Your new experience awaits. Try the new design now and help us make it even better

REVIEW article

Front. Cardiovasc. Med., 12 November 2025

Sec. Heart Valve Disease

Volume 12 - 2025 | https://doi.org/10.3389/fcvm.2025.1697099

This article is part of the Research TopicInvestigating the Role of Extracellular Vesicles in Cardiovascular Disease MechanismsView all 4 articles

Investigating the mechanisms of small extracellular vesicles in cardiovascular disease using the living myocardial slice platform

  • 1National Heart and Lung Institute, Imperial College London, London, United Kingdom
  • 2Section of Structural and Synthetic Biology, Department of Infectious Disease, Imperial College London, London, United Kingdom

Cardiovascular disease remains the leading cause of death worldwide. Extracellular vesicles (EVs) play a regulatory role in homeostasis, associated with their contribution to cell-cell communication. Recently, it has been confirmed that they also regulate the progression of cardiovascular disease. Specifically, myocardial injury induces an increase in the secretion of small extracellular vesicles (sEVs), both in the cardiac microenvironment and peripheral circulation. Small extracellular vesicles (sEVs) are lipid bilayer particles within the size range of 35–200 nm and are secreted by all cell types. Their high content of bioactive cargo—primarily miRNA—is altered in response to external stimuli, leading to behavioral changes of the recipient cells. In the context of cardiovascular disease, this change leads to acute and long term functional, structural, and biochemical effects on the myocardium. However, the mechanism behind the altered sEVs secretion and their changes in content in the context of cardiovascular disease is yet to be determined. That is partially due to the challenges associated with the isolation of cardiac-derived sEVs, which are essential for the investigation of the mechanisms behind cardiovascular disease progression. Living myocardial slices (LMS) provide an ideal platform for the isolation and investigation of sEVs function in the myocardium. Indeed, LMS not only maintain the cellular complexity and architecture of the native adult myocardium but can also be cultured over days/weeks without significant alterations in cardiac function, making them a reliable model for sEVs isolation and characterization at multiple timepoints. This review aims to summarize recent findings on the effect of sEVs on the onset and progression of cardiovascular disease and to discuss different methods for their isolation from LMSs and the investigation of their functional, structural, and biochemical effect on the myocardium.

1 Introduction

Cardiovascular disease (CVD) is amongst the leading causes of mortality globally (1). Common CVDs include arrhythmia, coronary artery diseases (CAD), valve disease, hypertrophy, stroke, congenital heart disease, and heart failure. In the UK, 7.6 million people are living with heart and circulatory diseases, and 175,000 deaths are reported annually. The cost of CVDs in the UK reaches the substantial amount of 12 billion pounds annually (2). The ageing population and the increase of risk factors associated with CVD, including diabetes, obesity, pulmonary hypertension, sedentary lifestyle, and stress, are causing a rise in CVD prevalence (3, 4). Effective treatment options are limited and involve surgical interventions—heart transplant, valve repair, coronary artery bypass graft (CABG) surgery, primary percutaneous coronary intervention (PPCI)—or medical therapy for the alleviation of symptoms (5, 6). Despite the recent advances in the research field, a lot of gaps still remain in the knowledge behind the molecular mechanisms that induce the onset and progression of CVD, while there is also lack of relevant in vivo and in vitro CVD models.

Extracellular vesicles (EVs)—particularly small EVs (sEVs)—have received attention in the cardiovascular field of research, due to their contribution to cell-cell communication. sEVs are known to be enriched with bioactive molecules and are released by donor cells to be taken up by target cells, where they deliver their cargo. Delivery of molecular contents by sEVs can influence gene expression and cell behavior of recipient cells both in health and disease. Their ability to translocate through the extracellular space and to deliver bioactive molecules in a targeted manner highlights their high potential in the field of therapeutics as tools for drug delivery (7). The shift in their content during disease conditions can provide insight in the presence of biomarkers in specific diseases, allowing their utilization as diagnostic or prognostic tools (79). Even though the sEV field is growing, a lot remains to be understood regarding the mechanisms behind the changes in sEV secretion and uptake and their participation in CVD, in part because of the lack of relevant experimental models. Thus, it is of high importance to develop relevant translational models for the investigation of the mechanistic role of sEVs in CVD.

The living myocardial slice (LMS) model is an ultra-thin layer of myocardium that maintains the multicellularity and extracellular matrix of the adult cardiac tissue, hence preserving the structural and electrophysiological properties of the tissue, whilst continuously secreting sEVs (10). Moreover, LMS can be manipulated to recreate disease models in vitro and investigate sEV dynamics at different timepoints of disease.

This review aims to summarize the recent advances on the effect of sEVs on the onset and progression of CVD, as well as to introduce and discuss the advantages of the LMS model as a powerful tool for the investigation of the mechanistic role of sEVs in CVD. Finally, it provides detailed methodology for cardiac sEV isolation from LMSs, techniques for cardiac sEV characterisation, and for the investigation of their functional, structural, and biochemical effects on CVDs using the human LMS model.

2 Biology of small extracellular vesicles

2.1 sEV definition and content

Extracellular vesicles (EVs) are heterogeneous lipid bilayer nanoparticles of several subtypes, sizes, and origins that contribute to intercellular communication in homeostasis and disease (11). The subtypes differ in terms of their size and biogenesis pathway and include small extracellular vesicles (sEVs) or exosomes, microvesicles, and apoptotic bodies. sEVs range from 35 to 200 nm, they are of endocytic origin and are released by all eukaryotic cell types (7). sEVs can be detected in body fluids, including sweat, blood, saliva, and urine (12).

Within their lipid bilayer, EVs carry bioactive molecules including DNA, proteins, lipids, and RNA, which upon uptake by recipient cells influence their behavior and mediate intercellular communication. microRNA (miRNA)—non-coding RNA that regulates gene expression post-transcriptionally—is the most abundant type of RNA contained in sEVs and has been established as a strong functional mediator in cellular communication (13). Other than their size, sEVs can be distinguished from other microvesicles by the presence of sEV-specific markers, which include the ESCRT complex, MVB-related protein Alix, heat-shock proteins, tumor susceptibility gene 101 protein (TSG101), clathrin, and transmembrane proteins of the tetraspanin family (CD9, CD63, CD81) (12).

2.2 sEV biogenesis pathway

The sEV biogenesis pathway is initiated by the inward budding of the cell membrane to form early endosomes (Figure 1). Bioactive molecules in the intracellular space are enclosed in the lipid membrane and then mature into late endosomes. Late endosomes form intraluminal vesicles (ILVs)—sEV precursors—through inward invagination and are packaged into multivesicular bodies (MVBs) (14). The molecular content and ubiquitination status of MVBs determines their fate, which is either degradation by lysosomes or fusion into the cell membrane to exocytose the sEVs in the extracellular space (15). The mechanisms involved in these processes are very complex and outside of the scope of this review. However, there are some excellent reviews where these mechanisms are explored further (12, 1618). Following detachment from the membrane, the sEVs retain their unique structure, including transmembrane proteins of the tetraspanin family (CD9, CD63, and CD81), which facilitate the loading of bioactive molecules in sEVs and play a crucial role in uptake mechanisms (12, 1518).

Figure 1
Illustration detailing the process of intercellular communication via exosomes between a donor and a recipient cell. The donor cell shows endocytosis, formation of early and late endosomes, and multivesicular bodies. Exosomes containing bioactive molecules are released through exocytosis. These exosomes interact with the recipient cell through fusion, phagocytosis, and direct binding, involving surface antigens and intracellular signaling. Exosome components include nucleic acids, proteins, lipids, adhesion molecules, and tetraspanin, with markers like TSG101 and Alix.

Figure 1. Biogenesis pathway of sEVs. sEVs are formed by the initial inward budding of the plasma membrane, generating the early endosomes. Bioactive molecules in the intracellular space are enclosed into late endosomes, forming intraluminal vesicles (ILVs) and packaged in multivesicular bodies (MVBs). The sEVs are released in the extracellular space by exocytosis to reach the recipient cell. Uptake mechanisms include phagocytosis, fusion, or direct binding with plasma membrane receptors. Adapted with permission from “Biogenesis and Cellular uptake of EVs” by Yuanwang Jia, Li Yu, Tieliang Ma, Wenrong Xu, Hui Qian, Yaoxiang Sun and Hui Shi, licensed under CC BY 4.0.

2.3 sEV uptake mechanisms

Once released from the cell, sEVs travel through the extracellular space to enter circulation and reach other tissues or to be taken up by target cells within the same tissue (19). Uptake mechanisms are achieved by the unique composition of receptors and antigens found in the lipid bilayer of sEVs (14) (Figure 1). Specifically, sEVs can directly fuse to the plasma membrane, delivering their content in the intracellular space of the recipient cell or directly bind to surface receptors and initiate intracellular signaling (20). In many cases, sEVs are taken up by the cell via endocytosis or phagocytosis, transferred in endosomes, and re-released in the intracellular space to secrete their contents or to undergo lysosomal degradation (21, 22).

2.4 sEV role in homeostasis and alterations in CVD

The miRNA content that is received by the recipient cell can affect its gene expression profile and in turn its phenotype. sEV content can differ depending on the cell type of origin and the state of the tissue releasing it—homeostatic or diseased. Under normal conditions, sEVs participate in cell-cell crosstalk to regulate major pathways, including proliferation, inflammation, angiogenesis, fibrosis, and apoptosis, to maintain homeostasis. Under stressful conditions or upon injury, the concentration and content of sEVs changes to impact the recipient cell's behavior accordingly as a response to the external stimulus. sEVs secreted by different cardiac cell types exert diverse influences on the recipient cells, due to their differential bioactive molecule content. These effects are structural, molecular, and eventually functional, and can vary depending on disease severity and stage. It is essential to investigate these effects to understand the role of sEVs on CVD progression (23).

3 sEV functional, structural, and biochemical alterations in CVD

3.1 sEVs increase in circulation in CVD

The role of sEVs in CVD progression is of high interest due to their participation in intercellular communication. The myocardium is comprised primarily of cardiomyocytes (CMs), cardiac fibroblasts (CFs), endothelial cells (ECs), vascular smooth muscle cells, and immune cells (24). The continuous secretion and uptake of sEVs leads to the interaction of the cardiac cell types and the influence of their behavior for the maintenance of homeostasis and for response to stressful conditions (25, 26). Depending on the cell type of origin and the conditions upon secretion, the sEV concentration and miRNA content will differ. Emanueli et al. demonstrated that following CABG surgery, the concentration of sEVs of cardiac origin and onto peripheral circulation rises and that their cardiac miRNA content is increased (27). It has also been confirmed that cardiac miRNA levels are increased in the plasma of acute myocardial infarction (MI) patients (9). More recently, this was also demonstrated in patients of ST-elevation myocardial infarction (STEMI)—the most severe type of acute MI. Following reperfusion via PPCI, the plasma of 42 patients was collected for circulating sEVs' isolation. Quantification of sEVs showed their increase in STEMI patients and in patients subjected to late reperfusion. Further investigations in sEV content and marker expression can be informative regarding their role in disease severity and progression. In this same study, it was observed that the sEV expression of platelet-specific markers showed a positive correlation with ejection fraction and a negative correlation with the degree of microvascular obstruction, evaluated with cardiac magnetic resonance (28). It is also interesting that depending on the clinical setting, the effect of sEVs may differ. This was demonstrated in a study investigating the effect of sEVs derived from the serum of diabetic mice on non-diabetic mouse models subjected to MI reperfusion (MI/R) injury. Specifically, the intramyocardial injection of sEVs aggravated the damage of MI/R, indicated by diminished functional recovery and an increase in scar size and CM death (29).

