Next Article in Journal
Integration of Human Protein Sequence and Protein-Protein Interaction Data by Graph Autoencoder to Identify Novel Protein-Abnormal Phenotype Associations
Previous Article in Journal
Contrasting Roles of Ethylene Response Factors in Pathogen Response and Ripening in Fleshy Fruit
Previous Article in Special Issue
Liver Regeneration by Hematopoietic Stem Cells: Have We Reached the End of the Road?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cellular Therapies in Pediatric Liver Diseases

Pediatric Liver GI and Nutrition Center and Mowat Labs, Institute of Liver Studies, King’s College London and King’s College Hospital, London SE5 9RS, UK
*
Author to whom correspondence should be addressed.
Cells 2022, 11(16), 2483; https://doi.org/10.3390/cells11162483
Submission received: 16 June 2022 / Revised: 30 July 2022 / Accepted: 6 August 2022 / Published: 10 August 2022

Abstract

:
Liver transplantation is the gold standard for the treatment of pediatric end-stage liver disease and liver based metabolic disorders. Although liver transplant is successful, its wider application is limited by shortage of donor organs, surgical complications, need for life long immunosuppressive medication and its associated complications. Cellular therapies such as hepatocytes and mesenchymal stromal cells (MSCs) are currently emerging as an attractive alternative to liver transplantation. The aim of this review is to present the existing world experience in hepatocyte and MSC transplantation and the potential for future effective applications of these modalities of treatment.

1. Introduction

Liver transplantation remains the accepted treatment for end-stage liver disease and many liver-based metabolic disorders. However, there remain constraints of scarcity of donor livers, high costs involved in transplantation, risks of complications post-operatively and the need for lifelong immunosuppression. The impact of liver transplantation on quality of life cannot be underestimated with a recent study suggesting that only 26% of a pediatric cohort achieved “meaningful survival” 20-years post-transplantation [1]. The advent of auxiliary liver transplantation, where a partial liver lobe is transplanted whilst leaving the native liver in situ, has shown that whole liver replacement is not necessary for the restoration of liver function in children with acute liver failure. Up to 70% of these patients can be weaned off immunosuppression due to spontaneous native liver regeneration. [2]. Hence alternative therapies like cellular therapies, gene therapies and use of small molecules are under constant review as alternatives to whole liver replacement.
Human primary hepatocyte transplantation (HTx) is the primary form of cell therapy in paediatric liver disease, having been studied extensively in animal models of human liver disease with several clinical series of human hepatocyte transplantation with encouraging results, for over 30 years. It is however, limited by the shortage of donor tissues, from which hepatocytes of good quality can be isolated, as well as the impact of rejection against allogenic cell graft in the longer term and the need for immunosuppression when used for the treatment of liver based metabolic disorders.
As such, other modalities of cell therapy are being considered, predominantly focusing on the therapeutic potential of stromal cells. Their use is particularly appealing due to the fact that the cells are readily available and have the potential to expand in vitro and in vivo. Some cells can also be isolated from the recipient itself with auto-transplantation avoiding the need for immunosuppression.
The aim of this review is to summarize the current state of the art for cell therapies in pediatric liver disease. We will highlight the advantages and disadvantages of each cellular modality, explore its present use in clinical trials and how potential limitations can be addressed.
Our review is restricted to primary human hepatocytes and MSC, which has the most current evidence in the pediatric population and not stem cell generated hepatocytes.

2. Hepatocyte Transplantation (HTx)

2.1. Biology and Origins

The hepatocyte is the functional unit of the liver, performing synthetic and detoxification functions. HTx has emerged as an alternative treatment modality to liver transplantation, providing missing hepatic function to patients once engrafted. Hepatocytes are generally isolated from donor livers unsuitable for transplantation due to prolonged warm or cold ischaemia times, steatosis, anatomic disparities and lately from neonatal donors. Cells are isolated under good manufacturing practice (GMP) using a three-step collagenase perfusion technique, which has been previously described [3]. Thereafter, hepatocytes are separated from other non-parenchymal cells using low speed centrifugation. Isolated hepatocytes can be effectively used either fresh or cryopreserved for later utilization
There are many advantages of HTx over orthotopic liver transplantation. It is less invasive and more economical, without the need for complex surgery. It also has the advantage of keeping the native liver in situ, allowing for potential future option of gene therapy or use of small molecule-based therapies. HTx can also be repeated if necessary, and has the advantage of being readily available, particularly when using cryopreserved cells. Physiological benefit is postulated when 5–10% of the liver mass is replaced, and thus multiple patients can benefit from one donor liver, saving other organs for patients who may benefit only from liver transplantation [4].
In 1977, Groth et al. demonstrated a significant decrease in hyperbilirubinaemia in a rat model for Crigler-Najjar syndrome Type I, 28 days after HTx [5]. A number of pre-clinical trials later, led to the first in human study of HTx in 1992. 10 patients with liver cirrhosis received autologous hepatocytes, but with no clear benefit [6].

2.2. HTx in Paediatric Liver Disease

Multiple clinical series have since been reported in patients with liver disease, with the most significant effects seen in patients with metabolic disorders (Table 1).

2.2.1. HTx for the Treatment of Metabolic Disorders

Crigler-Najjar syndrome, phenylketonuria and factor VII deficiency are all liver based single gene defects, which lead to a reduced or absent function of the encoded gene product. It has been estimated that a cell engraftment corresponding to ~ 5–10% of liver mass is sufficient to overcome the gene defect and improve clinical outcome [7]. Although liver transplantation is the only curative therapy for these diseases, HTx has a promising treatment potential by replacing the diseased hepatocytes and restoring necessary function. In 1998, the first sustained effect of HTx for a single gene defect was demonstrated in a 10-year-old girl with Crigler-Najjar syndrome Type I who was on the liver transplantation waiting list. After a dose of 7.5 × 109 hepatocytes, partial correction of hyperbilirubinemia was achieved and sustained for 11 months [8]. Since then, HTx has been used to treat numerous other genetic liver diseases with varying success (Table 1).
In our experience at the King’s College Hospital, London, similar to others, HTx for the treatment of liver-based metabolic defects has resulted in varying successes. We have shown promising success in using HTx for the treatment of Crigler-Najjar syndrome [9] ornithine transcarbamylase (OTC) deficiency [10], and inherited factor VII deficiency [9]. Although our HTx metabolic program is suitable as a bridge to transplantation, new approaches which can improve hepatocyte engraftment and sustained functionality are needed and are under development before HTx can become a standard of care for liver-based metabolic diseases.

