Skip to main content

Apoptotic mesenchymal stem cells and their secreted apoptotic extracellular vesicles: therapeutic applications and mechanisms

Abstract

Mesenchymal stem cells (MSCs), an accessible and less ethically controversial class of adult stem cells, have demonstrated significant efficacy in treating a wide range of diseases in both the preclinical and clinical phases. However, we do not yet have a clear understanding of the mechanisms by which MSCs exert their therapeutic effects in vivo. We found that the transplanted MSCs go an apoptotic fate within 24 h in vivo irrespective of the route of administration. Still, the short-term survival of MSCs do not affect their long-term therapeutic efficacy. An increasing number of studies have demonstrated that transplantation of apoptotic MSCs (ApoMSCs) show similar or even better efficacy than viable MSCs, including a variety of preclinical disease models such as inflammatory diseases, skin damage, bone damage, organ damage, etc. Although the exact mechanism has yet to be explored, recent studies have shown that transplanted MSCs undergo apoptosis in vivo and are phagocytosed by phagocytes, thereby exerting immunomodulatory effects. The apoptotic extracellular vesicles secreted by ApoMSCs (MSC-ApoEVs) play a significant role in promoting immunomodulation, endogenous stem cell regeneration, and angiogenesis due to their apoptotic properties and inheritance of molecular characteristics from their parental MSCs. On this basis, this review aims to deeply explore the therapeutic applications and mechanisms of ApoMSCs and their secretion of MSC-ApoEVs, as well as the challenges they face.

Background

The efficacy of mesenchymal stem cells (MSCs) in a wide range of preclinical disease models has been well documented over the past decades [1,2,3,4,5]. There is even an increasing number of clinical trial cases using MSCs as a therapeutic tool [6,7,8,9]. For a long time, it has been believed that surviving MSCs were essential for therapeutic efficacy and their therapeutic effects depended mainly on their differentiation properties, paracrine mechanisms, and intercellular surface molecular interactions [10,11,12,13,14,15]. However, recent studies have shown that transplanted MSCs may go into apoptosis within a short period of time because of the disease microenvironment such as hypoxia, ischemia, and inflammation, but apoptosis does not affect the efficacy of MSCs [16]. Thus, recent studies have proposed a new concept that apoptosis of MSCs in vivo plays a key role in the treatment of disease.

As early as 2005, thum et al. proposed a new immunomodulatory mechanism: the ‘dying stem cell’ hypothesis [17]. Specifically, the authors found that the viability of peripheral blood or whole bone marrow MSCs (BMMSCs) isolated from patients with acute myocardial infarction for transplantation ranged from 75 to 95%, implying that some of the cells had undergone apoptosis and necrosis prior to transplantation [17]. The influence of the myocardial infarction microenvironment on the viability of transplanted MSCs was implicated [17]. They proposed a new insight into the effect of transplanted MSCs on improving cardiac function: transplanted MSCs that underwent apoptosis in vivo would recruit immune cells and suppress inflammatory macrophages and dendritic cells [17]. Although this was hypothesized, a growing number of recent studies have confirmed this hypothesis. They found that MSCs underwent extensive apoptosis within hours of infusion and were subsequently engulfed by local myeloid phagocytes, which were the ultimate players in the anti-inflammatory response [18].

With more and more research, direct transplantation of apoptotic MSCs (ApoMSCs) induced in vitro were preferred by more and more researchers for more efficient treatment. It has been found that transplantation of ApoMSCs showed similar or even better efficacy than viable MSCs, which has been demonstrated in various preclinical diseases such as autoimmune diseases and inflammatory diseases [16, 19]. Studies applying anti-ApoMSCs on animal disease models revealed limited immunomodulatory effects and diminished efficacy.

MSCs may be exposed to the risk of pulmonary embolism due to their excessive size [20]. In response to the popularity of cell-free therapies, the apoptotic extracellular vesicles secreted by ApoMSCs (MSC-ApoEVs) have been used by an increasing number of researchers as a result of ApoMSCs [21]. Due to their phagocytic targeting like ApoMSCs and carrying abundant functional proteins and RNAs from their parental MSCs, they have the potential to be more direct therapeutic agents than ApoMSCs themselves. It has been demonstrated in preclinical disease models such as bone damage, skin damage, dental damage, diabetes, cancer, and inflammatory diseases [22]. MSC-ApoEVs have been explored as drug carriers due to their easy availability, high yield, and high drug loading rate [10, 23].

In this review, we first summarized the spontaneous apoptotic fate of transplanted MSCs in vivo provided evidence for the therapeutic efficacy of MSCs after apoptosis, and explored the causes of apoptosis occurs in MSCs. In addition, we summarized in detail the specific applications and mechanisms of action of ApoMSCs and their MSC-ApoEVs for therapy. Finally, we analyzed and concluded the challenges and prospects arising from the therapeutic application of ApoMSCs and MSC-ApoEVs.

Spontaneous apoptotic fate of transplanted MSCs

The fate of MSCs with different routes of administration for transplantation to undergo apoptosis in vivo has been confirmed by an increasing number of studies [24]. Intravenous injection was the most common, minimally invasive, and highly reproducible route of administration, with data suggesting that it accounted for 43% of clinical trials [25]. Various MSCs detection techniques such as MSCs constitutively expressing fluorescent proteins or luciferase, or MSCs labeled with fluorescent dyes or radiotracers were used to study the biodistribution of MSCs, which showed that intravenously infused MSCs rarely reached the transplantation damage site, and most MSCs were trapped in the lungs due to their inability to pass through the capillary system of the lungs and were later redistributed to liver and spleen, and almost none at other sites(< 0.1%) [20, 26, 27]. Different tissue culture or polymerase chain reaction (PCR) of MSCs-specific marker genes results showed that surviving MSCs rapidly decreased and disappeared within only 24 h [26, 28]. Interestingly, more than half of the fluorescence-positive MSCs in the lungs showed co-expression of calreticulin within 30 min after MSCs injection and exhibited a high level of expression of the apoptotic marker caspase 3 within 1 h [26, 29]. These suggested that intravenously injected MSCs underwent rapid apoptosis during their stay in the lungs [26, 29]. Among them, calreticulin is a marker that serves as a signal for the host immune system to consecutively induce phagocytosis [26]. After 24 h of intravenous infusion of MSCs, it was found that MSCs were contained in monocytes of predominantly non-classical Ly6Clow phenotype, and MSCs-containing monocytes were also detected systemically, further demonstrating that intravenously transplanted MSCs interacted with phagocytosis after apoptosis occurred [30].

In addition to the survival status of MSCs, it was found that most MSCs were no longer complete soon after transplantation [26]. Using microscopy to analyze the size changes of green fluorescent MSCs, it was found that fluorescent MSCs in transplanted mouse lungs appeared to be smaller, and Hoechst 33,342 nuclear staining showed that most of the nuclear signals were absent in fluorescence-positive MSCs after transplantation, indicating that MSCs continued to decrease in size and undergo fragmentation after transplantation [26]. Taken together, these suggested that MSCs underwent apoptosis, fragmentation, and phagocytosis by immune cells after intravenous transplantation.

Direct injection of MSCs into damaged tissues showed similar results to intravenous injection, which reported apoptosis of locally transplanted MSCs after 3–5 days and the appearance of almost all locally transplanted MSCs in tissue-specific phagocytes within a week [27, 31]. Preda et al. explored the fate of MSCs under different routes of administration by using a dual tracking system for MSCs, i.e., luciferase expression and VivoTrack680 labeling and in vivo optical imaging. It was found that MSCs infused into intraventricular, intrapancreatic, intrasplenic, and subcutaneous areas of diabetic mice activate hypoxic signaling pathways within one day after transplantation, followed by caspase 3-mediated apoptosis [16]. Immune cells were locally recruited at the transplantation site, resulting in macrophage phagocytosis of apoptotic MSCs [16].

Impact of spontaneous apoptotic MSCs on diseased animals

The short-term survival of MSCs did not seem to interfere with their effectiveness, as the beneficial effects of MSCs remained potent after apoptosis [30]. In graft-versus-host disease (GVHD) mice, 1 h after MSCs infusion, extensive apoptosis and perforin-dependent caspase activation were observed due to the presence of cytotoxic immune cells [32]. This was necessary for the MSCs to function as an immunosuppressor [32]. MSCs infusion effectively reduced GVHD effector cells infiltrating the spleen and lungs by up to 10% compared to untreated GVHD mice [32]. When MSCs were intravenously infused in acute liver-injured mice, MSCs underwent apoptosis within 2–4 h after [33]. With the elevation of phosphatidylserine (PS) levels on apoptotic MSCs, monocyte-derived macrophages were recruited for phagocytosis and shifted to the M2 phenotype [33]. The transplantation of spontaneously apoptotic MSCs was effective in ameliorating acute liver injury in mice after 12 h of treatment [33]. The area of hepatic necrosis was reduced, the serum levels of alanine aminotransferase and aspartate aminotransferase were restored to normal, and the number of apoptotic hepatocytes in mice was also significantly elevated [33]. In myocardial infarction mice, direct intracardiac injection of MSCs underwent apoptosis within 24 h [34]. However, the left ventricular function and the fibrotic area of cardiac infarcts were effectively improved in mice after 4 weeks of MSCs transplantation compared with phosphate buffered saline-treated controls [34].

Studies applying anti-ApoMSCs on animal disease models revealed limited immunomodulatory effects and diminished efficacy [29, 35]. Liu et al. established anti-ApoMSCs by pretreatment with the caspase 3 inhibitor Z-DEVD-FMK [35]. Although there was some therapeutic effect, the Z-DEVD-FMK-treated MSCs secreted less anti-inflammatory factor tumor necrosis factor-alpha- -stimulated gene 6 (TSG-6), and their ability to regulate inflammation and inhibit scar formation was reduced compared with viable MSCs and H2O2-treated ApoMSCs [35]. Among them, H2O2-treated ApoMSCs had the best therapeutic effects [35]. Pang et al. established anti-ApoMSCs to treat ovalbumin (OVA) -sensitized asthmatic mice by deleting BAK/BAX (key proteins for apoptosis) in MSCs through the caspase 9 ribonucleoprotein complexes introduced via electroporation [29]. Although they were efficacious in improving lung histology, the extent of treatment was reduced compared to the viable MSCs-treated group [29]. In terms of immunosuppressive capacity, anti-ApoMSCs were shown not to downregulate the proliferation of activated T cells in diseased mice [29]. Serum specific pro-inflammatory factors interleukin (IL)-5 and IL-13 were also less down-regulated compared to the viable MSCs-treated group [29]. These therapeutic results with anti-ApoMSCs further side-step the fact that apoptosis of MSCs was necessary for immunosuppression and therapeutic efficacy in vivo.

The causes of spontaneous apoptosis of transplanted MSCs

Currently, the specific cause of apoptosis of MSCs after transplantation is still unclear. It is considered that the microenvironment at disease conditions might influence the fate of transplanted MSCs by factors like cytotoxic immune cells, hypoxia, inflammatory factors, nutritional deficiencies, microorganisms, etc [36].

Galleu et al. speculated that apoptosis of transplanted MSCs involves the presence of cytotoxic immune cells such as CD8+ T cells and CD56+ natural killer (NK) cells [32]. In GVHD mice, it was found that the apoptosis of MSCs was induced only in diseased mice infused with CD8+ T cells, but not in those not infused with CD8+ T cells [32]. The same result was also obtained by using Mh/ perforin knockout mice (defective cytotoxic GVHD effector cells) as the recipients [32]. Similar cytotoxicity on MSCs observation was made in an in vitro co-culture study [32]. Peripheral blood mononuclear cells (PBMCs) were isolated from GVHD patients who had favorable therapeutic outcomes with MSCs treatment (noted as PBMC+) and co-cultured with human BMMSCs for 4 h [32]. In contrast to the co-culture of PBMCs collected from patients who had poor response to MSCs treatment (noted as PBMCs), apoptosis of MSCs in PBMCs+ was observed to be upregulated 4-fold by flow cytometry [32]. Among them, CD56+ NK cells and CD8+ T cells populations in the PBMCs+ were the cells responsible for initiating MSCs apoptosis [32]. Apoptosis of MSCs may be caused by these toxic immune cells via granzyme B release and perforin-dependent and Fas/FasL ((Fas ligand) pathways [32]. This result was similarly validated in PBMCs extracted from patients with Crohn’s disease [37]. Compared to PBMCs, the PBMC+ group induced more significant human adipose-derived MSCs (ADMSCs) to apoptosis and higher levels of prostaglandin E2 (PGE2) secreted from human ADMSCs [37]. Researchers believed that these cytotoxic immune cells may serve as a clinical biomarker to assist practitioners in predicting the treatment effect of MSCs therapy as precision medicine [32, 37].