Work has also been performed for larger EV types. A study exposed Langendorff-perfused rat hearts to ischemia and reperfusion and isolated microvesicles from the coronary perfusate to investigate their response to the injury. Quantification of an established EV marker confirmed an increase in the concentration of microvesicles (30). This was also demonstrated in mice subjected to acute MI by coronary artery ligation. In this study, EVs were isolated from LV tissue and large and small EVs were quantified separately, to confirm a higher concentration for the large EVs. These findings were also confirmed in clinical samples collected from patients that had undergone aortic valve replacement (31).

3.2 sEV content shifts in CVD

3.2.1 sEVs released by CMs

The altered sEV release and protein content from CMs in response to CVD can affect pathways of angiogenesis, apoptosis, inflammation, proliferation, and fibrosis (Figure 2). Gupta et al. first showed that CMs release sEVs. The aim of the study was to understand the secretion pathway of heat shock protein 60 (HSP60), which is a protein that protects tissues during injury. Their findings demonstrated that adult CMs secrete HSP60 via exosomes and that hypoxia-reoxygenation injury (HRI) increased the secretion of HSP60-containing exosomes by CMs by three times compared to healthy conditions (32).

Figure 2
Diagram illustrating interactions between cardiomyocytes, fibroblasts, and endothelial cells. Cardiomyocytes are affected by hypertrophy, apoptosis, inflammation, and fibrosis. Fibroblasts undergo proliferation, inflammation, and fibrosis. Endothelial cells experience proliferation and angiogenesis. Arrows indicate interactions between these cell types.

Figure 2. sEV-mediated regulation of biological processes. sEVs are produced by all cardiac cell types. Their content mediates intercellular communication, which results in the influence of several pathways, including the proliferation, inflammation, fibrosis, apoptosis, and angiogenesis. Arrows indicate the direction of sEVs released from cardiomyocytes (red), endothelial cells (blue), and fibroblasts (orange).

Ever since, others have shown that following ischemia-reperfusion injury (IRI), CMs secrete sEVs that play a role in cell-cell crosstalk. Chen et al. demonstrated that in a murine model of HRI, the triggering of the eNOS pathway—cardioprotective for ECs—can occur following the uptake of CM-derived sEVs by ECs (33). Importantly, the impact of miRNAs carried by CM-derived sEVs on other cardiac cell types has become a significant area of research, with many of these miRNAs now recognized as cardiac-specific. Anselmo et al. (34) recently demonstrated that the surface antigen CD172a is specifically expressed on EVs secreted by cardiomyocytes (CMs), enabling the identification of the cellular origin of sEVs. These sEVs were found to be enriched with cardiac-specific miRNAs and were produced in response to cellular stress triggered by hypoxic conditions. Additionally, the study revealed that patients with aortic stenosis who underwent transcatheter aortic valve replacement and had elevated levels of CD172a+ EVs in their peripheral blood showed improved survival outcomes (34).

The miRNAs that are released from CMs can have a significant role in disease progression. Yang et al. (35) demonstrated that hypoxic conditions lead to elevated levels of miR-208a in rat CMs. Furthermore, when exosomes from these hypoxic CMs were co-cultured with CFs, the CFs showed increased levels of miR-208a, which stimulated their proliferation and myofibroblast activation, thereby aggravating fibrosis (35). In terms of CM–EC communication, sEVs released from glucose-deprived CMs were found to be enriched in miR-17, miR-20, miR-23a, miR-23b, miR-30c, and miR-126-3p. Upon uptake by ECs, these miRNAs enhanced EC proliferation and promoted angiogenesis (36).

3.2.2 sEVs released by CFs

CFs play a key role in triggering and sustaining inflammation and tissue remodeling after cardiac injury. As part of this process, they secrete sEVs enriched with miRNAs that can modulate the behavior of other cell types. Bang et al. demonstrated that exosomes isolated from CFs of mice subjected to transverse aortic constriction, when applied to cultured CMs, induced hypertrophic response in the CMs. Further analysis confirmed that CMs internalised these CF-derived exosomes, which delivered miR-21_3p - a CF-specific miRNA known to drive stress-induced hypertrophy (106).

Cosme et al. conducted a study to assess how sEVs from CFs exposed to HRI affect CM viability. Proteomic comparisons between sEVs from control and hypoxic CFs revealed 71 proteins with significantly increased levels and 73 with decreased levels, many of which were associated with fibrotic and tissue remodeling pathways. The study also demonstrated that the impact of CF-derived sEVs on CMs varied depending on the state of the recipient cells upon administration. When applied to healthy CMs, the sEVs enhanced cell survival, whereas their application after HRI led to diminished viability (37).

Another study demonstrated that CF-derived sEVs released during HRI contain miR-133a. This miRNA was found to inhibit the expression of embryonic lethal abnormal vision like 1 (ELAVL1), a protein known for its pro-inflammatory role. By downregulating ELAVL1, miR-133a contributed to decreased inflammation and reduced apoptosis in CMs (38).

3.2.3 sEVs released by ECs

ECs are key components of blood vessels, playing a crucial role in mediating communication between the bloodstream and surrounding tissues. During ischemic events, blood flow is restricted, leading to EC injury and the subsequent release of sEVs (24). In one study, sEVs derived from stem cell-derived ECs exposed to hypoxic conditions were found to suppress apoptosis and promote angiogenesis. This, in turn, enhanced CM survival and contractile function in a human IRI heart-on-a-chip model (39).

A study examining cultured human ECs found that hypoxic conditions alter the proteomic composition of their secreted EVs. Notably, there was an increase in proteins such as lysyl oxidase-like 2 (LOXL2), fibronectin, and collagen, suggesting that these EVs play a role in promoting inflammation, with a particular emphasis on tissue remodeling and fibrotic processes (40).

Davidson et al. demonstrated that co-culturing human umbilical vein ECs with CMs prior to HRI significantly enhanced CM survival. This protective effect was attributed to the exosomal content released by ECs, which activated the ERK1/2 MAPK signaling pathway in the CMs (41). The existing literature is summarized in Table 1.

Table 1
www.frontiersin.org

Table 1. Overview of cardiac miRNA effects on proliferation, apoptosis, inflammation, angiogenesis, and fibrosis pathways.

3.3 Gaps in the sEV field

Despite the extensive literature in the role of sEVs in CVDs, there are still fundamental gaps in the field, both in the context of homeostasis and disease, that do not allow the understanding of sEVs' role in disease progression and prevention. Specifically, there are questions regarding the basic biology of EVs, due to the high heterogeneity of the molecular content and structure (12). Moreover, even though there has been an increased understanding behind the several sEV uptake mechanisms, which include surface receptor interactions with the recipient cell and by internalization of the sEVs by phagocytosis or clathrin-mediated uptake, it is still not well understood how these mechanisms change in response to disease. It is of high interest to establish the mechanistic shift of sEV secretion, uptake, and content change in CVD. The remaining questions are due to the challenges in studying sEVs and the lack of in vivo and in vitro experimental models that can overcome them (11).

4 Challenges in studying cardiac-derived sEVs

4.1 sEV isolation and characterisation

There are many factors that make sEV research challenging. Firstly, sEVs are nanosized particles that are difficult to isolate and quantify. Their small size obstructs the accurate determination of their identity and origin. Another challenge is their heterogeneity. EVs are a large population of particles of multiple subtypes, sizes, and origins (11). Even though each population expresses specific markers and is within specific size ranges, there is still an overlap. For example, sEVs express the tetraspanin proteins, but they are also expressed by microvesicles at lower levels (42). Additionally, the size limits are not strict. sEVs range from 30–200 nm, while microvesicles from 100–1,000 nm. Thus, it is essential that appropriate techniques are used to accurately isolate and characterize different EV populations, while also separating them from other non-vesicular populations (43). Finally, the molecular heterogeneity of sEVs is also a challenge, especially in the context of disease. The temporal dynamics of sEV secretion and content change of different cell types but also within the same at the different stages of disease progression make their characterisation more challenging. Recently, sEV separation and characterisation technologies have progressed to improve isolation purity and yield, however it still remains a challenge to achieve both quantity and specificity (11). These challenges can be minimized by following the minimal information for studies of extracellular vesicles (MISEV), which proposes strategies for the isolation and characterisation of EVs, depending on the source, type, and general characteristics (11, 43).

4.2 Lack of relevant experimental models

The gaps in understanding the role of sEVs in CVD are in part due to the lack of appropriate experimental models. When studying the functional role of sEVs, the choice of model is crucial, as it directly influences the quality, relevance, and interpretability of the sEVs produced. In the context of CVD, it is particularly important to be able to obtain pure cardiac-derived sEVs specifically. Consequently, the more physiologically relevant the model, the more representative and informative the sEVs obtained will be. There is a critical need for experimental systems that not only reflect the pathophysiology of CVD but also enable the isolation and investigation of myocardium specific-derived sEVs, to uncover their role in disease onset and progression.

Several experimental models have been employed to study sEVs in CVD, each showing defects in terms of physiological relevance and feasibility. Studies that use monolayers of cells fail to include the multicellular and extracellular matrix components of the cardiac tissue, making them minimalistic models (44). Consequently, the sEVs derived from these models are of lower numbers and from limited cell types. It is also important to consider the origin of cellular models. Many studies use cells that are derived from animal neonatal stem cells or induced pluripotent stem cells (iPSCs), which show substantial differences from adult human cardiac cells in terms of metabolism and Ca2+ handling and sEV secretion and content (45).

Engineered heart tissue, organoids, or organ-on-a-chip models are three dimensional (3D), hence eliminating the limitations of monolayers of cells. They include more cell types and extracellular components, thus incorporating an increased complexity in the model (46). However, most of these models can only be recreated using neonatal CMs and stem cell-derived CMs, who exhibit fetal maturity. Moreover, these models lack the biomimetic environment that is crucial for maintenance of the myocardium in vivo, such as mechanical load and electrical stimulation. These factors play a significant role in cardiac cell maturation and function and will reflect the profile of the sEVs they produce (47).

The performance of in vivo studies using animal models adds an increased complexity in the investigation, due to the involvement of inflammation and the participation and response of other organs, affecting the applicability of the results and subsequent conclusions (44). The secretion of sEVs from other organs increases the heterogeneity of the sEV populations isolated, complicating characterisation. Animal models also display differences in cardiac size and physiology, compared to humans, while CVD phenotypes may also vary (48). This makes the findings less relevant and translational.

5 The living myocardial slice model

5.1 Living myocardial slice: the model of intermediate complexity

The living myocardial slice (LMS) model is an ultra-thin layer of myocardium (300–350 μm). It is considered a model of intermediate complexity, as it maintains the multicellular and extracellular matrix components of adult cardiac tissue, while avoiding the high complexity of animal models. This suggests that they preserve the electrophysiological properties of the native tissue, as they represent its functional, structural, mechanical, and biochemical properties (10).

