Next Article in Journal
Deciphering Differential Life Stage Radioinduced Reproductive Decline in Caenorhabditis elegans through Lipid Analysis
Next Article in Special Issue
Three-Dimensional Osteogenic Differentiation of Bone Marrow Mesenchymal Stem Cells Promotes Matrix Metallopeptidase 13 (MMP13) Expression in Type I Collagen Hydrogels
Previous Article in Journal
Tumor Necrosis Factor’s Pathway in Crohn’s Disease: Potential for Intervention
Previous Article in Special Issue
Immunomodulatory Properties of Umbilical Cord Blood-Derived Small Extracellular Vesicles and Their Therapeutic Potential for Inflammatory Skin Disorders
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cell-Based Regeneration and Treatment of Liver Diseases

1
Department of Visceral, Transplant and Thoracic Surgery (VTT), Center of Operative Medicine, Daniel Swarovski Research Laboratory (DSL) and organLife Laboratory, Medical University of Innsbruck (MUI), 6020 Innsbruck, Austria
2
Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(19), 10276; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms221910276
Submission received: 19 August 2021 / Revised: 13 September 2021 / Accepted: 23 September 2021 / Published: 24 September 2021
(This article belongs to the Special Issue Stem Cell Activation in Adult Organism 2.0)

Abstract

:
The liver, in combination with a functional biliary system, is responsible for maintaining a great number of vital body functions. However, acute and chronic liver diseases may lead to irreversible liver damage and, ultimately, liver failure. At the moment, the best curative option for patients suffering from end-stage liver disease is liver transplantation. However, the number of donor livers required by far surpasses the supply, leading to a significant organ shortage. Cellular therapies play an increasing role in the restoration of organ function and can be integrated into organ transplantation protocols. Different types and sources of stem cells are considered for this purpose, but highly specific immune cells are also the focus of attention when developing individualized therapies. In-depth knowledge of the underlying mechanisms governing cell differentiation and engraftment is crucial for clinical implementation. Additionally, novel technologies such as ex vivo machine perfusion and recent developments in tissue engineering may hold promising potential for the implementation of cell-based therapies to restore proper organ function.

1. Introduction

The liver is the largest parenchymal organ, which controls a multitude of metabolic functions, including the homeostasis of carbohydrates, lipids, and proteins. It is the primary detoxifying organ [1] and is essential for a proper immune response against pathogens and the further recruitment of innate and adaptive immune cells [1,2]. This can be facilitated by low pressure and flow internal to the sinusoids [3]. Since abundant gut-derived pathogens and exogenous non-pathogenic molecules continuously enter the liver, several mechanisms contribute to prevent unexpected immune responses, establishing a balance between defense and the maintenance of self-tolerance [4,5,6]. The unique histoarchitecture and interactions of the different cell types are responsible for these very specific liver functions.
Despite significant achievements in surgical and pharmacological treatment options, liver diseases, in general, remain a leading cause of death. Severe liver disease resulting in end-stage organ failure may be caused by viral infections, cancer, or genetic predisposition. For many patients, the only treatment option is orthoptic liver transplantation (OLT) [7]. However, OLT is limited by a significant organ shortage, and OLT may not be suitable for specific diseases.
In this review, we first overview the mechanisms underlying the self-regenerating potential of the liver since the development of novel cell-based treatments relies on understanding these processes. Stem-cell-related approaches and cellular immunotherapies are then discussed for the treatment of liver diseases, taking into account ongoing clinical trials and current applications. We further highlight the importance of the chosen route of administration of these therapies and discuss arising novel technologies, which may be advantageous when introducing such innovative treatment protocols.

2. Liver Cell Types and Their Potential of Regeneration

2.1. Microanatomy of the Liver

Morphologically, the roughly hexagonal-shaped liver lobes are arranged around the sinusoids, harboring different cells (Figure 1). Every cell type of the liver is characterized by defined responsibilities, which collectively warrant an intact hepatic function at various levels [8]. Parenchymal cells account for 60–80% of the total liver mass. Hepatocytes are the most abundant type within this group, which are organized in cords lining the sinusoids. They play a central role in maintaining organ homeostasis by regulating physiological processes and metabolic activity linked to digestion, including the metabolization of carbohydrates and lipids and the production of bile. The majority of the remaining non-parenchymal cell population (NPC, 20–40% of the total liver mass) is composed of liver sinusoidal endothelial cells (LSECs, 10–20%) that form a permeable barrier between sinusoidal vessels and hepatocytes. Biliary epithelial cells (BECs), which constitute 5% of the total liver mass, will be described in detail in Section 2.1 [9]. Kupffer cells (KCs), also known as intravascular resident macrophages, account for 4–8%. They are located within the sinusoids and primarily perform immunological tasks. Hepatic stellate cells (SCs, 0.2–0.4%) are mesenchymal cells residing in the space of Disse, which is referred to as the space between LSECs and hepatic epithelial cells. SCs remain in a quiescent state; however, when triggered by liver damage, they become activated [1,2]. Moreover, circulating lymphocytes, consisting of T-cells (CD4+/CD8+), B-cells, natural killer (NK) cells, and NKT cells, are present. They interfere with antigen-presenting cells such as LSECs, KCs, and liver resident dendritic cells (DCs) or directly with hepatocytes, enabling adaptions of the immune response [2,10,11].

2.2. Biliary Epithelial Cells—The Lining of the Biliary Tree

The biliary tree is lined by BECs—also known as cholangiocytes [9,13,14,15]. With the enlargement of the bile ducts, the epithelial lining changes: BECs become larger in size, and their architecture changes from cuboidal to columnar. The common bile duct is lined by columnar epithelium with mucus-secreting cells [9,13,14,15].
BECs have several different functions: one of the BECs’ main functions is bile modification and transport. As mentioned above, bile is mainly produced by hepatocytes, although BECs can contribute to the production of the daily bile volume to a limited extent [16,17]. The bile is secreted into bile canaliculi, which are tubules formed between adjacent hepatocytes. They drain into the bile ducts, where the bile is further modified by BECs [1]. This entails a secretion of water and electrolytes (such as HCO3 and Cl) as well as the uptake of bile acids, glucose, and amino acids [18,19,20,21,22,23,24]. The secretion of HCO3- by BECs and the resulting alkaline environment is known as the “biliary HCO3-umbrella”. The HCO3 umbrella by the deprotonation of hydrophobic bile salt protects BECs from hydrophobic bile salt toxicity [25,26].

2.3. Mechanisms of Liver Cell Regeneration

Since a functioning liver is essential to maintaining body homeostasis, several compensatory regenerative mechanisms exist. Most experimental evidence of liver regeneration is based on animal models of partial hepatectomy. In addition, drug-induced acute liver injury has also been investigated in clinically relevant models of centrilobular liver necrosis and sterile inflammation, such as carbon tetrachloride, thioacetamide, or paracetamol overdose [27]. Both hepatocytes and BECs have been described to proliferate upon acute liver diseases. However, it has been observed that the ability of proliferation in hepatocytes gets lost in chronic diseases, which may also account for NPCs [28]. The molecular mechanism of liver regeneration was extensively reviewed recently [29,30]. Here, we focus on the fundamental mechanisms of liver regenerative potential based on hepatocytes and BECs.
Hepatocytes play a central role in liver regeneration. In this process, several transcription factors trigger mature hepatocytes to re-enter the cell cycle by progressing from the G1 to S phase [31] and, in turn, producing mitogenic growth factors to attract further hepatocytes and other cell types to start proliferating. Such molecules are the vascular endothelial growth factor (VEGF), angiopoietins 1 and 2 (mitogenic for LSECs), transforming growth factor alpha (TGFα) (mitogenic for endothelial cells, LSECs, and SCs), fibroblast growth factor 1 (FGF1) and FGF2 (mitogenic for SCs and LSECs), and the granulocyte–macrophage colony-stimulating factor (GM-CSF) (mitogenic for KCs) [28,32]. Hepatocyte proliferation is mainly controlled by extracellular signals through the tyrosine-protein kinase mesenchymal–epithelial transition factor (MET), which is the hepatocyte growth factor (HGF receptor), and EGFR (epidermal growth factor receptor). Upon hepatectomy, portal venous blood is a major initiator of liver regeneration by the increased shear stress on LSECs and by carrying signaling molecules like EGF, bile acids, and insulin [29,33]. Very early upon partial hepatectomy, the activation of metalloproteinases by the urokinase-type plasminogen activator [34,35] leads to the breakdown of some extracellular matrix components. Most importantly, extrahepatic produced HGF is liberated and activated, soon released in the peripheral blood as well, and provides mitogenic signaling [36]. Quite importantly, HGF or EGFR ligands (EGF, TGFα, heparin-binding EGF-like growth factor, and amphiregulin) induce hepatocyte proliferation in rodents if injected intravenously and in vitro [37]. In addition to HGF, interleukin-6 (IL6), tumor necrosis factor alpha (TNFα), bile acids, noradrenaline, leptin, and serotonin are soon released in the peripheral blood as well, all of which are known to be associated with liver regeneration, termed as auxiliary mitogens. In a uniquely concerted way, only a fraction of hepatocytes will undergo partial dedifferentiation at once, protecting the liver from acute liver failure by sustaining physiologic hepatocyte function in the remaining population [38].
The second key player for the self-regenerating potential of the liver is BECs, which are known as a highly plastic cell population [39]. In rodent models of impaired hepatocyte proliferation or loss of a large amount of liver mass, BECs have been shown to contribute to hepatocyte regeneration by differentiation into hepatocytes [40,41,42]. The plasticity of BECs is facilitated by intermediate transition through bipotential BECs, which have been suggested to be found mainly in the canals of Hering. Bipotential BECs expand during the so-called ductular reaction, which is mainly an expansion of cells of biliary origin observed in chronic liver disease, and can differentiate into both hepatocytes and BECs to enhance liver regeneration [11,43,44,45,46]. Bipotential BECs require NOTCH signaling to differentiate into BECs, while bipotential BEC to hepatocyte differentiation is promoted by Wnt signaling [47,48].
Different molecular pathways such as ten–eleven translocation 1 (TET1, a hydroxymethylcytosine deaminase) mediate changes in the DNA-methylome of BECs, which have been described to be crucial for this cell population to acquire their cellular plasticity [49]. One of the TET1 targets is the hippo-yes-associated protein (YAP)/PDZ-binding motif (TAZ) pathway [49]. YAP has likewise been identified as an important factor for BEC plasticity [39]. YAP mediates BEC self-renewal in liver homeostasis [39,50] and can also be induced by BEC exposure to bile acids [39,50]. This mechanism is hypothesized to contribute to BEC regeneration following acute injury or after partial hepatectomy in rodent models, which is associated with increased bile and bile acid secretion [51,52]. Furthermore, YAP-related upregulation of the biliary marker SRY-Box transcription factor 9 (Sox9) in hepatocytes can initiate hepatocyte-to-BEC differentiation [53,54,55,56].
Taken together, the regenerative potential of acute liver injury or disease is mainly due to hepatocytes re-entering the cell cycle. However, in chronic liver injury, the majority of hepatocytes are senescent [30]; this demands alternative therapy options.

