose gradient centrifugation method [203], named ginger-derived nanoparticles (GDNs), were shown to contain large amounts of ginger bioactive constituents (6-gingerol and 6-shogaol). These GDNs were taken up by IECs and macrophages [204]. Oral administration of GDNs naturally containing shogaol protects mice from alcohol-induced liver damage by activating NRF2 [203].
In support of this concept, dietary bovine milk exosomes modulate gut microbiota composition, especially increasing SCFA levels [205]. One of the main consequences of this modulation in mice is the maintenance of intestinal barrier function and the regulation of the intestinal immune system. Actually, maternal breast milk contains abundant exosomes that may function as signalosomes, inducing β-cell proliferation shortly after birth, whereas weaning and stopping the exposure to milk exosomes may represent the key signal for β-cell maturation [206]. However, continued consumption of bovine milk by Western societies, and hence continued exposure to milk exosomes, may have harmful effects on human health since other studies suggest that they also participate in the pathogenesis of metabolic diseases [207]. This is because the most abundant miRNAs in bovine milk exosomes, including miR-21 and miR-148a, disturb energy homeostasis by influencing adipogenesis and insulin secretion, hence adding a new perspective to the pathogenesis of T2D [206]. Alternatively, exosomes from the breast milk of mothers with a metabolic disease may impact infant health. Milk exosomes from T1D mothers are enriched in immune-modulating miRNAs, whose transference to the infant could lead to overactivation of the immune system [208]. Similarly, maternal obesity and gestational diabetes are associated with alterations in the content of specific exosomal miRNAs in human milk that may increase the adiposity of the infant [209,210].
All these data show the potential of using milk exosomes as a scalable, biocompatible, and cost-effective delivery system to enhance the efficacy of therapeutic miRNAs or other drugs that are not orally bioavailable. Milk-derived exosomes loaded with miR-31 have been tested as a novel system to promote angiogenesis, thus accelerating diabetic wound healing, one of the major health problems worldwide [211]. Most recently, milk EVs carrying the antidiabetic drug Liraglutide have been developed as the next-generation drug delivery platform for the treatment of diabetes [212].
Dietary plant EVs can also modify intestinal communication with distal tissues, including the placenta. Watermelon EVs affect the trophoblast proteome and key aspects of trophoblast behavior, including migration, thus recommending a maternal diet high in fruit and vegetables as it lowers the risk of fetal growth restriction [213]. In addition, dietary plant EVs influence human health by modulating gut microbiota and intestinal barrier function [214]. Moreover, EVs from oranges could be used for the treatment of intestinal complications associated with diet-induced obesity in mice [215]. Similarly, ginger EVs show therapeutic promise. In particular, GDNs block NLRP3 inflammasome assembly and activation, thus emerging as new potent agents to treat the pathogenesis of many diseases, including Alzheimer’s disease and T2D [216]. Recently, GDNs have been shown to regulate the intestinal immune system and improve gut barrier integrity through miRNA delivery in mice [217]. Moreover, manipulation of gut commensal L. rhamnosus through dietary GDNs may be a promising strategy to treat metabolic syndrome, and oral administration of GDNs enriched in miR-375 to HFD mice improves host glucose tolerance and insulin response [218].
There is a growing interest in therapeutically targeting the inflammatory response that underlies age-related chronic diseases, including obesity and T2D, as well as other pathologies, such as autoimmune diseases and neurodegenerative disorders, which have a serious impact on the lives of patients. Interestingly, celery exosomes suppress activated T lymphocytes in a dose-dependent manner, and miR-159a and miR-156c found in nut exosome-like nanoparticles suppress inflammation. These data suggest that plant EVs can be used as immunosuppressants to treat a variety of ailments [219,220].

4. Open Questions and Perspectives

4.1. Current Limitations in EV Characterization

All the reports that we have summarized point to a key role for EVs of different origins—plant, bacterial, and mammalian—in the maintenance of metabolic homeostasis and the etiopathology of metabolic diseases. Hence, EVs and bEVs display great potential as biomarkers to diagnose or monitor disease progress, therapeutic targets to alleviate many illnesses, and innovative therapeutic vectors to increase the bioavailability of different drugs [221,222]. However, standard techniques for the isolation of bacterial and eukaryotic vesicles from different types of samples should be well established before EVs reach the clinical setting as either biomarkers or therapeutic agents. Current methodologies for EV isolation do not discriminate between the different types of vesicles, such as MVs or exosomes, as no specific markers exist. Similarly, although some steps have been taken in that direction, there is a need to find strong tissue-specific markers that identify the source of EVs circulating in the blood. Selective isolation of EVs coming from metabolically relevant tissues such as the liver or pancreatic β-cells would provide us with more precise biomarkers than interrogating the whole pool of EVs circulating in blood.
Alternatively, bEVs from gut microbiota are frequently characterized from stool samples. Some authors identify the bacterial origin of fecal bEVs by amplifying DNA from the most common gut microbiota phyla. However, exhaustive characterization is still a challenge due to the complexity of the material. Whereas the characterization of bEVs from culture medium is relatively straightforward, the isolation and characterization of bEVs from the corporal fluids of the host are far more complex. The major obstacle is the lack of bEV universal markers and their similar size to mammalian EVs. Nevertheless, new methods are currently being developed to optimize bEV isolation from body fluids [223]. Additionally, the identification of bioactive molecules in bEVs from gut microbiota is still in its infancy and has been mainly focused on the analysis of the proteome [224]. However, as we have mentioned, bEVs also carry miRNA-like molecules that could potentially regulate gene expression in mammalian cells. Hence, a deeper knowledge of bEVs may provide a novel and unexpected insight into the pathobiology of diabetes.

4.2. Perspectives

Aside from optimizing and standardizing methods for EV isolation from different tissues, the gut microbiota, or food ingredients, extensive characterization of their cargo should be performed before using them as therapeutic agents or drug delivery vectors in humans. We and others have used EVs of different origins as carriers of artificial genetic material, miRNAs or siRNAs, for the treatment of obesity and diabetes in rodent models [22,66,225], but as mentioned, EVs from MSCs of different sources have different endogenous properties and induce differential effects on acceptor cells [111]. Hence, exhaustive characterization of EVs with potential therapeutic use would be key to optimizing their effects. Other authors have used food-derived EVs as carriers to deliver bioactive molecules to tissues [212,226], with the advantage that they can be administered orally; however, stronger evidence is still needed to ascertain what portion of EVs from food reach the circulation and directly affect the function of internal organs.
Of the miRNAs mentioned in the text, miR-122 is the most studied. miR-122 is a liver-enriched miRNA that participates in the regulation of cholesterol metabolism, and its circulating levels have been shown to positively correlate with liver injury and different features of glucose metabolism and the metabolic syndrome [227]. As mentioned, it also negatively correlates with the Firmicutes/Bacteroidetes ratio. Most of the studies point to a detrimental role of miR-122 overexpression in metabolism (Table 1), and we and others have shown the beneficial effects of inhibiting it [225,228]. Similarly, miR-155 is a pro-inflammatory miRNA that has been shown to be released from lymphocytes or macrophages and may participate in the development of insulin resistance in both type 1 and type 2 diabetes [229]. Alternatively, miR-223-3p has an anti-inflammatory function and participates in the regulation of lipid and cholesterol homeostasis, delays atherosclerotic plate formation, and decreases steatosis (Table 1) [230,231,232,233]. We recently showed that EV-miR-223-3p is significantly decreased in prediabetic subjects that go on to develop T2D in the next 4 years [234].
Biodistribution studies are also required to identify membrane molecules targeting delivery to specific cell types. This information is needed to take full advantage of EVs’ ability to deliver bioactive molecules and drugs, thereby decreasing unwanted secondary effects. Modification of the cells used to produce the EVs by overexpressing specific membrane proteins shows promise in this regard [235].
Additionally, we still need to better understand the mechanisms by which gut microbiota and the cells of the host communicate with each other and collaborate in maintaining energy homeostasis. Thus, more evidence is needed to know if bEVs with bioactive cargo directly affect the function of mammalian cells in the host, aside from inducing inflammation, and conversely, if eukaryotic EVs from the host are able to modulate the composition of the gut microbiota. In this respect, EVs from intestinal epithelial cells have been shown to carry miRNAs that can influence bacterial gene expression, thus controlling gut microbiota composition and homeostasis [236]. However, a deeper knowledge of inter-kingdom crosstalk would show if more distant organs of the host, such as the skeletal muscle or the liver, are also able to communicate with the gut microbiota via EVs. This would give us a mechanism by which lifestyle interventions or environmental factors other than food, such as exercise, fasting, or even depressive moods, may affect gut microbiota composition.

5. Conclusions

EVs can be found in all domains of life and act as complex signalosomes that mediate communication between the different tissues and kingdoms to coordinate cellular functions in order to maintain metabolic homeostasis in the host. Alterations of prokaryotic or eukaryotic EVs may lead to metabolic imbalance and disease, whereas their modulation could be used as a novel therapeutic strategy for the alleviation and treatment of metabolic diseases.