2.2.2. HTx for Treatment of Acute Liver Failure (ALF)

HTx has also shown some promising clinical success in the treatment of acute liver failure (ALF). ALF is a rapid decline in liver function over the course of days or weeks. Once ALF progresses in severity, the only medical treatment available is orthotopic liver transplantation, without which survival rates are very low [11]. HTx may be an effective treatment option whilst the patient waits for a donor liver to become available. HTx has been used to treat various cases of ALFs which have occurred following Dilantin [12], halothane [13] and multiple polysubstance misuse [12] showing promising improvements in encephalopathy and ammonia concentrations (Table 2). When human foetal hepatocytes were used to treat ALF, the overall survival of the treated group was 43% compared to 33% in the matched controls [14].
Although utilizing HTx for the treatment of ALF appears promising, it requires hepatocytes to be transplanted into an extremely harsh environment containing high levels of cellular necrosis and apoptosis [15]. As such, novel approaches are being examined to overcome this harsh microenvironment including implanting cells in alternative sites such as the peritoneum [16] and lymph nodes [17].
Another strategy to protect transplanted hepatocytes from the hostile microenvironment of a failing liver is to encapsulate the implanted cells within a biocompatible polymer. We have developed a technique using alginate to form hepatocyte microbeads [18] which can be infused into the peritoneal cavity of the patient, to temporarily replace the failing liver until regeneration can occur. These microbeads have been shown to be safe and, importantly, demonstrated promising efficacy in a pediatric cohort with ALF [19]. Since then, we have refined our hepatocyte microbead prototype. Progressing to a microbead which now involves multiple cell types and an improved hydrogel that better supports the cell function in vitro as well as in vivo, in preclinical studies (unpublished). Furthermore, a recent study has shown these types of alginate microbeads containing hepatocytes can be cryopreserved with some maintenance of hepatic functions once thawed, indicating the possibility of an off-the-shelf product being available for rapid treatment of ALFs [20]. These improved beads are currently being tested in Phase I/II clinical trial to show safety and efficacy (EudraCT Number: 2019-003916-29).
Table 1. Hepatocyte transplantation: clinical studies in patients with inborn errors of metabolism.
Table 1. Hepatocyte transplantation: clinical studies in patients with inborn errors of metabolism.
ReferenceLiver DiseasePatientsCell TypeRouteDoseOutcome
(Fox et al., 1998) [8]Crigler-Najjar syndrome type 11 female child (10 years)Fresh primary hepatocytes (5-year-old donor)Portal Vein7.5 × 107OLT after 4 years
(Darwish et al., 2004) [21]1 female child (8 years)Both fresh and cryopreserved primary hepatocytesPortal Vein7.5 × 107
(9 injections over 5 months)
OLT after 20 months
(Ambrosino et al., 2005) [22]1 male child (9 years)Fresh primary hepatocytes (47-year-old donor)Portal Vein7.5 × 107OLT after 5 months
(Dhawan et al., 2006) [23]1 male child (18 months); 1 female child (3 years)Cryopreserved primary hepatocytesPortal Vein4.3 × 107
2.1 × 107
OLT after 8 months
(Allen et al., 2008) [24]1 female child (8 years)Fresh primary hepatocytes (7-year-old donor)Portal Vein1.4 × 107OLT after 11 months
(Lysy et al., 2008) [25]1 female child (9 year); 1 female child (1 year)Both fresh and cryopreserved primary hepatocytesPorth-a-cath in jejunal vein; Broviac in portal vein6.1 × 107 (18 infusions from 3 different donors)
2.6 × 107 (14 infusions from 1 donor)
OLT after 6 months
OLT after 4 months
(Grossman et al., 1995) [26]Familial hypercholesterolemiaFive patients (7–41 years)Fresh primary hepatocytes transduced through retrovirus-mediated gene transfer for LDLR genePortal Vein1.0–3.2 × 107Variable and transient response
(Dhawan et al., 2004) [9]Factor VII deficiency1 child (3 months); 1 child (3 years)Both fresh and cryopreserved primary hepatocytesPortal Vein1.1 × 107
2.2 × 107
OLT after 7 months
OLT after 8 months
(Muraca et al., 2002) [27]Glycogen storage disease Type I1 female adult (47 years)Fresh primary hepatocytesPortal Vein2 × 1079 months after trans- plantation, patient on normal diet and can fast for 7 h without experiencing hypoglycaemia
(Lee et al., 2007) [28]1 male adult (18 years)Both fresh and cryopreserved primary hepatocytesPortal Vein2 × 109 for first infusion; 1 × 109 for second and 3 × 109 for final infusion250 days after HTx patient on a normal diet
(Sokal, Smets, Bourgois, Van Maldergem, et al., 2003) [29]Infantile Refsum’s disease1 female child (4 years)Both fresh and cryopreserved primary hepatocytesPortal Vein1.1 × 109 for first infusion; 1.4 × 108 and 9 × 107 on day 3, 1.84 × 108 and 2.43 × 108 on day 4, and 1.96 × 108 on day 5Continued metabolic improvement 1 year after HTx
(Dhawan et al., 2006) [23]Progressive familial intrahepatic cholestasis Type 22 children (18 months and 3 years)Fresh primary hepatocytesPortal Vein0.2 × 107
0.4 × 107
OLT after 5 months
OLT after 14 months
(S. C. Strom et al., 1997) [13]OTC deficiency1 male child (5 years)Fresh primary hepatocytesPortal Vein1 × 107Death 42 days later
(Horslen et al., 2003) [30,31]1 male child (10 h old)Both fresh and cryopreserved primary hepatocytesPortal Vein4 × 109 for first infusion; further 3.3 × 109 between days of life 37 and 51; 1.7 × 109 between days 113 and 116OLT at 6 months
(Stéphenne et al., 2005) [28]1 male child (14 months)Cryopreserved primary hepatocytesPortal Vein2.4 × 107
10 infusions over 16 weeks
OLT after 6 months
(Puppi et al., 2008) [10]1 male child (1 day)Both fresh and cryopreserved primary hepatocytesPortal Vein1.74 × 109; 7 infusions over the first month of life and 1 infusion at 5 months.APOLT at 7 months
(Meyburg et al., 2009) [32]1 male child (6 h); 1 male child (9 days)Cryopreserved primary hepatocytes from one donor (9 days old)Portal Vein9.4 × 108 in 3 infusions; 8.7 × 108 in 2 infusionsDeath at 4 months;
Listed for OLT 5 months after HTx
(Stéphenne et al., 2006) [33]ASL deficiency1 female child (3 years)Both fresh and cryopreserved primary hepatocytesPortal Vein1.7 × 109 in 7 infusions over 1 month period; 2.5 months after first infusion patent received a further 10 × 109 cells over 2 days; two months later a further 1× 109 cellsOLT after 18 months
(Meyburg et al., 2009) [32]CPS1 deficiency1 male child (10 weeks)
Both fresh and cryopreserved primary hepatocytesPortal Vein1.87 × 109 over 6 infusionsListed for OLT 7 months after HTx
(Meyburg et al., 2009) [32]Citrullinemia1 female child (3 years)Both fresh and cryopreserved primary hepatocytesPortal Vein1.89 × 109 over 4 infusionsProtein intake could be increased 10 months after HTx
Table 2. Hepatocyte transplantation: clinical studies in patients with Acute Liver Failure; OLT Orthotopic liver transplantation.
Table 2. Hepatocyte transplantation: clinical studies in patients with Acute Liver Failure; OLT Orthotopic liver transplantation.
ReferenceLiver DiseasePatients TreatedCell TypeRouteDoseOutcome
(Soriano et al., 1997) [34]Drug-induced liver failure16 years; 12 years; 10 yearsCryopreserved primary hepatocytesPortal Vein4 × 107–4 × 109Death on day 2; Death on day 7; Death on day 7
(Bilir et al., 2000) [35]1 female adult (32 years); 1 male adult (35 years); 1 male adult
(55 years)
Cryopreserved primary hepatocytesIntrasplenic1.3 × 109; 1 × 1010; 3.9 × 1010Death on day 14;
Death on day 20;
Death in 6 h
(Strom et al., 1999) [36]1 female teenager (13 years); 1 female adult
(43 years)
NAPortal VeinNADeath on day 4;
Death on day 35
(Fisher and Strom 2006) [37]1 female adult (27 years); 26 years; 21 years; 35 years; 35 years; 51 yearsNAIntrasplenic; Intrasplenic; Intrasplenic; Portal Vein; Portal Vein; Portal Vein2.8 × 107; 3 1.2 × 109; 3 infusions of 9 × 108,
9 × 108
and 2.5 × 107; 5.4 × 109; 3.7 × 109; 3.9 × 109
OLT on day 10;
OLT on day 2;
Death on day 1;
Death on day 18;
Full recovery;
Death on day 3
(Habibullah et al., 1994) [14]1 female adult (32 years); 1 male adult (29 years); 1 female adult (20 years); 1 female adult (20 years); 1 female adult (24 years)Fresh foetal hepatocytesIntraperitoneal6 × 107 kg body weightDeath in 30 h;
Death in 37 h;
Death in 48 h;
Full recovery;
Full recovery
(Fisher and Strom 2006) [37]Viral-induced acute liver failure4 years; 54 yearsNAPortal Vein2 infusions of 1.7 × 109; 6.6 × 109Death on day 2;
Death on day 7
(Bilir et al., 2000b) [35]1 female adult (29 years); 1 female adult (65 years)Cryopreserved primary hepatocytesPortal Vein and intrasplenic1 × 1010; 3 × 1010Death in 18 h.
Death on day 52
(Strom et al., 1999) [36]1 female adult (28 years); 1 female adult (37 years); 1 male adult
(43 years)
NAIntrasplenic; intrasplenic; Portal VeinNAOLT on day 3;
Death on day 5;
OLT on day 1
(Fisher et al., 2000b) [12]1 female adult (37 years)NAintrasplenic1.2 × 108Full recovery
(Habibullah et al., 1994b) [14]1 female adult (40 years)Fresh foetal hepatocytesIntraperitoneal
6 × 107/kg body weightDeath in 13 h
(Stephen C. Strom et al., 1997b) [38]1 female adult (40 years)Cryopreserved primary hepatocytesIntrasplenic7.5 × 106Death on Day 4 due to ICP monitor complications
(Soriano et al., 1997)Idiopathic acute liver failure3 years; 5 yearsCryopreserved primary hepatocytesPortal Vein4 × 109Full recovery;
OLT on day 4
(Fisher and Strom 2006) [37]3.5 months; 23 years; 48 yearsNAPortal Vein; intrasplenic; Portal Vein1.8 × 108; 2.86 × 108; 7.5 × 108OLT on day 1;
OLT on day 5 and death on day 13;
Death on day 1
(Habibullah et al., 1994a) [14]1 male child (8 years)Fresh foetal hepatocytesIntraperitoneal
60 × 106/kg body weightFull recovery
(Schneider et al., 2006) [39]Mushroom-poisoning-induced acute liver failure1 female (64 years)Cryopreserved primary hepatocytesPortal Vein8 × 109Full recovery
(Strom et al., 1999) [36]
Postsurgical acute liver failure1 male (69 years)NAIntrasplenicNADeath on day 2
(Khan et al., 2004) [40]Acute liver failure induced by acute fatty liver of pregnancy1 female (26 years)Fresh Foetal hepatocytesIntraperitoneal 3 × 108Full recovery
(Stephen C. Strom et al., 1997b) [38]Alpha 1 anti-trypsin1 female adult (52 years)Cryopreserved primary hepatocytesIntrasplenic2.2 × 107OLT on Day 2
(Stephen C. Strom et al., 1997b) [38]TPN/Sepsis1 male child (6 months)Cryopreserved primary hepatocytesIntrasplenic5.2 × 107Life support stopped on day 7