3. Cellular Therapies for Treatment of Liver Diseases and Induction of Regeneration

The primary malignancies and underlying circumstances that ultimately lead to liver failure are varied, requiring different therapy strategies. The transplantation of hepatocytes and BECs has been investigated for a variety of diseases, with the most promising results observed for metabolic disorders [57]. To generate those cells in vitro, stem cells are considered the ideal source. They are characterized by their ability of self-renewal and the potential of differentiation into multiple lineages. However, the pluripotency and, thus, the ability to form teratomas in vivo is a major concern and reason why the direct application or transplantation of stem cells has not been clinically approved yet [58]. It can be distinguished by autologous and allogeneic approaches. In the first-mentioned approach, the patient’s own cells are utilized, whereas, in the latter, donor cells are obtained and transferred to the recipient. Thus, allogeneic cell transplantation possesses the potential of immunogenicity. The most frequently used types are embryonic stem cells (ESCs), hematopoietic stem cells (HSCs), induced pluripotent stem cells (iPSCs), and mesenchymal stem cells (MSCs), whereas recent studies have mainly focused on the last two mentioned cells (Figure 2) [59,60]. On the other hand, resident cells of the liver can be directly targeted, whereas immune cells are of special interest within this approach. Especially in cancer, these therapies are utilized for autoimmune diseases and also after transplantation [2,4,5].

3.1. Stem Cells: Sources, Mechanism of Action, and Applications

3.1.1. Embryonal Stem Cells (ESCs)

Pluripotent embryonic stem cells possess an unlimited differentiation capacity and can be obtained in the early stages of embryonic development from blastocyst inner cell mass [61,62]. They can be utilized to derive hepatocytes in a four-step protocol following their natural path of development in vivo, which can be monitored through the expression of appropriate gene signatures. Differentiation is driven by interactions with components of several signaling cascades (e.g., activin A, FGF, bone morphogenetic protein 4 (BMP4), HGF) [63,64]. The therapeutic effect of such in-vitro-derived hepatocytes has been investigated in various studies. For example, ESC-derived hepatocytes led to the rescue of acute liver failure induced by acetaminophen in an experimental murine model [61,64]. Moreover, it has been observed that ESC-derived hepatocyte-like cells transport regeneration-promoting trophic factors promoting liver regeneration, and are involved in cell replacement [62]. ESCs may further be used for treatment of fibrosis caused by chronic liver injury. In this mean, the effect of ESC-derived macrophages was tested in a murine model of carbon tetrachloride (CCL4)-induced hepatic injury, which revealed a significant reduction of fibrosis. Simultaneously, fibrogenic myofibroblasts were downregulated, progenitor cells of the liver were activated at the site of injury, and KCs were repopulated. These results give a first insight into the therapeutical usability of ESC-derived macrophages [65]. Besides the application of ESC-derived cell types directly into the liver, the research field that aims to construct liver organoids has gained importance. Liver organoids could serve as an improved model to investigate various liver diseases and develop novel organoid-based therapies [66,67,68,69]. However, its clinical use is associated with ethical concerns due to the deviation procedure of ESCs.

3.1.2. Hematopoietic Stem Cells (HSCs)

Multipotent hematopoietic stem cells are derived from bone marrow, peripheral blood, or umbilical cord blood. In standard isolation protocols, the expression of the surface marker CD34+ is used, although it is well known that some of these populations may lack CD34+. The migration of the HSCs towards the liver is regulated by the interactions of various chemokines, cytokines, and proteolytic enzymes. In a mouse model, stromal-cell-derived factor 1 (SDF1), matrix metallopeptidase 9 (MMP9), and HGF have been reported to mediate homing to the site of injury [70]. Thereby, adhesion has been reported to be dependent on CD44 and is modulated by C-X-C motif chemokine receptor 3 (CXCR3) and CXCR4 [71]. Moreover, HSCs primed with granulocyte colony-stimulating factor (G-CSF) have been shown to not only selectively migrate to the site of injury but also to induce hepatocyte formation [62,72]. However, the underlying mechanism of differentiation of HSCs is not truly understood [73]. Khurana et al. demonstrated in a cell culture model that one driver is hepatocyte nuclear factor−4α (HNF4α) [74]. In further in-vitro studies, HGF [75,76], FGF2, and FGF4 [75] have also been identified as key players. On the other hand, several studies support the theory of cell fusion rather than differentiation [77,78,79]. Nevertheless, in animal but also clinical studies, reasonable results could be achieved. In a fibrotic mouse model induced by CCL4, the expression of MMP9 and MMP14, which are involved in the degradation of collagen fibers, resulting in a reduction of fibrosis, was observed. Moreover, MMP9 secrets HSC differentiation and proliferation stimulating factors (e.g., c-Kit) to reduce inflammatory reactions and also triggers movement towards the afflicted tissue [80]. In a clinical trial, 48 patients suffering from end-stage liver disease, either from hepatitis C (HCV) infection or due to autoimmunity, received injections of autologous CD34+ HSCs. Liver function significantly improved according to serum levels of albumin, the international normalized ratio (INR), and transaminases during 12 months of follow-up [81]. In a larger study including 90 patients, the effect was even enhanced by repeated CD34+ HSCs injections. A linear enhancement of the effect was reported during the 12-month follow-up, suggesting engraftment of the cells; however, the study lacked histological proof [82].
Figure 2. Different sources of stem cells and their mechanism of differentiation for cell-based therapies. ESCs (A) are isolated from the early blastocyst [61]. The sources of HSCs (B) and MSCs (C) include placenta, umbilical cord, bone marrow, and peripheral blood; for MSC, additionally, dental pulp, muscle, and adipose tissue can be utilized [60,83]. iPSCs (D) are derived from somatic cells by a reprogramming step. Triggered by different transcription and growth factors, all of them can be differentiated into hepatocytes.
Figure 2. Different sources of stem cells and their mechanism of differentiation for cell-based therapies. ESCs (A) are isolated from the early blastocyst [61]. The sources of HSCs (B) and MSCs (C) include placenta, umbilical cord, bone marrow, and peripheral blood; for MSC, additionally, dental pulp, muscle, and adipose tissue can be utilized [60,83]. iPSCs (D) are derived from somatic cells by a reprogramming step. Triggered by different transcription and growth factors, all of them can be differentiated into hepatocytes.
Ijms 22 10276 g002

3.1.3. Induced Pluripotent Stem Cells (iPSCs)

A special feature of iPSCs is their production by reprogramming adult somatic cells back to a pluripotent state. iPSCs may then be utilized to produce various cell types, including hepatocytes and BECs. To generate functional hepatocytes, phase-specific markers, growth factors, cytokines, and chemicals are necessary within a three-step transformation from human iPSCs to endodermal cells, hepatoblast-like cells (HLC), and, finally, differentiated hepatocyte-like cells. The majority of protocols include the activation of Activin A for endoderm differentiation. As a next step, treatment with BMP and FGF2 induces differentiation into HLCs, and, finally, HGF and oncostatin M (OSM) trigger maturation. Such hepatocyte-like cells are defined by the expression of hepatocyte-specific markers and, for example, are used to treat end-stage liver disease by repopulating damaged liver tissue [62,84]. In a murine model for cell replacement therapy, the therapeutic potential of transplanted iPSC-derived HLCs was examined in carbon-tetrachloride-induced liver injuries. The created hepatic lineage expressed hepatocyte-specific markers, stored glycogen and lipids, and stimulated metabolic activity. One week after transplantation, the incorporation and functional integration of HLCs into the liver was evident, leading to enhanced albumin serum levels and decreased levels of lactate dehydrogenase and bilirubin. Further to this, these mice showed improved survival and slightly reduced fibrosis during a period of 5 weeks after transplantation compared to untreated controls [84]. In another mouse study, hepatocyte-like cells integrated with an albumin reporter (referred to as iHep) produced from iPSCs were transplanted into fibrotic livers, which resulted in beneficial effects including a decrease in thioacetamide-induced fibrosis, liver cell apoptosis, and pro-inflammatory factors and improved liver function. Moreover, the number of proliferating hepatocytes and iHeps expressing albumin was elevated following transplantation, demonstrating their potential to promote regenerative processes [85]. Besides the creation of HLCs, human iPSCs are also used to generate mature KCs. Therefore, iPSCs are differentiated into macrophage precursors, which are then used to generate the induced KCs. These cells mostly displayed identical gene expression patterns and functional properties compared to primary adult human KCs. At the current stage of research, induced KCs are used in different co-culturing models (e.g., with hepatocytes, macrophage precursors, or iHeps) in vitro to study inflammation, test for hepatoxicity, or examine cholestasis formation [86]. In cell-free therapies, iPSCs are used for the production of EVs. In a murine model of liver injury, the injection of EVs displayed anti-fibrotic effects by reducing profibrogenic factors or inhibiting the profibrogenic activity of human stellate cells. These results indicate that iPSC-derived EVs may exert therapeutic potential for the treatment of chronic liver diseases where the formation of fibrosis is the main feature [87].

3.1.4. Mesenchymal Stem Cells (MSCs)

MSCs are multipotent stem cells and must fulfill at least the following criteria stated by the Society for Cellular Therapy [88]. First, MSCs must be plastic-adherent when maintained in standard culture conditions. Second, MSCs must express CD105, CD73, and CD90 and lack expression of CD45, CD34, CD14 or CD11b, CD79α or CD19 and HLA-DR surface molecules. Third, MSCs must differentiate into osteoblasts, adipocytes, and chondroblasts in vitro. They are further known for their high differentiation potential and low immunogenicity and can be isolated of different tissues, including bone marrow (bm), adipose tissue, muscle, peripheral blood, placenta, umbilical cord (uc), and dental pulp (dp) [83]. Differentiation into both hepatocytes and BECs has been described using different protocols in vitro with the use of specific factors such as HGF, FGF4, insulin-transferrin-sodium selenite (ITS), and insulin growth factor 1 (IGF1) [89,90,91]. However, the differentiation was not limited to in vitro studies. In a mouse model of biliary injury, the administration of bmMSCs led to the restoration of the injury through delta ligand-like 4 (DLL4) activation, which enhanced BEC differentiation [92]. Iwanaka et al. injected dpMSCs into CCL4-induced chronic liver fibrotic mice, which induced tissue regeneration due to in vivo differentiation [93].
Besides the differentiation into hepatocytes and BECs for tissue replacement, the regenerative effect of MSCs may mainly underlie paracrine effects. Upon administration, MSCs migrate to the injured tissue area and secrete growth factors related to liver regeneration, which, in turn, stimulate cell proliferation, enhance angiogenesis, and restrain apoptosis [62,94,95]. Furthermore, MSCs promote liver regeneration by immunomodulation due to the release of soluble factors (e.g., nitric oxide (NO), prostaglandin E2 (PGE2), IL6, or IL10). This, in turn, leads to the downregulation of T-cells, the inhibition of B-cells, or the advantageous modulation of liver injury, hence improving inflammatory macrophages. Additionally, MSCs possess immunosuppressive properties regulated by cytokines (e.g., IL1α, TNFα, or interferon gamma (IFNγ)) and interactions with chemokines (e.g., XCR4/5/7) [96,97,98]. Moreover, upon the transplantation of MSCs, anti-fibrotic effects have been reported. Quiescent SCs can differentiate into fibrogenic myofibroblasts in chronic liver injury, but due to the secretion of several trophic factors such as TNFα and IL10, collagen synthesis is attenuated. On the other hand, HGF and NGF induce the apoptosis of SCs [97,99]. Ultimately, anti-inflammatory reactions are also promoted by MSCs, which may reduce hepatocyte damage [97,98]. In a clinical trial of 43 patients suffering from acute-on-chronic liver failure caused by hepatitis B (HBV) infection, all 24 patients in the test group treated with ucMSCs showed an enhanced survival rate, increased albumin secretion, and reduced transaminases levels compared to the control group [100]. Analogously, favorable results have also been reported for ucMSCs in a clinical study on primary biliary cirrhosis [101]. Recently, MSCs are a prominent tool to create EVs such as apoptotic bodies or exosomes in cell-free therapies. Specifically, cell-type-specific exosomes containing bioactive molecules such as various RNA types, other nucleic acids, proteins, and lipids are widely used in therapeutical trials due to their advantageous functions (e.g., involvement in drug delivery, immune regulation, tissue repair/regeneration, angiogenesis, cell proliferation, collagen reduction, or metabolic activities). Current studies mainly focus on the therapeutic effects of bm- or uc-derived MSC-EVs on acute liver failure, liver fibrosis, and hepatocellular carcinoma treatment using cell culture or experimental models. Thus, further studies are needed to assess their applicability in humans [98,102].
Taken together, stem cells as a source for hepatocytes and various progenitors, which can be utilized for subsequent transplantation, show great potential. Numerous clinical trials on hepatocyte transplantation have been conducted or are ongoing currently; however, no treatment has been clinically approved to date. The majority of these studies have reported promising short time efficacies, but they could not maintain the efficacy long term [57]. The indications for hepatocyte transplantation include acute and acute-on-chronic liver failure [103]. Still, the majority of reports are on inherited metabolic disorders, including different types of urea cycle defects [104,105,106,107,108] and Crigler-Najjar syndrome [109,110,111]. However, randomized trials are lacking; thus, it is difficult to conclude on the efficacy [112]. On the other hand, stem cells may also be administered in an undifferentiated manner to exert their effect. For this purpose, MSCs are considered the most promising candidate since numerous clinical trials are currently ongoing. Recently, Smets et al. reported a safe and effective treatment of MSC therapy in 14 pediatric patients suffering from metabolic disorders [113]. They utilized a specifically manufactured cell suspension called HepaStem, which is also currently investigated for patients with acute-on-chronic liver failure (NCT04229901). In general, MSC-based therapies are considered safe, but there are also reports of side effects [102,114], indicating the need for further studies. Another challenge in the context of stem cell therapies, in general, is to enable cell engraftment, which has been reported to be rather poor in some studies [57,103].

