Stem cell exosomes: new hope and future potential for relieving liver fibrosis

Article information

Clin Mol Hepatol. 2025;31(2):333-349
Publication date (electronic) : 2024 November 7
doi : https://doi.org/10.3350/cmh.2024.0854
11 Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang Province, P. R. China
2Department of Cell biology, School of Medicine, Taizhou University, Taizhou, Zhejiang Province, P. R. China
3Department of Pathophysiology, School of Basic Medicine, Shenyang Medical College, Shenyang, Liaoning Province, P. R. China
Corresponding author : Chong Jin Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, #999, Donghai Avenue, Jiaojiang District, Taizhou City, Zhejiang Province, 318001, P. R. China Tel: +8613757689065, Fax: +86-0576-88664768, E-mail: 13757689065@163.com
Editor: Norifumi Kawada, Osaka City University, Japan
Received 2024 September 29; Revised 2024 October 30; Accepted 2024 November 5.

Abstract

Liver fibrosis is a chronic liver injury resulting from factors like viral hepatitis, autoimmune hepatitis, non-alcoholic steatohepatitis, fatty liver disease, and cholestatic liver disease. Liver transplantation is currently the gold standard for treating severe liver diseases. However, it is limited by a shortage of donor organs and the necessity for lifelong immunosuppressive therapy. Mesenchymal stem cells (MSCs) can differentiate into various liver cells and enhance liver function when transplanted into patients due to their differentiation and proliferation capabilities. Therefore, it can be used as an alternative therapy for treating liver diseases, especially for liver cirrhosis, liver failure, and liver transplant complications. However, due to the potential tumorigenic effects of MSCs, researchers are exploring a new approach to treating liver fibrosis using extracellular vesicles (exosomes) secreted by stem cells. Many studies show that exosomes released by stem cells can promote liver injury repair through various pathways, contributing to the treatment of liver fibrosis. In this review, we focus on the molecular mechanisms by which stem cell exosomes affect liver fibrosis through different pathways and their potential therapeutic targets. Additionally, we discuss the advantages of exosome therapy over stem cell therapy and the possible future directions of exosome research, including the prospects for clinical applications and the challenges to be overcome.

INTRODUCTION

Hepatic fibrosis (HF) is a chronic liver disease attributed to numerous underlying causes, such as viral hepatitis, nonalcoholic steatohepatitis (NASH), nonalcoholic fatty liver disease, autoimmune hepatitis, and metabolic disorders [1]. In the progression of HF, as it develops from stage F0 to stage F4 (i.e., cirrhosis), the clinical symptoms become increasingly pronounced with the gradual deterioration of the condition [2]. HF development is characterized by pathologic changes, such as hepatocellular necrosis, inflammation, oxidative stress (OS), and extracellular matrix deposition, which may eventually lead to cirrhosis [3]. Chronic liver disease and cirrhosis cause 2 million deaths per year globally [4]. However, HF is reversible, considering it is not at an advanced cirrhotic stage. Liver transplantation is the most effective treatment for severe HF and cirrhosis; however, numerous patients await transplantation, and there are limited donors. An assessment of global liver transplantation statistics suggested a significant increase in transplant numbers since 2015 [5]. In 2021, 9,234 liver transplants were reported in the U.S. Moreover, the post-transplantation mortalities were 13.3%, 18.6%, and 35.9% for 3, 5, and 10 years, respectively [6].

Stem cell therapy has been recognized as a promising therapeutic strategy for HF. This can be attributed to the shortage of liver transplantation donors, complications persisting after transplantation, and relatively high mortality. Stem cells are one of the key hotspots in regenerative medicine because of their self-regenerative and multipotential differentiation abilities. Mesenchymal stem cells (MSCs) are extracted from the adipose tissue, bone marrow, and umbilical cord blood in treatments and studies by comparing the efficiency and immunocompatibility of stem cell extraction and considering the ethical issues [7]. MSCs substantially improve liver function when treating liver diseases [8]. Additionally, they develop into different cell lines exhibiting anti-inflammatory and immunomodulatory capabilities, minimizing the need for immunosuppressive drugs [9]. Stem cell therapy is widely applied to musculoskeletal and neurological disorders, immune disorders, hematologic dysfunctions, and degenerative diseases [7]. However, researchers have not identified effective countermeasures because stem cell transplantation may cause cellular rejection, infusion reactions, and medical tumor implantation [10]. Stem cells act in a paracrine manner through their soluble extracellular vehicles (EVs). Moreover, exosomes are considered the miniature versions of their parent cells [11]. Thus, mesenchymal stem cell-derived exosomes (MSC-Exos) inherit similar therapeutic effects, such as anti-inflammatory, immunomodulatory, and tissue regeneration, from their parental cells [12]. These physiological properties are unique to stem cells and serve as the advantages of exosomes over other cellular exosomes when applied to clinical therapies. Moreover, exosomes exhibit a superior safety profile than MSC-derived cells. This is because they do not replicate and can be easily stored without losing their properties [13]. MSC-Exos are a favorable alternative to stem cell therapy regarding its efficacy and circumvent the drawbacks associated with stem cell therapy.

ORIGIN AND BIOLOGICAL PROPERTIES OF MSC-EXOS

EVs are categorized into exosomes, microvesicles, and apoptotic vesicles based on their size and origin [14]. Exosomes are small vesicles ranging between 40 nm and 150 nm; they are secreted by the cells [15]. Additionally, exosomes are the most well-defined EVs abundant in body fluids, such as blood, saliva, amniotic fluid, urine, and breast milk [16]. They act as a cellular messenger, participating in cellular communication and metabolism [17]. The mode of action of exosomes includes content delivery to the cell membrane through receptor-ligand interactions, endocytosis, or direct fusion, facilitating the mediation and regulation of cellular functions in the target cells [18]. Exosomes encompass several biologically active molecules, including proteins, mRNAs, and non-coding RNAs, such as microRNAs, which can transfer functions across different cell types and even species (Fig. 1) [19]. In addition to directly promoting tissue and organ regeneration, EVs regulate the microenvironment, which is deleterious to tissue injury repair [20]. Exosomes perform important biological functions in an organism.

Figure 1.

The nucleic acids and proteins produced by the nucleus and organelles, as well as substances taken up through endocytosis, together form early endosomes, which are then expelled from the cell through exocytosis. There are various functional substances present in the exocrine body and it s surface, such as proteins, DNA, miRNA, etc.

MSC-Exos exhibit some similarities and differences in protein expression and function with cell-secreted exosomes. Similar to other exosome formation, MSC-Exos are produced through “endocytosis-fusion-exocytosis” [21]. Similarly, MSC-Exos express proteins associated with exosome formation, such as membrane-associated proteins (CD9, CD63, CD81, major histocompatibility complex type 1 [MHC-I], and MHC-II), heat shock proteins (HSPs) (Hsp8, Hsp70, and Hsp90), and multivesicular bodies (ALG-2-interacting protein X, tumor susceptibility gene 101, and synthetic proteins) [22]. By contrast, they express several stem cell-specific proteins, such as CD29, CD44, and CD73 [23]. In addition to differential protein expression, MSC-Exos encompass numerous substances related to their biological origins, such as lipid rafts, saturated fatty acids, and specific ncRNAs. These substances are closely related to the regulation of target cells [24]. Regarding preservation, isolated MSC-Exos should be processed or frozen more immediately than other exosomes. This is because the storage temperature exerts a strong influence on its structure [25]. Functionally, MSC-Exos are central to promoting hepatocyte proliferation and maintaining hepatocyte function; they possess regulatory properties that allow the transport of functional proteins or RNAs to the target cells through physiological barriers for communication and regulation [26].