2.3. Future Directions of HTx

The routes of transplantation that have been used include intraportal, intraperitoneal or intrasplenic administration, with most centers using a dose of up to 2 × 108 cells/kg of body weight (into the portal circulation), as the recommended upper limit per infusion, with multiple infusions being done when higher number of cells are required. Whilst the safety has been well described with good short-term outcomes, the longer term improvement of hepatic function is suboptimal due to the poor engraftment of cells and rejection, in spite of immunosuppression [41].
The issue around cell engraftment is influenced by the quality of hepatocytes that are isolated, which tends to be poor particularly when they are isolated from liver tissues that are rejected for transplantation for prolonged ischaemic time, severe fatty liver, non-heart beating and older donors. It is important to evaluate the cell quality prior to transplantation—with many centers accepting cell viability of equal or higher than 60% for clinical uses using trypan blue. Other, more specific assays to detect apoptosis and metabolic function are also increasingly being used.
Good cell engraftment is key to HTx success. In an effort to enhance hepatocyte engraftment, multiple strategies have been carried out to precondition the recipient’s liver and give a selective advantage to the transplanted cells such as portal vein embolization—a safe and well-tolerated procedure [42], partial hepatectomy—used in two patients with Crigler-Najjar syndrome Type 1 [43] and liver irradiation—a promising new preconditioning approach [44]. These techniques have been used to varying degrees of success and a better understanding of the utility of these techniques would probably improve outcomes of HTx.
Transplanted hepatocyte loss is another important consideration in the failure of HTx. Allogenic hepatocytes are immunogenic, unlike the immune-privileged liver, which allows for graft tolerance and withdrawal of immunosuppression in some cases. Both the innate and adaptive immunity play an important role in the rejection of transplanted hepatocytes. A number of immunosuppressive protocols have been utilized and adapted for HTx, mainly consisting of steroids and calcineurin inhibitors, to varying degrees of success. The instant blood mediated inflammatory response (IBMIR) is a significant phenomenon that leads to substantial cell loss when infused into the portal circulation. Alpha-one –antitrypsin use in the hepatocyte infusion has shown promise to dampen this response.
The optimized protocol for production of GMP-grade alginate encapsulated microencapsulated hepatocytes, which have a semi-permeable polymerized structure, protects the cells from host immune attack. We previously demonstrated that the ultrapure sodium alginate that is used does not elicit immune activation in vitro. This approach allows cell transplantation without using immunosuppression [18].

3. Mesenchymal Stromal Cells (MSC) Transplantation

3.1. Biology and Origins

Friedenstein et al. first isolated and identified MSCs in 1968 [45]. They are a subtype of adult fibroblast-like cells with the potential for self-renewal and a high proliferative ability. They were originally identified in the bone marrow, but because of the small number of cells that are available (0.01 to 0.001% of total bone marrow cells) [46] and the difficulty of isolating cells from the bone marrow, scientists have explored alternative sources. They have been successfully isolated from different tissue sources including synovial membrane [47], adipose tissue (AT) [48], umbilical cord (UC) blood, amniotic fluid (AF) [49] and the placenta [50]. The common feature of MSCs is the ability to differentiate into adipocyte, cartilage and osteogenic tissue. UC sources of MSCs have been of particular interest, due to the various compartments that it can be isolated from including the umbilical vein, arteries and Wharton’s jelly as well as the fact that higher quantities of more primitive MSCs that are isolated from UC tissue compared to other tissues sources. It has also been shown that MSCs isolated from UC tend to exhibit a higher proliferative capacity compared to MSCs obtained from other sources [51]. There are significant differences in the MSCs differentiation potential from different sources, despite having similar phenotypic and antigenic profiles [52,53].

3.2. Mechanisms of Action in Liver Disease

3.2.1. Immunomodulation by MSCs

MSCs have the potential to modulate and repair injured tissue by changing toxic immune response through a range of mechanisms including direct cell–cell interactions with injured cells or remotely, through the release of paracrine factors [54]. MSCs have the added advantage of having reduced immunogenicity due to the lack of expression of Class II major histocompatibility antigens (MHC) when unprimed and the cells do not express the majority of molecules that are detectable for immune recognition such as CD80, CD86 and CD40.

Immuno-Modulatory Effect of MSCs on Adaptive Immunity

T-cell proliferation is inhibited in vitro by MSCs through the secretion of soluble factors or by the direct interaction with T-lymphocytes [55]. Molecules such as transforming growth factor β (TGF- β), hepatocyte growth factor, prostaglandin E2 (PGE2) [56] and indoleamine 2,3-dioxygenase [57] are secreted by MSCs with an immunomodulatory effect on T-cell activities. The secretion of these immunomodulatory molecules can differ according to the source of the MSCs [58]. The secretion of nitric oxide (NO) by MSCs also cause inhibition of the STAT5 pathways leading to the suppression of T cell proliferation [59]. The activation of T-cells is also thought to be suppressed by the release of matrix metalloproteinases (MMP) such as MMP-2 and MMP-9 [60]. MSCs also affect the generation and development of regulatory T-cells (T-regs), which can have a positive impact on the balance of immune damage during tissue injury. It is also worth noting that the inflammatory environment is particularly important in the interaction between MSCs and T-cells.
MSCs also play a role in the inhibition of B cell proliferation thereby reducing their production of immunoglobulin. In a study by Glennie et al., murine B cell proliferation was induced with CD40 and IL-4, but subsequent co-culture with MSCs significantly inhibited their proliferation [61]. There was also a consequent stimulation in immunoglobulin production after co-culture of B-cells [62]. They also tend to alter surface expression of chemokine receptors on B-cells—particularly in the expression of CXCR4, which has a role in homing and fate of MSCs [63].
Viral infections and tumor cells induce an immune response particularly involving natural killer cells [64]. The release of soluble factors such as PGE2 and TGF-β from MSCs as well as the cell–cell interactions has been shown to reduce IL-15 secretion from NK cells [55].