3.2. Immune Cell Therapies: A Novel Paradigm in the Development of Therapies

3.2.1. Regulatory T-Cells (Tregs)

Regulatory T-cells (Tregs) play multiple roles in the immunopathology of liver diseases. First of all, they are crucial in maintaining hepatic immunological self-tolerance. Normal liver contains a low frequency of CD8+ or CD4+ effector T-cells and Tregs; however, inflammatory and autoimmune liver diseases are associated with enrichment of these subsets of lymphocytes [115,116]. Tregs attenuate inflammation by suppressing effector T-cell proliferation and cytokine secretion, thereby avoiding chronic lobular or interface hepatitis, which may result in cirrhosis, liver failure, or hepatocellular carcinoma (HCC) [117]. Accordingly, deficiencies in Treg cell development or function result in uncontrolled immune responses and tissue destruction, leading to inflammatory disorders such as graft-versus-host disease (GvHD), transplant rejection, and autoimmune diseases. Tregs have been considered an attractive therapeutic candidate for restoring immune tolerance in autoimmune and autoinflammatory diseases and in the course of transplantation of hematopoietic stem cells and solid organs, with the intent to reduce or replace immunosuppressive drugs [118]. Overall, Treg cell infusions have been shown to be safe, even if the manufacturing of these cells in the context of chronic immunosuppressive treatments remains challenging. Thus, enhancing Treg frequency, along with administration of alloantigen-specific Tregs, may potentially be a future therapeutic option to maintain post-transplant tolerance [116,119].
Moreover, evidence suggests that a quantitative or functional deficiency in Tregs plays a key role in the pathogenesis of autoimmune hepatitis (AIH) and other autoimmune diseases [116,120,121]. In AIH, the pathogenetic effector CD4+ and CD8+ T-cell immune response to auto-antigen causes a loss of immunological tolerance to hepatocytes [116]. The standard treatment to achieve remission is represented by long-term immunosuppression; however, progression to end-stage liver disease occurs in 10–20% of patients [116]. Consequently, a shift towards regulatory dominance by applying cellular therapy to enrich hepatic Tregs could be a therapeutic strategy for restoring self-tolerance [116]. Targeting CXCR3, which is highly expressed in peripheral Tregs and enriched in inflamed liver tissue, as well as selectively expanding Tregs by IL2 or IL2/anti- IL2 complex administration, may be potential options to suppress ongoing hepatitis [116,122,123]. Additionally, Tregs were administered to protect against fulminant hepatitis by suppressing effector T-cells in mouse acute HBV infection [124]. The field is steadily moving forward, with several phase I clinical trials aiming to evaluate Treg cell therapy in AIH (NCT02704338) and liver transplantation [116,125]. Immunotherapy was investigated as a further therapeutic option for patients with HCC. On the other hand, FOXP3+ Tregs are one of the mechanisms of tumor-driven immune evasion due to their capacity to block tumor-specific CD8+ and CD4+ effector T-cells. Hence, attenuating Tregs may promote anti-tumor immunity [126]. The administration of blocking antibodies such as daclizumab, which reduces FOXP3+ CD25high CD45RAneg Treg numbers, or cytotoxic T-lymphocyte-associated antigen 4 (CTLA4), thereby depleting CD25+ Tregs, represents potential approaches to do so [127,128]. Tempering successful immunotherapy by degrading local Tregs while preserving host immune response may improve HCC therapy.

3.2.2. Chimeric Antigen Receptor-Engineered T-Cells (CAR-Ts)

Chimeric antigen receptor-engineered T-cell (CAR-T) therapy is a promising strategy in the development of novel and personalized cancer therapies; it is based on T-cell transfer after the stimulation, expansion, and reinfusion of peripheric antigen-specific T-cells derived from patient blood. So far, CAR-T therapy has been investigated in clinical trials to treat hematological diseases and solid tumors [4]. These cells, which are engineered to recognize specific tumor-associated antigens (TAA), consist of four components: (i) the external component, usually made by the single-chain variable fragment (scFv) domain; (ii) the space region or hinge, consisting of the IgG1 hinge-CH2-CH3 Fc domain connecting the scFv; (iii) the transmembrane region; and (iv) the intracellular signaling domain, which varies from one CAR-T generation to another [129]. Various CAR-T approaches for the treatment of HCC or HCC metastases or against different TAAs have been evaluated in pre-clinical models, showing increased anti-tumor efficacy. However, a lack of specific tumor antigens, limited trafficking, and the penetration of CAR-T cells to tumor sites, as well as the immunosuppressive tumor microenvironment, are still major hurdles to overcome [4]. Moreover, CAR-T therapy is associated with substantial side effects, such as tumor lysis syndrome and cytokine release syndrome. Both complications may be managed by enhancing the selectivity of CARs and controlling CAR-T activity in the future [130]. Currently, 22 clinical trials are investigating the application of CAR-T for HCC treatment, most of them targeting glypican 3 (GPC3), Mucin 1 (MUC), and carcinoembryonic antigen. To date, no clinical trials investigating the efficacy of CAR-T therapy to treat HCC have been completed [129,131,132].

3.2.3. Dendritic Cells (DCs)

DCs are bone-marrow-derived antigen-presenting cells, promoting self-tolerance in the healthy steady-state while regulating innate and adaptive immunity [133]. Recent approaches have demonstrated a role for infused regulatory DCs (DCreg) in reducing T-cell activation and coordinating CD4+ and CD8+ memory T-cell responses [134], particularly for the therapy of allograft rejection. The first in-human, single-center, open-label phase I/II study has been initiated to test the safety of a single infusion of donor-derived DCreg in de novo adult living donor liver transplant recipients. Furthermore, the impact of DCreg as a novel adjunct induction therapy to induce donor-specific T-cell hyper-responsiveness and to allow early withdrawal of immunosuppressive therapy will be tested in this study [135]. Strategies selectively targeting DCs to enhance their immune regulatory function still promise a potential therapeutic application [133].

3.2.4. Macrophages, Monocytes, and Kupffer Cells (KCs)

Hepatic macrophages are key players in various types of liver diseases and are delineated as promising targets to develop new therapies [136]. These include resident KCs and monocyte-derived macrophages (MoMFs). While KCs represent the main hepatic macrophages involved in homeostasis, hepatic damage results in massive infiltration of MoMFs into the injured liver, which contributes to augmenting liver inflammation, fibrosis, and HCC [137]. The targeting of hepatic macrophages by chemokines in order to regulate their recruitment, differentiation/polarization, and activation has been investigated in pre-clinical models [138]. Additionally, therapies based on inhibiting the intracellular inflammatory signaling pathways of KC have been explored [139]. The expression of Cadherin 11 (CDH11) in injured cells has been shown to regulate myofibroblast activation during the development of fibrosis. Consequently, CDH11 may represent the main target of macrophages for the treatment of liver fibrosis [140]. Moreover, approaches to limit monocyte recruitment to the liver, mostly based on interfering with the chemokine signaling for monocytes, have been investigated. Given the categorization of MoMFs in Ly6Chigh monocytes, causing organ impairment and Ly6Clow monocytes, which are restorative, a suggested strategy was to switch Ly6Chigh monocytes into Ly6Clow monocytes in order to achieve the restoration of proper organ function [141]. Taken together, the significant role of macrophages in liver diseases highlights them as ideal targets when developing novel cell-based therapies; however, the heterogeneity of this cell population represents a challenge in this context.
In summary, the most promising efforts in developing immune cell therapies to treat and cure liver diseases have been reported for the treatment of HCC. Several clinical phase I/II studies are currently ongoing, with most of them investigating the potential of CAR-T cell therapies [142].

4. Technological Developments for Improved Delivery and Engraftment of Cell-Based Therapeutics

Several factors have to be considered when developing cell-based therapies. A major hurdle is the route of administration since this is a critical parameter for the efficacy of such therapies. For in vivo injection, it can be distinguished between local and systemic delivery. The most prominent route is by portal vein or hepatic artery infusion; however, ectopic implantation, either into the spleen or peritoneum, has also been reported. It is important that the cells reach their target—the liver parenchyma—within 24 h since, otherwise, they will be cleared by macrophages [57,60,143]. Thus, the use of matrix components such as hyaluronic acid (HA), collagen, and laminin has been studied by different groups to enhance engraftment [144]. Nevi et al. coated hBTSCs with HA and administered those via the spleen into immunocompromised mice. Compared to the control group, the efficacy of the engraftment could be increased four-fold. Moreover, metabolic function according to serum albumin levels was also improved [145].
Recent advances in organoid technologies as a state-of-the-art 3D culture model are not only of interest to study diseases or for pharmacological screening but may also be utilized to be transplanted directly to improve organ function, as shown by a few experimental studies. For example, Kuijk et al. generated liver organoids of rat liver stem cells and subsequently transplanted them into a rat model of acute liver failure. In 7 out of 11 rats, proper cell engraftment was reported with improved liver function, as indicated by albumin secretion [146]. In another study, extrahepatic cholangiocyte organoids were able to reconstruct the biliary tree in a mouse model of extrahepatic biliary injury [147].
Nevertheless, OLT will most likely remain the only treatment/curative option for a variety of end-stage liver diseases in the near future. Thus, ex vivo treatment of marginal organs may help to overcome organ shortage. Machine perfusion of organs is not only a versatile tool for organ preservation but may also be utilized for the delivery of cells to promote liver regeneration and repair. In this context, normothermic machine perfusion (NMP) is of special interest. The main blood vessels of the liver are thereby cannulated to maintain a perfusion circuit, which is carried out at 37 °C to achieve close to physiological conditions. The therapeutic agents can be added directly into the perfusate to be delivered into the vasculature of the organ. The group of Yang et al. administered bmMSCs in a donation after circulatory death (DCD) rat model during NMP, which resulted in improved liver function, reduced histological damage, and restored mitochondrial function [148]. In another murine study, the supplementation of the perfusate with EV derived from human liver stem cells led to improved liver function compared to the control group [149]. Laing et al. perfused discarded human organs under normothermic conditions and delivered multipotent adult progenitor cells to the organs. They not only showed engraftment of the cells via confocal microscopy but, additionally, three out of six organs met the viability criteria for transplantation [150]. However, only a limited number of experiments have been reported for the liver so far, in contrast to other organs such as kidney and heart.
Recently, Sampaziotis et al. combined two of the most promising technologies to repair human livers declined for transplantation due to ischemic bile duct injury, indicated by a pH < 7.5: red fluorescence protein (RFP)-expressing cholangiocyte organoids (mechanically dissociated, 10 × 106 cells) were injected into the terminal branch of the intrahepatic ducts of three human livers at the start of NMP; at the same time, they were preserved at the perfusion device ex vivo for up to 100 h. In this experimental effort, the successful engraftment of REP organoids into the intrahepatic biliary tree was reported for the very first time when combining machine perfusion and 3D cell culture techniques. The organoids expressed key biliary markers and did not differentiate into other lineages. After 100 h of NMP, no signs of cholangiopathy were found [151]. While these promising results represent the very first step towards revolutionizing the concept of treating and even curing severe liver diseases, it may take another decade to fully implement this therapeutic option in clinical routine, leaving many questions unanswered at this point.