The LMS model can be prepared from several species, including rat, rabbit, lamb, pig, and human, as previously described (10, 44, 49, 105). The method of human LMS preparation involves different steps (Figure 3). Briefly, following heart explant and dissection, a left ventricular (LV) block of 1 cm3 is placed in a high-precision vibratome, which precision-cuts the LV in 300 μm thick layers. To generate the LMS, each layer is trimmed to achieve 1 cm2 slices, followed by the attachment of polyactic acid (PLA) rings perpendicular to the fibers' direction to not hinder contraction.

Figure 3
Series of images depict the process for the generation of the human LMS model using a vibratome. The first step (top left) shows an LV section, highlighting a specific tissue area. This area is the LV tissue block that is placed on the vibratome (top right). The vibratome precision-cuts the LV tissue block (bottom right) into 300-micrometer sections. Then follow the LV section trimming and ring attachment (bottom middle). The last image in the process (bottom left) depicts the LMS model ready for culture or experimentation.

Figure 3. Generation process of the human LMS model. The human donor heart is dissected to retrieve an LV block of 1 cm3. The vibratome precision-cuts the block to obtain layers of 300 μm thickness. The slice is aligned under the microscope for the positioning of the PLA rings perpendicular to the muscle fibers. The LMS is ready to culture.

LMS can then be cultured in biomimetic conditions using isometric chambers. Specifically, the LMSs are placed in customized stretchers that allow application of mechanical preload by stretching the slices at different sarcomere lengths. In that way, we provide a degree of mechanical load to closely recapitulate mechanical preload. The chambers can then be supplemented with pure O2 or 95% CO2 and media rich in hormones, nutrients, and salts, which is constantly circulated through the chamber. Finally, electrical stimulation is delivered throughout the culture period (10, 44). These culture conditions have been optimised to allow for the long-term survival of the LMS model, as its thin structure allows for sufficient O2 and metabolite diffusion (50). Under these conditions the LMS model continues to secrete sEVs throughout the entire culture duration, that can be later isolated from the media for further investigation.

5.2 Using the LMS to model CVD

The LMS model can be used to model CVD and investigate sEV dynamics at different stages of disease, allowing its use as a platform to investigate mechanisms of biological processes. The use of diseased or failing hearts will allow preservation of the pathological characteristics of the tissue, as they similarly maintain the functional and structural properties of the disease. Alternatively, the LMS model can be manipulated to recapitulate CVD, by either altering the culture conditions or artificially recreating the disease condition, depending on the desired phenotype (44, 51). In any case, it is important to consider that a single LV piece can generate a sufficient number of LMSs to allow for reproducible experiments and potential replacement of animal models.

The LMS model has been used to simulate different CVD. For example, manipulating the amount of preload during culture by overstretching the LMS using the stretchers results in the pathological effects observed in heart failure (51). Moreover, procedures can be performed on the LMSs to induce injury. Dries et al. used rat LMSs to induce cryoinjury and observe the effects after culture (52). Ischemia-reperfusion or hypoxia-reoxygenation injury can be induced by the manipulation of O2 levels in culture (53, 54).

5.3 Limitations of the LMS model

Despite the numerous advantages of the LMS model, there are a few points to consider. Firstly, the LMS model preparation requires practice of several months before obtaining appropriate and viable samples for experimentation. This is primarily due to the skills required for the dissection and slice trimming technique, appropriate positioning of the PLA rings, and careful handling of the LMSs during stretching for culture and recordings (44). Additionally, as the slicing progresses from endocardium to epicardium, there is increasing heterogeneity of cellular populations depending on the depth of the LV block. This may include increased variability and can be accounted for by the randomization of the LMSs. Moreover, even though the LMS platform retains the structure of microvessels, they are not functional as they are not perfused by blood and oxygen throughout (10). Another limitation includes the difficulty in identifying the appropriate O2 levels to account for lack of blood flow across vessels and the absence of hemoglobin to deliver O2 to the tissue. In fact, in the LMS, oxygenation is achieved through diffusion. To account for this variability, the use of O2 electrodes is recommended for the monitoring of the O2 concentration real-time, and it is worth considering in hypoxia studies.

6 Isolation and characterisation of cardiac sEVs from LMS

6.1 Isolation of sEVs from LMS

The isolation method of sEVs should be determined based on their source, biophysical and biochemical properties, specificity, and desired yield. The most widely used isolation and separation methods include differential ultracentrifugation, density gradient centrifugation, size exclusion chromatography (SEC), precipitation, ultrafiltration, label-free microfluidics, immune-precipitation and affinity precipitation (20). Each of these methods offers different advantages and limitations, associated with the yield, heterogeneity, and purity of the obtained sEVs. For example, precipitation and ultracentrifugation are associated with high yield, however they isolate a mixture of extracellular particles, reducing the specificity of the EVs obtained. Similarly, differential ultracentrifugation isolates EVs depending on their size, but co-isolates non-vesicular extracellular particles. Hence, it is suggested to combine different methods to exploit the advantages of multiple techniques (11, 43). The combination of ultrafiltration with size exclusion chromatography (SEC) achieves the efficient isolation of EVs from conditioned media. It has been previously demonstrated that the combination of ultrafiltration and SEC for the isolation of EVs from culture media achieves a similar or even higher yield and purity compared to ultracentrifugation (55, 56).

Following preparation and culture of LMS, the isolation of cardiac sEVs would be achieved from the conditioned medium, in which the LMSs are cultured. The ultrafiltration strategy involves the utilization of membranes that separate the sEVs based on their molecular weight cut-off (MWCO), which is estimated to be 10–100 kDa for sEVs (57, 58). The centrifugal concentrators contain a 10,000 kDa MWCO filtration membrane that, following centrifugation reduces the final volume of the media to less than 1 ml (59).

The concentrated media then undergoes SEC. SEC utilizes polymers to create a porous stationary phase in a chromatographic column, where sEVs are separated in different path lengths according to their size. The principle is that the larger particles pass through the column first, while the smaller particles are trapped in the pores, and therefore eluted in the later fractions. The principle of the isolation techniques is that they separate smaller from larger EVs. Thus, depending on the desired size and type of EV, columns of the appropriate recovery range should be utilized (20). For the collection of sEVs, the initial fractions of the eluate are discarded, while the last fractions are the sEV-enriched. Multiple studies have used SEC to isolate cardiac sEVs from the culture media of human iPSC-CMs and cardiac fibroblasts. Recently, a study isolating sEVs from the conditioned media of human cardiac fibroblasts used ultrafiltration, followed by SEC, to achieve a high yield of cardiac sEVs (55). The combination of these two techniques achieves high sEV yield, integrity, and specificity in a short time and with limited equipment required (11, 20, 56, 60).

6.2 Characterisation of isolated sEVs from LMS

According to the MISEV, the established sEV characterisation parameters include quantity, size, sEV marker expression, and shape (11, 43), as illustrated in Figure 4. These parameters ensure that the nanoparticles collected are of sEV identity.

Figure 4
Flowchart illustrating the process of small extracellular vesicle (sEV) analysis. It begins with sEV collection from conditioned media, followed by isolation using ultrafiltration and size exclusion chromatography (SEC). Characterization involves nanoparticle tracking analysis, NanoFCM with graphs of size and particle concentration, ExoView showing CD81, CD63, CD9, and transmission electron microscopy displaying vesicle images.

Figure 4. Overview of sEV isolation and characterisation methods. The LMS conditioned media is collected from the culture chamber. The sEV isolation is achieved via ultrafiltration and SEC. The isolated sEVs are characterised based on quantity and s size distribution, marker expression, and shape. Representative images of the output of NTA, NanoFCM, ExoView, and images of sEVs visualized under Transmission Electron Microscopy. “ExoView” reproduced with permission from “Representative ExoView images of indicated immunophenotypically captured and detected Serum 10 k xg EVs” and “Transmission Electron Microscopy” adapted with permission from “Quality control of EVs utilised for comparative NTA device analysis” by Daniel Bachurski, Maximiliane Schuldner, Phuong-Hien Nguyen, Alexandra Malz, Katrin S Reiners, Patricia C Grenzi, Felix Babatz, Astrid C Schauss, Hinrich P Hansen, Michael Hallek and Elke Pogge von Strandmann, licensed under CC BY-NC 4.0.

6.2.1 Nanoparticle tracking analysis (NTA)

Nanoparticle Tracking Analysis (NTA) is a widely used technique for the characterisation of sEVs. NTA tracks average concentration, average size, and particle-size distribution of nanoparticles. NTA uses the Nanosight NS300 (Malvern Instruments) to quantify concentration and size of nanoparticles ranging from 30 to 1,000 nm. The sEVs are irradiated with a laser beam and visualised by the light scattered. This technique also provides the fluorescent mode, which allows detection of individual fluorescently labelled nanoparticles (61). The instrument is equipped with three light sources of different wavelengths (450 nm, 520 nm, and 635 nm). The NTA software applies the Stokes-Einstein equation to measure the size of the particles (62). It achieves that by tracking the Brownian motion, which is defined as the movement caused by the random collisions of nanoparticles that are dispersed in a liquid. Specifically, the presence of sEVs in a sample is calculated using the equation D = kBT/6πηr, where D is the diffusion coefficient, kB is the Boltzmann constant, T is the absolute temperature, and η is the dynamic viscosity of the liquid, to determine r, which is the radius of the nanoparticle. For the acquisition of recordings, sEVs need to be diluted accordingly, to preserve osmolality of the sEVs and ensure accurate detection and quantification, according to the manufacturer (63). The NTA can been used for EV samples derived from various sources, including plasma, urine, and cardiac cells in culture (27, 64, 65).

6.2.2 Nano flow cytometry

Nano flow cytometry is another standardised method for EV quantification and size distribution analysis. The NanoFCM Flow NanoAnalyzer is a nano flow cytometer that uses light scattering to measure concentration and size distribution of nanoparticles ranging from 7 to 1,000 nm in size at a single nanoparticle level. The system also offers the option of fluorescence labeling, as it is equipped with lasers of multiple wavelengths. It is a great tool to identify specific sEV populations with the use of tetraspanin proteins, and with other markers depending on their origin. For example, it has been recently established that CD172a is a cardiac sEV-specific marker (34). This offers an additional characterisation step, allowing the quantification of sEV-sized particles that are positive for cardiac sEV markers.

Due to its high sensitivity, many attempts of optimisation are required to determine the concentration of sEVs that allow their optimal staining, as well as their detection and accurate measurement when using the NanoFCM Flow NanoAnalyzer. The optimisation process should take into account several parameters, including sEV size, concentration, chemical attributes, and markers tested. For the staining, it is recommended to test different antibody concentrations, sEV concentrations, incubation timings, and incubation temperatures. Following staining, the sample should be washed to remove free dye. Once optimised, the sample dilution used for NanoFCM Flow NanoAnalyzer recordings is also important.

This has been performed for sEVs isolated from various biofluids, cultured cells, and cells isolated from tissue (6668). NTA and nano flow cytometry individually provide high-sensitivity readouts, however their accuracy can be influenced by parameters including sample heterogeneity, source, and isolation method (69). Thus, the combination of NTA and nano flow cytometry eliminates or reduces the limitations associated with each method individually (70).

6.2.3 Exoview

The ExoView R100 is a system that is utilized to visualize, quantify, and analyze the expression of sEV markers at a single-particle level. It uses chips that contain antibody capture spots which, following the immunofluorescent staining of sEVs, indicate the presence and quantity of the tetraspanin markers attached to the chip. This system uses an interferometric reflectance imaging sensor, which achieves high resolution images to detect and count individual sEVs bound on the chip, measure their size, and quantify marker expression (71).