Immuno-Modulatory Effect of MSCs on Innate Immunity

Studies have suggested that MSCs trigger polarization of classical pro-inflammatory macrophages (M1) toward alterative macrophages (M2) that secrete anti-inflammatory cytokines in vivo and in vitro [65]. This polarization is secondary to the ability of MSCs to secrete soluble factors such as interleukin-10 (IL-10) and IL-1Ra, which have been shown to dampen liver injury by promoting M2 macrophage polarization [66]. Apart from this, MSCs also have the potential of increase survival of monocytes through the upregulation of CCL18, which indirectly mediates the ability of induce Treg formation by MSCs [67].
MSCs also affect dendritic cell function by blocking differentiation of antigen-presenting cells to monocytes and decreasing their expression of anti-inflammatory molecules such as IL-12, TNFα, and IFNγ, whilst also increasing the secretion of IL-10 which can induce regulatory T-cell numbers [68].

3.2.2. Anti-Fibrotic Activities of MSCs

The inflammatory and fibrotic processes in the liver are closely intertwined. In liver injury, pro-fibrotic factors such as TGF-β, platelet derived growth factor (PDGF), IL-13 and IL-4, are secreted by the resident immune cells. These play a crucial role in the activation and proliferation of hepatic stellate cells (HSCs), which transform into myofibroblasts. Myofibroblasts are responsible for the production of the extracellular matrix (ECM) in the liver [69,70,71,72]. The deposition of ECM, including collagen I, collagen III and collagen IV are pivotal in causing liver fibrosis.
The anti-fibrotic effects of MSCs can be divided into the direct and indirect effects on HSCs. Indirectly, MSCs work by modulating immune cell activity to regulate the activity of HSCs. MSCs tend to migrate to the sites of injury where they are exposed to inflammatory cytokines such as IFNγ and IL-1β [73]. There, the MSCs work by secreting various soluble mediators such as NO, PGE2, IDO, IL-6, IL-10 and HLA-γ, which results in the suppression of proliferation and activation of a variety of immune cells, and induction of Treg cells [74]. MSCs suppress immune cell activity thereby reducing fibrogenic processes and reducing ECM accumulation in liver disease.
The direct anti-fibrotic effects of MSCs on HSCs involve the inhibition of ECM production potential of HSCs and the induction of apoptosis of HSCs. MSCs secrete IL-10, HGF, TGF-β3 and TNFα, which inhibit the proliferation of HSCs and decrease ECM synthesis [71]. During direct co-culture of MSCs and HSCs, the Notch pathway is activated with significant suppression of HSC proliferation and α-SMA expression. In liver fibrosis, the activated HSCs express the tissue inhibitors of metalloproteinase (TIMP)-1 and TIMP-2, whereas MSCs are known to increase the expression of MMPs. In experimental models, this is generally associated with the resolution of fibrosis [75].

3.2.3. Hepatocyte-Like Differentiation of MSCs

We have discussed the potential of HTx in improving liver function and mitigating fibrosis in preclinical and clinical studies. There are several factors that can influence the hepatic differentiation of MSCs. Firstly, when MSCs are co-cultured with hepatocytes they can differentiate into hepatocyte-like cells [76]. This has been reported in rats, mice, sheep and humans [77,78]. When MSCs are treated with a combination of several growth factors, chemical compounds and cytokines (such as HGF, fibroblast growth factor 2 epidermal growth factor, oncostatin M, dexamethasone, insulin-transferrin-selenium and/or nicotinamide), there is an increase in the expression of hepatocyte markers such as albumin, α-fetoprotein, CK18, GATA4, CK19, and HNF-3β [79]. It has also been noted that hepatic stem/progenitor cells which are isolated from the adult human liver have a better potential at differentiation into hepatocytes compared to MSCs that are isolated from tissues other than the liver [79]. Future studies are still warranted improve efficacy and consistency of hepatic differentiation from MSCs.

3.3. MSCs in Paediatric Liver Disease

MSCs have been used in various clinical settings with varying degrees of success, including in the treatment of degenerative and immune-mediated diseases. There has been a significant increase in the number of clinical trials investigating the use of MSCs in both acute and chronic liver disease (Table 3). The question remains over whether MSCs are more effective than the conventional standard of care and which liver conditions are best suited for MSC treatment.
The majority of trials that have been published thus far have been in adults. The majority of these studies are focused on liver cirrhosis (n = 15), whilst four studies are related to liver failure and three studies are for complications after liver transplantation. The common thread for all these diseases is that they are end-stage liver diseases and the only effective treatment at this stage is liver transplantation, which is limited by the availability of organs, surgical complications, the need for immunosuppression and the high medical and surgical costs. Therefore, an effective alternative therapy such as MSC therapy is warranted in this group of patients.
Currently there are over 1000 clinical trials (1586 trials in adults and 322 trials in the paediatric population) that are associated with MSC cell therapies that are registered with www.clinicaltrials.gov (accessed on 15 June 2022). Additionally, MSC cell therapy has been employed to treat Bronchopulmonary Dysplasia (BPD) in premature infants (22) which showed MSC treatment was safe and feasible in a preterm cohort and demonstrated a significant reduction in cytokine levels and lower severity of BPD. This poises MSCs as a strong candidate for use in a clinical trial for treating paediatric liver diseases such as BA, following a display of efficacy in this study (23–25).