Author Contributions

Conceptualization and writing, C.C. and M.P. Writing—review and editing, C.C., A.N. and M.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by EFSD/Boehringer Ingelheim 2020, awarded to M.P., and Grant SED for Basic Research in Diabetes 2020, awarded to C.C.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Illustrations were created with images from Servier Medical Art, provided by Servier, licensed under a Creative Commons Attribution 3.0 unported license.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Chooi, Y.C.; Ding, C.; Magkos, F. The Epidemiology of Obesity. Metabolism 2019, 92, 6–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Khan, M.A.B.; Hashim, M.J.; King, J.K.; Govender, R.D.; Mustafa, H.; Kaabi, J. Epidemiology of Type 2 Diabetes–Global Burden of Disease and Forecasted Trends. J. Epidemiol. Glob. Health. 2020, 10, 107–111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Ortega, M.A.; Fraile-Martínez, O.; Naya, I.; García-Honduvilla, N.; Álvarez-Mon, M.; Buján, J.; Asúnsolo, Á.; de la Torre, B. Type 2 Diabetes Mellitus Associated with Obesity (Diabesity). The Central Role of Gut Microbiota and Its Translational Applications. Nutrients 2020, 12, 2749. [Google Scholar] [CrossRef] [PubMed]
  4. Holly, J.M.P.; Biernacka, K.; Maskell, N.; Perks, C.M. Obesity, Diabetes and COVID-19: An Infectious Disease Spreading From the East Collides With the Consequences of an Unhealthy Western Lifestyle. Front. Endocrinol. 2020, 11, 582870. [Google Scholar] [CrossRef] [PubMed]
  5. Wagner, R.; Heni, M.; Tabak, A.G.; Machann, J.; Schick, F.; Randrianarisoa, E.; Hrabě de Angelis, M.; Birkenfeld, A.L.; Stefan, N.; Peter, A.; et al. Pathophysiology-Based Subphenotyping of Individuals at Elevated Risk for Type 2 Diabetes. Nat. Med. 2021, 27, 49–57. [Google Scholar] [CrossRef]
  6. Udler, M.S.; Kim, J.; von Grotthuss, M.; Bonas-Guarch, S.; Cole, J.B.; Chiou, J.; Boehnke, M.; Laakso, M.; Atzmon, G.; Glaser, B.; et al. Type 2 Diabetes Genetic Loci Informed by Multi-Trait Associations Point to Disease Mechanisms and Subtypes: A Soft Clustering Analysis. PLoS Med. 2018, 15, e1002654. [Google Scholar] [CrossRef] [Green Version]
  7. Ahlqvist, E.; Storm, P.; Käräjämäki, A.; Martinell, M.; Dorkhan, M.; Carlsson, A.; Vikman, P.; Prasad, R.B.; Aly, D.M.; Almgren, P.; et al. Novel Subgroups of Adult-Onset Diabetes and Their Association with Outcomes: A Data-Driven Cluster Analysis of Six Variables. Lancet Diabetes Endocrinol. 2018, 6, 361–369. [Google Scholar] [CrossRef] [Green Version]
  8. Priest, C.; Tontonoz, P. Inter-Organ Cross-Talk in Metabolic Syndrome. Nat. Metab. 2019, 1, 1177–1188. [Google Scholar] [CrossRef]
  9. Bays, H.E.; González-Campoy, J.M.; Bray, G.A.; Kitabchi, A.E.; Bergman, D.A.; Schorr, A.B.; Rodbard, H.W.; Henry, R.R. Pathogenic Potential of Adipose Tissue and Metabolic Consequences of Adipocyte Hypertrophy and Increased Visceral Adiposity. Expert Rev. Cardiovasc. Ther. 2008, 6, 343–368. [Google Scholar] [CrossRef] [Green Version]
  10. Samuel, V.T.; Shulman, G.I. Nonalcoholic Fatty Liver Disease as a Nexus of Metabolic and Hepatic Diseases. Cell Metab. 2017, 27, 22–41. [Google Scholar] [CrossRef]
  11. Murphy, R.M.; Watt, M.J.; Febbraio, M.A. Metabolic Communication during Exercise. Nat. Metab. 2020, 2, 805–816. [Google Scholar] [CrossRef] [PubMed]
  12. Guirro, M.; Costa, A.; Gual-Grau, A.; Herrero, P.; Torrell, H.; Canela, N.; Arola, L. Effects from Diet-Induced Gut Microbiota Dysbiosis and Obesity Can Be Ameliorated by Fecal Microbiota Transplantation: A Multiomics Approach. PLoS ONE 2019, 14, e0218143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Qin, J.; Li, R.; Raes, J.; Arumugam, M.; Solvsten Burgdorf, K.; Manichanh, C.; Nielsen, T.; Pons, N.; Levenez, F.; Yamada, T.; et al. A Human Gut Microbial Gene Catalogue Established by Metagenomic Sequencing. Nature 2010, 464, 59–65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Gentile, C.L.; Weir, T.L. The Gut Microbiota at the Intersection of Diet and Human Health. Science 2018, 362, 776–780. [Google Scholar] [CrossRef] [Green Version]
  15. Lankelma, J.M.; Nieuwdorp, M.; de Vos, W.M.; Wiersinga, W.J. The Gut Microbiota in Internal Medicine: Implications for Health and Disease. Neth. J. Med. 2015, 73, 61–68. [Google Scholar] [PubMed]
  16. Cuevas-Sierra, A.; Ramos-Lopez, O.; Riezu-Boj, J.I.; Milagro, F.I.; Martinez, J.A. Diet, Gut Microbiota, and Obesity: Links with Host Genetics and Epigenetics and Potential Applications. Adv. Nutr. 2019, 10, S17–S30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Durack, J.; Lynch, S.V. The Gut Microbiome: Relationships with Disease and Opportunities for Therapy. J. Exp. Med. 2019, 216, 20–40. [Google Scholar] [CrossRef] [Green Version]
  18. Sharma, S.; Tripathi, P. Gut Microbiome and Type 2 Diabetes: Where We Are and Where to Go. J. Nutr. Biochem. 2019, 63, 101–108. [Google Scholar] [CrossRef]
  19. Valdes, A.M.; Walter, J.; Segal, E.; Spector, T.D. Role of the Gut Microbiota in Nutrition and Health. BMJ 2018, 361, 36–44. [Google Scholar] [CrossRef] [Green Version]
  20. Wang, H.; Lu, Y.; Yan, Y.; Tian, S.; Zheng, D.; Leng, D.; Wang, C.; Jiao, J.; Wang, Z.; Bai, Y. Promising Treatment for Type 2 Diabetes: Fecal Microbiota Transplantation Reverses Insulin Resistance and Impaired Islets. Front. Cell. Infect. Microbiol. 2020, 9, 455. [Google Scholar] [CrossRef]
  21. Woith, E.; Fuhrmann, G.; Melzig, M.F. Extracellular Vesicles-Connecting Kingdoms. Int. J. Mol. Sci. 2019, 20, 5695. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Castaño, C.; Novials, A.; Párrizas, M. Exosomes and Diabetes. Diabetes Metab. Res. Rev. 2019, 35, e3107. [Google Scholar] [CrossRef]
  23. 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]
  24. Toyofuku, M.; Nomura, N.; Eberl, L. Types and Origins of Bacterial Membrane Vesicles. Nat. Rev. Microbiol. 2019, 17, 13–24. [Google Scholar] [CrossRef] [PubMed]
  25. Layton, E.; Fairhurst, A.M.; Griffiths-Jones, S.; Grencis, R.K.; Roberts, I.S. Regulatory RNAs: A Universal Language for Inter-Domain Communication. Int. J. Mol. Sci. 2020, 21, 8919. [Google Scholar] [CrossRef] [PubMed]
  26. Blüher, M. Obesity: Global Epidemiology and Pathogenesis. Nat. Rev. Endocrinol. 2019, 15, 288–298. [Google Scholar] [CrossRef]
  27. Connolly, K.D.; Wadey, R.M.; Mathew, D.; Johnson, E.; Rees, D.A.; James, P.E. Evidence for Adipocyte-Derived Extracellular Vesicles in the Human Circulation. Endocrinology 2018, 159, 3259–3267. [Google Scholar] [CrossRef]
  28. Thomou, T.; Mori, M.A.; Dreyfuss, J.M.; Konishi, M.; Sakaguchi, M.; Wolfrum, C.; Rao, T.N.; Winnay, J.N.; Garcia-Martin, R.; Grinspoon, S.K.; et al. Adipose-Derived Circulating MiRNAs Regulate Gene Expression in Other Tissues. Nature 2017, 542, 450–455. [Google Scholar] [CrossRef] [Green Version]
  29. Flaherty, S.E., 3rd; Grijalva, A.; Xu, X.; Ables, E.; Nomani, A.; Ferrante, A.W., Jr. A Lipase-Independent Pathway of Lipid Release and Immune Modulation by Adipocytes. Science 2019, 363, 989–993. [Google Scholar] [CrossRef]
  30. Chevillet, J.R.; Kang, Q.; Ruf, I.K.; Briggs, H.A.; Vojtech, L.N.; Hughes, S.M.; Cheng, H.H.; Arroyo, J.D.; Meredith, E.K.; Gallichotte, E.N.; et al. Quantitative and Stoichiometric Analysis of the MicroRNA Content of Exosomes. Proc. Natl. Acad. Sci. USA 2014, 111, 14888–14893. [Google Scholar] [CrossRef]
  31. Li, Y.; He, X.; Li, Q.; Lai, H.; Zhang, H.; Hu, Z.; Li, Y.; Huang, S. EV-Origin: Enumerating the Tissue-Cellular Origin of Circulating Extracellular Vesicles Using ExLR Profile. Comput. Struct. Biotechnol. J. 2020, 18, 2851–2859. [Google Scholar] [CrossRef] [PubMed]
  32. Li, F.; Li, H.; Jin, X.; Zhang, Y.; Kang, X.; Zhang, Z.; Xu, M.; Qian, Z.; Ma, Z.; Gao, X.; et al. Adipose-Specific Knockdown of Sirt1 Results in Obesity and Insulin Resistance by Promoting Exosomes Release. Cell Cycle 2019, 18, 2067–2082. [Google Scholar] [CrossRef] [PubMed]
  33. Garcia-Martin, R.; Brandao, B.B.; Thomou, T.; Altindis, E.; Kahn, C.R. Tissue Differences in the Exosomal/Small Extracellular Vesicle Proteome and Their Potential as Indicators of Altered Tissue Metabolism. Cell Rep. 2022, 38, 110277. [Google Scholar] [CrossRef]
  34. Garcia-Martin, R.; Wang, G.; Brandão, B.B.; Marques Zanotto, T.; Shah, S.; Kumar Patel, S.; Schilling, B.; Kahn, C.R. MicroRNA Sequence Codes for Small Extracellular Vesicle Release and Cellular Retention. Nature 2022, 601, 446–451. [Google Scholar] [CrossRef] [PubMed]
  35. Ying, W.; Gao, H.; Castellani, F.; Dos, G.; Jin, Z.; Ly, C.; Olefsky, J.M.; Ying, W.; Gao, H.; Castellani, F.; et al. MiR-690, an Exosomal-Derived MiRNA from M2- Polarized Macrophages, Improves Insulin Sensitivity in Obese Mice Article from M2-Polarized Macrophages, Improves Insulin Sensitivity in Obese Mice. Cell Metab. 2021, 33, 781–790. [Google Scholar] [CrossRef]
  36. Ying, W.; Riopel, M.; Bandyopadhyay, G.; Dong, Y.; Birmingham, A.; Seo, J.B.; Ofrecio, J.M.; Wollam, J.; Hernandez-Carretero, A.; Fu, W.; et al. Adipose Tissue Macrophage-Derived Exosomal MiRNAs Can Modulate in Vivo and in Vitro Insulin Sensitivity. Cell 2017, 171, 372–384. [Google Scholar] [CrossRef] [Green Version]
  37. Jalabert, A.; Reininger, L.; Berger, E.; Coute, Y.; Meugnier, E.; Forterre, A.; Errazuriz-Cerda, E.; Geloen, A.; Aouadi, M.; Bouzakri, K.; et al. Profiling of Ob/Ob Mice Skeletal Muscle Exosome-like Vesicles Demonstrates Combined Action of MiRNAs, Proteins and Lipids to Modulate Lipid Homeostasis in Recipient Cells. Sci. Rep. 2022, 11, 21626. [Google Scholar] [CrossRef]
  38. Ling, D.; Song, H.; Shuo, L.; Wang, L.; Xie, P.; Li, W.; Liu, J.; Tong, Y.; Zhang, C.-Y.; Jiang, X.; et al. Gonadal White Adipose Tissue-Derived Exosomasl MiR-222 Promotes Obesity-Associated Insulin Resistance. Aging 2020, 12, 22719–22742. [Google Scholar]
  39. Crewe, C.; Joffin, N.; Rutkowski, J.M.; Kim, M.; Zhang, F.; Towler, D.A.; Gordillo, R.; Scherer, P.E. An Endothelial-to-Adipocyte Extracellular Vesicle Axis Governed by Metabolic State. Cell 2018, 175, 695–708. [Google Scholar] [CrossRef] [Green Version]
  40. Song, Y.; Li, H.; Ren, X.; Li, H.; Feng, C. SNHG9, Delivered by Adipocyte-Derived Exosomes, Alleviates Inflammation and Apoptosis of Endothelial Cells through Suppressing TRADD Expression. Eur. J. Pharmacol. 2020, 872, 172977. [Google Scholar] [CrossRef]
  41. Obata, Y.; Kita, S.; Koyama, Y.; Fukuda, S.; Takeda, H.; Takahashi, M.; Fujishima, Y.; Nagao, H.; Masuda, S.; Tanaka, Y.; et al. Adiponectin/T-Cadherin System Enhances Exosome Biogenesis and Decreases Cellular Ceramides by Exosomal Release. JCI Insight 2018, 3, e99680. [Google Scholar] [CrossRef] [PubMed]