The ExoView has been used extensively for the characterization of EVs isolated from tissues including cardiac, foreskin, and lung fibroblasts, neuronal stem cells, but also from fluids, including plasma and bronchoalveolar lavage fluid (7275).

6.2.4 Transmission electron microscopy

Transmission electron microscopy (TEM) is used as another common step of sEV characterisation. This is particularly important for the identification of the characteristic shape of sEVs, while also enabling the evaluation of the presence of sEV markers (43). It is a widely used method for the visualization and characterization of nanoparticles, as it offers the acquisition of higher magnification than the light microscope (76). TEM has been performed for the visualization of sEVs isolated from the media of cultured CMs of adult mouse hearts, but also from human plasma (66, 77). However, it is a high-cost technique and requires expertise.

6.3 Application of sEVs on LMS

Following quantification and characterisation of LMS-derived sEVs, it is important to investigate their mechanistic effect on LMS. This can be achieved by the application of sEVs on LMS. The sEVs are absorbed by the LMS and can influence them, according to the stage of disease they were collected at, as well as the health state of the LMS—healthy or diseased. A potential technique for the application of sEVs on LMS would be their administration on the air-liquid interface of the LMS (Figure 5). Specifically, media can be removed to expose the top surface of the LMS to air, while the bottom surface will still be in contact with the media. The volume containing the desired sEV concentration can be applied on the LMS. For this stage, dose-response experiments of sEVs on LMS are recommended, for the determination of the optimal sEV concentration. Electrical stimulation, media circulation, and oxygenation should be interrupted, to avoid dispersion of the sEVs in the media. Following absorption, the media can be added on the chamber and electrical stimulation, media circulation, and oxygenation can be resumed.

Figure 5
Diagram of a culture chamber with labeled components. The LMS framework is indicated above the air-liquid interface, which separates the media from the air. A stretcher is shown within the chamber, alongside PLA rings. Arrows point to each component for clarity.

Figure 5. Setup of the air-liquid interface of sEVs treatment on LMS. Media is removed from the culture chamber to expose the top surface of the LMS to air. The sEVs are applied on the surface for their absorption by the LMS. Electrical stimulation, media circulation, and oxygenation are interrupted. Following absorption, media is added to achieve the desired volume and electrical stimulation, media circulation, and oxygenation are resumed.

7 Investigation of functional, structural, and biochemical effects of sEVs on LMS

7.1 Investigation of functional effects of sEVs on LMS

The set of experiments that can be performed in terms of function, structure, and molecular profile of LMSs following sEV treatment are summarized in Figure 6. Functional studies are essential to understand the health state of the tissue and to investigate the effect of disease. Function can be evaluated by looking at contraction kinetics, live Ca2+ imaging, and conduction velocity. The investigation of the functional properties of LMS has been described previously but has not been performed on human LMS that have been treated with LMS-derived cardiac sEVs. However, the protocols and principles remain the same.

Figure 6
Diagram summarizing studies on small extracellular vesicles (sEV) and their treatment on LMS. The left portion outlines biochemical studies on sEVs, including RNA extraction and miRNA analysis. A central graphic shows sEV treatment on cardiac muscle tissue. The right portion includes functional studies on LMS, with graphs and imaging of contractility and conduction velocity, and structural studies using immunofluorescence staining to investigate differential gene expression.

Figure 6. Overview of functional, structural, and biochemical studies on LMS treated with sEVs. The investigation of differential miRNA expression on sEVs can be performed by miRNA arrays. Following the treatment of LMS with cardiac sEVs, functional studies (contractility, live Ca2+ imaging, and conduction velocity), structural studies, and biochemical studies can be carried out on the LMS. Contractility recording adapted with permission from “Contractility recordings and representative confocal images of living myocardial slice” by Filippo Perbellini and Thomas Thum, licensed under CC BY-NC. Live Ca2+ adapted with permission from “Viability in freshly sliced LMS and after 24h of culture” by Eef Dries, Ifigeneia Bardi, Raquel Nunez-Toldra, Bram Meijlink and Cesare M. Terracciano, licensed under CC BY 4.0. Conduction velocity reproduced with permission from “Conduction, connexins, β-adrenergic stimulation and arrhythmogenesis in rat myocardial slices cultured for 24 h. a, b Representative images of longitudinal and transverse conduction maps of a 0 h rat myocardial slice. Black arrows show myocardial fibre direction. Yellow star = location of the stimulating electrode. Area of conduction map: 4.9 × 4.9 cm” by Samuel A. Watson, James Duff, Ifigeneia Bardi, Magdalena Zabielska, Santosh S. Atanur, Richard J. Jabbour, André Simon, Alejandra Tomas, Ryszard T. Smolenski, Sian E. Harding, Filippo Perbellini and Cesare M. Terracciano, licensed under CC BY 4.0. Immunofluorescence staining adapted with permission from “Imaging LMS” by S. A. Watson, A. Dendorfer, T. Thum and F. Perbellini, licensed under under CC BY 4.0. Investigation of differential gene expression adapted with permission from “Electromechanical regulation of gene expression in rat myocardial slices cultured for 24 h” by Samuel A. Watson, James Duff, Ifigeneia Bardi, Magdalena Zabielska, Santosh S. Atanur, Richard J. Jabbour, André Simon, Alejandra Tomas, Ryszard T. Smolenski, Sian E. Harding, Filippo Perbellini and Cesare M. Terracciano, licensed under CC BY 4.0.

7.1.1 Contractility studies

Functional studies for the contractility and kinetics evaluation of the LMSs are performed using the force transducer (FT) apparatus. The FT setup in our laboratory has been updated to provide biomimetic conditions, while allowing real-time measurements of functional parameters (10). The FT is wired to an amplifier that conveys the force signal to the transducer for processing.

From the force transient, parameters such as passive force, which measures the diastolic force generated upon stretch, and the active force, which expresses the maximum systolic force generated upon systole can be extrapolated. These values are normalised by the cross-sectional area of each LMS. Kinetics parameters include the time to peak (TTP), time to 50% and 90% decay, and the decay rate constant (tau) (10, 44).

The recording of these parameters is measured at multiple stretches of the sarcomere, by manipulating the length of the LMS. It is particularly interesting to investigate the functional changes under different sarcomere lengths, as according to the Frank-Starling Law, increasing the stretch of the sarcomere in healthy myocytes within physiological ranges increases the force they generate upon contraction (80).

Upon the start of the experiment, the slack length of the LMS is measured, and is gradually stretched horizontally to achieve different lengths throughout the recording. Specifically, the functional parameters of the LMS are recorded and measured at unloaded, 5%, 10%, 15%, 20%, 22% (physiological), 25%, and 30% stretch (10, 44).

Functional studies of LMS have been performed extensively in our laboratory for several disease models. Rat LMS have been used to evaluate the functional effects of the inhibition of calcium/calmodulin-dependent protein kinase II in cryo-injured LMS (52). Another study has investigated the effect of different sarcomere lengths following 24 h in culture on the contraction amplitude of rat LMS. In the same study, human failing hearts were used for the generation of LMS to evaluate the effect of electromechanical stimulation on their function. It was reported that the optimal preload applied in culture is equivalent to a sarcomere length of 2.2 μm (78). From these studies, it is evident that the LMS model can be used for the performance of contractility experiments, while providing an insight for many models of disease.

7.1.2 Live Ca2+ imaging studies

The evaluation of live Ca2+ imaging provides valuable information regarding the electrophysiological capacities of the LMS in homeostasis and injury, including Ca2+ transient kinetics and intracellular Ca2+ concentrations. These parameters can be affected by disease, thus providing an insight into the effects of CVD progression. This is achieved with the use of fluorescent indicators that are loaded in the cells and upon illumination, will fluoresce with every contractile cycle of the LMS.

Common fluorescent probes include Fluo-4AM and Fluo-8AM, which have been previously applied in rat LMSs (52). Other Ca2+ indicators include Rhod-AM, which has been used for swine, rabbit, and human models (8183). It is important to note that Rhoad2AM can also detect mitochondrial Ca2+ activity, thus Fluo-4AM or Fluo-8AM might be more reliable (84). Also, during the incubation with the fluorescent indicator, the LMS should remain on the stretcher to maintain the physiological load. It is important to use blebbistatin or 2,3-butanedione monoxime to inhibit the activity of myosin (79), thus allowing the successful loading of the indicator.

Recently, Ca2+ kinetics were evaluated in a human LMS model. Specifically, the researchers were interested in the electrophysiological changes that occur in the LMS model following long-term culture of approximately 10 days. Thus, they used Rhoad2AM to evaluate the Ca2+ handling properties of the LMS and were able to identify an improved function of the LMS following culture, confirmed by the increased Ca2+ transient amplitude and reduced duration. This shows that the investigation of Ca2+ dynamics is possible with human LMS (83). Even though the effects of the application of LMS-derived cardiac sEVs on human LMSs on the Ca2+ activity have not been previously evaluated, the protocol should be the same and can be applied following sEV application and desired culture duration.

7.1.3 Conduction velocity studies

Conduction velocity (CV) studies estimate the speed at which the electrical impulse travels through the cardiac tissue, to induce an action potential. The investigation of CV is essential, as it has shown to be influenced in CVD (85). The LMS model can be used as a platform to study CV by its attachment on a multi-electrode array (MEA) system. CV can be measured longitudinally or transversally to myofiber orientation. In our laboratory, conduction velocity studies have been optimized for rat and human LMSs (78, 86). Briefly, the MEA used is the MEA1060 (Multi Channel Systems), which contains 60 microelectrodes in an 8 × 8 arrangement. The LMS is attached in the center of the system, in combination with a holder to improve its interaction with the electrodes. During the recording, the sample is perfused with oxygenated Tyrode solution and maintained at 37°C. Electrical stimulation at a frequency of 1 Hz from different MEA microelectrodes and with the lowest voltage and duration to achieve the threshold allows field potential recording for CV evaluation. Arrhythmia and spontaneous field potentials can also be studied. As with contractility and Ca2+ studies, this protocol can be repeated following sEV application.

7.2 Investigation of structural effects of sEVs on LMS

7.2.1 Immunostaining of LMS treated with sEVs

Prior to application on LMS, sEVs can be stained to offer the option of visualization on the LMS for structural studies. There are staining kits that have been widely used. For LMS-derived cardiac sEVs, the ExoGlow (System Biosciences) kit has been successful. It uses a fluorescent dye that stains the lipid bilayer of sEVs and has been used extensively for EV research (87).

For investigation of sEV uptake and biodistribution on the LMS, the cellular components of the tissue should be stained with appropriate cell-specific membrane dyes. The structural effect of cardiac-derived sEVs on the LMS can be evaluated via the immunofluorescence staining of multiple cell types. This will allow measurement of several parameters that can be affected by the pathways influenced by sEV content. Recommendations include WGA for evaluation of hypertrophy, collagen and vimentin for fibrosis and stromal cell proliferation, connexin-43 for gap junction formation between cells, isolectin B-4 and CD31 for angiogenesis, TUNEL for apoptosis, a-actinin for CM characterisation and measurement of sarcomere length, and any cellular processes of interest. Quantification of these proteins can be performed by protein extraction and Western blot, following optimised protocols for LMS (10, 44, 51, 52).