3.3.1. MSC Therapy for Biliary Atresia

Biliary atresia (BA) is a progressive fibrosing obstructive cholangiopathy, involving the extrahepatic and intrahepatic biliary systems to varying degrees [80]. Clinical presentation tends to be in the neonatal period and is characterised by direct or conjugated hyperbilirubinaemia, acholic stools, dark urine, variable levels of hepatosplenomegaly and progressive liver failure. Infants who are untreated, rapidly develop progressive fibrosis, leading to portal hypertension and end-stage liver disease, invariably resulting in death within the first two years of life. It has an overall incidence of 1 in 15,000 to 1 in 20,000 in Europe and North America [81].
The aetiology of BA remains incompletely understood but it is likely that there are a number of different mechanisms and factors that contribute to the final common pathway that is identified as the BA phenotype. At King’s College Hospital, we have described a number of distinct variants including a syndromic form—biliary atresia splenic malformation (BASM), cystic BA and more recently, CMV IgM +ve associated BA [82]. The majority of cases, however, do not have any other defining characteristics and are referred to as isolated BA. Environmental and genetic susceptibility factors are thought to interact and orchestrate disease pathogenesis. The key factors that are involved are infectious and immunologic processes [83].
Pathologically, the histologic evidence of lymphomononuclear inflammatory cells infiltrating the vicinity of injured interlobular bile ducts and within the duct epithelium suggests, that in BA, immunologic mechanisms play an important role in bile duct damage [84]. Newly expressed or altered antigens on the surface of bile duct epithelium emerge after an initial viral or toxic insult to the biliary epithelium. This is then presented by macrophages to naïve T lymphocytes. Primed Th1 lymphocytes then organise an immune response through the release of proinflammatory cytokines (IL-2, IFN-γ, TNF-α and IL-12) and recruiting cytotoxic T-cells [85], ultimately causing bile duct epithelial injury. This eventually results in scarring and obliteration of the bile ducts seen in BA.
Early diagnosis is pivotal for the best outcomes from the surgical intervention that is known as Kasai portoenterostomy (KPE). The surgery involves the removal of the atretic extrahepatic biliary tree in an attempt to re-establish bile flow to the intestine by creating a Roux en-Y intestinal conduit. The outcome following this surgery is highly variable in the literature as well as in real world experience, with effective drainage achieved in just over 50% of children [86].
Unfortunately, even with KPE, significant progression of fibrosis and the development of cirrhosis occurs in the majority of patients, requiring liver transplant. BA remains the leading indication for liver transplantation (LT) in children, and at present, no other successful medical treatments have been identified. Despite the major drive to ameliorate the fibro-inflammatory progression in BA, to ultimately reduce the need for LT, no efficacious immunomodulatory treatment for BA is currently available.
Adjuvant medical therapies are a controversial area with arguably marginal evidence to support its use, with institutions around the world adopting individual protocols that are based on experience. As inflammation is thought to be a key factor in the pathogenesis of BA, corticosteroids have been trialled in multiple studies. A randomised, double blind, placebo-controlled trial using low-dose corticosteroids that was conducted in the UK with 71 infants showed no difference in the clearance of jaundice or improvement in the native liver survival/reduction in need for transplantation [87]. Intravenous immunoglobuline (IVIg) administration was trialled as an adjuvant therapy in BA patients in North America as a multicentre Phase Ib/II clinical trial, however failed to show any beneficial effect. As such, it is not routinely used in BA patients post-KPE [88]. None of the treatments above have shown significant improvements in terms of clearance of jaundice, native liver survival or reduction in liver transplantation rates. As such, a safe and efficacious treatment modality is pertinent in this group of patients to improve clinical outcomes.
MSCs as we discussed, preferentially home to damaged tissue, where they serve as a reservoir of growth factors and regenerative molecules. They initiate immunomodulation by targeting a range of innate and adaptive immune cells by direct interaction and by secreting soluble factors [89]. Furthermore, MSCs exhibit anti-fibrotic properties and can directly and indirectly inhibit hepatic stellate cell activation in biliary atresia which contributes to the ongoing biliary atresia. The low immunogenicity that MSCs possess enables allogeneic cells to be used as “off-the-shelf” products. Their safety for use as advanced therapy medicinal products (ATMP) has been widely evaluated.
Lei et al. investigated the anti-fibrotic potential of bone marrow-derived MSCs on liver fibrosis induced by BA [90]. An inflammatory cholangiopathy is induced in the murine model of BA using RRV infection in the newborn BALB/c mice. The pathological phenotype is similar to that in human BA. They demonstrated that BM-MSCs treatment significantly restored liver enzymatic function and bilirubin metabolism and inhibited oxidative stress and alleviated liver fibrosis of the RRV-induced murine model of BA [90]. Collagen IV and COL1A1 were also highly expressed in the liver tissue of the murine model group but BM-MSC treatment significantly reduced their expression. α-SMA expression was also elevated in the BA group but clearly reduced in the BM-MSC treatment group using immunohistochemical staining and Western blotting of liver samples. Pro- inflammatory factors such as TNF-α and TGF-β1 were also significantly inhibited in the BM-MSC group.
Currently there is one Phase I/II clinical trial registered investigating the use of UC-MSC transplantation in patients with BA. Two doses of 1 million MSCs per kg body weight that will be administered at an interval of 6 months.

3.3.2. MSC Therapy for Acute Liver Failure

In acute liver failure, there is rapid loss of function with tissue necrosis and treatment is focused on restoring liver function and preventing disease progression. MSCs have the potential to provide restoration of liver function [91]. The use of MSCs in ALF has been studied in pre-clinical models of mice, rats and monkeys. In a murine model of ALF secondary to acetaminophen poisoning, human UC-MSCs were transplanted intravenously resulting in significantly improved survival rates and alleviated hepatic injury [92]. In a rat ALF model, MSCs were shown to prevent the release of liver injury biomarkers, with recovery of the liver structure [93]. The transplantation of MSCs that were co-cultured with hepatocytes also provided better restoration of liver function, which results in lower levels of aspartate aminotransferase, total bilirubin and alanine aminotransferase (ALT). In another pre-clinical ALF non-human primate model, Guo et al. showed that the early infusion of UC-MSCs could significantly improve hepatic histology, systemic homeostasis and better survival [94]. IL-6 seems to play an important role in initiating and accelerating ALF, whilst UC-MSCs seem to disrupt this inflammatory cascade by inhibiting monocyte activation. Based on these studies, MSCs may have a significant beneficial impact in ALF or as a bridge to transplantation.
The potential of MSCs as a valid treatment modality in replacing hepatocytes in injured liver and effectively rescuing experimental liver failure and contributing to liver regeneration has been demonstrated by many studies (Table 3) and ongoing clinical trials (15 trials currently registered). More research is needed in the realms of sourcing of cells, dosages and the routes of administration to improve the outcomes of this treatment.

3.3.3. MSC Therapy for Cirrhosis

At the crux of liver fibrosis is the transdifferentiation of quiescent HSCs to myofibroblastic HSCs. Regulating HSC activation is a potential therapeutic target for liver fibrosis. As previously discussed, MSCs have the potential to inhibit HSC activation through suppression of their proliferation and stimulating apoptosis by increasing pro-apoptotic proteins. MSCs can also replace dying hepatocytes by differentiating into hepatocyte-like cells. MSCs also exert their anti-inflammatory properties by suppressing the synthesis of pro-inflammatory cytokines such as TNFα, IFNγ and IL-17 and promoting the production of anti-inflammatory cytokines such as IL-4 and IL-10.
Most clinical trials (Table 3) have shown that MSC-based therapies seem to have a beneficial effect on liver fibrosis. In adult studies of alcoholic liver cirrhosis, BM—MSCs significantly improved liver function and reduced the accumulation of collagen [95,96]. In liver cirrhosis secondary to HBV, MSCs were shown to alleviate the expression of fibrotic markers and significantly reduce MELD scores at 48 weeks, whilst also expressing immunomodulatory properties [97]. Whilst little to no side effects were reported in these clinical trials, two studies suggested no significant effect of MSCs in patients with liver cirrhosis [98,99].
Therefore, it remains necessary to conduct, larger-scale clinical trials (50 trials currently registered) covering various conditions leading to cirrhosis, using a range of doses and frequencies, and routes of administration to establish the effectiveness of MSCs in the treatment of liver cirrhosis.
Table 3. Mesenchymal stromal cell transplantation: clinical studies in patients with liver disease.
Table 3. Mesenchymal stromal cell transplantation: clinical studies in patients with liver disease.
ReferenceLiver DiseaseType of Clinical TrialNo of Patients TreatedNo of Control PatientsSource of MSCsRouteDoseOutcomeAdverse Events
Liver Cirrhosis
Mohamadnejad et al., 2007 [100]Decompensated liver cirrhosisI40Autologous, bone marrowIntravenous Peripheral vein31.73 × 106Improvement in MELD score n = 2, QoL improved in n = 4No side effects
Kharaziha et al., 2009 [101]End stage liver diseaseI–II80Autologous iliac crestIntravenous Peripheral vein or portal vein3–5 × 107, twiceImprovement in MELD scoreNo side effects
Zhang et al., 2012 [97]Decompensated liver cirrhosisPaired controlled study3015UC-MSCIntravenous, peripheral vein0.5 × 106 cells/kg bodyReduction in ascites, liver function and MELD scoreNo side effects
El-Ansary et al., 2012 [102]Liver cirrhosis secondary to hepatitis CPhase II RCT1510Bone marrow, autologousIntravenous1 × 106 cells/kg bodyImprovement in liver function, MELD scoreNo side effects
Mohamadnejad et al., 2013 [98]Decompensated cirrhosisRCT1512Bone marrow, autologousIntravenous, peripheral vein1.95 × 107 cellsNo significant differenceNo side effects
Amin et al., 2013 [103]Liver cirrhosis secondary to hepatitis CNo control group200Bone marrow, autologousIntrasplenic10 × 106Significant improvement in liver function testsNo side effects
Jang et al., 2013 [95]Alcoholic cirrhosisPhase II clinical trial110Autologous, bone marrowPeripheral veinNAImprovement in histological appearance, Child score and decrease of TGFb1,a-SMANo side effects
Wang et al., 2013 [104]Primary biliary cirrhosisSingle arm trial70UC-MSCsIntravenous, peripheral vein0.5 × 106 cells/kg, three timesImprovement in fatigue and pruritus, and decrease in alkaline phosphatase and GGTNo side effects
Salama et al., 2014 [105]End-stage liver disease Hepatitis CRCT2020Autologous, bone marrowPeripheral veinNAImprovement in synthetic function and liver function etstsNo side effects
Xu et al., 2014 [106]Liver cirrhosis Hepatitis BRCT2019Autologous, bone marrowHepatic arteryNAImprovement in liver function, increased Treg/Th17 ratioNo side effects
Kantarcioglu et al., 2015 [99]Liver cirrhosisNo control group120Autologous, bone marrowPeripheral vein, intravenous1 × 106 cells/kgImprovement in MELD score n = 8, no change in liver regeneration or fibrosis at 6mNo side effects
Suk et al., 2016 [96]Alcoholic liver cirrhosisPhase 2 RCT3718Autologous, bone marrowHepatic artery5 × 107
once vs twice
Improvement in fibrosis quantification and Child scoreNo side effects
Sakai et al., 2016 [107]Liver cirrhosisPhase I40Adipose tissue MSCHepatic artery6.6 × 105 cells/kgImprovement in liver function, HGF and IL6 increasedNo side effects
Liang et al., 2017 [108]Liver cirrhosis secondary to autoimmune diseaseNo control260UC-MSC, cord blood MSC, bone marrow MSCPeripheral intravenous1 × 106/kgImprovement in liver function and MELD scoreNo side effects
Fang et al., 2018 [109]Decompensated liver cirrhosis, hepatitis BRCT5053UC-MSCsPeripheral intravenous(4.0–4.5) × 108Improvement in Child score, MELD and liver functionNo sie effects
Liver Failure
Peng et al., 2011 [110]Hepatitis B liver failureRCT53105Autologous, bone marrowHepatic ArteryNAGood short term efficacy, no significant long term improvementNo side effects
Amer et al., 2011 [111]Hepatitis C liver failureRCT2020Autologous, bone marrowIntrasplenic, intrahepatic2 × 107 hepatic lineage committed cellsSigniifcant improvement in ascites, albumin, Child score, MELD scoreNo side effects
Shi et al., 2012 [112]Acute on chronic liver failureParallel controlled trial2419UC-MSCsIntravenous, peripheral vein0.5 × 106 UC-MSCs per kilogram, three timesSignificant increase in survival rates, improvement in MELD scoreNo side effects
Lin et al., 2017 [113]Acute on chronic liver failure, Hepatitis BRCT5456Allogenic, bone marrowPeripheral, intravenous1.0 to 10 × 105 cells/kg
four times
Improved 24 week survival, liver function and decrease in severe infectionsNo side effects
Liver Transplantation
Shi et al., 2017 [114]Allograft liver rejectionRCT1413UC-MSCsPeripheral intravenous1 × 106/kg body weight (once or multiple times in n = 1)Improvement in liver function, histology and increased Treg/Th17 ratioNo side effects
Detry et al., 2017 [115]Liver transplantationPhase I-II1019Allogenic, bone marrowPeripheral intravenous1.5–3 × 106/kgNo improvement in toleranceNo side effects
Zhang et al., 2016 [116]Ischaemic biliary lesion after liver transplanatationPhase 1 RCT1270UC-MSCIntravenous peripheral1.0 × 106 MSCs per kilogramImprovement in liver function test and improvement survivalNo side effects