  42. Li, X.; Ballantyne, L.L.; Yu, Y.; Funk, C.D. Perivascular Adipose Tissue-Derived Extracellular Vesicle MiR-221-3p Mediates Vascular Remodeling. FASEB J. 2019, 33, 12704–12722. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Huang, X.Y.; Chen, J.X.; Ren, Y.; Fan, L.C.; Xiang, W.; He, X.J. Exosomal MiR-122 Promotes Adipogenesis and Aggravates Obesity through the VDR/SREBF1 Axis. Obesity 2022, 30, 666–679. [Google Scholar] [CrossRef] [PubMed]
  44. Mleczko, J.; Ortega, F.J.; Falcon-Perez, J.M.; Wabitsch, M.; Fernandez-Real, J.M.; Mora, S. Extracellular Vesicles from Hypoxic Adipocytes and Obese Subjects Reduce Insulin-Stimulated Glucose Uptake. Mol. Nutr. Food Res. 2018, 62, 1700917. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Yang, W.; Tu, H.; Tang, K.; Huang, H.; Ou, S.; Wu, J. MiR-3064 in Epicardial Adipose-Derived Exosomes Targets Neuronatin to Regulate Adipogenic Differentiation of Epicardial Adipose Stem Cells. Front. Cardiovasc. Med. 2021, 8, 709079. [Google Scholar] [CrossRef] [PubMed]
  46. Pan, Y.; Hui, X.; Chong Hoo, R.L.; Ye, D.; Cheung Chan, C.Y.; Feng, T.; Wang, Y.; Ling Lam, K.S.; Xu, A. Adipocyte-Secreted Exosomal MicroRNA-34a Inhibits M2 Macrophage Polarization to Promote Obesity-Induced Adipose Inflammation. J. Clin. Investig. 2019, 129, 834–849. [Google Scholar] [CrossRef] [Green Version]
  47. Zhang, D.; Yao, X.; Teng, Y.; Zhao, T.; Lin, L.; Li, Y.; Shang, H.; Jin, Y.; Jin, Q. Adipocytes-Derived Exosomal MicroRNA-1224 Inhibits M2 Macrophage Polarization in Obesity-Induced Adipose Tissue Inflammation via MSI2-Mediated Wnt/β-Catenin Axis. Mol. Nutr. Food Res. 2022, 66, e2100889. [Google Scholar] [CrossRef]
  48. Xie, Z.; Wang, X.; Liu, X.; Du, H.; Sun, C.; Shao, X.; Tian, J.; Gu, X.; Wang, H.; Tian, J.; et al. Adipose-Derived Exosomes Exert Proatherogenic Effects by Regulating Macrophage Foam Cell Formation and Polarization. J. Am. Heart Assoc. 2018, 7, e007442. [Google Scholar] [CrossRef]
  49. Yu, Y.; Du, H.; Wei, S.; Feng, L.; Li, J.; Yao, F.; Zhang, M.; Hatch, G.M.; Chen, L. Adipocyte-Derived Exosomal MiR-27a Induces Insulin Resistance in Skeletal Muscle through Repression of PPARγ. Theranostics 2018, 8, 2171–2188. [Google Scholar] [CrossRef]
  50. Crewe, C.; Funcke, J.B.; Li, S.; Joffin, N.; Gliniak, C.M.; Ghaben, A.L.; An, Y.A.; Sadek, H.A.; Gordillo, R.; Akgul, Y.; et al. Extracellular Vesicle-Based Interorgan Transport of Mitochondria from Energetically Stressed Adipocytes. Cell Metab. 2021, 33, 1853–1868.e11. [Google Scholar] [CrossRef]
  51. Man, W.; Song, X.; Xiong, Z.; Gu, J.; Lin, J.; Gu, X.; Yu, D.; Li, C.; Jiang, M.; Zhang, X.; et al. Exosomes Derived from Pericardial Adipose Tissues Attenuate Cardiac Remodeling Following Myocardial Infarction by Adipsin-Regulated Iron Homeostasis. Front. Cardiovasc. Med. 2022, 9, 2408. [Google Scholar] [CrossRef]
  52. Itokazu, M.; Onodera, Y.; Mori, T.; Inoue, S.; Yamagishi, K.; Moritake, A.; Iwawaki, N.; Shigi, K.; Takehara, T.; Higashimoto, Y.; et al. Adipose-Derived Exosomes Block Muscular Stem Cell Proliferation in Aged Mouse by Delivering MiRNA Let-7d-3p That Targets Transcription Factor HMGA2. J. Biol. Chem. 2022, 298, 102098. [Google Scholar] [CrossRef]
  53. Dang, S.Y.; Leng, Y.; Wang, Z.X.; Xiao, X.; Zhang, X.; Wen, T.; Gong, H.Z.; Hong, A.; Ma, Y. Exosomal Transfer of Obesity Adipose Tissue for Decreased MiR-141-3p Mediate Insulin Resistance of Hepatocytes. Int. J. Biol. Sci. 2019, 15, 351–368. [Google Scholar] [CrossRef]
  54. Li, Y.; Luan, Y.; Li, J.; Song, H.; Li, Y.; Qi, H.; Sun, B.; Zhang, P.; Wu, X.; Liu, X.; et al. Exosomal MiR-199a-5p Promotes Hepatic Lipid Accumulation by Modulating MST1 Expression and Fatty Acid Metabolism. Hepatol. Int. 2020, 14, 1057–1074. [Google Scholar] [CrossRef]
  55. Wang, J.; Li, L.; Zhang, Z.; Zhang, X.; Zhu, Y.; Zhang, C.; Bi, Y. Extracellular Vesicles Mediate the Communication of Adipose Tissue with Brain and Promote Cognitive Impairment Associated with Insulin Resistance. Cell Metab. 2022, 34, 1264–1279. [Google Scholar] [CrossRef]
  56. Zhang, Y.; Yu, M.; Dong, J.; Wu, Y.; Tian, W. Nucleophosmin3 Carried by Small Extracellular Vesicles Contribute to White Adipose Tissue Browning. J. Nanobiotechnol. 2022, 20, 165. [Google Scholar] [CrossRef]
  57. Zhang, Y.; Cai, Y.; Zhang, H.; Zhang, J.; Zeng, Y.; Fan, C.; Zou, S.; Wu, C.; Fang, S.; Li, P.; et al. Brown Adipose Tissue Transplantation Ameliorates Diabetic Nephropathy through the MiR-30b Pathway by Targeting Runx1. Metabolism 2021, 125, 154916. [Google Scholar] [CrossRef]
  58. Zhao, H.; Shang, Q.; Pan, Z.; Bai, Y.; Li, Z.; Zhang, H.; Zhang, Q.; Guo, C.; Zhang, L.; Wang, Q. Exosomes From Adipose-Derived Stem Cells Attenuate Adipose Inflammation and Obesity Through Polarizing M2 Macrophages and Beiging in White Adipose Tissue. Diabetes 2018, 67, 235–247. [Google Scholar] [CrossRef] [Green Version]
  59. Lee, K.S.; Lee, J.; Kim, H.K.; Yeom, S.H.; Woo, C.H.; Jung, Y.J.; Yun, Y.E.; Park, S.Y.; Han, J.; Kim, E.; et al. Extracellular Vesicles from Adipose Tissue-Derived Stem Cells Alleviate Osteoporosis through Osteoprotegerin and MiR-21-5p. J. Extracell. Vesicles 2021, 10, e12152. [Google Scholar] [CrossRef]
  60. Niu, Q.; Wang, T.; Wang, Z.; Wang, F.; Huang, D.; Sun, H.; Liu, H. Adipose-Derived Mesenchymal Stem Cell-Secreted Extracellular Vesicles Alleviate Non-Alcoholic Fatty Liver Disease via Delivering MiR-223-3p. Adipocyte 2022, 11, 572–587. [Google Scholar] [CrossRef]
  61. Liu, W.; Yuan, Y.; Liu, D. Extracellular Vesicles from Adipose-Derived Stem Cells Promote Diabetic Wound Healing via the PI3K-AKT-MTOR-HIF-1α Signaling Pathway. Tissue. Eng. Regen. Med. 2021, 18, 1035–1044. [Google Scholar] [CrossRef]
  62. Wang, J.; Wu, H.; Peng, Y.; Zhao, Y.; Qin, Y.; Zhang, Y.; Xiao, Z. Hypoxia Adipose Stem Cell-Derived Exosomes Promote High-Quality Healing of Diabetic Wound Involves Activation of PI3K/Akt Pathways. J. Nanobiotechnol. 2021, 19, 202. [Google Scholar] [CrossRef] [PubMed]
  63. Liu, T.; Sun, Y.-C.; Cheng, P.; Shao, H.-G. Adipose Tissue Macrophage-Derived Exosomal MiR-29a Regulates Obesity-Associated Insulin Resistance. Biochem. Biophys. Res. Commun. 2019, 515, 352–358. [Google Scholar] [CrossRef] [PubMed]
  64. Qian, B.; Yang, Y.; Tang, N.; Wang, J.; Sun, P.; Yang, N.; Chen, F.; Wu, T.; Sun, T.; Li, Y.; et al. M1 Macrophage-Derived Exosomes Impair Beta Cell Insulin Secretion via MiR-212-5p by Targeting SIRT2 and Inhibiting Akt/GSK-3β/β-Catenin Pathway in Mice. Diabetologia 2021, 64, 2037–2051. [Google Scholar] [CrossRef]
  65. Gao, H.; Luo, Z.; Jin, Z.; Ji, Y.; Ying, W. Adipose Tissue Macrophages Modulate Obesity-Associated β Cell Adaptations through Secreted MiRNA-Containing Extracellular Vesicles. Cells 2021, 10, 2451. [Google Scholar] [CrossRef]
  66. Castaño, C.; Mirasierra, M.; Vallejo, M.; Novials, A.; Parrizas, M. Delivery of Muscle-Derived Exosomal MiRNAs Induced by HIIT Improves Insulin Sensitivity through down-Regulation of Hepatic FoxO1 in Mice. Proc. Natl. Acad. Sci. USA 2020, 117, 30335–30343. [Google Scholar] [CrossRef]
  67. Vechetti, I.J.; Peck, B.D.; Wen, Y.; Walton, R.G.; Valentino, T.R.; Alimov, A.P.; Dungan, C.M.; van Pelt, D.W.; von Walden, F.; Alkner, B.; et al. Mechanical Overload-Induced Muscle-Derived Extracellular Vesicles Promote Adipose Tissue Lipolysis. FASEB J. 2021, 35, e21644. [Google Scholar] [CrossRef]
  68. Di, W.; Amdanee, N.; Zhang, W.; Zhou, Y. Long-Term Exercise-Secreted Extracellular Vesicles Promote Browning of White Adipocytes by Suppressing MiR-191a-5p. Life Sci. 2020, 263, 118464. [Google Scholar] [CrossRef]
  69. Shao, X.; Gong, W.; Wang, Q.; Wang, P.; Shi, T.; Mahmut, A.; Qin, J.; Yao, Y.; Yan, W.; Chen, D.; et al. Atrophic Skeletal Muscle Fibre-Derived Small Extracellular Vesicle MiR-690 Inhibits Satellite Cell Differentiation during Ageing. J. Cachex. Sarcopenia Muscle 2022, 13, 3163–3180. [Google Scholar] [CrossRef]
  70. Jalabert, A.; Vial, G.; Guay, C.; Wiklander, O.P.B.; Nordin, J.Z.; Aswad, H.; Forterre, A.; Meugnier, E.; Pesenti, S.; Regazzi, R.; et al. Exosome-like Vesicles Released from Lipid-Induced Insulin-Resistant Muscles Modulate Gene Expression and Proliferation of Beta Recipient Cells in Mice. Diabetologia 2016, 59, 1049–1058. [Google Scholar] [CrossRef] [Green Version]
  71. Kim, K.; Kang, J.K.; Jung, Y.H.; Lee, S.B.; Rametta, R.; Dongiovanni, P.; Valenti, L.; Pajvani, U.B. Adipocyte PHLPP2 Inhibition Prevents Obesity-Induced Fatty Liver. Nat. Commun. 2021, 12, 1822. [Google Scholar] [CrossRef] [PubMed]
  72. Jung, J.W.; Kim, J.E.; Kim, E.; Lee, H.; Lee, H.; Shin, E.A.; Lee, J.W. Liver-Originated Small Extracellular Vesicles with TM4SF5 Target Brown Adipose Tissue for Homeostatic Glucose Clearance. J. Extracell. Vesicles 2022, 11, e12262. [Google Scholar] [CrossRef] [PubMed]
  73. Ji, Y.; Luo, Z.; Gao, H.; dos Reis, F.C.G.; Bandyopadhyay, G.; Jin, Z.; Manda, K.A.; Isaac, R.; Yang, M.; Fu, W.; et al. Hepatocyte-Derived Exosomes from Early Onset Obese Mice Promote Insulin Sensitivity through MiR-3075. Nat. Metab. 2021, 3, 1163–1174. [Google Scholar] [CrossRef]
  74. Wang, Y.; Jin, P.; Liu, J.; Xie, X. Exosomal MicroRNA-122 Mediates Obesity-Related Cardiomyopathy through Suppressing Mitochondrial ADP-Ribosylation Factor-like 2. Clin. Sci. 2019, 133, 1871–1881. [Google Scholar] [CrossRef] [PubMed]
  75. Lou, J.; Wu, J.; Feng, M.; Dang, X.; Wu, G.; Yang, H.; Wang, Y.; Li, J.; Zhao, Y.; Shi, C.; et al. Exercise Promotes Angiogenesis by Enhancing Endothelial Cell Fatty Acid Utilization via Liver-Derived Extracellular Vesicle MiR-122-5p. J. Sport Health Sci. 2022, 11, 495–508. [Google Scholar] [CrossRef]
  76. Castaño, C.; Kalko, S.; Novials, A.; Párrizas, M. Obesity-Associated Exosomal MiRNAs Modulate Glucose and Lipid Metabolism in Mice. Proc. Natl. Acad. Sci. USA 2018, 115, 12158–12163. [Google Scholar] [CrossRef] [Green Version]
  77. Zhao, J.; Hu, L.; Gui, W.; Xiao, L.; Wang, W.; Xia, J.; Fan, H.; Li, Z.; Zhu, Q.; Hou, X.; et al. Hepatocyte TGF-β Signaling Inhibiting WAT Browning to Promote NAFLD and Obesity Is Associated With Let-7b-5p. Hepatol. Commun. 2022, 6, 1301–1321. [Google Scholar] [CrossRef]
  78. He, Y.; Rodrigues, R.M.; Wang, X.; Seo, W.; Ma, J.; Hwang, S.; Fu, Y.; Trojnár, E.; Mátyás, C.; Zhao, S.; et al. Neutrophil-to-Hepatocyte Communication via LDLR-Dependent MiR-223–Enriched Extracellular Vesicle Transfer Ameliorates Nonalcoholic Steatohepatitis. J. Clin. Invest. 2021, 131, e141513. [Google Scholar] [CrossRef]
  79. Xu, H.; Du, X.; Xu, J.; Zhang, Y.; Tian, Y.; Liu, G.; Wang, X.; Ma, M.; Duid, W.; Liu, Y.; et al. Pancreatic β Cell MicroRNA-26a Alleviates Type 2 Diabetes by Improving Peripheral Insulin Sensitivity and Preserving β Cell Function. PLoS Biol. 2020, 18, e3000603. [Google Scholar] [CrossRef]
  80. Burillo, J.; Fernández-Rhodes, M.; Piquero, M.; López-Alvarado, P.; Menéndez, J.C.; Jiménez, B.; González-Blanco, C.; Marqués, P.; Guillén, C.; Benito, M. Human Amylin Aggregates Release within Exosomes as a Protective Mechanism in Pancreatic β Cells: Pancreatic β-Hippocampal Cell Communication. Biochim. Biophys. Acta Mol. Cell Res. 2021, 1868, 118971. [Google Scholar] [CrossRef]