5. Conclusions

Cellular therapies are a promising alternative that may replace orthoptic liver transplantation for a variety of liver diseases in the future or at least bridge the time to transplantation. Interdisciplinary efforts are made to enhance their efficacy by combining recent achievements in the fields of tissue engineering and machine perfusion, which may enhance the number of organs for transplantation and may also enable the regeneration of the patients’ own organs in the future. The concept of ex vivo organ treatment and regeneration is not limited to liver diseases but can also be applied to other organs such as heart, kidney, and lung. A better understanding of the underlying mechanisms of the various diseases, together with drug engineering approaches, has led to the development of novel immunomodulatory drugs, which is a great achievement towards personalized medicine.

Author Contributions

Conceptualization, J.H. and T.H.; writing—original draft preparation, J.H., V.H., H.E., A.T.M., M.F. and T.H.; writing—review and editing, D.Ö., J.T., S.S. and T.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

We thank all members of the organLife team at the Department of Visceral, Transplant, and Thoracic Surgery. We would like to acknowledge that the figures were created using BioRender.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AFPAlpha-1-fetoprotein
AIHAutoimmune hepatitis
BECBiliary epithelial cells
bmBone marrow
BMPBone morphogenic protein
CAR-TChimeric antigen receptor-engineered T cells
CCL4Carbon tetrachloride
CDH11Cadherin 11
CPMCarboxypeptidase M
CTLA4Cytotoxic T lymphocyte-associated antigen 4
CXCRC-X-C motif chemokine receptor
DCDendritic cells
DCregRegulatory DC
DEXDexamethasone
DLL4Delta-ligand like 4
dpDental pulp
EGFREpidermal growth factor receptor
ESCEmbryonic stem cell
EVExtracellular vesicles
FGFFibroblast growth factor
FoxA2Forkhead-box protein A2
G-CSFGranulocyte colony-stimulating factor
GATA4Gata-binding protein 4
GM-CSFGranulocyte-monocyte colony-stimulating factor
GPC3Glypican 3
GvHDGraft-versus-host disease
HAHyaluronic acid
HBCHepatitis B
HCCHepatocellular carcinoma
HCVHepatitis C
HGFHepatocyte growth factor
HLCHepatoblast-like cells
HNF4αHepatocyte nuclear factor 4α
HSCHematopoietic stem cell
IGF1Insulin growth factor 1
iHepInduced hepatocyte-like cells
ILInterleukin
INFγInterferon gamma
INRInternational normalized ratio
iPSCInduced pluripotent stem cell
ITSInsulin-transferrin-sodium selenite
KCKupffer cells
LSECLiver sinusoidal endothelial cells
MMPMatrix metallopeptidase
MoMFMonocyte-derived macrophage
MSCMesenchymal stem cell
MUCMucin 1
NMPNormothermic machine perfusion
NONitric oxide
NPCNon-parenchymal cells
OLTOrthoptic liver transplantation
Oct4Octamer binding transcription factor 4
PEG2Prostaglansin E2
RFPRed flourescene protein
SCHepatic stellate cells
scFvSingle chain variable fragment
SDF1Stromal-cell-derived factor 1
SoxSRY-Box Transcription Factor
TAATumor-assocated antigens
TAZPDZ-minding motif
TET1Ten-eleven translocation 1
TGFαTransforming growth factor alpha
TIMP1Inhibitor of metalloprotease 1
TNFαTumor necrosis factor alpha
TregsRegulatory T cells
ucumbilical cord
VEGFVascular endothelial growth factor
YAPYes-associated protein