3.4. Future Directions of MSC Therapy in Liver Disease

As described, multiple clinical trials have verified the safety of MSCs in liver disease with therapeutic efficacy. However, the main issue remains the homing ability of MSCs for which in-depth studies are underway to identify relevant mechanisms of MSC homing and to explore new strategies to improve this.
The key areas of focus include: (1) transplantation route: intravenous infusion of MSCs is easy, economical and can be performed on numerous occasions, but the proportion of cell colonization via this route is low. Portal vein injection may be suitable in patients without risk of portal hypertension, whilst non-systematic homing can be done through intrasplenic puncture injection, the amount is limited each time, and there is an increased risk of bleeding. Hepatic artery injection is also used but is not ideal for multiple treatments. There are still limitations in terms of routes of MSC transplantation and a more individualized case-by-case approach may be appropriate. (2) Optimization of MSC culture conditions: pre-treatment of MSCs have the potential to improve therapeutic efficacy of MSCs, but can affect the phenotypic and paracrine functions, therefore further research is required before clinical use. (3) Modifications of MSCs: this is an active area of research through gene editing and chemical modifications. These methods however may cause biosafety issues and preclinical studies are still needed to safely use these cells [117].
Whilst the safety has been established, the clinical application of MSCs in are still fragmented in terms of the cell source, dose, route, optimal time of inclusion and curative effect. MSCs in clinical use are typically cryopreserved and compared to freshly isolated MSCs, have poorer homing ability and shortens their durability, survival in vivo and tissue repair [118]. Further studies are still needed to investigate the homing mechanism of MSCs and various strategies to improve this, particularly in the pediatric population.