  81. Guay, C.; Kruit, J.K.; Rome, S.; Menoud, V.; Mulder, N.L.; Jurdzinski, A.; Mancarella, F.; Sebastiani, G.; Donda, A.; Gonzalez, B.J.; et al. Lymphocyte-Derived Exosomal MicroRNAs Promote Pancreatic β Cell Death and May Contribute to Type 1 Diabetes Development. Cell Metab. 2019, 29, 348–361.e6. [Google Scholar] [CrossRef] [PubMed]
  82. Phu, T.A.; Ng, M.; Vu, N.K.; Bouchareychas, L.; Raffai, R.L. IL-4 Polarized Human Macrophage Exosomes Control Cardiometabolic Inflammation and Diabetes in Obesity. Mol. Ther. 2022, 30, 2274–2297. [Google Scholar] [CrossRef] [PubMed]
  83. de Silva, N.; Samblas, M.; Martínez, J.A.; Milagro, F.I. Effects of Exosomes from LPS-Activated Macrophages on Adipocyte Gene Expression, Differentiation, and Insulin-Dependent Glucose Uptake. J. Physiol. Biochem. 2018, 74, 559–568. [Google Scholar] [CrossRef] [PubMed]
  84. Gesmundo, I.; Pardini, B.; Gargantini, E.; Gamba, G.; Birolo, G.; Fanciulli, A.; Banfi, D.; Congiusta, N.; Favaro, E.; Deregibus, M.C.; et al. Adipocyte-Derived Extracellular Vesicles Regulate Survival and Function of Pancreatic β Cells. JCI Insight 2021, 6, e141962. [Google Scholar] [CrossRef]
  85. Ge, Q.; Xie, X.; Chen, X.; Huang, R.; Rui, C.X.; Zhen, Q.; Hu, R.; Wu, M.; Xiao, X.; Li, X. Circulating Exosome-like Vesicles of Humans with Nondiabetic Obesity Impaired Islet β-Cell Proliferation, Which Was Associated with Decreased Omentin-1 Protein Cargo. Genes Dis. 2022, 9, 1099–1113. [Google Scholar] [CrossRef]
  86. Gu, H.; Yang, K.; Shen, Z.; Jia, K.; Liu, P.; Pan, M.; Sun, C. ER Stress-Induced Adipocytes Secrete-Aldo-Keto Reductase 1B7-Containing Exosomes That Cause Nonalcoholic Steatohepatitis in Mice. Free Radic. Biol. Med. 2021, 163, 220–233. [Google Scholar] [CrossRef]
  87. Fuchs, A.; Samovski, D.; Smith, G.I.; Cifarelli, V.; Farabi, S.S.; Yoshino, J.; Pietka, T.; Chang, S.W.; Ghosh, S.; Myckatyn, T.M.; et al. Associations Among Adipose Tissue Immunology, Inflammation, Exosomes and Insulin Sensitivity in People With Obesity and Nonalcoholic Fatty Liver Disease. Gastroenterology 2021, 161, 968–981.e12. [Google Scholar] [CrossRef]
  88. Gan, L.; Xie, D.; Liu, J.; Bond Lau, W.; Christopher, T.A.; Lopez, B.; Zhang, L.; Gao, E.; Koch, W.; Ma, X.L.; et al. Small Extracellular Microvesicles Mediated Pathological Communications Between Dysfunctional Adipocytes and Cardiomyocytes as a Novel Mechanism Exacerbating Ischemia/Reperfusion Injury in Diabetic Mice. Circulation 2020, 141, 968–983. [Google Scholar] [CrossRef] [PubMed]
  89. Gao, J.; Li, X.; Wang, Y.; Cao, Y.; Yao, D.; Sun, L.; Qin, L.; Qiu, H.; Zhan, X. Adipocyte-Derived Extracellular Vesicles Modulate Appetite and Weight through MTOR Signalling in the Hypothalamus. Acta Physiol. 2020, 228, e13339. [Google Scholar] [CrossRef] [PubMed]
  90. Liu, Y.; Wang, Y.; Wang, C.; Shi, R.; Zhou, X.; Li, Z.; Sun, W.; Zhao, L.; Yuan, L. Maternal Obesity Increases the Risk of Fetal Cardiac Dysfunction via Visceral Adipose Tissue Derived Exosomes. Placenta 2021, 105, 85–93. [Google Scholar] [CrossRef]
  91. Wang, J.; Wu, Y.; Guo, J.; Fei, X.; Yu, L.; Ma, S. Adipocyte-Derived Exosomes Promote Lung Cancer Metastasis by Increasing MMP9 Activity via Transferring MMP3 to Lung Cancer Cells. Oncotarget 2017, 8, 81880–81891. [Google Scholar] [CrossRef] [PubMed]
  92. Clement, E.; Lazar, I.; Attané, C.; Carrié, L.; Dauvillier, S.; Ducoux-Petit, M.; Esteve, D.; Menneteau, T.; Moutahir, M.; le Gonidec, S.; et al. Adipocyte Extracellular Vesicles Carry Enzymes and Fatty Acids That Stimulate Mitochondrial Metabolism and Remodeling in Tumor Cells. EMBO J. 2020, 39, e102525. [Google Scholar] [CrossRef] [PubMed]
  93. Fontana, F.; Anselmi, M.; Carollo, E.; Sartori, P.; Procacci, P.; Carter, D.; Limonta, P. Adipocyte-Derived Extracellular Vesicles Promote Prostate Cancer Cell Aggressiveness by Enabling Multiple Phenotypic and Metabolic Changes. Cells 2022, 11, 2388. [Google Scholar] [CrossRef] [PubMed]
  94. La Camera, G.; Gelsomino, L.; Malivindi, R.; Barone, I.; Panza, S.; de Rose, D.; Giordano, F.; D’Esposito, V.; Formisano, P.; Bonofiglio, D.; et al. Adipocyte-Derived Extracellular Vesicles Promote Breast Cancer Cell Malignancy through HIF-1α Activity. Cancer Lett. 2021, 521, 155–168. [Google Scholar] [CrossRef]
  95. Lazar, I.; Clement, E.; Carrié, L.; Esteve, D.; Dauvillier, S.; Moutahir, M.; Dalle, S.; Delmas, V.; Andrieu-Abadie, N.; Larue, L.; et al. Adipocyte Extracellular Vesicles Decrease P16INK4A in Melanoma: An Additional Link between Obesity and Cancer. J. Invest. Dermatol. 2022, 142, 2488–2498.e8. [Google Scholar] [CrossRef]
  96. Seale, P.; Bjork, B.; Yang, W.; Kajimura, S.; Chin, S.; Kuang, S.; Scimè, A.; Devarakonda, S.; Conroe, H.M.; Erdjument-Bromage, H.; et al. PRDM16 Controls a Brown Fat/Skeletal Muscle Switch. Nature 2008, 454, 961–967. [Google Scholar] [CrossRef] [Green Version]
  97. An, Y.; Wang, G.; Diao, Y.; Sun, H.; Wang, H.; Wu, Z. A Molecular Switch Regulating Cell Fate Choice between Muscle Progenitor Cells and Brown Adipocytes. Dev. Cell 2017, 41, 382–391. [Google Scholar] [CrossRef] [Green Version]
  98. Tseng, Y.-H.; Kokkotou, E.; Schulz, T.J.; Huang, T.L.; Winnay, J.N.; Taniguchi, C.M.; Tran, T.T.; Suzuki, R.; Espinoza, D.O.; Yamamoto, Y.; et al. New Role of Bone Morphogenetic Protein 7 in Brown Adipogenesis and Energy Expenditure. Nature 2008, 454, 1000–1004. [Google Scholar] [CrossRef]
  99. Villarroya, F.; Cereijo, R.; Villarroya, J.; Giralt, M. Brown Adipose Tissue as a Secretory Organ. Nat. Rev. Endocrinol. 2017, 13, 26–35. [Google Scholar] [CrossRef]
  100. Nedergaard, J.; Cannon, B. The Browning of White Adipose Tissue: Some Burning Issues. Cell Metab. 2014, 20, 396–407. [Google Scholar] [CrossRef] [Green Version]
  101. Hong, P.; Wu, Y.; Zhang, Q.; Liu, P.; Zhang, S.; Yu, M.; Tian, W. Identification of Thermogenesis-Related LncRNAs in Small Extracellular Vesicles Derived from Adipose Tissue. BMC Genom. 2022, 23, 660. [Google Scholar] [CrossRef] [PubMed]
  102. Zhou, X.; Li, Z.; Qi, M.; Zhao, P.; Duan, Y.; Yang, G.; Yuan, L. Brown Adipose Tissue-Derived Exosomes Mitigate the Metabolic Syndrome in High Fat Diet Mice. Theranostics 2020, 10, 8197–8210. [Google Scholar] [CrossRef] [PubMed]
  103. Zhao, H.; Chen, X.; Hu, G.; Li, C.; Guo, L.; Zhang, L.; Sun, F.; Xia, Y.; Yan, W.; Cui, Z.; et al. Small Extracellular Vesicles From Brown Adipose Tissue Mediate Exercise Cardioprotection. Circ. Res. 2022, 130, 1490–1506. [Google Scholar] [CrossRef] [PubMed]
  104. Wu, B.; Feng, J.; Guo, J.; Wang, J.; Xiu, G.; Xu, J.; Ning, K.; Ling, B.; Fu, Q.; Xu, J. ADSCs-Derived Exosomes Ameliorate Hepatic Fibrosis by Suppressing Stellate Cell Activation and Remodeling Hepatocellular Glutamine Synthetase-Mediated Glutamine and Ammonia Homeostasis. Stem Cell Res. Ther. 2022, 13, 494. [Google Scholar] [CrossRef] [PubMed]
  105. Cheng, L.; Yu, P.; Li, F.; Jiang, X.; Jiao, X.; Shen, Y.; Lai, X. Human Umbilical Cord-Derived Mesenchymal Stem Cell-Exosomal MiR-627-5p Ameliorates Non-Alcoholic Fatty Liver Disease by Repressing FTO Expression. Hum. Cell 2021, 34, 1697–1708. [Google Scholar] [CrossRef]
  106. Jin, J.; Shi, Y.; Gong, J.; Zhao, L.; Li, Y.; He, Q.; Huang, H. Exosome Secreted from Adipose-Derived Stem Cells Attenuates Diabetic Nephropathy by Promoting Autophagy Flux and Inhibiting Apoptosis in Podocyte. Stem Cell Res. Ther. 2019, 10, 95. [Google Scholar] [CrossRef] [Green Version]
  107. Duan, Y.; Luo, Q.; Wang, Y.; Ma, Y.; Chen, F.; Zhu, X.; Shi, J. Adipose Mesenchymal Stem Cell-Derived Extracellular Vesicles Containing MicroRNA-26a-5p Target TLR4 and Protect against Diabetic Nephropathy. J. Biol. Chem. 2020, 295, 12868–12884. [Google Scholar] [CrossRef]
  108. Kang, Y.; Song, Y.; Luo, Y.; Song, J.; Li, C.; Yang, S.; Guo, J.; Yu, J.; Zhang, X. Exosomes Derived from Human Umbilical Cord Mesenchymal Stem Cells Ameliorate Experimental Non-Alcoholic Steatohepatitis via Nrf2/NQO-1 Pathway. Free Radic. Biol. Med. 2022, 192, 25–36. [Google Scholar] [CrossRef]
  109. Chen, M.-T.; Zhao, Y.-T.; Zhou, L.-Y.; Li, M.; Zhang, Q.; Han, Q.; Xiao, X.-H. Exosomes Derived from Human Umbilical Cord Mesenchymal Stem Cells Enhance Insulin Sensitivity in Insulin Resistant Human Adipocytes. Curr. Med. Sci. 2021, 41, 87–93. [Google Scholar] [CrossRef]
  110. Sun, Y.; Shi, H.; Yin, S.; Ji, C.; Zhang, X.; Zhang, B.; Wu, P.; Shi, Y.; Mao, F.; Yan, Y.; et al. Human Mesenchymal Stem Cell Derived Exosomes Alleviate Type 2 Diabetes Mellitus through Reversing Peripheral Insulin Resistance and Relieving β-Cell Destruction. ACS Nano 2017, 12, 7613–7628. [Google Scholar] [CrossRef]
  111. Pomatto, M.; Gai, C.; Negro, F.; Cedrino, M.; Grange, C.; Ceccotti, E.; Togliatto, G.; Collino, F.; Tapparo, M.; Figliolini, F.; et al. Differential Therapeutic Effect of Extracellular Vesicles Derived by Bone Marrow and Adipose Mesenchymal Stem Cells on Wound Healing of Diabetic Ulcers and Correlation to Their Cargoes. Int. J. Mol. Sci. 2021, 22, 3851. [Google Scholar] [CrossRef] [PubMed]
  112. Horiguchi, M.; Okada, Y.; Turudome, Y.; Ushijima, K. Exosome Degeneration in Mesenchymal Stem Cells Derived from Patients with Type 1 Diabetes Mellitus. Int. J. Mol. Sci. 2021, 22, 10906. [Google Scholar] [CrossRef] [PubMed]
  113. Su, T.; Xiao, Y.; Xiao, Y.; Guo, Q.; Li, C.; Huang, Y.; Deng, Q.; Wen, J.; Zhou, F.; Luo, X.H. Bone Marrow Mesenchymal Stem Cells-Derived Exosomal MiR-29b-3p Regulates Aging-Associated Insulin Resistance. ACS Nano 2019, 13, 2450–2462. [Google Scholar] [CrossRef]
  114. Trovato, E.; di Felice, V.; Barone, R. Extracellular Vesicles: Delivery Vehicles of Myokines. Front. Physiol. 2019, 10, 1–13. [Google Scholar] [CrossRef] [Green Version]
  115. Karstoft, K.; Pedersen, B.K. Skeletal Muscle as a Gene Regulatory Endocrine Organ. Curr. Opin. Clin. Nutr. Metab. Care 2016, 19, 270–275. [Google Scholar] [CrossRef] [PubMed]
  116. Sullivan, B.P.; Nie, Y.; Evans, S.; Kargl, C.K.; Hettinger, Z.R.; Garner, R.T.; Hubal, M.J.; Kuang, S.; Stout, J.; Gavin, T.P. Obesity and Exercise Training Alter Inflammatory Pathway Skeletal Muscle Small Extracellular Vesicle MicroRNAs. Exp. Physiol. 2022, 107, 462–475. [Google Scholar] [CrossRef]
  117. Whitham, M.; Parker, B.L.; Friedrichsen, M.; Hingst, J.R.; Hjorth, M.; Hughes, W.E.; Egan, C.L.; Cron, L.; Watt, K.I.; Kuchel, R.P.; et al. Extracellular Vesicles Provide a Means for Tissue Crosstalk during Exercise. Cell Metab. 2018, 27, 237–251. [Google Scholar] [CrossRef] [Green Version]
  118. Mullen, M.; Williams, K.; LaRocca, T.; Duke, V.; Hambright, W.S.; Ravuri, S.K.; Bahney, C.S.; Ehrhart, N.; Huard, J. Mechanical Strain Drives Exosome Production, Function, and MiRNA Cargo in C2C12 Muscle Progenitor Cells. J. Orthop. Res. 2022. [Google Scholar] [CrossRef]
  119. Wang, D.; Zhang, X.; Li, Y.; Jia, L.; Zhai, L.; Wei, W.; Zhang, L.; Jiang, H.; Bai, Y. Exercise-Induced Browning of White Adipose Tissue and Improving Skeletal Muscle Insulin Sensitivity in Obese/Non-Obese Growing Mice: Do Not Neglect Exosomal MiR-27a. Front. Nutr. 2022, 9, 1276. [Google Scholar] [CrossRef]
  120. Takafuji, Y.; Kawao, N.; Ohira, T.; Mizukami, Y.; Okada, K.; Jo, J.-I.; Tabata, Y.; Kaji, H. Extracellular Vesicles Secreted from Mouse Muscle Cells Improve Delayed Bone Repair in Diabetic Mice. Endocr. J. 2022, 56, 0918–8959. [Google Scholar] [CrossRef]
  121. Takafuji, Y.; Tatsumi, K.; Kawao, N.; Okada, K.; Muratani, M.; Kaji, H. MicroRNA-196a-5p in Extracellular Vesicles Secreted from Myoblasts Suppresses Osteoclast-like Cell Formation in Mouse Cells. Calcif. Tissue Int. 2021, 108, 364–376. [Google Scholar] [CrossRef] [PubMed]
  122. Nie, Y.; Sato, Y.; Garner, R.T.; Kargl, C.; Wang, C.; Kuang, S.; Gilpin, C.J.; Gavin, T.P. Skeletal Muscle-Derived Exosomes Regulate Endothelial Cell Functions via Reactive Oxygen Species-Activated Nuclear Factor-ΚB Signalling. Exp. Physiol. 2019, 104, 1262–1273. [Google Scholar] [CrossRef] [PubMed]
  123. Wu, J.; Dong, T.; Chen, T.; Sun, J.; Luo, J.; He, J.; Wei, L.; Zeng, B.; Zhang, H.; Li, W.; et al. Hepatic Exosome-Derived MiR-130a-3p Attenuates Glucose Intolerance via Suppressing PHLPP2 Gene in Adipocyte. Metabolism 2020, 103, 154006. [Google Scholar] [CrossRef] [PubMed]
  124. Fabregat, I.; Moreno-Càceres, J.; Sánchez, A.; Dooley, S.; Dewidar, B.; Giannelli, G.; ten Dijke, P. TGF-β Signalling and Liver Disease. FEBS J. 2016, 283, 2219–2232. [Google Scholar] [CrossRef] [Green Version]
  125. Ogino, N.; Takahashi, H.; Nagaoka, K.; Harada, Y.; Kubo, M.; Miyagawa, K.; Kusanaga, M.; Oe, S.; Honma, Y.; Harada, M.; et al. Possible Contribution of Hepatocyte Secretion to the Elevation of Plasma Exosomal Arginase-1 in High-Fat Diet-Fed Mice. Life Sci. 2021, 278, 119588. [Google Scholar] [CrossRef]
  126. Royo, F.; Moreno, L.; Mleczko, J.; Palomo, L.; Gonzalez, E.; Cabrera, D.; Cogolludo, A.; Perez Vizcaino, F.; Van-Liempd, S.; Falcon-Perez, J.M. Hepatocyte-Secreted Extracellular Vesicles Modify Blood Metabolome and Endothelial Function by an Arginase-Dependent Mechanism. Sci. Rep. 2017, 7, 42798. [Google Scholar] [CrossRef] [Green Version]
  127. Shah, R.; Murthy, V.; Pacold, M.; Danielson, K.; Tanriverdi, K.; Larson, M.G.; Hanspers, K.; Pico, A.; Mick, E.; Reis, J.; et al. Extracellular RNAs Are Associated with Insulin Resistance and Metabolic Phenotypes. Diabetes Care 2017, 40, 546–553. [Google Scholar] [CrossRef] [Green Version]
  128. Das, A.; Basu, S.; Bandyopadhyay, D.; Mukherjee, K.; Datta, D.; Chakraborty, S.; Jana, S.; Adak, M.; Bose, S.; Chakrabarti, S.; et al. Inhibition of Extracellular Vesicle-Associated MMP2 Abrogates Intercellular Hepatic MiR-122 Transfer to Liver Macrophages and Curtails Inflammation. iScience 2021, 24, 103428. [Google Scholar] [CrossRef]
  129. Wan, L.; Xia, T.; Du, Y.; Liu, J.; Xie, Y.; Zhang, Y.; Guan, F.; Wu, J.; Wang, X.; Shi, C.; et al. Exosomes from Activated Hepatic Stellate Cells Contain GLUT1 and PKM2: A Role for Exosomes in Metabolic Switch of Liver Nonparenchymal Cells. FASEB J. 2019, 33, 8530–8542. [Google Scholar] [CrossRef]
  130. Chidester, S.; Livinski, A.A.; Fish, A.F.; Joseph, P.V. The Role of Extracellular Vesicles in β-Cell Function and Viability: A Scoping Review. Front. Endocrinol. 2020, 11, 375. [Google Scholar] [CrossRef]
  131. Sun, Y.; Mao, Q.; Shen, C.; Wang, C.; Jia, W. Exosomes from β-Cells Alleviated Hyperglycemia and Enhanced Angiogenesis in Islets of Streptozotocin-Induced Diabetic Mice. Diabetes Metab. Syndr. Obes. 2019, 12, 2053–2064. [Google Scholar] [CrossRef] [PubMed]
  132. Ribeiro, D.; Horvath, I.; Heath, N.; Hicks, R.; Forslöw, A.; Wittung-Stafshede, P. Extracellular Vesicles from Human Pancreatic Islets Suppress Human Islet Amyloid Polypeptide Amyloid Formation. Proc. Natl. Acad. Sci. USA 2017, 114, 11127–11132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Lakhter, A.J.; Pratt, R.E.; Moore, R.E.; Doucette, K.K.; Maier, B.F.; Dimeglio, L.A.; Sims, E.K. Beta Cell Extracellular Vesicle MiR-21-5p Cargo Is Increased in Response to Inflammatory Cytokines and Serves as a Biomarker of Type 1 Diabetes. Diabetologia 2018, 61, 1124–1134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Javeed, N.; Her, T.K.; Brown, M.R.; Vanderboom, P.; Rakshit, K.; Egan, A.M.; Vella, A.; Lanza, I.; Matveyenko, A.v. Pro-Inflammatory β Cell Small Extracellular Vesicles Induce β Cell Failure through Activation of the CXCL10/CXCR3 Axis in Diabetes. Cell Rep. 2021, 36, 109613. [Google Scholar] [CrossRef]
  135. Cao, M.; Isaac, R.; Yan, W.; Ruan, X.; Jiang, L.; Wan, Y.; Wang, J.; Wang, E.; Caron, C.; Neben, S.; et al. Cancer-Cell-Secreted Extracellular Vesicles Suppress Insulin Secretion through MiR-122 to Impair Systemic Glucose Homeostasis and Contribute to Tumour Growth. Nat. Cell Biol. 2022, 24, 954–967. [Google Scholar] [CrossRef]
  136. Chambers, E.S.; Preston, T.; Frost, G.; Morrison, D.J. Role of Gut Microbiota-Generated Short-Chain Fatty Acids in Metabolic and Cardiovascular Health. Curr. Nutr. Rep. 2018, 7, 198–206. [Google Scholar] [CrossRef] [Green Version]
  137. Virtue, A.T.; McCright, S.J.; Wright, J.M.; Jimenez, M.T.; Mowel, W.K.; Kotzin, J.J.; Joannas, L.; Basavappa, M.G.; Spencer, S.P.; Clark, M.L.; et al. The Gut Microbiota Regulates White Adipose Tissue Inflammation and Obesity via a Family of MicroRNAs. Sci. Transl. Med. 2019, 11, eaav1892. [Google Scholar] [CrossRef]
  138. Kim, B.; Choi, H.N.; Yim, J.E. Effect of Diet on the Gut Microbiota Associated with Obesity. J. Obes. Metab. Syndr. 2019, 28, 216–224. [Google Scholar] [CrossRef] [Green Version]
  139. Rinninella, E.; Cintoni, M.; Raoul, P.; Lopetuso, L.R.; Scaldaferri, F.; Pulcini, G.; Miggiano, G.A.D.; Gasbarrini, A.; Mele, M.C. Food Components and Dietary Habits: Keys for a Healthy Gut Microbiota Composition. Nutrients 2019, 11, 2393. [Google Scholar] [CrossRef] [Green Version]
  140. Kong, C.; Gao, R.; Yan, X.; Huang, L.; Qin, H. Probiotics Improve Gut Microbiota Dysbiosis in Obese Mice Fed a High-Fat or High-Sucrose Diet. Nutrition 2019, 60, 175–184. [Google Scholar] [CrossRef]
  141. Kim, M.S.; Hwang, S.S.; Park, E.J.; Bae, J.W. Strict Vegetarian Diet Improves the Risk Factors Associated with Metabolic Diseases by Modulating Gut Microbiota and Reducing Intestinal Inflammation. Environ. Microbiol. Rep. 2013, 5, 765–775. [Google Scholar] [CrossRef] [PubMed]
  142. Rosés, C.; Cuevas-Sierra, A.; Quintana, S.; Riezu-Boj, J.I.; Alfredo Martínez, J.; Milagro, F.I.; Barceló, A. Gut Microbiota Bacterial Species Associated with Mediterranean Diet-Related Food Groups in a Northern Spanish Population. Nutrients 2021, 13, 636. [Google Scholar] [CrossRef] [PubMed]
  143. Menni, C.; Jackson, M.A.; Pallister, T.; Steves, C.J.; Spector, T.D.; Valdes, A.M. Gut Microbiome Diversity and High-Fibre Intake Are Related to Lower Long-Term Weight Gain. Int. J. Obes. 2017, 41, 1099–1105. [Google Scholar] [CrossRef] [Green Version]
  144. Li, L.; Li, C.; Lv, M.; Hu, Q.; Guo, L.; Xiong, D. Correlation between Alterations of Gut Microbiota and MiR-122-5p Expression in Patients with Type 2 Diabetes Mellitus. Ann. Transl. Med. 2020, 8, 1481. [Google Scholar] [CrossRef] [PubMed]
  145. Noble, E.E.; Hsu, T.M.; Kanoski, S.E. Gut to Brain Dysbiosis: Mechanisms Linking Western Diet Consumption, the Microbiome, and Cognitive Impairment. Front. Behav. Neurosci. 2017, 11, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Agus, A.; Denizot, J.; Thévenot, J.; Martinez-Medina, M.; Massier, S.; Sauvanet, P.; Bernalier-Donadille, A.; Denis, S.; Hofman, P.; Bonnet, R.; et al. Western Diet Induces a Shift in Microbiota Composition Enhancing Susceptibility to Adherent-Invasive, E. Coli Infection and Intestinal Inflammation. Sci. Rep. 2016, 6, 19032. [Google Scholar] [CrossRef] [Green Version]
  147. Chang, C.J.; Lu, C.C.; Lin, C.S.; Martel, J.; Ko, Y.F.; Ojcius, D.M.; Wu, T.R.; Tsai, Y.H.; Yeh, T.S.; Lu, J.J.; et al. Antrodia Cinnamomea Reduces Obesity and Modulates the Gut Microbiota in High-Fat Diet-Fed Mice. Int. J. Obes. 2018, 42, 231–243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Dao, M.C.; Everard, A.; Aron-Wisnewsky, J.; Sokolovska, N.; Prifti, E.; Verger, E.O.; Kayser, B.D.; Levenez, F.; Chilloux, J.; Hoyles, L.; et al. Akkermansia Muciniphila and Improved Metabolic Health during a Dietary Intervention in Obesity: Relationship with Gut Microbiome Richness and Ecology. Gut 2016, 65, 426–436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Whisner, C.M.; Maldonado, J.; Dente, B.; Krajmalnik-Brown, R.; Bruening, M. Diet, Physical Activity and Screen Time but Not Body Mass Index Are Associated with the Gut Microbiome of a Diverse Cohort of College Students Living in University Housing: A Cross-Sectional Study. BMC Microbiol. 2018, 18, 210. [Google Scholar] [CrossRef]
  150. Maniar, K.; Moideen, A.; Bhattacharyya, R.; Banerjee, D. Metformin Exerts Anti-Obesity Effect via Gut Microbiome Modulation in Prediabetics: A Hypothesis. Med. Hypotheses 2017, 104, 117–120. [Google Scholar] [CrossRef]
  151. Lee, H.; Lee, Y.; Kim, J.; An, J.; Lee, S.; Kong, H.; Song, Y.; Lee, C.K.; Kim, K. Modulation of the Gut Microbiota by Metformin Improves Metabolic Profiles in Aged Obese Mice. Gut Microbes 2018, 9, 155–165. [Google Scholar] [CrossRef]
  152. Myronovych, A.; Peck, B.C.E.; An, M.; Zhu, J.; Warm, A.; Kupe, A.; Lubman, D.M.; Seeley, R.J. Intestinal Extracellular Vesicles Are Altered by Vertical Sleeve Gastrectomy. Am. J. Physiol. Gastrointest. Liver Physiol. 2021, 320, G153–G165. [Google Scholar] [CrossRef] [PubMed]
  153. Huh, Y.J.; Seo, J.Y.; Nam, J.; Yang, J.; McDowell, A.; Kim, Y.K.; Lee, J.H. Bariatric/Metabolic Surgery Induces Noticeable Changes of Microbiota and Their Secreting Extracellular Vesicle Composition in the Gut. Obes. Surg. 2019, 29, 2470–2484. [Google Scholar] [CrossRef] [PubMed]
  154. Stark, C.M.; Susi, A.; Emerick, J.; Nylund, C.M. Antibiotic and Acid-Suppression Medications during Early Childhood Are Associated with Obesity. Gut 2019, 68, 62–69. [Google Scholar] [CrossRef] [PubMed]
  155. Cuesta, C.M.; Guerri, C.; Urena, J.; Pascual, M. Role of Microbiota-Derived Extracellular Vesicles in Gut-Brain Communication. Int. J. Mol. Sci. 2021, 22, 4235. [Google Scholar] [CrossRef] [PubMed]
  156. Tan, J.; Ni, D.; Taitz, J.; Pinget, G.V.; Read, M.; Senior, A.; Wali, J.A.; Elnour, R.; Shanahan, E.; Wu, H.; et al. Dietary Protein Increases T-Cell-Independent SIgA Production through Changes in Gut Microbiota-Derived Extracellular Vesicles. Nat. Commun. 2022, 13, 4336. [Google Scholar] [CrossRef] [PubMed]
  157. Choi, Y.; Kwon, Y.; Kim, D.K.; Jeon, J.; Jang, S.C.; Wang, T.; Ban, M.; Kim, M.H.; Jeon, S.G.; Kim, M.S.; et al. Gut Microbe-Derived Extracellular Vesicles Induce Insulin Resistance, Thereby Impairing Glucose Metabolism in Skeletal Muscle. Sci. Rep. 2015, 5, 15878. [Google Scholar] [CrossRef] [Green Version]
  158. Tulkens, J.; Vergauwen, G.; van Deun, J.; Geeurickx, E.; Dhondt, B.; Lippens, L.; de Scheerder, M.A.; Miinalainen, I.; Rappu, P.; de Geest, B.G.; et al. Increased Levels of Systemic LPS-Positive Bacterial Extracellular Vesicles in Patients with Intestinal Barrier Dysfunction. Gut 2020, 69, 191–193. [Google Scholar] [CrossRef] [Green Version]
  159. Ashrafian, F.; Behrouzi, A.; Shahriary, A.; Badi, S.A.; Davari, M.; Khatami, S.; Jamnani, F.R.; Fateh, A.; Vaziri, F.; Siadat, S.D. Comparative Study of Effect of Akkermansia Muciniphila and Its Extracellular Vesicles on Toll-like Receptors and Tight Junction. Gastroenterol. Hepatol. Bed Bench 2019, 12, 163–168. [Google Scholar]
  160. Hiippala, K.; Barreto, G.; Burrello, C.; Diaz-Basabe, A.; Suutarinen, M.; Kainulainen, V.; Bowers, J.R.; Lemmer, D.; Engelthaler, D.M.; Eklund, K.K.; et al. Novel Odoribacter Splanchnicus Strain and Its Outer Membrane Vesicles Exert Immunoregulatory Effects in Vitro. Front. Microbiol. 2020, 11, 575455. [Google Scholar] [CrossRef]
  161. Alvarez, C.S.; Gimenez, R.; Canas, M.A.; Vera, R.; Diaz-Garrido, N.; Badia, J.; Baldoma, L. Extracellular Vesicles and Soluble Factors Secreted by Escherichia Coli Nissle 1917 and ECOR63 Protect against Enteropathogenic E. Coli-Induced Intestinal Epithelial Barrier Dysfunction. BMC Microbiol. 2019, 19, 166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Kang, C.S.; Ban, M.; Choi, E.J.; Moon, H.G.; Jeon, J.S.; Kim, D.K.; Park, S.K.; Jeon, S.G.; Roh, T.Y.; Myung, S.J.; et al. Extracellular Vesicles Derived from Gut Microbiota, Especially Akkermansia Muciniphila, Protect the Progression of Dextran Sulfate Sodium-Induced Colitis. PLoS ONE 2013, 8, e76520. [Google Scholar] [CrossRef] [PubMed]
  163. Fábrega, M.J.; Rodríguez-Nogales, A.; Garrido-Mesa, J.; Algieri, F.; Badía, J.; Giménez, R.; Gálvez, J.; Baldomà, L. Intestinal Anti-Inflammatory Effects of Outer Membrane Vesicles from Escherichia Coli Nissle 1917 in DSS-Experimental Colitis in Mice. Front. Microbiol. 2017, 8, 1274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Shen, Y.; Torchia, M.L.G.; Lawson, G.W.; Karp, C.L.; Ashwell, J.D.; Mazmanian, S.K. Outer Membrane Vesicles of a Human Commensal Mediate Immune Regulation and Disease Protection. Cell Host Microbe 2012, 12, 509–520. [Google Scholar] [CrossRef] [Green Version]
  165. Alpdundar Bulut, E.; Bayyurt Kocabas, B.; Yazar, V.; Aykut, G.; Guler, U.; Salih, B.; Surucu Yilmaz, N.; Ayanoglu, I.C.; Polat, M.M.; Akcali, K.C.; et al. Human Gut Commensal Membrane Vesicles Modulate Inflammation by Generating M2-like Macrophages and Myeloid-Derived Suppressor Cells. J. Immunol. 2020, 205, 2707–2718. [Google Scholar] [CrossRef]
  166. Patten, D.A.; Hussein, E.; Davies, S.P.; Humphreys, P.N.; Collett, A. Commensal-Derived OMVs Elicit a Mild Proinflammatory Response in Intestinal Epithelial Cells. Microbiology 2017, 163, 702–711. [Google Scholar] [CrossRef]
  167. Cañas, M.A.; Fábrega, M.J.; Giménez, R.; Badia, J.; Baldomà, L. Outer Membrane Vesicles From Probiotic and Commensal Escherichia Coli Activate NOD1-Mediated Immune Responses in Intestinal Epithelial Cells. Front. Microbiol. 2018, 9, 498. [Google Scholar] [CrossRef] [Green Version]
  168. Moosavi, S.M.; Akhavan Sepahi, A.; Mousavi, S.F.; Vaziri, F.; Siadat, S.D. The Effect of Faecalibacterium Prausnitzii and Its Extracellular Vesicles on the Permeability of Intestinal Epithelial Cells and Expression of PPARs and ANGPTL4 in the Caco-2 Cell Culture Model. J. Diabetes Metab. Disord. 2020, 19, 1061–1069. [Google Scholar] [CrossRef]
  169. Hickey, C.A.; Kuhn, K.A.; Donermeyer, D.L.; Porter, N.T.; Jin, C.; Cameron, E.A.; Jung, H.; Kaiko, G.E.; Wegorzewska, M.; Malvin, N.P.; et al. Colitogenic Bacteroides Thetaiotaomicron Antigens Access Host Immune Cells in a Sulfatase-Dependent Manner via Outer Membrane Vesicles. Cell Host Microbe 2015, 17, 672–680. [Google Scholar] [CrossRef] [Green Version]
  170. Chelakkot, C.; Choi, Y.; Kim, D.K.; Park, H.T.; Ghim, J.; Kwon, Y.; Jeon, J.; Kim, M.S.; Jee, Y.K.; Gho, Y.S.; et al. Akkermansia Muciniphila-Derived Extracellular Vesicles Influence Gut Permeability through the Regulation of Tight Junctions. Exp. Mol. Med. 2018, 50, e450. [Google Scholar] [CrossRef] [Green Version]
  171. Ashrafian, F.; Shahriary, A.; Behrouzi, A.; Moradi, H.R.; Keshavarz Azizi Raftar, S.; Lari, A.; Hadifar, S.; Yaghoubfar, R.; Ahmadi Badi, S.; Khatami, S.; et al. Akkermansia Muciniphila-Derived Extracellular Vesicles as a Mucosal Delivery Vector for Amelioration of Obesity in Mice. Front. Microbiol. 2019, 10, 2155. [Google Scholar] [CrossRef] [PubMed]
  172. Ashrafian, F.; Keshavarz Azizi Raftar, S.; Lari, A.; Shahryari, A.; Abdollahiyan, S.; Moradi, H.R.; Masoumi, M.; Davari, M.; Khatami, S.; Omrani, M.D.; et al. Extracellular Vesicles and Pasteurized Cells Derived from Akkermansia Muciniphila Protect against High-Fat Induced Obesity in Mice. Microb. Cell Fact. 2021, 20, 219. [Google Scholar] [CrossRef] [PubMed]
  173. Lee, K.E.; Kim, J.K.; Han, S.K.; Lee, D.Y.; Lee, H.J.; Yim, S.V.; Kim, D.H. The Extracellular Vesicle of Gut Microbial Paenalcaligenes Hominis Is a Risk Factor for Vagus Nerve-Mediated Cognitive Impairment. Microbiome 2020, 8, 107. [Google Scholar] [CrossRef] [PubMed]
  174. Seyama, M.; Yoshida, K.; Yoshida, K.; Fujiwara, N.; Ono, K.; Eguchi, T.; Kawai, H.; Guo, J.; Weng, Y.; Haoze, Y.; et al. Outer Membrane Vesicles of Porphyromonas Gingivalis Attenuate Insulin Sensitivity by Delivering Gingipains to the Liver. Biochim. Biophys. Acta Mol. Basis Dis. 2020, 1866, 165731. [Google Scholar] [CrossRef] [PubMed]
  175. Choi, J.W.; Kim, S.C.; Hong, S.H.; Lee, H.J. Secretable Small RNAs via Outer Membrane Vesicles in Periodontal Pathogens. J. Dent. Res. 2017, 96, 458–466. [Google Scholar] [CrossRef] [PubMed]
  176. Han, E.-C.; Choi, S.-Y.; Lee, Y.; Park, J.-W.; Hong, S.-H.; Lee, H.-J. Extracellular RNAs in Periodontopathogenic Outer Membrane Vesicles Promote TNF-a Production in Human Macrophages and Cross the Blood-Brain Barrier in Mice. FASEB J. 2019, 33, 13412–13422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Gao, H.; Jin, Z.; Tang, K.; Ji, Y.; Suarez, J.; Suarez, J.A.; e Rocha, K.C.; Zhang, D.; Dillmann, W.H.; Mahata, S.K.; et al. Microbial DNA Enrichment Promotes Adrenomedullary Inflammation, Catecholamine Secretion, and Hypertension in Obese Mice. J. Am. Heart Assoc. 2022, 11, e024561. [Google Scholar] [CrossRef]
  178. Luo, Z.; Ji, Y.; Zhang, D.; Gao, H.; Jin, Z.; Yang, M.; Ying, W. Microbial DNA Enrichment Promotes Liver Steatosis and Fibrosis in the Course of Non-Alcoholic Steatohepatitis. Acta Physiol. 2022, 235, e13827. [Google Scholar] [CrossRef]
  179. Luo, Z.; Ji, Y.; Gao, H.; Gomes Dos Reis, F.C.; Bandyopadhyay, G.; Jin, Z.; Ly, C.; Chang, Y.J.; Zhang, D.; Kumar, D.; et al. CRIg + Macrophages Prevent Gut Microbial DNA-Containing Extracellular Vesicle-Induced Tissue Inflammation and Insulin Resistance. Gastroenterololy 2021, 160, 863–874. [Google Scholar] [CrossRef]
  180. Gao, H.; Luo, Z.; Ji, Y.; Tang, K.; Jin, Z.; Ly, C.; Sears, D.D.; Mahata, S.; Ying, W. Accumulation of Microbial DNAs Promotes to Islet Inflammation and β Cell Abnormalities in Obesity in Mice. Nat. Commun. 2022, 13, 565. [Google Scholar] [CrossRef]
  181. Choi, J.H.; Moon, C.M.; Shin, T.S.; Kim, E.K.; McDowell, A.; Jo, M.K.; Joo, Y.H.; Kim, S.E.; Jung, H.K.; Shim, K.N.; et al. Lactobacillus Paracasei-Derived Extracellular Vesicles Attenuate the Intestinal Inflammatory Response by Augmenting the Endoplasmic Reticulum Stress Pathway. Exp. Mol. Med. 2020, 52, 423–437. [Google Scholar] [CrossRef] [PubMed]
  182. Vargoorani, M.E.; Modarressi, M.H.; Vaziri, F.; Motevaseli, E.; Siadat, S.D. Stimulatory Effects of Lactobacillus Casei Derived Extracellular Vesicles on Toll-like Receptor 9 Gene Expression and Cytokine Profile in Human Intestinal Epithelial Cells. J. Diabetes Metab. Disord. 2020, 19, 223–231. [Google Scholar] [CrossRef] [PubMed]
  183. Ghaderi, F.; Sotoodehnejadnematalahi, F.; Hajebrahimi, Z.; Fateh, A.; Siadat, S.D. Effects of Active, Inactive, and Derivatives of Akkermansia Muciniphila on the Expression of the Endocannabinoid System and PPARs Genes. Sci. Rep. 2022, 12, 12301. [Google Scholar] [CrossRef] [PubMed]
  184. Kim, K.; Lee, S.; Park, S.C.; Kim, N.E.; Shin, C.; Lee, S.K.; Jung, Y.; Yoon, D.; Kim, H.; Kim, S.; et al. Role of an Unclassified Lachnospiraceae in the Pathogenesis of Type 2 Diabetes: A Longitudinal Study of the Urine Microbiome and Metabolites. Exp. Mol. Med. 2022, 54, 1125–1132. [Google Scholar] [CrossRef]
  185. Nah, G.; Park, S.C.; Kim, K.; Kim, S.; Park, J.; Lee, S.; Won, S. Type-2 Diabetics Reduces Spatial Variation of Microbiome Based on Extracellur Vesicles from Gut Microbes across Human Body. Sci. Rep. 2019, 9, 20136. [Google Scholar] [CrossRef] [Green Version]
  186. Cerdó, T.; García-Santos, J.A.; Bermúdez, M.G.; Campoy, C. The Role of Probiotics and Prebiotics in the Prevention and Treatment of Obesity. Nutrients 2019, 11, 635. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Park, S.; Ji, Y.; Jung, H.Y.; Park, H.; Kang, J.; Choi, S.H.; Shin, H.; Hyun, C.K.; Kim, K.T.; Holzapfel, W.H. Lactobacillus Plantarum HAC01 Regulates Gut Microbiota and Adipose Tissue Accumulation in a Diet-Induced Obesity Murine Model. Appl. Microbiol. Biotechnol. 2017, 101, 1605–1614. [Google Scholar] [CrossRef]
  188. Huber-Ruano, I.; Calvo, E.; Mayneris-Perxachs, J.; Rodríguez-Peña, M.-M.; Ceperuelo-Mallafré, V.; Cedó, L.; Núñez-Roa, C.; Miro-Blanch, J.; Arnoriaga-Rodríguez, M.; Balvay, A.; et al. Orally Administered Odoribacter Laneus Improves Glucose Control and Inflammatory Profile in Obese Mice by Depleting Circulating Succinate. Microbiome 2021, 10, 135. [Google Scholar] [CrossRef]
  189. Çelik, M.N.; Ünlü Söğüt, M. Probiotics Improve Chemerin Levels and Metabolic Syndrome Parameters in Obese Rats. Balkan Med. J. 2019, 36, 270–275. [Google Scholar]
  190. Rodovalho, V.D.R.; Luz, B.S.R.D.; Rabah, H.; do Carmo, F.L.R.; Folador, E.L.; Nicolas, A.; Jardin, J.; Briard-Bion, V.; Blottière, H.; Lapaque, N.; et al. Extracellular Vesicles Produced by the Probiotic Propionibacterium Freudenreichii CIRM-BIA 129 Mitigate Inflammation by Modulating the NF-ΚB Pathway. Front. Microbiol. 2020, 11, 1544. [Google Scholar] [CrossRef]
  191. Kim, M.H.; Choi, S.J.; Choi, H.I.; Choi, J.P.; Park, H.K.; Kim, E.K.; Kim, M.J.; Moon, B.S.; Min, T.K.; Rho, M.; et al. Lactobacillus Plantarum-Derived Extracellular Vesicles Protect Atopic Dermatitis Induced by Staphylococcus Aureus-Derived Extracellular Vesicles. Allergy Asthma Immunol. Res. 2018, 10, 516–532. [Google Scholar] [CrossRef]
  192. Müller, L.; Kuhn, T.; Koch, M.; Fuhrmann, G. Stimulation of Probiotic Bacteria Induces Release of Membrane Vesicles with Augmented Anti-Inflammatory Activity. ACS Appl. Bio. Mater. 2021, 4, 3739–3748. [Google Scholar] [CrossRef] [PubMed]
  193. Tandon, D.; Haque, M.M.; Gote, M.; Jain, M.; Bhaduri, A.; Dubey, A.K.; Mande, S.S. A Prospective Randomized, Double-Blind, Placebo-Controlled, Dose-Response Relationship Study to Investigate Efficacy of Fructo-Oligosaccharides (FOS) on Human Gut Microflora. Sci. Rep. 2019, 9, 5473. [Google Scholar] [CrossRef] [PubMed]
  194. Wiese, M.; Bashmakov, Y.; Chalyk, N.; Nielsen, D.S.; Krych, Ł.; Kot, W.; Klochkov, V.; Pristensky, D.; Bandaletova, T.; Chernyshova, M.; et al. Prebiotic Effect of Lycopene and Dark Chocolate on Gut Microbiome with Systemic Changes in Liver Metabolism, Skeletal Muscles and Skin in Moderately Obese Persons. Biomed. Res. Int. 2019, 2019, 4625279. [Google Scholar] [CrossRef] [PubMed]
  195. Pérez-Bermúdez, P.; Blesa, J.; Soriano, J.M.; Marcilla, A. Extracellular Vesicles in Food: Experimental Evidence of Their Secretion in Grape Fruits. Eur. J. Pharm. Sci. 2017, 98, 40–50. [Google Scholar] [CrossRef] [PubMed]
  196. Rome, S. Biological Properties of Plant-Derived Extracellular Vesicles. Food Funct. 2019, 10, 529–538. [Google Scholar] [CrossRef]
  197. Tomé-Carneiro, J.; Fernández-Alonso, N.; Tomás-Zapico, C.; Visioli, F.; Iglesias-Gutierrez, E.; Dávalos, A. Breast Milk MicroRNAs Harsh Journey towards Potential Effects in Infant Development and Maturation. Lipid Encapsulation Can Help. Pharmacol. Res. 2018, 132, 21–32. [Google Scholar] [CrossRef]
  198. Feng, X.; Chen, X.; Zheng, X.; Zhu, H.; Qi, Q.; Liu, S.; Zhang, H.; Che, J. Latest Trend of Milk Derived Exosomes: Cargos, Functions, and Applications. Front. Nutr. 2021, 8, 855. [Google Scholar] [CrossRef]
  199. Salem, M.; Rohani, S.; Gillies, E.R. Curcumin, a Promising Anti-Cancer Therapeutic: A Review of Its Chemical Properties, Bioactivity and Approaches to Cancer Cell Delivery. RSC Adv. 2014, 4, 10815. [Google Scholar] [CrossRef]
  200. Wolf, T.; Baier, S.R.; Zempleni, J. The Intestinal Transport of Bovine Milk Exosomes Is Mediated by Endocytosis in Human Colon Carcinoma Caco-2 Cells and Rat Small Intestinal IEC-6 Cells. J. Nutr. 2015, 145, 2201–2206. [Google Scholar] [CrossRef] [Green Version]
  201. Munir, J.; Lee, M.; Ryu, S. Exosomes in Food: Health Benefits and Clinical Relevance in Diseases. Adv. Nutr. 2020, 11, 687–696. [Google Scholar] [CrossRef] [PubMed]
  202. Manca, S.; Upadhyaya, B.; Mutai, E.; Desaulniers, A.T.; Cederberg, R.A.; White, B.R.; Zempleni, J. Milk Exosomes Are Bioavailable and Distinct MicroRNA Cargos Have Unique Tissue Distribution Patterns. Sci. Rep. 2018, 8, 11321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Zhuang, X.; Deng, Z.B.; Mu, J.; Zhang, L.; Yan, J.; Miller, D.; Feng, W.; McClain, C.J.; Zhang, H.G. Ginger-Derived Nanoparticles Protect against Alcohol-Induced Liver Damage. J. Extracell. Vesicles 2015, 4, 28713. [Google Scholar] [CrossRef] [PubMed]
  204. Zhang, M.; Viennois, E.; Prasad, M.; Zhang, Y.; Wang, L.; Zhang, Z.; Han, M.K.; Xiao, B.; Xu, C.; Srinivasan, S.; et al. Edible Ginger-Derived Nanoparticles: A Novel Therapeutic Approach for the Prevention and Treatment of Inflammatory Bowel Disease and Colitis-Associated Cancer. Biomaterials 2016, 101, 321–340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Tong, L.; Hao, H.; Zhang, X.; Zhang, Z.; Lv, Y.; Zhang, L.; Yi, H. Oral Administration of Bovine Milk-Derived Extracellular Vesicles Alters the Gut Microbiota and Enhances Intestinal Immunity in Mice. Mol. Nutr. Food Res. 2020, 64, 1901251. [Google Scholar] [CrossRef]
  206. 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]
  207. Melnik, B.C.; Schmitz, G. Exosomes of Pasteurized Milk: Potential Pathogens of Western Diseases. J. Transl. Med. 2019, 17, 3. [Google Scholar] [CrossRef] [Green Version]
  208. Mirza, A.H.; Kaur, S.; Nielsen, L.B.; Størling, J.; Yarani, R.; Roursgaard, M.; Mathiesen, E.R.; Damm, P.; Svare, J.; Mortensen, H.B.; et al. Breast Milk-Derived Extracellular Vesicles Enriched in Exosomes From Mothers With Type 1 Diabetes Contain Aberrant Levels of MicroRNAs. Front. Immunol. 2019, 10, 2543. [Google Scholar] [CrossRef]
  209. Shah, K.B.; Chernausek, S.D.; Garman, L.D.; Pezant, N.P.; Plows, J.F.; Kharoud, H.K.; Demerath, E.W.; Fields, D.A. Human Milk Exosomal MicroRNA: Associations with Maternal Overweight/Obesity and Infant Body Composition at 1 Month of Life. Nutrients 2021, 13, 1091. [Google Scholar] [CrossRef]
  210. Shah, K.B.; Fields, D.A.; Pezant, N.P.; Kharoud, H.K.; Gulati, S.; Jacobs, K.; Gale, C.A.; Kharbanda, E.O.; Nagel, E.M.; Demerath, E.W.; et al. Gestational Diabetes Mellitus Is Associated with Altered Abundance of Exosomal MicroRNAs in Human Milk. Clin. Ther. 2022, 44, 172–185. [Google Scholar] [CrossRef]
  211. 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] [PubMed]
  212. Xu, M.; Chen, G.; Dong, Y.; Yang, J.; Liu, Y.; Song, H.; Song, H.; Wang, Y. Liraglutide-Loaded Milk Exosomes Lower Blood Glucose When Given by Sublingual Route. Chem. Med. Chem. 2022, 17, e202100758. [Google Scholar] [CrossRef] [PubMed]
  213. Timms, K.; Holder, B.; Day, A.; Mclaughlin, J.; Forbes, K.A.; Westwood, M. Watermelon-Derived Extracellular Vesicles Influence Human Ex Vivo Placental Cell Behavior by Altering Intestinal Secretions. Mol. Nutr. Food Res. 2022, 66, 2200013. [Google Scholar] [CrossRef] [PubMed]
  214. Díez-Sainz, E.; Lorente-Cebrián, S.; Aranaz, P.; Riezu-Boj, J.I.; Martínez, J.A.; Milagro, F.I. Potential Mechanisms Linking Food-Derived MicroRNAs, Gut Microbiota and Intestinal Barrier Functions in the Context of Nutrition and Human Health. Front. Nutr. 2021, 8, 586564. [Google Scholar] [CrossRef]
  215. Berger, E.; Colosetti, P.; Jalabert, A.; Meugnier, E.; Wiklander, O.P.B.; Jouhet, J.; Errazurig-Cerda, E.; Chanon, S.; Gupta, D.; Rautureau, G.J.P.; et al. Use of Nanovesicles from Orange Juice to Reverse Diet-Induced Gut Modifications in Diet-Induced Obese Mice. Mol. Ther. Methods Clin. Dev. 2020, 18, 880–892. [Google Scholar] [CrossRef]
  216. Chen, X.; Zhou, Y.; Yu, J. Exosome-like Nanoparticles from Ginger Rhizomes Inhibited NLRP3 Inflammasome Activation. Mol. Pharm. 2019, 16, 2690–2699. [Google Scholar] [CrossRef]
  217. 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] [Green Version]
  218. Kumar, A.; Ren, Y.; Sundaram, K.; Mu, J.; Sriwastva, M.K.; Dryden, G.W.; Lei, C.; Zhang, L.; Yan, J.; Zhang, X.; et al. MiR-375 Prevents High-Fat Diet-Induced Insulin Resistance and Obesity by Targeting the Aryl Hydrocarbon Receptor and Bacterial Tryptophanase (TnaA) Gene. Theranostics 2021, 11, 4061–4077. [Google Scholar] [CrossRef]
  219. Taşlı, P.N. Usage of Celery Root Exosome as an Immune Suppressant; Lipidomic Characterization of Apium Graveolens Originated Exosomes and Its Suppressive Effect on PMA/Ionomycin Mediated CD4+ T Lymphocyte Activation. J. Food Biochem. 2022, 46, e14393. [Google Scholar] [CrossRef]
  220. Aquilano, K.; Ceci, V.; Gismondi, A.; de Stefano, S.; Iacovelli, F.; Faraonio, R.; di Marco, G.; Poerio, N.; Minutolo, A.; Minopoli, G.; et al. Adipocyte Metabolism Is Improved by TNF Receptor-Targeting Small RNAs Identified from Dried Nuts. Commun. Biol. 2019, 2, 317. [Google Scholar] [CrossRef] [Green Version]
  221. Antimisiaris, S.G.; Mourtas, S.; Marazioti, A. Exosomes and Exosome-Inspired Vesicles for Targeted Drug Delivery. Pharm. 2018, 10, 218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Liang, Y.; Duan, L.; Lu, J.; Xia, J. Engineering Exosomes for Targeted Drug Delivery. Theranostics 2021, 11, 3183–3195. [Google Scholar] [CrossRef] [PubMed]
  223. Tulkens, J.; de Wever, O.; Hendrix, A. Analyzing Bacterial Extracellular Vesicles in Human Body Fluids by Orthogonal Biophysical Separation and Biochemical Characterization. Nat. Protoc. 2020, 15, 40–67. [Google Scholar] [CrossRef]
  224. Nunez Lopez, Y.O.; Iliuk, A.; Petrilli, A.M.; Glass, C.; Casu, A.; Pratley, R.E. Proteomics and Phosphoproteomics of Circulating Extracellular Vesicles Provide New Insights into Diabetes Pathobiology. Int. J. Mol. Sci. 2022, 23, 5779. [Google Scholar] [CrossRef] [PubMed]
  225. Castaño, C.; Meza-Ramos, A.; Batlle, M.; Guasch, E.; Novials, A.; Párrizas, M. Treatment with EV-MiRNAs Alleviates Obesity-Associated Metabolic Dysfunction in Mice. Int. J. Mol. Sci. 2022, 23, 14920. [Google Scholar] [CrossRef] [PubMed]
  226. Keshtkar, S.; Azarpira, N.; Ghahremani, M.H. Mesenchymal Stem Cell-Derived Extracellular Vesicles: Novel Frontiers in Regenerative Medicine. Stem. Cell Res. Ther. 2018, 9, 63. [Google Scholar] [CrossRef]
  227. Thakral, S.; Ghoshal, K. MiR-122 Is a Unique Molecule with Great Potential in Diagnosis, Prognosis of Liver Disease, and Therapy Both as MiRNA Mimic and Antimir. Curr. Gene Ther. 2015, 15, 142–150. [Google Scholar] [CrossRef] [Green Version]
  228. Krützfeldt, J.; Rajewsky, N.; Braich, R.; Rajeev, K.G.; Tuschl, T.; Manoharan, M.; Stoffel, M. Silencing of MicroRNAs in Vivo with Antagomirs. Nature 2005, 438, 685–689. [Google Scholar] [CrossRef]
  229. Mahesh, G.; Biswas, R. MicroRNA-155: A Master Regulator of Inflammation. J. Interferon Cytokine Res. 2019, 39, 321–330. [Google Scholar] [CrossRef]
  230. Yuan, X.; Berg, N.; Lee, J.W.; Le, T.-T.; Neudecker, V.; Jing, N.; Eltzschig, H. MicroRNA MiR-223 as Regulator of Innate Immunity. J. Leukoc. Biol. 2018, 104, 515–524. [Google Scholar] [CrossRef]
  231. Zhang, M.W.; Shen, Y.J.; Shi, J.; Yu, J.G. MiR-223-3p in Cardiovascular Diseases: A Biomarker and Potential Therapeutic Target. Front. Cardiovasc. Med. 2021, 7, 422. [Google Scholar] [CrossRef] [PubMed]
  232. Ye, D.; Zhang, T.; Lou, G.; Liu, Y. Role of MiR-223 in the Pathophysiology of Liver Diseases. Exp. Mol. Med. 2018, 50, 128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Aziz, F. The Emerging Role of MiR-223 as Novel Potential Diagnostic and Therapeutic Target for Inflammatory Disorders. Cell Immunol. 2016, 303, 1–6. [Google Scholar] [CrossRef]
  234. Parrizas, M.; Mundet, X.; Castano, C.; Canivell, S.; Cos, X.; Brugnara, L.; Giraldez-Garcia, C.; Regidor, E.; Mata-Cases, M.; Franch-Nadal, J.; et al. MiR-10b and MiR-223-3p in Serum Microvesicles Signal Progression from Prediabetes to Type 2 Diabetes. J. Endocrinol. Invest. 2020, 43, 451–459. [Google Scholar] [CrossRef] [PubMed]
  235. Liu, Y.; Li, D.; Liu, Z.; Zhou, Y.; Chu, D.; Li, X.; Jiang, X.; Hou, D.; Chen, X.; Chen, Y.; et al. Targeted Exosome-Mediated Delivery of Opioid Receptor Mu SiRNA for the Treatment of Morphine Relapse. Sci. Rep. 2015, 5, 17543. [Google Scholar] [CrossRef] [Green Version]
  236. Liu, S.; da Cunha, A.P.; Rezende, R.M.; Cialic, R.; Wei, Z.; Bry, L.; Comstock, L.E.; Gandhi, R.; Weiner, H.L. The Host Shapes the Gut Microbiota via Fecal MicroRNA. Cell Host Microbe 2016, 19, 32–43. [Google Scholar] [CrossRef]
Figure 1. EV biogenesis in plants, mammals, and bacteria. Aside from apoptotic bodies generated upon programmed cell death, plant and mammalian cells release two main types of EVs: MVs resulting from plasma membrane evaginations and exosomes formed when the membrane of early endosomes invaginates in the intracellular space, engulfing a portion of the cytoplasm and giving rise to a multivesicular body (MVB) that may then fuse with lysosomes to recycle cellular components or with the plasma membrane, hence releasing the exosomes to the extracellular space. Alternatively, Gram-negative bacteria have two routes for bEV formation: blebbing of the outer membrane caused by cell cover disturbances leads to the production of OMVs that do not contain cytoplasmic components, whereas explosive cell lysis initiated by phage-derived endolysin that degrades the peptidoglycan cell wall results in cells bursting and releasing membrane fragments that self-assemble into OIMVs, which contain cytoplasmic components. In Gram-positive bacteria, endolysin triggers bubbling cell death, which is induced by DNA-damaging stress and results in CMVs that contain membrane and cytoplasmic components.
Figure 1. EV biogenesis in plants, mammals, and bacteria. Aside from apoptotic bodies generated upon programmed cell death, plant and mammalian cells release two main types of EVs: MVs resulting from plasma membrane evaginations and exosomes formed when the membrane of early endosomes invaginates in the intracellular space, engulfing a portion of the cytoplasm and giving rise to a multivesicular body (MVB) that may then fuse with lysosomes to recycle cellular components or with the plasma membrane, hence releasing the exosomes to the extracellular space. Alternatively, Gram-negative bacteria have two routes for bEV formation: blebbing of the outer membrane caused by cell cover disturbances leads to the production of OMVs that do not contain cytoplasmic components, whereas explosive cell lysis initiated by phage-derived endolysin that degrades the peptidoglycan cell wall results in cells bursting and releasing membrane fragments that self-assemble into OIMVs, which contain cytoplasmic components. In Gram-positive bacteria, endolysin triggers bubbling cell death, which is induced by DNA-damaging stress and results in CMVs that contain membrane and cytoplasmic components.
Ijms 24 02071 g001
Figure 2. EVs and their cargo participate in inter-organ crosstalk to regulate metabolic homeostasis. EVs released by many cell types can be captured by acceptor cells in an autocrine and paracrine manner, initiating biological responses. In addition, EVs can go into the circulation and reach other organs, driving inter-organ crosstalk. M1: M1 macrophages; M2: M2 macrophages. See text for details.
Figure 2. EVs and their cargo participate in inter-organ crosstalk to regulate metabolic homeostasis. EVs released by many cell types can be captured by acceptor cells in an autocrine and paracrine manner, initiating biological responses. In addition, EVs can go into the circulation and reach other organs, driving inter-organ crosstalk. M1: M1 macrophages; M2: M2 macrophages. See text for details.
Ijms 24 02071 g002
Figure 3. Impact of dietary patterns on gut microbiota composition and bEV profile and its effects on metabolic diseases. Unhealthy dietary patterns contribute to obesity and T2D by promoting gut microbiota dysbiosis. The increase in detrimental bEVs may then affect gut barrier permeability and intestinal inflammation. After gaining access to the systemic circulation, bEVs may disturb peripheral organ function, thus impairing metabolic homeostasis and promoting inflammation, thereby increasing the severity of metabolic disorders. Conversely, an increase of beneficial bEVs through a plant-based diet or probiotic supplements could be a strategy to treat metabolic disorders by counteracting dysbiosis, gut permeability, inflammation, metabolic homeostasis disturbances, and nervous system imbalance. See text for details.
Figure 3. Impact of dietary patterns on gut microbiota composition and bEV profile and its effects on metabolic diseases. Unhealthy dietary patterns contribute to obesity and T2D by promoting gut microbiota dysbiosis. The increase in detrimental bEVs may then affect gut barrier permeability and intestinal inflammation. After gaining access to the systemic circulation, bEVs may disturb peripheral organ function, thus impairing metabolic homeostasis and promoting inflammation, thereby increasing the severity of metabolic disorders. Conversely, an increase of beneficial bEVs through a plant-based diet or probiotic supplements could be a strategy to treat metabolic disorders by counteracting dysbiosis, gut permeability, inflammation, metabolic homeostasis disturbances, and nervous system imbalance. See text for details.
Ijms 24 02071 g003
Figure 4. EVs found in edible vegetables or milk, or released by the gut microbiota and the different organs, participate in a multi-directional, inter-kingdom crosstalk that is required to coordinate metabolic processes and maintain homeostasis in the host. See text for details.
Figure 4. EVs found in edible vegetables or milk, or released by the gut microbiota and the different organs, participate in a multi-directional, inter-kingdom crosstalk that is required to coordinate metabolic processes and maintain homeostasis in the host. See text for details.
Ijms 24 02071 g004
Table 1. Summary of the studies mentioned in the text dealing with EV-mediated inter-tissue communication. See text for details.
Table 1. Summary of the studies mentioned in the text dealing with EV-mediated inter-tissue communication. See text for details.
Source of EVsRelevant CargoTarget Cell/TissueEffectRef
ECsPlasma componentsAdipocytesSignaling[39]
AdipocytesSNHG9ECsAnti-inflammation[40]
AdipocytesmiR-221-3pVSMCsVascular remodeling[42]
AdipocytesmiR-122PreadipocytesAdipogenesis[43]
AdipocytesmiR-3604PreadipocytesReduced adipogenesis[45]
AdipocytesmiR-34,
miR-1224
MacrophagesM2 polarization inhibition[46,47]
AdipocytesNDMacrophagesFoam cell formation[48]
AdipocytesmiR-222,
miR-27a
MyocytesLower insulin sensitivity[38,49]
AdipocytesMitochondriaCardiomyocytesAntioxidant[50]
AdipocytesAdipsinCardiomyocytesAttenuated remodeling[51]
Adipocyteslet-7d-3pMuscle stem cellsDecreased proliferation[52]
AdipocytesmiR-199a,
miR-141-3p
HepatocytesHepatic steatosis and lower insulin sensitivity[53,54]
AdipocytesmiR-9-3pBrainCognitive impairment[55]
Brown adipocytesNPM3WATBrowning[56]
Brown adipocytesmiR-30bKidneyReduced fibrosis[57]
Adipose MSCsSTAT3MacrophagesM2 polarization[58]
Adipose MSCsOPGMacrophagesDecreased osteoclast differentiation[59]
Adipose MSCsmiR-627,
miR-223-3p
HepatocytesDecreased steatosis[60]
Adipose MSCsmiR-21-3p,
miR-126
FibroblastsAccelerated wound healing[61,62]
M1 macrophagesmiR-155,
miR-29a
Adipocytes, hepatocytes, myocytesLower insulin sensitivity[36,63]
M1 macrophagesmiR-212,
miR-155
Pancreatic β-cellsDecreased insulin secretion[64,65]
M2 macrophagesmiR-690Adipocytes, hepatocytes, myocytesHigher insulin sensitivity[35]
MusclemiR-133bLiverHigher insulin sensitivity[66]
MusclemiR-1WATIncreased lipolysis[67]
MusclemiR-191a,
miR-27a-3p
WATBrowning[68]
MusclemiR-690Muscle stem cellsInhibition of differentiation[69]
MusclemiR-16Pancreatic β-cellsIncreased proliferation[70]
LivermiR-130a-3pAdipocytesHigher insulin sensitivity[71]
LiverTM4SF5Brown adipocytesHigher insulin sensitivity[72]
LivermiR-3075Adipocytes, hepatocytes, myocytesHigher insulin sensitivity[73]
LivermiR-122CardiomyocytesImpaired mitochondrial function[74]
LivermiR-122ECsAngiogenesis[75]
HepatocytesmiR-122MacrophagesIncreased inflammation[76]
Hepatocyteslet−7bBrown adipocytesDecreased thermogenesis[77]
NeutrophilmiR-223-3pHepatocytesDecreased inflammation and hepatic steatosis[78]
Pancreatic β-cellsmiR-26aPancreatic β-cellsImproved insulin secretion[79]
Pancreatic β-cellsIAPPHippocampusMitochondrial fission[80]
T lymphocytesmiR-155Pancreatic β-cellsApoptosis[81]
Table 2. Summary of the studies mentioned in the text dealing with bacteria-host EV-mediated inter-kingdom communication. See text for details.
Table 2. Summary of the studies mentioned in the text dealing with bacteria-host EV-mediated inter-kingdom communication. See text for details.
Source of EVsRelevant CargoTarget Cell/TissueEffectRef
A. muciniphilaNDIECsAnti-inflammatory[159]
A. muciniphilaNDIECsGut fortification[170]
A. muciniphilaNDLiver, ATAnti-obesity[171,172]
A. muciniphila, EcN1917NDGutColitis protection[162,163]
O. splanchnicusNDIECsAnti-inflammatory[160]
B. FragilisPSADCsColitis protection[164]
B. thetaiotaomicronanSMEGutColitis[169]
P. hominisLPSHippocampusCognitive impairment[173]
P. panacisLPSAdipose, muscleInsulin resistance[157]
P. pentosaceusNDMacrophagesImmuno-suppression[165]
P. gingivalisProteasesLiverImpaired glucose metabolism[174]
P. gingivalis, T. denticola, A. actinomycetemcomitansmiRNAsT cellsAnti-inflammatory[175]
A. actinomycetemcomitansmiRNA-likeMacrophagesNeuro-inflammatory[176]
E. coliC25NDIECsPro-inflammatory[166]
EcN1917, ECOR12NDIECsPro-inflammatory[167]
EcN1917, ECOR63NDIECsAnti-inflammatory[161]
F. prausnitziiNDIECsPro-inflammatory[168]
GutmbDNAMacrophages, Adrenal cellsHypertension[177]
GutmbDNAHepatocytesLiver fibrosis[178]
GutmbDNALiver, adipose, musclePro-inflammatory[179]
GutDNAβ-cellInsulin resistance[180]
L. paracaseiNDIECsColitis protection[181]
L. caseiNDIECsAnti-inflammatory[182]
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

Castaño, C.; Novials, A.; Párrizas, M. An Overview of Inter-Tissue and Inter-Kingdom Communication Mediated by Extracellular Vesicles in the Regulation of Mammalian Metabolism. Int. J. Mol. Sci. 2023, 24, 2071. https://doi.org/10.3390/ijms24032071

AMA Style

Castaño C, Novials A, Párrizas M. An Overview of Inter-Tissue and Inter-Kingdom Communication Mediated by Extracellular Vesicles in the Regulation of Mammalian Metabolism. International Journal of Molecular Sciences. 2023; 24(3):2071. https://doi.org/10.3390/ijms24032071

Chicago/Turabian Style

Castaño, Carlos, Anna Novials, and Marcelina Párrizas. 2023. "An Overview of Inter-Tissue and Inter-Kingdom Communication Mediated by Extracellular Vesicles in the Regulation of Mammalian Metabolism" International Journal of Molecular Sciences 24, no. 3: 2071. https://doi.org/10.3390/ijms24032071

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