Stained sEVs, with combination of cellular staining, allows the investigation of sEV biodistribution on the LMS. Biodistribution evaluation is of particular interest in sEV applications, as it can inform about the tropism of the sEVs on the LMS. Specifically, quantification of sEVs within different cell types can indicate their preferential uptake by specific cell types in a multicellular environment and can inform about their uptake at specific timepoints of disease.

7.3 Investigation of molecular effects of sEVs on LMS

7.3.1 miRNA profiling and bioinformatics analysis

The miRNA content of cardiac sEVs is of particular interest in the CVD field, as miRNAs play a major role in the regulation of gene expression during health and disease. miRNAs are post-transcriptional regulators of gene expression, and their levels change dynamically in different stages of CVD (12). The miRNA content of sEVs reflects the state of the donor cell and gives an insight into the molecular changes that happen to cells in health and disease. Individual miRNA are potent mediators of disease progression, and identifying specific sEV miRNAs can give insights into disease mechanisms (88). The LMS model plays a key role because it gives us the ability to isolate myocardium-specific sEVs of adult tissue and with the contribution of all different cardiac cell types (44). The application of sEVs derived from LMS can provide a deeper understanding of their functional and structural effects on different CVDs and of their role in cardiac intercellular communication.

To investigate the cargo of sEVs, the RNA should be extracted and applied on miRNA arrays. miRNA arrays work as standard polymerase chain reaction (PCR) panels that allow the profiling of hundreds of miRNAs from small RNA quantities and allow the quantification of their relative expression in a given sample (89). Following analysis of the miRNA array, the upregulated and downregulated miRNAs can be validated by quantitative PCR (qPCR) and inserted into bioinformatics databases to identify potential target genes for downstream analysis (90).

Several studies have investigated the miRNA content of cardiac sEVs from different sources, with the aim to uncover differential miRNA expression. A recent study investigated the miRNA profile of sEVs derived from induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) from dilated cardiomyopathy patients. The iPSCs were differentiated into CMs in culture and the secreted sEVs were isolated from the conditioned media. miRNA profiling confirmed the upregulation of miR-218-5p, which is associated with increased fibrosis through the activation of TGF-β signaling pathway (91). In another study, the RNA extraction of sEVs secreted from cultured adult mouse CMs allowed for the profiling and identification of 1,520 miRNA, of which 423 could be associated with a biological network (77). Another study performed profiling of EV-derived miRNA from the plasma of patients with aortic valve stenosis, to investigate the mechanism behind valvular cell calcification. Circulating sEV miRNAs were profiled using a miRNA array that identified the presence of 254 miRNAs, of which the 51 were differentially expressed between aortic valve stenosis and healthy patients. The upregulated miR-455-3p and downregulated miR-103a-3p were linked with pathways that can influence the calcification pathway in aortic valve stenonis patients (92). These studies show that the identification of changes in specific miRNAs can lead to their linking with specific pathways, thus explaining the progression of disease phenotypes.

A recent study compared the miRNA profile of cardiac EVs secreted from murine healthy and failing hearts, using a bioinformatic approach. Following validation with miRNA array and qPCR, 7 upregulated and 3 downregulated miRNAs were found. Bioinformatics analysis confirmed that the unregulated miRNAs were involved in cell cycle, signal transduction, and apoptosis pathways, while the downregulated were associated with cell cycle, apoptosis, and transcription and translation processes (93). This is another example of the importance of linking miRNA changes to genetic pathways and biological processes, to explain the changes observed in disease.

LMS are an unprecedented tool for the study of sEVs in cardiovascular physiology and disease, offering a wide range of potential applications. One of the potential applications with the LMS model would be the investigation of the gene expression profile of the cells within the LMS. Specifically, the isolation of different cell types of the LMS for the performance of bulk sequencing combined with bioinformatics analyses can confirm whether the miRNA changes have an effect on the gene expression levels of their target genes (93). This information can explain the structural and functional changes during disease progression. The LMS model offers unique applications for studies using sEVs. Firstly, it can be used for investigating the tropism of sEVs and potentially identifying their preferential site of uptake in the heart. Secondly, it can be used to track which specific cell type in this multicellular platform releases the most sEVs in health and disease. Future advancements can enhance delivery through engineering them with specific receptors or ligands, based on their native characteristics (94).

These studies highlight that investigating differential expression of miRNAs is a critical step for gaining a deeper understanding of the mechanism driving CVD progression. Transitioning this research from animal or iPSC-derived models to human LMS models is a significant advancement, as it enhances the relevance and translational potential of the findings to potentially improve the prognosis, prevention, or treatment of CVD.

8 Clinical and translational implications

It is important to highlight the growing significance of sEV research and its potential impact on the cardiovascular field. It is established that sEVs regulate intercellular communication on homeostasis and disease (19). This role makes them important tools for the diagnosis, treatment, and prevention of disease. Firstly, due to their altered levels in disease, circulating sEVs can act as biomarkers for diagnosis and prognosis. Their high potential as biomarkers is associated with their stability upon isolation and their presence in virtually any body fluid. Moreover, the content of sEVs portrays health state of the cells that secrete them and can enter circulation, thus enabling their detection (95). Additionally, the content of sEVs is protected by their membrane and is not degraded or dispersed (88). An example of an established CVD biomarker is cardiac troponin, which is elevated following acute MI. Similar trends have been observed with cardiac miRNAs. Plasma samples from patients that had undergone CABG surgery exhibited an increase in miR-499 levels, making it a biomarker of perioperative MI (96). A cohort analysis with data from 2,500 patients identified miR-499 and miR-208b to be displaying comparable specificity to that of cardiac troponin T as a biomarker of MI (97). The LMS model can be used to validate these findings, using adult human tissue in order to identify novel biomarkers of CVD with a higher translational potential.

Advancements in the synthetic biology and bioengineering field allow the use of sEVs as therapeutics' delivery tools (98, 99). In this context, the sEVs can be engineered for targeted drug delivery (7). Specifically, tools can be used for the loading of sEVs with bioactive molecules that can be delivered to the recipient cells, functioning as a therapeutic (100). Advances in bioengineering also allow addition of cell-specific ligands to target specific cells (94). Another advancement is the use of Y-box proteins to sort miRNAs into sEVs in cells (101). However, these techniques require refinement prior to translation, as there are risks associated with disturbing sEV structure and content, thus affecting their ability to travel through the extracellular environment and to the target cell. Transfection has been performed for the loading of sEVs secreted from cardiac stromal cells with miR-21-5p—a known downregulated miRNA in heart failure—to stimulate the regeneration of the damaged myocardium (102). Another study used electroporation to load sEVs with anti-inflammatory agents, achieving a 20% loading efficiency and observing a reduction in inflammation in a model of atherosclerosis (103).

9 Conclusion

CVD is the leading cause of death globally. The high complexity of CVD combined with the lack of appropriate experimental models has prevented the understanding of the mechanisms that contribute to its detrimental effects. This has led to the lack of efficient preventative methods and effective treatment options (1, 3).

The sEV field has seen significant advances in recent years. The increasing understanding of their participation in cellular processes of homeostasis and disease has become more evident in multiple fields (19). In the cardiovascular field, sEV miRNA content has been confirmed to influence cellular function, structure, and molecular profile, affecting the progression of CVD (91, 97, 104). The mechanism behind the concentration and content changes of sEVs in response to external stimuli—including disease—have not been fully uncovered. This is related to the lack of relevant experimental models that allow isolation of a pure population of cardiac-specific sEVs, as well as the inaccuracies of the instruments for the detection and measurement of nano-sized particles.

This review acts as a guide to introduce LMS-derived cardiac sEVs that are of pure cardiac origin and secreted by the multicellular environment that composes the adult human myocardium. This is a significant step forward compared to studies that isolate sEVs from body fluids, where the source of sEVs is difficult to determine. The multicellular LMS platform offers a more efficient way to isolate pure cardiac sEVs, without the involvement of other organs (44). Meanwhile, the electrical stimulation, oxygenation, and mechanical preload conditions, combined with the circulation of media rich in hormones and vitamins, provide a highly physiological environment for the LMS. This environment could act as a confounder on the quantity of sEVs secreted and on their content, which could potentially show great differences in comparison with previously reported minimalistic models. The procedures mentioned above can allow the isolation of sEVs that are produced by cardiac tissue during health and disease conditions, opening avenues for further investigation on their functional, structural, and molecular influence on the myocardium. In conclusion, the growing sEV field shows high potential in the development of the CVD field, offering new strategies for diagnosis and treatment of cardiac disease. The combination of cardiac sEVs with the LMS model can offer a more relevant and translational model for the understanding of the mechanistic role of sEVs in the functional, structural, and biochemical effects of different CVDs.

Author contributions

FK: Writing – review & editing, Writing – original draft. LN: Writing – review & editing. RK: Writing – review & editing. PF: Writing – review & editing. CT: Writing – review & editing.

Funding

The author(s) declare that financial support was received for the research and/or publication of this article. Financial support provided by the British Heart Foundation (FS/PhD/24/29460).

Acknowledgments

We are grateful to the British Heart Foundation for financial support (FS/PhD/24/29460).

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The author(s) declare that no Generative AI was used in the creation of this manuscript.

Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.

Publisher's note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Tajabadi M, Goran Orimi H, Ramzgouyan MR, Nemati A, Deravi N, Beheshtizadeh N, et al. Regenerative strategies for the consequences of myocardial infarction: chronological indication and upcoming visions. Biomed Pharmacother. (2022) 146:112584. doi: 10.1016/j.biopha.2021.112584

PubMed Abstract | Crossref Full Text | Google Scholar

3. Savarese G, Becher PM, Lund LH, Seferovic P, Rosano GMC, Coats AJS. Global burden of heart failure: a comprehensive and updated review of epidemiology. Cardiovasc Res. (2022) 118(17):3272–87. doi: 10.1093/cvr/cvac013

Crossref Full Text | Google Scholar

4. Pencina MJ, Navar AM, Wojdyla D, Sanchez RJ, Khan I, Elassal J, et al. Quantifying importance of major risk factors for coronary heart disease. Circulation. (2019) 139(13):1603–11. doi: 10.1161/CIRCULATIONAHA.117.031855

PubMed Abstract | Crossref Full Text | Google Scholar

5. Yuan J, Yang H, Liu C, Shao L, Zhang H, Lu K, et al. Microneedle patch loaded with exosomes containing MicroRNA-29b prevents cardiac fibrosis after myocardial infarction. Adv Healthcare Mater. (2023) 12(13):e2202959. doi: 10.1002/adhm.202202959

Crossref Full Text | Google Scholar

6. Hausenloy DJ, Yellon DM. Myocardial ischemia-reperfusion injury: a neglected therapeutic target. J Clin Invest. (2013) 123(1):92–100. doi: 10.1172/JCI62874

Crossref Full Text | Google Scholar

7. Shanmuganathan M, Vughs J, Noseda M, Emanueli C. Exosomes: basic biology and technological advancements suggesting their potential as ischemic heart disease therapeutics. Front Physiol. (2018) 9:1159. doi: 10.3389/fphys.2018.01159