It has also been claimed that apoptosis of transplanted MSCs in vivo occurred due to hypoxia at the damaged site [16]. In diabetes mice, Preda et al. found that MSCs transplanted in the pancreas transiently triggered hypoxia-induced signaling pathways and activated caspase 3/7-mediated apoptosis in the presence of pro-inflammatory factors such as tumor necrosis factor-alpha (TNF-α) and interferon-gamma (IFN-γ) [16]. Interestingly, the authors found that both hypoxia and pro-inflammatory factors are essential for apoptosis [16]. In periodontitis mice, the expression level of the hypoxia-related factor hypoxia-inducible factor 1 alpha (HIF-1α) was significantly elevated in periodontitis tissues, demonstrating the presence of a hypoxic microenvironment, which induced apoptosis in the transplanted MSCs [38].

MSCs have been shown to treat a variety of inflammatory diseases, but few have explored the impact of the inflammatory environment on the fate of transplanted MSCs. Li et al. demonstrated that the presence of IFN-γ and TNF-α stimulated the high expression of inducible nitric oxide synthase in MSCs, which contributed to the production of intracellular nitric oxide (NO), thus up-regulating the expression of Fas on the surface of MSCs and aggravating endoplasmic reticulum stress to induce apoptosis [39].

In the ischemic heart, exogenous MSCs were exposed to a variety of pro-apoptotic or cytotoxic factors such as inadequate blood supply, hypoxic pressure, and inflammation triggered by tissue damage that may produce oxidative stress, cytotoxic free radicals, and proteins, all of which may trigger apoptosis [40]. After infusion of MSCs, any sites, and environments they pass through may induce any form of cell death, so the mechanism of apoptosis in transplanted MSCs in vivo remains a major challenge that is complex and still needs to be explored in depth.

Therapeutic applications of ApoMSCs and MSC-ApoEVs

Therapeutic applications of ApoMSCs

A growing number of studies have demonstrated that apoptosis of MSCs in vivo was necessary for their therapeutic effects. Therefore, infusion of ApoMSCs in vivo may be a more direct and efficient therapeutic option. Currently, the efficacy of ApoMSCs in various preclinical disease models such as sepsis, acute tissue injury, acute inflammation, autoimmune disease, and proliferative scarring of the skin has shown exciting results, as shown in Table 1.

Table 1 Typical therapeutic applications of ApoMSCs from different sources

Sepsis is commonly associated with the persistence of infection-induced immune dysregulation, with a strong attack of the hyperinflammatory response leading to functional organ dysfunction [41, 42]. Chang et al. effectively upregulated the survival rate of septic rats by using apoptotic ADMSCs(Apo-ADMSCs) [43, 44]. They effectively ameliorated disease-related cardiac, hepatic, pulmonary, and renal injuries through anti-inflammatory, anti-apoptotic, and antioxidant effects [43, 44]. To explore the acute immune response, the study observed peripheral blood and spleen-derived serum [43, 44]. It was found that circulating TNF-α levels in peripheral blood decreased significantly after treatment with Apo-ADMSCs, as well as the levels of toxic T-lymphocytes [43, 44].

In acute lung injury, apoptotic human umbilical cord MSCs ( Apo-hUCMSCs) have been shown to have better therapeutic effects than viable hUCMSCs [45]. ApoMSCs effectively reduced neutrophil infiltration and total protein infiltration in rat lungs, and improved alveolar-capillary membrane integrity [45]. The expression levels of relevant inflammatory factors such as the pro-inflammatory factor INF-γ decreased and the anti-inflammatory factor IL-4 increased in lung tissues [45].

Many studies have shown that melatonin could be effective in maintaining body function and homeostasis by down-regulating inflammatory and oxidative responses [46]. Interestingly, Yip et al. combined melatonin with Apo-ADMSCs for the treatment of acute pulmonary ischemia-reperfusion rats, and found that the therapeutic efficacy was superior to that of the melatonin group, and even superior to that of the melatonin-combined-viable MSCs group [47]. Combined treatment with melatonin and Apo-ADMSCs better-ameliorated hypoxemia and pulmonary arterial hypertension in the diseased mice [47]. Both inflammatory and oxidative protein markers were significantly decreased in the lung tissues, while antioxidant indicator levels increased [47]. The study also revealed anti-apoptotic effects in the tissues, such as a significant decrease in the levels of apoptotic markers phosphorylated H2AX (γ-H2AX), cleaved caspase 3, and poly (ADP-ribose) polymerase proteins (PARP), as well as an increase in the protein levels of anti-apoptotic protein markers such as Bcl-2 [47]. Recent studies have shown that ApoMSCs have more potent anti-inflammatory and oxidative stress-reducing properties than viable MSCs, which partly explains why melatonin works better in combination with Apo-ADMSCs [43, 44]. Overall, these studies offered better prospects for the treatment of ApoMSCs. See Fig. 1 for details.

Fig. 1
figure 1

Therapeutic applications of ApoMSCs and MSC-ApoEVs

MSCs induced apoptosis in vitro in various ways such as STS, inflammatory factor, H2O2, hypoxia, UVC light, and serum free. ApoMSCs and MSC-ApoEVs have great potential for the treatment of a wide range of diseases. Among them, for the characterization of MSC-ApoEVs, they were found to express apoptosis-related markers and EVs-related markers.

Therapeutic applications of MSC-ApoEVs

Biological properties of MSC-ApoEVs

MSCs underwent apoptosis during which they released apoptotic extracellular vesicles, which resulted from apoptosis [21]. This predicted that MSC-ApoEVs may play a similarly important role as ApoMSCs in the treatment of disease [21]. Before summarizing the therapeutic applications of MSC-ApoEVs, it is necessary to introduce the biological properties of MSC-ApoEVs.

Extracellular vesicles (EVs) are membrane-bound particles released by cells into the extracellular space [48]. EVs have played an important role in the diagnosis and treatment of various diseases due to the carrying of a wide range of molecules derived from the parental cells, such as lipids, proteins, nucleic acids, and metabolites [49]. Even EVs are increasingly being used as drug-delivery vehicles due to their favorable safety and unique biodistribution capabilities [50].

Classically, EVs can be classified into three subtypes depending on their origin: exosomes (Exos) (produced by endosomes, = 30–150 nm in diameter), microvesicles (MVs) (generated at the plasma membrane from its outward budding, ranging in diameter from less than 100 nm to a few micrometers) and apoptotic extracellular vesicles (ApoEVs) (produced by apoptotic cells, = 50–5000 nm in diameter) [23, 51, 52]. EVs produced by viable cells, including Exos and MVs, have been extensively studied. However, most people still limit their knowledge of ApoEVs to the level of the apoptotic bodies (ApoBDs) with large diameters [53,54,55,56]. Currently, ApoEVs can be classified into three subtypes according to their biogenesis and size: ApoBDs (=1–5 μm in diameter), microvesicle-like ApoEVs (ApoMVs) (=100–1000 nm in diameter), and exosome-like ApoEVs (ApoExos) (< 150 nm in diameter) [52, 57].

The biogenesis of ApoBDs is mainly related to actomyosin reorganization in the late stages of apoptosis, where the plasma membrane blebbing, protrusions, and fragmentation produce membrane-bound ApoBDs of varying sizes, which prevents the loss of cellular contents, especially some pro-inflammatory substances [55, 58].

For the biogenesis of ApoMVs, they may be generated during the early stages of apoptosis and by a mechanism like MVs production from viable cells: plasma membrane budding, but the exact molecular regulatory mechanism is not yet clear [57, 59].

ApoExos are also like Exos production from viable cells: they are released upon fusion of the multivesicular body with the plasma membrane [57, 60]. ApoExos were analyzed using transmission electron microscopy, where multivesicular endosomes containing internal vesicles were visible [60]. The characterization revealed the expression of Exos-associated protein markers such as CD63 and CD9 [60]. The mechanism of the formation may involve the sphingosine-1-phosphate/sphingosine-1-phosphate receptors signaling pathway, the autophagy-lysosomal pathway, or the Gasdermins-mediated pathway [60,61,62].

Functionally, ApoEVs, as key messengers released by apoptotic cells, not only recruit peripheral phagocytes for effective removal of apoptotic substances but also contain a variety of biomolecules, including microRNAs and proteins, to facilitate intercellular communication [57]. ApoEVs derived from a variety of different cells, such as MSCs, immune cells, and endothelial cells, have been found to play important roles in efferocytosis, immunomodulation, cell proliferation and survival, and angiogenesis [52, 63]. Here, we focus on MSC-ApoEVs.

Unlike the method of collecting viable MSC-EVs for therapy by centrifugation at 120,000 g, the isolation of MSC-ApoEVs for therapy is currently collected mainly by centrifugation at 16,000 g. Transmission electron microscopy and nanoparticle tracking analysis showed a larger and wider range of diameters than viable MSC-EVs, generally ranging from 150 to 1000 nm. Notably, the method of isolation was unable to distinguish between EVs from healthy, stressed, etc. with the same density and particle size, which highlight the limitations of isolating and purifying MSC-ApoEVs. They were characterized to express apoptosis-related specific surface markers such as PS, calreticulin (CRT), and component 1q (C1q) and to express apoptosis-specific proteins including cleaved caspase 3. Even EVs-related markers such as transmembrane/lipid-bound protein (e.g., CD63, CD9, CD81, Flotillin1) and cytosolic protein (e.g., tumor susceptibility gene 101 (TSG101), Alix) were characterized in MSC-ApoEVs. It predicted that the biogenesis of MSC-ApoEVs for therapy may involve exosome-like biogenesis. Related markers of parental MSCs: CD29(+), CD44(+), CD90(+), CD34(-) and CD45(-) were also characterized, as shown in Table 2.

Table 2 Characterization of MSC-ApoEVs

It has been shown that MSC-ApoEVs contain a variety of contents such as proteins, DNA, RNA, lipids, and metabolites from the parental MSCs, suggesting their therapeutic potential in cell-to-cell communication [23]. Proteomic analysis of ApoEVs derived from different sources of MSCs, such as human BMMSCs and human ADMSCs, revealed that 80% of the total proteins of the parental MSCs were present in MSC-ApoEVs, which were higher than viable MSC-EVs [78, 85]. They were enriched with many functional proteins related to cell behavior, cell metabolism, cellular transport, and regulation of various diseases [78, 85]. Furthermore, previous studies have shown significant similarity in total RNA profiles between ApoEVs and corresponding parental osteoclasts [86]. Combined with the fact that MSC-ApoEVs carry apoptosis-related markers, it predicted an immunomodulatory capacity similar to the parental ApoMSCs that can induce phagocytosis by phagocytes.

In conclusion, MSC-ApoEVs have the potential to be a more direct therapeutic agent than ApoMSCs, which is more in line with the current trend of advocating cell-free therapy. More and more studies are now demonstrating the therapeutic potential of MSC-ApoEVs, as shown in Table 3.