The application of MSC-Exos to regenerative medicine appears to be promising for treating tissue wounds by mediating intercellular communication in skin regeneration. Thus, MSC-Exos are central to skin wound healing by reducing inflammatory responses, promoting proliferation, inhibiting apoptosis, and enhancing angiogenesis [27]. The proteins and miRNAs in exosomes regulate cellular metabolism in the liver, thereby altering the microenvironment in target cells. The mediated cell-cell communication alters tumor growth, cell migration, antiviral infections, and hepatocyte regeneration. Thus, the application of MSC-Exos as a diagnostic or therapeutic tool has excessive potential for liver diseases [28]. In summary, MSC-Exos have good prospects for clinical applications and offer an alternative to stem cell-based therapies. This article elucidates investigations on MSC-Exos in regulating HF through multiple pathways and trends for future research on exosome therapeutics.

MECHANISM OF ACTION UNDERLYING EXOSOMES IN HF TREATMENT

MSC-Exos modulate macrophage polarization to alleviate HF

HF development can be attributed to the activation of hepatic stellate cells (HSC). However, immune cells, particularly macrophages, affect HSC activation. Macrophages are highly plastic and can be functionally divided into two major subpopulations, namely, classically activated macrophages (CAM, M1-macrophages) and alternatively activated macrophages (AAM, M2-macrophages) [29]. In vitro, M1-macrophages are induced by lipopolysaccharides (LPS) and interferon-γ (IFN-γ); they exhibit pro-inflammatory activity. By contrast, M2 macrophages are induced by interleukin (IL)-4 and IL-13; they exhibit anti-inflammatory activity [30]. Macrophages and fibroblasts are central to all types of tissue fibrosis, which begins with the recruitment of numerous pro-fibrotic cytokines and chemokines [31]. For example, M1 macrophages secrete several pro-inflammatory cytokines, such as IL-1b, inducible nitric oxide synthase, and tumor necrosis factor-α (TNF-α) that contribute to fibrosis. Contrarily, M2-macrophages are anti-inflammatory macrophages, which primarily produce anti-inflammatory factors, such as IL-10, transforming growth factor β (TGF-β), and arginase 1. Moreover, they serve as fibrinolytic upon a reduction of the influences that cause inflammation [31].

MSC-derived extracellular vesicles (MSC-EVs) alleviate HF through multiple pathways, including the modulation of M1-macrophage polarization to M2-macrophages. Exosomes have been identified as effective paracrine mediators of the antifibrotic effects of MSCs. Researchers have proposed miR-148a as a potential therapeutic target for HF. Mechanistically, in exosomes, miR-148a targets KLF6 directly to regulate macrophage polarization via the Janus kinase/signal transducers and activators of the transcription pathway [32]. Moreover, MSC-EVs effectively switch the polarization state of macrophages from the M1 to the M2 phenotype to alleviate HF [32]. Additionally, MSC-Exos consist of IL-10, which acts as an anti-inflammatory agent in hepatic macrophages and reduces liver injury [33]. The intravenous transplantation of human umbilical cord MSCs (HucMSCs) into methionine- and choline-deficient (MCD) NASH mice improves the appearance of MCD-induced body weight loss and liver injury. This is because exosome-induced macrophages produce anti-inflammatory factors and reduce inflammatory cytokines in the liver tissues, thus alleviating HF and improving the body state in mice [34].

Apart from the direct application of MSC-Exos to the liver, stem cells can be pretreated using cytokines. Stem cells act as a “drug reservoir” for exosomes, yielding more therapeutic exosomes upon cytokine action. For example, HucMSC pretreatment with interleukin 6 significantly upregulates anti-inflammatory miR-455-3p levels in the secreted exosomes. Moreover, the exosomes are translocated to macrophages, inhibiting macrophage overactivation and exerting anti-inflammatory activity by targeting phosphoinositide-3-kinase regulatory subunit 1 [35]. Upon activation by inflammatory vesicle agonists, the trans-Golgi network (TGN) disassembles to form the dispersed trans-Golgi network (dTGN), and dTGN-enriched cells are recruited for a nucleotide-binding domain, leucine-rich–containing family, pyrin domain–containing-3 (NLRP3) [36]. After TNF-α pretreatment, HucMSCs release numerous exosomes enriched in miR-299-3p. Additionally, exosome translocation to macrophages prevents the disassembly of TGNs to form dTGNs, thereby inhibiting the recruitment and activation of NLRP3 inflammatory vesicles to exert anti-inflammatory effects [37]. In summary, regulating the polarization state and anti-inflammatory activity of macrophages using exosomes is central to preventing HF progression (Fig. 2).

Figure 2.

LPS and IFN-γ induce M1 macrophages, whereas IL-4 and IL-13 induce M2 macrophages. Exosomes containing specific miRNAs derived from stem cells can modulate the polarization of macrophages. This modulation enables pro-inflammatory M1 macrophages to transform into anti-inflammatory M2 macrophages.

MSC-Exos modulate autophagy to alleviate HF

Autophagy is the process of self-digestion and metabolism that maintains homeostasis in the internal environment. Autophagy involves the breakdown and reuse of organelles, proteins, and macromolecules in the cytoplasm. Moreover, cellular autophagy plays an important physiological role in recycling substances to adapt to metabolic stress [38]. Cellular autophagy plays a dual role in the liver. In the liver, HSC activation promotes HF and plays an anti-fibrotic role by reducing hepatocyte death or regulating macrophages [39]. Autophagy is critical for maintaining intrahepatic homeostasis in both parenchymal (hepatocytes) and nonparenchymal (HSCs), sinusoidal endothelial cells, and Kupffer cells (KCs) cells inside the liver [40]. Collectively, autophagy has been recognized as an anti-HF pathway in the liver because it contributes to liver homeostasis by removing misfolded proteins, damaged organelles, and lipid droplets, which are central to energy homeostasis and cytoplasmic content control [41]. Exosome formation and cellular autophagy share similar signaling pathways and molecular mechanisms; moreover, they demonstrate a substantial interaction [42]. In the liver, exosome formation and cellular autophagy tend to be inversely related. Cellular autophagy inhibits the sustained and enhanced release of EVs, for example, in alcoholic liver disease. Therefore, EV release from the hepatocytes increases, whereas hepatocyte autophagy is inhibited [43]. HSC stimulation using platelet-derived growth factors inhibits HSC autophagy and enhances EV release [44].

Interaction between the cellular production of exosomes and autophagy affects HF. thioacetamide stimulation elevates the stress protein Tribbles homolog 3 (TRIB3) and interacts with the selective autophagy receptor Sequestosome 1 (SQSTM1), causing SQSTM1 aggregate accumulation and impeding autophagy. TRIB3-mediated autophagy inhibition not only prevents autophagic degradation but also promotes hepatocyte secretion by exosomes enriched with inhibin subunit beta A/activin A. Eventually, it causes HSC migration, proliferation, and activation [45]. However, disrupted interaction between TRIB3-SQSTM1 and specific helical peptides restores autophagy in the hepatocytes and HSC, exerting a protective effect against HF [45]. Researchers have investigated the protective role of MSC-Exos in the liver against HF by modulating autophagy. Bone marrow mesenchymal stem cell-derived exosomes (BMSCExos) in hepatocytes increase the expression of autophagy marker proteins LC3 and Beclin-1 and autophagosome formation. Additionally, BMSC-Exos attenuate hepatocyte apoptosis by promoting autophagy [46]. Exosomes secreted by adipose-derived-MSCs (AD-MSCs) acted on a mouse model of carbon tetrachloride (CCl4)-induced HF. miR-181-5p in the exosomes reduced HSC activation by directly inhibiting the STAT3/B-cell lymphoma 2/Beclin1 pathway and increased autophagy, thereby reducing HF [47]. In summary, autophagy by MSC-Exo-targeted modulation is a feasible strategy for HF treatment (Fig. 3).

Figure 3.

The interaction between exosomes and autophagy significantly impacts the progression of liver fibrosis. (I) To summarize, autophagy occurs in both parenchymal and non-parenchymal cells of the liver. This process can help mitigate liver fibrosis. (II) When autophagy in hepatocytes is inhibited, the release of exosomes increases. (III) When platelet-derived growth factor activates hepatic stellate cells, blocking their autophagy leads to an increase in exosome release. (IV) AMSC-Exosomes and BMSC-Exosomes help prevent liver fibrosis by promoting the process of autophagy.

