259
Views
0
CrossRef citations to date
0
Altmetric
REVIEW

Mesenchymal Stem Cell–Derived Exosomes in Various Chronic Liver Diseases: Hype or Hope?

, , , , , , , & show all
Pages 171-189 | Received 12 Sep 2023, Accepted 27 Dec 2023, Published online: 10 Jan 2024

Abstract

Chronic liver conditions are associated with high mortality rates and have a large adverse effect on human well-being as well as a significant financial burden. Currently, the only effective treatment available for the effects of liver failure and cirrhosis resulting from the progression of several chronic liver diseases is liver transplantation carried out at the original location. This implies that developing novel and effective treatments is imperative. Regenerative medicine has long been associated with stem cell therapy. Mesenchymal stem cells (MSCs), a type of cell with great differentiation potential, have become the preferred source for stem cell therapy. According to recent studies, MSCs’ paracrine products—rather than their capacity for differentiation—play a significant therapeutic effect. MSC exosomes, a type of extracellular vesicle (MSC-EV), came into view as the paracrine substances of MSCs. According to research, MSC exosomes can maintain tissue homeostasis, which is necessary for healthy tissue function. All tissues contain them, and they take part in a variety of biological activities that support cellular activity and tissue regeneration in order to preserve tissue homeostasis. The outcomes support the use of MSCs and the exosomes they produce as a therapeutic option for a range of diseases. This review provides a brief overview of the source of MSC-EVs and outlines their physiological roles and biochemical capabilities. The elucidation of the role of MSC-EVs in the recovery and repair of hepatic tissues, as well as their contribution to maintaining tissue homeostasis, is discussed in relation to different chronic liver diseases. This review aims to provide new insights into the unique roles that MSC-EVs play in the treatment of chronic liver diseases.

Introduction

Numerous pathogenic factors, such as viruses, chemicals, and autoimmune reactions, and the excessive consumption of alcohol can result in both acute and chronic liver damage, which leads to inflammation.Citation1 The liver cannot replace damaged liver cells, which leads to the development of serious problems like hepatic encephalopathy and hepatorenal syndrome, as well as symptoms including jaundice and poor blood coagulation.Citation2 This, in turn, can result in cirrhosis, liver failure, and, potentially, liver cancer, with a mortality rate ranging from 50% to 90%.Citation3 Current therapies for many liver diseases, however, are limited. In the case of advanced liver disease, especially cirrhosis and liver failure, the only possible treatment is a liver transplant performed in the patient’s body.Citation4

However, the limited availability of liver donors, exorbitant expenses, complications after surgery, and the risk of organ rejection are significant deterrents to transplantation.Citation5 As a result, researchers from all around the world have been exploring alternate types of therapy that can reverse fibrosis, decrease liver inflammation and necrosis, and promote the regeneration of liver cells.Citation6,Citation7

MSCs are found in various tissues and are not restricted to mesodermal organs such as the bone marrow, adipose tissue, muscle, or bone.Citation8,Citation9 They can also be extracted from the brain, spleen, liver, kidneys, lungs, thymus, and pancreas.Citation10–12 Owing to their remarkable ability to regenerate, in recent times, MSCs have undergone clinical testing for a broader spectrum of disease indications,Citation13 and the utilization of MSCs in clinical trials has seen tremendous growth.Citation14,Citation15 MSCs are easily extracted from adult tissues and have a strong in vitro expansion capacity.Citation16,Citation17

Despite being found in a variety of tissues, MSCs share comparable phenotypic characteristics, and some may even have extra features that are indicative of the source tissues.Citation17–19 MSCs can transform into a minimum of three different cell types, namely, adipocytes, chondrocytes, and osteocytes.Citation19,Citation20 Because MSCs are able to distinguish, their use in clinical trials has expanded substantially.Citation21 MSCs are now well recognized as a type of precursor cells that support the stroma and have the ability to differentiate into stroma-supporting cells. They also produce pro-stromal factors and diverse cell growth factors.Citation22–25

MSCsCitation26 play a role in maintaining the hematopoietic stem cell balance within the matrix and also contribute to the maintenance of microenvironmental homeostasis.Citation27 Self-renewal and differentiation maintain equilibrium between vital and static hematopoietic stem cells within this microenvironment.Citation28 MSCs are becoming increasingly important in the study of liver diseases due to their capacity for self-renewal and multi-directional differentiation. These cells can be obtained from different tissues, including bone marrow, umbilical cord, and adipose tissue. In addition, they have immunomodulatory, apoptosis-inhibition, cell-regeneration, and anti-fibrotic properties.Citation13,Citation19,Citation29–33 The liver, being a crucial organ in regulating immunity and metabolism, contains numerous immune cells, including myeloid and lymphoid cells. Disruption of hepatic immune homeostasis is often linked to various liver diseases.Citation34,Citation35 Recent clinical investigations have shown that therapies utilizing MSCs can reduce liver damage, improve liver function, and promote the regeneration of liver tissue.Citation36,Citation37

However, with the growing use and study of MSCs, concerns have emerged regarding their theoretical basis related to their ability to differentiate. Multiple studies have shown that although MSC therapy leads to the restoration of function, only a small number of cells achieve differentiation into appropriate tissues after MSC transplantation.Citation38,Citation39 Moreover, the therapeutic effect of transplanted MSCs does not depend on the closeness of the transplantation location to the injury site.Citation40 Currently, there is a prevailing opinion that the immunomodulatory function of MSCs is predominantly accomplished through the paracrine pathway. This pathway leads to the reduction of damage and promotes the process of healing.Citation41,Citation42 Extracellular vesicles (EVs) are released by phospholipid bilayer-containing cells and play a crucial role in facilitating cell-to-cell communication by carrying membrane and cytoplasmic proteins, lipids, and RNA.Citation24,Citation40,Citation43–45 MSC-derived EVs (MSC-EVs) are significantly smaller and more readily obtainable and storable compared to MSCs.Citation46 Following in vivo intravenous administration, MSC-EVs primarily accumulate in the liver. The occurrence of liver diseases is intimately associated with an imbalance in immunological homeostasis, which is closely related to the function of MSC-EVs.Citation47 Hence, researchers are investigating the utilization of MSC-EVs as a substitute for MSCs in liver ailments.Citation48 This review examines the advancements in the utilization of MSC-EVs in treating various chronic liver ailments.

Overview of MSCs

MSCs are pluripotent cells present in nearly all types of postnatal organs and tissues.Citation49 Moreover, MSCs possess a robust capacity for migration.Citation11 MSCs possess strong immunomodulatory properties.Citation50 MSCs are being increasingly utilized in clinical trials that focus on tissue repair and regeneration owing to their immunomodulatory and cell survival-enhancing characteristics.Citation51 Nevertheless, in contrast to expectations, numerous preclinical investigations have reported that MSCs do not integrate effectively into tissues in significant quantities, and the duration of integration is inadequate.Citation12

MSCs possess strong immunomodulatory properties, effectively suppressing T cells, B cells, and natural killer (NK) cells in addition to their ability to differentiate in multiple directions.Citation52 Presently, there is substantial evidence backing the therapeutic and immunomodulatory roles of MSCs, primarily relying on paracrine effects and the release of secretory components. Therefore, many scholars are showing interest in the paracrine functions of MSCs, focusing specifically on paracrine secretions such as EVs.Citation53,Citation54 According to their findings, MSCs are capable of producing and releasing numerous growth factors, chemokines, and cytokines, which, in turn, have an impact on the neighboring cells.Citation20 In 2005, Gnecchi et al proposed that MSCs can release protective substances.Citation55 They intramyocardially injected the culture supernatants from MSCs with high Akt gene expression, as well as MSCs alone, into an animal model of acute infarction. Both animal groups showed a decrease in the myocardial infarction area to a similar extent, thereby validating their hypothesis.Citation56 Furthermore, it has been reported that MSCs enhance microcirculation and produce various anti-apoptotic proteins, including Bcl-2 and Akt, thereby averting cell death.Citation57,Citation58 Numerous studies have confirmed the likelihood of the hypothesis that the healing benefits of MSCs are attributable to their paracrine effects rather than their differentiation effects.

Recently, studies have shown that under appropriate conditions, MSCs can be differentiated into hepatocyte-like cells (HLCs), providing new ideas for the treatment of liver diseases. Liver-specific miR-122 can be efficiently transfected into MSCs and activate their differentiation into HLCs. Transplantation of these engineered MSCs can treat acute liver failure by replenishing hepatocyte function.Citation59 Various liver diseases can lead to hepatic failure, and obtaining a sufficient number of functional hepatocytes is the key to liver regeneration.Citation60 Since the functions of primary hepatocytes are unstable, directing the differentiation of MSCs and other stem cells into HLCs, as well as developing and utilizing more stable and safer extracellular vesicle products, are the directions that researchers are paying attention to. These studies have provided a theoretical basis for the application of MSCs in the treatment of liver diseases.

Overview of MSC-EVs

Research on MSCs’ release of extracellular vesicles has advanced significantly to date, and it has been shown that MSCs secrete a variety of extracellular vesicles, including exosomes, microvesicles, and microparticles.Citation61 Studies indicate that MSC-EVs have a significant impact on the transfer and regulation of intercellular information.Citation47,Citation62 Scientists initially conducted thorough investigations into the mechanism of MSC-EVs generation. The study determined that the generation of MSC-EVs predominantly takes place through the endoplasmic reticulum and multivesicular body pathways.Citation63 The endoplasmic reticulum pathway involves the formation of vesicles on the endoplasmic reticulum, which bind to membrane transport proteins (eg, MVB) via fusion and are ultimately released outside the cell.Citation64,Citation65 The multivesicular body pathway includes the discharge of MSC-EVs by shedding vesicles directly from the cellular membrane in reaction to the equilibrium of the plasma membrane.Citation66 Gaining knowledge about these pathways provides a basis for further investigation into the biological functions of MSC-EVs.

In addition, a comprehensive examination of the components of MSC-EVs was carried out. A multitude of bioactive constituents, including proteins, nucleic acids, and lipids, have been identified inside MSC-EVs.Citation48 Proteomics analysis revealed that MSC-EVs contain a diverse range of operational proteins, such as cell adhesion agents, enzymes that degrade the extracellular matrix, and factors that modulate the immune system.Citation67 Furthermore, MSC-EVs contain miRNAs, mRNAs, and various nucleic acids in abundance, which have the potential to influence gene expression and cellular functions when transferred to recipient cells.Citation68 MSC-EVs demonstrate various biological capabilities and potential uses in medical practice.Citation69 Based on empirical evidence, MSC-EVs have been demonstrated to exhibit anti-inflammatory, anti-fibrotic, angiogenesis-promoting, and immune-modulating properties.Citation70

Furthermore, extensive research has been conducted on the use of MSC-EVs to manage diverse ailments, including cardiovascular disorders, neurological conditions, and immune-related diseases.Citation71–73 The potential applications of MSC-EVs in disease treatment and tissue regeneration are being investigated through preclinical and clinical trials, which have shown promising results.