Table 3 Typical therapeutic applications of MSC-ApoEVs from different sources

Direct therapeutic applications

In mice with dorsal wound injury, BMMSCs transplanted directly at the wound site promoted skin wound healing but also underwent apoptosis within a short period of time [67]. Direct treatment of wounds with BMMSC-ApoEVs dissolved in the hydrogel Pluronic F (PF)-127 resulted in a reduction of the scar area, a more complete skin structure, the production of newly formed epithelium and hair follicles, and even a reduction of the inflammatory cell infiltration was observed [67]. Type 2 diabetes is often associated with poor wound healing, which can be difficult to treat. However, research has shown that applying hydrogel containing hUCMSC-ApoEVs to wounds in diabetic mice effectively accelerated wound healing [64]. Interestingly, ADMSC-ApoEVs of different diameter sizes showed different efficacy, and it was found that small diameter ADMSC-ApoEVs (less than 200 nm) were significantly more efficacious than large diameter ADMSC-ApoEVs (greater than 500 nm) [65]. Meanwhile, the age of the ADMSCs donor showed differences in the treatment of skin injuries. Although both young and old ADMSC-ApoEVs induced skin healing and reduced scar formation, the younger ADMSC-ApoEVs were more effective [87]. In addition to direct treatment at the wound site, it was found that intravenous injection of BMMSC-ApoEVs in wound-injured mice resulted in the accumulation of BMMSC-ApoEVs in the liver, skin, spleen, and lungs, with peak levels of BMMSC-ApoEVs occurring on the third day and significantly decreasing on the seventh-day post-infusion [68]. The study focused on the regression of BMMSC-ApoEVs at the skin injury and found that fluorescently labeled BMMSC-ApoEVs showed accumulation at the skin injury in mice within 12 h but disappeared on day 7 [68]. It may be related to the metabolism of BMMSC-ApoEVs by the epidermis and hair follicles of the skin as shown by structure illumination microscopy analysis, and the results were further confirmed in the symbiotic model [68]. The accumulation of BMMSC-ApoEVs at the site of skin damage effectively activated skin and hair follicle MSCs, thus effectively promoting skin healing and hair growth [68, 88].

MSC-ApoEVs have been shown to be effective in the treatment of bone damage. Zhu et al. found that whole-body fluorescence imaging of intravenously injected BMMSC-ApoEVs showed that BMMSC-ApoEVs were predominantly enriched in the liver, whereas the enrichment of BMMSC-ApoEVs in the femur gradually increased over time [75]. Injections of BMMSC-ApoEVs significantly ameliorated bone loss induced by primary (aged mice) and secondary (ovariectomy (OVX) mice) osteoporotic mice, with increased bone mass and improved bone microarchitecture in the mice [75]. Even in situ treatment with scaffolds in combination with BMMSC-ApoEVs was effective in promoting new bone generation in rats with cranial bone defects [75]. Interestingly, BMMSC-ApoEVs at different apoptotic stages differed in promoting osteogenesis. It has been claimed that BMMSC-ApoEVs from mid-apoptotic stages had the strongest regenerative effect relative to early and late apoptotic stages, which may be related to the differences in the substances contained in BMMSC-ApoEVs produced at different apoptotic stages [89]. Many studies have shown that a hypoxic environment can enhance the biological functions of MSCs [90]. On this basis, Ding et al. utilized a modified gelatin matrix/3D printed extracellular mesenchyme scaffold complex as a carrier to inject ADMSC-ApoEVs induced to apoptosis after hypoxic incubation directly into the damaged joint cavity exhibited better cartilage repair and regeneration than ADMSC-ApoEVs generated under normoxic conditions [79]. It may be related to the fact that ADMSC-ApoEVs generated under hypoxia were superior to those generated under normoxic conditions in terms of production, microRNA (miRNA), and functional proteins [79]. This engineering of 3D printed cells in combination with hypoxic preconditioning treatment protocol offers greater advantages and possibilities for the treatment of MSC-ApoEVs.

Of course, MSC-ApoEVs can also be an excellent treatment for dental injury diseases. In periodontitis mice, gingival sulcus injection of BMMSC-ApoEVs significantly ameliorated their alveolar bone destruction [38]. In a pulp removal model, treatment with stem cells from human exfoliated deciduous teeth (SHED)-ApoEVs promoted pulp revascularization and tissue regeneration [81]. In addition, MSC-ApoEVs showed great potential in the treatment of diseases such as hemophilia A [78], diabetes mellitus [91], myocardial infarction [34], autoimmune disorders [92, 93], sepsis [74], noisy hearing loss [85], cancer [69, 77], uterine adhesions [94], and aging [72].

Drug delivery therapy

In addition to direct therapeutic effects, MSC-ApoEVs can be used as packaged drug carriers for therapy. The apoptotic property of MSC-ApoEVs induces efferocytosis, which can be utilized to enable nanomedicine-loaded monocytes/macrophages to naturally target the damaged site for therapy [10]. It has been demonstrated that systemically infused MSC-ApoEVs can be home to hepatic macrophages [95]. Xiang et al. exploited this homing property and combined the antibacterial function of copper-doped carbon dots (Cu-CDs) to effectively enhance the ability of hepatic macrophage endocytosis of bacteria in septic mice by loading Cu-CD onto the entire surface of BMMSC-ApoEVs via electrostatic action [95]. In this case, small GTPase Rab5, which was enriched in BMMSC-ApoEVs, played an important role in the internalization of bacteria by hepatic macrophages and promoted the maturation of early phagolysosomes [95].

In a study by Yang et al., it was found that high concentrations of alpha-mangostin (α-M), a drug with anti-inflammatory, antioxidant, neuroprotective, and cardiovascular protective efficacy, were able to induce apoptosis in hUCMSCs, producing hUCMSC-ApoEVs enriched with α-M (α-M/ApoEVs) [96]. Surface modification of α-M/ApoEVs by co-administration of matrix metalloproteinase-activated cell-penetrating peptide could target brain damage and be phagocytosed by brain macrophage microglia [96]. More effective treatment of ischaemic stroke compared to untreated hUCMSC-ApoEVs [96].

Bortezomib (BTZ) is often used as a therapeutic agent for multiple myeloma, but side effects such as peripheral neurotoxicity, nephrotoxicity, and leukopenia are often associated with its treatment [97,98,99]. Cao et al. found that co-culturing Apo-BMMSCs with bortezomib, the naturally occurring apoptotic encapsulation system of Apo-BMMSCs can encapsulate bortezomib in BMMSC-ApoEVs [77]. The drug loading rate was higher than conventional sonication loading [77]. Bortezomib-loaded BMMSC-ApoEVs not only reduced the toxicity of BTZ but also produced better anti-multiple myeloma effects compared to natural BMMSC-ApoEVs and BTZ administered alone [77]. This may be a result of the combined effect of the small Rab GTPase Rab7 (a key regulator of vesicle maturation and secretion) and BTZ in BMMSC-ApoEVs [77].

The potential of MSC-ApoEVs in the treatment of bone damage has been demonstrated [71, 79]. Gui et al. affixed a bone-targeting peptide (Asp-Ser-Ser)6 ((DSS)6) to the surface of BMMSC-ApoEVs, and co-loaded ubiquitin ligase RING finger protein (RNF) 146 onto BMMSCs by adenoviral transduction to obtain functional BMMSC-ApoEVs [100]. This effectively improved the bone-targeting ability of BMMSC-ApoEVs and induced the osteogenic capacity of endogenous MSCs [100].

Therapeutic mechanisms of ApoMSCs

ApoMSCs and efferocytosis

The efficient clearance of apoptotic cells by phagocytes is also known as “efferocytosis” [101]. In most tissues, efferocytosis is carried out by either highly phagocytic tissue-resident professional phagocytes (e.g., macrophages and dendritic cells) or less phagocytic non-professional phagocytes (which can be neighboring cells such as epithelial cells and fibroblasts) [101]. Phagocytes have positive anti-inflammatory and immunosuppressive effects on apoptotic cells after efferocytosis [101]. ApoMSCs can also interact with phagocytes to exert anti-inflammatory mechanisms. See Fig. 2 for details.

Fig. 2
figure 2

Therapeutic mechanisms of ApoMSCs

MSCs transplanted in vivo undergo apoptosis for various reasons such as inflammatory factor, hypoxia, microorganisms, cytotoxic immune cells, and nutritional deficiencies. Mechanisms of therapeutic effects of ApoMSCs. (1) ApoMSCs and efferocytosis: ApoMSCs induce macrophages to exert immunomodulatory effects by binding to the macrophage surface receptor MerTK via surface PS, in which ApoMSCs attract more CCR2 chemokines to recruit more macrophages. Macrophages regulated by ApoMSCs promote their own M2 polarisation and inhibit M1 polarisation, inhibit neutrophil and T-cell activation. (2) Release of soluble factors by ApoMSCs: MSCs secrete a variety of soluble factors after apoptosis, e.g. TSG-6, PGE2, PAI-1, M-CSF, IL-33, paracrine factors inhibit tissue inflammation, promote macrophage M2 polarisation and inhibit T-cell activation. (3) Release of MSC-ApoEVs: See Fig. 3 for details.

In GVDH mice, apoptosis BMMSCs (Apo-BMMSCs) induced efferocytosis by recipient macrophages and produced indoleamine 2,3-dioxygenase (IDO) ameliorating the infiltration of immunotoxic cells [32]. IDO is an immunosuppressive molecule that promotes the conversion of tryptophan to kynurenine and locally reduces the concentration of the essential amino acid tryptophan, which has been shown to reduce T-cell proliferation, thereby inhibiting T-cell-mediated immune responses [32, 102].

In allergic asthmatic mice, efferocytosis of Apo-BMMSCs induced alterations in the metabolic and inflammatory pathways of alveolar macrophages, i.e., a shift in the M2 phenotype and secretion of IL-10, which acted as an immunosuppressor to reduce the number of pulmonary eosinophils and decrease the severity of the disease [29]. Macrophages are characterized by remarkable plasticity and versatility, capable of switching from one phenotype to another, i.e., M1 phenotype to M2 phenotype [103]. M1 macrophages often promote the production of reactive oxygen species (ROS) and nitrogen intermediates, and the production of inflammatory factors such as IL-1 beta(β), TNF-α, and IL-6, which contribute to pro-inflammatory, bactericidal, and anti-tumour effects [103]. And M2 macrophages can produce IL-10, transforming growth factor-beta (TGF-β), vascular endothelial growth factor (VEGF), etc., which is conducive to the regulation of immunity, inhibition of inflammation, and promotion of angiogenesis [103]. He et al. found that Apo-BMMSCs ameliorated acute liver injury by binding to the macrophage surface Mer tyrosine kinase (MerTK) receptor via cell-surface PS and undergoing an M2 phenotypic shift by recruiting Ly-6Chigh monocyte-derived macrophages partly via chemokine receptor C–C motif chemokine receptor 2 (CCR2) as well as producing IL-10 [33]. Interestingly, direct administration of PS liposomes mimicking Apo-BMMSCs exerted similar protective effects [33].

Release of soluble factors by ApoMSCs

In addition to efferocytosis, ApoMSCs can also directly secrete immunosuppressive factors to exert therapeutic effects. In an in vitro cellular assay, human ADMSCs were exposed to anti-Fas activating antibody and recombinant granzyme B to induce apoptosis and their transcriptional analysis revealed that, among a series of up-regulated genomes present in Apo-ADMSCs, immunomodulatory genes were the most prominently up-regulated, which mainly contained: IL-6, leukemia inhibitory factor, tumor necrosis factor alpha-induced protein 6, and cyclooxygenase 2 encoding gene prostaglandin-endoperoxide synthase 2, followed by the apoptotic genome [37]. Specifically, the authors found that Apo-ADMSCs were induced to produce a series of immunosuppressive molecules after caspase activation under the regulation of the NF (nuclear factor)-kappa(κ)B pathway, among which PGE2 was essential for Apo-ADMSCs to directly inhibit T cells activation [37]. This was similarly demonstrated in mice with acute colitis, where the addition of induced Caspase 9 MSCs with a suicidal apoptosis gene could inhibit T-cell activation by secreting PGE2 to promote weight regain, improved survival, and decreased inflammation, such as a decrease in serum levels of TNF-α and IL-6, in the diseased mice [19]. In rabbits with proliferative scarring, direct injection of H2O2-induced Apo-BMMSCs produced more anti-inflammatory factors, such as TSG-6, through the caspase 3 activation pathway, which effectively down-regulated pro-inflammatory/pro-fibrotic mediators, such as monocyte chemotactic protein-1, macrophage inflammatory protein-1 beta, IL-6, and TGF-β1, in the tissues, and ameliorated the formation of scarring [35].