MSC-Exos regulate pro-inflammatory cytokine secretion to alleviate HF

Inflammatory cytokines are central to HF development. Numerous inflammatory cytokines, such as IL-6, TGF-β, TNF-α, and IL-1, promote HF [48]. They promote the abnormal proliferation and deposition of hepatic connective tissue, thereby exacerbating HF [49]. Therefore, clinicians should control the production and activation of inflammatory cytokines to prevent HF development and progression.

MSC-Exos regulate TGF-β secretion

The biological functions of TGF-β involve tissue repair and embryonic development [50]. TGF-β plays a regulatory role in cell proliferation, senescence, apoptosis, inflammatory response, tissue fibrosis, and aging [51]. It modulates senescence-associated fibrosis by regulating inflammation, DNA damage, OS, and cellular senescence [52]. These processes help maintain the dynamic homeostasis of tissues and organs. Researchers have identified >30 isoforms of TGF-β, a major pro-fibrotic cytokine; TGF-β1 is the most relevant isoform to HF [53]. It accelerates HF by promoting HSC activation through the TGF-β1/Smads signaling pathway [54]. Some findings appear promising for inhibiting TGF-β expression through MSC-Exos’ action. For example, Wharton’s jelly, attached to the umbilical cord, contains abundant MSCs. It can differentiate into adipocytes, osteoblasts, chondrocytes, neurons, and hepatocytes [55]. The mRNA expression levels of NOX1, NOX2, and NOX4 genes, which are associated with TGF-β, were significantly reduced in LX2 cells within 24 h of treatment using exosomes (40 µg/mL or 50 µg/mL) derived from Wharton’s jelly MSCs. Furthermore, TGF-β expression was significantly inhibited under similar conditions [56]. Therefore, MSC-Exos in liver cells will supposedly inhibit TGF-β expression, thus alleviating HF and becoming a novel treatment target.

Exosomes of different cellular origins affect TGF-β expression in HF. For example, umbilical cord-derived MSCs (UCMSCs) inhibit signaling pathways by suppressing TGF-β expression, inhibiting HSC activation, attenuating HF, and reducing protein expression related to cellular senescence and cell proliferation [57]. Exosomes secreted by AD-MSCs comprising overexpressed miRNA-181-5p inhibit TGF-β1-induced upregulation of pro-fibrotic genes, thereby attenuating hepatic injury. Additionally, they significantly downregulate the expression of type I collagen, α-smooth muscle actin, and fibronectin in hepatic tissues, thereby inhibiting HF [47]. In vitro, applying human embryonic MSC-Exos enriched with miR-6766-3p to human HSC (LX2) significantly reduces pro-fibrotic marker expression. miR-6766-3p inhibits TGFβRII expression to reduce Smads signaling, eventually reducing HSC activation and preventing HF onset or development [58]. MSC-Exo attenuates HF by inhibiting TGF-β expression. Additionally, exosomes produced by other cells exert similar effects. For example, in vivo and in vitro experiments have suggested that exosomes derived from natural killer (NK) cells inhibit TGF-β1-induced proliferation, CCl4-induced HF in mice, and the activation of human HSC lineage cells (Fig. 4) [59].

Figure 4.

Exosomes of different cellular origins can alleviate liver fibrosis via regulating TGF-β/Smad pathway.

Regulating IL-6 secretion by MSC-Exos

IL-6 is the predominant cytokine associated with inflammatory diseases [60]. As a key pro-inflammatory cytokine, it has numerous physiological functions, such as regulating cell proliferation and differentiation [61]. IL-6 exerts a bidirectional effect on HF, promoting inflammation production that causes hepatocyte necrosis and apoptosis and protecting the hepatocytes against injury at appropriate concentrations [62]. Therefore, IL-6 can be applied to liver lesion treatment by selectively inhibiting the IL-6 pathway and decreasing expression [63]. However, MSC-Exos can regulate various cytokines, such as IL-6, thereby intervening in HF onset and progression [64].

MSC-Exos regulate inflammatory cytokine secretion by modulating the degree of gene expression [65]. Adipose-derived mesenchymal MSC-Exos were administered to an LPS/D-Gal-induced acute liver failure mouse model. miR17 in exosomes reduced inflammatory marker expression, such as TNF-α, IL-6, IL-1β, and IFN-γ, by regulating the expression of genes related to inflammatory cytokines, thereby attenuating liver injury [66]. Exosomes isolated from UCMSCs, either infused alone or combined with poly sterols, inhibited mRNA expression of IL-6 in a mouse model of liver failure. Furthermore, the combination of UCMSC-derived exosomes with poly sterols is preferable to exosomes alone; both attenuate hepatic injury in HF mice by modulating the IL-6/STAT3 signaling pathway [67]. These pathways facilitate decreasing the secretion of inflammatory cytokines and liver injury and attenuating HF. MSC-Exos can prevent liver injury by regulating IL-6 and its signaling pathway. Moreover, upon applying IL-6 to HucMSCs, the resulting exosomes were enriched in miR-455-3p. This in turn effectively reduced serum inflammatory cytokine levels and attenuated liver injury [35].

In addition to TGF-β and IL-6, MSC-Exos regulate several cytokines, such as chemokines, IL-7, IL-1β, TNF-α, IFN-γ, and other inflammatory cytokines [68]. During HF progression, MSC-Exos interact with inflammatory cytokines and co-regulate a series of changes to suppress inflammation, improve hepatic function, promote hepatocyte regeneration, and alleviate HF in the liver.

MSC-Exos regulate OS to alleviate HF

OS refers to the excessive production of highly reactive molecules, such as reactive oxygen radicals (ROS) and reactive nitrogen radicals (RNS), in the body upon subjecting an organism to harmful stimuli [69]. In OS, oxidation exceeds oxidant scavenging, and the oxidative and antioxidant systems are out of balance, leading to tissue damage [70]. The liver is an important site for ROS production and is continuously exposed to different toxicants or reactive metabolites (both ROS and RNS) [71]. ROS accumulation in the liver induces hepatocyte dysfunction or death, which eventually leads to the release of injury-associated molecules. Simultaneously, nonparenchymal cells, such as KCs, HSCs, and newly recruited immune cells, are activated and produce pro-fibrotic and pro-inflammatory mediators [72]. Therefore, OS is one of the chief causative factors of liver diseases and can lead to common liver diseases [73]. MSC-derived exosomes are rich in bioactive molecules with numerous bioregulatory functions that suggestively improve the efficacy of treating OS-related diseases [74].

MSC-Exos exert stronger anti-OS effects in the liver and, thus, prevent liver injury in HF treatment. To verify the definite effects of MSC-Exos, researchers have compared the antioxidant effects of human umbilical cord blood MSC-Exo (HucMSC-Exo) with that of bifendate (DDB), a commonly used hepatoprotective agent, in a CCL4-induced liver injury mouse model. HucMSC-Exo exerted more significant antioxidant and hepatoprotective effects, compared with DDB [75]. Moreover, HucMSC-Exo exerted antioxidant and anti-apoptotic effects upon injection into a mouse model of hepatic injury by the tail vein or orally. Additionally, it promoted the recovery of hepatic oxidative damage and prevented hepatic failure by delivering glutathione peroxidase 1 [76]. Oxygen transport and iron homeostasis are closely related [77]. Thus, abundant iron in the cells and tissues disrupts redox homeostasis and catalyzes ROS production, leading to OS. Eventually, it interacts with iron death at the cellular level and is considered an iron-dependent form of cell death [78]. Benzyl chloride 1 (BECN1) is a key regulator of ferroptosis [79]. An intravenous injection of MSC-ex significantly reduced HSC activation and collagen deposition in the fibrotic livers of experimental mice. Mechanistically, BECN1 delivered via MSC-Exos promoted iron death and OS in HSCs, thereby preventing HF [80].

Additionally, the exosomes of cells across different species play a biological role. Similar to exosomes secreted by mammalian cells, edible plant-derived exosome-like nanoparticles serve as extracellular messengers to mediate intercellular communication and regulate interspecies communication through their products (e.g., miRNAs, biologically active lipids, and proteins) [81]. Researchers have screened blueberry-derived exosomes-like nanoparticles; they attenuated rotenone-induced OS in human HepG2 cells and high-fat diet-fed C57BL/6 mice to mitigate liver injury [82]. Exosomes obtained from multiple sources may protect from liver injury by regulating OS as well as HF.

COMPARING THE ADVANTAGES OF EXOSOME AND STEM CELL THERAPIES

Clinical trials have elucidated issues with the safety and stability of BMSCs that affect clinical treatment progression, clinical staging, and determining infusion volumes upon using BMSCs for the treatment of chronic liver disease, including HF and cirrhosis [83]. Several researchers have investigated the safety of stem cell therapy, which primarily focuses on topical treatments, such as ocular administration [84]. No substantial abnormality has been identified in its local administration, and the therapeutic effect has been satisfactory. However, the safety of systemic MSC injection in humans remains unclear. In a few experiments on stem cell infusion therapy, substantial MSCs accumulated in the lungs of humans and animals upon infusion into the body; most MSCs were eliminated within the subsequent 3 to 6 days. Moreover, <1% of MSCs were distributed in various organs, including the spleen, liver, and bone marrow, after 1 week [85]. Thus, stem cells do not remain stable in the body for a long duration after infusion because of rapid clearance. Moreover, stem cell therapy has drawbacks and negative effects. For example, UCMSC therapy reduces pro-inflammatory cytokine production, such as TNF-α, IFN-γ, IL-6, IL-1β, TGF-β1, and IL-4. By contrast, it increases anti-inflammatory cytokine production, such as IL-10 [86]. Furthermore, it poses the risk of medically derived tumors, such as post-transplant lymphoproliferative disease. This transplantation complication is the leading cause of cancer-related deaths after solid organ or allogeneic hematopoietic stem cell transplantation [87]. In summary, despite several examples of MSC application in clinical studies, maintaining constant and effective quantity and quality control is challenging.

Using exosomes for disease treatment can effectively avoid some issues associated with stem cell therapies and achieve good therapeutic effects. For example, in a study involving 40 patients with chronic kidney disease, 20 were randomly selected to receive stem cell exosome treatment, while the others received a placebo. After a one-year monitoring period, it was found that members of the treatment group showed significant improvements in kidney function and inflammatory status compared to the control group [88]. For acute or chronic graft-versus-host disease (GvHD) caused by stem cell transplantation, researchers used exosomes derived from the supernatant of BM-MSCs to treat GvHD patients, resulting in significant improvement in symptoms; the amount of diarrhea was significantly reduced, and skin and mucosal damage showed marked improvement [89]. At the same time, to observe whether exosome therapy has adverse reactions, in a clinical trial, 24 healthy volunteers inhaled ASC-Exos via nebulization, followed by multiple biological examinations such as blood parameters, liver and kidney function, lactate dehydrogenase, immunoglobulin concentration, and electrocardiograms. All volunteers showed good tolerance to the infusion, and no adverse reactions occurred [90]. Furthermore, as of now, there are still several clinical trials regarding exosomes that are in the monitoring phase, and their application potential is gradually being confirmed (Table 1) [91].

Clinical applications of exosomes

The specific advantages of exosome therapy are as follows: Exosomes are physiologically stabilized by a double-layer plasma membrane. Moreover, the component substances are protected from the action of immune cells and digestive enzymes, resulting in low immunogenicity [92]. Exosomes deliver different proteins to the target cells and efficiently deliver numerous growth factors and those that regulate cellular responses to the liver [8]. Exosome targeting can achieve good therapeutic effects at smaller drug concentrations, displaying favorable application value [93]. Additionally, the use of human-derived MSCs encounters ethical issues in some countries, despite no ethical issues with UCMSCs. This is because the umbilical cord is considered biological waste and is discarded after birth [94]. However, these concerns are related to considering MSCs from embryos. Nonetheless, exosomes from stem cells instead of cells pose fewer ethical challenges [95]. Thus, MSC-Exos treatment exerts a more favorable therapeutic effect. Exosome therapy is safer than stem cell therapy because of non-ethical constraints, greater therapeutic stability, and the absence of defects inherent to stem cell therapy. Therefore, it serves as an effective alternative to stem cell therapy.

CHALLENGES AND PROSPECTS FOR EXOSOME THERAPY

Exosomes have been developed as drug delivery vehicles or extensively studied as biomarkers. Particularly, MSC-EVs have been used in several clinical studies. However, researchers should further investigate the long-term effects of MSC-EVs. For example, differences in the therapeutic effects of MSC-secreted exosomes from different sources should be evaluated for immunomodulation, antifibrosis, and liver regeneration potential to determine the optimal therapeutic strategy [98]. Because exosomes are secreted by cells, pretreatment before exosome collection may generate a product with better therapeutic efficacy. In one study, ASCs secreted exosomes with specific functions by overexpressing miRNA-181-5p; the resulting exosomes could be targeted for transport into the fibrotic liver [47]. Thus, exosomes can be used to deliver potent molecules released by MSC, whereas those secreted by genetically altered stem cells can be targeted for delivery [99]. EVs are cellular products involved in almost all physiological activities, including cell proliferation, migration, and apoptosis. They are closely associated with immune surveillance and immune cell activation [100]. Thus, exosomes can regulate disease progression. Researchers are exploring the role of exosomes in diagnosing liver disease. Exosome detection may serve as a novel diagnostic indicator for liver disease by facilitating an understanding of liver pathology [101]. Moreover, MSC-derived exosomes exert anti-inflammatory, immunomodulatory, and therapeutic effects [102]. As a novel antigenic material, MSC displays good immune properties; therefore, MSC-Exos can be applied to vaccine development [103]. MSCs secrete EVs in large quantities, which facilitate exploring MSC-Exos as a novel vaccine vector [104]. Conventional liposome-delivered vaccines can cause adverse immunogenic reactions. By contrast, EV vectors effectively avoid such reactions [105,106].

Research on the clinical potential of exosomes is underway. For example, no unified standard has been proposed for exosome preparation. Moreover, researchers have developed separation methods, including ultracentrifugation, density gradient centrifugation, polymer precipitation, ultrafiltration, size-exclusion chromatography, and immunoaffinity methods, based on the exosome size, density, and immunophenotype. Nonetheless, the yield and physicochemical properties of separated exosomes depend on the isolation method and cannot be identical [107]. For example, ultracentrifugation is a common method that produces exosomes with high purity but low yield. At the same time, when developing exosome-based therapies, it is crucial to ensure that processes adhering to good manufacturing practices and strict quality control measures are employed to produce exosome preparations that meet clinical standards [108]. Strict quality control is also crucial for reproducible research in academic studies [109]. Recently, the International Society for Extracellular Vesicles (ISEV) has established a set of standards and guidelines for extracellular vesicle research, aimed at providing a standardized framework for the clinical application and scientific research of extracellular vesicles [110]. Thus, MSC-EV separation methods must be standardized in the future [111]. Researchers should improve and standardize established separation protocols to increase the yield and purity of the samples. BMSC-Exos treatment is considered a “double-edged sword” [112]. Despite the therapeutic effects, the safety, adverse effects, and antagonistic outcomes of exosome treatment are unclear [113]. Therefore, MSC-EVs should be applied cautiously in the future because the role of MSC-EVs in disease development has not been elucidated completely. In summary, exosomes are promising for vaccine development, disease diagnosis, and treatment; however, clinicians should consider their immunologic properties. In the future, researchers should explore the application value of exosomes from different cellular sources or those secreted after cellular pretreatment as well as discover more efficient and stable exosome isolation methods to obtain the ideal purity and yield.

SUMMARY

In summary, effective treatments for HF caused by chronic liver injury because of multiple etiologies remain limited. However, HF treatment using MSC-Exos appears as a novel approach with promising clinical applications. EVs secreted by stem cells mediate intercellular communication and contribute to HF onset and progression. HF treatment using MSC-Exo can repair the liver tissue by polarizing M1-type macrophages into M2-type macrophages, which exert their anti-inflammatory effects. Additionally, MSC-Exos reduce the secretion of inflammatory cytokines, such as IL-6, TGF-β, and TNF-α, and inhibit HSC activation by regulating HSC autophagy, thus preventing HF. MSC-Exos can alleviate HF by regulating OS. In summary, MSC-Exos plays a regulatory role in alleviating HF. However, research on the role of MSC-Exos in HF is underway. This warrants additional studies and in-depth investigations to elucidate their therapeutic effects against HF and the complex mechanisms underlying exosome-mediated therapy for HF.

Existing studies have not elucidated the adverse effects of MSC-Exo completely. Thus, we intend to explore their therapeutic effects and adverse effects on HF treatment in the future. Exosome preparation involves high cost and low yield, and the preparation process is not unified. Thus, we intend to study exosome preparations from different species or cell sources. We aim to identify exosome preparations that are abundant, simple, cost-effective, and stable. This study may be important for developing novel therapeutic strategies for HF. Additionally, regulating stem cells to secrete exosomes with targeted therapeutic functions by targeting stem cell genes to produce exosomes with specific functions may offer a novel approach to developing exosome agents. Exosome therapy, an emerging research topic, holds promise for exerting therapeutic effects similar to stem cell therapy while overcoming the associated limitations and adverse effects. In the future, an in-depth exploration of its characteristics can provide a good reference for exosome development.

Exosomes are expected to serve as carriers for delivering drugs. The application of exosomes in vaccine research and development may prevent immunogenic reactions. In the future, we aim to explore the feasibility of their application to vaccine formulations and investigate the immunological properties of exosomes as antigens, which will contribute to developing novel carrier vaccines.

Despite the therapeutic potential of MSC-Exos for HF, further advancements are required for a feasible clinical application. Exploring the specific mechanisms by which exosomes treat HF will provide valuable insights for developing novel therapeutic strategies. Researchers should conduct clinical trials using multiple sources of stem cells and exosomes, providing a theoretical basis for the continued advancement of exosomes in treating liver disease and, eventually, improving patient prognosis. In conclusion, exploring the role of MSC-Exos in HF has substantial clinical value.

Notes

Authors’ contribution

LHL: conceptualization & supervision & writing-review and editing; YJL: conceptualization & writing-original draft; KPW: conceptualization & writing-original draft; JGM: conceptualization & writing-review and editing; ZYW: Draw Figures & writing-review; HJ: conceptualization & writingoriginal draft; CJ: conceptualization & supervision & writing-review and editing. All authors read and approved the final manuscript.

Acknowledgements

This study was supported by Zhejiang Provincial Natural Science Foundation of China [No. LY23H030002], Medical Science and Technology Project of Zhejiang Province [No. 2023KY412] and Liver Cancer Early Warning and Early Intervention Technology Innovation Team of Taizhou Central Hospital (Taizhou University Hospital).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AD-MSCs

adipose-derived-MSCs

BECN1

Benzyl chloride 1

dTGN

dispersed trans-Golgi network

EVs

extracellular vehicles

GvHD

graft-versus-host disease

HF

hepatic fibrosis

HSC

hepatic stellate cells

HSPs

heat shock proteins

HucMSCs

human umbilical cord MSCs

IFN-γ

interferon-γ

IL

interleukin

KCs

Kupffer cells

LPS

lipopolysaccharides

MHC-I

major histocompatibility complex type 1

MSC-EVs

MSC-derived extracellular vesicles

MSC-Exos

mesenchymal stem cell-derived exosomes

MSCs

Mesenchymal stem cells

NASH

nonalcoholic steatohepatitis

NK

natural killer

NLRP3

nucleotide-binding domain

OS

oxidative stress

RNS

reactive nitrogen radicals

ROS

reactive oxygen radicals

SQSTM1

Sequestosome 1

TGF-β

transforming growth factor β

TGN

trans-Golgi network

TNF-α

tumor necrosis factor-α

TRIB3

Tribbles homolog 3

UCMSCs

umbilical cord-derived MSCs

References

1. Hernandez-Gea V, Friedman SL. Pathogenesis of liver fibrosis. Annu Rev Pathol 2011;6:425–456.
2. Poisson J, Lemoinne S, Boulanger C, Durand F, Moreau R, Valla D, et al. Liver sinusoidal endothelial cells: physiology and role in liver diseases. J Hepatol 2017;66:212–227.
3. Bataller R, Brenner DA. Liver fibrosis. J Clin Invest 2005;115:209–218.
4. Moon AM, Singal AG, Tapper EB. Contemporary epidemiology of chronic liver disease and cirrhosis. Clin Gastroenterol Hepatol 2020;18:2650–2666.
5. Trebicka J, Bork P, Krag A, Arumugam M. Utilizing the gut microbiome in decompensated cirrhosis and acute-on-chronic liver failure. Nat Rev Gastroenterol Hepatol 2021;18:167–180.
6. Kwong AJ, Ebel NH, Kim WR, Lake JR, Smith JM, Schladt DP, et al. OPTN/SRTR 2021 annual data report: liver. Am J Transplant 2023;23(2 Suppl 1):S178–S263.
7. Hoang DM, Pham PT, Bach TQ, Ngo ATL, Nguyen QT, Phan TTK, et al. Stem cell-based therapy for human diseases. Signal Transduct Target Ther 2022;7:272.
8. Hazrati A, Malekpour K, Soudi S, Hashemi SM. Mesenchymal stromal/stem cells and their extracellular vesicles application in acute and chronic inflammatory liver diseases: emphasizing on the anti-fibrotic and immunomodulatory mechanisms. Front Immunol 2022;13:865888.
9. Uccelli A, Moretta L, Pistoia V. Mesenchymal stem cells in health and disease. Nat Rev Immunol 2008;8:726–736.
10. Zakrzewski W, Dobrzyński M, Szymonowicz M, Rybak Z. Stem cells: past, present, and future. Stem Cell Res Ther 2019;10:68.
11. Vizoso FJ, Eiro N, Cid S, Schneider J, Perez-Fernandez R. Mesenchymal stem cell secretome: toward cell-free therapeutic strategies in regenerative medicine. Int J Mol Sci 2017;18:1852.
12. Tan F, Li X, Wang Z, Li J, Shahzad K, Zheng J. Clinical applications of stem cell-derived exosomes. Signal Transduct Target Ther 2024;9:17.
13. Wen S, Dooner M, Cheng Y, Papa E, Del Tatto M, Pereira M, et al. Mesenchymal stromal cell-derived extracellular vesicles rescue radiation damage to murine marrow hematopoietic cells. Leukemia 2016;30:2221–2231.
14. van Niel G, D’Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol 2018;19:213–228.
15. Zhang Y, Liu Y, Liu H, Tang WH. Exosomes: biogenesis, biologic function and clinical potential. Cell Biosci 2019;9:19.
16. Morrison TJ, Jackson MV, Cunningham EK, Kissenpfennig A, McAuley DF, O’Kane CM, et al. Mesenchymal stromal cells modulate macrophages in clinically relevant lung injury models by extracellular vesicle mitochondrial transfer. Am J Respir Crit Care Med 2017;196:1275–1286.
17. Isaac R, Reis FCG, Ying W, Olefsky JM. Exosomes as mediators of intercellular crosstalk in metabolism. Cell Metab 2021;33:1744–1762.
18. Gurung S, Perocheau D, Touramanidou L, Baruteau J. The exosome journey: from biogenesis to uptake and intracellular signalling. Cell Commun Signal 2021;19:47.
19. Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, Lötvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol 2007;9:654–659.
20. Xiao X, Xu M, Yu H, Wang L, Li X, Rak J, et al. Mesenchymal stem cell-derived small extracellular vesicles mitigate oxidative stress-induced senescence in endothelial cells via regulation of miR-146a/Src. Signal Transduct Target Ther 2021;6:354.
21. Harrell CR, Jovicic N, Djonov V, Arsenijevic N, Volarevic V. Mesenchymal stem cell-derived exosomes and other extracellular vesicles as new remedies in the therapy of inflammatory diseases. Cells 2019;8:1605.
22. Du Z, Wu T, Liu L, Luo B, Wei C. Extracellular vesicles-derived miR-150-5p secreted by adipose-derived mesenchymal stem cells inhibits CXCL1 expression to attenuate hepatic fibrosis. J Cell Mol Med 2021;25:701–715.
23. Yu B, Zhang X, Li X. Exosomes derived from mesenchymal stem cells. Int J Mol Sci 2014;15:4142–4157.
24. Shen Z, Huang W, Liu J, Tian J, Wang S, Rui K. Effects of mesenchymal stem cell-derived exosomes on autoimmune diseases. Front Immunol 2021;12:749192.
25. Maroto R, Zhao Y, Jamaluddin M, Popov VL, Wang H, Kalubowilage M, et al. Effects of storage temperature on airway exosome integrity for diagnostic and functional analyses. J Extracell Vesicles 2017;6:1359478.
26. Lou G, Chen Z, Zheng M, Liu Y. Mesenchymal stem cell-derived exosomes as a new therapeutic strategy for liver diseases. Exp Mol Med 2017;49e346.
27. Hu P, Yang Q, Wang Q, Shi C, Wang D, Armato U, et al. Mesenchymal stromal cells-exosomes: a promising cell-free therapeutic tool for wound healing and cutaneous regeneration. Burns Trauma 2019;7:38.
28. Sato K, Meng F, Glaser S, Alpini G. Exosomes in liver pathology. J Hepatol 2016;65:213–221.
29. Krenkel O, Tacke F. Liver macrophages in tissue homeostasis and disease. Nat Rev Immunol 2017;17:306–321.
30. Orecchioni M, Ghosheh Y, Pramod AB, Ley K. Macrophage polarization: different gene signatures in M1(LPS+) vs. classically and M2(LPS-) vs. alternatively activated macrophages. Front Immunol 2019;10:1084.
31. Setten E, Castagna A, Nava-Sedeño JM, Weber J, Carriero R, Reppas A, et al. Understanding fibrosis pathogenesis via modeling macrophage-fibroblast interplay in immune-metabolic context. Nat Commun 2022;13:6499.
32. Tian S, Zhou X, Zhang M, Cui L, Li B, Liu Y, et al. Mesenchymal stem cell-derived exosomes protect against liver fibrosis via delivering miR-148a to target KLF6/STAT3 pathway in macrophages. Stem Cell Res Ther 2022;13:330.
33. Zhang Y, Zhang X, Zhang H, Song P, Pan W, Xu P, et al. Mesenchymal stem cells derived extracellular vesicles alleviate traumatic hemorrhagic shock induced hepatic injury via IL-10/PTPN22-mediated M2 Kupffer cell polarization. Front Immunol 2021;12:811164.
34. Shi Y, Yang X, Wang S, Wu Y, Zheng L, Tang Y, et al. Human umbilical cord mesenchymal stromal cell-derived exosomes protect against MCD-induced NASH in a mouse model. Stem Cell Res Ther 2022;13:517.
35. Shao M, Xu Q, Wu Z, Chen Y, Shu Y, Cao X, et al. Exosomes derived from human umbilical cord mesenchymal stem cells ameliorate IL-6-induced acute liver injury through miR-455-3p. Stem Cell Res Ther 2020;11:37.
36. Chen J, Chen ZJ. PtdIns4P on dispersed trans-Golgi network mediates NLRP3 inflammasome activation. Nature 2018;564:71–76.
37. Zhang S, Jiang L, Hu H, Wang H, Wang X, Jiang J, et al. Pretreatment of exosomes derived from hUCMSCs with TNF-α ameliorates acute liver failure by inhibiting the activation of NLRP3 in macrophage. Life Sci 2020;246:117401.
38. Levine B, Kroemer G. Biological functions of autophagy genes: a disease perspective. Cell 2019;176:11–42.
39. Ueno T, Komatsu M. Autophagy in the liver: functions in health and disease. Nat Rev Gastroenterol Hepatol 2017;14:170–184.
40. Hazari Y, Bravo-San Pedro JM, Hetz C, Galluzzi L, Kroemer G. Autophagy in hepatic adaptation to stress. J Hepatol 2020;72:183–196.
41. Allaire M, Rautou PE, Codogno P, Lotersztajn S. Autophagy in liver diseases: time for translation? J Hepatol 2019;70:985–998.
42. Qiao L, Hu J, Qiu X, Wang C, Peng J, Zhang C, et al. LAMP2A, LAMP2B and LAMP2C: similar structures, divergent roles. Autophagy 2023;19:2837–2852.
43. Babuta M, Furi I, Bala S, Bukong TN, Lowe P, Catalano D, et al. Dysregulated autophagy and lysosome function are linked to exosome production by micro-RNA 155 in alcoholic liver disease. Hepatology 2019;70:2123–2141.
44. Gao J, Wei B, de Assuncao TM, Liu Z, Hu X, Ibrahim S, et al. Hepatic stellate cell autophagy inhibits extracellular vesicle release to attenuate liver fibrosis. J Hepatol 2020;73:1144–1154.
45. Zhang XW, Zhou JC, Peng D, Hua F, Li K, Yu JJ, et al. Disrupting the TRIB3-SQSTM1 interaction reduces liver fibrosis by restoring autophagy and suppressing exosome-mediated HSC activation. Autophagy 2020;16:782–796.
46. Zhao S, Liu Y, Pu Z. Bone marrow mesenchymal stem cell-derived exosomes attenuate D-GaIN/LPS-induced hepatocyte apoptosis by activating autophagy in vitro. Drug Des Devel Ther 2019;13:2887–2897.
47. Qu Y, Zhang Q, Cai X, Li F, Ma Z, Xu M, et al. Exosomes derived from miR-181-5p-modified adipose-derived mesenchymal stem cells prevent liver fibrosis via autophagy activation. J Cell Mol Med 2017;21:2491–2502.
48. Tilg H, Wilmer A, Vogel W, Herold M, Nölchen B, Judmaier G, et al. Serum levels of cytokines in chronic liver diseases. Gastroenterology 1992;103:264–274.
49. Tacke F, Puengel T, Loomba R, Friedman SL. An integrated view of anti-inflammatory and antifibrotic targets for the treatment of NASH. J Hepatol 2023;79:552–566.
50. Batlle E, Massagué J. Transforming growth factor-β signaling in immunity and cancer. Immunity 2019;50:924–940.
51. Martinez FJ, Collard HR, Pardo A, Raghu G, Richeldi L, Selman M, et al. Idiopathic pulmonary fibrosis. Nat Rev Dis Primers 2017;3:17074.
52. Frangogiannis N. Transforming growth factor-β in tissue fibrosis. J Exp Med 2020;217e20190103.
53. Peng D, Fu M, Wang M, Wei Y, Wei X. Targeting TGF-β signal transduction for fibrosis and cancer therapy. Mol Cancer 2022;21:104.
54. Muñoz-Félix JM, González-Núñez M, López-Novoa JM. ALK1-Smad1/5 signaling pathway in fibrosis development: friend or foe? Cytokine Growth Factor Rev 2013;24:523–537.
55. Troyer DL, Weiss ML. Wharton’s jelly-derived cells are a primitive stromal cell population. Stem Cells 2008;26:591–599.
56. Afarin R, Behdarvand T, Shakerian E, Salehipour Bavarsad S, Rashidi M. Exosomes of Whartons’ jelly mesenchymal stem cell reduce the NOX genes in TGF-β-induced hepatic fibrosis. Iran J Basic Med Sci 2022;25:1498–1503.
57. Huang YJ, Cao J, Lee CY, Wu YM. Umbilical cord blood plasma-derived exosomes as a novel therapy to reverse liver fibrosis. Stem Cell Res Ther 2021;12:568.
58. Wang N, Li X, Zhong Z, Qiu Y, Liu S, Wu H, et al. 3D hESC exosomes enriched with miR-6766-3p ameliorates liver fibrosis by attenuating activated stellate cells through targeting the TGFβRII-SMADS pathway. J Nanobiotechnology 2021;19:437.
59. Wang L, Wang Y, Quan J. Exosomes derived from natural killer cells inhibit hepatic stellate cell activation and liver fibrosis. Hum Cell 2020;33:582–589.
60. Aletaha D, Kerschbaumer A, Kastrati K, Dejaco C, Dougados M, McInnes IB, et al. Consensus statement on blocking interleukin-6 receptor and interleukin-6 in inflammatory conditions: an update. Ann Rheum Dis 2023;82:773–787.
61. Garbers C, Heink S, Korn T, Rose-John S. Interleukin-6: designing specific therapeutics for a complex cytokine. Nat Rev Drug Discov 2018;17:395–412.
62. Kroy DC, Beraza N, Tschaharganeh DF, Sander LE, Erschfeld S, Giebeler A, et al. Lack of interleukin-6/glycoprotein 130/signal transducers and activators of transcription-3 signaling in hepatocytes predisposes to liver steatosis and injury in mice. Hepatology 2010;51:463–473.
63. Schmidt-Arras D, Rose-John S. IL-6 pathway in the liver: from physiopathology to therapy. J Hepatol 2016;64:1403–1415.
64. Yuan YG, Wang JL, Zhang YX, Li L, Reza AMMT, Gurunathan S. Biogenesis, composition and potential therapeutic applications of mesenchymal stem cells derived exosomes in various diseases. Int J Nanomedicine 2023;18:3177–3210.
65. Wang C, Xu M, Fan Q, Li C, Zhou X. Therapeutic potential of exosome-based personalized delivery platform in chronic inflammatory diseases. Asian J Pharm Sci 2023;18:100772.
66. Liu Y, Lou G, Li A, Zhang T, Qi J, Ye D, et al. AMSC-derived exosomes alleviate lipopolysaccharide/d-galactosamine-induced acute liver failure by miR-17-mediated reduction of TXNIP/NLRP3 inflammasome activation in macrophages. EBioMedicine 2018;36:140–150.
67. Deng C, Hu J, He L, Ge L, Wu N, Xie M, et al. Daucosterol combined with umbilical cord mesenchymal stem cell-derived exosomes can alleviate liver damage in liver failure mice by regulating the IL-6/STAT3 signaling pathway. Cancer Biol Ther 2023;24:2184150.
68. Psaraki A, Ntari L, Karakostas C, Korrou-Karava D, Roubelakis MG. Extracellular vesicles derived from mesenchymal stem/stromal cells: the regenerative impact in liver diseases. Hepatology 2022;75:1590–1603.
69. Sies H, Jones DP. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat Rev Mol Cell Biol 2020;21:363–383.
70. Bisht S, Faiq M, Tolahunase M, Dada R. Oxidative stress and male infertility. Nat Rev Urol 2017;14:470–485.
71. Jaeschke H, Ramachandran A. Reactive oxygen species in the normal and acutely injured liver. J Hepatol 2011;55:227–228.
72. Tanaka M, Miyajima A. Liver regeneration and fibrosis after inflammation. Inflamm Regen 2016;36:19.
73. Allameh A, Niayesh-Mehr R, Aliarab A, Sebastiani G, Pantopoulos K. Oxidative stress in liver pathophysiology and disease. Antioxidants (Basel) 2023;12:1653.
74. Zhang W, Wang T, Xue Y, Zhan B, Lai Z, Huang W, et al. Research progress of extracellular vesicles and exosomes derived from mesenchymal stem cells in the treatment of oxidative stress-related diseases. Front Immunol 2023;14:1238789.
75. Jiang W, Tan Y, Cai M, Zhao T, Mao F, Zhang X, et al. Human umbilical cord MSC-derived exosomes suppress the development of CCl4-induced liver injury through antioxidant effect. Stem Cells Int 2018;2018:6079642.
76. Yan Y, Jiang W, Tan Y, Zou S, Zhang H, Mao F, et al. hucMSC exosome-derived GPX1 is required for the recovery of hepatic oxidant injury. Mol Ther 2017;25:465–479.
77. Shah YM, Xie L. Hypoxia-inducible factors link iron homeostasis and erythropoiesis. Gastroenterology 2014;146:630–642.
78. Galaris D, Barbouti A, Pantopoulos K. Iron homeostasis and oxidative stress: an intimate relationship. Biochim Biophys Acta Mol Cell Res 2019;1866:118535.
79. Kang R, Zhu S, Zeh HJ, Klionsky DJ, Tang D. BECN1 is a new driver of ferroptosis. Autophagy 2018;14:2173–2175.
80. Tan Y, Huang Y, Mei R, Mao F, Yang D, Liu J, et al. HucMSC-derived exosomes delivered BECN1 induces ferroptosis of hepatic stellate cells via regulating the xCT/GPX4 axis. Cell Death Dis 2022;13:319.
81. Zheng JS, Sharp SJ, Imamura F, Chowdhury R, Gundersen TE, Steur M, et al. Association of plasma biomarkers of fruit and vegetable intake with incident type 2 diabetes: EPIC-InterAct case-cohort study in eight European countries. BMJ 2020;370:m2194.
82. Zhao WJ, Bian YP, Wang QH, Yin F, Yin L, Zhang YL, et al. Blueberry-derived exosomes-like nanoparticles ameliorate nonalcoholic fatty liver disease by attenuating mitochondrial oxidative stress. Acta Pharmacol Sin 2022;43:645–658.
83. Yang X, Li Q, Liu W, Zong C, Wei L, Shi Y, et al. Mesenchymal stromal cells in hepatic fibrosis/cirrhosis: from pathogenesis to treatment. Cell Mol Immunol 2023;20:583–599.
84. Damala M, Sahoo A, Pakalapati N, Singh V, Basu S. Preclinical evaluation of efficacy and safety of human limbus-derived stromal/mesenchymal stem cells with and without alginate encapsulation for future clinical applications. Cells 2023;12:876.
85. Sanchez-Diaz M, Quiñones-Vico MI, Sanabria de la Torre R, Montero-Vílchez T, Sierra-Sánchez A, Molina-Leyva A, et al. Biodistribution of mesenchymal stromal cells after administration in animal models and humans: a systematic review. J Clin Med 2021;10:2925.
86. He Y, Guo X, Lan T, Xia J, Wang J, Li B, et al. Human umbilical cord-derived mesenchymal stem cells improve the function of liver in rats with acute-on-chronic liver failure via downregulating Notch and Stat1/Stat3 signaling. Stem Cell Res Ther 2021;12:396.
87. Friman TK, Jäämaa-Holmberg S, Åberg F, Helanterä I, Halme M, Pentikäinen MO, et al. Cancer risk and mortality after solid organ transplantation: a population-based 30-year cohort study in Finland. Int J Cancer 2022;150:1779–1791.
88. Nassar W, El-Ansary M, Sabry D, Mostafa MA, Fayad T, Kotb E, et al. Umbilical cord mesenchymal stem cells derived extracellular vesicles can safely ameliorate the progression of chronic kidney diseases. Biomater Res 2016;20:21.
89. Kordelas L, Rebmann V, Ludwig AK, Radtke S, Ruesing J, Doeppner TR, et al. MSC-derived exosomes: a novel tool to treat therapy-refractory graft-versus-host disease. Leukemia 2014;28:970–973.
90. Shi MM, Yang QY, Monsel A, Yan JY, Dai CX, Zhao JY, et al. Preclinical efficacy and clinical safety of clinical-grade nebulized allogenic adipose mesenchymal stromal cells-derived extracellular vesicles. J Extracell Vesicles 2021;10e12134.
91. Lotfy A, AboQuella NM, Wang H. Mesenchymal stromal/stem cell (MSC)-derived exosomes in clinical trials. Stem Cell Res Ther 2023;14:66.
92. Cho E, Nam GH, Hong Y, Kim YK, Kim DH, Yang Y, et al. Comparison of exosomes and ferritin protein nanocages for the delivery of membrane protein therapeutics. J Control Release 2018;279:326–335.
93. Fernández-Messina L, Rodríguez-Galán A, de Yébenes VG, Gutiérrez-Vázquez C, Tenreiro S, Seabra MC, et al. Transfer of extracellular vesicle-microRNA controls germinal center reaction and antibody production. EMBO Rep 2020;21e48925.
94. Huang PY, Shih YH, Tseng YJ, Ko TL, Fu YS, Lin YY. Xenograft of human umbilical mesenchymal stem cells from Wharton’s jelly as a potential therapy for rat pilocarpine-induced epilepsy. Brain Behav Immun 2016;54:45–58.
95. Hastuti S, Idroes R, Imran I, Ramli Y, Abas AH, Tallei TE. hUMSC vs. hUMSC-exosome: which one is better for epilepsy? Pharmaceuticals (Basel) 2022;15:1247.
96. Sengupta V, Sengupta S, Lazo A, Woods P, Nolan A, Bremer N. Exosomes derived from bone marrow mesenchymal stem cells as treatment for severe COVID-19. Stem Cells Dev 2020;29:747–754.
97. Yu H, Huang T, Lu WW, Tong L, Chen D. Osteoarthritis pain. Int J Mol Sci 2022;23:4642.
98. Bloor AJC, Patel A, Griffin JE, Gilleece MH, Radia R, Yeung DT, et al. Production, safety and efficacy of iPSC-derived mesenchymal stromal cells in acute steroid-resistant graft versus host disease: a phase I, multicenter, open-label, dose-escalation study. Nat Med 2020;26:1720–1725.
99. Li M, Jiang Y, Hou Q, Zhao Y, Zhong L, Fu X. Potential preactivation strategies for improving therapeutic efficacy of mesenchymal stem cells: current status and future prospects. Stem Cell Res Ther 2022;13:146.
100. Liu B, Jin Y, Yang J, Han Y, Shan H, Qiu M, et al. Extracellular vesicles from lung tissue drive bone marrow neutrophil recruitment in inflammation. J Extracell Vesicles 2022;11e12223.
101. Masyuk AI, Masyuk TV, Larusso NF. Exosomes in the pathogenesis, diagnostics and therapeutics of liver diseases. J Hepatol 2013;59:621–625.
102. Lin Z, Wu Y, Xu Y, Li G, Li Z, Liu T. Mesenchymal stem cell-derived exosomes in cancer therapy resistance: recent advances and therapeutic potential. Mol Cancer 2022;21:179.
103. Khatri M, Richardson LA, Meulia T. Mesenchymal stem cell-derived extracellular vesicles attenuate influenza virus-induced acute lung injury in a pig model. Stem Cell Res Ther 2018;9:17.
104. Zhang X, Zhang H, Gu J, Zhang J, Shi H, Qian H, et al. Engineered extracellular vesicles for cancer therapy. Adv Mater 2021;33e2005709.
105. Wu L, Xie W, Li Y, Ni Q, Timashev P, Lyu M, et al. Biomimetic nanocarriers guide extracellular ATP homeostasis to remodel energy metabolism for activating innate and adaptive immunity system. Adv Sci (Weinh) 2022;9e2105376.
106. Marar C, Starich B, Wirtz D. Extracellular vesicles in immunomodulation and tumor progression. Nat Immunol 2021;22:560–570.
107. Hu T, Wolfram J, Srivastava S. Extracellular vesicles in cancer detection: hopes and hypes. Trends Cancer 2021;7:122–133.
108. Whitford W, Guterstam P. Exosome manufacturing status. Future Med Chem 2019;11:1225–1236.
109. Lötvall J, Hill AF, Hochberg F, Buzás EI, Di Vizio D, Gardiner C, et al. Minimal experimental requirements for definition of extracellular vesicles and their functions: a position statement from the International Society for Extracellular Vesicles. J Extracell Vesicles 2014;3:26913.
110. Ha DH, Kim HK, Lee J, Kwon HH, Park GH, Yang SH, et al. Mesenchymal stem/stromal cell-derived exosomes for immunomodulatory therapeutics and skin regeneration. Cells 2020;9:1157.
111. Li J, Tan M, Xiang Q, Zhou Z, Yan H. Thrombin-activated platelet-derived exosomes regulate endothelial cell expression of ICAM-1 via microRNA-223 during the thrombosis-inflammation response. Thromb Res 2017;154:96–105.
112. Vakhshiteh F, Atyabi F, Ostad SN. Mesenchymal stem cell exosomes: a two-edged sword in cancer therapy. Int J Nanomedicine 2019;14:2847–2859.
113. Rezaie J, Feghhi M, Etemadi T. A review on exosomes application in clinical trials: perspective, questions, and challenges. Cell Commun Signal 2022;20:145.

Article information Continued

Figure 1.

The nucleic acids and proteins produced by the nucleus and organelles, as well as substances taken up through endocytosis, together form early endosomes, which are then expelled from the cell through exocytosis. There are various functional substances present in the exocrine body and it s surface, such as proteins, DNA, miRNA, etc.

Figure 2.

LPS and IFN-γ induce M1 macrophages, whereas IL-4 and IL-13 induce M2 macrophages. Exosomes containing specific miRNAs derived from stem cells can modulate the polarization of macrophages. This modulation enables pro-inflammatory M1 macrophages to transform into anti-inflammatory M2 macrophages.

Figure 3.

The interaction between exosomes and autophagy significantly impacts the progression of liver fibrosis. (I) To summarize, autophagy occurs in both parenchymal and non-parenchymal cells of the liver. This process can help mitigate liver fibrosis. (II) When autophagy in hepatocytes is inhibited, the release of exosomes increases. (III) When platelet-derived growth factor activates hepatic stellate cells, blocking their autophagy leads to an increase in exosome release. (IV) AMSC-Exosomes and BMSC-Exosomes help prevent liver fibrosis by promoting the process of autophagy.

Figure 4.

Exosomes of different cellular origins can alleviate liver fibrosis via regulating TGF-β/Smad pathway.

Table 1.

Clinical applications of exosomes

Disease Number of participants in the experiment Exosome origin Application effect Ref.
Chronic kidney disease 40 Umbilical cord mesenchymal stem cells During the 12-month study period, significant improvements were observed in eGFR (estimated glomerular filtration rate), serum creatinine levels, blood urea content, and UACR (urine microalbumin to creatinine ratio). [88]
Graft-versushost disease (GvHD) 1 Bone marrow mesenchymal stem cells After MSC Exos treatment, the symptoms of GvHD significantly alleviated. Within two weeks, there was a noticeable control effect on the GvHD of the skin and mucous membranes, which remained stable for the following four months. [89]
COVID-19 24 Bone marrow mesenchymal stem cells The patient survival rate is 83%; among them, approximately 71% achieved complete recovery, 13% are in a stable condition despite being critically ill, and 16% died due to nontreatment-related reasons. [96]
Osteoarthritis 33 Bone marrow mesenchymal stem cells A 6-month follow-up observation of patients' recovery progress showed an average pain intensity reduction of 77%, an average decrease of 80% in the disability index, an average improvement of 76% in lower limb function scale scores, an average increase of 51% in upper limb function scale scores, and a significant improvement in quality of life or daily activity capabilities. [97]
Health volunteer 24 Adipose-derived mesenchymal stem cells No adverse reactions. [90]