Currently, exosomes are the most characterized extracellular vesicles as they have more biologically and biochemically defined parameters that can be detected in conventional laboratories.Citation74 Exosomes range from 40–100 nm in size, have a density of 1.10–1.18 g/mL on a sucrose gradient, and are linked to marker proteins such as Alix and Tsg101, as well as tetrameric transmembrane proteins including CD9, CD63, and CD81.Citation75,Citation76 Exosomes, derived from introns,Citation76 are the sole extracellular vesicles that are currently recognized. Introns form vesicles with several layers and a large number of luminal vesicles by invaginating the endosomal membrane.Citation76 When they fuse with the cell membrane, luminal vesicles are released to form exosomes.Citation77 This distinguishes the process of exosome formation from that of other extracellular vesicle types. Through extensive examination of MSC-EVs, it has been established that these minuscule extracellular vesicles have a significant impact on the communication and regulation between cells.Citation76 Additional investigation will reveal the precise molecular mechanism of MSC-EVs, improve their composition, and broaden their potential uses in a therapeutic context. This has the potential to provide innovative strategies and techniques for enhancing the management of chronic liver disease and promoting tissue regeneration.Citation78

Functions of MSC-EVs

MSC-EVs have received extensive research attention as a novel extracellular messaging medium.Citation79 The primary role of exosomes is to transmit cellular elements from secretory cells to receptor cells in one direction, thereby controlling the activity of the latter.Citation80 Similarly, MSC-EVs are responsible for intercellular communication.Citation81 Exosomes derived from MSCs can control the activity and role of immune cells through the transportation of diverse regulatory proteins, miRNAs, and other biologically active substances.Citation82 Clinical investigations demonstrate that MSC-EVs can perform an immunomodulatory effect by suppressing T cell activation, regulating macrophage polarization, and modifying B cell activity.Citation83 Treatment of autoimmune liver diseasesCitation84 relies on this immunomodulatory property.

Furthermore, MSC-EVs can secrete bioactive compounds that consist of anti-inflammatory proteins and miRNAs, thereby controlling inflammatory reactions and preventing the release of inflammatory mediators.Citation85 Studies show that MSC-EVs can mitigate the inflammatory pathological process, thereby mitigating the progression of inflammatory liver condition.Citation86 Moreover, MSC-EVs can transport numerous biologically active substances that enhance the growth of cells and the formation of new blood vessels, ultimately aiding in tissue healing and renewal.Citation87 Several studies have indicated the role of MSC-EVs in the regeneration of cardiovascular, neural, hepatic, skeletal, and various other tissues.Citation84,Citation88–90 Tissue regeneration function could be a novel approach to treating liver tissue injury. In addition, MSC-EVs, as a natural nanoparticle, have potential applications in drug delivery.Citation91 Researchers have modified the composition and internal structure of exosomes to enable the precise delivery of drugs to specific cells and tissues.Citation92 Targeted therapy and personalized medicine may be based on this drug delivery mechanism to treat liver diseases. MSC-EVs generally have a wide range of activities, such as immunomodulation, inflammation reduction, tissue regeneration promotion, and drug delivery facilitationCitation79,Citation91,Citation93,Citation94 (). More studies on MSC-EVs can lead to a better understanding of their molecular mechanisms, which will facilitate the development of more effective clinical implementation strategies and provide new therapeutic prospects for the treatment of disease and tissue regeneration.

Figure 1 Functions of MSC-EVs. MSC-EVs can transport a variety of bioactive substances, promote cell growth and the formation of new blood vessels, and control the activity and role of immune cells. In addition, MSC-EV, as a natural nanoparticle, has potential applications in drug delivery.

Note: This figure is originally drawn by Figdraw platform (www.figdraw.com).
Figure 1 Functions of MSC-EVs. MSC-EVs can transport a variety of bioactive substances, promote cell growth and the formation of new blood vessels, and control the activity and role of immune cells. In addition, MSC-EV, as a natural nanoparticle, has potential applications in drug delivery.

MSC-EVs in the Treatment of Liver Diseases

Lately, a growing body of research has demonstrated the profound promise of MSC-EVs made from MSCs as a medicinal strategy for treating liver conditions.Citation95 In the human body, the liver is the primary organ responsible for metabolism and immune function.Citation96 The majority of the blood that enters the liver is from the portal vein instead of the hepatic artery.Citation97 The liver parenchyma receives blood primarily through the periportal vessels and is then drained from the liver parenchyma by the central hepatic vein through the intricate system of hepatic sinusoids.Citation98 Hepatic sinusoids serve as anatomical locations that maintain immune homeostasis.Citation99 Lymphocyte extravasation is promoted by the prolonged contact between lymphocytes and antigen-presenting cells due to the sluggish circulation in the hepatic sinusoids.Citation100 Besides hepatocytes and cholangiocytes, there are nonparenchymal cells such as hepatic stellate cells (HSCs) and hepatic sinusoidal endothelial cells, as well as numerous immune cells such as T cells, macrophages, and dendritic cells in the liver.Citation101,Citation102 Liver diseases are typically linked to an imbalance in immune equilibrium, and this immune control includes the varied functionality of macrophages and dendritic cells, the proportion of various T-cell subsets (such as T helper [Th]17 and regulatory T [Treg] cells), the equilibrium between inflammatory and anti-inflammatory cytokines, and other factors related to immunity, which align perfectly with the role of MSC-EVs.Citation99,Citation103,Citation104 MSC-EVs primarily accumulate in the liver when administered intravenously in vivo.Citation68 Through immunomodulation, the regulation of MSV-EVs can lead to a decrease in the release of interleukin (IL)-6 and IL-1β by macrophages, and a decrease in the expression of CD154 by CD4+ T cells. This reduction in cytokine release and CD154 expression could potentially alleviate hepatic inflammation and provide relief in various liver diseases.Citation63,Citation69,Citation105

Role of MSC-EVs in Liver Diseases

This section provides an overview of preclinical investigations on MSC-EVs as a treatment modality for liver diseases such as liver failure, steatohepatitis linked to metabolism, autoimmune hepatitis (AIH), hepatic fibrosis, and hepatic ischemia reperfusion injury (HIRI). summarizes the specific role of MSC-EVs in different liver diseases. The emphasis is on understanding the molecular mechanisms and regulation of MSC-EVs, as discussed in relevant studies.Citation48,Citation106,Citation107

Table 1 Is a Summary of the Specific Role of MSC-EVs in Different Diseases

MSC-EVs and Acute Liver Injury

The two main pathophysiological features of liver failure that impact the organism as a whole and the liver specifically are immunological dysfunction and inflammation.Citation138 The rapid occurrence of apoptosis and necrosis in a large number of liver cells due to severe immune damage is followed by the development of ischemia, hypoxia, and endotoxemia. This sequence of events drives and speeds up the advancement of liver failure, commonly referred to as the “triple whammy” theory of liver failure.Citation138–140 Membrane permeability increases in the injured hepatocytes in the early stages of liver injury. This results in the release of molecules linked to damage-associated intracellular patterns. These molecules then bind to Toll-like receptors, triggering the activation of downstream signaling pathways such as c-jun amino terminal kinase, mitogen-activated protein kinase (MAPK), nuclear factor-κB (NF-κB), and signal transducer and activator of transcription 3.

Consequently, inflammatory cells are induced to infiltrate the affected area, further exacerbating liver failure through the promotion of tumor necrosis factor (TNF)-α, IL-1β, IL-6, IL-8, and other inflammatory factors, as well as cascade reactions.Citation141 Exosomes derived from various MSCs have been used recently by researchers to treat various liver failure models, proving that they can protect against liver damage.Citation142 Yan et alCitation143 administered human umbilical cord MSC-derived exosomes (ucMSC-Ex) at low (8 mg/kg), medium (16 mg/kg), and high (32 mg/kg) doses to mice with carbon tetrachloride (CCl4)-induced liver failure. Large areas of steatosis and hepatocellular necrosis were found in the liver tissues of mice administered the low dose, whereas the liver tissue lesions in mice in the medium- and high-dose groups were significantly alleviated. EVs promoted recovery from hepatic oxidative injury by delivering GPX1. Besides, several animal experiments have confirmed that the protective role of MSC-Ex in various models of liver failure included inhibition of inflammation, anti-apoptosis effects, and attenuation of oxidative stress.Citation144

Human endometrial MSC-EVs have been found to alleviate liver function and reduce hepatocyte apoptosis in a mouse model of ALI, potentially by migrating to the injured liver and suppressing inflammatory responses.Citation112 Bone-marrow-derived MSC-EVs could also accumulate in the injured liver tissue and attenuate damage after systemic administration in a lethal mouse model of acute liver failure, with improved survival rates compared to controls.Citation113 The therapeutic effects were partly mediated by a highly enriched Y RNA fragment in MSC-EVs.

Adipose tissue-derived MSC-EVs (AMSC-Exos) could protect against inflammasome-induced inflammation in acute liver failure by shuttling miR-17 into hepatic macrophages and suppressing NLRP3 activation via downregulating TXNIP.Citation115 This highlights the involvement of exosomal microRNAs as important therapeutic agents. AMSC-Exos may suppress the production of inflammatory cytokines and restore liver function by targeting NLRP3-regulated inflammation. Another study found human umbilical cord MSC-EVs attenuated NLRP3 inflammasome activation and reduced liver injury markers in a mouse model of acute liver failure, likely via dampening inflammatory responses of macrophages.Citation114 The effects were associated with decreased expression of NLRP3, Caspase-1, IL-1β and IL-6. Furthermore, systemic administration of BMSC-EVs activated regenerative mechanisms and inhibited apoptosis to protect against toxicant-induced acute liver injury in mice.Citation116 The hepatoprotective effects involved upregulation of proliferation and cell cycle-related genes.

In summary, accumulating evidence has characterized the therapeutic potentials of MSC-EVs in various models of acute liver damage, which are exerted through suppression of inflammasome-mediated inflammation responses, activation of hepatocyte regeneration, and inhibition of cell death pathways. Further research is warranted to understand better the responsible bioactive molecules shuttled by MSC-EVs and optimize treatment strategies.