Crossref Full Text | Google Scholar

8. Owen A, Stary CM, Gross ER. Exosomes as perioperative therapeutics to limit organ injury. Br J Anaesth. (2023) 130(3):248–50. doi: 10.1016/j.bja.2022.12.014

PubMed Abstract | Crossref Full Text | Google Scholar

9. D'Alessandra Y, Devanna P, Limana F, Straino S, Di Carlo A, Brambilla PG, et al. Circulating microRNAs are new and sensitive biomarkers of myocardial infarction. Eur Heart J. (2010) 31(22):2765–73. doi: 10.1093/eurheartj/ehq167

PubMed Abstract | Crossref Full Text | Google Scholar

10. Pitoulis FG, Watson SA, Perbellini F, Terracciano CM. Myocardial slices come to age: an intermediate complexity in vitro cardiac model for translational research. Cardiovasc Res. (2020) 116(7):1275–87. doi: 10.1093/CVR/CVZ341

PubMed Abstract | Crossref Full Text | Google Scholar

11. Welsh JA, Goberdhan DCI, O'Driscoll L, Buzas EI, Blenkiron C, Bussolati B, et al. Minimal information for studies of extracellular vesicles (MISEV2023): from basic to advanced approaches. J Extracell Vesicles. (2024) 13(2):e12404. doi: 10.1002/jev2.12404

PubMed Abstract | Crossref Full Text | Google Scholar

12. Du Y, Wu L, Wang L, Reiter RJ, Lip GYH, Ren J. Extracellular vesicles in cardiovascular diseases: from pathophysiology to diagnosis and therapy. Cytokine Growth Factor Rev. (2023) 74:40–55. doi: 10.1016/j.cytogfr.2023.09.006

PubMed Abstract | Crossref Full Text | Google Scholar

13. O'Brien J, Hayder H, Zayed Y, Peng C. Overview of microRNA biogenesis, mechanisms of actions, and circulation. Front Endocrinol. (2018) 9:402. doi: 10.3389/fendo.2018.00402

Crossref Full Text | Google Scholar

14. Zhang L, Liu J, Zhou C. Current aspects of small extracellular vesicles in pain process and relief. Biomater Res. (2023) 27(1):78. doi: 10.1186/s40824-023-00417-3

PubMed Abstract | Crossref Full Text | Google Scholar

15. Han QF, Li WJ, Hu KS, Gao J, Zhai WL, Yang JH, et al. Exosome biogenesis: machinery, regulation, and therapeutic implications in cancer. Mol Cancer. (2022b) 21(1):207. doi: 10.1186/s12943-022-01671-0

PubMed Abstract | Crossref Full Text | Google Scholar

16. Larios J, Mercier V, Roux A, Gruenberg J. ALIX- And ESCRT-III-dependent sorting of tetraspanins to exosomes. J Cell Biol. (2020) 219(3):e201904113. doi: 10.1083/jcb.201904113

Crossref Full Text | Google Scholar

17. Sinha S, Hoshino D, Hong NH, Kirkbride KC, Grega-Larson NE, Seiki M, et al. Cortactin promotes exosome secretion by controlling branched actin dynamics. J Cell Biol. (2016) 214(2):197–213. doi: 10.1083/jcb.201601025

PubMed Abstract | Crossref Full Text | Google Scholar

18. Yu Z, Shi M, Stewart T, Fernagut PO, Huang Y, Tian C, et al. Reduced oligodendrocyte exosome secretion in multiple system atrophy involves SNARE dysfunction. Brain. (2021) 143(6):1780–97. doi: 10.1093/BRAIN/AWAA110

PubMed Abstract | Crossref Full Text | Google Scholar

19. Han C, Yang J, Sun J, Qin G. Extracellular vesicles in cardiovascular disease: biological functions and therapeutic implications. Pharmacol Ther. (2022) 233:108025. doi: 10.1016/j.pharmthera.2021.108025

Crossref Full Text | Google Scholar

20. Jia Y, Yu L, Ma T, Xu W, Qian H, Sun Y, et al. Small extracellular vesicles isolation and separation: current techniques, pending questions and clinical applications. Theranostics. (2022) 12(15):6548–75. doi: 10.7150/thno.74305

Crossref Full Text | Google Scholar

21. Joshi BS, de Beer MA, Giepmans BNG, Zuhorn IS. Endocytosis of extracellular vesicles and release of their cargo from endosomes. ACS Nano. (2020) 14(4):4444–55. doi: 10.1021/acsnano.9b10033

PubMed Abstract | Crossref Full Text | Google Scholar

22. Mulcahy LA, Pink RC, Carter DRF. Routes and mechanisms of extracellular vesicle uptake. J Extracell Vesicles. (2014) 3(1):24641. doi: 10.3402/jev.v3.24641

Crossref Full Text | Google Scholar

23. Das D, Jothimani G, Banerjee A, Dey A, Duttaroy AK, Pathak S. A brief review on recent advances in diagnostic and therapeutic applications of extracellular vesicles in cardiovascular disease. Int J Biochem Cell Biol. (2024) 173:106616. doi: 10.1016/j.biocel.2024.106616

PubMed Abstract | Crossref Full Text | Google Scholar

24. Litviňuková M, Talavera-López C, Maatz H, Reichart D, Worth CL, Lindberg EL, et al. Cells of the adult human heart. Nature. (2020) 588(7838):466–72. doi: 10.1038/s41586-020-2797-4

Crossref Full Text | Google Scholar

25. Zhang Z, Zou Y, Song C, Cao K, Cai K, Chen S, et al. Advances in the study of exosomes in cardiovascular diseases. J Adv Res. (2024) 66:133–53. doi: 10.1016/j.jare.2023.12.014

PubMed Abstract | Crossref Full Text | Google Scholar

26. Wang C, Li Z, Liu Y, Yuan L. Exosomes in atherosclerosis: performers, bystanders, biomarkers, and therapeutic targets. Theranostics. (2021) 11(8):3996–4010. doi: 10.7150/THNO.56035

Crossref Full Text | Google Scholar

27. Emanueli C, Shearn AIU, Laftah A, Fiorentino F, Reeves BC, Beltrami C, et al. Coronary artery-bypass-graft surgery increases the plasma concentration of exosomes carrying a cargo of cardiac microRNAs: an example of exosome trafficking out of the human heart with potential for cardiac biomarker discovery. PLoS One. (2016) 11(4):e0154274. doi: 10.1371/journal.pone.0154274

PubMed Abstract | Crossref Full Text | Google Scholar

28. Zarà M, Baggiano A, Amadio P, Campodonico J, Gili S, Annoni A, et al. Circulating small extracellular vesicles reflect the severity of myocardial damage in STEMI patients. Biomolecules. (2023) 13(10):1470. doi: 10.3390/biom13101470

Crossref Full Text | Google Scholar

29. 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: 10.1161/CIRCULATIONAHA.119.042640

Crossref Full Text | Google Scholar

30. Giricz Z, Varga ZV, Baranyai T, Sipos P, Pálóczi K, Kittel Á, et al. Cardioprotection by remote ischemic preconditioning of the rat heart is mediated by extracellular vesicles. J Mol Cell Cardiol. (2014) 68:75–8. doi: 10.1016/j.yjmcc.2014.01.004

PubMed Abstract | Crossref Full Text | Google Scholar

31. Loyer X, Zlatanova I, Devue C, Yin M, Howangyin KY, Klaihmon P, et al. Intra-cardiac release of extracellular vesicles shapes inflammation following myocardial infarction short communication. Circ Res. (2018) 123(1):100–6. doi: 10.1161/CIRCRESAHA.117.311326

Crossref Full Text | Google Scholar

32. Gupta S, Knowlton AA. HSP60 Trafficking in adult cardiac myocytes: role of the exosomal pathway. Am J Physiol Heart Circ Physiol. (2007) 292(6):H3052–6. doi: 10.1152/ajpheart.01355.2006

PubMed Abstract | Crossref Full Text | Google Scholar

33. Chen G, Xu C, Gillette TG, Huang T, Huang P, Li Q, et al. Cardiomyocyte-derived small extracellular vesicles can signal eNOS activation in cardiac microvascular endothelial cells to protect against ischemia/reperfusion injury. Theranostics. (2020) 10(25):11754–74. doi: 10.7150/thno.43163

PubMed Abstract | Crossref Full Text | Google Scholar

34. Anselmo A, Frank D, Papa L, Viviani Anselmi C, Di Pasquale E, Mazzola M, et al. Myocardial hypoxic stress mediates functional cardiac extracellular vesicle release. Eur Heart J. (2021) 42(28):2780–92. doi: 10.1093/eurheartj/ehab247

PubMed Abstract | Crossref Full Text | Google Scholar

35. Yang J, Yu X, Xue F, Li Y, Liu W, Zhang S. Exosomes derived from cardiomyocytes promote cardiac fibrosis via myocyte-fibroblast cross-talk. Am J Transl Res. (2018) 10(12):4350–66. PMID: 30662677.

Google Scholar

36. Garcia NA, Ontoria-Oviedo I, González-King H, Diez-Juan A, Sepúlveda P. Glucose starvation in cardiomyocytes enhances exosome secretion and promotes angiogenesis in endothelial cells. PLoS One. (2015) 10(9):e0138849. doi: 10.1371/journal.pone.0138849

Crossref Full Text | Google Scholar

37. Cosme J, Guo H, Hadipour-Lakmehsari S, Emili A, Gramolini AO. Hypoxia-induced changes in the fibroblast secretome, exosome, and whole-cell proteome using cultured, cardiac-derived cells isolated from neonatal mice. J Proteome Res. (2017) 16(8):2836–47. doi: 10.1021/acs.jproteome.7b00144

Crossref Full Text | Google Scholar

38. Liu N, Xie L, Xiao P, Chen X, Kong W, Lou Q, et al. Cardiac fibroblasts secrete exosome microRNA to suppress cardiomyocyte pyroptosis in myocardial ischemia/reperfusion injury. Mol Cell Biochem. (2022) 477(4):1249–60. doi: 10.1007/s11010-021-04343-7

Crossref Full Text | Google Scholar

39. Yadid M, Lind JU, Ardoña HAM, Sheehy SP, Dickinson LE, Eweje F, et al. Endothelial extracellular vesicles contain protective proteins and rescue ischemia-reperfusion injury in a human heart-on-chip. Sci Transl Med. (2020) 12(565):eaax8005. doi: 10.1126/scitranslmed.aax8005

Crossref Full Text | Google Scholar

40. de Jong OG, Verhaar MC, Chen Y, Vader P, Gremmels H, Posthuma G, et al. Cellular stress conditions are reflected in the protein and RNA content of endothelial cell-derived exosomes. J Extracell Vesicles. (2012) 1(1):18396. doi: 10.3402/jev.v1i0.18396

Crossref Full Text | Google Scholar

41. Davidson SM, Riquelme JA, Zheng Y, Vicencio JM, Lavandero S, Yellon DM. Endothelial cells release cardioprotective exosomes that may contribute to ischaemic preconditioning. Sci Rep. (2018) 8(1):15885. doi: 10.1038/s41598-018-34357-z

PubMed Abstract | Crossref Full Text | Google Scholar

42. Mathieu M, Névo N, Jouve M, Valenzuela JI, Maurin M, Verweij FJ, et al. Specificities of exosome versus small ectosome secretion revealed by live intracellular tracking of CD63 and CD9. Nat Commun. (2021) 12(1):4389. doi: 10.1038/s41467-021-24384-2

Crossref Full Text | Google Scholar

43. Théry C, Witwer KW, Aikawa E, Alcaraz MJ, Anderson JD, Andriantsitohaina R., et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the international society for extracellular vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles. (2018) 7(1):1535750. doi: 10.1080/20013078.2018.1535750

Crossref Full Text | Google Scholar

44. Watson SA, Scigliano M, Bardi I, Ascione R, Terracciano CM, Perbellini F. Preparation of viable adult ventricular myocardial slices from large and small mammals. Nat Protoc. (2017) 12(12):2623–39. doi: 10.1038/nprot.2017.139

Crossref Full Text | Google Scholar

45. Mitcheson JS, Hancox JC, Levi AJ. Action potentials, ion channel currents and transverse tubule density in adult rabbit ventricular myocytes maintained for 6 days in cell culture. Pflugers Arch Eur J Physiol. (1996) 431(6):814–27. doi: 10.1007/s004240050073

Crossref Full Text | Google Scholar

46. Richards DJ, Li Y, Kerr CM, Yao J, Beeson GC, Coyle RC, et al. Human cardiac organoids for the modelling of myocardial infarction and drug cardiotoxicity. Nat Biomed Eng. (2020) 4(4):446–62. doi: 10.1038/s41551-020-0539-4

Crossref Full Text | Google Scholar

47. Mohr E, Thum T, Bär C. Accelerating cardiovascular research: recent advances in translational 2D and 3D heart models. Eur J Heart Fail. (2022) 24(10):1778–91. doi: 10.1002/ejhf.2631

Crossref Full Text | Google Scholar

48. van der Velden J, Asselbergs FW, Bakkers J, Batkai S, Bertrand L, Bezzina CR, et al. Animal models and animal-free innovations for cardiovascular research: current status and routes to be explored. Consensus document of the ESC working group on myocardial function and the ESC working group on cellular biology of the heart. Cardiovasc Res. (2022) 118(15):3016–51. doi: 10.1093/cvr/cvab370

Crossref Full Text | Google Scholar

49. Fischer C, Milting H, Fein E, Reiser E, Lu K, Seidel T, et al. Long-term functional and structural preservation of precision-cut human myocardium under continuous electromechanical stimulation in vitro. Nat Commun. (2019) 10(1):117. doi: 10.1038/s41467-018-08003-1

Crossref Full Text | Google Scholar

50. van der Geest JSA, De Boer TP, Terracciano CM, Thum T, Dendorfer A, Doevendans PA, et al. Living myocardial slices: walking the path towards standardization. Cardiovasc Res. (2025) 121(7):1011–23. doi: 10.1093/cvr/cvaf079

PubMed Abstract | Crossref Full Text | Google Scholar

51. Pitoulis FG, Nunez-Toldra R, Xiao K, Kit-Anan W, Mitzka S, Jabbour RJ, et al. Remodelling of adult cardiac tissue subjected to physiological and pathological mechanical load in vitro. Cardiovasc Res. (2022) 118(3):814–27. doi: 10.1093/cvr/cvab084

Crossref Full Text | Google Scholar

52. Dries E, Bardi I, Nunez-Toldra R, Meijlink B, Terracciano CM. Camkii inhibition reduces arrhythmogenic ca2+ events in subendocardial cryoinjured rat living myocardial slices. J Gen Physiol. (2021) 153(6):e202012737. doi: 10.1085/jgp.202012737

PubMed Abstract | Crossref Full Text | Google Scholar

53. Waleczek FJG, Cipriano G, Haas JA, Garg A, Pfanne A, Just A, et al. Prolonged hypoxia in rat living myocardial slices affects function, expression, and structure. Int J Mol Sci (2025) 26(1):218. doi: 10.3390/ijms26010218

Crossref Full Text | Google Scholar

54. Pilcher LC, Hancock E, Neeli A, Sckolnick M, Caporizzo MA, Palmer BM, et al. Loss of Snord116 protects cardiomyocyte kinetics during ischemic stress. J Mol Cell Cardiol Plus. (2025) 11:100291. doi: 10.1016/j.jmccpl.2025.100291

PubMed Abstract | Crossref Full Text | Google Scholar

55. Wang BX, Nicastro L, Couch L, Kit-Anan W, Downing B, Macleod KT, et al. Extracellular vesicles from human cardiac fibroblasts modulate calcium cycling in human stem cell-derived cardiomyocytes. Cells. (2022) 11(7):1171. doi: 10.3390/cells11071171

Crossref Full Text | Google Scholar

56. Benedikter BJ, Bouwman FG, Vajen T, Heinzmann ACA, Grauls G, Mariman EC, et al. Ultrafiltration combined with size exclusion chromatography efficiently isolates extracellular vesicles from cell culture media for compositional and functional studies. Sci Rep. (2017) 7(1):15297. doi: 10.1038/s41598-017-15717-7

PubMed Abstract | Crossref Full Text | Google Scholar

57. Hendrix A, Lippens L, Pinheiro C, Théry C, Martin-Jaular L, Lötvall J, et al. Extracellular vesicle analysis. Nat Rev Methods Primers. (2023) 3(1):56. doi: 10.1038/s43586-023-00240-z

Crossref Full Text | Google Scholar

58. Liangsupree T, Multia E, Riekkola ML. Modern isolation and separation techniques for extracellular vesicles. J Chromatogr, A. (2021) 1636:461773. doi: 10.1016/j.chroma.2020.461773

PubMed Abstract | Crossref Full Text | Google Scholar

59. Dong L, Zieren RC, Horie K, Kim CJ, Mallick E, Jing Y, et al. Comprehensive evaluation of methods for small extracellular vesicles separation from human plasma, urine and cell culture medium. J Extracell Vesicles. (2020) 10(2):e12044. doi: 10.1002/jev2.12044

Crossref Full Text | Google Scholar

60. Baranyai T, Herczeg K, Onódi Z, Voszka I, Módos K, Marton N, et al. Isolation of exosomes from blood plasma: qualitative and quantitative comparison of ultracentrifugation and size exclusion chromatography methods. PLoS One. (2015) 10(12):e0145686. doi: 10.1371/journal.pone.0145686

PubMed Abstract | Crossref Full Text | Google Scholar

61. Kowkabany G, Bao Y. Nanoparticle tracking analysis: an effective tool to characterize extracellular vesicles. Molecules. (2024) 29(19):4672. doi: 10.3390/molecules29194672

Crossref Full Text | Google Scholar

62. Comfort N, Cai K, Bloomquist TR, Strait MD, Ferrante AW, Baccarelli AA. Nanoparticle tracking analysis for the quantification and size determination of extracellular vesicles. J Visualized Exp. (2021) (169):10.3791/62447. doi: 10.3791/62447

Crossref Full Text | Google Scholar

63. Gardiner C, Ferreira YJ, Dragovic RA, Redman CWG, Sargent IL. Extracellular vesicle sizing and enumeration by nanoparticle tracking analysis. J Extracell Vesicles. (2013) 2(1):19671. doi: 10.3402/jev.v2i0.19671

Crossref Full Text | Google Scholar

64. Hinzman CP, Jayatilake M, Bansal S, Fish BL, Li Y, Zhang Y, et al. An optimized method for the isolation of urinary extracellular vesicles for molecular phenotyping: detection of biomarkers for radiation exposure. J Transl Med. (2022) 20(1):199. doi: 10.1186/s12967-022-03414-7

Crossref Full Text | Google Scholar

65. Caller T, Rotem I, Shaihov-Teper O, Lendengolts D, Schary Y, Shai R, et al. Small extracellular vesicles from infarcted and failing heart accelerate tumor growth. Circulation. (2024) 149(22):1729–48. doi: 10.1161/CIRCULATIONAHA.123.066911

Crossref Full Text | Google Scholar

66. Preußer C, Stelter K, Tertel T, Linder M, Helmprobst F, Szymanski W, et al. Isolation of native EVs from primary biofluids—free-flow electrophoresis as a novel approach to purify ascites-derived EVs. J. Extracell. Biol. (2022) 1(12):e71. doi: 10.1002/jex2.71

Crossref Full Text | Google Scholar

67. Guo J, Wu C, Lin X, Zhou J, Zhang J, Zheng W, et al. Establishment of a simplified dichotomic size-exclusion chromatography for isolating extracellular vesicles toward clinical applications. J Extracell Vesicles. (2021) 10(11):e12145. doi: 10.1002/jev2.12145

PubMed Abstract | Crossref Full Text | Google Scholar

68. Chen P, Tang S, Gao H, Zhang H, Chen C, Fang Z, et al. Wharton’s jelly mesenchymal stem cell-derived small extracellular vesicles as natural nanoparticles to attenuate cartilage injury via microRNA regulation. Int J Pharm. (2022) 623:121952. doi: 10.1016/j.ijpharm.2022.121952

Crossref Full Text | Google Scholar

69. Shearn AIU, Aday S, Ben-Aicha S, Carnell-Morris P, Siupa A, Angelini GD, et al. Analysis of neat biofluids obtained during cardiac surgery using nanoparticle tracking analysis: methodological considerations. Front Cell Dev Biol. (2020) 8:367. doi: 10.3389/fcell.2020.00367

Crossref Full Text | Google Scholar

70. Mladenović D, Brealey J, Peacock B, Koort K, Zarovni N. Quantitative fluorescent nanoparticle tracking analysis and nano-flow cytometry enable advanced characterization of single extracellular vesicles. J Extracell Biol. (2025) 4(1):e70031. doi: 10.1002/jex2.70031

Crossref Full Text | Google Scholar

71. Daaboul GG, Gagni P, Benussi L, Bettotti P, Ciani M, Cretich M, et al. Digital detection of exosomes by interferometric imaging. Sci Rep. (2016) 6:37246. doi: 10.1038/srep37246

PubMed Abstract | Crossref Full Text | Google Scholar

72. Breitwieser K, Koch LF, Tertel T, Proestler E, Burgers LD, Lipps C, et al. Detailed characterization of small extracellular vesicles from different cell types based on tetraspanin composition by ExoView R100 platform. Int J Mol Sci. (2022) 23(15):8544. doi: 10.3390/ijms23158544

Crossref Full Text | Google Scholar

73. Katsur M, He Z, Vinokur V, Corteling R, Yellon DM, Davidson SM. Exosomes from neuronal stem cells may protect the heart from ischaemia/reperfusion injury via JAK1/2 and gp130. J Cell Mol Med. (2021) 25(9):4455–65. doi: 10.1111/jcmm.16515

PubMed Abstract | Crossref Full Text | Google Scholar

74. Price J, Gardiner C, Harrison P. Platelet-enhanced plasma: characterization of a novel candidate resuscitation fluid’s extracellular vesicle content, clotting parameters, and thrombin generation capacity. Transfusion. (2021) 61(7):2179–94. doi: 10.1111/trf.16423

Crossref Full Text | Google Scholar

75. Mahida RY, Price J, Lugg ST, Li H, Parekh D, Scott A, et al. CD14-positive Extracellular vesicles in bronchoalveolar lavage fluid as a new biomarker of acute respiratory distress syndrome. Am J Physiol Lung Cell Mol Physiol. (2022) 322(4):L617–24. doi: 10.1152/ajplung.00052.2022