Apoptotic cell-induced alterations in macrophage polarization of the M2 phenotype have been demonstrated in various types of apoptotic cells, but correlates secreted by ApoMSCs gave them a further predominant advantage [104,105,106]. In ischemic muscle injury mice, after induction of apoptosis by three different cell types such as BMMSCs, murine fibroblast cell line NIH 3T3 and murine macrophage cell line RAW 264.7, apoptotic supernatants of BMMSCs and murine fibroblast cell line NIH 3T3 significantly down-regulated TNF-α produced by macrophages activation, whereas apoptotic supernatants of murine macrophage cell line RAW 264.7 required 8–16 times more cells to produce the same efficacy as the other two [107]. This may be related to the different expression content of functional proteins in the cells [107]. In the apoptotic supernatant of BMMSCs, various proteins associated with the polarization of M2 macrophage anti-inflammatory phenotype, such as plasminogen activator inhibitor 1 (PAI-1), macrophage-colony stimulating factor (M-CSF), and IL-33, were highly expressed [107]. Although there are fewer comparative studies in this area, they still illustrate the excellent therapeutic potential of ApoMSCs.

Release of MSC-ApoEVs

Immunomodulatory capacity of MSC-ApoEVs

Recent studies have shown that MSC-ApoEVs, as metabolic secretions of ApoMSCs, played a similar role in inducing macrophage phagocytosis to exert positive anti-inflammatory and immunosuppressive effects. It has been found that BMMSC-ApoEVs expressed ‘eat’ me signaling: calreticulin on their surface, which attracted phagocytosis by hepatic macrophages [91]. This effectively restored hepatic macrophage homeostasis and allowed them to exert anti-inflammatory effects [91]. It played an important role in high-fat-induced type 2 diabetic mice, where the infusion of BMMSC-ApoEVs accumulated in the liver effectively improved glucose tolerance, attenuated insulin resistance, and ameliorated hepatic steatosis, among others [91]. The anti-inflammatory effect of MSC-ApoEVs was also demonstrated in acute endometrial injury mice. In situ treatment of hUCMSC-ApoEVs combined with hyaluronic acid hydrogel enhanced macrophage recruitment and phagocytosis and increased the number of M2 macrophages, resulting in high levels of IL-10 production [94]. It effectively increased the endometrial thickness and the number of endometrial glands, reduced the fibrotic area, improved the endometrial tolerance and ultimately promoted the restoration of fertility [94]. See Fig. 3 for details.

Fig. 3
figure 3

Therapeutic mechanisms of MSC-ApoEVs

(1) Immunomodulatory effects: MSC-ApoEVs can regulate the activation of macrophages, neutrophils, and T cells. (2) Rescue damaged MSCs: functional proteins and miRNAs carried by MSC-ApoEVs can promote self-renewal and differentiation of endogenously damaged MSCs. (3) Promote apoptosis of diseased cells: induction of apoptosis of diseased cells by Fas/FasL. (4) Promote angiogenesis: ECs undergo proliferation, migration, and autophagy through functional proteins or miRNA and PD1/PDL1 from MSC-ApoEVs and thus angiogenesis is promoted.

In vitro, BMMSC-ApoEVs obtained after TGF-β1/IFN-γ-licensed BMMSCs-induced apoptosis effectively suppressed lipopolysaccharide (LPS)-stimulated inflammatory responses in THP-1 [76]. This may be related to the fact that BMMSC-ApoEVs were partially phagocytosed by pro-inflammatory macrophages through the phosphatidylinositol 3-kinase pathway, effectively inhibiting the secretion of pro-inflammatory factors such as TNF-α and IL-1β [76]. In a similar study, it was found that BMMSC-ApoEVs may inhibit inflammation in pro-inflammatory macrophages via the AMPK/SIRT1/NF-κB pathway, as demonstrated in pro-inflammatory bone marrow-derived macrophages induced by porphyromonas gingivalis derived LPS [108].

More and more studies have shown that MSC-ApoEVs are indispensable for regulating macrophage anti-inflammatory polarisation in terms of the microRNAs they carry. Li et al. found that miRNA (miR)-21–5p carried in ADMSCs-ApoEVs induced M2 polarisation in macrophages and enhanced skin wound healing by targeting the kruppel like factor 6 (KLF6) gene [109]. Similarly, recently Mao et al. found that miR-20a-5p carried in ADMSCs-ApoEVs effectively ameliorated diabetic rat-induced skin injury by promoting macrophage M2 polarization by regulating the JAK-STAT1 signaling pathway [80].

The immunomodulatory mechanism of MSC-ApoEVs, in addition to its significant role in regulating the macrophage M1/M2 phenotypic transition, still played an important potential in regulating other pro-inflammatory behaviors of macrophages such as the occurrence of pyroptosis and anaerobic glycolysis. During skin wound repair in type 2 diabetes mice, a large number of macrophages located in the injured area underwent pyroptosis due to oxidative stress, resulting in intense inflammation [61]. In situ treatment with hUCMSC-ApoEVs can effectively improve wound healing by decreasing macrophage NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome production, which is upstream of pyroptosis [61]. Jiang et al. found that hUCMSC-ApoEVs-expressed programmed cell death 1 ligand 1 (PDL1) caused macrophages to undergo a metabolic switch from glycolysis to oxidative phosphorylation (OXPHOS) through the Erk pathway downstream of the PDL1- programmed cell death protein 1 (PD1) axis, thereby shifting from pro-inflammatory to anti-inflammatory state [85]. The application of hUCMSC-ApoEVs effectively ameliorated the inflammation of lung tissues in acutely lung-injured mice [85].

MSC-ApoEVs played an important role in regulating T cell activation. In vitro, BMMSC-ApoEV derived from TGF-β1/ IFN-γ-licensed MSCs inhibited T cell proliferation and reduced activated CD69+ T cells [76]. However, the induction of Tregs was promoted and the expression of immunomodulatory CD73+ T cells was maintained [76]. Efferocytosis induced by apoptotic cells in macrophages often produces anti-inflammatory mediators that inhibit the immune response and which play an important role in suppressing T-cells activation [76]. However, it has recently been shown that the expression of PS on the surface of MSC-ApoEVs can directly inhibit T-cells activation [92]. Previous studies have shown that PS specific to the surface of apoptotic cells often binds directly or indirectly to phagocytic receptors such as the TIM (T-cell immunoglobulin domain and mucin domain) family and TAM (TYRO3、AXL and MERTK) family to exert anti-inflammatory effects [101]. Wang et al. found that after in vivo infusion of BMMSC-ApoEVs in systemic lupus erythematosus mice, in the absence of macrophages, BMMSC-ApoEVs inhibited T cell activation through direct contact between surface PS and TIM-3 receptors on the surface of activated CD4+ T cells [92]. This effectively contributed to the reduction of IFN-γ and IL-2 levels from serum and the improvement of the corresponding clinical symptoms in lupus and arthritis [92].

MSC-ApoEVs had great potential in regulating neutrophil activation [74]. It has been claimed that MSC-ApoEVs can alter the form of neutrophil death, thereby inhibiting the activity [74]. In septic mice, neutrophils underwent a unique form of cell death, namely neutrophil extracellular traps (NETs) consisting of citrullinated histone H3 and myeloperoxidase (NETosis) [74]. Intravenous administration of BMMSC-ApoEVs switched the mode of neutrophil death from NETosis to apoptosis via the Fas/FasL pathway [74]. This effectively inhibited the infiltration of neutrophils and monocytes into distal organs, thereby improving the survival rate of septic mice [74] Interestingly, BMMSC-ApoEVs and NETosis can be attracted to each other by electrostatic interactions [74]. Specifically, positively charged NETs, which were mainly concentrated in the bone marrow and spleen, could be effectively regulated by attracting negatively charged BMMSC-ApoEVs to contact them [74].

MSC-ApoEVs rescued damaged endogenous MSCs

Most adult organs contain regenerative stem cells, which are essential for maintaining tissue homeostasis and promoting repair after injury [110]. Apoptosis-induced compensatory proliferation-induced regenerative repair plays a crucial role in the proliferation of neighboring cells, especially neighboring stem cells [111]. Previous studies have shown that the specific mechanism of apoptosis-induced compensatory proliferation (AICP) was mainly dependent on caspase activation and driven by a paracrine mechanism: secretion of mitogens [112]. Recently, it has been shown that mitogenic signals generated by apoptosis-induced compensatory proliferation may be utilized by neighboring MSCs in the form of ApoEVs, thus exerting a reparative effect. Brock et al. found that in zebrafish epidermis, MSC-ApoEVs containing Wnt8a were generated in a caspase-activated manner after the apoptosis of basal stem cells [113]. Phagocytosis of MSC-ApoEVs was performed by neighboring MSCs to activate the Wnt signaling pathway and induce the proliferation of MSCs in the epidermis [113]. Recently, it was found that zebrafish epidermal basal stem cells induced apoptosis can generate MSC-ApoEVs carrying macrophage inhibitory factor (MIF) on their surface [114]. It could promote the proliferation of neighboring epidermal basal MSCs by regulating the MIF/CD74/ERK signaling axis [114].ROS is usually regarded as deleterious to the cells, however, ROS plays an important role in AICP and tissue regeneration [115, 116]. On this basis, it has been found that low concentrations of BMMSC-ApoEVs could promote the production of beneficial ROS by recipient cells and promote BMMSCs proliferation, migration, and osteogenic differentiation through activation of JNK signaling [89]. In an in vitro cellular study, it was found that dental pulp stem cells (DPSC)-ApoEVs may promote DPSCs osteogenic differentiation by activating the ERK1/2 signaling pathway [83].

As MSC-ApoEVs, they themselves carried MSCs regulatory-related factors to play important roles in the self-renewal and differentiation ability of damaged endogenous MSCs, such as microRNA. Among them, miR-1324, miR-328-3p, and miR-145a-5p carried by MSC-ApoEVs have a major role in the promotion of bone injury diseases [71, 75, 93]. In Zhu et al.‘s study, the amelioration of bone damage may be related to the release of miR-1324 from BMMSC-ApoEVs [75]. Specifically, miR-1324 inhibited the expression of the target gene Sorting Nexin 14 (SNX14), which activated the SMAD 1/5 pathway in endogenous BMMSCs [75]. This effectively promoted BMMSCs osteogenesis and inhibited osteoclast differentiation [75]. In another study, Liu et al. found that BMMSC-ApoEVs could promote self-renewal and differentiation by activating the Wnt/β- catenin pathway in endogenous MSCs [93]. This may be related to the presence of RNF146 and miR-328-3p in BMMSC-ApoEVs to downregulate Axin1 [93]. Activation of the wnt/β-catenin pathway also upregulated FasL expression in endogenous MSCs, contributing to osteoclast apoptosis [93]. In a recent study, Zhang et al. found that BMMSC-ApoEVs can downregulate TGF-β/Smad 2/3 signaling in OVX MSCs via miR-145a-5p [71]. miR-145a-5p also activated Wnt/β-catenin-linker protein signaling through downregulation of TGF-β receptor 2 (TGF-βR2) and Dickkopf 1 (DKK1) [71]. These promoted the self-renewal and differentiation of endogenous MSCs and effectively ameliorated osteoporosis in mice [71]. However, it has also been reported that the specific Wnt/β-catenin signaling was not as effective in improving osteoporosis in mice. It has also been reported that hsa-miR-4485-3p, which is specifically present in BMMSC/ADMSC-ApoEVs, inhibits osteogenic differentiation and promotes adipogenic differentiation of MSCs by regulating the AKT signaling pathway [117]. Taking advantage of this property, customized MSC-ApoEVs knocking down hsa-miR-4485-3p demonstrated better bone regeneration therapeutic effects [117].