4. Conclusions: Future of Cell Therapy

In this review, we have explored the recent advances in studies using hepatocyte and MSC therapies. The use of these modalities requires further optimization from bench to bedside. Important existing issues including preparative measures, optimal timing of injections, longevity of transplanted cells and improved engraftment are key.
The future probably lies in cell-free transplantation. We believe that co-culture techniques like hepatocytes and MSCs could be replaced by small molecules or exosomes that mediate the hepatotrophic, anti-apoptotic and immunomodulatory effects of MSCs. MSCs are known to secrete a variety of factors such as cytokines/chemokines, free nucleic acids, extracellular vesicles and lipids in response to physiological or pathological stimuli. These derived secretomes and extracellular vesicles have similar therapeutic function to MSC-based therapies. Similarly, proregenerative molecules could replace hepatocyte transplantation for acute liver failure. For single gene defects gene therapy will be the future.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Vimalesvaran, S.; Souza, L.N.; Deheragoda, M.; Samyn, M.; Day, J.; Verma, A.; Vilca-Melendez, H.; Rela, M.; Heaton, N.; Dhawan, A. Outcomes of Adults Who Received Liver Transplant as Young Children. EClinicalMedicine 2021, 38, 100987. [Google Scholar] [CrossRef] [PubMed]
  2. Faraj, W.; Dar, F.; Bartlett, A.; Melendez, H.V.; Marangoni, G.; Mukherji, D.; Vergani, G.M.; Dhawan, A.; Heaton, N.; Rela, M. Auxiliary Liver Transplantation for Acute Liver Failure in Children. Ann. Surg. 2010, 251, 351–356. [Google Scholar] [CrossRef] [PubMed]
  3. Puppi, J.; Strom, S.C.; Hughes, R.D.; Bansal, S.; Castell, J.V.; Dagher, I.; Ellis, E.C.; Nowak, G.; Ericzon, B.G.; Fox, I.J.; et al. Improving the Techniques for Human Hepatocyte Transplantation: Report from a Consensus Meeting in London. Cell Transpl. 2012, 21, 1–10. [Google Scholar] [CrossRef] [PubMed]
  4. Iansante, V.; Chandrashekran, A.; Dhawan, A. Cell-Based Liver Therapies: Past, Present and Future. Philos. Trans. R Soc. Lond. B Biol. Sci. 2018, 373, 20170229. [Google Scholar] [CrossRef]
  5. Groth, C.G.; Arborgh, B.; Bjorken, C.; Sundberg, B.; Lundgren, G. Correction of Hyperbilirubinemia in the Glucuronyltransferase-Deficient Rat by Intraportal Hepatocyte Transplantation. Transpl. Proc. 1977, 9, 313–316. [Google Scholar]
  6. Mito, M.; Kusano, M.; Kawaura, Y. Hepatocyte Transplantation in Man. Transpl. Proc. 1992, 24, 3052–3053. [Google Scholar] [CrossRef]
  7. Barahman, M.; Asp, P.; Roy-Chowdhury, N.; Kinkhabwala, M.; Roy-Chowdhury, J.; Kabarriti, R.; Guha, C. Hepatocyte Transplantation: Quo Vadis? Int. J. Radiat. Oncol. Biol. Phys. 2019, 103, 922–934. [Google Scholar] [CrossRef]
  8. 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]
  9. Dhawan, A.; Mitry, R.R.; Hughes, R.D.; Lehec, S.; Terry, C.; Bansal, S.; Arya, R.; Wade, J.J.; Verma, A.; Heaton, N.D.; et al. Hepatocyte Transplantation for Inherited Factor Vii Deficiency. Transplantation 2004, 78, 1812–1814. [Google Scholar] [CrossRef]
  10. 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. Transpl. 2008, 8, 452–457. [Google Scholar] [CrossRef]
  11. Hussaini, S.H.; Oldroyd, B.; Stewart, S.P.; Soo, S.; Roman, F.; Smith, M.A.; Pollard, S.; Lodge, P.; O’grady, J.G.; Losowsky, M.S. Effects of Orthotopic Liver Transplantation on Body Composition. Liver 1998, 18, 173–179. [Google Scholar] [CrossRef]
  12. Fisher, R.A.; Bu, D.; Thompson, M.; Tisnado, J.; Prasad, U.; Sterling, R.; Posner, M.; Strom, S. Defining Hepatocellular Chimerism in a Liver Failure Patient Bridged with Hepatocyte Infusion. Transplantation 2000, 69, 303–307. [Google Scholar] [CrossRef]
  13. Strom, S.C.; Fisher, R.A.; Rubinstein, W.S.; Barranger, J.A.; Towbin, R.B.; Charron, M.; Mieles, L.; Pisarov, L.A.; Dorko, K.; Thompson, M.T.; et al. Transplantation of Human Hepatocytes. Transpl. Proc. 1997, 29, 2103–2106. [Google Scholar] [CrossRef]
  14. Habibullah, C.M.; Syed, I.H.; Qamar, A.; Taher-Uz, Z. Human Fetal Hepatocyte Transplantation in Patients with Fulminant Hepatic Failure. Transplantation 1994, 58, 951–952. [Google Scholar] [CrossRef]
  15. Minnis-Lyons, S.E.; Ferreira-Gonzalez, S.; Aleksieva, N.; Man, T.Y.; Gadd, V.L.; Williams, M.J.; Guest, R.V.; Lu, W.Y.; Dwyer, B.J.; Jamieson, T.; et al. Notch-Igf1 Signaling During Liver Regeneration Drives Biliary Epithelial Cell Expansion and Inhibits Hepatocyte Differentiation. Sci. Signal 2021, 14, eaay9185. [Google Scholar] [CrossRef]
  16. Nagaki, M.; Kano, T.; Muto, Y.; Yamada, T.; Ohnishi, H.; Moriwaki, H. Effects of Intraperitoneal Transplantation of Microcarrier-Attached Hepatocytes on D-Galactosamine-Induced Acute Liver Failure in Rats. Gastroenterol. Jpn. 1990, 25, 78–87. [Google Scholar] [CrossRef]
  17. Hoppo, T.; Komori, J.; Manohar, R.; Stolz, D.B.; Lagasse, E. Rescue of Lethal Hepatic Failure by Hepatized Lymph Nodes in Mice. Gastroenterology 2011, 140, 656–666 e652. [Google Scholar] [CrossRef] [Green Version]
  18. Mitry, R.R.; Jitraruch, S.; Iansante, V.; Dhawan, A. Alginate Encapsulation of Human Hepatocytes and Assessment of Microbeads. Methods Mol. Biol. 2017, 1506, 273–281. [Google Scholar] [CrossRef]
  19. Dhawan, A.; Chaijitraruch, N.; Fitzpatrick, E.; Bansal, S.; Filippi, C.; Lehec, S.C.; Heaton, N.D.; Kane, P.; Verma, A.; Hughes, R.D.; et al. Alginate Microencapsulated Human Hepatocytes for the Treatment of Acute Liver Failure in Children. J. Hepatol. 2020, 72, 877–884. [Google Scholar] [CrossRef]
  20. Jitraruch, S.; Dhawan, A.; Hughes, R.D.; Filippi, C.; Lehec, S.C.; Glover, L.; Mitry, R.R. Cryopreservation of Hepatocyte Microbeads for Clinical Transplantation. Cell Transpl. 2017, 26, 1341–1354. [Google Scholar] [CrossRef] [Green Version]
  21. Darwish, A.A.; Sokal, E.; Stephenne, X.; Najimi, M.; De Goyet Jde, V.; Reding, R. Permanent Access to the Portal System for Cellular Transplantation Using an Implantable Port Device. Liver Transpl. 2004, 10, 1213–1215. [Google Scholar] [CrossRef]
  22. 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 Transpl. 2005, 14, 151–157. [Google Scholar] [CrossRef]
  23. Dhawan, A.; Mitry, R.R.; Hughes, R.D. Hepatocyte Transplantation for Liver-Based Metabolic Disorders. J. Inherit. Metab. Dis. 2006, 29, 431–435. [Google Scholar] [CrossRef]
  24. 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]
  25. Lysy, P.A.; Najimi, M.; Stephenne, X.; Bourgois, A.; Smets, F.; Sokal, E.M. Liver Cell Transplantation for Crigler-Najjar Syndrome Type I: Update and Perspectives. World J. Gastroenterol. 2008, 14, 3464–3470. [Google Scholar] [CrossRef]
  26. Grossman, M.; Rader, D.J.; Muller, D.W.; Kolansky, D.M.; Kozarsky, K.; Clark, B.J., 3rd; Stein, E.A.; Lupien, P.J.; Brewer, H.B., Jr.; Raper, S.E.; et al. A Pilot Study of Ex Vivo Gene Therapy for Homozygous Familial Hypercholesterolaemia. Nat. Med. 1995, 1, 1148–1154. [Google Scholar] [CrossRef]
  27. Muraca, M.; Gerunda, G.; Neri, D.; Vilei, M.T.; Granato, A.; Feltracco, P.; Meroni, M.; Giron, G.; Burlina, A.B. Hepatocyte Transplantation as a Treatment for Glycogen Storage Disease Type 1a. Lancet 2002, 359, 317–318. [Google Scholar] [CrossRef]