References

  1. Trefts, E.; Gannon, M.; Wasserman, D.H. The liver. Curr. Biol. 2017, 27, R1147–R1151. [Google Scholar] [CrossRef]
  2. Racanelli, V.; Rehermann, B. The liver as an immunological organ. Hepatology 2006, 43, S54–S62. [Google Scholar] [CrossRef]
  3. Guillot, A.; Tacke, F. Liver Macrophages: Old Dogmas and New Insights. Hepatol. Commun. 2019, 3, 730–743. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Chen, Y.; Chang-Yong, E.; Gong, Z.W.; Liu, S.; Wang, Z.X.; Yang, Y.S.; Zhang, X.W. Chimeric antigen receptor-engineered T-cell therapy for liver cancer. Hepatobiliary Pancreat. Dis. Int. 2018, 17, 301–309. [Google Scholar] [CrossRef] [PubMed]
  5. Aravalli, R.N.; Steer, C.J. Immune-Mediated Therapies for Liver Cancer. Genes 2017, 8, 76. [Google Scholar] [CrossRef] [Green Version]
  6. Pinato, D.J.; Guerra, N.; Fessas, P.; Murphy, R.; Mineo, T.; Mauri, F.A.; Mukherjee, S.K.; Thursz, M.; Wong, C.N.; Sharma, R.; et al. Immune-based therapies for hepatocellular carcinoma. Oncogene 2020, 39, 3620–3637. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Asrani, S.K.; Devarbhavi, H.; Eaton, J.; Kamath, P.S. Burden of liver diseases in the world. J. Hepatol. 2019, 70, 151–171. [Google Scholar] [CrossRef]
  8. Ding, C.; Li, Y.; Guo, F.; Jiang, Y.; Ying, W.; Li, D.; Yang, D.; Xia, X.; Liu, W.; Zhao, Y.; et al. A Cell-type-resolved Liver Proteome. Mol. Cell Proteom. 2016, 15, 3190–3202. [Google Scholar] [CrossRef] [Green Version]
  9. Strazzabosco, M.; Fabris, L. Functional anatomy of normal bile ducts. Anat. Rec. 2008, 291, 653–660. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Kubes, P.; Jenne, C. Immune Responses in the Liver. Annu. Rev. Immunol. 2018, 36, 247–277. [Google Scholar] [CrossRef]
  11. Doherty, D.G. Immunity, tolerance and autoimmunity in the liver: A comprehensive review. J. Autoimmun. 2016, 66, 60–75. [Google Scholar] [CrossRef]
  12. Lu, W.Y.; Bird, T.G.; Boulter, L.; Tsuchiya, A.; Cole, A.M.; Hay, T.; Guest, R.V.; Wojtacha, D.; Man, T.Y.; Mackinnon, A.; et al. Hepatic progenitor cells of biliary origin with liver repopulation capacity. Nat. Cell Biol. 2015, 17, 971–983. [Google Scholar] [CrossRef]
  13. Brunner, S.M.; Junger, H.; Ruemmele, P.; Schnitzbauer, A.A.; Doenecke, A.; Kirchner, G.I.; Farkas, S.A.; Loss, M.; Scherer, M.N.; Schlitt, H.J.; et al. Bile duct damage after cold storage of deceased donor livers predicts biliary complications after liver transplantation. J. Hepatol. 2013, 58, 1133–1139. [Google Scholar] [CrossRef]
  14. Tabibian, J.H.; Masyuk, A.I.; Masyuk, T.V.; O’Hara, S.P.; LaRusso, N.F. Physiology of cholangiocytes. Compr. Physiol. 2013, 3, 541–565. [Google Scholar]
  15. Keplinger, K.M.; Bloomston, M. Anatomy and embryology of the biliary tract. Surg. Clin. N. Am. 2014, 94, 203–217. [Google Scholar] [CrossRef]
  16. Alpini, G.; Lenzi, R.; Sarkozi, L.; Tavoloni, N. Biliary physiology in rats with bile ductular cell hyperplasia. Evidence for a secretory function of proliferated bile ductules. J. Clin. Investig. 1988, 81, 569–578. [Google Scholar] [CrossRef] [Green Version]
  17. Alpini, G.; Lenzi, R.; Zhai, W.R.; Slott, P.A.; Liu, M.H.; Sarkozi, L.; Tavoloni, N. Bile secretory function of intrahepatic biliary epithelium in the rat. Am. J. Physiol. 1989, 257, G124–G133. [Google Scholar] [CrossRef] [PubMed]
  18. Alpini, G.; Ueno, Y.; Glaser, S.S.; Marzioni, M.; Phinizy, J.L.; Francis, H.; Lesage, G. Bile acid feeding increased proliferative activity and apical bile acid transporter expression in both small and large rat cholangiocytes. Hepatology 2001, 34, 868–876. [Google Scholar] [CrossRef] [PubMed]
  19. Alpini, G.; Roberts, S.; Kuntz, S.M.; Ueno, Y.; Gubba, S.; Podila, P.V.; LeSage, G.; LaRusso, N.F. Morphological, molecular, and functional heterogeneity of cholangiocytes from normal rat liver. Gastroenterology 1996, 110, 1636–1643. [Google Scholar] [CrossRef] [PubMed]
  20. Alpini, G.; Glaser, S.; Robertson, W.; Rodgers, R.E.; Phinizy, J.L.; Lasater, J.; LeSage, G.D. Large but not small intrahepatic bile ducts are involved in secretin-regulated ductal bile secretion. Am. J. Physiol. 1997, 272, G1064–G1074. [Google Scholar] [CrossRef]
  21. Martinez-Anso, E.; Castillo, J.E.; Diez, J.; Medina, J.F.; Prieto, J. Immunohistochemical detection of chloride/bicarbonate anion exchangers in human liver. Hepatology 1994, 19, 1400–1406. [Google Scholar] [CrossRef]
  22. Glaser, S.S.; Gaudio, E.; Rao, A.; Pierce, L.M.; Onori, P.; Franchitto, A.; Francis, H.L.; Dostal, D.E.; Venter, J.K.; DeMorrow, S.; et al. Morphological and functional heterogeneity of the mouse intrahepatic biliary epithelium. Lab. Investig. 2009, 89, 456–469. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Lazaridis, K.N.; Pham, L.; Vroman, B.; de Groen, P.C.; LaRusso, N.F. Kinetic and molecular identification of sodium-dependent glucose transporter in normal rat cholangiocytes. Am. J. Physiol. 1997, 272, G1168–G1174. [Google Scholar] [CrossRef]
  24. Ballatori, N.; Jacob, R.; Barrett, C.; Boyer, J.L. Biliary catabolism of glutathione and differential reabsorption of its amino acid constituents. Am. J. Physiol. 1988, 254, G1–G7. [Google Scholar] [CrossRef]
  25. Hohenester, S.; Wenniger, L.M.; Paulusma, C.C.; van Vliet, S.J.; Jefferson, D.M.; Elferink, R.P.; Beuers, U. A biliary HCO3- umbrella constitutes a protective mechanism against bile acid-induced injury in human cholangiocytes. Hepatology 2012, 55, 173–183. [Google Scholar] [CrossRef]
  26. Hohenester, S.; Maillette de Buy Wenniger, L.; Jefferson, D.M.; Oude Elferink, R.P.; Beuers, U. Biliary bicarbonate secretion constitutes a protective mechanism against bile acid-induced injury in man. Dig. Dis. 2011, 29, 62–65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Bhushan, B.; Apte, U. Liver Regeneration after Acetaminophen Hepatotoxicity: Mechanisms and Therapeutic Opportunities. Am. J. Pathol. 2019, 189, 719–729. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Michalopoulos, G.K. Liver regeneration. J. Cell Physiol. 2007, 213, 286–300. [Google Scholar] [CrossRef]
  29. Michalopoulos, G.K.; Bhushan, B. Liver regeneration: Biological and pathological mechanisms and implications. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 40–55. [Google Scholar] [CrossRef]
  30. Campana, L.; Esser, H.; Huch, M.; Forbes, S. Liver regeneration and inflammation: From fundamental science to clinical applications. Nat. Rev. Mol. Cell Biol. 2021. [Google Scholar] [CrossRef]
  31. Fausto, N.; Campbell, J.S.; Riehle, K.J. Liver regeneration. Hepatology 2006, 43, S45–S53. [Google Scholar] [CrossRef] [PubMed]
  32. Michalopoulos, G.K. Principles of liver regeneration and growth homeostasis. Compr. Physiol. 2013, 3, 485–513. [Google Scholar]
  33. Marubashi, S.; Sakon, M.; Nagano, H.; Gotoh, K.; Hashimoto, K.; Kubota, M.; Kobayashi, S.; Yamamoto, S.; Miyamoto, A.; Dono, K.; et al. Effect of portal hemodynamics on liver regeneration studied in a novel portohepatic shunt rat model. Surgery 2004, 136, 1028–1037. [Google Scholar] [CrossRef] [PubMed]
  34. Mars, W.M.; Liu, M.-L.; Kitson, R.P.; Goldfarb, R.H.; Gabauer, M.K.; Michalopoulos, G.K. Immediate Early Detection of Urokinase Receptor After Partial Hepatectomy and Its Implications for Initiation of Liver Regeneration. Hepatology 1995, 21, 1695–1701. [Google Scholar] [PubMed]
  35. Kim, T.-H.; Mars, W.M.; Stolz, D.B.; Michalopoulos, G.K. Expression and Activation of Pro-MMP-2 and Pro-MMP-9 during Rat Liver Regeneration. Hepatology 2000, 31, 75–82. [Google Scholar] [CrossRef]
  36. Lindroos, P.M.; Zarnegar, R.; Michalopoulos, G.K. Hepatocyte Growth Factor (Hepatopoietin A) Rapidly Increases in Plasma before DNA Synthesis and Liver Regeneration Stimulated by Partial Hepatectomy and Carbon Tetrachloride Administration. Hepatology 1991, 13, 744–750. [Google Scholar] [CrossRef]
  37. Patijn, G.A.; Lieber, A.; Schowalter, D.B.; Schwall, R.; Kay, M.A. Hepatocyte Growth Factor Induces Hepatocyte Proliferation In Vivo and Allows for Effiecient Retroviral-Mediated Gene Transfer in Mice. Hepatology 1998, 28, 707–716. [Google Scholar] [CrossRef] [PubMed]
  38. Gilgenkrantz, H.; Collin de l’Hortet, A. Understanding Liver Regeneration: From Mechanisms to Regenerative Medicine. Am. J. Pathol. 2018, 188, 1316–1327. [Google Scholar] [CrossRef] [Green Version]
  39. Pepe-Mooney, B.J.; Dill, M.T.; Alemany, A.; Ordovas-Montanes, J.; Matsushita, Y.; Rao, A.; Sen, A.; Miyazaki, M.; Anakk, S.; Dawson, P.A.; et al. Single-Cell Analysis of the Liver Epithelium Reveals Dynamic Heterogeneity and an Essential Role for YAP in Homeostasis and Regeneration. Cell Stem Cell 2019, 25, 23–38.e28. [Google Scholar] [CrossRef] [PubMed]
  40. He, J.; Lu, H.; Zou, Q.; Luo, L. Regeneration of liver after extreme hepatocyte loss occurs mainly via biliary transdifferentiation in zebrafish. Gastroenterology 2014, 146, 789–800.e788. [Google Scholar] [CrossRef]
  41. Raven, A.; Lu, W.Y.; Man, T.Y.; Ferreira-Gonzalez, S.; O’Duibhir, E.; Dwyer, B.J.; Thomson, J.P.; Meehan, R.R.; Bogorad, R.; Koteliansky, V.; et al. Cholangiocytes act as facultative liver stem cells during impaired hepatocyte regeneration. Nature 2017, 547, 350–354. [Google Scholar] [CrossRef] [PubMed]
  42. Speicher, T.; Siegenthaler, B.; Bogorad, R.L.; Ruppert, R.; Petzold, T.; Padrissa-Altes, S.; Bachofner, M.; Anderson, D.G.; Koteliansky, V.; Fässler, R.; et al. Knockdown and knockout of β1-integrin in hepatocytes impairs liver regeneration through inhibition of growth factor signalling. Nat. Commun. 2014, 5, 3862. [Google Scholar] [CrossRef] [Green Version]
  43. Huch, M.; Bonfanti, P.; Boj, S.F.; Sato, T.; Loomans, C.J.M.; van de Wetering, M.; Sojoodi, M.; Li, V.S.W.; Schuijers, J.; Gracanin, A.; et al. Unlimited in vitro expansion of adult bi-potent pancreas progenitors through the Lgr5/R-spondin axis. EMBO J. 2013, 32, 2708–2721. [Google Scholar] [CrossRef] [Green Version]
  44. Shin, S.; Upadhyay, N.; Greenbaum, L.E.; Kaestner, K.H. Ablation of Foxl1-Cre-labeled hepatic progenitor cells and their descendants impairs recovery of mice from liver injury. Gastroenterology 2015, 148, 192–202.e193. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Espanol-Suner, R.; Carpentier, R.; Van Hul, N.; Legry, V.; Achouri, Y.; Cordi, S.; Jacquemin, P.; Lemaigre, F.; Leclercq, I.A. Liver progenitor cells yield functional hepatocytes in response to chronic liver injury in mice. Gastroenterology 2012, 143, 1564–1575.e1567. [Google Scholar] [CrossRef]