In addition to miRNAs in MSC-ApoEVs, several functional protein molecules played important roles in regulating endogenous stem cell injury. In a model of aging-associated bone loss, Lei et al. found that young BMMSC-ApoEVs carried high levels of Ras-related GTP binding protein 7 (Rab 7) that acted on receptor-aged MSCs to restore autolysosomal lysosome formation [118]. It activated cellular autophagy, whereas promoted regeneration and osteogenic and lipogenic differentiation in MSCs [118]. This was confirmed in the Li et al. study, BMMSC-ApoEVs could carry a large number of Ras proteins to activate the Ras/Raf1/Mek/Erk pathway [119]. It promoted osteogenesis and bone formation of MSCs in vivo and in vitro, and effectively improved osteoporosis in mice [119]. MSC-ApoEVs have been shown to promote skin MSCs regeneration in skin injury, but some studies have shown that embryonic stem cells-derived ApoEVs (ESC-ApoEVs) showed better efficacy than hUCMSC-ApoEVs [88]. It may be related to the fact that ESC-ApoEVs expressed more functional nuclear proteins from their parental MSCs, such as SOX2, which can activate the Hippo signaling pathway of skin MSCs [88].

Recently, it has also been shown that ESC-ApoEVs were significantly better than hUCMSC-ApoEVs in the treatment of senile osteoporosis [120]. This may be related to the fact that ESC-ApoEVs carried high levels of functional proteins associated with the maintenance of mitochondrial homeostasis [120]. Specifically, protein mass spectrometry analysis showed that ESC-ApoEVs highly expressed TCOF1, a nucleolar factor that regulates nucleolar transcription, compared with hUCMSC-ApoEVs [120]. In vitro, TCOF1 in ESC-ApoEVs rescued mitochondrial dysfunction in senescent BMMSCs by upregulating the mitochondrial protein FLVCR1 [120]. However, this was reversed by the knockdown of TCOF1 in ESC-ApoEVs using siRNA, and FLVCR1 levels were also decreased in senescent BMMSCs [120].

Angiogenesis promoted by MSC-ApoEVs

Angiogenesis is an essential step in tissue repair due to the ability of blood vessels to transport nutrients and oxygen to support cells at the wound site [121]. Recently, more and more studies have shown that MSC-ApoEVs play an influential role in the therapy of angiogenic diseases. Angiogenesis often contributes to skin wound healing, and in skin-injured rats, ADMSC-ApoEVs effectively induced endothelial cells(ECs) to proliferate, migrate, and undergo angiogenesis, thereby reducing the area of wound injury and accelerating wound healing [87]. However, compared with normal MSC-ApoEVs oxygen stress-treated MSC-ApoEVs (Oxi-MSC-ApoEVs) were more effective in promoting angiogenesis at the wound site, which may be related to the fact that Oxi-MSC-ApoEVs contained a large amount of miR-210-3p [84]. miR-210-3p promoted ECs migration and activation by activating AKT signaling [84].

The dental pulp is a vessel-rich tissue in stable root canals, and in pulp-in-situ injured Beagle dogs, ApoEVs derived from DPSC -ApoEVs could be phagocytosed by ECs to transfer mitochondrial Tu translation elongation factor (TUFM) [81]. TUFM could specifically activate the endogenous ECs’ autophagy pathway via the autophagy pathway of the transcription factor EB (TFEB), which promotes angiogenesis [81]. Ultimately, accelerated revascularization promoted efficient regeneration of the dental pulp, which was rendered ineffective by the addition of apoptosis inhibitors [81]. This mechanism was demonstrated in mice with ischemic heart disease, i.e. myocardial infarction [34]. Direct intramyocardial injection of BMMSC-ApoEVs induced autophagy in ECs after being phagocytosed by recipient ECs, thereby enhancing angiogenesis and recovery of cardiac function [34]. This may be related to the fact that donor BMMSC-ApoEVs promoted TFEB-mediated lysosomal function, which upregulated autophagy in recipient ECs to promote angiogenesis [34]. It also involved AKT-mediated activation of the VEGF signaling pathway and NO generation [34].

The PD1/PDL1 axis has been reported to regulate glycolytic metabolism during inflammation, however, recent studies have found that it also has great potential in regulating ECs glycolytic metabolism [82, 122]. In ischemic retinopathy mice, ECs at the retinopathy site highly expressed PDL1 due to hypoxia, whereas ApoEVs derived from SHED-ApoEVs superficially expressed high levels of PD1 [82]. SHED-apoVs could inhibit ECs’ anaerobic glycolysis via the PD1-PDL1 pathway to promote angiogenesis [82].

Apoptosis promoted by MSC-ApoEVs in pathogenic cells

As BMMSC/ADMSC-ApoEVs, the apoptosis-typical receptor Fas carried on their surface could activate platelet coagulation by binding to the platelet surface FasL, thereby rescuing hemophilia A mice with factor VIII knockout [78]. However, the surface of MSC-ApoEVs, in addition to carrying the apoptosis-associated factor Fas, was highly expressed in FasL, which may be related to the inheritance of properties from parental MSCs, which played an important role in the induction of apoptosis [123, 124]. In addition to the previously mentioned alteration of neutrophil death pattern by BMMSC-ApoEVs through Fas/FasL, in multiple myeloma mice, BMMSC-ApoEVs utilized their FasL to directly activate the calcium channel-induced Fas membrane transport pathway in multiple myeloma cells to cause apoptosis of multiple myeloma cells and extended the survival rate of multiple myeloma mice [69, 74] It has even been found that BMMSC-ApoEVs could promote apoptosis of myoblasts and then cause them to release myoblast-derived ApoEVs, which could release creatine through activated Pannexin 1 channels and eventually increased the fusion of myoblasts to promote muscle regeneration [70].

Maintenance of homeostasis of other tissues by MSC-ApoEVs

Surely, some of the functional molecules carried by MSC-ApoEVs have also shown remarkable efficacy in the treatment of other diseases such as aging, and in the maintenance of organ homeostasis such as the liver, ovaries, ears, and teeth.

It has been demonstrated that BMMSC/hUCMSC/ESC-ApoEVs can inherit nuclear proteins from their parental MSCs, which was also confirmed by Huang et al. They found that MSC-ApoEVs could inherit a variety of DNA repair-related proteins from parental MSCs, including PARP1, Rad51, Rad52, XRCC4-like factor, Proliferating Cell Nuclear Antigen, damage-specific DNA binding protein 1, X-ray Repair Cross Complementing Protein 1, and Recombinant Prominin 1, which were higher than viable MSC-EVs [72]. Among them, PARP1 played a key role in MSC-ApoEVs-mediated DNA repair, which could effectively ameliorate premature senescence and DNA damage induced by lethal dose irradiation in apoptosis-deficient mice [72].

Defective apoptosis resulted in hepatic injury in mice, however, infusion of BMMSC/hUCMSC-ApoEVs effectively protected hepatocyte health and maintained hepatic homeostasis, which may be related to the presence of membrane proteins on the surface of MSC-ApoEVs that can interact with hepatocytes [125]. Specifically, MSC-ApoEVs were endocytosed by hepatocytes via Gal(Galactose) /GalNAc (N-acetylgalactosamine) -ASGPR (asialoglycoprotein receptor) and formed ApoEVs-Golgi complex (AGC) by binding to the hepatocyte Golgi surface receptor syntaxin 16(STX16) via membrane surface vesicle-associated membrane protein 3(VAMP3) [125]. The AGC promoted microtubule acetylation by regulating α-microtubule protein N-acetyltransferase 1, thereby facilitating the cytoplasmic division of hepatocytes [125]. This effectively promoted liver regeneration and effectively saved acute liver failure [125].

Naturally, MSC-ApoEVs also played an instrumental role in the maintenance of ovarian homeostasis [73]. It has been shown that apoptosis defects caused the development of polycystic changes and oocyte damage in ovary mice, while BMMSC-ApoEVs effectively ameliorated follicular dysfunction [73]. This was attributed to the fact that BMMSC-ApoEVs inherited the Wnt membrane receptor inhibitor protein RNF43 from parental MSCs [73]. Specifically, BMMSC-ApoEVs could deliver the functional protein RNF43 to the theca cells (TCs) through a characteristic vesicle-cell membrane integration mechanism. And then interacted with the Wnt receptor frizzled-4 (FZD4), which could effectively down-regulated aberrantly activated Wnt/β-catenin signaling in the TCs [73]. The restored Wnt/β-catenin signaling down-regulated reactively activated DKK1 levels in the oocyte regions, thereby effectively rescuing the down-regulated Wnt/β-catenin signaling in the oocytes [73]. This played an important role in promoting ovarian folliculogenesis [73]. The functional pathway of the NPPC/cGMP/PDE3A/cAMP cascade in follicles was also restored [73]. In addition to apoptosis-deficient mice, the amelioration of ovarian dysfunction by apoptotic follicles was demonstrated in dehydroepiandrosterone-induced polycystic ovarian syndrome mice and in mice with aging-associated ovarian dysfunction [73].

Furthermore, MSC-ApoEVs have tremendous potential for the treatment of hearing impairment [85]. Proteomic analysis of hUCMSC-ApoEVs revealed that they were rich in various oxidoreductases and signal transduction and activators of transcription 3 (STAT3), a transcription factor that responds to various cytokines or growth factors [85]. It was found that hUCMSC-ApoEVs could reduce oxidative stress and ameliorate noise-induced hearing loss in mice by promoting nuclear translocation of activated forkhead box o3 (FOXO3a) and upregulation of superoxide dismutase 2 via STAT3 in damaged cells [85].

In periodontitis mice, gingival sulcus injection of hypoxia-induced BMMSC-ApoEVs better induced targeted phagocytosis of BMMSC-ApoEVs by osteoclasts since both BMMSC-ApoEVs and osteoblasts express dendritic cell-specific transmembrane protein (DC-STAMP) (a membrane fusion regulator) [38]. Meanwhile, miR-223-3p (a key regulator of osteoclast differentiation and bone resorption) in BMMSC-ApoEVs effectively ameliorated alveolar bone destruction by targeting Itgb1 to attenuate osteoclast formation and reduced bone resorption [38].

Challenges and prospects

Most of the existing reports suggested that apoptosis of transplanted MSCs in vivo may be induced by the microenvironment in which they are transplanted, such as hypoxia, inflammatory factors, nutritional deficiencies, cytotoxic immune cells, microorganisms, etc [36]. However, based on the currently available data, it is still difficult to know the clear mechanism, which limits the exploration of both in vitro expansion methods and in vivo therapeutic mechanisms for apoptosis-dependent MSCs therapy. In other studies, MSCs pretreated with inflammatory factors or hypoxia could also exert favorable therapeutic effects. In vitro, IFN-γ-stimulated hUCMSCs could effectively inhibit NK cells activation and resist NK-mediated cytotoxicity through the production of IDO [126]. In hindlimb ischemic mice, hypoxia preconditioned MSCs activated the HIF-1α/GRP78/Akt signaling axis to repair mouse injury [127]. Surprisingly, some studies have shown that pretreatment with the inflammatory factor IFN-γ upregulated apoptosis-related genes such as Fas and interferon regulatory factor 1 [39, 128]. Hypoxia has also been used in many studies as a method to induce apoptosis [38, 79]. This evidence suggested that the same mechanism of downstream driving effect may exist between the significant therapeutic effects of inflammatory factors and hypoxic preconditioning and the efficacy produced by cell-induced apoptosis [18]. However, it is also necessary to note that in many cases ApoMSCs therapy relies on tissue-resident macrophages, but the state of macrophages such as chemotaxis and numbers are difficult to predict in different diseases, which is a factor to be considered in the use of apoptotic MSCs therapy.