Crossref Full Text | Google Scholar

76. Corona ML, Hurbain I, Raposo G, van Niel G. Characterization of extracellular vesicles by transmission electron microscopy and immunolabeling electron microscopy. Methods Mol Biol. (2023) 2668:33–43. doi: 10.1007/978-1-0716-3203-1_4

Crossref Full Text | Google Scholar

77. Waldenström A, Gennebäck N, Hellman U, Ronquist G. Cardiomyocyte microvesicles contain DNA/RNA and convey biological messages to target cells. PLoS One. (2012) 7(4):e34653. doi: 10.1371/journal.pone.0034653

PubMed Abstract | Crossref Full Text | Google Scholar

78. Watson SA, Duff J, Bardi I, Zabielska M, Atanur SS, Jabbour RJ, et al. Biomimetic electromechanical stimulation to maintain adult myocardial slices in vitro. Nat Commun. (2019) 10(1):2168. doi: 10.1038/s41467-019-10175-3

Crossref Full Text | Google Scholar

79. Watson SA, Dendorfer A, Thum T, Perbellini F. A practical guide for investigating cardiac physiology using living myocardial slices. Basic Res Cardiol. (2020) 115(6):61. doi: 10.1007/s00395-020-00822-y

Crossref Full Text | Google Scholar

80. Huxley AF, Niedergerke R. Structural changes in muscle during contraction: interference microscopy of living muscle fibres. Nature. (1954) 173(4412):971–3. doi: 10.1038/173971a0

Crossref Full Text | Google Scholar

81. Omichi C, Lamp ST, Lin SF, Yang J, Baher A, Zhou S, et al. Intracellular ca dynamics in ventricular fibrillation. Am J Physiol Heart Circ Physiol. (2004) 286(5):H1836–44. doi: 10.1152/ajpheart.00123.2003

PubMed Abstract | Crossref Full Text | Google Scholar

82. Myles RC, Wang L, Kang C, Bers DM, Ripplinger CM. Local β-adrenergic stimulation overcomes source-sink mismatch to generate focal arrhythmia. Circ Res. (2012) 110(11):1454–64. doi: 10.1161/CIRCRESAHA.111.262345

PubMed Abstract | Crossref Full Text | Google Scholar

83. van der Geest JSA, Benavente ED, van Ham WB, Doevendans PA, van Laake LW, de Boer TP, et al. Characterization of human living myocardial slices culture-induced adaptations: a translational perspective. J Mol Cell Cardiol Plus. (2025a) 13:100465. doi: 10.1016/j.jmccpl.2025.100465

PubMed Abstract | Crossref Full Text | Google Scholar

84. Kosmach A, Roman B, Sun J, Femnou A, Zhang F, Liu C, et al. Monitoring mitochondrial calcium and metabolism in the beating MCU-KO heart. Cell Rep. (2021) 37(3):109846. doi: 10.1016/j.celrep.2021.109846

PubMed Abstract | Crossref Full Text | Google Scholar

85. Honarbakhsh S, Schilling RJ, Orini M, Providencia R, Keating E, Finlay M, et al. Structural remodeling and conduction velocity dynamics in the human left atrium: relationship with reentrant mechanisms sustaining atrial fibrillation. Heart Rhythm. (2019) 16(1):18–25. doi: 10.1016/j.hrthm.2018.07.019

PubMed Abstract | Crossref Full Text | Google Scholar

86. Camelliti P, Al-Saud SA, Smolenski RT, Al-Ayoubi S, Bussek A, Wettwer E, et al. Adult human heart slices are a multicellular system suitable for electrophysiological and pharmacological studies. J Mol Cell Cardiol. (2011) 51(3):390–8. doi: 10.1016/j.yjmcc.2011.06.018

PubMed Abstract | Crossref Full Text | Google Scholar

87. Ben-Aicha S, Anwar M, Vilahur G, Martino F, Kyriazis PG, de Winter N, et al. Small extracellular vesicles in the pericardium modulate macrophage immunophenotype in coronary artery disease. JACC Basic Transl Sci. (2024) 9(9):1057–72. doi: 10.1016/j.jacbts.2024.05.003

Crossref Full Text | Google Scholar

88. Reiss AB, Ahmed S, Johnson M, Saeedullah U, De Leon J. Exosomes in cardiovascular disease: from mechanism to therapeutic target. Metabolites. (2023) 13(4):479. doi: 10.3390/metabo13040479

PubMed Abstract | Crossref Full Text | Google Scholar

89. Liu CG, Calin GA, Volinia S, Croce CM. MicroRNA expression profiling using microarrays. Nat Protoc. (2008) 3(4):563–78. doi: 10.1038/nprot.2008.14

PubMed Abstract | Crossref Full Text | Google Scholar

90. Sticht C, De La Torre C, Parveen A, Gretz N. Mirwalk: an online resource for prediction of microrna binding sites. PLoS One. (2018) 13(10):e0206239. doi: 10.1371/journal.pone.0206239

PubMed Abstract | Crossref Full Text | Google Scholar

91. Fu X, Mishra R, Chen L, Arfat MY, Sharma S, Kingsbury T, et al. Exosomes mediated fibrogenesis in dilated cardiomyopathy through a MicroRNA pathway. iScience. (2023) 26(2):105963. doi: 10.1016/j.isci.2023.105963

Crossref Full Text | Google Scholar

92. Khandagale A, Corcoran P, Dan Y, Isaksson A, James S, Siegbahn A, et al. Plasma-derived extracellular vesicle-propagated microRNA from aortic stenosis patients render pro-calcifying effects on valve interstitial cells. J Am Coll Cardiol Basic Trans Sci. (2025) 10(9):101327. doi: 10.1016/j.jacbts.2025.101327

Crossref Full Text | Google Scholar

93. Oh JG, Lee P, Gordon RE, Sahoo S, Kho C, Jeong D. Analysis of extracellular vesicle miRNA profiles in heart failure. J Cell Mol Med. (2020) 24(13):7214–27. doi: 10.1111/jcmm.15251

PubMed Abstract | Crossref Full Text | Google Scholar

94. de Abreu RC, Fernandes H, da Costa Martins PA, Sahoo S, Emanueli C, Ferreira L. Native and bioengineered extracellular vesicles for cardiovascular therapeutics. Nat Rev Cardiol. (2020) 17(11):685–97. doi: 10.1038/s41569-020-0389-5

Crossref Full Text | Google Scholar

95. Weber B, Franz N, Marzi I, Henrich D, Leppik L. Extracellular vesicles as mediators and markers of acute organ injury: current concepts. Eur J Trauma Emerg Surg. (2022) 48(3):1525–44. doi: 10.1007/s00068-021-01607-1

PubMed Abstract | Crossref Full Text | Google Scholar

96. Yao Y, Du J, Cao X, Wang Y, Huang Y, Hu S, et al. Plasma levels of microRNA-499 provide an early indication of perioperative myocardial infarction in coronary artery bypass graft patients. PLoS One. (2014) 9(8):e104618. doi: 10.1371/journal.pone.0104618

Crossref Full Text | Google Scholar

97. Xue S, Zhu W, Liu D, Su Z, Zhang L, Chang Q, et al. Circulating miR-26a-1, miR-146a and miR-199a-1 are potential candidate biomarkers for acute myocardial infarction. Mol Med. (2019) 25(1):18. doi: 10.1186/s10020-019-0086-1

PubMed Abstract | Crossref Full Text | Google Scholar

98. Kelwick RJR, Webb AJ, Heliot A, Segura CT, Freemont PS. Opportunities to accelerate extracellular vesicle research with cell-free synthetic biology. J Extracell Biol. (2023) 2(5):e90. doi: 10.1002/jex2.90

Crossref Full Text | Google Scholar

99. Yang C, Xue Y, Duan Y, Mao C, Wan M. Extracellular vesicles and their engineering strategies, delivery systems, and biomedical applications. J Controlled Release. (2024) 365:1089–123. doi: 10.1016/j.jconrel.2023.11.057

Crossref Full Text | Google Scholar

100. O'Brien K, Breyne K, Ughetto S, Laurent LC, Breakefield XO. RNA Delivery by extracellular vesicles in mammalian cells and its applications. Nat Rev Mol Cell Biol. (2020) 21(10):585–606. doi: 10.1038/s41580-020-0251-y

Crossref Full Text | Google Scholar

101. Shurtleff MJ, Temoche-Diaz MM, Karfilis KV, Ri S, Schekman R. Y-box protein 1 is required to sort microRNAs into exosomes in cells and in a cell-free reaction. eLife. (2016) 5:e19276. doi: 10.7554/eLife.19276

Crossref Full Text | Google Scholar

102. Qiao L, Hu S, Liu S, Zhang H, Ma H, Huang K, et al. MicroRNA-21-5p dysregulation in exosomes derived from heart failure patients impairs regenerative potential. J Clin Invest. (2019) 129(6):2237–50. doi: 10.1172/JCI123135

PubMed Abstract | Crossref Full Text | Google Scholar

103. Wu G, Zhang J, Zhao Q, Zhuang W, Ding J, Zhang C, et al. Molecularly engineered macrophage-derived exosomes with inflammation tropism and intrinsic heme biosynthesis for atherosclerosis treatment. Angew Chemie Int Ed. (2020) 59(10):4068–74. doi: 10.1002/anie.201913700

Crossref Full Text | Google Scholar

104. Nail HM, Chiu CC, Leung CH, Ahmed MMM, Wang HMD. Exosomal miRNA-mediated intercellular communications and immunomodulatory effects in tumor microenvironments. J Biomed Sci. (2023) 30(1): 69. doi: 10.1186/s12929-023-00964-w

PubMed Abstract | Crossref Full Text | Google Scholar

105. Caliandro R, Husetić A, Ligtermoet ML, Boender AR, Zentilin L, Boink GJJ, et al. Living myocardial slices as a model for testing cardiac pro-reparative gene therapies. Mol Ther. (2025) 33(7):2990–6. doi: 10.1016/j.ymthe.2025.03.033

PubMed Abstract | Crossref Full Text | Google Scholar

106. Bang C, Batkai S, Dangwal S, Gupta SK, Foinquinos A, Holzmann A, et al. Cardiac fibroblast-derived microRNA passenger strand-enriched exosomes mediate cardiomyocyte hypertrophy. J Clin Investig. (2014) 124(5):2136–46. doi: 10.1172/JCI70577

Crossref Full Text | Google Scholar

Keywords: cardiovascular disease, extracellular vesicles, living myocardial slices, small extracellular vesicles, mechanisms, cell-cell crosstalk

Citation: Koutentaki F, Nicastro L, Kelwick RJR, Freemont PS and Terracciano CM (2025) Investigating the mechanisms of small extracellular vesicles in cardiovascular disease using the living myocardial slice platform. Front. Cardiovasc. Med. 12:1697099. doi: 10.3389/fcvm.2025.1697099

Received: 1 September 2025; Accepted: 16 October 2025;
Published: 12 November 2025.

Edited by:

Jona Benjamin Krohn, Heidelberg University Hospital, Germany

Reviewed by:

Sveva Bollini, University of Genoa, Italy
Dominika Bernáth-Nagy, Semmelweis University, Hungary

Copyright: © 2025 Koutentaki, Nicastro, Kelwick, Freemont and Terracciano. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Cesare M. Terracciano, Yy50ZXJyYWNjaWFub0BpbXBlcmlhbC5hYy51aw==

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.