  46. Suzuki, A.; Sekiya, S.; Buscher, D.; Izpisua Belmonte, J.C.; Taniguchi, H. Tbx3 controls the fate of hepatic progenitor cells in liver development by suppressing p19ARF expression. Development 2008, 135, 1589–1595. [Google Scholar] [CrossRef] [Green Version]
  47. Lu, J.; Zhou, Y.; Hu, T.; Zhang, H.; Shen, M.; Cheng, P.; Dai, W.; Wang, F.; Chen, K.; Zhang, Y.; et al. Notch Signaling Coordinates Progenitor Cell-Mediated Biliary Regeneration Following Partial Hepatectomy. Sci. Rep. 2016, 6, 22754. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Boulter, L.; Govaere, O.; Bird, T.G.; Radulescu, S.; Ramachandran, P.; Pellicoro, A.; Ridgway, R.A.; Seo, S.S.; Spee, B.; Van Rooijen, N.; et al. Macrophage-derived Wnt opposes Notch signaling to specify hepatic progenitor cell fate in chronic liver disease. Nat. Med. 2012, 18, 572–579. [Google Scholar] [CrossRef] [Green Version]
  49. Aloia, L.; McKie, M.A.; Vernaz, G.; Cordero-Espinoza, L.; Aleksieva, N.; van den Ameele, J.; Antonica, F.; Font-Cunill, B.; Raven, A.; Aiese Cigliano, R.; et al. Epigenetic remodelling licences adult cholangiocytes for organoid formation and liver regeneration. Nat. Cell Biol. 2019, 21, 1321–1333. [Google Scholar] [CrossRef]
  50. Yimlamai, D.; Christodoulou, C.; Galli, G.G.; Yanger, K.; Pepe-Mooney, B.; Gurung, B.; Shrestha, K.; Cahan, P.; Stanger, B.Z.; Camargo, F.D. Hippo pathway activity influences liver cell fate. Cell 2014, 157, 1324–1338. [Google Scholar] [CrossRef] [Green Version]
  51. Fukano, S.; Saitoh, Y.; Uchida, K.; Akiyoshi, T.; Takeda, K.i. Bile acid metabolism in partially hepatectomized rats. Steroids 1985, 45, 209–227. [Google Scholar] [CrossRef]
  52. Lesage, G.; Glaser, S.S.; Gubba, S.; Robertson, W.E.; Phinizy, J.L.; Lasater, J.; Rodgers, R.E.; Alpini, G. Regrowth of the rat biliary tree after 70% partial hepatectomy is coupled to increased secretin-induced ductal secretion. Gastroenterology 1996, 111, 1633–1644. [Google Scholar] [CrossRef] [Green Version]
  53. Planas-Paz, L.; Sun, T.; Pikiolek, M.; Cochran, N.R.; Bergling, S.; Orsini, V.; Yang, Z.; Sigoillot, F.; Jetzer, J.; Syed, M.; et al. YAP, but Not RSPO-LGR4/5, Signaling in Biliary Epithelial Cells Promotes a Ductular Reaction in Response to Liver Injury. Cell Stem Cell 2019, 25, 39–53.e10. [Google Scholar] [CrossRef] [PubMed]
  54. Font-Burgada, J.; Shalapour, S.; Ramaswamy, S.; Hsueh, B.; Rossell, D.; Umemura, A.; Taniguchi, K.; Nakagawa, H.; Valasek, M.A.; Ye, L.; et al. Hybrid Periportal Hepatocytes Regenerate the Injured Liver without Giving Rise to Cancer. Cell 2015, 162, 766–779. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Han, X.; Wang, Y.; Pu, W.; Huang, X.; Qiu, L.; Li, Y.; Yu, W.; Zhao, H.; Liu, X.; He, L.; et al. Lineage Tracing Reveals the Bipotency of SOX9+ Hepatocytes during Liver Regeneration. Stem Cell Rep. 2019, 12, 624–638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Yimlamai, D.; Fowl, B.H.; Camargo, F.D. Emerging evidence on the role of the Hippo/YAP pathway in liver physiology and cancer. J. Hepatol. 2015, 63, 1491–1501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Iansante, V.; Mitry, R.R.; Filippi, C.; Fitzpatrick, E.; Dhawan, A. Human hepatocyte transplantation for liver disease: Current status and future perspectives. Pediatr. Res. 2018, 83, 232–240. [Google Scholar] [CrossRef] [Green Version]
  58. Zhou, G.P.; Jiang, Y.Z.; Sun, L.Y.; Zhu, Z.J. Therapeutic effect and safety of stem cell therapy for chronic liver disease: A systematic review and meta-analysis of randomized controlled trials. Stem Cell Res. Ther. 2020, 11, 419. [Google Scholar] [CrossRef]
  59. Cantz, T.; Manns, M.P.; Ott, M. Stem cells in liver regeneration and therapy. Cell Tissue Res. 2008, 331, 271–282. [Google Scholar] [CrossRef] [Green Version]
  60. Tsolaki, E.; Yannaki, E. Stem cell-based regenerative opportunities for the liver: State of the art and beyond. World J. Gastroenterol. 2015, 21, 12334–12350. [Google Scholar] [CrossRef]
  61. Boyd, A.; Newsome, P.; Lu, W.Y. The role of stem cells in liver injury and repair. Expert Rev. Gastroenterol. Hepatol. 2019, 13, 623–631. [Google Scholar] [CrossRef] [PubMed]
  62. Wang, J.; Sun, M.; Liu, W.; Li, Y.; Li, M. Stem Cell-Based Therapies for Liver Diseases: An Overview and Update. Tissue Eng. Regen. Med. 2019, 16, 107–118. [Google Scholar] [CrossRef] [PubMed]
  63. Hannan, N.R.; Segeritz, C.P.; Touboul, T.; Vallier, L. Production of hepatocyte-like cells from human pluripotent stem cells. Nat. Protoc. 2013, 8, 430–437. [Google Scholar] [CrossRef] [Green Version]
  64. Tolosa, L.; Caron, J.; Hannoun, Z.; Antoni, M.; López, S.; Burks, D.; Castell, J.V.; Weber, A.; Gomez-Lechon, M.J.; Dubart-Kupperschmitt, A. Transplantation of hESC-derived hepatocytes protects mice from liver injury. Stem Cell Res. Ther. 2015, 6, 246. [Google Scholar] [CrossRef] [Green Version]
  65. Haideri, S.S.; McKinnon, A.C.; Taylor, A.H.; Kirkwood, P.; Starkey Lewis, P.J.; O’Duibhir, E.; Vernay, B.; Forbes, S.; Forrester, L.M. Injection of embryonic stem cell derived macrophages ameliorates fibrosis in a murine model of liver injury. NPJ Regen. Med. 2017, 2, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Prior, N.; Inacio, P.; Huch, M. Liver organoids: From basic research to therapeutic applications. Gut 2019, 68, 2228–2237. [Google Scholar] [CrossRef] [Green Version]
  67. Wang, S.; Wang, X.; Tan, Z.; Su, Y.; Liu, J.; Chang, M.; Yan, F.; Chen, J.; Chen, T.; Li, C.; et al. Human ESC-derived expandable hepatic organoids enable therapeutic liver repopulation and pathophysiological modeling of alcoholic liver injury. Cell Res. 2019, 29, 1009–1026. [Google Scholar] [CrossRef] [PubMed]
  68. Ramli, M.N.B.; Lim, Y.S.; Koe, C.T.; Demircioglu, D.; Tng, W.; Gonzales, K.A.U.; Tan, C.P.; Szczerbinska, I.; Liang, H.; Soe, E.L.; et al. Human Pluripotent Stem Cell-Derived Organoids as Models of Liver Disease. Gastroenterology 2020, 159, 1471–1486.e1412. [Google Scholar] [CrossRef]
  69. Mun, S.J.; Ryu, J.S.; Lee, M.O.; Son, Y.S.; Oh, S.J.; Cho, H.S.; Son, M.Y.; Kim, D.S.; Kim, S.J.; Yoo, H.J.; et al. Generation of expandable human pluripotent stem cell-derived hepatocyte-like liver organoids. J. Hepatol. 2019, 71, 970–985. [Google Scholar] [CrossRef]
  70. Kollet, O.; Shivtiel, S.; Chen, Y.-Q.; Suriawinata, J.; Thung, S.N.; Dabeva, M.D.; Kahn, J.; Spiegel, A.; Dar, A.; Samira, S.; et al. HGF, SDF-1, and MMP-9 are involved in stress-induced human CD34+ stem cell recruitment to the liver. J. Clin. Investig. 2003, 112, 160–169. [Google Scholar] [CrossRef]
  71. Crosby, H.A.; Lalor, P.F.; Ross, E.; Newsome, P.N.; Adams, D.H. Adhesion of human haematopoietic (CD34+) stem cells to human liver compartments is integrin and CD44 dependent and modulated by CXCR3 and CXCR4. J. Hepatol. 2009, 51, 734–749. [Google Scholar] [CrossRef]
  72. Lee, J.Y.; Hong, S.H. Hematopoietic Stem Cells and Their Roles in Tissue Regeneration. Int. J. Stem Cells 2020, 13, 1–12. [Google Scholar] [CrossRef] [Green Version]
  73. Lagasse, E.; Connors, H.; Al-Dhalimy, M.; Reitsma, M.; Dohse, M.; Osborne, L.; Wang, X.; Finegold, M.; Weissman, I.L.; Grompe, M. Purified hematopoietic stem cells can differentiate into hepatocytes in vivo. Nat. Med. 2000, 6, 1229–1234. [Google Scholar] [CrossRef]
  74. Khurana, S.; Jaiswal, A.K.; Mukhopadhyay, A. Hepatocyte nuclear factor-4alpha induces transdifferentiation of hematopoietic cells into hepatocytes. J. Biol. Chem. 2010, 285, 4725–4731. [Google Scholar] [CrossRef] [Green Version]
  75. Sellamuthu, S.; Manikandan, R.; Thiagarajan, R.; Babu, G.; Dinesh, D.; Prabhu, D.; Arulvasu, C. In vitro trans-differentiation of human umbilical cord derived hematopoietic stem cells into hepatocyte like cells using combination of growth factors for cell based therapy. Cytotechnology 2011, 63, 259–268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Lazaro, C.A.; Croager, E.J.; Mitchell, C.; Campbell, J.S.; Yu, C.; Foraker, J.; Rhim, J.A.; Yeoh, G.C.; Fausto, N. Establishment, characterization, and long-term maintenance of cultures of human fetal hepatocytes. Hepatology 2003, 38, 1095–1106. [Google Scholar] [CrossRef] [PubMed]
  77. Vassilopoulos, G.; Wang, P.-R.; Russel, D. Transplanted Bone Marrow Regenerates Liver By Cell Fusion. Nature 2003, 422, 901–904. [Google Scholar] [CrossRef] [PubMed]
  78. Wang, X.; Willenbring, H.; Akkari, Y.; Torimaru, Y.; Foster, M.; Al-Dhalimy, M.; Lagasse, E.; Finegold, M.; Olson, S.; Grompe, M. Cell Fusion Is The Principal Source Of Bone-Marrow-Derived Hepatocytes. Nature 2003, 422, 893–897. [Google Scholar] [CrossRef]
  79. Lizier, M.; Castelli, A.; Montagna, C.; Lucchini, F.; Vezzoni, P.; Faggioli, F. Cell fusion in the liver, revisited. World J. Hepatol. 2018, 10, 213–221. [Google Scholar] [CrossRef] [PubMed]
  80. Sakaida, I.; Terai, S.; Yamamoto, N.; Aoyama, K.; Ishikawa, T.; Nishina, H.; Okita, K. Transplantation of bone marrow cells reduces CCl4-induced liver fibrosis in mice. Hepatology 2004, 40, 1304–1311. [Google Scholar] [CrossRef]
  81. Salama, H.; Zekri, A.R.; Zern, M.; Bahnassy, A.; Loutfy, S.; Shalaby, S.; Vigen, C.; Burke, W.; Mostafa, M.; Medhat, E.; et al. Autologous hematopoietic stem cell transplantation in 48 patients with end-stage chronic liver diseases. Cell Transplant. 2010, 19, 1475–1486. [Google Scholar] [CrossRef] [PubMed]
  82. Zekri, A.R.; Salama, H.; Medhat, E.; Musa, S.; Abdel-Haleem, H.; Ahmed, O.S.; Khedr, H.A.; Lotfy, M.M.; Zachariah, K.S.; Bahnassy, A.A. The impact of repeated autologous infusion of haematopoietic stem cells in patients with liver insufficiency. Stem Cell Res. Ther. 2015, 6, 118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Pittenger, M.F.; Discher, D.E.; Peault, B.M.; Phinney, D.G.; Hare, J.M.; Caplan, A.I. Mesenchymal stem cell perspective: Cell biology to clinical progress. NPJ Regen. Med. 2019, 4, 22. [Google Scholar] [CrossRef] [Green Version]
  84. Asgari, S.; Moslem, M.; Bagheri-Lankarani, K.; Pournasr, B.; Miryounesi, M.; Baharvand, H. Differentiation and transplantation of human induced pluripotent stem cell-derived hepatocyte-like cells. Stem Cell Rev. Rep. 2013, 9, 493–504. [Google Scholar] [CrossRef]