MSC-EVs and Metabolism-Associated Fatty Liver Disease (MAFLD)

MAFLD is a condition where there is an accumulation of fat in liver tissues even in the absence of significant alcohol consumption or related metabolic disorders.Citation145 Presently, MAFLD poses a major challenge. The latest data suggest that its occurrence is on the rise worldwide, reaching 24% and resulting in a substantial financial burden.Citation146 A study found that human umbilical cord MSC-EVs containing miR-627-5p could improve insulin tolerance, alleviate liver injury, modulate glucose/lipid metabolism and reduce lipid deposition in an MAFLD rat model.Citation136 The effects were associated with miR-627-5p targeting and suppressing Fat Mass and Obesity associated (FTO) gene expression. In vitro, miR-627-5p overexpressing EVs also promoted cell viability, inhibited palmitic acid-induced apoptosis of LO-2 hepatocytes, and regulated a panel of metabolism-related genes. The data support miR-627-5p enriched MSC-EVs as a promising approach to improve MAFLD progression by enhancing metabolic profiles and reducing liver damage.

Another study reported that adipose-derived MSC-EVs delivering anti-fibrotic miR-223-3p could attenuate lipid accumulation and liver fibrosis by targeting E2F1, which plays a key role in hepatic stellate cell activation.Citation137 The anti-fibrotic effects were validated in a mouse model of diet-induced MAFLD. This highlights the therapeutic potential of functional MSC-EVs achieved by manipulating exosomal microRNA cargo. MSC-EVs can also regulate lipid metabolism. In addition, some studies show that MSC-EVs can enhance the process of fatty acid oxidation and hinder the synthesis of fatty acids, ultimately leading to decreased fat accumulation.Citation147

Furthermore, MSC-EVs affect the differentiation and viability of adipocytes and decrease the inflammatory reaction in adipose tissues.Citation148 The advancement of MAFLD is known to be frequently accompanied by hepatic fibrosis, and MSC-EVs can impede the progression of liver fibrosis.Citation137 According to studies, MSC-EVs can regulate the activity of matrix metalloproteinases and reduce the formation and accumulation of collagen in the liver, which decreases the severity of fibrosis.Citation149

In conclusion, MSC-EVs have emerged as a promising therapeutic agent for MAFLD, with a range of beneficial activities demonstrated in preclinical studies. These activities include resolving metabolic disorders, suppressing inflammation, inhibiting fibrosis progression, and promoting liver regeneration. Further research is required to understand the underlying mechanisms better and facilitate the translation of findings into clinical practice.

MSC-EVs and AIH

AIH is a chronic, progressive liver disease caused by autoimmune disorders. It is characterized by liver inflammation, infiltration of lymphocytes and plasma cells, and elevated ALT, AST, and immunoglobulin G (IgG) levels in the blood.Citation150 The clinical diagnosis of AIH relies on liver biopsy to evaluate for interfacial hepatitis, rosette nodules, and lymphocytic infiltration.Citation151 The current initial therapy for AIH involves the use of corticosteroids with or without azathioprine.Citation152 Although the precise origin of AIH is unknown, scientists believe that a mix of environmental variables, genetic susceptibility, and molecular mimicry may be responsible for lowering autoimmune tolerance.Citation153,Citation154

The primary method of treating AIH at the moment is either hormone therapy alone or in conjunction with immunosuppressants. It has significant drawbacks and limitations, such as prolonged treatment duration, adverse responses, insufficient patient adherence, and failure to stop the advancement of liver fibrosis and cirrhosis, even though it may, to some extent, increase patients’ chances of survival.Citation152 Therefore, the treatment of AIH has always been a clinical focus owing to these challenges.

MSC-EVs has emerged as a promising cell-free therapy for autoimmune hepatitis. Studies have shown that MSC-exosomes can attenuate liver injury and inflammation in mouse models of autoimmune hepatitis induced by concanavalin A or liver antigen S100.Citation110,Citation111 The anti-inflammatory and hepatoprotective effects of MSC-exosomes are comparable to mesenchymal stem cell transplantation.Citation110 Tracking experiments revealed that MSC-exosomes could target the injured liver after injection. MSC-exosomes can transfer microRNAs, such as miR-223-3p, to hepatocytes and immune cells like macrophages in the liver, regulating cell death, inflammation and immune responses.Citation109 It also inhibited the activation of NLRP3 inflammasome and caspase-1 activation, thus reducing IL-1β production and pyroptosis.Citation108 MSC-exosomes modulate the polarization of macrophages from pro-inflammatory M1 phenotype to anti-inflammatory M2 phenotypes and increase the number and function of regulatory T cells in the liver through immunosuppressive molecules like TGF-β.

Furthermore, therapeutic molecules like dexamethasone can be loaded into MSC exosomes to enhance the treatment outcome. The drug-loaded exosomes exhibit better liver distribution and anti-inflammatory effects.Citation111 MSC-exosomes show promise as a cell-free therapy for AIH through multiple mechanisms of action. Further investigations are warranted to promote the clinical translation of MSC-exosomes.

MSC-EVs and Liver Fibrosis

Liver fibrosis is a degenerative condition that occurs as a result of long-term liver damage. It is a persistent inflammatory response characterized by the replacement of normal liver tissue with fibrous tissue. Many chronic liver diseases, such as hepatitis B and hepatitis C, fatty liver, and alcoholic liver disease, often lead to liver fibrosis. Additionally, the presence of extensive fibrosis may result in cirrhosis and liver insufficiency.Citation155 HSCs that are activated in the liver are the primary fibroblasts and have a crucial function in the development of liver fibrosis. Normally, HSCs stay dormant and store vitamin A and bilirubin. However, when the liver is damaged, HSCs become active and transform into cell types linked to fibrosis. HSCs that have been activated release collagen, fibronectin, and other substances that stimulate the production of collagen fibers, resulting in the development of liver tissue fibrosis.Citation156

Angiogenesis plays a crucial role in the development of liver fibrosis, resulting in the development of new blood vessels with a supply of oxygen and vital nutrients.Citation157 Activation of vascular endothelial cells by vascular endothelial growth factors (eg, vascular endothelial growth factor-A) and fibronectin-inducing factors (eg, transforming growth factor β) leads to the stimulation of abnormal vasculogenesis and perivascular inflammatory reactions.Citation158

However, the primary cause of liver fibrosisCitation157 is the persistent inflammatory reaction resulting from long-term liver disease. Inflammation-causing agents include cytokines (TNF-α, IL-1, IL-6) and chemokines (alcohol, compounds from oxidative stress). These substances induce hepatocellular damage and apoptosis by stimulating the development of liver fibrosis.Citation40 Furthermore, the accumulation of collagen and other matrix components in a fibrosed liver modifies the organization and performance of the liver. Activated HSCs release matrix metalloproteinase inhibitors, which inhibit matrix metalloproteinase activity, leading to matrix accumulation and deposition and promoting the progression of fibrosis.Citation132

In addition to the mechanisms mentioned above, several other molecular pathways, including the TGF-β/Smad pathway, Wnt/β-catenin pathway, and NF-κB pathway, are also considered significant in the development of liver fibrosis. Numerous preclinical investigations have recently reported the anti-fibrotic properties of MSC-EVs in the liver,Citation159 lungs,Citation160 kidneys,Citation161 and heart.Citation162 As stated previously, the progression of liver fibrosis is intricately linked to a persistent inflammatory reaction, and MSC-EVs exert anti-inflammatory effects. MSC-EVs can suppress the synthesis of inflammatory cytokines, including TNF-α and IL-1β, leading to a decrease in inflammation.Citation141 The transformation of epithelial cells to mesenchymal cells can be inhibited by MSC-EVs, leading to the amelioration of CCl4-induced hepatic fibrosis.Citation128

Furthermore, the activation of HSCs can be significantly inhibited by MSC-EVs. The primary feature of hepatic fibrosis is the accumulation of collagen and other matrix components. MSC-EVs can reduce the progression of fibrosis by suppressing collagen production and regulating matrix metalloproteinase activity.Citation132 MSC-EVs can directly act on hepatic stellate cells to inhibit their activation and proliferation, thereby reducing the expression of collagen and other fibrosis-related genes.Citation127,Citation128,Citation133 Suppressing the excessive proliferation of hepatic stellate cells and the over-deposition of collagen is an important mechanism for the anti-fibrotic effects of MSC-EVs. MSC-EVs can also reduce the accumulation and activation of inflammatory cells, such as macrophages in the liver,Citation125,Citation130 since inflammatory responses and immune cell activation are closely related to the process of liver fibrosis. By inhibiting inflammatory signaling pathways such as NF-kB,Citation125,Citation131,Citation133 MSC-EVs can exert inhibitory effects on fibrosis. In addition, MSC-EVs can inhibit EMT processes and protect hepatocytes from apoptosis.Citation128,Citation129

The occurrence of EMT can lead to an increase in fibroblasts, while hepatocyte apoptosis is also a feature of liver fibrosis. MSC-EVs reduced EMT by upregulating markers such as E-cadherin. Some studies have indicated that the therapeutic effects of MSC-EVs are superior to MSC themselvesCitation131,Citation132 because EVs can avoid some of the problems of cell therapy, such as the risk of embolism. Surface-engineered EVs can improve targeting,Citation134 while sustained-release formulations can prolong the action time of EVs in target organs.Citation129 Furthermore, miRNAs enriched in MSC-EVs, such as miR-223 and miR-486,Citation109,Citation127,Citation133 are also involved in their anti-fibrotic mechanisms. These miRNAs can downregulate hepatic stellate cell activation-related genes to exert effects. miR-150-5p can inhibit hepatic stellate cell activation by targeting CXCL1.Citation135

Therefore, MSCs and their secreted EVs exert synergistic effects through multiple pathways, inhibiting activation of hepatic stellate cells and inflammatory cells, reducing EMT, protecting hepatocytes and other mechanisms to treat liver fibrosis.

(). This provides a theoretical basis for MSC-EVs to become a new extracellular therapy for liver fibrosis, but further research and clinical validation are still needed.

Figure 2 Mechanisms of mesenchymal stem cells in treating liver fibrosis. MSC-EVs can inhibit liver fibrosis by reducing inflammatory response, suppressing hepatic stellate cell activation and proliferation, and promoting hepatocyte regeneration.

Note: This figure is originally drawn by Figdraw platform (www.figdraw.com).
Figure 2 Mechanisms of mesenchymal stem cells in treating liver fibrosis. MSC-EVs can inhibit liver fibrosis by reducing inflammatory response, suppressing hepatic stellate cell activation and proliferation, and promoting hepatocyte regeneration.

MSC-EVs and HIRI

HIRI is a tissue injury and inflammatory response caused by hepatic ischemia (insufficient blood supply) and reperfusion (blood resupply). Liver surgery, liver transplantation, and shock often result in HIRI. Severe HIRI can result in liver failure and organ damage.Citation163 The intricate process of HIRI entails interactions between several cellular types and chemical communication channels. Ischemia impairs cellular activity and disturbs energy metabolism in the liver by depriving it of oxygen and nutrients.Citation164 Ischemia also leads to a decrease in intracellular ATP levels, resulting in elevated intracellular calcium ion levels, compromised mitochondrial function, and the development of oxidative stress.Citation165

When blood is being resupplied to the liver during reperfusion, several negative effects are associated with this process. Reperfusion worsens oxidative stress, which generates significant amounts of reactive oxygen radicals and inflammatory agents. These changes cause apoptosis, necrosis, and inflammatory responses, all of which contribute to tissue damage.Citation164 Oxygen radicals and inflammatory mediators cause lipid peroxidation of cell membranes, protein oxidation, and DNA damage, which worsen cellular damage and inflammatory responses.Citation165

Studies show that CD4+ T lymphocytes have a crucial function in initiating hepatic inflammatory reactions in response to HIRI. Following reperfusion, a notable increase is observed in the proliferation, infiltration, and aggregation of CD4+ T cells that have specificity for antigens in ischemia-affected tissues. CD154, a cell-surface protein weighing 32–39 kDa and belonging to the TNF superfamily, is abundantly expressed by activated CD4+ T cells. This protein can enhance the immune response, promote platelet production, and worsen hepatocellular injury by interacting with CD40/CD154. Low levels of CD154 are typically expressed during normal circumstances. CD4+ T cell activation leads to the continuous synthesis and expression of a significant level of CD154 on the cell surface. CD154 binds to CD40 on B cells, natural killer (NK) cells, dendritic cells, macrophages, basophils, and eosinophils, triggering the activation of downstream transcription factors that stimulate cytokine production.

Furthermore, CD154 is a protein that can undergo rapid degradation over a short period.Citation105 Hence, early therapeutic strategies are essential to regulate the manifestation of CD154 on hepatic CD4+ T cells to alleviate HIRI and decrease the complications and mortality in OLT. Thus, the hepatoprotective effects of MSC-EVs are attributed to the suppression of CD154 expression on CD4+ T cells in the liver. Additional mechanistic investigations revealed that chaperone proteins that include the TCP1 subunit 2 in MSC-EVs were moved to CD4+ T cells. These cells are responsible for controlling the calcium influx/NFAT1 signaling pathway, affecting the synthesis and expression of CD154.Citation123

Moreover, MSC-EVs can reduce serum aminotransferase levels, alleviate tissue necrosis, increase the number of Ki-67-positive hepatocytes, and inhibit the transcription of inflammation-related genes, demonstrating regenerative repair potential.Citation118,Citation124 Compared to unfractionated conditioned medium, the fractionated EVs secreted by MSCs have greater cytoprotective and restorative capacities.Citation118 In mouse IRI models, MSC-EVs can reduce apoptotic and caspase-3 positive cells, decrease inflammatory factor mRNA levels, increase hepatocyte viability, and inhibit oxidative stress and NF-ĸB activation, thereby suppressing inflammatory responses and cell apoptosis.Citation119,Citation120 This is related to the antioxidant enzyme MnSOD carried and delivered by MSC-EVs.Citation120 Exogenous MSC-EVs can transport IL-10 into the liver, be taken up by Kupffer cells, induce PTPN22 expression, polarize Kupffer cells towards an anti-inflammatory phenotype, and alleviate inflammation and liver injury.Citation121 Studies have demonstrated that miR-1246 originating from MSC-EVs interacts with GSK 3β in liver cells, significantly suppressing Wntl, Wnt 3a, and β-catenin expression. This interaction activates the Wnt/β-catenin signaling pathway and reduces the production of HIRI-induced TNF-α, IL-6, and IL-1β.

Consequently, this reduction in inflammation alleviates HIRI.Citation122 The exploration of employing MSC-EVs to enhance HIRI is presently in its preliminary stages, thus raising several unresolved inquiries. Further research is required to conduct comprehensive investigations into the mechanisms and lasting impacts of MSC-EVs in the treatment of HIRI.

Limitations of MSC-EVs and Improvements in Clinical Applications

Pluripotent stem cells can be employed to acquire MSC-EVs, which exhibit potential for clinical application. However, within a clinical setting, MSC-EVs are linked to specific disadvantages that require attention. Moreover, the methods for manufacturing MSC-EVs have not yet been well standardized. The exploitation of these resources in a clinical setting is impeded by the obstacles presented by their diversity and limited length of use. Different techniques of preparation produce various batches of MSC-EVs, each possessing unique features and functions. Therefore, it is crucial to create standardized preparation processes in order to ensure consistency and reliability when utilized for clinical purposes.

Significant challenges also lie in the concentration and purification of MSC-EVs. The commonly used method of isolating extracellular vesicles (EVs), ultracentrifugation, is ineffective for characterizing individual EVs. To address this issue, a number of innovations in EV isolation techniques are currently being employed. These include techniques such as fluorescent marking and subsequent analysis using high-resolution flow cytometry,Citation166 specialized flow cytometry,Citation167 and the use of laser tweezers and Raman spectroscopyCitation168 for quantitative and qualitative assessment. The purity and functionality of MSC-EVs may be affected by the presence of other cell types or impurities during the current techniques used for isolation and enrichment. Hence, it is imperative to improve the efficacies of the separation and enrichment techniques to enhance the purity of MSC-EVs.

Additionally, the long-term viability of MSC-EVs is also a matter of concern. Possible alterations in the functionalities of MSC-EVs may occur as a consequence of several conditions, including temperature, pH, and oxygen levels, in both in vitro and in vivo settings. These changes have the potential to lead to a reduction or complete cessation of their functions. Recent studies focus on delivering EVs locally using tissue-engineered substances, which could be a potential approach to enhancing their practical use. Polymer networks in hydrogels have a three-dimensional arrangement that enables them to absorb substantial quantities of water or biofluids.Citation169 Hydrogels exhibit biocompatibility and remarkable mechanical characteristics.Citation170

Furthermore, hydrogels can maintain EV release, boost their stability, and enhance their efficacy.Citation48 Thus, the identification of a suitable modifier may enhance the durability of MSC-EVs. Nevertheless, our understanding of the processes by which MSC-EVs exert their effects and their biological activities is limited. Notwithstanding this limitation, MSC-EVs are recognized for their anti-inflammatory, anti-fibrotic, and regeneration-promoting properties. Therefore, further investigation is necessary to further understand the roles of MSC-EVs in a therapeutic context.

Summary and Future Prospects

The utilization of MSCs is critical in the management of disorders that alter tissue function, as emerging data indicates that the compounds they produce have a therapeutic impact. MSC-EVs, being the primary constituent of MSC paracrine substances, can protect against different types of chronic liver ailments. The liver offers a range of cell types that can serve as recipient cells, including hepatic macrophages for exosomes.Citation171 Recently, small sample clinical studies have been conducted to explore the safety and initial effectiveness of exosomes in the treatment of cirrhosis (ChiCTR2300075676). Nevertheless, the evaluation of MSC-EVs in clinical studies remains limited, and a significant disparity exists before their utilization in a therapeutic environment. In clinical trials involving MSC-EVs, it is crucial to address the challenges of optimizing the culture conditions of MSC, establishing standardized protocols for extracting and identifying EVs, and determining the optimal disease states that could benefit from treatment.Citation172 Coupling gene editing tools or various biomaterials to tissue-specific extracellular vesicles may enable precise modulation of target genes to improve therapeutic efficacy.Citation173 For example, loading CRISPR-Cas9 ribonucleoproteins into extracellular vesicles derived from activated hepatic stellate cells enabled targeted delivery to liver tissue.Citation174 This extracellular vesicle-mediated gene editing system specifically accumulated in the liver in vivo and exhibited significant therapeutic potential in models of acute liver injury, chronic liver fibrosis and hepatocellular carcinoma.

In conclusion, MSC-EVs have great potential for treating liver disease. These problems will undoubtedly be answered as research on MSC and its EVs progresses, opening up exciting new therapeutic opportunities for the treatment of chronic liver diseases. In the future, we anticipate that MSC-EVs will be a useful therapeutic approach for treating liver disorders.

Author Contributions

All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

Disclosure

The authors declare that they have no competing interests.

Additional information

Funding

This review was supported by Jinhua Municipal Science and Technology Bureau Key Project (No. 2023-3-091) and Jinhua Municipal Central Hospital Science and Technology Project (No. JY2022-1-04).

References

  • Wu Y, Min J, Ge C, et al. Interleukin 22 in Liver Injury, Inflammation and Cancer. Int J Biol Sci. 2020;16(13):2405–2413. doi:10.7150/ijbs.38925
  • Carmona C, Claxton L, O’Brien A, Hebditch V. Cirrhosis in over 16s: assessment and management-updated summary of NICE guidance. BMJ. 2023;383:2598. doi:10.1136/bmj.p2598
  • Sobeh M, Hamza MS, Ashour ML, et al. A polyphenol-rich fraction from Eugenia uniflora exhibits antioxidant and hepatoprotective activities in vivo. Pharmaceuticals. 2020;13(5):84.
  • Comarmond C, Cacoub P, Saadoun D. Treatment of chronic hepatitis C-associated cryoglobulinemia vasculitis at the era of direct-acting antivirals. Therap Adv Gastroenterol. 2020;13:1756284820942617. doi:10.1177/1756284820942617
  • Udompap P, Kim D, Kim WR. Current and Future Burden of Chronic Nonmalignant Liver Disease. Clin Gastroenterol Hepatol. 2015;13(12):2031–2041. doi:10.1016/j.cgh.2015.08.015
  • Starkey Lewis P, Campana L, Aleksieva N, et al. Alternatively activated macrophages promote resolution of necrosis following acute liver injury. J Hepatol. 2020;73(2):349–360. doi:10.1016/j.jhep.2020.02.031
  • Caldez MJ, Bjorklund M, Kaldis P. Cell cycle regulation in NAFLD: when imbalanced metabolism limits cell division. Hepatol Int. 2020;14(4):463–474. doi:10.1007/s12072-020-10066-6
  • Minami T, Aoyagi K, Kawahara A, et al. Evaluation of the expression of bone marrow-derived mesenchymal stem cells and cancer-associated fibroblasts in the stroma of gastric cancer tissue. Ann Gastroenterol Surg. 2020;4(4):464–474. doi:10.1002/ags3.12347
  • Freedman BR, Mooney DJ. Biomaterials to Mimic and Heal Connective Tissues. Adv Mater. 2019;31(19):e1806695. doi:10.1002/adma.201806695
  • Gomzikova MO, Aimaletdinov AM, Bondar OV, et al. Immunosuppressive properties of cytochalasin B-induced membrane vesicles of mesenchymal stem cells: comparing with extracellular vesicles derived from mesenchymal stem cells. Sci Rep. 2020;10(1):10740. doi:10.1038/s41598-020-67563-9
  • Li Q, Chen X, Li J. Marrow-derived mesenchymal stem cells regulate the inflammatory response and repair alveolar type II epithelial cells in acute lung injury of rats. J Int Med Res. 2020;48(4):300060520909027. doi:10.1177/0300060520909027
  • Li Z, Gong X, Li D, Yang X, Shi Q, Ju X. Intratracheal Transplantation of Amnion-Derived Mesenchymal Stem Cells Ameliorates Hyperoxia-Induced Neonatal Hyperoxic Lung Injury via Aminoacyl-Peptide Hydrolase. Int J Stem Cells. 2020;13(2):221–236. doi:10.15283/ijsc19110
  • Ni Z, Zhou S, Li S, et al. Exosomes: roles and therapeutic potential in osteoarthritis. Bone Res. 2020;8:25. doi:10.1038/s41413-020-0100-9
  • Solis MA, I M, Correa R, Huang LLH. Stem cells as a potential therapy for diabetes mellitus: a call-to-action in Latin America. Diabetol Metab Syndr. 2019;11:20. doi:10.1186/s13098-019-0415-0
  • Takayama Y, Kusamori K, Katsurada Y, Obana S, Itakura S, Nishikawa M. Efficient delivery of mesenchymal stem/stromal cells to injured liver by surface PEGylation. Stem Cell Res Ther. 2023;14(1):216. doi:10.1186/s13287-023-03446-w
  • Karaahmet F, Kocaman SA. Endothelial progenitor cells and mesenchymal stem cells to overcome vascular deterioration and cytokine storm in critical patients with COVID-19. Med Hypotheses. 2020;144:109973. doi:10.1016/j.mehy.2020.109973
  • Seyedrazizadeh S-Z, Poosti S, Nazari A, et al. Extracellular vesicles derived from human ES-MSCs protect retinal ganglion cells and preserve retinal function in a rodent model of optic nerve injury. Stem Cell Res Ther. 2020;11(1):203. doi:10.1186/s13287-020-01702-x
  • Xu B, Yuan F-Z, Lin L, et al. The Higher Inherent Therapeutic Potential of Biomaterial-Based hDPSCs and hEnSCs for Pancreas Diseases. Front Bioeng Biotechnol. 2020;8:636. doi:10.3389/fbioe.2020.00636
  • Wang J, Fu X, Yan Y, et al. In vitro differentiation of rhesus macaque bone marrow- and adipose tissue-derived MSCs into hepatocyte-like cells. Exp Ther Med. 2020;20(1):251–260. doi:10.3892/etm.2020.8676
  • Mishra VK, Shih -H-H, Parveen F, et al. Identifying the Therapeutic Significance of Mesenchymal Stem Cells. Cells. 2020;9(5):1145.
  • Fan T, Qu R, Yu Q, et al. Bioinformatics analysis of the biological changes involved in the osteogenic differentiation of human mesenchymal stem cells. J Cell Mol Med. 2020;24(14):7968–7978. doi:10.1111/jcmm.15429
  • Allameh A, Ahmadi-Ashtiani HR, Maleki N. Glutathione-related inflammatory signature in hepatocytes differentiated from the progenitor mesenchymal stem cells. Heliyon. 2020;6(6):e04149. doi:10.1016/j.heliyon.2020.e04149
  • Yao Z, Liu H, Yang M, et al. Bone marrow mesenchymal stem cell-derived endothelial cells increase capillary density and accelerate angiogenesis in mouse hindlimb ischemia model. Stem Cell Res Ther. 2020;11(1):221. doi:10.1186/s13287-020-01710-x
  • Balbi C, Costa A, Barile L, Bollini S. Message in a Bottle: upgrading Cardiac Repair into Rejuvenation. Cells. 2020;9(3):548.
  • Zhang J, Xie B, Hashimoto K. Current status of potential therapeutic candidates for the COVID-19 crisis. Brain Behav Immun. 2020;87:59–73. doi:10.1016/j.bbi.2020.04.046
  • Krambs JR, Abou Ezzi G, Yao J-C, Link DC. Canonical signaling by TGF family members in mesenchymal stromal cells is dispensable for hematopoietic niche maintenance under basal and stress conditions. PLoS One. 2020;15(5):e0233751. doi:10.1371/journal.pone.0233751
  • Wang L, Zhang L, Liang X, et al. Adipose Tissue-Derived Stem Cells from Type 2 Diabetics Reveal Conservative Alterations in Multidimensional Characteristics. Int J Stem Cells. 2020;13(2):268–278. doi:10.15283/ijsc20028
  • Chen J, Li M, Liu A-Q, et al. Gli1+ Cells Couple with Type H Vessels and Are Required for Type H Vessel Formation. Stem Cell Reports. 2020;15(1):110–124. doi:10.1016/j.stemcr.2020.06.007
  • Parhizkar Roudsari P, Alavi-Moghadam S, Payab M, et al. Auxiliary role of mesenchymal stem cells as regenerative medicine soldiers to attenuate inflammatory processes of severe acute respiratory infections caused by COVID-19. Cell Tissue Bank. 2020;21(3):405–425. doi:10.1007/s10561-020-09842-3
  • Basiri A, Pazhouhnia Z, Beheshtizadeh N, Hoseinpour M, Saghazadeh A, Rezaei N. Regenerative Medicine in COVID-19 Treatment: real Opportunities and Range of Promises. Stem Cell Rev Rep. 2021;17(1):163–175. doi:10.1007/s12015-020-09994-5
  • Yu C, Peall IW, Pham SH, Okolicsanyi RK, Griffiths LR, Haupt LM. Syndecan-1 Facilitates the Human Mesenchymal Stem Cell Osteo-Adipogenic Balance. Int J Mol Sci. 2020;21(11):145.
  • Carvalheiro T, Zimmermann M, Radstake TRDJ, Marut W. Novel insights into dendritic cells in the pathogenesis of systemic sclerosis. Clin Exp Immunol. 2020;201(1):25–33. doi:10.1111/cei.13417
  • Fan D, Zeng M, Xia Q, et al. Efficacy and safety of umbilical cord mesenchymal stem cells in treatment of cesarean section skin scars: a randomized clinical trial. Stem Cell Res Ther. 2020;11(1):244. doi:10.1186/s13287-020-01695-7
  • van Best N, Rolle-Kampczyk U, Schaap FG, et al. Bile acids drive the newborn’s gut microbiota maturation. Nat Commun. 2020;11(1):3692. doi:10.1038/s41467-020-17183-8
  • Alqahtani SA, Schattenberg M. Liver injury in COVID-19: the current evidence. United Eur Gastroenterol J. 2020;8(5):509–519. doi:10.1177/2050640620924157
  • Furuta T, Furuya K, Zheng Y-W, Oda T. Novel alternative transplantation therapy for orthotopic liver transplantation in liver failure: a systematic review. World J Transplant. 2020;10(3):64–78. doi:10.5500/wjt.v10.i3.64
  • Petryk N, Shevchenko O. Mesenchymal Stem Cells Anti-Inflammatory Activity in Rats: proinflammatory Cytokines. J Inflamm Res. 2020;13:293–301. doi:10.2147/JIR.S256932
  • Lotfi M, Hamblin MR, Rezaei N. COVID-19: transmission, prevention, and potential therapeutic opportunities. Clin Chim Acta. 2020;508:254–266. doi:10.1016/j.cca.2020.05.044
  • Ullah M, Liu DD, Rai S, et al. Reversing Acute Kidney Injury Using Pulsed Focused Ultrasound and MSC Therapy: a Role for HSP-Mediated PI3K/AKT Signaling. Mol Ther Methods Clin Dev. 2020;17:683–694. doi:10.1016/j.omtm.2020.03.023
  • Cai J, Wu J, Wang J, et al. Extracellular vesicles derived from different sources of mesenchymal stem cells: therapeutic effects and translational potential. Cell Biosci. 2020;10:69. doi:10.1186/s13578-020-00427-x
  • Ikeda T, Nishita M, Hoshi K, Honda T, Kakeji Y, Minami Y. Mesenchymal stem cell-derived CXCL16 promotes progression of gastric cancer cells by STAT3-mediated expression of Ror1. Cancer Sci. 2020;111(4):1254–1265. doi:10.1111/cas.14339
  • Watanabe T, Tsuchiya A, Takeuchi S, et al. Development of a non-alcoholic steatohepatitis model with rapid accumulation of fibrosis, and its treatment using mesenchymal stem cells and their small extracellular vesicles. Regen Ther. 2020;14:252–261. doi:10.1016/j.reth.2020.03.012
  • Arnold P, Li W, et al. Joint Reconstituted Signaling of the IL-6 Receptor via Extracellular Vesicles. Cells. 2020;9(5):1307.
  • Krammer TL, Mayr M, Hackl M. microRNAs as promising biomarkers of platelet activity in antiplatelet therapy monitoring. Int J Mol Sci. 2020;21:254.
  • Shimizu A, Sawada K, Kimura T. Pathophysiological Role and Potential Therapeutic Exploitation of Exosomes in Ovarian Cancer. Cells. 2020;9(4):814.
  • Campos A, Leyton L, Quest AFG. Caveolin-1 function at the plasma membrane and in intracellular compartments in cancer. Cancer Metastasis Rev. 2020;39(2):435–453. doi:10.1007/s10555-020-09890-x
  • Seo Y, Kim H-S, Hong I-S. Stem Cell-Derived Extracellular Vesicles as Immunomodulatory Therapeutics. Stem Cells Int. 2019;2019:5126156. doi:10.1155/2019/5126156
  • Tsiapalis D, O’Driscoll L. Mesenchymal Stem Cell Derived Extracellular Vesicles for Tissue Engineering and Regenerative Medicine Applications. Cells. 2020;9(4):991.
  • Ryu J-S, Seo SY, Jeong E-J, et al. Ganglioside GM3 Up-Regulate Chondrogenic Differentiation by Transform Growth Factor Receptors. Int J Mol Sci. 2020;21(6):548.
  • Lee DB, Verstraete FJM, Arzi B. An Update on Feline Chronic Gingivostomatitis. Vet Clin North Am Small Anim Pract. 2020;50(5):973–982. doi:10.1016/j.cvsm.2020.04.002
  • Volleman TNE, Schol J, Morita K, Sakai D, Watanabe M. Wnt3a and wnt5a as Potential Chondrogenic Stimulators for Nucleus Pulposus Cell Induction: a Comprehensive Review. Neurospine. 2020;17(1):19–35. doi:10.14245/ns.2040040.020
  • Hutchings G, Janowicz K, Moncrieff L, et al. The Proliferation and Differentiation of Adipose-Derived Stem Cells in Neovascularization and Angiogenesis. Int J Mol Sci. 2020;21(11):991.
  • Bao X, Wang J, Zhou G, et al. Extended in vitro culture of primary human mesenchymal stem cells downregulates Brca1-related genes and impairs DNA double-strand break recognition. FEBS Open Bio. 2020;10(7):1238–1250. doi:10.1002/2211-5463.12867
  • Shi R, Lian W, Jin Y, et al. Role and effect of vein-transplanted human umbilical cord mesenchymal stem cells in the repair of diabetic foot ulcers in rats. Acta Biochim Biophys Sin (Shanghai). 2020;52(6):620–630. doi:10.1093/abbs/gmaa039
  • Gnecchi M, He H, Liang OD, et al. Paracrine action accounts for marked protection of ischemic heart by Akt-modified mesenchymal stem cells. Nat Med. 2005;11(4):367–368.
  • Gnecchi M, He H, Noiseux N, et al. Evidence supporting paracrine hypothesis for Akt-modified mesenchymal stem cell-mediated cardiac protection and functional improvement. FASEB J. 2006;20(6):661–669.
  • Okazaki T, Magaki T, Takeda M, et al. Intravenous administration of bone marrow stromal cells increases survivin and Bcl-2 protein expression and improves sensorimotor function following ischemia in rats. Neurosci Lett. 2008;430(2):109–114.
  • Wang S-P, Wang Z-H, Peng D-Y, S-M L, Wang H, Wang X-H. Therapeutic effect of mesenchymal stem cells in rats with intracerebral hemorrhage: reduced apoptosis and enhanced neuroprotection. Mol Med Rep. 2012;6(4):848–854. doi:10.3892/mmr.2012.997
  • Wei H, Li F, Xue T, et al. MicroRNA-122-functionalized DNA tetrahedron stimulate hepatic differentiation of human mesenchymal stem cells for acute liver failure therapy. Bioact Mater. 2023;28:50–60. doi:10.1016/j.bioactmat.2023.04.024
  • Hu C, Li L. In vitro culture of isolated primary hepatocytes and stem cell-derived hepatocyte-like cells for liver regeneration. Protein Cell. 2015;6(8):562–574. doi:10.1007/s13238-015-0180-2
  • Z-H L, Wang Y-L, Wang H-J, J-H W, Tan Y-Z. Rapamycin-Preactivated Autophagy Enhances Survival and Differentiation of Mesenchymal Stem Cells After Transplantation into Infarcted Myocardium. Stem Cell Rev Rep. 2020;16(2):344–356. doi:10.1007/s12015-020-09952-1
  • Zhang Y, Li Y, Li W, et al. Therapeutic Effect of Human Umbilical Cord Mesenchymal Stem Cells at Various Passages on Acute Liver Failure in Rats. Stem Cells Int. 2018;2018:7159465. doi:10.1155/2018/7159465
  • Zhou X, Jin N, Wang F, Chen B. Mesenchymal stem cells: a promising way in therapies of graft-versus-host disease. Cancer Cell Int. 2020;20:114. doi:10.1186/s12935-020-01193-z
  • Buono L, Scalabrin S, De Iuliis M, et al. Mesenchymal Stem Cell-Derived Extracellular Vesicles Protect Human Corneal Endothelial Cells from Endoplasmic Reticulum Stress-Mediated Apoptosis. Int J Mol Sci. 2021;22(9):4930.
  • Moayedfard Z, Sani F, Alizadeh A, Bagheri Lankarani K, Zarei M, Azarpira N. The role of the immune system in the pathogenesis of NAFLD and potential therapeutic impacts of mesenchymal stem cell-derived extracellular vesicles. Stem Cell Res Ther. 2022;13(1):242. doi:10.1186/s13287-022-02929-6
  • 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(1):17. doi:10.1186/s13287-018-0774-8
  • Wu R, Fan X, Wang Y, et al. Mesenchymal Stem Cell-Derived Extracellular Vesicles in Liver Immunity and Therapy. Front Immunol. 2022;13:833878. doi:10.3389/fimmu.2022.833878
  • Zhao M, Liu S, Wang C, et al. Mesenchymal Stem Cell-Derived Extracellular Vesicles Attenuate Mitochondrial Damage and Inflammation by Stabilizing Mitochondrial DNA. ACS Nano. 2021;15(1):1519–1538. doi:10.1021/acsnano.0c08947
  • Branscome H, Paul S, Yin D, et al. Use of Stem Cell Extracellular Vesicles as a “Holistic” Approach to CNS Repair. Front Cell Dev Biol. 2020;8:455. doi:10.3389/fcell.2020.00455
  • Yin L, Liu X, Shi Y, et al. Therapeutic Advances of Stem Cell-Derived Extracellular Vesicles in Regenerative Medicine. Cells. 2020;9(3):548.
  • Li Q, Huang Z, Wang Q, et al. Targeted immunomodulation therapy for cardiac repair by platelet membrane engineering extracellular vesicles via hitching peripheral monocytes. Biomaterials. 2022;284:121529. doi:10.1016/j.biomaterials.2022.121529
  • Aneesh A, Liu A, Moss HE, et al. Emerging concepts in the treatment of optic neuritis: mesenchymal stem cell-derived extracellular vesicles. Stem Cell Res Ther. 2021;12(1):594. doi:10.1186/s13287-021-02645-7
  • Fujii S, Miura Y. Immunomodulatory and Regenerative Effects of MSC-Derived Extracellular Vesicles to Treat Acute GVHD. Stem Cells. 2022;40(11):977–990. doi:10.1093/stmcls/sxac057
  • Harrell CR, Jovicic N, Djonov V, Volarevic V. Therapeutic Use of Mesenchymal Stem Cell-Derived Exosomes: from Basic Science to Clinics. Pharmaceutics. 2020;12(5):991.
  • Qiu L, Wang J, Chen M, Chen F, Tu W. Exosomal microRNA‑146a derived from mesenchymal stem cells increases the sensitivity of ovarian cancer cells to docetaxel and taxane via a LAMC2‑mediated PI3K/Akt axis. Int J Mol Med. 2020;46(2):609–620. doi:10.3892/ijmm.2020.4634
  • Wang M, Yu F, Li P, Wang K. Emerging Function and Clinical Significance of Exosomal circRNAs in Cancer. Mol Ther Nucleic Acids. 2020;21:367–383. doi:10.1016/j.omtn.2020.06.008
  • Gao S, Zhu H, Zuo X, Luo H. Cathepsin G and Its Role in Inflammation and Autoimmune Diseases. Arch Rheumatol. 2018;33(4):498–504. doi:10.5606/ArchRheumatol.2018.6595
  • Lu T, Zhang J, Cai J, et al. Extracellular vesicles derived from mesenchymal stromal cells as nanotherapeutics for liver ischaemia-reperfusion injury by transferring mitochondria to modulate the formation of neutrophil extracellular traps. Biomaterials. 2022;284:121486. doi:10.1016/j.biomaterials.2022.121486
  • Pan L-F, Niu Z-Q, Ren S, et al. Could extracellular vesicles derived from mesenchymal stem cells be a potential therapy for acute pancreatitis-induced cardiac injury? World J Stem Cells. 2023;15(7):654–664. doi:10.4252/wjsc.v15.i7.654
  • Qian X, An N, Ren Y, Yang C, Zhang X, Li L. Immunosuppressive Effects of Mesenchymal Stem Cells-derived Exosomes. Stem Cell Rev Rep. 2021;17(2):411–427. doi:10.1007/s12015-020-10040-7
  • Cha K-Y, Cho W, Park S, et al. Generation of bioactive MSC-EVs for bone tissue regeneration by tauroursodeoxycholic acid treatment. J Control Release. 2023;354:45–56. doi:10.1016/j.jconrel.2022.12.053
  • Pu Q, Xiu G, Sun J, Liu P, Ling B. Progress on the effect of mesenchymal stem cell derived exosomes on multiple organ dysfunction in sepsis. Zhonghua Wei Zhong Bing Ji Jiu Yi Xue. 2021;33(6):757–760. doi:10.3760/cma.j.cn121430-20200908-00620
  • Wang J-H, Liu X-L, Sun J-M, Yang J-H, D-H X, Yan -S-S. Role of mesenchymal stem cell derived extracellular vesicles in autoimmunity: a systematic review. World J Stem Cells. 2020;12(8):879–896. doi:10.4252/wjsc.v12.i8.879
  • Gupta S, Pinky V, et al. Comparative Evaluation of Anti-Fibrotic Effect of Tissue Specific Mesenchymal Stem Cells Derived Extracellular Vesicles for the Amelioration of CCl4 Induced Chronic Liver Injury. Stem Cell Rev Rep. 2022;18(3):1097–1112. doi:10.1007/s12015-021-10313-9
  • Angioni R, Liboni C, Herkenne S, et al. CD73+ extracellular vesicles inhibit angiogenesis through adenosine A2B receptor signalling. J Extracell Vesicles. 2020;9(1):1757900. doi:10.1080/20013078.2020.1757900
  • Xin D, Li T, Chu X, Ke H, Liu D, Wang Z. MSCs-extracellular vesicles attenuated neuroinflammation, synapse damage and microglial phagocytosis after hypoxia-ischemia injury by preventing osteopontin expression. Pharmacol Res. 2021;164:105322. doi:10.1016/j.phrs.2020.105322
  • Pu Y, Li C, Qi X, et al. Extracellular Vesicles from NMN Preconditioned Mesenchymal Stem Cells Ameliorated Myocardial Infarction via miR-210-3p Promoted Angiogenesis. Stem Cell Rev Rep. 2023;19(4):1051–1066. doi:10.1007/s12015-022-10499-6
  • Wu Y, Peng W, Fang M, Wu M, Wu M. MSCs-Derived Extracellular Vesicles Carrying miR-212-5p Alleviate Myocardial Infarction-Induced Cardiac Fibrosis via NLRC5/VEGF/TGF-β1/SMAD Axis. J Cardiovasc Transl Res. 2022;15(2):302–316. doi:10.1007/s12265-021-10156-2
  • Han M, Cao Y, Xue H, et al. Neuroprotective Effect of Mesenchymal Stromal Cell-Derived Extracellular Vesicles Against Cerebral Ischemia-Reperfusion-Induced Neural Functional Injury: a Pivotal Role for AMPK and JAK2/STAT3/NF-ΰB Signaling Pathway Modulation. Drug Des Devel Ther. 2020;14:2865–2876. doi:10.2147/DDDT.S248892
  • Gholami L, Nooshabadi VT, Shahabi S, et al. Extracellular vesicles in bone and periodontal regeneration: current and potential therapeutic applications. Cell Biosci. 2021;11(1):16. doi:10.1186/s13578-020-00527-8
  • Rezaie J, Nejati V, Mahmoodi M, Ahmadi M. Mesenchymal stem cells derived extracellular vesicles: a promising nanomedicine for drug delivery system. Biochem Pharmacol. 2022;203:115167. doi:10.1016/j.bcp.2022.115167
  • Gentile P, Sterodimas A. Adipose Stem Cells (ASCs) and Stromal Vascular Fraction (SVF) as a Potential Therapy in Combating (COVID-19)-Disease. Aging Dis. 2020;11(3):465–469. doi:10.14336/AD.2020.0422
  • Abreu H, Canciani E, Raineri D, Cappellano G, Rimondini L, Chiocchetti A. Extracellular Vesicles in Musculoskeletal Regeneration: modulating the Therapy of the Future. Cells. 2021;11(1).
  • Liao Z, Liu C, Wang L, Sui C, Zhang H. Therapeutic Role of Mesenchymal Stem Cell-Derived Extracellular Vesicles in Female Reproductive Diseases. Front Endocrinol (Lausanne). 2021;12:665645. doi:10.3389/fendo.2021.665645
  • Zhang J, Lu T, Xiao J, et al. MSC-derived extracellular vesicles as nanotherapeutics for promoting aged liver regeneration. J Control Release. 2023;356:402–415. doi:10.1016/j.jconrel.2023.02.032
  • Diniz AB, Antunes M, Lacerda V, et al. Imaging and immunometabolic phenotyping uncover changes in the hepatic immune response in the early phases of NAFLD. JHEP Rep. 2020;2(4):100117. doi:10.1016/j.jhepr.2020.100117
  • Kjã¦rgaard K, Sandahl TD, Frisch K, et al. Intravenous and oral copper kinetics, biodistribution and dosimetry in healthy humans studied by [64Cu]copper PET/CT. EJNMMI Radiopharm Chem. 2020;5(1):15. doi:10.1186/s41181-020-00100-1
  • Fahey S, Dempsey E, Long A. The role of chemokines in acute and chronic hepatitis C infection. Cell Mol Immunol. 2014;11(1):25–40. doi:10.1038/cmi.2013.37
  • Patel SR, Lundgren TS, Spencer HT, Doering CB. The Immune Response to the fVIII Gene Therapy in Preclinical Models. Front Immunol. 2020;11:494. doi:10.3389/fimmu.2020.00494
  • Gottwick C, Carambia A, Herkel J. Harnessing the liver to induce antigen-specific immune tolerance. Semin Immunopathol. 2022;44(4):475–484. doi:10.1007/s00281-022-00942-8
  • Lin L, Gong H, Li R, et al. Nanodrug with ROS and pH Dual-Sensitivity Ameliorates Liver Fibrosis via Multicellular Regulation. Adv Sci. 2020;7(7):1903138. doi:10.1002/advs.201903138
  • Horng J-H, Lin W-H, C-R W, et al. HBV X protein-based therapeutic vaccine accelerates viral antigen clearance by mobilizing monocyte infiltration into the liver in HBV carrier mice. J Biomed Sci. 2020;27(1):70. doi:10.1186/s12929-020-00662-x
  • Van Herck M, Vonghia L, Kwanten WJ, et al. Diet Reversal and Immune Modulation Show Key Role for Liver and Adipose Tissue T Cells in Murine Nonalcoholic Steatohepatitis. Cell Mol Gastroenterol Hepatol. 2020;10(3):467–490. doi:10.1016/j.jcmgh.2020.04.010
  • Asadipour M, Fazeli P, Zohouri M, et al. IL-18 in Blood Serum of Hepatitis C Patients Might be of Predictive Value for Individual Outcomes. Infect Disord Drug Targets. 2021;21(3):389–393. doi:10.2174/1871526520666200707113401
  • De Luca L, Trino S, Laurenzana I, et al. Mesenchymal Stem Cell Derived Extracellular Vesicles: a Role in Hematopoietic Transplantation? Int J Mol Sci. 2017;18(5):852.
  • Cai P, Mu Y, Olveda RM, Ross AG, Olveda DU, McManus DP. Serum Exosomal miRNAs for Grading Hepatic Fibrosis Due to Schistosomiasis. Int J Mol Sci. 2020;21:5548.
  • Morã¡n L, Cubero FJ. Extracellular vesicles in liver disease and beyond. World J Gastroenterol. 2018;24(40):4519–4526. doi:10.3748/wjg.v24.i40.4519
  • Chen L, F-B L, Chen D-Z, et al. BMSCs-derived miR-223-containing exosomes contribute to liver protection in experimental autoimmune hepatitis. Mol Immunol. 2018;93:38–46. doi:10.1016/j.molimm.2017.11.008
  • F-B L, Chen D-Z, Chen L, et al. Attenuation of Experimental Autoimmune Hepatitis in Mice with Bone Mesenchymal Stem Cell-Derived Exosomes Carrying MicroRNA-223-3p. Mol Cells. 2019;42(12):906–918. doi:10.14348/molcells.2019.2283
  • Tamura R, Uemoto S, Tabata Y. Immunosuppressive effect of mesenchymal stem cell-derived exosomes on a concanavalin A-induced liver injury model. Inflamm Regen. 2016;36:26. doi:10.1186/s41232-016-0030-5
  • Zhao J, Li Y, Jia R, Wang J, Shi M, Wang Y. Mesenchymal Stem Cells-Derived Exosomes as Dexamethasone Delivery Vehicles for Autoimmune Hepatitis Therapy. Front Bioeng Biotechnol. 2021;9:650376. doi:10.3389/fbioe.2021.650376
  • Chen L, Xiang B, Wang X, Xiang C. Exosomes derived from human menstrual blood-derived stem cells alleviate fulminant hepatic failure. Stem Cell Res Ther. 2017;8(1):9. doi:10.1186/s13287-016-0453-6
  • Haga H, Yan IK, Takahashi K, Matsuda A, Patel T. Extracellular Vesicles from Bone Marrow-Derived Mesenchymal Stem Cells Improve Survival from Lethal Hepatic Failure in Mice. Stem Cells Transl Med. 2017;6(4):1262–1272. doi:10.1002/sctm.16-0226
  • Jiang L, Zhang S, Hu H, et al. Exosomes derived from human umbilical cord mesenchymal stem cells alleviate acute liver failure by reducing the activity of the NLRP3 inflammasome in macrophages. Biochem Biophys Res Commun. 2019;508(3):735–741. doi:10.1016/j.bbrc.2018.11.189
  • Liu Y, Lou G, Li A, 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. doi:10.1016/j.ebiom.2018.08.054
  • Tan CY, Lai RC, Wong W, Dan YY, Lim S-K, Ho HK. Mesenchymal stem cell-derived exosomes promote hepatic regeneration in drug-induced liver injury models. Stem Cell Res Ther. 2014;5(3):76. doi:10.1186/scrt465
  • Zhang S, Jiang L, Hu H, 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. doi:10.1016/j.lfs.2020.117401
  • Damania A, Jaiman D, Teotia AK, Kumar A. Mesenchymal stromal cell-derived exosome-rich fractionated secretome confers a hepatoprotective effect in liver injury. Stem Cell Res Ther. 2018;9(1):31. doi:10.1186/s13287-017-0752-6
  • Haga H, Yan IK, Borrelli DA, et al. Extracellular vesicles from bone marrow-derived mesenchymal stem cells protect against murine hepatic ischemia/reperfusion injury. Liver Transpl. 2017;23(6):791–803. doi:10.1002/lt.24770
  • Yao J, Zheng J, Cai J, et al. Extracellular vesicles derived from human umbilical cord mesenchymal stem cells alleviate rat hepatic ischemia-reperfusion injury by suppressing oxidative stress and neutrophil inflammatory response. FASEB J. 2019;33(2):1695–1710. doi:10.1096/fj.201800131RR
  • Zhang Y, Zhang X, Zhang H, 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. doi:10.3389/fimmu.2021.811164
  • Xie K, Liu L, Chen J, Liu F. Exosomes derived from human umbilical cord blood mesenchymal stem cells improve hepatic ischemia reperfusion injury via delivering miR-1246. Cell Cycle. 2019;18(24):3491–3501. doi:10.1080/15384101.2019.1689480
  • Zheng J, Lu T, Zhou C, et al. Extracellular Vesicles Derived from Human Umbilical Cord Mesenchymal Stem Cells Protect Liver Ischemia/Reperfusion Injury by Reducing CD154 Expression on CD4+ T Cells via CCT2. Adv Sci. 2020;7(18):1903746. doi:10.1002/advs.201903746
  • Anger F, Camara M, Ellinger E, et al. Human Mesenchymal Stromal Cell-Derived Extracellular Vesicles Improve Liver Regeneration After Ischemia Reperfusion Injury in Mice. Stem Cells Dev. 2019;28(21):1451–1462. doi:10.1089/scd.2019.0085
  • Angioni R, Cal㬠B, Vigneswara V, et al. Administration of Human MSC-Derived Extracellular Vesicles for the Treatment of Primary Sclerosing Cholangitis: preclinical Data in MDR2 Knockout Mice. Int J Mol Sci. 2020;21(22):8874.
  • Han HS, Lee H, You D, et al. Human adipose stem cell-derived extracellular nanovesicles for treatment of chronic liver fibrosis. J Control Release. 2020;320:328–336. doi:10.1016/j.jconrel.2020.01.042
  • Kim J, Lee C, Shin Y, et al. sEVs from tonsil-derived mesenchymal stromal cells alleviate activation of hepatic stellate cells and liver fibrosis through miR-486-5p. Mol Ther. 2021;29(4):1471–1486. doi:10.1016/j.ymthe.2020.12.025
  • Li T, Yan Y, Wang B, et al. Exosomes derived from human umbilical cord mesenchymal stem cells alleviate liver fibrosis. Stem Cells Dev. 2013;22(6):845–854. doi:10.1089/scd.2012.0395
  • Mardpour S, Ghanian MH, Sadeghi-Abandansari H, et al. Hydrogel-Mediated Sustained Systemic Delivery of Mesenchymal Stem Cell-Derived Extracellular Vesicles Improves Hepatic Regeneration in Chronic Liver Failure. ACS Appl Mater Interfaces. 2019;11(41):37421–37433. doi:10.1021/acsami.9b10126