In this review, we also explored MSC-ApoEVs, the administration of which may be a favorable cell-free alternative treatment due to the inheritance of important molecules and properties from the parental MSCs. Although viable MSC-EVs have been found to be therapeutically effective, available technologies are still limited in the number of viable MSC-EVs that can be produced in vitro [129]. In contrast, MSC-ApoEVs are readily available and have a high production rate. It has been found that BMMSC-ApoEVs were produced ten times more than viable BMMSC-EVs [92]. Furthermore, BMMSCs produced 245 times more BMMSC-ApoEVs in vitro compared to the same number of T cells [10, 92]. Yang et al. found that the same number of ADMSCs produced six times and four times more ADMSC-ApoEVs particles and proteins, respectively than viable ADMSC-EVs within a 12-hour period [65]. However, MSC-ApoEVs therapy has been an emerging therapeutic modality in the last few years, and we have yet to thoroughly investigate the complexity and heterogeneity of its contents and the stability of its amplification in vitro. We found that some studies have used gels such as PF-127, gelatin methacrylate, and modified gelatin such as the synthesis of norbornene-modified gelatin, and the synthesis of tetrazine-modified gelatin to slow down the degradation rate of MSC-ApoEVs when mixed with them [64, 65, 79]. Even the tissue engineering approach of constructing 3D-printed extracellular matrix environments in vitro further improved the efficacy of MSC-ApoEVs [79]. The adoption of such engineered MSC-ApoEVs offers more possibilities for its developmental applications.

Questions remain regarding the therapeutic options for ApoMSCs and MSC-ApoEVs. In vitro, it has been found that it was ApoMSC, but not MSC-ApoEVs, that were able to produce PGE2 to inhibit T-cell activation, furthermore, in vivo MSCs were shown to undergo apoptosis by means of cell-to-cell contact [37]. It has been shown that the anti-inflammatory effects of apoptotic cells during efferocytosis are closely related to the metabolites they produce at different stages of efferocytosis. Even though receptors that induce efferocytosis are present on the surfaces of both ApoMSCs and MSC-ApoEVs, some differences in the therapeutic mechanisms of these two types of cells may remain, which will require a contextual analysis of the utilization of ApoMSCs/ MSC-ApoEVs in future therapies.

Conclusions

There is growing evidence of the potential therapeutic and even drug delivery capacity of ApoMSCs and MSC-ApoEVs. In this review, we concluded the therapeutic mechanism of ApoMSCs, found that they played significant roles in immunomodulation, such as inducing macrophage polarization into anti-inflammatory phenotype and producing immunosuppressive secretome, and tissue regeneration, such as through secretion of MSC-ApoEVs carrying regenerative molecules, as well as in the promotion of angiogenesis, inducing apoptosis of diseased cells, and maintaining tissue homeostasis. This evidence provides new ideas for investigators of MSCs-based therapies. Among them, we found that MSC-ApoEVs are broadly promising for therapeutic applications due to their inheritance of important properties from their parental MSCs, as well as their easy availability and high yield. However, we have not yet clarified the specific mechanism of apoptosis of transplanted MSCs in vivo, which limits the exploration of both in vitro expansion methods and in vivo therapeutic mechanisms that rely on ApoMSCs for therapeutic purposes. Moreover, the therapeutic application of ApoMSCs and MSC-ApoEVs is still in the early stages, and further research and exploration of the differences between the two and their respective advantages are still necessary.

Data availability

Not applicable.

Abbreviations

MSCs:

Mesenchymal stem cells

BMMSCs:

Bone marrow mesenchymal stem cells

ApoMSCs:

Apoptotic mesenchymal stem cells

MSC-ApoEVs:

The apoptotic extracellular vesicles secreted by ApoMSCs

PCR:

Polymerase chain reaction

GVHD:

Graft-versus-host disease

PS:

Phosphatidylserine

TSG-6:

Tumor necrosis factor -alpha -stimulated gene 6

OVA:

Ovalbumin

IL-5:

Interleukin-5

NK cells:

Natural killer cells

PBMCs:

Peripheral blood mononuclear cells

FasL:

Fas ligand

ADMSCs:

Adipose-derived mesenchymal stem cells

PGE2:

Prostaglandin E2

TNF-α:

Tumor necrosis factor -alpha

IFN-γ:

Interferon-gamma

HIF-1α:

Hypoxia-inducible factor 1 alpha

NO:

Nitric oxide

hUCMSCs:

Human umbilical cord mesenchymal stem cells

γ-H2AX:

phosphorylated H2AX

PARP:

Poly (ADP-ribose) polymerase proteins

EVs:

Extracellular vesicles

Exos:

Exosomes

MVs:

Microvesicles

ApoBDs:

Apoptotic bodies

ApoMVs:

Microvesicle-like ApoEVs

ApoExos:

Exosome-like ApoEVs

CRT:

Calreticulin

CIq:

Component 1q

TSG101:

Tumor susceptibility gene 101

PF:

Pluronic F

OVX:

ovariectomy

miRNA:

MicroRNA

SHED:

human exfoliated deciduous teeth

Cu-CDs:

Copper-doped carbon dots

α-M:

Alpha-mangostin

BTZ:

Bortezomib

RNF 146:

Ring finger protein 146

IDO:

Indoleamine 2,3-dioxygenase

ROS:

Reactive oxygen species

TGF-β:

Transforming growth factor-beta

VEGF:

Vascular endothelial growth factor

MerTK:

Mer tyrosine kinase

CCR2:

C–C motif chemokine receptor 2

NF-κB:

Nuclear factor-kappa B

PAI-1:

Plasminogen activator inhibitor 1

M-CSF:

Macrophage-colony stimulating factor

LPS:

Lipopolysaccharide

miR:

MiRNA

KLF6:

Kruppel like factor 6

NLRP3:

NOD-, LRR-, and pyrin domain-containing protein 3

PDL1:

Programmed cell death 1 ligand 1

OXPHOS:

Oxidative phosphorylation

PD1:

Programmed cell death protein 1

TIM:

T-cell immunoglobulin domain and mucin domain

TAM:

Tyro3、Axl and MerTK

NETs:

Neutrophil extracellular traps

NETosis:

Nets consisting of citrullinated histone h3 and myeloperoxidase

AICP:

Apoptosis-Induced Compensatory Proliferation

MIF:

Macrophage inhibitory factor

DPSC:

Dental pulp stem cells

SNX14:

Sorting Nexin 14

TGF-βR2:

TGF-β receptor 2

DKK1:

Dickkopf 1

Rab 7:

Ras-related GTP binding protein 7

ESCs:

Embryonic stem cells

ECs:

Endothelial cells

Oxi-MSC-ApoEVs:

Oxygen stress-treated MSC-ApoEVs

TUFM:

Mitochondrial tu translation elongation factor

TFEB:

Transcription factor EB

Gal:

Galactose

NAc:

N-acetylgalactosamine

ASGPR:

Asialoglycoprotein receptor

FZD4:

Frizzled-4

TCs:

Theca cells

STAT3:

Signal transduction and activators of transcription 3

FOXO3a:

Forkhead box o3

SOD2:

Superoxide dismutase 2

DC-STAMP:

Dendritic cell-specific transmembrane protein

GelMA:

Gelatin methacrylate

GelNb:

Norbornene-modified gelatin

GelTR:

Tetrazine-modified gelatin

hBMMSCs:

Human bone marrow mesenchymal stem cells

WJMSCs:

Wharton’ Jelly mesenchymal stem cells

mBMMSCs:

Murine bone marrow mesenchymal stem cells

mADMSCs:

Murine adipose-derived mesenchymal stem cells

hDPSCs:

Human dental pulp stem cells

rBMMSCs:

Rabbit bone marrow mesenchymal stem cells

hADMSCs:

Human adipose-derived mesenchymal stem cells

iPSCs:

Induced pluripotent stem cells

i.v.:

Intravenous

i.p.:

Intraperitoneal

STS:

Staurosporine

UVC:

Ultraviolet C

Treg:

Regulatory T cells

BMD:

Bone mineral density

BV/TV:

Bone volume/total volume

References

  1. Lan T, Luo M, Wei X. Mesenchymal stem/stromal cells in cancer therapy. J Hematol Oncol. 2021;14(1):195.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Zhu Y, et al. Application of mesenchymal stem cell therapy for aging frailty: from mechanisms to therapeutics. Theranostics. 2021;11(12):5675–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Velarde F, et al. Mesenchymal stem cell-mediated transfer of mitochondria: mechanisms and functional impact. Cell Mol Life Sci. 2022;79(3):177.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Abdolmohammadi K, et al. Mesenchymal stem cell-based therapy as a new therapeutic approach for acute inflammation. Life Sci. 2023;312:121206.

    Article  CAS  PubMed  Google Scholar 

  5. Xu H, et al. Mesenchymal stem cells-based cell-free Therapy Targeting Neuroinflammation. Aging Dis. 2024;15(3):965–76.

    PubMed  PubMed Central  Google Scholar 

  6. Huang Y, Wu Q, Tam PKH. Immunomodulatory mechanisms of mesenchymal stem cells and their potential clinical applications. Int J Mol Sci, 2022. 23(17).

  7. Margiana R, et al. Clinical application of mesenchymal stem cell in regenerative medicine: a narrative review. Stem Cell Res Ther. 2022;13(1):366.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Copp G, Robb KP, Viswanathan S. Culture-expanded mesenchymal stromal cell therapy: does it work in knee osteoarthritis? A pathway to clinical success. Cell Mol Immunol. 2023;20(6):626–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Hoang DM, et al. Stem cell-based therapy for human diseases. Signal Transduct Target Ther. 2022;7(1):272.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Lin R, Zhang T, Gao J. Apoptotic vesicles of MSCs: the natural therapeutic agents and bio-vehicles for Targeting Drug Delivery. Small. 2023;19(47):e2301671.

    Article  PubMed  Google Scholar 

  11. Ding JY, et al. Mesenchymal stem cell-derived extracellular vesicles in skin wound healing: roles, opportunities and challenges. Mil Med Res. 2023;10(1):36.

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Malekpour K, et al. Mechanisms behind therapeutic potentials of mesenchymal stem cell mitochondria transfer/delivery. J Control Release. 2023;354:755–69.

    Article  CAS  PubMed  Google Scholar 

  13. Cunningham CJ, Redondo-Castro E, Allan SM. The therapeutic potential of the mesenchymal stem cell secretome in ischaemic stroke. J Cereb Blood Flow Metab. 2018;38(8):1276–92.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Chang C, et al. Effects of mesenchymal stem cell-derived paracrine signals and their delivery strategies. Adv Healthc Mater. 2021;10(7):e2001689.

    Article  PubMed  Google Scholar 

  15. Yagi H, et al. Mesenchymal stem cells: mechanisms of immunomodulation and homing. Cell Transpl. 2010;19(6):667–79.

    Article  Google Scholar 

  16. Preda MB, et al. Short lifespan of syngeneic transplanted MSC is a consequence of in vivo apoptosis and immune cell recruitment in mice. Cell Death Dis. 2021;12(6):566.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Thum T, et al. The dying stem cell hypothesis: immune modulation as a novel mechanism for progenitor cell therapy in cardiac muscle. J Am Coll Cardiol. 2005;46(10):1799–802.

    Article  CAS  PubMed  Google Scholar 

  18. Giacomini C, et al. The critical role of apoptosis in mesenchymal stromal cell therapeutics and implications in homeostasis and normal tissue repair. Cell Mol Immunol. 2023;20(6):570–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Romecín PA, et al. Robust in Vitro and in vivo immunosuppressive and anti-inflammatory properties of Inducible Caspase-9-mediated apoptotic mesenchymal Stromal/Stem cell. Stem Cells Transl Med. 2022;11(1):88–96.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Sanchez-Diaz M et al. Biodistribution of Mesenchymal Stromal Cells after administration in animal models and humans: a systematic review. J Clin Med, 2021. 10(13).

  21. Wen J, et al. Comparison of immunotherapy mediated by apoptotic bodies, microvesicles and exosomes: apoptotic bodies’ unique anti-inflammatory potential. J Transl Med. 2023;21(1):478.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Zhu Y, Chen X, Liao Y. Mesenchymal stem cells-derived apoptotic extracellular vesicles (ApoEVs): mechanism and application in tissue regeneration. Stem Cells. 2023;41(9):837–49.

    Article  PubMed  Google Scholar 

  23. Zhou M, et al. Apoptotic bodies for advanced drug delivery and therapy. J Control Release. 2022;351:394–406.

    Article  CAS  PubMed  Google Scholar 

  24. Harrell CR, Volarevic V. Apoptosis: a friend or foe in mesenchymal stem cell-based immunosuppression. Adv Protein Chem Struct Biol. 2021;126:39–62.

    Article  CAS  PubMed  Google Scholar 

  25. Kabat M, et al. Trends in mesenchymal stem cell clinical trials 2004–2018: is efficacy optimal in a narrow dose range? Stem Cells Transl Med. 2020;9(1):17–27.

    Article  CAS  PubMed  Google Scholar 

  26. Leibacher J, et al. Human mesenchymal stromal cells undergo apoptosis and fragmentation after intravenous application in immune-competent mice. Cytotherapy. 2017;19(1):61–74.

    Article  CAS  PubMed  Google Scholar 

  27. Zheng D, et al. Secondary lymphoid organs in Mesenchymal Stromal Cell Therapy: more than just a Filter. Front Immunol. 2022;13:892443.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Eggenhofer E, et al. Mesenchymal stem cells are short-lived and do not migrate beyond the lungs after intravenous infusion. Front Immunol. 2012;3:297.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Pang SHM et al. Mesenchymal stromal cell apoptosis is required for their therapeutic function. Nat Commun, 2021. 12(1).

  30. de Witte SFH, et al. Immunomodulation by therapeutic mesenchymal stromal cells (MSC) is triggered through phagocytosis of MSC by Monocytic cells. Stem Cells. 2018;36(4):602–15.

    Article  PubMed  Google Scholar 

  31. Wu HW et al. The Effects of Programmed Cell Death of Mesenchymal Stem Cells on the Development of Liver Fibrosis. Stem Cells Int, 2023. 2023: p. 4586398.

  32. Galleu A et al. Apoptosis in mesenchymal stromal cells induces in vivo recipient-mediated immunomodulation. Sci Transl Med, 2017. 9(416).

  33. He X, et al. Spontaneous apoptosis of cells in therapeutic stem cell preparation exert immunomodulatory effects through release of phosphatidylserine. Signal Transduct Target Ther. 2021;6(1):270.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Liu H, et al. Donor MSCs release apoptotic bodies to improve myocardial infarction via autophagy regulation in recipient cells. Autophagy. 2020;16(12):2140–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Liu S, et al. Mesenchymal stem cells prevent hypertrophic scar formation via inflammatory regulation when undergoing apoptosis. J Invest Dermatol. 2014;134(10):2648–57.

    Article  CAS  PubMed  Google Scholar 

  36. Kholodenko IV, et al. Apoptotic MSCs and MSC-Derived apoptotic bodies as New Therapeutic Tools. Curr Issues Mol Biol. 2022;44(11):5153–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Cheung TS, et al. Apoptosis in mesenchymal stromal cells activates an immunosuppressive secretome predicting clinical response in Crohn’s disease. Mol Ther. 2023;31(12):3531–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Li X, et al. Mesenchymal stem cell-derived apoptotic bodies alleviate alveolar bone destruction by regulating osteoclast differentiation and function. Int J Oral Sci. 2023;15(1):51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Li X, et al. IFNγ and TNFα synergistically induce apoptosis of mesenchymal stem/stromal cells via the induction of nitric oxide. Stem Cell Res Ther. 2019;10(1):18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Geng YJ. Molecular mechanisms for cardiovascular stem cell apoptosis and growth in the hearts with atherosclerotic coronary disease and ischemic heart failure. Ann N Y Acad Sci. 2003;1010:687–97.

    Article  CAS  PubMed  Google Scholar 

  41. Vincent JL. Current sepsis therapeutics. EBioMedicine. 2022;86:104318.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Zhang YY, Ning BT. Signaling pathways and intervention therapies in sepsis. Signal Transduct Target Ther. 2021;6(1):407.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Chang CL, et al. Impact of apoptotic adipose-derived mesenchymal stem cells on attenuating organ damage and reducing mortality in rat sepsis syndrome induced by cecal puncture and ligation. J Transl Med. 2012;10:244.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Sung PH, et al. Apoptotic adipose-derived mesenchymal stem cell therapy protects against lung and kidney injury in sepsis syndrome caused by cecal ligation puncture in rats. Stem Cell Res Ther. 2013;4(6):155.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Liu FB, Lin Q, Liu ZW. A study on the role of apoptotic human umbilical cord mesenchymal stem cells in bleomycin-induced acute lung injury in rat models. Eur Rev Med Pharmacol Sci. 2016;20(5):969–82.

    PubMed  Google Scholar 

  46. Chitimus DM et al. Melatonin’s Impact on Antioxidative and Anti-Inflammatory Reprogramming in Homeostasis and Disease. Biomolecules, 2020. 10(9).

  47. Yip HK, et al. Melatonin treatment improves adipose-derived mesenchymal stem cell therapy for acute lung ischemia-reperfusion injury. J Pineal Res. 2013;54(2):207–21.

    Article  CAS  PubMed  Google Scholar 

  48. van Niel G, et al. Challenges and directions in studying cell-cell communication by extracellular vesicles. Nat Rev Mol Cell Biol. 2022;23(5):369–82.

    Article  PubMed  Google Scholar 

  49. Poupardin R, et al. Advances in Extracellular Vesicle Research over the past Decade: source and isolation method are connected with Cargo and function. Adv Healthc Mater. 2024;13(19):e2303941.

    Article  PubMed  Google Scholar 

  50. Rädler J, et al. Exploiting the biogenesis of extracellular vesicles for bioengineering and therapeutic cargo loading. Mol Ther. 2023;31(5):1231–50.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Dixson AC, et al. Context-specific regulation of extracellular vesicle biogenesis and cargo selection. Nat Rev Mol Cell Biol. 2023;24(7):454–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Zou X, et al. Advances in biological functions and applications of apoptotic vesicles. Cell Commun Signal. 2023;21(1):260.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Das S, Lyon CJ, Hu T. A Panorama of Extracellular Vesicle Applications: from biomarker detection to therapeutics. ACS Nano. 2024;18(14):9784–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Kumar MA, et al. Extracellular vesicles as tools and targets in therapy for diseases. Signal Transduct Target Ther. 2024;9(1):27.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Atkin-Smith GK, Poon IKH. Disassembly of the dying: mechanisms and functions. Trends Cell Biol. 2017;27(2):151–62.

    Article  CAS  PubMed  Google Scholar 

  56. Santavanond JP, et al. Apoptotic bodies: mechanism of formation, isolation and functional relevance. Subcell Biochem. 2021;97:61–88.

    Article  CAS  PubMed  Google Scholar 

  57. Shi B, Phan TK, Poon IKH. Extracellular vesicles from the dead: the final message. Trends Cell Biol; 2024.

  58. Bedoui S, Herold MJ, Strasser A. Emerging connectivity of programmed cell death pathways and its physiological implications. Nat Rev Mol Cell Biol. 2020;21(11):678–95.

    Article  CAS  PubMed  Google Scholar 

  59. Ainola M, et al. Activation of plasmacytoid dendritic cells by apoptotic particles - mechanism for the loss of immunological tolerance in Sjögren’s syndrome. Clin Exp Immunol. 2018;191(3):301–10.

    Article  CAS  PubMed  Google Scholar 

  60. Park SJ, et al. Molecular mechanisms of biogenesis of apoptotic exosome-like vesicles and their roles as damage-associated molecular patterns. Proc Natl Acad Sci U S A. 2018;115(50):E11721–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Hur J, et al. Role of gasdermin E in the Biogenesis of apoptotic cell-derived exosomes. J Immunol. 2023;210(12):1974–89.

    Article  CAS  PubMed  Google Scholar 

  62. Beillevaire D, et al. Autolysosomes and caspase-3 control the biogenesis and release of immunogenic apoptotic exosomes. Cell Death Dis. 2022;13(2):145.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Yu L, et al. Apoptotic bodies: bioactive treasure left behind by the dying cells with robust diagnostic and therapeutic application potentials. J Nanobiotechnol. 2023;21(1):218.

    Article  Google Scholar 

  64. Wang Y, et al. Umbilical cord mesenchymal stem cell-derived apoptotic extracellular vesicles ameliorate cutaneous wound healing in type 2 diabetic mice via macrophage pyroptosis inhibition. Stem Cell Res Ther. 2023;14(1):257.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Yang J, et al. ApoSEVs-Mediated modulation of versatile target cells promotes Diabetic Wound Healing: unveiling a Promising Strategy. Int J Nanomed. 2023;18:6955–77.

    Article  CAS  Google Scholar 

  66. Jiang T, et al. Apoptotic bodies inhibit inflammation by PDL1-PD1-mediated macrophage metabolic reprogramming. Cell Prolif. 2024;57(1):e13531.

    Article  CAS  PubMed  Google Scholar 

  67. Liu J, et al. Apoptotic bodies derived from mesenchymal stem cells promote cutaneous wound healing via regulating the functions of macrophages. Stem Cell Res Ther. 2020;11(1):507.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Ma L, et al. Apoptotic extracellular vesicles are metabolized regulators nurturing the skin and hair. Bioact Mater. 2023;19:626–41.

    CAS  PubMed  Google Scholar 

  69. Wang J, et al. Apoptotic extracellular vesicles ameliorate multiple myeloma by restoring Fas-mediated apoptosis. ACS Nano. 2021;15(9):14360–72.

    Article  CAS  PubMed  Google Scholar 

  70. Ye Q, et al. MSCs-derived apoptotic extracellular vesicles promote muscle regeneration by inducing Pannexin 1 channel-dependent creatine release by myoblasts. Int J Oral Sci. 2023;15(1):7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Zhang R et al. Apoptotic vesicles rescue impaired mesenchymal stem cells and their therapeutic capacity for osteoporosis by restoring miR-145a-5p deficiency. Res Sq, 2024.

  72. Huang Z, et al. Apoptotic vesicles are required to repair DNA damage and suppress premature cellular senescence. J Extracell Vesicles. 2024;13(4):e12428.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Fu Y, et al. Mesenchymal stem cell-derived apoptotic vesicles ameliorate impaired ovarian folliculogenesis in polycystic ovary syndrome and ovarian aging by targeting WNT signaling. Theranostics. 2024;14(8):3385–403.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Ou Q, et al. Electrostatic charge-mediated apoptotic vesicle Biodistribution attenuates Sepsis by switching Neutrophil NETosis to apoptosis. Small. 2022;18(20):e2200306.

    Article  PubMed  Google Scholar 

  75. Zhu Y, et al. Apoptotic vesicles regulate bone metabolism via the miR1324/SNX14/SMAD1/5 signaling Axis. Small. 2023;19(16):e2205813.

    Article  PubMed  Google Scholar 

  76. Wang J, et al. Immunomodulatory function of licensed human bone marrow mesenchymal stromal cell-derived apoptotic bodies. Int Immunopharmacol. 2023;125Pt A:p111096.

    Article  Google Scholar 

  77. Cao Z, et al. Encapsulation of Nano-Bortezomib in apoptotic stem cell-derived vesicles for the treatment of multiple myeloma. Small. 2023;19(40):e2301748.

    Article  PubMed  Google Scholar 

  78. Zhang X, et al. Proteomic analysis of MSC-derived apoptotic vesicles identifies Fas inheritance to ameliorate haemophilia a via activating platelet functions. J Extracell Vesicles. 2022;11(7):e12240.

    Article  PubMed  PubMed Central  Google Scholar 

  79. Ding Z, et al. Apoptotic extracellular vesicles derived from hypoxia-preconditioned mesenchymal stem cells within a modified gelatine hydrogel promote osteochondral regeneration by enhancing stem cell activity and regulating immunity. J Nanobiotechnol. 2024;22(1):74.

    Article  CAS  Google Scholar 

  80. Mao J, et al. Balancing macrophage polarization via stem cell-derived apoptotic bodies for diabetic wound healing. Med. 2024;5(2):148–e1688.