  85. Choi, J.S.; Jeong, I.S.; Park, Y.J.; Kim, S.W. HGF and IL-10 expressing ALB::GFP reporter cells generated from iPSCs show robust anti-fibrotic property in acute fibrotic liver model. Stem Cell Res. Ther. 2020, 11, 332. [Google Scholar] [CrossRef]
  86. Tasnim, F.; Xing, J.; Huang, X.; Mo, S.; Wei, X.; Tan, M.H.; Yu, H. Generation of mature kupffer cells from human induced pluripotent stem cells. Biomaterials 2019, 192, 377–391. [Google Scholar] [CrossRef]
  87. Povero, D.; Pinatel, E.M.; Leszczynska, A.; Goyal, N.P.; Nishio, T.; Kim, J.; Kneiber, D.; de Araujo Horcel, L.; Eguchi, A.; Ordonez, P.M.; et al. Human induced pluripotent stem cell-derived extracellular vesicles reduce hepatic stellate cell activation and liver fibrosis. JCI Insight 2019, 5, e125652. [Google Scholar] [CrossRef]
  88. Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.; Krause, D.; Deans, R.; Keating, A.; Prockop, D.; Horwitz, E. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006, 8, 315–317. [Google Scholar] [CrossRef]
  89. Snykers, S.; Vanhaecke, T.; Papeleu, P.; Luttun, A.; Jiang, Y.; Vander Heyden, Y.; Verfaillie, C.; Rogiers, V. Sequential exposure to cytokines reflecting embryogenesis: The key for in vitro differentiation of adult bone marrow stem cells into functional hepatocyte-like cells. Toxicol. Sci. 2006, 94, 330–341, discussion 235–339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Ayatollahi, M.; Soleimani, M.; Tabei, S.Z.; Kabir Salmani, M. Hepatogenic differentiation of mesenchymal stem cells induced by insulin like growth factor-I. World J. Stem Cells 2011, 3, 113–121. [Google Scholar] [CrossRef]
  91. Afshari, A.; Shamdani, S.; Uzan, G.; Naserian, S.; Azarpira, N. Different approaches for transformation of mesenchymal stem cells into hepatocyte-like cells. Stem Cell Res. Ther. 2020, 11, 54. [Google Scholar] [CrossRef] [Green Version]
  92. Sun, S.; Yuan, L.; An, Z.; Shi, D.; Xin, J.; Jiang, J.; Ren, K.; Chen, J.; Guo, B.; Zhou, X.; et al. DLL4 restores damaged liver by enhancing hBMSC differentiation into cholangiocytes. Stem Cell Res. 2020, 47, 101900. [Google Scholar] [CrossRef] [PubMed]
  93. Iwanaka, T.; Yamaza, T.; Sonoda, S.; Yoshimaru, K.; Matsuura, T.; Yamaza, H.; Ohga, S.; Oda, Y.; Taguchi, T. A model study for the manufacture and validation of clinical-grade deciduous dental pulp stem cells for chronic liver fibrosis treatment. Stem Cell Res. Ther. 2020, 11, 134. [Google Scholar] [CrossRef]
  94. Lee, J.; Choi, J.; Kang, S.; Kim, J.; Lee, R.; So, S.; Yoon, Y.I.; Kirchner, V.A.; Song, G.W.; Hwang, S.; et al. Hepatogenic Potential and Liver Regeneration Effect of Human Liver-derived Mesenchymal-Like Stem Cells. Cells 2020, 9, 1521. [Google Scholar] [CrossRef]
  95. Li, T.; Xia, M.; Gao, Y.; Chen, Y.; Xu, Y. Human umbilical cord mesenchymal stem cells: An overview of their potential in cell-based therapy. Expert Opin Biol. Ther. 2015, 15, 1293–1306. [Google Scholar] [CrossRef] [PubMed]
  96. Hu, C.; Wu, Z.; Li, L. Mesenchymal stromal cells promote liver regeneration through regulation of immune cells. Int. J. Biol. Sci. 2020, 16, 893–903. [Google Scholar] [CrossRef]
  97. Kang, S.H.; Kim, M.Y.; Eom, Y.W.; Baik, S.K. Mesenchymal Stem Cells for the Treatment of Liver Disease: Present and Perspectives. Gut Liver 2020, 14, 306–315. [Google Scholar] [CrossRef] [PubMed]
  98. Tsuchiya, A.; Takeuchi, S.; Watanabe, T.; Yoshida, T.; Nojiri, S.; Ogawa, M.; Terai, S. Mesenchymal stem cell therapies for liver cirrhosis: MSCs as "conducting cells" for improvement of liver fibrosis and regeneration. Inflamm. Regen. 2019, 39, 18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Eom, Y.W.; Shim, K.Y.; Baik, S.K. Mesenchymal stem cell therapy for liver fibrosis. Korean J. Intern. Med. 2015, 30, 580–589. [Google Scholar] [CrossRef]
  100. Shi, M.; Zhang, Z.; Xu, R.; Lin, H.; Fu, J.; Zou, Z.; Zhang, A.; Shi, J.; Chen, L.; Lv, S.; et al. Human mesenchymal stem cell transfusion is safe and improves liver function in acute-on-chronic liver failure patients. Stem Cells Transl. Med. 2012, 1, 725–731. [Google Scholar] [CrossRef] [PubMed]
  101. Wang, L.; Li, J.; Liu, H.; Li, Y.; Fu, J.; Sun, Y.; Xu, R.; Lin, H.; Wang, S.; Lv, S.; et al. Pilot study of umbilical cord-derived mesenchymal stem cell transfusion in patients with primary biliary cirrhosis. J. Gastroenterol. Hepatol. 2013, 28 (Suppl. S1), 85–92. [Google Scholar] [CrossRef] [PubMed]
  102. 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, 49, e346. [Google Scholar] [CrossRef]
  103. Forbes, S.J.; Gupta, S.; Dhawan, A. Cell therapy for liver disease: From liver transplantation to cell factory. J. Hepatol. 2015, 62, S157–S169. [Google Scholar] [CrossRef] [Green Version]
  104. Puppi, J.; Tan, N.; Mitry, R.R.; Hughes, R.D.; Lehec, S.; Mieli-Vergani, G.; Karani, J.; Champion, M.P.; Heaton, N.; Mohamed, R.; et al. Hepatocyte transplantation followed by auxiliary liver transplantation--a novel treatment for ornithine transcarbamylase deficiency. Am. J. Transplant. 2008, 8, 452–457. [Google Scholar] [CrossRef]
  105. Soltys, K.A.; Setoyama, K.; Tafaleng, E.N.; Soto Gutierrez, A.; Fong, J.; Fukumitsu, K.; Nishikawa, T.; Nagaya, M.; Sada, R.; Haberman, K.; et al. Host conditioning and rejection monitoring in hepatocyte transplantation in humans. J. Hepatol. 2017, 66, 987–1000. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Meyburg, J.; Das, A.M.; Hoerster, F.; Lindner, M.; Kriegbaum, H.; Engelmann, G.; Schmidt, J.; Ott, M.; Pettenazzo, A.; Luecke, T.; et al. One liver for four children: First clinical series of liver cell transplantation for severe neonatal urea cycle defects. Transplantation 2009, 87, 636–641. [Google Scholar] [CrossRef]
  107. Stephenne, X.; Najimi, M.; Sibille, C.; Nassogne, M.C.; Smets, F.; Sokal, E.M. Sustained engraftment and tissue enzyme activity after liver cell transplantation for argininosuccinate lyase deficiency. Gastroenterology 2006, 130, 1317–1323. [Google Scholar] [CrossRef] [PubMed]
  108. Meyburg, J.; Opladen, T.; Spiekerkotter, U.; Schlune, A.; Schenk, J.P.; Schmidt, J.; Weitz, J.; Okun, J.; Burger, F.; Omran, T.B.; et al. Human heterologous liver cells transiently improve hyperammonemia and ureagenesis in individuals with severe urea cycle disorders. J. Inherit. Metab. Dis. 2018, 41, 81–90. [Google Scholar] [CrossRef]
  109. Ambrosino, G.; Varotto, S.; Strom, S.C.; Guariso, G.; Franchin, E.; Miotto, D.; Caenazzo, L.; Basso, S.; Carraro, P.; Valente, M.L.; et al. Isolated Hepatocyte Transplantation for Crigler-Najjar Syndrome Type 1. Cell Transplant. 2005, 14, 151–157. [Google Scholar] [CrossRef]
  110. Fox, I.J.; Chowdhury, J.R.; Kaufman, S.S.; Goertzen, T.C.; Chowdhury, N.R.; Warkentin, P.I.; Dorko, K.; Sauter, B.V.; Strom, S.C. Treatment Of The Crigler-Najjar Syndrome Type I With Hepatocyte Transplantation. N. Engl. J. Med. 1998, 338, 1422–1426. [Google Scholar] [CrossRef] [PubMed]
  111. Allen, K.J.; Mifsud, N.A.; Williamson, R.; Bertolino, P.; Hardikar, W. Cell-mediated rejection results in allograft loss after liver cell transplantation. Liver Transpl. 2008, 14, 688–694. [Google Scholar] [CrossRef]
  112. Hansel, M.C.; Gramignoli, R.; Skvorak, K.J.; Dorko, K.; Marongiu, F.; Blake, W.; Davila, J.; Strom, S.C. The history and use of human hepatocytes for the treatment of liver diseases: The first 100 patients. Curr. Protoc Toxicol. 2014, 62, 14.12.1-23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Smets, F.; Dobbelaere, D.; McKiernan, P.; Dionisi-Vici, C.; Broue, P.; Jacquemin, E.; Lopes, A.I.; Goncalves, I.; Mandel, H.; Pawlowska, J.; et al. Phase I/II Trial of Liver-derived Mesenchymal Stem Cells in Pediatric Liver-based Metabolic Disorders: A Prospective, Open Label, Multicenter, Partially Randomized, Safety Study of One Cycle of Heterologous Human Adult Liver-derived Progenitor Cells (HepaStem) in Urea Cycle Disorders and Crigler-Najjar Syndrome Patients. Transplantation 2019, 103, 1903–1915. [Google Scholar] [PubMed]
  114. Lee, S.M.; Lee, S.D.; Wang, S.Z.; Sarkar, D.; Lee, H.M.; Khan, A.; Bhati, C.; Sharma, A.; Kumaran, V.; Bruno, D.; et al. Effect of mesenchymal stem cell in liver regeneration and clinical applications. Hepatoma Res. 2021, 7, 53. [Google Scholar] [CrossRef]
  115. Norris, S.; Collins, C.; Doherty, D.G.; Smith, F.; McEntee, G.; Traynor, O.; Nolan, N.; Hegarty, J.; O’Farrelly, C. Resident human hepatic lymphocytes are phenotypically different from circulating lymphocytes. J. Hepatol. 1998, 28, 84–90. [Google Scholar] [CrossRef]
  116. Oo, Y.H.; Sakaguchi, S. Regulatory T-cell directed therapies in liver diseases. J. Hepatol. 2013, 59, 1127–1134. [Google Scholar] [CrossRef] [Green Version]
  117. Mieli-Vergani, G.; Vergani, D. Autoimmune hepatitis. Nat. Rev. Gastroenterol. Hepatol. 2011, 8, 320–329. [Google Scholar] [CrossRef] [PubMed]
  118. Ferreira, L.M.R.; Muller, Y.D.; Bluestone, J.A.; Tang, Q. Next-generation regulatory T cell therapy. Nat. Rev. Drug Discov. 2019, 18, 749–769. [Google Scholar] [CrossRef] [Green Version]
  119. Yu, J.; Liu, Z.; Li, C.; Wei, Q.; Zheng, S.; Saeb-Parsy, K.; Xu, X. Regulatory T Cell Therapy Following Liver Transplantation. Liver Transpl. 2021, 27, 264–280. [Google Scholar] [CrossRef]
  120. Longhi, M.S.; Ma, Y.; Grant, C.R.; Samyn, M.; Gordon, P.; Mieli-Vergani, G.; Vergani, D. T-regs in autoimmune hepatitis-systemic lupus erythematosus/mixed connective tissue disease overlap syndrome are functionally defective and display a Th1 cytokine profile. J. Autoimmun. 2013, 41, 146–151. [Google Scholar] [CrossRef]
  121. Longhi, M.S.; Ma, Y.; Bogdanos, D.P.; Cheeseman, P.; Mieli-Vergani, G.; Vergani, D. Impairment of CD4(+)CD25(+) regulatory T-cells in autoimmune liver disease. J. Hepatol. 2004, 41, 31–37. [Google Scholar] [CrossRef] [PubMed]
  122. Curbishley, S.M.; Eksteen, B.; Gladue, R.P.; Lalor, P.; Adams, D.H. CXCR 3 activation promotes lymphocyte transendothelial migration across human hepatic endothelium under fluid flow. Am. J. Pathol. 2005, 167, 887–899. [Google Scholar] [CrossRef] [Green Version]
  123. Létourneau, S.; van Leeuwen, E.M.; Krieg, C.; Martin, C.; Pantaleo, G.; Sprent, J.; Surh, C.D.; Boyman, O. IL-2/anti-IL-2 antibody complexes show strong biological activity by avoiding interaction with IL-2 receptor alpha subunit CD25. Proc. Natl. Acad. Sci. USA 2010, 107, 2171–2176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Stross, L.; Günther, J.; Gasteiger, G.; Asen, T.; Graf, S.; Aichler, M.; Esposito, I.; Busch, D.H.; Knolle, P.; Sparwasser, T.; et al. Foxp3+ regulatory T cells protect the liver from immune damage and compromise virus control during acute experimental hepatitis B virus infection in mice. Hepatology 2012, 56, 873–883. [Google Scholar] [CrossRef]