  • Ohara M, Ohnishi S, Hosono H, et al. Extracellular Vesicles from Amnion-Derived Mesenchymal Stem Cells Ameliorate Hepatic Inflammation and Fibrosis in Rats. Stem Cells Int. 2018;2018:3212643. doi:10.1155/2018/3212643
  • Rong X, Liu J, Yao X, Jiang T, Wang Y, Xie F. Human bone marrow mesenchymal stem cells-derived exosomes alleviate liver fibrosis through the Wnt/β-catenin pathway. Stem Cell Res Ther. 2019;10(1):98. doi:10.1186/s13287-019-1204-2
  • Rostom DM, Attia N, Khalifa HM, Abou Nazel MW, El Sabaawy EA. The Therapeutic Potential of Extracellular Vesicles Versus Mesenchymal Stem Cells in Liver Damage. Tissue Eng Regen Med. 2020;17(4):537–552. doi:10.1007/s13770-020-00267-3
  • Wang N, Li X, Zhong Z, 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(1):437. doi:10.1186/s12951-021-01138-2
  • You DG, Oh BH, Nguyen VQ, et al. Vitamin A-coupled stem cell-derived extracellular vesicles regulate the fibrotic cascade by targeting activated hepatic stellate cells in vivo. J Control Release. 2021;336:285–295. doi:10.1016/j.jconrel.2021.06.031
  • 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(2):701–715. doi:10.1111/jcmm.16119
  • Cheng L, Yu P, Li F, et al. Human umbilical cord-derived mesenchymal stem cell-exosomal miR-627-5p ameliorates non-alcoholic fatty liver disease by repressing FTO expression. Hum Cell. 2021;34(6):1697–1708. doi:10.1007/s13577-021-00593-1
  • Niu Q, Wang T, Wang Z, et al. Adipose-derived mesenchymal stem cell-secreted extracellular vesicles alleviate non-alcoholic fatty liver disease via delivering miR-223-3p. Adipocyte. 2022;11(1):572–587. doi:10.1080/21623945.2022.2098583
  • Engelmann C, Zhang IW. Mechanisms of immunity in acutely decompensated cirrhosis and acute-on-chronic liver failure. Liver Int. 2023. doi:10.1111/liv.15644
  • Yu Z, Li J, Ren Z, et al. Switching from Fatty Acid Oxidation to Glycolysis Improves the Outcome of Acute-On-Chronic Liver Failure. Adv Sci. 2020;7(7):1902996. doi:10.1002/advs.201902996
  • Cao P, Chen Q, Shi C, Wang L, Gong Z. Fusobacterium nucleatum promotes the development of acute liver failure by inhibiting the NAD+ salvage metabolic pathway. Gut Pathog. 2022;14(1):29. doi:10.1186/s13099-022-00503-2
  • Zhang S, Hou Y, Yang J, et al. Application of mesenchymal stem cell exosomes and their drug-loading systems in acute liver failure. J Cell Mol Med. 2020;24(13):7082–7093. doi:10.1111/jcmm.15290
  • Fang X, Gao F, Yao Q, et al. Pooled Analysis of Mesenchymal Stromal Cell-Derived Extracellular Vesicle Therapy for Liver Disease in Preclinical Models. J Pers Med. 2023;13(3):441.
  • Yan Y, Jiang W, Tan Y, et al. hucMSC Exosome-Derived GPX1 Is Required for the Recovery of Hepatic Oxidant Injury. Mol Ther. 2017;25(2):465–479. doi:10.1016/j.ymthe.2016.11.019
  • Dong X, Feng X, Liu J, et al. Characteristics of Intestinal Microecology during Mesenchymal Stem Cell-Based Therapy for Mouse Acute Liver Injury. Stem Cells Int. 2019;2019:2403793. doi:10.1155/2019/2403793
  • Heeren J, Scheja L. Metabolic-associated fatty liver disease and lipoprotein metabolism. Mol Metab. 2021;50:101238. doi:10.1016/j.molmet.2021.101238
  • Zhang J-B. Therapeutic Effect of Prolyl Endopeptidase Inhibitor in High-fat Diet-induced Metabolic Dysfunction-associated Fatty Liver Disease. J Clin Transl Hepatol. 2023;11(5):1035–1049. doi:10.14218/JCTH.2022.00110
  • Gao Z, Zhang C, Peng F, et al. Hypoxic mesenchymal stem cell-derived extracellular vesicles ameliorate renal fibrosis after ischemia-reperfusion injure by restoring CPT1A mediated fatty acid oxidation. Stem Cell Res Ther. 2022;13(1):191. doi:10.1186/s13287-022-02861-9
  • Wei S, Li A, Zhang L, Du M. GROWTH AND DEVELOPMENT SYMPOSIUM: STEM AND PROGENITOR CELLS IN ANIMAL GROWTH: long noncoding RNAs in adipogenesis and adipose development of meat animals12. J Anim Sci. 2019;97(6):2644–2657. doi:10.1093/jas/skz114
  • Grange C, Tritta S, Tapparo M, et al. Stem cell-derived extracellular vesicles inhibit and revert fibrosis progression in a mouse model of diabetic nephropathy. Sci Rep. 2019;9(1):4468. doi:10.1038/s41598-019-41100-9
  • Komori A. Recent updates on the management of autoimmune hepatitis. Clin Mol Hepatol. 2021;27(1):58–69. doi:10.3350/cmh.2020.0189
  • Covelli C, Sacchi D, Sarcognato S, et al. Pathology of autoimmune hepatitis. Pathologica. 2021;113(3):185–193. doi:10.32074/1591-951X-241
  • Chung Y, Rahim MN, Graham JJ, Zen Y, Heneghan MA. An update on the pharmacological management of autoimmune hepatitis. Expert Opin Pharmacother. 2021;22(11):1475–1488. doi:10.1080/14656566.2021.1895747
  • Martinon F, Mayor A, Tschopp J. The inflammasomes: guardians of the body. Annu Rev Immunol. 2009;27:229–265. doi:10.1146/annurev.immunol.021908.132715
  • Vanaja SK, Rathinam VAK, Fitzgerald KA. Mechanisms of inflammasome activation: recent advances and novel insights. Trends Cell Biol. 2015;25(5):308–315. doi:10.1016/j.tcb.2014.12.009
  • Liu X, Mi X, Wang Z, et al. Ginsenoside Rg3 promotes regression from hepatic fibrosis through reducing inflammation-mediated autophagy signaling pathway. Cell Death Dis. 2020;11(6):454. doi:10.1038/s41419-020-2597-7
  • Joseph J. Serum Marker Panels for Predicting Liver Fibrosis - An Update. Clin Biochem Rev. 2020;41(2):67–73. doi:10.33176/AACB-20-00002
  • Ponziani FR, Nicoletti A, Gasbarrini A, Pompili M. Diagnostic and therapeutic potential of the gut microbiota in patients with early hepatocellular carcinoma. Ther Adv Med Oncol. 2019;11:1758835919848184. doi:10.1177/1758835919848184
  • Qiu B-F, Zhang G-Q. Effect of the transdifferentiation of BECs into myofibroblasts on the pathogenesis of secondary cholestatic hepatic fibrosis. Exp Ther Med. 2019;17(4):2769–2776. doi:10.3892/etm.2019.7234
  • Bruno S, Pasquino C, Herrera Sanchez MB, et al. HLSC-Derived Extracellular Vesicles Attenuate Liver Fibrosis and Inflammation in a Murine Model of Non-alcoholic Steatohepatitis. Mol Ther. 2020;28(2):479–489. doi:10.1016/j.ymthe.2019.10.016
  • Zhou J, Lin Y, Kang X, Liu Z, Zhang W, Xu F. microRNA-186 in extracellular vesicles from bone marrow mesenchymal stem cells alleviates idiopathic pulmonary fibrosis via interaction with SOX4 and DKK1. Stem Cell Res Ther. 2021;12(1):96. doi:10.1186/s13287-020-02083-x
  • Wang B, Yao K, Huuskes BM, et al. Mesenchymal Stem Cells Deliver Exogenous MicroRNA-let7c via Exosomes to Attenuate Renal Fibrosis. Mol Ther. 2016;24(7):1290–1301. doi:10.1038/mt.2016.90
  • Wang S, Li L, Liu T, Jiang W, Hu X. miR-19a/19b-loaded exosomes in combination with mesenchymal stem cell transplantation in a preclinical model of myocardial infarction. Regener Med. 2020;15(6):1749–1759. doi:10.2217/rme-2019-0136
  • Guan Y, Yao W, Yi K, et al. Nanotheranostics for the Management of Hepatic Ischemia-Reperfusion Injury. Small. 2021;17(23):e2007727. doi:10.1002/smll.202007727
  • Lee HM, Kim T, Choi HJ, et al. Influence of intraoperative oxygen content on early postoperative graft dysfunction in living donor liver transplantation: a STROBE-compliant retrospective observational study. Medicine (Baltimore). 2020;99(21):e20339. doi:10.1097/MD.0000000000020339
  • Valdã©s S, Paredes SD, Garcã-A Carreras C, et al. S-Adenosylmethionine Decreases Bacterial Translocation, Proinflammatory Cytokines, Oxidative Stress and Apoptosis Markers in Hepatic Ischemia-Reperfusion Injury in Wistar Rats. Antioxidants (Basel). 2023;12(8)4930.
  • van der Vlist EJ, Stoorvogel W, Arkesteijn GJA, Wauben MHM. Fluorescent labeling of nano-sized vesicles released by cells and subsequent quantitative and qualitative analysis by high-resolution flow cytometry. Nat Protoc. 2012;7(7):1311–1326. doi:10.1038/nprot.2012.065
  • Pospichalova V, Svoboda J, Dave Z, et al. Simplified protocol for flow cytometry analysis of fluorescently labeled exosomes and microvesicles using dedicated flow cytometer. J Extracell Vesicles. 2015;4:25530. doi:10.3402/jev.v4.25530
  • Smith ZJ, Lee C, Rojalin T, et al. Single exosome study reveals subpopulations distributed among cell lines with variability related to membrane content. J Extracell Vesicles. 2015;4:28533. doi:10.3402/jev.v4.28533
  • Hoffman AS. Hydrogels for biomedical applications. Ann N Y Acad Sci. 2001;944:62–73.
  • Xu N, Xu J, Zheng X, Hui J. Preparation of Injectable Composite Hydrogels by Blending Poloxamers with Calcium Carbonate-Crosslinked Sodium Alginate. ChemistryOpen. 2020;9(4):451–458. doi:10.1002/open.202000040
  • Okusha Y, Eguchi T, Tran MT, et al. Extracellular Vesicles Enriched with Moonlighting Metalloproteinase Are Highly Transmissive, Pro-Tumorigenic, and Trans-Activates Cellular Communication Network Factor (CCN2/CTGF): CRISPR against Cancer. Cancers (Basel). 2020;12(4):548.
  • Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science. 2020;367:6478. doi:10.1126/science.aau6977
  • Li F, Zhang J, Yi K, et al. Delivery of Stem Cell Secretome for Therapeutic Applications. ACS Appl Bio Mater. 2022;5(5):2009–2030. doi:10.1021/acsabm.1c01312
  • Wan T, Zhong J, Pan Q, Zhou T, Ping Y, Liu X. Exosome-mediated delivery of Cas9 ribonucleoprotein complexes for tissue-specific gene therapy of liver diseases. Sci Adv. 2022;8(37):eabp9435. doi:10.1126/sciadv.abp9435