    Article  CAS  PubMed  Google Scholar 

  81. Li Z, et al. Apoptotic vesicles activate autophagy in recipient cells to induce angiogenesis and dental pulp regeneration. Mol Ther. 2022;30(10):3193–208.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Jing Y et al. Apoptotic vesicles modulate endothelial metabolism and ameliorate ischemic retinopathy via PD1/PDL1 Axis. Adv Healthc Mater, 2024: p. e2303527.

  83. Yang K et al. Apoptotic vesicles derived from Dental Pulp Stem cells promote bone formation through the ERK1/2 signaling pathway. Biomedicines, 2024. 12(4).

  84. Zhao Q, et al. Biogenerated Oxygen-Related Environmental stressed apoptotic vesicle targets endothelial cells. Adv Sci (Weinh). 2024;11(20):e2306555.

    Article  PubMed  Google Scholar 

  85. Huang X, et al. Apoptotic vesicles resist oxidative damage in noise-induced hearing loss through activation of FOXO3a-SOD2 pathway. Stem Cell Res Ther. 2023;14(1):88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Ma Q, et al. Osteoclast-derived apoptotic bodies show extended biological effects of parental cell in promoting bone defect healing. Theranostics. 2020;10(15):6825–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Yan K, et al. The Effect of Age on the regenerative potential of adipose stem-cell-derived apoptotic extracellular vesicles in rat skin Wound Healing. Int J Med Sci. 2024;21(8):1529–40.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Qu Y, et al. Apoptotic vesicles inherit SOX2 from pluripotent stem cells to accelerate wound healing by energizing mesenchymal stem cells. Acta Biomater. 2022;149:258–72.

    Article  CAS  PubMed  Google Scholar 

  89. Li M, et al. BMSC-Derived ApoEVs promote Craniofacial Bone Repair via ROS/JNK Signaling. J Dent Res. 2022;101(6):714–23.

    Article  CAS  PubMed  Google Scholar 

  90. Yang Y, Lee EH, Yang Z. Hypoxia-conditioned mesenchymal stem cells in tissue regeneration application. Tissue Eng Part B Rev. 2022;28(5):966–77.

    Article  CAS  PubMed  Google Scholar 

  91. Zheng C, et al. Apoptotic vesicles restore liver macrophage homeostasis to counteract type 2 diabetes. J Extracell Vesicles. 2021;10(7):e12109.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Wang R, et al. Apoptotic vesicles ameliorate lupus and arthritis via phosphatidylserine-mediated modulation of T cell receptor signaling. Bioact Mater. 2023;25:472–84.

    CAS  PubMed  Google Scholar 

  93. Liu D, et al. Circulating apoptotic bodies maintain mesenchymal stem cell homeostasis and ameliorate osteopenia via transferring multiple cellular factors. Cell Res. 2018;28(9):918–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Xin L, et al. In situ delivery of apoptotic bodies derived from mesenchymal stem cells via a hyaluronic acid hydrogel: a therapy for intrauterine adhesions. Bioact Mater. 2022;12:107–19.

    CAS  PubMed  Google Scholar 

  95. Xiang L, et al. Carbon dot-loaded apoptotic vesicles improve the liver Kupffer cell-mediated Antibacterial Effect to synergistically alleviate Sepsis. ACS Nano. 2024;18(26):16726–42.

    Article  CAS  PubMed  Google Scholar 

  96. You Y, et al. Tailored apoptotic vesicle delivery platform for inflammatory regulation and tissue repair to ameliorate ischemic stroke. ACS Nano. 2023;17(9):8646–62.

    Article  CAS  PubMed  Google Scholar 

  97. Zhang S, et al. Bortezomib-based consolidation or maintenance therapy for multiple myeloma: a meta-analysis. Blood Cancer J. 2020;10(3):33.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Scott K, et al. Bortezomib for the treatment of multiple myeloma. Cochrane Database Syst Rev. 2016;4(4):pCd010816.

    Google Scholar 

  99. Cengiz Seval G, Beksac M. The safety of bortezomib for the treatment of multiple myeloma. Expert Opin Drug Saf. 2018;17(9):953–62.

    Article  CAS  PubMed  Google Scholar 

  100. Gui L, et al. Bone-targeting peptide and RNF146 modified apoptotic extracellular vesicles alleviate osteoporosis. Int J Nanomed. 2024;19:471–88.

    Article  CAS  Google Scholar 

  101. Mehrotra P, Ravichandran KS. Drugging the efferocytosis process: concepts and opportunities. Nat Rev Drug Discov. 2022;21(8):601–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Dolšak A, Gobec S, Sova M. Indoleamine and tryptophan 2,3-dioxygenases as important future therapeutic targets. Pharmacol Ther. 2021;221:107746.

    Article  PubMed  Google Scholar 

  103. Zhou HC, et al. Lactic acid in macrophage polarization: the significant role in inflammation and cancer. Int Rev Immunol. 2022;41(1):4–18.

    Article  CAS  PubMed  Google Scholar 

  104. Snodgrass RG, et al. Efferocytosis potentiates the expression of arachidonate 15-lipoxygenase (ALOX15) in alternatively activated human macrophages through LXR activation. Cell Death Differ. 2021;28(4):1301–16.

    Article  CAS  PubMed  Google Scholar 

  105. Liang X, et al. Phosphatidylserine released from apoptotic cells in tumor induces M2-like macrophage polarization through the PSR-STAT3-JMJD3 axis. Cancer Commun (Lond). 2022;42(3):205–22.

    Article  PubMed  Google Scholar 

  106. Chen Y, et al. Rubicon promotes the M2 polarization of Kupffer cells via LC3-associated phagocytosis-mediated clearance to improve liver transplantation. Cell Immunol. 2022;378:104556.

    Article  CAS  PubMed  Google Scholar 

  107. Huang W, et al. Soluble components from mesenchymal stromal cell processing exert anti-inflammatory effects and facilitate ischemic muscle regeneration. Cytotherapy. 2023;25(4):375–86.

    Article  CAS  PubMed  Google Scholar 

  108. Ye Q, et al. Apoptotic extracellular vesicles alleviate Pg-LPS induced inflammatory responses of macrophages via AMPK/SIRT1/NF-kappaB pathway and inhibit osteoclast formation. J Periodontol. 2022;93(11):1738–51.

    Article  CAS  PubMed  Google Scholar 

  109. Li J, et al. Apoptotic bodies extracted from adipose mesenchymal stem cells carry microRNA-21-5p to induce M2 polarization of macrophages and augment skin wound healing by targeting KLF6. Burns. 2022;48(8):1893–908.

    Article  PubMed  Google Scholar 

  110. Brunet A, Goodell MA, Rando TA. Ageing and rejuvenation of tissue stem cells and their niches. Nat Rev Mol Cell Biol. 2023;24(1):45–62.

    Article  CAS  PubMed  Google Scholar 

  111. Rajagopalan K et al. Understanding the molecular mechanism of regeneration through apoptosis-induced compensatory proliferation studies - updates and future aspects. Apoptosis, 2024.

  112. Boland K, Flanagan L, Prehn JH. Paracrine control of tissue regeneration and cell proliferation by Caspase-3. Cell Death Dis. 2013;4(7):e725.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Brock CK, et al. Stem cell proliferation is induced by apoptotic bodies from dying cells during epithelial tissue maintenance. Nat Commun. 2019;10(1):1044.

    Article  PubMed  PubMed Central  Google Scholar 

  114. Essien SA, Ahuja I, Eisenhoffer GT. Macrophage Migration Inhibitory Factor on Apoptotic Extracellular Vesicles Regulates Compensatory Proliferation. bioRxiv, 2023.

  115. Farrell L, et al. Actin remodeling mediates ROS production and JNK activation to drive apoptosis-induced proliferation. PLoS Genet. 2022;18(12):e1010533.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Diwanji N, Bergmann A. An unexpected friend - ROS in apoptosis-induced compensatory proliferation: implications for regeneration and cancer. Semin Cell Dev Biol. 2018;80:74–82.

    Article  CAS  PubMed  Google Scholar 

  117. Cheng Y, et al. Tailored apoptotic vesicles promote bone regeneration by releasing the osteoinductive brake. Int J Oral Sci. 2024;16(1):31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Lei F, et al. Apoptotic vesicles rejuvenate mesenchymal stem cells via Rab7-mediated autolysosome formation and alleviate bone loss in aging mice. Nano Res. 2022;16(1):822–33.

    Article  Google Scholar 

  119. Li M, et al. Extracellular vesicles from apoptotic BMSCs ameliorate osteoporosis via transporting regenerative signals. Theranostics. 2024;14(9):3583–602.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Qu Y, et al. Apoptotic metabolites ameliorate bone aging phenotypes via TCOF1/FLVCR1-mediated mitochondrial homeostasis. J Nanobiotechnol. 2024;22(1):549.

    Article  CAS  Google Scholar 

  121. Shi Y, et al. Mesenchymal stem cell-derived extracellular vesicles: a new impetus of promoting angiogenesis in tissue regeneration. Cytotherapy. 2019;21(5):497–508.

    Article  CAS  PubMed  Google Scholar 

  122. Helou DG, et al. PD-1 pathway regulates ILC2 metabolism and PD-1 agonist treatment ameliorates airway hyperreactivity. Nat Commun. 2020;11(1):3998.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Akiyama K, et al. Mesenchymal-stem-cell-induced immunoregulation involves FAS-ligand-/FAS-mediated T cell apoptosis. Cell Stem Cell. 2012;10(5):544–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Atsuta I, et al. Mesenchymal stem cells inhibit multiple myeloma cells via the Fas/Fas ligand pathway. Stem Cell Res Ther. 2013;4(5):111.

    Article  PubMed  PubMed Central  Google Scholar 

  125. Sui B, et al. Apoptotic vesicular metabolism contributes to Organelle Assembly and safeguards Liver Homeostasis and Regeneration. Gastroenterology. 2024;167(2):343–56.

    Article  CAS  PubMed  Google Scholar 

  126. Noone C, et al. IFN-γ stimulated human umbilical-tissue-derived cells potently suppress NK activation and resist NK-mediated cytotoxicity in vitro. Stem Cells Dev. 2013;22(22):3003–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Hu C, Li L. Preconditioning influences mesenchymal stem cell properties in vitro and in vivo. J Cell Mol Med. 2018;22(3):1428–42.

    Article  PubMed  PubMed Central  Google Scholar 

  128. Porta C, et al. Interferons alpha and gamma induce p53-dependent and p53-independent apoptosis, respectively. Oncogene. 2005;24(4):605–15.

    Article  CAS  PubMed  Google Scholar 

  129. Wang Y, et al. Delivering antisense oligonucleotides across the blood-brain barrier by Tumor Cell-Derived Small apoptotic bodies. Adv Sci (Weinh). 2021;8(13):2004929.

    Article  CAS  PubMed  Google Scholar 

  130. Laing AG, et al. Immune modulation by apoptotic dental pulp stem cells in vivo. Immunotherapy. 2018;10(3):201–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Ou Q, et al. Apoptosis releases hydrogen sulfide to inhibit Th17 cell differentiation. Cell Metab. 2024;36(1):78–e895.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors declare that we have not use AI-generated work in this manuscript.

Funding

This study was supported by the Wuhan University of Science and Technology startup fund (Chu Tian Scholars Program) (grant number X22020024), the open laboratory fund of Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration (grant number 2022kqhm005), the Hubei Provincial Health and Health Commission Research Project (grant number WJ2023M121), and the National Natural Science Foundation of China (grant number U22A20314).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualized and supervised the work: RXW wrote the manuscript and revised the content. JF revised the content and draw the images (created with BioRender.com). HJH, XJL, XXW, JMY modified the language. QSY, and YH provided constructive comments on the review. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Qingsong Ye or Yan He.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, R., Fu, J., He, J. et al. Apoptotic mesenchymal stem cells and their secreted apoptotic extracellular vesicles: therapeutic applications and mechanisms. Stem Cell Res Ther 16, 78 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13287-025-04211-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13287-025-04211-x

Keywords