  125. Safinia, N.; Grageda, N.; Scottà, C.; Thirkell, S.; Fry, L.J.; Vaikunthanathan, T.; Lechler, R.I.; Lombardi, G. Cell Therapy in Organ Transplantation: Our Experience on the Clinical Translation of Regulatory T Cells. Front. Immunol. 2018, 9, 354. [Google Scholar] [CrossRef]
  126. Fu, J.; Xu, D.; Liu, Z.; Shi, M.; Zhao, P.; Fu, B.; Zhang, Z.; Yang, H.; Zhang, H.; Zhou, C.; et al. Increased regulatory T cells correlate with CD8 T-cell impairment and poor survival in hepatocellular carcinoma patients. Gastroenterology 2007, 132, 2328–2339. [Google Scholar] [CrossRef]
  127. Rech, A.J.; Mick, R.; Martin, S.; Recio, A.; Aqui, N.A.; Powell, D.J.; Colligon, T.A.; Trosko, J.A.; Leinbach, L.I.; Pletcher, C.H.; et al. CD25 blockade depletes and selectively reprograms regulatory T cells in concert with immunotherapy in cancer patients. Sci. Transl. Med. 2012, 4, 134ra162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Sutmuller, R.P.; van Duivenvoorde, L.M.; van Elsas, A.; Schumacher, T.N.; Wildenberg, M.E.; Allison, J.P.; Toes, R.E.; Offringa, R.; Melief, C.J. Synergism of cytotoxic T lymphocyte-associated antigen 4 blockade and depletion of CD25(+) regulatory T cells in antitumor therapy reveals alternative pathways for suppression of autoreactive cytotoxic T lymphocyte responses. J. Exp. Med. 2001, 194, 823–832. [Google Scholar] [CrossRef] [Green Version]
  129. Dal Bo, M.; De Mattia, E.; Baboci, L.; Mezzalira, S.; Cecchin, E.; Assaraf, Y.G.; Toffoli, G. New insights into the pharmacological, immunological, and CAR-T-cell approaches in the treatment of hepatocellular carcinoma. Drug Resist. Update 2020, 51, 100702. [Google Scholar] [CrossRef]
  130. Wang, Z.; Wu, Z.; Liu, Y.; Han, W. New development in CAR-T cell therapy. J. Hematol. Oncol. 2017, 10, 53. [Google Scholar] [CrossRef] [Green Version]
  131. Burga, R.A.; Thorn, M.; Point, G.R.; Guha, P.; Nguyen, C.T.; Licata, L.A.; DeMatteo, R.P.; Ayala, A.; Joseph Espat, N.; Junghans, R.P.; et al. Liver myeloid-derived suppressor cells expand in response to liver metastases in mice and inhibit the anti-tumor efficacy of anti-CEA CAR-T. Cancer Immunol. Immunother. 2015, 64, 817–829. [Google Scholar] [CrossRef] [Green Version]
  132. Gao, H.; Li, K.; Tu, H.; Pan, X.; Jiang, H.; Shi, B.; Kong, J.; Wang, H.; Yang, S.; Gu, J.; et al. Development of T cells redirected to glypican-3 for the treatment of hepatocellular carcinoma. Clin. Cancer Res. 2014, 20, 6418–6428. [Google Scholar] [CrossRef] [Green Version]
  133. Thomson, A.W.; Ezzelarab, M.B. Regulatory dendritic cells: Profiling, targeting, and therapeutic application. Curr. Opin. Organ. Transplant. 2018, 23, 538–545. [Google Scholar] [CrossRef]
  134. Morelli, A.E.; Thomson, A.W. Orchestration of transplantation tolerance by regulatory dendritic cell therapy or in-situ targeting of dendritic cells. Curr. Opin Organ. Transplant. 2014, 19, 348–356. [Google Scholar] [CrossRef] [Green Version]
  135. Thomson, A.W.; Humar, A.; Lakkis, F.G.; Metes, D.M. Regulatory dendritic cells for promotion of liver transplant operational tolerance: Rationale for a clinical trial and accompanying mechanistic studies. Hum. Immunol. 2018, 79, 314–321. [Google Scholar] [CrossRef] [PubMed]
  136. van der Heide, D.; Weiskirchen, R.; Bansal, R. Therapeutic Targeting of Hepatic Macrophages for the Treatment of Liver Diseases. Front. Immunol. 2019, 10, 2852. [Google Scholar] [CrossRef] [Green Version]
  137. Tacke, F.; Zimmermann, H.W. Macrophage heterogeneity in liver injury and fibrosis. J. Hepatol. 2014, 60, 1090–1096. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Triantafyllou, E.; Woollard, K.J.; McPhail, M.J.W.; Antoniades, C.G.; Possamai, L.A. The Role of Monocytes and Macrophages in Acute and Acute-on-Chronic Liver Failure. Front. Immunol. 2018, 9, 2948. [Google Scholar] [CrossRef]
  139. Loomba, R.; Lawitz, E.; Mantry, P.S.; Jayakumar, S.; Caldwell, S.H.; Arnold, H.; Diehl, A.M.; Djedjos, C.S.; Han, L.; Myers, R.P.; et al. The ASK1 inhibitor selonsertib in patients with nonalcoholic steatohepatitis: A randomized, phase 2 trial. Hepatology 2018, 67, 549–559. [Google Scholar] [CrossRef] [PubMed]
  140. Pedroza, M.; To, S.; Smith, J.; Agarwal, S.K. Cadherin-11 contributes to liver fibrosis induced by carbon tetrachloride. PLoS ONE 2019, 14, e0218971. [Google Scholar] [CrossRef] [Green Version]
  141. Maradana, M.R.; Yekollu, S.K.; Zeng, B.; Ellis, J.; Clouston, A.; Miller, G.; Talekar, M.; Bhuyan, Z.A.; Mahadevaiah, S.; Powell, E.E.; et al. Immunomodulatory liposomes targeting liver macrophages arrest progression of nonalcoholic steatohepatitis. Metabolism 2018, 78, 80–94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Rochigneux, P.; Chanez, B.; De Rauglaudre, B.; Mitry, E.; Chabannon, C.; Gilabert, M. Adoptive Cell Therapy in Hepatocellular Carcinoma: Biological Rationale and First Results in Early Phase Clinical Trials. Cancers 2021, 13, 271. [Google Scholar] [CrossRef] [PubMed]
  143. Siefert, J.; Hillebrandt, K.H.; Moosburner, S.; Podrabsky, P.; Geisel, D.; Denecke, T.; Unger, J.K.; Sawitzki, B.; Gul-Klein, S.; Lippert, S.; et al. Hepatocyte Transplantation to the Liver via the Splenic Artery in a Juvenile Large Animal Model. Cell Transplant. 2019, 28, 14S–24S. [Google Scholar] [CrossRef] [PubMed]
  144. Ridola, L.; Bragazzi, M.C.; Cardinale, V.; Carpino, G.; Gaudio, E.; Alvaro, D. Cholangiocytes: Cell transplantation. Biochim. Biophys. Acta Mol. Basis Dis. 2018, 1864, 1516–1523. [Google Scholar] [CrossRef] [PubMed]
  145. Nevi, L.; Carpino, G.; Costantini, D.; Cardinale, V.; Riccioni, O.; Di Matteo, S.; Melandro, F.; Berloco, P.B.; Reid, L.; Gaudio, E.; et al. Hyaluronan coating improves liver engraftment of transplanted human biliary tree stem/progenitor cells. Stem Cell Res. Ther. 2017, 8, 68. [Google Scholar] [CrossRef] [PubMed]
  146. Kuijk, E.W.; Rasmussen, S.; Blokzijl, F.; Huch, M.; Gehart, H.; Toonen, P.; Begthel, H.; Clevers, H.; Geurts, A.M.; Cuppen, E. Generation and characterization of rat liver stem cell lines and their engraftment in a rat model of liver failure. Sci. Rep. 2016, 6, 22154. [Google Scholar] [CrossRef]
  147. Sampaziotis, F.; Justin, A.W.; Tysoe, O.C.; Sawiak, S.; Godfrey, E.M.; Upponi, S.S.; Gieseck, R.L., 3rd; de Brito, M.C.; Berntsen, N.L.; Gomez-Vazquez, M.J.; et al. Reconstruction of the mouse extrahepatic biliary tree using primary human extrahepatic cholangiocyte organoids. Nat. Med. 2017, 23, 954–963. [Google Scholar] [CrossRef]
  148. Yang, L.; Cao, H.; Sun, D.; Hou, B.; Lin, L.; Shen, Z.Y.; Song, H.L. Bone marrow mesenchymal stem cells combine with normothermic machine perfusion to improve rat donor liver quality-the important role of hepatic microcirculation in donation after circulatory death. Cell Tissue Res. 2020, 381, 239–254. [Google Scholar] [CrossRef] [PubMed]
  149. Rigo, F.; De Stefano, N.; Navarro-Tableros, V.; David, E.; Rizza, G.; Catalano, G.; Gilbo, N.; Maione, F.; Gonella, F.; Roggio, D.; et al. Extracellular Vesicles from Human Liver Stem Cells Reduce Injury in an Ex Vivo Normothermic Hypoxic Rat Liver Perfusion Model. Transplantation 2018, 102, e205–e210. [Google Scholar] [CrossRef]
  150. Laing, R.W.; Stubblefield, S.; Wallace, L.; Roobrouck, V.D.; Bhogal, R.H.; Schlegel, A.; Boteon, Y.L.; Reynolds, G.M.; Ting, A.E.; Mirza, D.F.; et al. The Delivery of Multipotent Adult Progenitor Cells to Extended Criteria Human Donor Livers Using Normothermic Machine Perfusion. Front. Immunol. 2020, 11, 1226. [Google Scholar] [CrossRef]
  151. Sampaziotis, F.; Muraro, D.; Tysoe, O.C.; Sawiak, S.; Beach, T.E.; Godfrey, E.M.; Upponi, S.S.; Brevini, T.; Wesley, B.T.; Garcia-Bernardo, J.; et al. Cholangiocyte organoids can repair bile ducts after transplantation in the human liver. Science 2021, 371, 839–846. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Histoarchitecture of the liver. The portal triad is composed of the bile duct (A), the portal vein (B), and the hepatic artery (C) [8]. The blood flows from the portal vein and hepatic artery through the sinusoids (E) towards the central vein (F), surrounded by fenestrated LSECs. Lymphocytes and KCs reside intravascular, whereas SCs are found in the space of Disse (D) [1]. BECs line the bile duct, where they modify the bile produced by hepatocytes. The junctional region between the bile duct and hepatocytes, known as the canal of Hering (G), harbors bipotential BECs [12].
Figure 1. Histoarchitecture of the liver. The portal triad is composed of the bile duct (A), the portal vein (B), and the hepatic artery (C) [8]. The blood flows from the portal vein and hepatic artery through the sinusoids (E) towards the central vein (F), surrounded by fenestrated LSECs. Lymphocytes and KCs reside intravascular, whereas SCs are found in the space of Disse (D) [1]. BECs line the bile duct, where they modify the bile produced by hepatocytes. The junctional region between the bile duct and hepatocytes, known as the canal of Hering (G), harbors bipotential BECs [12].
Ijms 22 10276 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Hofmann, J.; Hackl, V.; Esser, H.; Meszaros, A.T.; Fodor, M.; Öfner, D.; Troppmair, J.; Schneeberger, S.; Hautz, T. Cell-Based Regeneration and Treatment of Liver Diseases. Int. J. Mol. Sci. 2021, 22, 10276. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms221910276

AMA Style

Hofmann J, Hackl V, Esser H, Meszaros AT, Fodor M, Öfner D, Troppmair J, Schneeberger S, Hautz T. Cell-Based Regeneration and Treatment of Liver Diseases. International Journal of Molecular Sciences. 2021; 22(19):10276. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms221910276

Chicago/Turabian Style

Hofmann, Julia, Verena Hackl, Hannah Esser, Andras T. Meszaros, Margot Fodor, Dietmar Öfner, Jakob Troppmair, Stefan Schneeberger, and Theresa Hautz. 2021. "Cell-Based Regeneration and Treatment of Liver Diseases" International Journal of Molecular Sciences 22, no. 19: 10276. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms221910276

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop