Abstract
Recent studies have demonstrated that extracellular vesicles (EVs) serve powerful and complex functions in metabolic regulation and metabolic-associated disease, although this field of research is still in its infancy. EVs are released into the extracellular space from all cells and carry a wide range of cargo including miRNAs, mRNA, DNA, proteins and metabolites that have robust signaling effects in receiving cells. EV production is stimulated by all major stress pathways and, as such, has a role in both restoring homeostasis during stress and perpetuating disease. In metabolic regulation the dominant stress signal is a lack of energy due either nutrient deficits or damaged mitochondria from nutrient excess. This stress signal is termed “energetic stress”, which triggers a robust and evolutionarily conserved response that engages major cellular stress pathways, the ER unfolded protein response, the hypoxia response, the antioxidant response and autophagy. This review proposes the model that energetic stress is the dominant stimulator of EV release with a focus on metabolically important cells such has hepatocytes, adipocyte, myocytes and pancreatic β-cells. Furthermore, this review will discuss how the cargo in stress-stimulated EVs regulate metabolism in receiving cells in both beneficial and detrimental ways.
Introduction
Recognizing the complexity of inter-cellular regulation of metabolism
Metabolism in multicellular organisms is tightly regulated through the concerted actions of multiple specialized cells. In vertebrates these specialized cells make up organ systems that preform specific functions that benefit the organism. For example, adipocytes store excess nutrients for future release in times of sparsity, hepatocytes package lipids from the diet into a usable form for other tissues and produce glucose to sustain the energetic requirements of the organism during fasting, and collections of neurons in the hypothalamus regulate feeding behavior and energy expenditure. Although the simplistic goal of metabolism-regulating mechanisms is to provide all cells in the organism with adequate fuel to carry out their functions, accomplishing this goal is a highly complex endeavor. Energy supplying organs like adipose tissue, liver and hypothalamus must be able to match energy output with the energy demand in the system. This includes sensing shifts in energy expenditure caused by energetically expensive processes like exercise, reproduction, and illness and responding by altering feeding behavior, blood nutrient availability and basal metabolic rate. The inter-organ regulation of metabolism is traditionally described by the release of hormones from multiple organs with receptor-mediated effects on metabolism in hormone-sensitive cells. The classic endocrine organs that secrete metabolism-regulating hormones are the thyroid gland, adrenal gland and pancreas, but other organs such as the adipose tissue and gastrointestinal tract also secrete an impressive array of hormones. Adipose tissue releases adipokines that span protein hormones, bioactive lipids and metabolites (1). The most well studied are leptin and adiponectin. Leptin is released from adipocytes in the fed state when adipocytes are actively storing energy. Its major action is on the brain to suppress food intake (1). Adiponectin is a strong insulin-sensitizing and anti-lipotoxic hormone that decreases gluconeogenesis in the liver and promotes fatty acid oxidation in skeletal muscle (1). Furthermore, in recent years the gastrointestinal tract has been shown to release over 20 peptide hormones such as ghrelin, glucose-dependent insulinotropic peptide (GIP) and glucagon-like peptides (GLPs; (2)). The release of these hormones is dictated by the gut nutrient composition and can act on various cell types to regulate gut motility, appetite control and maintenance of glucose homeostasis (2). Therefore, under any given physiological state there is a diverse composite of signaling molecules from various organs, that facilitate the tight regulation of systemic metabolism. Dysregulation of this inter-cellular communication can lead to obesity and metabolic syndrome or exacerbate already established metabolic disease (2–4).
The Immergence of Extracellular Vesicles in Metabolic Regulation: Compounding Complexity
Extracellular vesicles (EVs) are highly heterogeneous membranous vesicles released into the extracellular space by most cells. EVs carry signaling proteins, mRNA, DNA, miRNAs, bioactive lipids, and metabolites (5). These EVs can function both to release unwanted or damaged material from the cell, and to initiate communication with other cells (6). As would be expected from such a diverse collection of cargo, EVs can have a more powerful and global effect on cellular signaling than hormone-mediated signaling from a single receptor. To date, several types of EVs have been described and classified by their intracellular origins. The initial discoveries of EVs identified exosomes, microvesicles and apoptotic bodies (5). Exosomes are produced in the multivesicular endosome (MVE) by the budding of the limiting membrane to form intraluminal vesicles (ILVs). Fusion of the MVE with the plasma membrane releases these 30–150 nm ILVs into the extracellular space, which are then termed exosomes (5, 7). Microvesicles, also called ectosomes, are formed by the direct budding of the plasma membrane (ectocytosis; (8)) and tend to be much larger than exosomes (50nm-1000nm; (5)). Whereas exosomes and microvesicles are released from healthy cells, apoptotic bodies are produced at the plasma membrane by dying cells. Additional classes of EVs have recently been discovered to originate from the endolysosomal system including those produced by lysosomal exocytosis, and secretory autophagy (9–11). In recent years EVs have been demonstrated to regulate metabolism, opening a new field of research. EV signaling has been called the next frontier in endocrinology (12), representing an unexplored network of metabolic regulation that may act synergistically or independently of classic hormones.
Recent studies have demonstrated a link between soluble hormone action and EV production. The adipokines adiponectin and resistin have been detected in circulating EVs at trace amounts (13). There is some evidence that EV-associated resistin can have signaling effects despite being a small fraction of the total resistin levels in circulation (14). However, the adipokines adiponectin and leptin have also been found to directly regulation EV production. Obata et. al. demonstrated that circulating adiponectin binds to T-cadherin on the surface of endothelial cells and stimulates EV production and release (15). These EVs contain endocytosed adiponectin-T-cadherin complexes and are highly enriched for ceramides. The authors suggest this is a mechanism by which adiponectin acts to clear the endothelial cell of toxic ceramide species. Leptin has also been shown to stimulate sEV production in breast cancer cell lines (16). This occurs through the post-transcription upregulation of TSG101, a critical member of the endosomal sorting complex (ESCRT-1). Glucagon is one of few examples outside of adipokines, of the crosstalk between classic hormones and EVs. Glucagon stimulates endothelial cell endocytosis of BSA-bound cargo from circulation, incorporates these molecules into sEVs, and enhances sEV production (17). These studies open the exciting possibility of robust and intricate cross-talked between soluble hormones and EV-mediated signaling.
Although little is known about how EVs signal to regulate metabolism under physiological conditions, EVs clearly influence the pathogenesis of obesity, metabolic syndrome, and associated comorbidities (18, 19). In fact, EVs can modulate the pathology of a wide variety of diseases including cardiovascular disease, cancer, mental disorders, and neurodegeneration, each of which have a metabolic component (20–22). The reason that EVs are so active in disease likely stems from the inherent nature of EVs as stress-stimulated signaling mechanisms.
Stress-stimulated EV Production
Many cells release EVs basally, however, various cellular stresses result in higher EV release with altered cargo. For example, stresses such as inflammation, fatty acids, hypoxia, thermal stress, osmotic stress, oxidative stress, genotoxic stress, pH and nutrient deprivation all result in enhanced EV release and altered EV cargo in many cell types (23–31). Well defined cellular stress responses are induced by the above-mentioned stimuli: the unfolded protein response (UPR) to ER stress, the antioxidant response to oxidative stress, and hypoxia response to low oxygen tension. These stress pathways have been linked directly to altered EV production and/or composition. For example, endothelial cells treated with thapsigargin or dithiothreitol to directly induce ER stress did not display changes in microvesicle production, but microvesicles were able to induce ER stress in untreated endothelial cells (32). In cancer cells, chemical induction of ER stress results in higher EV release that carry pro-inflammatory signals (33, 34). Likewise, hepatocytes treated with palmitate, or thapsigargin released more EVs, which was dependent on key proteins in the ER stress signaling pathway, IRE1α and XPB1s (35). In the context of oxidative stress, pro-oxidant conditions result in enhanced small EV release in HEK293 cells and senescent cells altering the cargo to be pro-inflammatory and pro-proliferative respectively (36, 37). These effects could be prevented with the antioxidant molecule N-acetyl-cysteine (NAC). In adipocytes, oxidative stress induced by mitochondrial electron transport chain inhibitors significantly increased small EV release (38). This effect was not seen when adipocytes were treated with buthionine sulphoximine (BSO), a compound that causes oxidative stress by depleting cellular glutathione (38). This suggests that ROS-mediated small EV release is likely due to mitochondria specific oxidative stress. Lastly, the key protein mediator of the hypoxia stress response, HIF1α, is activated by ROS to induce EV secretion in HIV-1 infected CD4+ T cells (39). Activation of HIF1α during ischemia in kidney tubular epithelial cells or HEK293 cells also results in increased shedding of EVs with altered miRNA composition (40, 41). Therefore, increased production and altered packaging of EVs is tied to major stress responses.
Stress-induced EV signaling is by far the most well studied in cancer research. Cancer cells undergo all the above-mentioned stressors to various degrees depending on the stage and whether they are associated with a solid tumor. Not much is known about stress stimulated EVs in metabolic regulation, however, many of these same stress states can be found in multiple organs under obese and diabetic conditions. This review will discuss energetic stress as underlying most stress responses that are linked to EV release and explore the functions of these stress associated EVs in receiving cells.
An Energetic Perspective of Stress
Hijacking old pathways for modern life
Nutrient acquisition is one of the most formidable and sustained environmental pressures throughout the evolutionary history of all organisms. Chemical energy from consumed nutrients is the principal requirement for life. As such, starvation elicits one of the strongest survival responses by slowing basal metabolism to preserve energy, promoting systemic reliance on adipose-derived lipids for energy to spare glucose for the brain, and initiating intense food-seeking behavior. Nutrient deprivation can be the result of starvation, but also from intense energy consumption, as in the case of exercise, or from a physical restriction of nutrient delivery to the tissue, as with expanding tissues that outpace angiogenesis. At the cellular level, nutrient deprivation activates multiple stress response pathways in many cell types, the ER UPR, the antioxidant response, hypoxia response and autophagy (Figure 1A) (42–48). Although these stress responses may be temporally regulated in the cell, they are highly interconnected. For example, protein folding in the ER is highly sensitive to ATP and calcium levels and reliant on a strict redox environment to allow for disulfide bonds to form (49). As the ER and mitochondria are physically connected, mitochondrial dysfunction can induce the ER stress response by altering ATP levels and generating ROS (50, 51). The reverse is also true, ER dysfunction can cause mitochondrial dysfunction and oxidative stress (50). The ER can generate hydrogen peroxide through oxidative protein folding (52). This ER-generated ROS can trigger the ER stress response, which in turn can activate antioxidant pathways involved in the oxidative stress response (53). Furthermore, ROS is also generated under hypoxic conditions and is known to activate the integrated stress response, which includes the UPR (54). Lastly, downstream of all these stress pathways is the activation of autophagy, a degradative process requiring specialized machinery to engulf damaged cytoplasmic components, including whole organelles. The autophagy machinery delivers this captured material to the lysosome, which digests and recycles cellular components. It is generally accepted that autophagy is a highly conserved process to maintain homeostasis during nutrient depletion by salvaging fuel for ATP generation and building blocks for anabolic reactions (55). These interacting stress responses have the purpose of restoring the cell to homeostasis and repairing any damage that occurred during nutrient stress.
Figure 1.

Convergent energetic stress responses of obesity, starvation, and exercise. A. Energetic stress induced by obesity, starvation and exercise results in low amino acid levels, low energy charge, high NAD+ levels, high ROS and increased circulating lipids. Each of these stress signals activates the respective sensor enzyme as indicated or amplifies other stress signals. B. The stress signals and sensor pathways activate highly interconnected stress response pathways: antioxidant, hypoxia, ER UPR and autophagy. These pathways are known regulators of EV release, however, how they interact to accomplish this is not understood. C. The convergent regulation of the endolysosomal system by stress pathways may account for the net increase in EV release during energetic stress.
In modern Western society there is no restraint on calorie procurement so humans rarely, if ever, must exercise starvation-induced cellular responses. Instead, a significant portion of humans have become obese. Interestingly, the same stress responses are activated by nutrient deprivation and nutrient excess in metabolic tissues (Figure 1A). Obesity stimulates the ER UPR, antioxidant pathways and hypoxia response in the liver, adipose tissue, and muscle in mice (56–67). This can be mimicked by simply providing mice with an infusion of fatty acids (68–71), suggesting these stress responses can be directly activated by excess fatty acids. In both fasting and obesity free fatty acids rise in circulation due to adipocyte lipolysis or adipose tissue dysfunction respectively. Tissues are forced to rely on fatty acids for ATP synthesis, in part because there is either no insulin signal, as in the case of fasting, or the tissues are insulin resistant, as in obesity. Fatty acids are energy-rich and require oxygen to be oxidized in the mitochondria to yield ATP. However, excess fatty acids can lead to significant mitochondrial dysfunction and reduce the cell’s ATP-generating capacity. Mitochondria respiring on fatty acids produce more free radicals than those utilizing glucose (72). Fatty acids can be oxidatively damaged forming highly reactive lipid species that are toxic to most cells. Furthermore, fatty acids can uncouple the mitochondria and inhibit oxidative phosphorylation (73, 74). In addition, the oxygen consumed during enhanced oxidation of fatty acids in adipocytes have been shown to produce a local “pseudohypoxia”, activating the major hypoxia stress response, HIF1α (66, 75). Lastly, long chain fatty acids in cell culture or obesity in vivo have been shown to activate autophagy in pancreatic β cells (76, 77). Palmitate was also found to stimulate autophagy in MEFs, which seems to be a common finding in cultured cells (78, 79). However, the effect of obesity on autophagy is highly dependent on the effected tissue. Autophagy has been reported to be increased in adipose tissue but decreased in other organs like the heart and liver (79). Therefore, fatty acids and their actions in mitochondria are at least one link between the convergent cellular stress responses of starvation and obesity: the ER UPR, antioxidant response, hypoxic response and autophagy (Figure 1A). As obesity is a new problem in our evolutionary history, it would seem that cells are hijacking ancient starvation-induced stress pathways to adapt to the modern stress of over-nutrition. However, this re-purposing of signaling is not ideal. Starvation is generally short lived, on the order of days, so the collateral damage of strict fatty acid utilization for ATP production is an acceptable trade-off. In the case of obesity, excess lipids can circulate throughout the body for years to a lifetime resulting in compounding oxidative stress and inflammation in multiple organs, leading to chronic disease.
Energetic stress: one stress to rule them all
From this perspective, the same stress pathways organisms have evolved to combat nutrient deprivation can be harnessed imperfectly to adapt to overnutrition. Every cellular process needs energy and disruption of the cells ability to acquire it, due to a lack of energetic substrates or overwhelming the mitochondrial capacity, elicits a stress response. Therefore, it can be said that both the nutrient stress of deficiency, or surplus, can be characterized generally as “energetic stress”. Energetic stress can be identified by low cellular energy status (low ATP/ADP and NADH/NAD+ ratios), limiting biosynthetic metabolites and high mitochondrial ROS generation due to dysfunctional mitochondria, or limiting oxygen. Induction of an “energetic stress response” activates a network of discrete, but interconnected stress responses that include all major stress pathways, as mentioned above. On the other hand, energy demand is significantly increased during stress to mount, sustain, and recover from stress responses (80). Thus, if a stress response did not originate from energic defects it is still likely accompanied by some degree of energetic stress. For example, DNA damage stimulates mitochondrial oxygen consumption to produce the required ATP for repair but this, in turn, results in oxidative stress-induced mitochondrial damage, inducing the mitochondrial stress response, mitophagy (81, 82). Induction of mitophagy following DNA damage was found to be essential for maintaining mitochondrial ATP production to support DNA repair (82). In addition, AMP-activated protein kinase (AMPK) activation is integral to the DNA damage repair pathways (83). AMPK is a key regulator of cellular bioenergetics being directly activated by AMP and ADP under conditions of low energy charge (84). The activity of AMPK stimulates the restoration of cellular ATP by enhancing glucose and fatty acid catabolism, activating autophagy, and inhibiting energy-consuming processes like cell growth and biosynthesis (83). Phosphorylation of Thr172 by LKB1 is required for AMPK activation (84). Conformational changes induced by AMP or ADP binding to the regulatory subunits of AMPK enhances phosphorylation and suppresses de-phosphorylation, allowing for sustained activation (85). These studies suggest that energetic stress is a consequence of the DNA damage response and the adaptation to which rewires cellular bioenergetics to support repair processes. This is likely a phenomenon that contributes other energy demanding stress responses.
Energetic Stress and EV Production
EV release is significantly regulated by energetic stress in metabolic cells. In starving mice, the total circulating EVs and adipose tissue EVs are increased (17, 27, 86). A similar phenomenon was observed after a bout of intense exercise in humans (87–89). Likewise, in both mice and human obesity total circulating EVs and adipose tissue EVs are increased (38, 86, 90, 91). Although, there are no reliable techniques to accurately identify the cell type origin of circulating EVs, it is estimated that skeletal muscle contributes a significant amount of EVs to the circulating pool (92). In stress conditions, the cells that release more EVs into the blood are likely those that are the most effected by, or the most sensitive to, the given energetic stress. In obesity this is adipocytes, myocytes and hepatocytes, and in exercise it is myocytes, which all display increased EV production under the defined energetic stress (23, 38, 93–99). It is interesting to note that EV release in response to energetic stress is a seemingly dichotomous process. Much more energy is required to restore the biomass lost to the cell in EVs in addition to that required for the stress response itself. Because EV release accompanies all forms of stress, we can speculate the benefit of doing so is worth the energetic cost. For example, one function of EVs is thought to be disposal of unwanted material. Therefore, under energetic stress the cell could extrude damaged cellular components to lessen the toxicity and burden of repair. Additionally, as discussed later in this review, EVs can act as “help me” signals, suggesting the recruitment of resources is worth the loss in biomass.
Signals and Sensors for EV Release During Energetic Stress
Whether EV release is triggered by the energetic stress perse (low ATP/ADP, low NADH/NAD+ or high ROS) or the multiple stress pathways activated under energetic stress is still unknown. However, there is evidence that EV production is regulated by proteins that directly sense the energetic state of the cell and function to match the metabolic activity of the cell with nutrient and energy availability: the mechanistic target if rapamycin complex 1 (MTORC1) and AMPK. MTORC1 has been described as a “metabolic rheostat”, sensing nutrient levels and growth signals, and modulating growth pathways and anabolic processes accordingly (100). This sensing function of MTORC1 is downstream of multiple pathways that are modulated by amino acids, ATP, oxygen, glucose, nucleotides and other nutrients (100). When these pathways signal adequate building block availability, MTORC1 is activated to promote anabolism, cell growth and proliferation and simultaneously attenuating catabolic processes like autophagy (100). AMPK and MTORC1 antagonize each other in function by direct or indirect inhibition (101). Under conditions where energy is limiting (high (AMP+ADP)/ATP ratio), AMPK is activated to enhance ATP generating pathways and recycling pathways like autophagy and inhibit anabolic pathways, which includes the direct phosphorylation and inhibition of MTORC1(84). The reciprocal regulation of these kinases situates them at the center of cellular metabolic decisions. Of high importance during metabolic stress is the judgment of whether to induce the “self-eating” pathway of autophagy. Both AMPK and MTORC1 are considered master regulators of this processes either by inducing or inhibiting autophagic flux respectively (84, 100).
MTORC1 inhibition through rapamycin treatment has been shown to significantly enhance EV release in mouse embryonic fibroblasts (MEFs; (27)). Likewise, activation of MTORC1 by silencing the tuberous sclerosis complex 1 and 2 genes (TSC1/TSC2) results in suppression of EV release (27). This data is consistent with the notion that energetic stress, where MTORC1 is inhibited, stimulates EV release. However, Yan et. al. reported a conflicting finding, that AMPK inhibition in adipocytes stimulates EV release and AMPK activation reduces EV release (102). A similar finding was reported in cancer cell lines and immortalized endothelial cells (103). Under a given physiological condition these kinases should have the opposite activation state. For example, starvation is a strong activating stimulus for AMPK and inhibitory for MTORC1. However, based on the available data, in this scenario activated AMPK would be working to suppress EV production while, simultaneously, inhibited MTORC1 would stimulate EV release, resulting in a net EV release of zero or an amount skewed to the stronger signal. While these data clearly demonstrate a role for enzymes that are directly regulated by energetic stress in EV production and release, it is unclear how these pathways interact to do so. It is possible these seemingly incompatible results can be explained by differences in the cell types used between the studies. Additionally, there may be dose-dependent or time-dependent effects that are not accounted for by the experimental design of these studies. Finally, these pathways may be independently regulating sub-populations of EVs.
Interestingly, inhibition of MTORC1 in HeLa results in increased EV release but does not result in changes to the proteome or miRNA profile of EVs (27). In fact, 97.48% of the EV proteome was unaltered in response to rapamycin treatment. Likewise, the miRNA profile of EVs from rapamycin treated cells was indistinguishable from those of control cells (27). Zou et. al. concluded that MTORC1 may regulate EV release but not cargo selection. A similar data set has not been published for AMPK knockout cells. It would be interesting to determine if AMPK regulates EV packaging in addition to secretion.
Another indicator of low energetic status of a cell has been linked to EV release, the NADH/NAD+ ratio. The quintessential sensors of NAD status are the NAD+-dependent deacetylases, sirtuins (SIRTs). This is a family of 7 proteins in mammals that play a substantial role in many metabolic processes and diseases (104). SIRT1 is the most well studied sirtuin protein. It is localized in the nucleus where it deacetylates various proteins such as histones, transcription factors, DNA repair proteins and metabolic enzymes (104). SIRT1, like all sirtuins, is activated by NAD+ accumulation, which occurs in muscle, liver and white adipose tissue during, nutrient deprivation, caloric restriction or exercise (104). Activated SIRT1 mounts a strong adaptive pathway that has been shown and protect the cell from stressors, enhance oxidative metabolism and to extend lifespan (104). As with AMPK, the sirtuins are important positive regulators of autophagy during nutrient stress (105). Adipocyte-specific knockout of SIRT1 results in increased circulated sEVs, which contributes to the whole-body phenotype of insulin resistance and increased fat mass (106). Likewise, in HEK293 cells, knockout of SIRT1 or SIRT2 resulted in enhanced sEV release and altered cargo loading through distinct mechanisms (107). Knockdown of SIRT1 but not SIRT6 and 7 resulted in a significant increase in sEV release in breast cancer cells (108). These data suggest that SIRT1 and 2 may act as negative regulators of EV release. Manipulation of sirtuin expression also effects the cargo loading of EVs. SIRT2 interacts with L-type lectin, LMAN2, which regulates the trafficking of proteins between the trans-Golgi Network (TGN) and the endosome. Knockdown of SIRT1 or SIRT2 in HEK293 cells resulted in release of LMAN2, a Golgi-resident protein, in exosomes as well as another marker protein commonly found in exosomes, GPRC5B (107). Likewise, SIRT1 knockdown in breast cancer cells resulted in a highly altered EV proteome that was enriched with ubiquitinated proteins (108). Therefore, SIRT1 may regulate the recruitment of proteins to EVs via altering their subcellular trafficking.
A drop in cellular energy charge may modulate EV production through MTORC1, AMPK and the sirtuins but ROS production is also a candidate for initiating EV release during energetic stress. The mitochondria are the main cellular source of free radicals, which is a natural biproduct of oxidative phosphorylation. However, dysfunction of mitochondria results in ROS production that exceeds the cell’s antioxidant capacity and leads to oxidative stress. ROS has been reported to stimulate EV release in several cell types. Treatment of adipocytes with non-toxic doses of mitochondrial electron transport chain inhibitors or palmitate, which are all known stimulators of ROS generation, significantly enhance sEV release (38). This was also shown in vivo where mitochondrial dysfunction was induced by overexpression of mitochondrial ferritin (FtMT), overexpression of mitochondrially targeted amyloid precursor protein β (APPβ), or knockout out SOD2 specifically in adipocytes in a doxycycline-inducible system (38). In each condition, circulating sEVs was increased. In another study, the treatment of HEK293 cells with the calcium ionophore A23187 enhances sEV release by generating free radicals (36). Polycyclic aromatic hydrocarbon-induced ROS has also been shown to stimulate EV release in hepatocytes (109). Additionally, mitochondrial ROS is known to activate AMPK indirectly through its effect on ATP production, potentially effecting EV release through AMPK (110). MTORC1 is also activated by low levels of ROS, but inhibited by high levels of ROS, or long-term ROS exposure (111). Li et. al. found that this biphasic effect was dependent on cell type and mediated through the activities of AMPK and protein phosphatase 2A (PP2A) on the MTORC1 regulatory subunit raptor (111). Lastly, ROS activates HIF1α by stabilizing the protein through inhibiting its upstream negative regulator prolyl hydroxylase (PHD), directly activating HIF1α by nitrosylation, or promoting HIF1α transcription (112). Activation of HIF1α is associated with increased EV release (39, 41).
Energetic stress clearly results in increased EV release from multiple cell types, but based on the above-mentioned studies, the molecular mechanism for this phenomenon is unclear. The main initiating signals of energetic stress are low energy charge (high AMP, ADP, NAD+) and high ROS production. These are sensed by AMPK, MTORC1, sirtuins and HIF1α, which are all, in turn, activated in response to energic stress. Although each of these pathways are associated with EV release, their direction of regulation is inconsistent with the general observation of increased EV release under energetic stress. For example, AMPK and sirtuins are activated by low energy charge, but negatively regulate EV release (102, 103, 106, 108). In contrast, MTORC1 inhibition and HIF1α activation, as occurs during nutrient deprivation and high ROS levels, promotes EV release (27, 39). Therefore, under any given condition the pathways that are the most dominant regulators of EV release are likely dependent on the cell type, the specific energetic stress stimulus, the extent of energetic stress, and the amount of time the cell has remained with un-resolved energetic stress. Therefore, much work is still required to understand the interactions between these energetic stress sensing pathways that modulate EV production (Figure 1B).
Effector Processes for EV Release During Energetic Stress: Autophagy and Lysosomal Activity
All signaling pathways that lead to EV production and release must end up in the endolysosomal system, except for those that enhance ectosome production at the plasma membrane. The endolysosomal system is made up of a collection of molecularly defined, but dynamic membrane compartments that regulate the degradation or release of various cellular components. The cellular components that end up in this degrative pathway originate from outside the cell through endocytosis, the plasma membrane as materials that escape recycling vesicles, and damaged cellular macromolecules and organelles that enter through autophagy (113). In recent years, EVs have been shown to be released from multiple compartments in the endolysosomal system. The first are exosomes, which are released after fusion of the MVE with the plasma membrane, an alternative pathway to fusion of the MVE with the lysosome for degradation of its contents (9). Additionally, EVs and soluble proteins can be released from autophagic vesicles through what has been termed secretory autophagy. Autophagosomes are doubled membraned structures that are formed during autophagic engulfment of cellular components. During degrative autophagy, the autophagosome fuses with the lysosome forming a degrading compartment. During secretory autophagy, the autophagosome can fuse with the plasma membrane, releasing its contents into the extracellular space (114, 115). This is considered to happen due to failure of the degrative pathway (9). However, it is important to mention this autophagosome-dependent mechanism of unconventional secretion is it not well understood and likely much more complex than simple fusion of the autophagosome with the plasma membrane. For example, the secretion of IL-1β has been found to occur through secretory autophagy (114), however, this cytoplasmic protein, devoid of a signal peptide, translocates across a membrane into the lumen of a vesicle intermediate that is thought to become the autophagosome(116). Interestingly, IL-1β does not enter the cytoplasmic interior that is formed during autophagosome engulfment of cytoplasmic components. Instead, it localizes to the intermembrane space of the double-membraned autophagosome (116). Zhang et. al. found that, because of this topology, release of soluble IL-1β is guaranteed to occur by either autophagosome fusion with the plasma membrane or through autophagosome fusion with MVEs and then subsequent fusion with the plasma membrane. Amphisome is the term given to this hybrid structure produced by fusion of autophagosomes with the late endosomes or MVEs. Amphisome contents can be degraded by fusion with the lysosome or released into the extracellular space (117, 118). Lastly, the lysosome itself can fuse with the plasma membrane and secrete its contents (9). It has been suggested that lysosomal exocytosis and MVE fusion with the plasma membrane is triggered by impaired acidification of these compartments (119, 120). Therefore, specific forms of EVs arise by these different secretory pathways, although we are far from being able to experimentally separate and characterize these populations. However, it seems to be consistent that the degradative and secretory functions of these pathways antagonize each other. For example, signaling pathways that enhance autophagy and lysosomal activity, which includes adequate acidification of the various compartments, results in reduced EV release. Whereas stimuli that disrupt the full execution of the degrative pathway results in enhanced EV release (Figure 1C).
One of the early studies identifying this tug of war was that published by Hessvik et. al. who demonstrated that inhibition or knockdown of Pikfyve increased exosome release via secretory autophagy (121). Pikfyve is a lipid kinase that generates PI(3,5)P2, a lipid species required for proper MVE fusion with the lysosome and lysosomal function (121, 122). Likewise, depletion of PI2P by disrupting the function of the PI3P-synthesizing kinase Vps34 resulted in impaired lysosomal degradation and autophagy, which lead to enhanced exosome release (123). In the context of energetic stress, ROS has been shown to disrupt lysosomal activity. Centrosome amplification causes ROS production in pancreatic cancer cells, which directly inhibits lysosomal activity, causing increased exosome secretion (124). In addition, several sensors of energetic stress are known to modulate autophagy or lysosomal activity. For example, the inhibition of EV release in SIRT1 depleted cells is thought to occur by suppression of autophagy in adipocytes or reduced lysosomal function in breast cancer cells (106, 108). In addition, AMPK is well known to activate autophagy through phosphorylation of ULK1 and MTORC1 to inhibit autophagy by phosphorylation of ULK1. Therefore, the combined effect of energetic stress pathways on both the initiation of autophagy and lysosomal degradation and full execution of these processes (eg. ensuring sufficient acidification of the compartment takes place) may be a large determinant of EV release. Furthermore, convergence of multiple stress response pathways on the regulation of the endolysosomal system may explain why such a variety of stresses induce EV release.
The Effect of Energetic Stress-induced EVs on Receiving Cells
EVs can act on receiving cells at two levels. EVs can interact with the plasma membrane to induce cell receptor signal transduction without entry into the cell (5). Additionally, EVs can enter the cell and offload cargo to induce functional cellular changes. EV uptake can be accomplished by receptor-mediated endocytosis, micropinocytosis, phagocytosis, caveolin or clathrin-dependent uptake, or direct fusion with the plasma membrane (5). The signaling effects of energetic-stress induced EVs from metabolic cells on target cells can be characterized as 1) a warning, 2) collateral damage, 3) protective, or 4) recruitment. Warning EVs originate in a sensor cell that is undergoing energetic stress and provides a chemical signal to receiving cells to initiate preparation for the potential spread of the stress stimuli. EVs induce collateral damage when they spread disease from one cell to another. EVs can also, carry protective molecules that alleviate stress in receiving cells. Lastly, EVs can be released by stressed cells that recruit resources from surrounding cells to aid in stress resolution.
Warning Signal: the benefits of the by-stander effect
Threats to a single cell are likely potential threats to all cells in a multicellular organism. The cell types that are the most sensitive to a given stress can act as “sensors”, relaying information to other cells about rises in stress stimuli (Figure 2A). The adipocyte is a prime example of a sensor cell. The function of the adipocyte is to sense incoming calories and modulate systemic metabolism and feeding behavior accordingly through secretion of adipokines (1). The adipocyte converts excess calories to neutral lipid for storage in the adipocyte lipid droplet. This storage function is essential to protect other organs from lipotoxicity. This is the most evident in lipodystrophy, where adipocytes do not develop, resulting in hyperlipidemia, organ inflammation and oxidative stress. Lipodystrophy is associated significant comorbidities including non-alcoholic fatty liver disease and cardiovascular disease (125). In obesity, adipocytes become dysfunctional, resulting in many of the same comorbidities. Interestingly, within 1 day of high fat feeding in mice, ROS production in adipose tissue is significantly increased (38). During the first 3 days of high fat feeding adipocyte mitochondria become uncoupled, which enhances oxygen consumption and stimulates a pseudo-hypoxia state where HIF1α is activated (66, 75). Under both acute and chronic high fat feeding adipose tissue releases more sEVs (38). This suggests adipocytes are particularly sensitive to the effects of nutrient overload and respond by releasing EVs. There are potentially cell populations in each tissue that play this sensing function and release EVs that transfer stress traits, including damage and stress responses, to surrounding cells. This effect has been termed “the bystander effect” (126). Based on available data, the bystander effect can have beneficial effects by enhancing cell defenses to foster resilience, or it can cause excessive damage and apoptosis. The former effect can be thought of as a warning signal, a mild stress to get the receiving cell ready for a potentially lethal insult.
Figure 2.

Categories describing the effect of energetic stress-induced EVs on receiving cells. A. Cells that are sensitive to a specific stress, such as adipocytes, can send out warning signals via EVs that enhance the resilience of the receiving cell. B. Stressed cells can also release EVs contain damage and pathology-spreading molecules. C. EVs from support cells, particularly stem cells, can produce EVs that contain inherently protective molecules such as antioxidant enzymes. These EVs protect organs such as the kidney, brain, and liver from oxidative stress and aging. D. EVs from stressed cells have a capacity to recruit needed resources from other cells. This is exemplified by cancer cells or myocytes using EVs to request fatty acids from adipocytes as a source of energy. In addition, these EVs signal to attract immune cells, in the case of obese adipose tissue, and endothelial cells, in the case of a tumor, obese adipose tissue and exercising muscle.
One of the most surprising stress signals is the packaging of oxidatively damaged, but respiration competent mitochondrial fragments into EVs. Obesity or palmitate treatment results in increased incorporation of damaged mitochondria into white adipocyte sEVs, which travel to the heart and induce a burst of ROS (38). This results in compensatory antioxidant signaling in the heart that protects cardiomyocytes from acute oxidative stress. A single injection of sEVs from energetically stressed adipocytes was demonstrated to limit cardiac ischemia/reperfusion injury in mice (38). These mitochondrial fragments have been detected in isolated sEVs from the serum or plasma of obese mice and humans (38). This suggests, that in the obese condition, where adipocytes become dysfunctional, sEV-associated mitochondria are relaying a pro-oxidant signal to other organs with the outcome of stimulating cell defense pathway preparation for a future insult. Such an insult could be an ischemic event in the heart, as is common in obese individuals, but could also be a warning of the lipotoxicity to come as adipocyte function continues to deteriorate over the progression of obesity. Although the outcome of adipocyte-derived sEV-associated mitochondria is a warning signal, it likely originates as a cell-autonomous protective mechanism. The “mitochondrial particles” where found to be mitochondrial derived vesicles (MDVs). MDV formation is a mitochondrial quality control mechanism that is independent of autophagy and mitophagy machinery yet is reliant on the actions of Pink and Parkin proteins (127). MDVs are formed in response to oxidative stress, allowing for the sequestration of oxidatively damaged mitochondrial components into a budding structure that is released as an MDV (127). MDVs are transported to the late endosome and eventually to the lysosome, which completes this quality control pathway (127). However, escape of MDVs from the endolysosomal system as EVs has been shown (38, 128). It appears that most of the mitochondria released from the adipocyte is taken up by macrophages in the tissue (129). Brown adipocytes also release damaged mitochondria in large EVs in response to cold-induced mitochondrial stress or treatment with FCCP, a mitochondrial uncoupling compound (130). These extruded mitochondria are also taken up by macrophages in the tissue. If the macrophages fail to clear these EVs brown adipocytes can take up their own damaged mitochondria, which impairs the thermogenic capcity of the cell (130). Furthermore, a similar but distinct mechanism for the extrusion of damaged mitochondria has been reported in cardiomyocytes. Damaged mitochondria are released in the autophagy-dependent EV subtype, exophers, which are taken up and degraded by cardiac macrophages (10). This “outsourcing of mitophagy” to macrophages by EVs has also been described in in mesenchymal stem cell (MSC) mitochondrial maintenance (131). Therefore, the mitochondria that enter circulation were likely destined for degradation by tissue resident macrophages but escaped this fate through unknown mechanisms. Interestingly, in the obese state mitochondria (naked or EV-enclosed) seem to display a preference for leaving white adipose tissue instead of being taken up by cells in the tissue (132). A major cause of this is the long-chain fatty acids found in a lard-based high fat diets inhibit the uptake of extracellular mitochondria by tissue-resident macrophages and so mitochondria are diverted into the blood (132). This is not true in brown adipose tissue where lard-based HFD increased the transfer of adipocyte mitochondria to tissue macrophages(132). As such, the origin of a mitochondria-mediated systemic warning signal may be tissue specific and a beneficial biproduct of cell autonomous quality control mechanisms. Although, this is likely not an obesity-specific stress response as mitochondrial protein content is also increased in circulating sEVs of melanoma patients (133).
Other stresses are well known to cause a bystander effect through EVs. Cells exposed to thermal stress release EVs that are sufficient to induce DNA damage in surrounding cells (134). Although this does result in some cell death, the remaining cells are more resistant to subsequent stresses (134). ER stress is also potentially transmitted between cells through the transfer of spliced XBP1 mRNA, a major initiating transcription factor in the ER stress response (135). A similar effect can be seen in response to oxidative stress. Oxidative stress during dorsal root ganglia damage induces macrophages EV production that transfers the ROS-generating enzyme NADPH oxidase 2 (NOX2) to the injured neuron (136). NOX2 stimulates P13K/AKT signaling in neurons through oxidative inhibition of PTEN, resulting in axonal regeneration (136).
There is no warning signal more apt than that of a dying cell. Apoptotic bodies or apoptotic EVs (ApoEVs) are produced by membrane blebbing during apoptosis and carry bioactive molecules (137). ApoEVs can be considered a warning signal, but it does not seem to be through perpetuating damage. Instead of causing toxicity, as may be expected, ApoEVs strongly promote growth, proliferation, and survival of surrounding cells. For example, dying endothelial cells release ApoEVs that are taken up by endothelial progenitor cells, where their contents stimulate proliferation and differentiation (138). Macrophage LPS-stimulated ApoEV release had a similar proliferative effect on epithelial cells through the transfer of miR-221 and miR-222 (139). Likewise, ApoEVs from dying epithelial cells also promote expansion of stem cells through the transfer of Wnt8a (140). Bone marrow mesenchymal stem cells (MSCs) have also been shown to engulf ApoEVs from circulation resulting in activation of the Wnt/β-catenin pathway, which plays a critical role in self-renewal and differentiation of MSCs (141). Liu et. al. demonstrated that dying transplanted MSCs in a myocardial infarction model released ApoEVs that induced lysosomal activity and autophagy in endothelial cells (142). This resulted in survival and proliferation of endothelial cells to improve cardiac recovery (142). The final example comes from the bone remodeling literature. Osteoclast ApoEvs induce osteoblast differentiation by activating the PI3K/AKT/MTORC1 pathway (143). Although the studies on ApoEVs are not extensive, the available results are consistently pointing toward the inherent nature of these EVs as warning signals to stimulate cell survival and tissue regeneration.
Collateral damage: the dark side of the bystander effect
It is likely that all warning signals may become pathogenic over time or throughout disease progression, particularly those that originate as damaged cellular components (Figure 2B). Furthermore, EVs have been shown to pass pathology-promoting traits between cells (144). Adipose tissue EVs are one of the best examples of a warning signal that can turn pathogenic. As mentioned above, mitochondrial stress simulates adipocytes to release sEVs that act as a beneficial warning signal to the heart (38). In contrast, EVs from adipocytes harvested from diabetic mice, on a chronic high fat diet, were shown to exacerbate myocardial ischemia/reperfusion injury in lean, non-diabetic mice (145). Mechanistically, adipocyte sEVs carried miR-130b-3p, which targeted AMPK, Birc6 and UCP3 in cardiomyocytes, thereby blunting protective pathways. Adipose tissue EVs also target the hippocampus, and if derived from diet-induced obese mice or humans with type 2 diabetes, these EVs promote synaptic damage and cognitive impairment though transfer of miRNAs (146). Adipocyte EVs from obese mice or adipocytes treated with high glucose and fatty acids to stimulate insulin resistance also have a significantly negative impact on surrounding adipose tissue cells. Insulin resistant adipocyte sEVs and large EVs can induce monocyte migration and differentiation into pro-inflammatory M1-like macrophages (86, 147–150). Concomitantly, adipocyte sEVs suppress the polarization of macrophages into M2-like, anti-inflammatory macrophages via miR-34a (151). Insulin resistant adipocyte or obese adipose tissue EVs can also provoke pathologic angiogenesis and increase atherosclerotic plaque burden (150, 152). Lastly, metabolically stressed adipocytes or obese adipose tissue EVs can induce inflammation and insulin resistance in muscle and liver cells (153–155), as well as dysregulate fibrogenic signaling in hepatocytes (155, 156). This is also true in humans, where both plasma and adipose tissue-derived sEVs from obese humans with nonalcoholic fatty liver disease induce insulin resistance in myotubes and hepatocytes (157). The insulin resistance phenotype is generated by direct effects of adipocyte EVs on myocytes and hepatocytes but may also be caused indirectly through effects on pancreatic β-cell insulin secretion. Gesmundo et. al. reported that healthy 3T3-L1 adipocyte EVs enhanced the proliferation, survival, and insulin secretion activity of the INS-1E β-cell line and human islets (158). In contrast, EVs from inflamed adipocytes promoted β-cell death and dysfunction in the presence or absence of pro-inflammatory cytokines (158). Finally, induction of ER stress in adipocytes stimulates the release of EVs that contain also-keto-reductase 1B7 (Akr1b7), a mediator of lipid metabolism is the liver (159). These adipocyte Ark1b7- containing EVs cause hepatic steatosis, inflammation in fibrosis in the liver, all of which lead to NASH in mice (159).
Adipocytes are not the only cells that spread pathology through EVs in obesity. M1-like macrophages from obese, inflamed adipose tissue can produce EVs that induce whole body insulin resistance, and blunt insulin-stimulated AKT phosphorylation in myocytes and hepatocytes at least partially through transfer of miR-155 (160). MiR29a was also found to be a major insulin resistance-promoting RNA from obese adipose tissue macrophage EVs (161). These effects are thought to be dependent on the miRNA cargo of adipocyte EVs. Therefore, the effects of EVs from adipose tissue cells in obesity and type 2 diabetes contribute to the severe insulin resistance and inflammation that is characteristic of the disease.
Hepatocyte EV actions also fall into the category of a beneficial warning signal that turns pathologic over the progression of obesity. Early during the onset of obesity, hepatocytes release miR-3075-containing EVs that promote insulin sensitivity in metabolic cells by downregulating fatty acid 2-hyroxylase (96). However, in chronic obesity hepatocyte EVs promote insulin resistance through activation of pro-inflammatory macrophages (96). Likewise, EVs produced by cultured hepatocytes in lipotoxic conditions to mimic obesity attract macrophages, activate inflammatory pathways in macrophages and endothelial cells and promote angiogenesis (99, 162–164). In addition, HepG2 cells treated with fatty acids and the HIF1α-stabilizing compound cobalt chloride released EVs that stimulated a fibrosis-related transcriptional profile in hepatic stellate cells (165). Therefore, the data suggests that during chronic obesity lipid laden hepatocytes produce EVs that promote local inflammation and fibrosis, key features of NAFLD.
Protection: guardian EVs
In some cases, EVs can be powerfully protective for receiving cells under stress, not through hormesis, as with warning signals, but through the transfer of inherently protective molecules like antioxidants and pro-survival signals (Figure 2C). These guardian EVs generally originate from stem cells, a type of cell that is integral to tissue maintenance and regeneration following injury. MSCs and adipose tissue-derived stem cells (ADSCs) are important mediators of protective EV-mediated signaling. Aging is a physiological process where regeneration of tissues is impaired due to a loss of stem cell function, metabolic disturbances and increased cellular senescence (166). Senescent cells are post-mitotic, resistant to apoptosis, metabolically active and secrete an array of molecules that promote inflammation, tissue damage, and impede stem cell activity (166). Interestingly, treatment of aged, senescent stem cells with young human MSC-derived EVs rescued stem cell function, and dramatically increase the lifespan and healthspan of the short-lived ERCC1-deficient mice (166). This effect was attributed to a reduction in the number of senescent cells allowing for improved tissue recovery from stress and augmented regeneration. MSC-sEVs also carry miR-146a, which suppresses Src phosphorylation in endothelial cells, resulting in reduced oxidative stress-induced senescence and enhanced angiogenesis (167). EVs can also correct the metabolic defects of aging. Yoshida et. al. demonstrated that circulating eNAMPT levels decrease with age, an enzyme that, in the blood, is exclusively associated with EVs. eNAMPT produces NAD+, a coenzyme that is essential for ATP production, and the levels of which are reduced with age. Injection of eNAMPT-containing EVs from young mice or from eNAMPT-overexpressing adipocytes significantly increased the lifespan of mice, corrected metabolic defects associated with aging, improved exercise tolerance and increased NAD+ biosynthesis in hypothalamic neurons (168). Any beneficial effect of endogenous EV during aging is likely hindered as EV levels decline with age and carry pathological, not protective cargo (169, 170).
MSCs and ADSCs EVs have been shown to have potent antioxidant characteristics. MSC-derived EVs protect hippocampal neurons from the oxidative stress associated with amyloid-β oligomers in Alzheimer’s disease (171). These protective EVs contain catalase, an antioxidant enzyme that converts the pro-oxidant hydrogen peroxide to oxygen and water. MSC-EVs are also enriched with antioxidant miRNAs that increase catalase, superoxide dismutase, and glutathione peroxidase activities in cultured cells to reduce ROS generation and protect hippocampal neurons from lasting effects of seizure damage (172). Furthermore, EVs from MSCs have been shown to reduced inflammation, oxidative stress, and apoptosis in a mouse model of colitis via suppression of ROS production and increased antioxidant capacity in the colon (173). MSC-sEVs protect the kidney from ischemic injury by stimulating the antioxidant transcription factor NRF2 (174). Similarly, MSC-EVs protect the heart during ischemia by promoting angiogenesis, reducing ROS, attenuating fibrosis, correcting cellular bioenergetics and modulating inflammation (175–179) ADSC-derived EVs have also been shown to attenuate cardiomyocyte death in response to oxidative stress (180). In the lung, ADSC EVs have been shown to attenuate the ROS produced by environmental toxin PM2.5 by upregulating NRF2 (181). These antioxidant features of ADSC EVs have also been implicated in protection against ultraviolet B-induced damage of the skin, and attenuation of inflammation and oxidative stress in macrophages (182, 183).
It is important to note that most of the studies that demonstrate a protective effect of EVs were conducted using healthy, unstimulated, or pharmacologically treated stem cells. These studies provide interesting ideas for therapeutic approaches and may provide evidence that stem cells in distal organs, that are not affected by a specific stress, may provide a beneficial signal to affected organs. However, stem cells are important for local maintenance of tissue homeostasis and thus, the actions of EVs from MSCs under the same stress as the rest of the cells in a microenvironment are important to consider. For example, an ischemic event may affect large portions of an organ. Several studies have determined that stem cells subjected to hypoxia in vitro still have protective effects on in vivo ischemic disease. Uptake of EVs from hypoxia conditioned ADSCs or MSCs attenuates epithelial oxidative stress and promotes angiogenesis respectively (184, 185). A similar result was reported with in vivo studies. ADSCs preconditioned with anoxia limited myocardial ischemia/reperfusion injury when injected into mice (186). Lastly, EVs from hypoxia-treated ADSCs promoted recovery of renal function following ischemic injury by promoting angiogenesis and mitigating oxidative stress (187). These studies suggest that either healthy or stressed stem cells release EVs that have protective effects on receiving cells.
Stem cells and other support cells can also transfer healthy mitochondria to surrounding cells that may be deficient. This occurs tunneling nanotubules, cell fusion, GAP junction and EVs (188). Although there are few examples of healthy mitochondria transfer between cells via EVs, those that we have suggest this process can be powerfully protective for the receiving cell with mitochondrial deficiencies. In the lungs, EV-associated mitochondria are transferred from MSCs to alveolar cells during LPS stress (189). The result is increased ATP production in alveolar cells, increasing survival rates in mice following LPS lung instillation. Similarly, cancer-associated fibroblasts release EVs that contain mitochondria. These mitochondria are taken up by dormant breast cancer cells resulting in stimulation oxidative phosphorylation and escape from dormancy (190). A recent study has demonstrated the presence of what seems to be healthy, respiration competent mitochondria in human plasma (191). By electron microscopy, this study demonstrated that this population of mitochondria was not enclosed in a phospholipid membrane and therefore not associated with EVs. It would be interesting to identify any function differences between extracellular naked mitochondria and those in EVs. Further work is required to determine which conditions or which cell types are prone to releasing healthy mitochondria, which may have protective outcomes, vs damaged mitochondria, which may act as a warning signal or pathology-spreading process as discussed above.
EVs released by muscle during exercise have also been shown to display guardian functions. The systemic effects of exercise have long been considered potently beneficial to the health of rodents and humans (192). Although the mechanisms are not understood, both the release of hormones called mitokines and EVs from muscle during exercise is thought to play a role (192). Several studies have now demonstrated increased circulating EVs following exercise that are enriched with skeletal muscle-specific miRNAs and proteins in humans and mice (87, 89, 193–195). Furthermore, exercise intensity positively correlates with total circulating EVs in rats (196). However, it is important to note that not all exercise-induced EVs in circulation are from muscle cells. In addition to myocytes, platelets, endothelial progenitors, endothelial cells, and leukocytes also contribute EVs to circulation during exercise (197). Therefore, it is challenging to ascribe a beneficial function to EVs from any one cell type, so many studies focus on the total blood EV population. Vechetti et. al. compiled a list of mRNAs identified in the literature to be associated with exercise induced EVs and ran a prediction analysis to identify target genes (192). Gene ontology analysis showed that the two most significant biological processes were response to reactive oxygen species and insulin secretion (192). Exercise has been reported to improve β-cell function and increase antioxidant enzyme activity in circulation, suggesting EVs may mediate these protective effects (192). Furthermore, exercise-induced EV cargo is thought to promote angiogenesis, modulate immune function, enhance insulin sensitivity and stimulate regeneration (192, 195). The overall beneficial effects of circulating EVs during exercise is exemplified by a study, which demonstrates that an intramyocardial injection of EVs isolated from the plasma of exercised mice significantly protects mice from myocardial ischemia/reperfusion injury (198). This occurs through activation of the ERK1/2 pathway and Hsp27, by mitigating oxidative stress in cardiomyocytes (198).
EVs released from the liver following acute high fat feeding can also have a have a protective function. Zhao et. al. demonstrated that liver triglyceride content is increased within 6 hours of high fat feeding, whereas adipose tissue lipid storage lags to 12 hours (95). During those 6 hours of lipid overload hepatocytes produce more EVs that stimulate differentiation of preadipocytes and lipogenesis resulting in increased fat pad mass. This form of adipose tissue expansion, where new adipocytes are recruited, is considered healthy adipose tissue remodeling that protects other organs from lipotoxicity (199). Recently, Jung et. al. demonstrated hepatocytes treated with high glucose release EVs containing transmembrane 4 L six family member 5 (TM4SF5), which stimulates brown adipose tissue gluocose uptake (200). Therefore, hepatocyte EVs may not directly protect the receiving cell, but the signaling outcome in white and brown adipocytes results in protection of the whole organism from both lipotoxicity and glucose toxicity.
Recruitment: calling the calvary
The function of EVs in the recruitment of resources is greatly understudied, but there are examples in the literature. Under physiological and pathological conditions, cells request energy from adipocytes via EVs (Figure 2D). During resistance training, the mechanical load stimulates skeletal muscle to release EVs into circulation, which are preferentially taken up by the visceral adipose tissue (201). miR-1 in these muscle EVs stimulates lipolysis in adipocytes by repressing Tfap2α expression and thereby stimulating β adrenergic signaling. The fatty acids liberated through this process are utilized by the muscle tissue but also systemically for energy. Exercise may be the only physiological example of this phenomenon, however, cancer cells that originate in a wide range of tissues display an aptitude for energy recruitment from adipocytes. As cancer cells are under intense energetic stress because of the energetic burden of proliferation, adipocyte-derived lipids are considered highly advantageous to cancer cell growth and survival (202). Lewis lung carcinoma cells (LLC) shed EVs that stimulate adipocyte lipolysis in vitro and in vivo (203–205). Mechanistically, these EVs carry parathyroid hormone-related protein (PTHrP), which stimulates lipolysis by activating the protein kinase A (PKA) signaling pathway (203). LLC-derived EVs can also carry IL-6, which promotes lipolysis through activation of the STAT3 pathway (205). Pancreatic cancer cells stimulate adipocyte lipolysis through EVs carrying adrenomedullin (AM; (206)). EV-associated AM functions through its receptor, ADMR, to activate hormone sensitive lipase (HSL) via p38/ERK1/2 signaling. Likewise, miRNAs in EVs from breast cancer cells rewire adipocyte metabolism, which results in increased release of fatty acids, pyruvate, lactate, and ketone bodies (207). Therefore, EVs can signal to recruit energy from adipocytes during nutrient stress.
EVs can also draft support cells into areas in the tissue where they are required. The immunomodulatory effect of EVs can contribute to disease, however, the purpose of this function is to recruit immune cells to clean up toxic material in damaged tissue or fight off infection. In obesity, there is substantial infiltration of pro-inflammatory macrophages into adipose tissue, which attempt to repair the tissue following the death of adipocytes. Although this can have detrimental effects, inflammatory processes are essential for proper adaptation of the adipose tissue to nutrient overload (208). EVs from palmitate-stressed adipocyte have been described as “find me” signals which recruit monocytes and macrophages to the adipose tissue (94). This effect has also been demonstrated in several other studies (86, 147–150). Immune cells are not the only support cells that are responsive to EVs. EVs produced by hypoxic MSCs stimulates new blood vessel formation which is required for re-supplying the effected tissue with nutrients and oxygen (185). This is a key process in regeneration following ischemic injury. Likewise, cardiomyocytes release EVs during ischemia that promote angiogenesis partially though the transfer of miR-222 and miR-143 to endothelial cells (209). Hypoxia is also a common condition in expanding tumors. Cancer cells release EVs that stimulate angiogenesis to alleviate hypoxia and supply the tumor with energy (210). The peudo hypoxia and energetic stress induced in muscle during exercise is thought to stimulate EVs that have angiogenic potential (211). Interestingly, the liver produces EVs during exercise that promote angiogenesis in skeletal muscle by miR-122–5p (212). Therefore, EVs released under energetic stress can function as a beacon to recruit energetic resources or support cells.
Conclusion and Future Directions
The true complexity of intercellular metabolic regulation has now become apparent in light of the burgeoning field of EV research. EV release is stimulated by multiple stress pathways and as such, EVs are key mediators of pathogenesis in many disease states. A connecting feature of all major stress pathways is energetic stress, which is either as a cause of stress signaling, or the consequence. The molecular characteristics of energetic stress include high AMP, ADP, NAD+, and ROS levels. Each of these stress signals directly and robustly regulate the activity of enzymes that sense the energetic status of the cell: AMPK, MTORC1, SIRT1 and HIF1α. These sensors, in turn modulate EV production and release through mechanisms that are not well understood. A likely candidate effector pathway for stress induced EV release is regulation of the endolysosomal system, a pathway that is downstream of all the mentioned sensor enzymes and major cellular stress pathways. Once released, stress induced EVs signal to regulate the metabolism of surrounding cells or distal organs through surprisingly complex mechanisms. Energetic stress induced EVs function to 1) send a chemical warning to prepare the metabolic infrastructure of the receiving cell for impending insults, 2) pass on disease traits to surrounding cells, 3) protect surrounding cells from stress, or 4) recruit energy and support cells to the EV-producing cell.
There are multiple areas where we lack knowledge of how EVs signal in metabolism. The first is how EV release is regulated by energetic stress. Specifically, how stress signals, sensor enzymes and effector pathways interact to produce a net positive release of EVs. This will likely have to be studied in a cell-type and stress-type specific way. In addition, we have to understand how stress-stimulated EVs signal to regulate metabolism in both physiological and pathologic conditions. This will mean defining networks of cells that communicate via EVs, profile cell type-specific EVs, and determine the functional outcome of these signaling axis in health and disease. Examining these novel EV-mediated signals may provide new possibilities for targeted treatment of metabolic disorders in the years to come.
Didactic Synopsis.
Extracellular Vesicles (EVs) are nano-sized vesicles released by all cells that carry signaling molecules like proteins, lipids, metabolites, and RNA species. EVs can have a strong effect on the function of a receiving cell.
EVs from metabolically important cells can regulate metabolism in target cells, however, little is known mechanistically. Of particular importance is the need to understand the role EVs have in promoting pathology in obesity and type 2 diabetes.
EV release is stimulated by major forms of cellular stress that all coincide with or are driven by energetic stress. Energetic stress is characterized by the inability of the cell to maintain adequate ATP levels due to reduced availability of oxidizable substrates or mitochondrial dysfunction.
Major nutrient sensing proteins like AMPK, MTORC1, sirtuins and HIF1α have been shown to influence EV release. Downstream of most of these pathways is autophagy, which too regulates stress-induced EV release.
Stress-induced EVs can act as a warning signal to other cells, propagate disease, protect the receiving cell from stress or recruit support cells to the area.
Understanding the regulation of EV production under stress and the cell type specific signaling that stress-induced EVs elicit may open opportunities for new therapeutic targets to combat the pathologies associated with obesity.
References
- 1.Funcke JB, Scherer PE. Beyond adiponectin and leptin: adipose tissue-derived mediators of inter-organ communication. J Lipid Res. 2019;60(10):1648–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Murphy KG, Bloom SR. Gut hormones and the regulation of energy homeostasis. Nature. 2006;444(7121):854–9. [DOI] [PubMed] [Google Scholar]
- 3.Timper K, Bruning JC. Hypothalamic circuits regulating appetite and energy homeostasis: pathways to obesity. Dis Model Mech. 2017;10(6):679–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Crewe C, An YA, Scherer PE. The ominous triad of adipose tissue dysfunction: inflammation, fibrosis, and impaired angiogenesis. J Clin Invest. 2017;127(1):74–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.van Niel G, D’Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol. 2018;19(4):213–28. [DOI] [PubMed] [Google Scholar]
- 6.Harding C, Heuser J, Stahl P. Endocytosis and intracellular processing of transferrin and colloidal gold-transferrin in rat reticulocytes: demonstration of a pathway for receptor shedding. Eur J Cell Biol. 1984;35(2):256–63. [PubMed] [Google Scholar]
- 7.Pan BT, Teng K, Wu C, Adam M, Johnstone RM. Electron microscopic evidence for externalization of the transferrin receptor in vesicular form in sheep reticulocytes. J Cell Biol. 1985;101(3):942–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Stein JM, Luzio JP. Ectocytosis caused by sublytic autologous complement attack on human neutrophils. The sorting of endogenous plasma-membrane proteins and lipids into shed vesicles. Biochem J. 1991;274 (Pt 2):381–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Buratta S, Tancini B, Sagini K, Delo F, Chiaradia E, Urbanelli L, et al. Lysosomal Exocytosis, Exosome Release and Secretory Autophagy: The Autophagic- and Endo-Lysosomal Systems Go Extracellular. Int J Mol Sci. 2020;21(7). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Nicolas-Avila JA, Lechuga-Vieco AV, Esteban-Martinez L, Sanchez-Diaz M, Diaz-Garcia E, Santiago DJ, et al. A Network of Macrophages Supports Mitochondrial Homeostasis in the Heart. Cell. 2020;183(1):94–109 e23. [DOI] [PubMed] [Google Scholar]
- 11.Medina DL, Fraldi A, Bouche V, Annunziata F, Mansueto G, Spampanato C, et al. Transcriptional activation of lysosomal exocytosis promotes cellular clearance. Dev Cell. 2011;21(3):421–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Das Gupta A, Krawczynska N, Nelson ER. Extracellular Vesicles-The Next Frontier in Endocrinology. Endocrinology. 2021;162(9). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Phoonsawat W, Aoki-Yoshida A, Tsuruta T, Sonoyama K. Adiponectin is partially associated with exosomes in mouse serum. Biochem Biophys Res Commun. 2014;448(3):261–6. [DOI] [PubMed] [Google Scholar]
- 14.Rong B, Feng R, Liu C, Wu Q, Sun C. Reduced delivery of epididymal adipocyte-derived exosomal resistin is essential for melatonin ameliorating hepatic steatosis in mice. J Pineal Res. 2019;66(4):e12561. [DOI] [PubMed] [Google Scholar]
- 15.Obata Y, Kita S, Koyama Y, Fukuda S, Takeda H, Takahashi M, et al. Adiponectin/T-cadherin system enhances exosome biogenesis and decreases cellular ceramides by exosomal release. JCI Insight. 2018;3(8). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Giordano C, Gelsomino L, Barone I, Panza S, Augimeri G, Bonofiglio D, et al. Leptin Modulates Exosome Biogenesis in Breast Cancer Cells: An Additional Mechanism in Cell-to-Cell Communication. J Clin Med. 2019;8(7). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Crewe C, Joffin N, Rutkowski JM, Kim M, Zhang F, Towler DA, et al. An Endothelial-to-Adipocyte Extracellular Vesicle Axis Governed by Metabolic State. Cell. 2018;175(3):695–708 e13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Isaac R, Reis FCG, Ying W, Olefsky JM. Exosomes as mediators of intercellular crosstalk in metabolism. Cell Metab. 2021;33(9):1744–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Crewe C, Scherer PE. Intercellular and interorgan crosstalk through adipocyte extracellular vesicles. Rev Endocr Metab Disord. 2021. [DOI] [PubMed] [Google Scholar]
- 20.Saeedi S, Israel S, Nagy C, Turecki G. The emerging role of exosomes in mental disorders. Transl Psychiatry. 2019;9(1):122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Xu R, Rai A, Chen M, Suwakulsiri W, Greening DW, Simpson RJ. Extracellular vesicles in cancer - implications for future improvements in cancer care. Nat Rev Clin Oncol. 2018;15(10):617–38. [DOI] [PubMed] [Google Scholar]
- 22.Hill AF. Extracellular Vesicles and Neurodegenerative Diseases. J Neurosci. 2019;39(47):9269–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Durcin M, Fleury A, Taillebois E, Hilairet G, Krupova Z, Henry C, et al. Characterisation of adipocyte-derived extracellular vesicle subtypes identifies distinct protein and lipid signatures for large and small extracellular vesicles. J Extracell Vesicles. 2017;6(1):1305677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Hedlund M, Nagaeva O, Kargl D, Baranov V, Mincheva-Nilsson L. Thermal- and oxidative stress causes enhanced release of NKG2D ligand-bearing immunosuppressive exosomes in leukemia/lymphoma T and B cells. PLoS One. 2011;6(2):e16899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Parvanian S, Zha H, Su D, Xi L, Jiu Y, Chen H, et al. Exosomal Vimentin from Adipocyte Progenitors Protects Fibroblasts against Osmotic Stress and Inhibits Apoptosis to Enhance Wound Healing. Int J Mol Sci. 2021;22(9). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Parolini I, Federici C, Raggi C, Lugini L, Palleschi S, De Milito A, et al. Microenvironmental pH is a key factor for exosome traffic in tumor cells. J Biol Chem. 2009;284(49):34211–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Zou W, Lai M, Zhang Y, Zheng L, Xing Z, Li T, et al. Exosome Release Is Regulated by mTORC1. Adv Sci (Weinh). 2019;6(3):1801313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.DeClercq V, d’Eon B, McLeod RS. Fatty acids increase adiponectin secretion through both classical and exosome pathways. Biochim Biophys Acta. 2015;1851(9):1123–33. [DOI] [PubMed] [Google Scholar]
- 29.Sano S, Izumi Y, Yamaguchi T, Yamazaki T, Tanaka M, Shiota M, et al. Lipid synthesis is promoted by hypoxic adipocyte-derived exosomes in 3T3-L1 cells. Biochem Biophys Res Commun. 2014;445(2):327–33. [DOI] [PubMed] [Google Scholar]
- 30.Kucharzewska P, Christianson HC, Welch JE, Svensson KJ, Fredlund E, Ringner M, et al. Exosomes reflect the hypoxic status of glioma cells and mediate hypoxia-dependent activation of vascular cells during tumor development. Proc Natl Acad Sci U S A. 2013;110(18):7312–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Abramowicz A, Widlak P, Pietrowska M. Different Types of Cellular Stress Affect the Proteome Composition of Small Extracellular Vesicles: A Mini Review. Proteomes. 2019;7(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Osman A, El-Gamal H, Pasha M, Zeidan A, Korashy HM, Abdelsalam SS, et al. Endoplasmic Reticulum (ER) Stress-Generated Extracellular Vesicles (Microparticles) Self-Perpetuate ER Stress and Mediate Endothelial Cell Dysfunction Independently of Cell Survival. Front Cardiovasc Med. 2020;7:584791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Collett GP, Redman CW, Sargent IL, Vatish M. Endoplasmic reticulum stress stimulates the release of extracellular vesicles carrying danger-associated molecular pattern (DAMP) molecules. Oncotarget. 2018;9(6):6707–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Buratta S, Shimanaka Y, Costanzi E, Ni S, Urbanelli L, Kono N, et al. Lipotoxic stress alters the membrane lipid profile of extracellular vesicles released by Huh-7 hepatocarcinoma cells. Sci Rep. 2021;11(1):4613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Kakazu E, Mauer AS, Yin M, Malhi H. Hepatocytes release ceramide-enriched pro-inflammatory extracellular vesicles in an IRE1alpha-dependent manner. J Lipid Res. 2016;57(2):233–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Mancek-Keber M, Frank-Bertoncelj M, Hafner-Bratkovic I, Smole A, Zorko M, Pirher N, et al. Toll-like receptor 4 senses oxidative stress mediated by the oxidation of phospholipids in extracellular vesicles. Sci Signal. 2015;8(381):ra60. [DOI] [PubMed] [Google Scholar]
- 37.Takasugi M, Okada R, Takahashi A, Virya Chen D, Watanabe S, Hara E. Small extracellular vesicles secreted from senescent cells promote cancer cell proliferation through EphA2. Nat Commun. 2017;8:15729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Crewe C, Funcke JB, Li S, Joffin N, Gliniak CM, Ghaben AL, et al. Extracellular vesicle-based interorgan transport of mitochondria from energetically stressed adipocytes. Cell Metab. 2021;33(9):1853–68 e11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Duette G, Pereyra Gerber P, Rubione J, Perez PS, Landay AL, Crowe SM, et al. Induction of HIF-1alpha by HIV-1 Infection in CD4(+) T Cells Promotes Viral Replication and Drives Extracellular Vesicle-Mediated Inflammation. mBio. 2018;9(5). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Li ZL, Lv LL, Tang TT, Wang B, Feng Y, Zhou LT, et al. HIF-1alpha inducing exosomal microRNA-23a expression mediates the cross-talk between tubular epithelial cells and macrophages in tubulointerstitial inflammation. Kidney Int. 2019;95(2):388–404. [DOI] [PubMed] [Google Scholar]
- 41.Muniz-Garcia A, Romero M, Falcomicronn-Perez JM, Murray P, Zorzano A, Mora S. Hypoxia-induced HIF1alpha activation regulates small extracellular vesicle release in human embryonic kidney cells. Sci Rep. 2022;12(1):1443. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Iurlaro R, Puschel F, Leon-Annicchiarico CL, O’Connor H, Martin SJ, Palou-Gramon D, et al. Glucose Deprivation Induces ATF4-Mediated Apoptosis through TRAIL Death Receptors. Mol Cell Biol. 2017;37(10). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Zhang YK, Wu KC, Klaassen CD. Genetic activation of Nrf2 protects against fasting-induced oxidative stress in livers of mice. PLoS One. 2013;8(3):e59122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Wasselin T, Zahn S, Maho YL, Dorsselaer AV, Raclot T, Bertile F. Exacerbated oxidative stress in the fasting liver according to fuel partitioning. Proteomics. 2014;14(16):1905–21. [DOI] [PubMed] [Google Scholar]
- 45.Lettieri-Barbato D, Minopoli G, Caggiano R, Izzo R, Santillo M, Aquilano K, et al. Fasting Drives Nrf2-Related Antioxidant Response in Skeletal Muscle. Int J Mol Sci. 2020;21(20). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Basse AL, Isidor MS, Winther S, Skjoldborg NB, Murholm M, Andersen ES, et al. Regulation of glycolysis in brown adipocytes by HIF-1alpha. Sci Rep. 2017;7(1):4052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Nishimoto A, Kugimiya N, Hosoyama T, Enoki T, Li TS, Hamano K. HIF-1alpha activation under glucose deprivation plays a central role in the acquisition of anti-apoptosis in human colon cancer cells. Int J Oncol. 2014;44(6):2077–84. [DOI] [PubMed] [Google Scholar]
- 48.Zhang G, Wang X, Rothermel BA, Lavandero S, Wang ZV. The integrated stress response in ischemic diseases. Cell Death Differ. 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Szegezdi E, Logue SE, Gorman AM, Samali A. Mediators of endoplasmic reticulum stress-induced apoptosis. EMBO Rep. 2006;7(9):880–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Bouman L, Schlierf A, Lutz AK, Shan J, Deinlein A, Kast J, et al. Parkin is transcriptionally regulated by ATF4: evidence for an interconnection between mitochondrial stress and ER stress. Cell Death Differ. 2011;18(5):769–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Booth DM, Enyedi B, Geiszt M, Varnai P, Hajnoczky G. Redox Nanodomains Are Induced by and Control Calcium Signaling at the ER-Mitochondrial Interface. Mol Cell. 2016;63(2):240–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Borges CR, Lake DF. Oxidative protein folding: nature’s knotty challenge. Antioxid Redox Signal. 2014;21(3):392–5. [DOI] [PubMed] [Google Scholar]
- 53.Cullinan SB, Diehl JA. PERK-dependent activation of Nrf2 contributes to redox homeostasis and cell survival following endoplasmic reticulum stress. J Biol Chem. 2004;279(19):20108–17. [DOI] [PubMed] [Google Scholar]
- 54.Liu L, Wise DR, Diehl JA, Simon MC. Hypoxic reactive oxygen species regulate the integrated stress response and cell survival. J Biol Chem. 2008;283(45):31153–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Galluzzi L, Pietrocola F, Levine B, Kroemer G. Metabolic control of autophagy. Cell. 2014;159(6):1263–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Ozcan U, Cao Q, Yilmaz E, Lee AH, Iwakoshi NN, Ozdelen E, et al. Endoplasmic reticulum stress links obesity, insulin action, and type 2 diabetes. Science. 2004;306(5695):457–61. [DOI] [PubMed] [Google Scholar]
- 57.Kawasaki N, Asada R, Saito A, Kanemoto S, Imaizumi K. Obesity-induced endoplasmic reticulum stress causes chronic inflammation in adipose tissue. Sci Rep. 2012;2:799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Malhi H, Kaufman RJ. Endoplasmic reticulum stress in liver disease. J Hepatol. 2011;54(4):795–809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Zhao T, Wu K, Hogstrand C, Xu YH, Chen GH, Wei CC, et al. Lipophagy mediated carbohydrate-induced changes of lipid metabolism via oxidative stress, endoplasmic reticulum (ER) stress and ChREBP/PPARgamma pathways. Cell Mol Life Sci. 2020;77(10):1987–2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Yuzefovych LV, LeDoux SP, Wilson GL, Rachek LI. Mitochondrial DNA damage via augmented oxidative stress regulates endoplasmic reticulum stress and autophagy: crosstalk, links and signaling. PLoS One. 2013;8(12):e83349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Furukawa S, Fujita T, Shimabukuro M, Iwaki M, Yamada Y, Nakajima Y, et al. Increased oxidative stress in obesity and its impact on metabolic syndrome. J Clin Invest. 2004;114(12):1752–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Ochiai D, Goda N, Hishiki T, Kanai M, Senoo-Matsuda N, Soga T, et al. Disruption of HIF-1alpha in hepatocytes impairs glucose metabolism in diet-induced obesity mice. Biochem Biophys Res Commun. 2011;415(3):445–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Lima GA, Anhe GF, Giannocco G, Nunes MT, Correa-Giannella ML, Machado UF. Contractile activity per se induces transcriptional activation of SLC2A4 gene in soleus muscle: involvement of MEF2D, HIF-1a, and TRalpha transcriptional factors. Am J Physiol Endocrinol Metab. 2009;296(1):E132–8. [DOI] [PubMed] [Google Scholar]
- 64.Zelzer E, Levy Y, Kahana C, Shilo BZ, Rubinstein M, Cohen B. Insulin induces transcription of target genes through the hypoxia-inducible factor HIF-1alpha/ARNT. EMBO J. 1998;17(17):5085–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.He Q, Gao Z, Yin J, Zhang J, Yun Z, Ye J. Regulation of HIF-1{alpha} activity in adipose tissue by obesity-associated factors: adipogenesis, insulin, and hypoxia. Am J Physiol Endocrinol Metab. 2011;300(5):E877–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Seo JB, Riopel M, Cabrales P, Huh JY, Bandyopadhyay GK, Andreyev AY, et al. Knockdown of Ant2 Reduces Adipocyte Hypoxia And Improves Insulin Resistance in Obesity. Nat Metab. 2019;1(1):86–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Shao M, Hepler C, Zhang Q, Shan B, Vishvanath L, Henry GH, et al. Pathologic HIF1alpha signaling drives adipose progenitor dysfunction in obesity. Cell Stem Cell. 2021;28(4):685–701 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Boden G, Song W, Duan X, Cheung P, Kresge K, Barrero C, et al. Infusion of glucose and lipids at physiological rates causes acute endoplasmic reticulum stress in rat liver. Obesity (Silver Spring). 2011;19(7):1366–73. [DOI] [PubMed] [Google Scholar]
- 69.Nivala AM, Reese L, Frye M, Gentile CL, Pagliassotti MJ. Fatty acid-mediated endoplasmic reticulum stress in vivo: differential response to the infusion of Soybean and Lard Oil in rats. Metabolism. 2013;62(5):753–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Borradaile NM, Han X, Harp JD, Gale SE, Ory DS, Schaffer JE. Disruption of endoplasmic reticulum structure and integrity in lipotoxic cell death. J Lipid Res. 2006;47(12):2726–37. [DOI] [PubMed] [Google Scholar]
- 71.Woodworth-Hobbs ME, Perry BD, Rahnert JA, Hudson MB, Zheng B, Russ Price S. Docosahexaenoic acid counteracts palmitate-induced endoplasmic reticulum stress in C2C12 myotubes: Impact on muscle atrophy. Physiol Rep. 2017;5(23). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Rindler PM, Plafker SM, Szweda LI, Kinter M. High dietary fat selectively increases catalase expression within cardiac mitochondria. J Biol Chem. 2013;288(3):1979–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Wojtczak L, Schonfeld P. Effect of fatty acids on energy coupling processes in mitochondria. Biochim Biophys Acta. 1993;1183(1):41–57. [DOI] [PubMed] [Google Scholar]
- 74.Bernardi P, Penzo D, Wojtczak L. Mitochondrial energy dissipation by fatty acids. Mechanisms and implications for cell death. Vitam Horm. 2002;65:97–126. [DOI] [PubMed] [Google Scholar]
- 75.Lee YS, Kim JW, Osborne O, Oh DY, Sasik R, Schenk S, et al. Increased adipocyte O2 consumption triggers HIF-1alpha, causing inflammation and insulin resistance in obesity. Cell. 2014;157(6):1339–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Ebato C, Uchida T, Arakawa M, Komatsu M, Ueno T, Komiya K, et al. Autophagy is important in islet homeostasis and compensatory increase of beta cell mass in response to high-fat diet. Cell Metab. 2008;8(4):325–32. [DOI] [PubMed] [Google Scholar]
- 77.Komiya K, Uchida T, Ueno T, Koike M, Abe H, Hirose T, et al. Free fatty acids stimulate autophagy in pancreatic beta-cells via JNK pathway. Biochem Biophys Res Commun. 2010;401(4):561–7. [DOI] [PubMed] [Google Scholar]
- 78.Tan SH, Shui G, Zhou J, Li JJ, Bay BH, Wenk MR, et al. Induction of autophagy by palmitic acid via protein kinase C-mediated signaling pathway independent of mTOR (mammalian target of rapamycin). J Biol Chem. 2012;287(18):14364–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Zhang Y, Sowers JR, Ren J. Targeting autophagy in obesity: from pathophysiology to management. Nat Rev Endocrinol. 2018;14(6):356–76. [DOI] [PubMed] [Google Scholar]
- 80.Picard M, McEwen BS, Epel ES, Sandi C. An energetic view of stress: Focus on mitochondria. Front Neuroendocrinol. 2018;49:72–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Qin L, Fan M, Candas D, Jiang G, Papadopoulos S, Tian L, et al. CDK1 Enhances Mitochondrial Bioenergetics for Radiation-Induced DNA Repair. Cell Rep. 2015;13(10):2056–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Dan X, Babbar M, Moore A, Wechter N, Tian J, Mohanty JG, et al. DNA damage invokes mitophagy through a pathway involving Spata18. Nucleic Acids Res. 2020;48(12):6611–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Szewczuk M, Boguszewska K, Kazmierczak-Baranska J, Karwowski BT. The role of AMPK in metabolism and its influence on DNA damage repair. Mol Biol Rep. 2020;47(11):9075–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Mihaylova MM, Shaw RJ. The AMPK signalling pathway coordinates cell growth, autophagy and metabolism. Nat Cell Biol. 2011;13(9):1016–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Oakhill JS, Steel R, Chen ZP, Scott JW, Ling N, Tam S, et al. AMPK is a direct adenylate charge-regulated protein kinase. Science. 2011;332(6036):1433–5. [DOI] [PubMed] [Google Scholar]
- 86.Flaherty SE 3rd, Grijalva A, Xu X, Ables E, Nomani A, Ferrante AW Jr. A lipase-independent pathway of lipid release and immune modulation by adipocytes. Science. 2019;363(6430):989–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Guescini M, Canonico B, Lucertini F, Maggio S, Annibalini G, Barbieri E, et al. Muscle Releases Alpha-Sarcoglycan Positive Extracellular Vesicles Carrying miRNAs in the Bloodstream. PLoS One. 2015;10(5):e0125094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Whitham M, Parker BL, Friedrichsen M, Hingst JR, Hjorth M, Hughes WE, et al. Extracellular Vesicles Provide a Means for Tissue Crosstalk during Exercise. Cell Metab. 2018;27(1):237–51 e4. [DOI] [PubMed] [Google Scholar]
- 89.Fruhbeis C, Helmig S, Tug S, Simon P, Kramer-Albers EM. Physical exercise induces rapid release of small extracellular vesicles into the circulation. J Extracell Vesicles. 2015;4:28239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Castano C, Kalko S, Novials A, Parrizas M. Obesity-associated exosomal miRNAs modulate glucose and lipid metabolism in mice. Proc Natl Acad Sci U S A. 2018;115(48):12158–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Deng ZB, Poliakov A, Hardy RW, Clements R, Liu C, Liu Y, et al. Adipose tissue exosome-like vesicles mediate activation of macrophage-induced insulin resistance. Diabetes. 2009;58(11):2498–505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Estrada AL, Valenti ZJ, Hehn G, Amorese AJ, Williams NS, Balestrieri NP, et al. Extracellular vesicle secretion is tissue-dependent ex vivo and skeletal muscle myofiber extracellular vesicles reach the circulation in vivo. Am J Physiol Cell Physiol. 2022;322(2):C246–C59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Wadey RM, Connolly KD, Mathew D, Walters G, Rees DA, James PE. Inflammatory adipocyte-derived extracellular vesicles promote leukocyte attachment to vascular endothelial cells. Atherosclerosis. 2019;283:19–27. [DOI] [PubMed] [Google Scholar]
- 94.Eguchi A, Mulya A, Lazic M, Radhakrishnan D, Berk MP, Povero D, et al. Microparticles release by adipocytes act as “find-me” signals to promote macrophage migration. PLoS One. 2015;10(4):e0123110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Zhao Y, Zhao MF, Jiang S, Wu J, Liu J, Yuan XW, et al. Liver governs adipose remodelling via extracellular vesicles in response to lipid overload. Nat Commun. 2020;11(1):719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Ji Y, Luo Z, Gao H, Dos Reis FCG, Bandyopadhyay G, Jin Z, et al. Hepatocyte-derived exosomes from early onset obese mice promote insulin sensitivity through miR-3075. Nat Metab. 2021;3(9):1163–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Aswad H, Forterre A, Wiklander OP, Vial G, Danty-Berger E, Jalabert A, et al. Exosomes participate in the alteration of muscle homeostasis during lipid-induced insulin resistance in mice. Diabetologia. 2014;57(10):2155–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Nielsen MH, Sabaratnam R, Pedersen AJT, Hojlund K, Handberg A. Acute Exercise Increases Plasma Levels of Muscle-Derived Microvesicles Carrying Fatty Acid Transport Proteins. J Clin Endocrinol Metab. 2019;104(10):4804–14. [DOI] [PubMed] [Google Scholar]
- 99.Hirsova P, Ibrahim SH, Krishnan A, Verma VK, Bronk SF, Werneburg NW, et al. Lipid-Induced Signaling Causes Release of Inflammatory Extracellular Vesicles From Hepatocytes. Gastroenterology. 2016;150(4):956–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Valvezan AJ, Manning BD. Molecular logic of mTORC1 signalling as a metabolic rheostat. Nat Metab. 2019;1(3):321–33. [DOI] [PubMed] [Google Scholar]
- 101.Holczer M, Hajdu B, Lorincz T, Szarka A, Banhegyi G, Kapuy O. A Double Negative Feedback Loop between mTORC1 and AMPK Kinases Guarantees Precise Autophagy Induction upon Cellular Stress. Int J Mol Sci. 2019;20(22). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Yan C, Tian X, Li J, Liu D, Ye D, Xie Z, et al. A High-Fat Diet Attenuates AMPK alpha1 in Adipocytes to Induce Exosome Shedding and Nonalcoholic Fatty Liver Development In Vivo. Diabetes. 2021;70(2):577–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Ludwig N, Yerneni SS, Menshikova EV, Gillespie DG, Jackson EK, Whiteside TL. Simultaneous Inhibition of Glycolysis and Oxidative Phosphorylation Triggers a Multi-Fold Increase in Secretion of Exosomes: Possible Role of 2’3’-cAMP. Sci Rep. 2020;10(1):6948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Houtkooper RH, Pirinen E, Auwerx J. Sirtuins as regulators of metabolism and healthspan. Nat Rev Mol Cell Biol. 2012;13(4):225–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Lee IH. Mechanisms and disease implications of sirtuin-mediated autophagic regulation. Exp Mol Med. 2019;51(9):1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Li F, Li H, Jin X, Zhang Y, Kang X, Zhang Z, et al. Adipose-specific knockdown of Sirt1 results in obesity and insulin resistance by promoting exosomes release. Cell Cycle. 2019;18(17):2067–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Lee BR, Sanstrum BJ, Liu Y, Kwon SH. Distinct role of Sirtuin 1 (SIRT1) and Sirtuin 2 (SIRT2) in inhibiting cargo-loading and release of extracellular vesicles. Sci Rep. 2019;9(1):20049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Latifkar A, Ling L, Hingorani A, Johansen E, Clement A, Zhang X, et al. Loss of Sirtuin 1 Alters the Secretome of Breast Cancer Cells by Impairing Lysosomal Integrity. Dev Cell. 2019;49(3):393–408 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.van Meteren N, Lagadic-Gossmann D, Podechard N, Gobart D, Gallais I, Chevanne M, et al. Extracellular vesicles released by polycyclic aromatic hydrocarbons-treated hepatocytes trigger oxidative stress in recipient hepatocytes by delivering iron. Free Radic Biol Med. 2020;160:246–62. [DOI] [PubMed] [Google Scholar]
- 110.Hinchy EC, Gruszczyk AV, Willows R, Navaratnam N, Hall AR, Bates G, et al. Mitochondria-derived ROS activate AMP-activated protein kinase (AMPK) indirectly. J Biol Chem. 2018;293(44):17208–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Li M, Zhao L, Liu J, Liu A, Jia C, Ma D, et al. Multi-mechanisms are involved in reactive oxygen species regulation of mTORC1 signaling. Cell Signal. 2010;22(10):1469–76. [DOI] [PubMed] [Google Scholar]
- 112.Movafagh S, Crook S, Vo K. Regulation of hypoxia-inducible factor-1a by reactive oxygen species: new developments in an old debate. J Cell Biochem. 2015;116(5):696–703. [DOI] [PubMed] [Google Scholar]
- 113.Repnik U, Cesen MH, Turk B. The endolysosomal system in cell death and survival. Cold Spring Harb Perspect Biol. 2013;5(1):a008755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Dupont N, Jiang S, Pilli M, Ornatowski W, Bhattacharya D, Deretic V. Autophagy-based unconventional secretory pathway for extracellular delivery of IL-1beta. EMBO J. 2011;30(23):4701–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Duran JM, Anjard C, Stefan C, Loomis WF, Malhotra V. Unconventional secretion of Acb1 is mediated by autophagosomes. J Cell Biol. 2010;188(4):527–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Zhang M, Kenny SJ, Ge L, Xu K, Schekman R. Translocation of interleukin-1beta into a vesicle intermediate in autophagy-mediated secretion. Elife. 2015;4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Gordon PB, Seglen PO. Prelysosomal convergence of autophagic and endocytic pathways. Biochem Biophys Res Commun. 1988;151(1):40–7. [DOI] [PubMed] [Google Scholar]
- 118.Chen YD, Fang YT, Cheng YL, Lin CF, Hsu LJ, Wang SY, et al. Exophagy of annexin A2 via RAB11, RAB8A and RAB27A in IFN-gamma-stimulated lung epithelial cells. Sci Rep. 2017;7(1):5676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Guo H, Chitiprolu M, Roncevic L, Javalet C, Hemming FJ, Trung MT, et al. Atg5 Disassociates the V1V0-ATPase to Promote Exosome Production and Tumor Metastasis Independent of Canonical Macroautophagy. Dev Cell. 2017;43(6):716–30 e7. [DOI] [PubMed] [Google Scholar]
- 120.Miao Y, Li G, Zhang X, Xu H, Abraham SN. A TRP Channel Senses Lysosome Neutralization by Pathogens to Trigger Their Expulsion. Cell. 2015;161(6):1306–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Hessvik NP, Overbye A, Brech A, Torgersen ML, Jakobsen IS, Sandvig K, et al. PIKfyve inhibition increases exosome release and induces secretory autophagy. Cell Mol Life Sci. 2016;73(24):4717–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Sharma G, Guardia CM, Roy A, Vassilev A, Saric A, Griner LN, et al. A family of PIKFYVE inhibitors with therapeutic potential against autophagy-dependent cancer cells disrupt multiple events in lysosome homeostasis. Autophagy. 2019;15(10):1694–718. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Miranda AM, Lasiecka ZM, Xu Y, Neufeld J, Shahriar S, Simoes S, et al. Neuronal lysosomal dysfunction releases exosomes harboring APP C-terminal fragments and unique lipid signatures. Nat Commun. 2018;9(1):291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Adams SD, Csere J, D’Angelo G, Carter EP, Romao M, Arnandis T, et al. Centrosome amplification mediates small extracellular vesicle secretion via lysosome disruption. Curr Biol. 2021;31(7):1403–16 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Vigouroux C, Caron-Debarle M, Le Dour C, Magre J, Capeau J. Molecular mechanisms of human lipodystrophies: from adipocyte lipid droplet to oxidative stress and lipotoxicity. Int J Biochem Cell Biol. 2011;43(6):862–76. [DOI] [PubMed] [Google Scholar]
- 126.Hall EJ. The bystander effect. Health Phys. 2003;85(1):31–5. [DOI] [PubMed] [Google Scholar]
- 127.Soubannier V, McLelland GL, Zunino R, Braschi E, Rippstein P, Fon EA, et al. A vesicular transport pathway shuttles cargo from mitochondria to lysosomes. Curr Biol. 2012;22(2):135–41. [DOI] [PubMed] [Google Scholar]
- 128.Todkar K, Chikhi L, Desjardins V, El-Mortada F, Pepin G, Germain M. Selective packaging of mitochondrial proteins into extracellular vesicles prevents the release of mitochondrial DAMPs. Nat Commun. 2021;12(1):1971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Brestoff JR, Wilen CB, Moley JR, Li Y, Zou W, Malvin NP, et al. Intercellular Mitochondria Transfer to Macrophages Regulates White Adipose Tissue Homeostasis and Is Impaired in Obesity. Cell Metab. 2021;33(2):270–82 e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Rosina M, Ceci V, Turchi R, Chuan L, Borcherding N, Sciarretta F, et al. Ejection of damaged mitochondria and their removal by macrophages ensure efficient thermogenesis in brown adipose tissue. Cell Metab. 2022;34(4):533–48 e12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Phinney DG, Di Giuseppe M, Njah J, Sala E, Shiva S, St Croix CM, et al. Mesenchymal stem cells use extracellular vesicles to outsource mitophagy and shuttle microRNAs. Nat Commun. 2015;6:8472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Borcherding N, Jia W, Giwa R, Field RL, Moley JR, Kopecky BJ, et al. Dietary lipids inhibit mitochondria transfer to macrophages to divert adipocyte-derived mitochondria into the blood. Cell Metab. 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Jang SC, Crescitelli R, Cvjetkovic A, Belgrano V, Olofsson Bagge R, Sundfeldt K, et al. Mitochondrial protein enriched extracellular vesicles discovered in human melanoma tissues can be detected in patient plasma. J Extracell Vesicles. 2019;8(1):1635420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Bewicke-Copley F, Mulcahy LA, Jacobs LA, Samuel P, Akbar N, Pink RC, et al. Extracellular vesicles released following heat stress induce bystander effect in unstressed populations. J Extracell Vesicles. 2017;6(1):1340746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Hosoi T, Nakashima M, Ozawa K. Incorporation of the Endoplasmic Reticulum Stress-Induced Spliced Form of XBP1 mRNA in the Exosomes. Front Physiol. 2018;9:1357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Hervera A, De Virgiliis F, Palmisano I, Zhou L, Tantardini E, Kong G, et al. Reactive oxygen species regulate axonal regeneration through the release of exosomal NADPH oxidase 2 complexes into injured axons. Nat Cell Biol. 2018;20(3):307–19. [DOI] [PubMed] [Google Scholar]
- 137.Li M, Liao L, Tian W. Extracellular Vesicles Derived From Apoptotic Cells: An Essential Link Between Death and Regeneration. Front Cell Dev Biol. 2020;8:573511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Hristov M, Erl W, Linder S, Weber PC. Apoptotic bodies from endothelial cells enhance the number and initiate the differentiation of human endothelial progenitor cells in vitro. Blood. 2004;104(9):2761–6. [DOI] [PubMed] [Google Scholar]
- 139.Zhu Z, Zhang D, Lee H, Menon AA, Wu J, Hu K, et al. Macrophage-derived apoptotic bodies promote the proliferation of the recipient cells via shuttling microRNA-221/222. J Leukoc Biol. 2017;101(6):1349–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Brock CK, Wallin ST, Ruiz OE, Samms KM, Mandal A, Sumner EA, et al. Stem cell proliferation is induced by apoptotic bodies from dying cells during epithelial tissue maintenance. Nat Commun. 2019;10(1):1044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Liu D, Kou X, Chen C, Liu S, Liu Y, Yu W, et al. Circulating apoptotic bodies maintain mesenchymal stem cell homeostasis and ameliorate osteopenia via transferring multiple cellular factors. Cell Res. 2018;28(9):918–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Liu H, Liu S, Qiu X, Yang X, Bao L, Pu F, et al. Donor MSCs release apoptotic bodies to improve myocardial infarction via autophagy regulation in recipient cells. Autophagy. 2020;16(12):2140–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Ma Q, Liang M, Wu Y, Ding N, Duan L, Yu T, et al. Mature osteoclast-derived apoptotic bodies promote osteogenic differentiation via RANKL-mediated reverse signaling. J Biol Chem. 2019;294(29):11240–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Sousa D, Lima RT, Vasconcelos MH. Intercellular Transfer of Cancer Drug Resistance Traits by Extracellular Vesicles. Trends Mol Med. 2015;21(10):595–608. [DOI] [PubMed] [Google Scholar]
- 145.Gan L, Xie D, Liu J, Bond Lau W, Christopher TA, Lopez B, 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(12):968–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Wang J, Li L, Zhang Z, Zhang X, Zhu Y, Zhang C, et al. Extracellular vesicles mediate the communication of adipose tissue with brain and promote cognitive impairment associated with insulin resistance. Cell Metab. 2022;34(9):1264–79 e8. [DOI] [PubMed] [Google Scholar]
- 147.Camino T, Lago-Baameiro N, Bravo SB, Sueiro A, Couto I, Santos F, et al. Vesicles Shed by Pathological Murine Adipocytes Spread Pathology: Characterization and Functional Role of Insulin Resistant/Hypertrophied Adiposomes. Int J Mol Sci. 2020;21(6). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Zhang Y, Mei H, Chang X, Chen F, Zhu Y, Han X. Adipocyte-derived microvesicles from obese mice induce M1 macrophage phenotype through secreted miR-155. J Mol Cell Biol. 2016;8(6):505–17. [DOI] [PubMed] [Google Scholar]
- 149.Song M, Han L, Chen FF, Wang D, Wang F, Zhang L, et al. Adipocyte-Derived Exosomes Carrying Sonic Hedgehog Mediate M1 Macrophage Polarization-Induced Insulin Resistance via Ptch and PI3K Pathways. Cell Physiol Biochem. 2018;48(4):1416–32. [DOI] [PubMed] [Google Scholar]
- 150.Xie Z, Wang X, Liu X, Du H, Sun C, Shao X, et al. Adipose-Derived Exosomes Exert Proatherogenic Effects by Regulating Macrophage Foam Cell Formation and Polarization. J Am Heart Assoc. 2018;7(5). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Pan Y, Hui X, Hoo RLC, Ye D, Chan CYC, Feng T, et al. Adipocyte-secreted exosomal microRNA-34a inhibits M2 macrophage polarization to promote obesity-induced adipose inflammation. J Clin Invest. 2019;129(2):834–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Wang F, Chen FF, Shang YY, Li Y, Wang ZH, Han L, et al. Insulin resistance adipocyte-derived exosomes aggravate atherosclerosis by increasing vasa vasorum angiogenesis in diabetic ApoE(−/−) mice. Int J Cardiol. 2018;265:181–7. [DOI] [PubMed] [Google Scholar]
- 153.Kranendonk ME, Visseren FL, van Herwaarden JA, Nolte-’t Hoen EN, de Jager W, Wauben MH, et al. Effect of extracellular vesicles of human adipose tissue on insulin signaling in liver and muscle cells. Obesity (Silver Spring). 2014;22(10):2216–23. [DOI] [PubMed] [Google Scholar]
- 154.Yu Y, Du H, Wei S, Feng L, Li J, Yao F, et al. Adipocyte-Derived Exosomal MiR-27a Induces Insulin Resistance in Skeletal Muscle Through Repression of PPARgamma. Theranostics. 2018;8(8):2171–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Crewe C, Chen S, Bu D, Gliniak CM, Wernstedt Asterholm I, Yu XX, et al. Deficient Caveolin-1 Synthesis in Adipocytes Stimulates Systemic Insulin-independent Glucose Uptake via Extracellular Vesicles. Diabetes. 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Koeck ES, Iordanskaia T, Sevilla S, Ferrante SC, Hubal MJ, Freishtat RJ, et al. Adipocyte exosomes induce transforming growth factor beta pathway dysregulation in hepatocytes: a novel paradigm for obesity-related liver disease. J Surg Res. 2014;192(2):268–75. [DOI] [PubMed] [Google Scholar]
- 157.Fuchs A, Samovski D, Smith GI, Cifarelli V, Farabi SS, Yoshino J, et al. Associations Among Adipose Tissue Immunology, Inflammation, Exosomes and Insulin Sensitivity in People With Obesity and Nonalcoholic Fatty Liver Disease. Gastroenterology. 2021;161(3):968–81 e12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Gesmundo I, Pardini B, Gargantini E, Gamba G, Birolo G, Fanciulli A, et al. Adipocyte-derived extracellular vesicles regulate survival and function of pancreatic beta cells. JCI Insight. 2021;6(5). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Gu H, Yang K, Shen Z, Jia K, Liu P, Pan M, et al. ER stress-induced adipocytes secrete-aldo-keto reductase 1B7-containing exosomes that cause nonalcoholic steatohepatitis in mice. Free Radic Biol Med. 2021;163:220–33. [DOI] [PubMed] [Google Scholar]
- 160.Ying W, Riopel M, Bandyopadhyay G, Dong Y, Birmingham A, Seo JB, et al. Adipose Tissue Macrophage-Derived Exosomal miRNAs Can Modulate In Vivo and In Vitro Insulin Sensitivity. Cell. 2017;171(2):372–84 e12. [DOI] [PubMed] [Google Scholar]
- 161.Liu T, Sun YC, Cheng P, Shao HG. Adipose tissue macrophage-derived exosomal miR-29a regulates obesity-associated insulin resistance. Biochem Biophys Res Commun. 2019;515(2):352–8. [DOI] [PubMed] [Google Scholar]
- 162.Povero D, Eguchi A, Niesman IR, Andronikou N, de Mollerat du Jeu X, Mulya A, et al. Lipid-induced toxicity stimulates hepatocytes to release angiogenic microparticles that require Vanin-1 for uptake by endothelial cells. Sci Signal. 2013;6(296):ra88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Jiang F, Chen Q, Wang W, Ling Y, Yan Y, Xia P. Hepatocyte-derived extracellular vesicles promote endothelial inflammation and atherogenesis via microRNA-1. J Hepatol. 2020;72(1):156–66. [DOI] [PubMed] [Google Scholar]
- 164.Tomita K, Kabashima A, Freeman BL, Bronk SF, Hirsova P, Ibrahim SH. Mixed Lineage Kinase 3 Mediates the Induction of CXCL10 by a STAT1-Dependent Mechanism During Hepatocyte Lipotoxicity. J Cell Biochem. 2017;118(10):3249–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Hernandez A, Reyes D, Geng Y, Arab JP, Cabrera D, Sepulveda R, et al. Extracellular vesicles derived from fat-laden hepatocytes undergoing chemical hypoxia promote a pro-fibrotic phenotype in hepatic stellate cells. Biochim Biophys Acta Mol Basis Dis. 2020;1866(10):165857. [DOI] [PubMed] [Google Scholar]
- 166.Dorronsoro A, Santiago FE, Grassi D, Zhang T, Lai RC, McGowan SJ, et al. Mesenchymal stem cell-derived extracellular vesicles reduce senescence and extend health span in mouse models of aging. Aging Cell. 2021;20(4):e13337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Xiao X, Xu M, Yu H, Wang L, Li X, Rak J, et al. Mesenchymal stem cell-derived small extracellular vesicles mitigate oxidative stress-induced senescence in endothelial cells via regulation of miR-146a/Src. Signal Transduct Target Ther. 2021;6(1):354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Yoshida M, Satoh A, Lin JB, Mills KF, Sasaki Y, Rensing N, et al. Extracellular Vesicle-Contained eNAMPT Delays Aging and Extends Lifespan in Mice. Cell Metab. 2019;30(2):329–42 e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Alibhai FJ, Lim F, Yeganeh A, DiStefano PV, Binesh-Marvasti T, Belfiore A, et al. Cellular senescence contributes to age-dependent changes in circulating extracellular vesicle cargo and function. Aging Cell. 2020;19(3):e13103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Eitan E, Green J, Bodogai M, Mode NA, Baek R, Jorgensen MM, et al. Age-Related Changes in Plasma Extracellular Vesicle Characteristics and Internalization by Leukocytes. Sci Rep. 2017;7(1):1342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.de Godoy MA, Saraiva LM, de Carvalho LRP, Vasconcelos-Dos-Santos A, Beiral HJV, Ramos AB, et al. Mesenchymal stem cells and cell-derived extracellular vesicles protect hippocampal neurons from oxidative stress and synapse damage induced by amyloid-beta oligomers. J Biol Chem. 2018;293(6):1957–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Luo Q, Xian P, Wang T, Wu S, Sun T, Wang W, et al. Antioxidant activity of mesenchymal stem cell-derived extracellular vesicles restores hippocampal neurons following seizure damage. Theranostics. 2021;11(12):5986–6005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Yang J, Liu XX, Fan H, Tang Q, Shou ZX, Zuo DM, et al. Extracellular Vesicles Derived from Bone Marrow Mesenchymal Stem Cells Protect against Experimental Colitis via Attenuating Colon Inflammation, Oxidative Stress and Apoptosis. PLoS One. 2015;10(10):e0140551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Zhang G, Zou X, Huang Y, Wang F, Miao S, Liu G, et al. Mesenchymal Stromal Cell-Derived Extracellular Vesicles Protect Against Acute Kidney Injury Through Anti-Oxidation by Enhancing Nrf2/ARE Activation in Rats. Kidney Blood Press Res. 2016;41(2):119–28. [DOI] [PubMed] [Google Scholar]
- 175.Viola M, de Jager SCA, Sluijter JPG. Targeting Inflammation after Myocardial Infarction: A Therapeutic Opportunity for Extracellular Vesicles? Int J Mol Sci. 2021;22(15). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Lai RC, Arslan F, Lee MM, Sze NS, Choo A, Chen TS, et al. Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury. Stem Cell Res. 2010;4(3):214–22. [DOI] [PubMed] [Google Scholar]
- 177.Zhang LL, Xiong YY, Yang YJ. The Vital Roles of Mesenchymal Stem Cells and the Derived Extracellular Vesicles in Promoting Angiogenesis After Acute Myocardial Infarction. Stem Cells Dev. 2021;30(11):561–77. [DOI] [PubMed] [Google Scholar]
- 178.Sanchez-Sanchez R, Gomez-Ferrer M, Reinal I, Buigues M, Villanueva-Badenas E, Ontoria-Oviedo I, et al. miR-4732–3p in Extracellular Vesicles From Mesenchymal Stromal Cells Is Cardioprotective During Myocardial Ischemia. Front Cell Dev Biol. 2021;9:734143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Arslan F, Lai RC, Smeets MB, Akeroyd L, Choo A, Aguor EN, et al. Mesenchymal stem cell-derived exosomes increase ATP levels, decrease oxidative stress and activate PI3K/Akt pathway to enhance myocardial viability and prevent adverse remodeling after myocardial ischemia/reperfusion injury. Stem Cell Res. 2013;10(3):301–12. [DOI] [PubMed] [Google Scholar]
- 180.Liu Z, Xu Y, Wan Y, Gao J, Chu Y, Li J. Exosomes from adipose-derived mesenchymal stem cells prevent cardiomyocyte apoptosis induced by oxidative stress. Cell Death Discov. 2019;5:79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Gao Y, Huang X, Lin H, Zhao M, Liu W, Li W, et al. Adipose mesenchymal stem cell-derived antioxidative extracellular vesicles exhibit anti-oxidative stress and immunomodulatory effects under PM2.5 exposure. Toxicology. 2021;447:152627. [DOI] [PubMed] [Google Scholar]
- 182.Xu P, Xin Y, Zhang Z, Zou X, Xue K, Zhang H, et al. Extracellular vesicles from adipose-derived stem cells ameliorate ultraviolet B-induced skin photoaging by attenuating reactive oxygen species production and inflammation. Stem Cell Res Ther. 2020;11(1):264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Shen K, Jia Y, Wang X, Zhang J, Liu K, Wang J, et al. Exosomes from adipose-derived stem cells alleviate the inflammation and oxidative stress via regulating Nrf2/HO-1 axis in macrophages. Free Radic Biol Med. 2021;165:54–66. [DOI] [PubMed] [Google Scholar]
- 184.Gessner A, Koch B, Klann K, Fuhrmann DC, Farmand S, Schubert R, et al. Characterization of Extracellular Vesicles from Preconditioned Human Adipose-Derived Stromal/Stem Cells. Int J Mol Sci. 2021;22(6). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Almeria C, Weiss R, Roy M, Tripisciano C, Kasper C, Weber V, et al. Hypoxia Conditioned Mesenchymal Stem Cell-Derived Extracellular Vesicles Induce Increased Vascular Tube Formation in vitro. Front Bioeng Biotechnol. 2019;7:292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Mao C, Li D, Zhou E, Gao E, Zhang T, Sun S, et al. Extracellular vesicles from anoxia preconditioned mesenchymal stem cells alleviate myocardial ischemia/reperfusion injury. Aging (Albany NY). 2021;13(4):6156–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Collino F, Lopes JA, Correa S, Abdelhay E, Takiya CM, Wendt CHC, et al. Adipose-Derived Mesenchymal Stromal Cells Under Hypoxia: Changes in Extracellular Vesicles Secretion and Improvement of Renal Recovery after Ischemic Injury. Cell Physiol Biochem. 2019;52(6):1463–83. [DOI] [PubMed] [Google Scholar]
- 188.Torralba D, Baixauli F, Sanchez-Madrid F. Mitochondria Know No Boundaries: Mechanisms and Functions of Intercellular Mitochondrial Transfer. Front Cell Dev Biol. 2016;4:107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Islam MN, Das SR, Emin MT, Wei M, Sun L, Westphalen K, et al. Mitochondrial transfer from bone-marrow-derived stromal cells to pulmonary alveoli protects against acute lung injury. Nat Med. 2012;18(5):759–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Sansone P, Savini C, Kurelac I, Chang Q, Amato LB, Strillacci A, et al. Packaging and transfer of mitochondrial DNA via exosomes regulate escape from dormancy in hormonal therapy-resistant breast cancer. Proc Natl Acad Sci U S A. 2017;114(43):E9066–E75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Al Amir Dache Z, Otandault A, Tanos R, Pastor B, Meddeb R, Sanchez C, et al. Blood contains circulating cell-free respiratory competent mitochondria. FASEB J. 2020;34(3):3616–30. [DOI] [PubMed] [Google Scholar]
- 192.Vechetti IJ Jr., Valentino T, Mobley CB, McCarthy JJ. The role of extracellular vesicles in skeletal muscle and systematic adaptation to exercise. J Physiol. 2021;599(3):845–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Lovett JAC, Durcan PJ, Myburgh KH. Investigation of Circulating Extracellular Vesicle MicroRNA Following Two Consecutive Bouts of Muscle-Damaging Exercise. Front Physiol. 2018;9:1149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.D’Souza RF, Woodhead JST, Zeng N, Blenkiron C, Merry TL, Cameron-Smith D, et al. Circulatory exosomal miRNA following intense exercise is unrelated to muscle and plasma miRNA abundances. Am J Physiol Endocrinol Metab. 2018;315(4):E723–E33. [DOI] [PubMed] [Google Scholar]
- 195.de Mendonca M, Rocha KC, de Sousa E, Pereira BMV, Oyama LM, Rodrigues AC. Aerobic exercise training regulates serum extracellular vesicle miRNAs linked to obesity to promote their beneficial effects in mice. Am J Physiol Endocrinol Metab. 2020;319(3):E579–E91. [DOI] [PubMed] [Google Scholar]
- 196.Oliveira GP Jr., Porto WF, Palu CC, Pereira LM, Petriz B, Almeida JA, et al. Effects of Acute Aerobic Exercise on Rats Serum Extracellular Vesicles Diameter, Concentration and Small RNAs Content. Front Physiol. 2018;9:532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Brahmer A, Neuberger EWI, Simon P, Kramer-Albers EM. Considerations for the Analysis of Small Extracellular Vesicles in Physical Exercise. Front Physiol. 2020;11:576150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Bei Y, Xu T, Lv D, Yu P, Xu J, Che L, et al. Exercise-induced circulating extracellular vesicles protect against cardiac ischemia-reperfusion injury. Basic Res Cardiol. 2017;112(4):38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Vishvanath L, Gupta RK. Contribution of adipogenesis to healthy adipose tissue expansion in obesity. J Clin Invest. 2019;129(10):4022–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Jung JW, Kim JE, Kim E, Lee H, Lee H, Shin EA, et al. Liver-originated small extracellular vesicles with TM4SF5 target brown adipose tissue for homeostatic glucose clearance. J Extracell Vesicles. 2022;11(9):e12262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Vechetti IJ Jr., Peck BD, Wen Y, Walton RG, Valentino TR, Alimov AP, et al. Mechanical overload-induced muscle-derived extracellular vesicles promote adipose tissue lipolysis. FASEB J. 2021;35(6):e21644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Nieman KM, Kenny HA, Penicka CV, Ladanyi A, Buell-Gutbrod R, Zillhardt MR, et al. Adipocytes promote ovarian cancer metastasis and provide energy for rapid tumor growth. Nat Med. 2011;17(11):1498–503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Hu W, Xiong H, Ru Z, Zhao Y, Zhou Y, Xie K, et al. Extracellular vesicles-released parathyroid hormone-related protein from Lewis lung carcinoma induces lipolysis and adipose tissue browning in cancer cachexia. Cell Death Dis. 2021;12(1):134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Hu W, Ru Z, Xiao W, Xiong Z, Wang C, Yuan C, et al. Adipose tissue browning in cancer-associated cachexia can be attenuated by inhibition of exosome generation. Biochem Biophys Res Commun. 2018;506(1):122–9. [DOI] [PubMed] [Google Scholar]
- 205.Hu W, Ru Z, Zhou Y, Xiao W, Sun R, Zhang S, et al. Lung cancer-derived extracellular vesicles induced myotube atrophy and adipocyte lipolysis via the extracellular IL-6-mediated STAT3 pathway. Biochim Biophys Acta Mol Cell Biol Lipids. 2019;1864(8):1091–102. [DOI] [PubMed] [Google Scholar]
- 206.Sagar G, Sah RP, Javeed N, Dutta SK, Smyrk TC, Lau JS, et al. Pathogenesis of pancreatic cancer exosome-induced lipolysis in adipose tissue. Gut. 2016;65(7):1165–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Wu Q, Li J, Li Z, Sun S, Zhu S, Wang L, et al. Exosomes from the tumour-adipocyte interplay stimulate beige/brown differentiation and reprogram metabolism in stromal adipocytes to promote tumour progression. J Exp Clin Cancer Res. 2019;38(1):223. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 208.Wernstedt Asterholm I, Tao C, Morley TS, Wang QA, Delgado-Lopez F, Wang ZV, et al. Adipocyte inflammation is essential for healthy adipose tissue expansion and remodeling. Cell Metab. 2014;20(1):103–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Ribeiro-Rodrigues TM, Laundos TL, Pereira-Carvalho R, Batista-Almeida D, Pereira R, Coelho-Santos V, et al. Exosomes secreted by cardiomyocytes subjected to ischaemia promote cardiac angiogenesis. Cardiovasc Res. 2017;113(11):1338–50. [DOI] [PubMed] [Google Scholar]
- 210.Kuriyama N, Yoshioka Y, Kikuchi S, Azuma N, Ochiya T. Extracellular Vesicles Are Key Regulators of Tumor Neovasculature. Front Cell Dev Biol. 2020;8:611039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Di Credico A, Izzicupo P, Gaggi G, Di Baldassarre A, Ghinassi B. Effect of Physical Exercise on the Release of Microparticles with Angiogenic Potential. Applied Sciences. 2020;10(14):4871. [Google Scholar]
- 212.Lou J, Wu J, Feng M, Dang X, Wu G, Yang H, et al. Exercise promotes angiogenesis by enhancing endothelial cell fatty acid use via liver-derived extracellular vesicle miR-122–5p. J Sport Health Sci. 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