  28. Bhattacharya, K.; Orton, R.C.; Qi, X.; Mundy, H.; Morley, D.W.; Champion, M.P.; Eaton, S.; Tester, R.F.; Lee, P.J. A Novel Starch for the Treatment of Glycogen Storage Diseases. J. Inherit. Metab. Dis. 2007, 30, 350–357. [Google Scholar] [CrossRef]
  29. Sokal, E.M.; Smets, F.; Bourgois, A.; Van Maldergem, L.; Buts, J.P.; Reding, R.; Bernard Otte, J.; Evrard, V.; Latinne, D.; Vincent, M.F.; et al. Hepatocyte Transplantation in a 4-Year-Old Girl with Peroxisomal Biogenesis Disease: Technique, Safety, and Metabolic Follow-Up. Transplantation 2003, 76, 735–738. [Google Scholar] [CrossRef] [Green Version]
  30. Horslen, S.P.; Mccowan, T.C.; Goertzen, T.C.; Warkentin, P.I.; Cai, H.B.; Strom, S.C.; Fox, I.J. Isolated Hepatocyte Transplantation in an Infant with a Severe Urea Cycle Disorder. Pediatrics 2003, 111, 1262–1267. [Google Scholar] [CrossRef] [Green Version]
  31. Stephenne, X.; Najimi, M.; Smets, F.; Reding, R.; De Ville De Goyet, J.; Sokal, E.M. Cryopreserved Liver Cell Transplantation Controls Ornithine Transcarbamylase Deficient Patient While Awaiting Liver Transplantation. Am. J. Transpl. 2005, 5, 2058–2061. [Google Scholar] [CrossRef] [PubMed]
  32. 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] [PubMed]
  33. 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]
  34. Soriano, H.E.; Wood, R.P.; Kang, D.C.; Ozaki, C.F.; Finegold, M.J.; Bischoff, F.C.; Reid, B.S.; Ferry, G.D. Hepatocellular Transplantation (Hct) in Children with Fulminant Liver Failure (Flf). Hepatology 1997, 26, 443. [Google Scholar]
  35. Bilir, B.M.; Guinette, D.; Karrer, F.; Kumpe, D.A.; Krysl, J.; Stephens, J.; Mcgavran, L.; Ostrowska, A.; Durham, J. Hepatocyte Transplantation in Acute Liver Failure. Liver Transpl. 2000, 6, 32–40. [Google Scholar] [CrossRef]
  36. Strom, S.C.; Chowdhury, J.R.; Fox, I.J. Hepatocyte Transplantation for the Treatment of Human Disease. Semin. Liver Dis. 1999, 19, 39–48. [Google Scholar] [CrossRef]
  37. Fisher, R.A.; Strom, S.C. Human Hepatocyte Transplantation: Worldwide Results. Transplantation 2006, 82, 441–449. [Google Scholar] [CrossRef]
  38. Strom, S.C.; Fisher, R.A.; Thompson, M.T.; Sanyal, A.J.; Cole, P.E.; Ham, J.M.; Posner, M.P. Hepatocyte Transplantation as a Bridge to Orthotopic Liver Transplantation in Terminal Liver Failure. Transplantation 1997, 63, 559–569. [Google Scholar] [CrossRef]
  39. Schneider, A.; Attaran, M.; Meier, P.N.; Strassburg, C.; Manns, M.P.; Ott, M.; Barthold, M.; Arseniev, L.; Becker, T.; Panning, B. Hepatocyte Transplantation in an Acute Liver Failure Due to Mushroom Poisoning. Transplantation 2006, 82, 1115–1116. [Google Scholar] [CrossRef]
  40. Khan, A.A.; Habeeb, A.; Parveen, N.; Naseem, B.; Babu, R.P.; Capoor, A.K.; Habibullah, C.M. Peritoneal Transplantation of Human Fetal Hepatocytes for the Treatment of Acute Fatty Liver of Pregnancy: A Case Report. Trop. Gastroenterol. 2004, 25, 141–143. [Google Scholar]
  41. Dhawan, A.; Puppi, J.; Hughes, R.D.; Mitry, R.R. Human Hepatocyte Transplantation: Current Experience and Future Challenges. Nat. Rev. Gastroenterol. Hepatol. 2010, 7, 288–298. [Google Scholar] [CrossRef]
  42. Dagher, I.; Boudechiche, L.; Branger, J.; Coulomb-Lhermine, A.; Parouchev, A.; Sentilhes, L.; Lin, T.; Groyer-Picard, M.T.; Vons, C.; Hadchouel, M.; et al. Efficient Hepatocyte Engraftment in a Nonhuman Primate Model after Partial Portal Vein Embolization. Transplantation 2006, 82, 1067–1073. [Google Scholar] [CrossRef]
  43. Jorns, C.; Nowak, G.; Nemeth, A.; Zemack, H.; Mork, L.M.; Johansson, H.; Gramignoli, R.; Watanabe, M.; Karadagi, A.; Alheim, M.; et al. De Novo Donor-Specific Hla Antibody Formation in Two Patients with Crigler-Najjar Syndrome Type I Following Human Hepatocyte Transplantation with Partial Hepatectomy Preconditioning. Am. J. Transplant. 2016, 16, 1021–1030. [Google Scholar] [CrossRef]
  44. 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] [Green Version]
  45. Friedenstein, A.J.; Petrakova, K.V.; Kurolesova, A.I.; Frolova, G.P. Heterotopic of Bone Marrow. Analysis of Precursor Cells for Osteogenic and Hematopoietic Tissues. Transplantation 1968, 6, 230–247. [Google Scholar] [CrossRef]
  46. Gronthos, S.; Zannettino, A.C.; Hay, S.J.; Shi, S.; Graves, S.E.; Kortesidis, A.; Simmons, P.J. Molecular and Cellular Characterisation of Highly Purified Stromal Stem Cells Derived from Human Bone Marrow. J. Cell Sci. 2003, 116, 1827–1835. [Google Scholar] [CrossRef] [Green Version]
  47. De Bari, C.; Dell’accio, F.; Tylzanowski, P.; Luyten, F.P. Multipotent Mesenchymal Stem Cells from Adult Human Synovial Membrane. Arthritis Rheum. 2001, 44, 1928–1942. [Google Scholar] [CrossRef]
  48. Zuk, P.A.; Zhu, M.; Mizuno, H.; Huang, J.; Futrell, J.W.; Katz, A.J.; Benhaim, P.; Lorenz, H.P.; Hedrick, M.H. Multilineage Cells from Human Adipose Tissue: Implications for Cell-Based Therapies. Tissue Eng. 2001, 7, 211–228. [Google Scholar] [CrossRef] [Green Version]
  49. Antonucci, I.; Stuppia, L.; Kaneko, Y.; Yu, S.; Tajiri, N.; Bae, E.C.; Chheda, S.H.; Weinbren, N.L.; Borlongan, C.V. Amniotic Fluid as a Rich Source of Mesenchymal Stromal Cells for Transplantation Therapy. Cell Transpl. 2011, 20, 789–795. [Google Scholar] [CrossRef]
  50. Lee, O.K.; Kuo, T.K.; Chen, W.M.; Lee, K.D.; Hsieh, S.L.; Chen, T.H. Isolation of Multipotent Mesenchymal Stem Cells from Umbilical Cord Blood. Blood 2004, 103, 1669–1675. [Google Scholar] [CrossRef] [Green Version]
  51. Baksh, D.; Yao, R.; Tuan, R.S. Comparison of Proliferative and Multilineage Differentiation Potential of Human Mesenchymal Stem Cells Derived from Umbilical Cord and Bone Marrow. Stem Cells 2007, 25, 1384–1392. [Google Scholar] [CrossRef] [Green Version]
  52. Secunda, R.; Vennila, R.; Mohanashankar, A.M.; Rajasundari, M.; Jeswanth, S.; Surendran, R. Isolation, Expansion and Characterisation of Mesenchymal Stem Cells from Human Bone Marrow, Adipose Tissue, Umbilical Cord Blood and Matrix: A Comparative Study. Cytotechnology 2015, 67, 793–807. [Google Scholar] [CrossRef]
  53. Yoo, K.H.; Jang, I.K.; Lee, M.W.; Kim, H.E.; Yang, M.S.; Eom, Y.; Lee, J.E.; Kim, Y.J.; Yang, S.K.; Jung, H.L.; et al. Comparison of Immunomodulatory Properties of Mesenchymal Stem Cells Derived from Adult Human Tissues. Cell Immunol. 2009, 259, 150–156. [Google Scholar] [CrossRef]
  54. Bunnell, B.A.; Betancourt, A.M.; Sullivan, D.E. New Concepts on the Immune Modulation Mediated by Mesenchymal Stem Cells. Stem Cell Res. Ther. 2010, 1, 34. [Google Scholar] [CrossRef] [Green Version]
  55. Klyushnenkova, E.; Mosca, J.D.; Zernetkina, V.; Majumdar, M.K.; Beggs, K.J.; Simonetti, D.W.; Deans, R.J.; Mcintosh, K.R. T Cell Responses to Allogeneic Human Mesenchymal Stem Cells: Immunogenicity, Tolerance, and Suppression. J. Biomed. Sci. 2005, 12, 47–57. [Google Scholar] [CrossRef]
  56. Aggarwal, S.; Pittenger, M.F. Human Mesenchymal Stem Cells Modulate Allogeneic Immune Cell Responses. Blood 2005, 105, 1815–1822. [Google Scholar] [CrossRef] [Green Version]
  57. Meisel, R.; Zibert, A.; Laryea, M.; Gobel, U.; Daubener, W.; Dilloo, D. Human Bone Marrow Stromal Cells Inhibit Allogeneic T-Cell Responses by Indoleamine 2,3-Dioxygenase-Mediated Tryptophan Degradation. Blood 2004, 103, 4619–4621. [Google Scholar] [CrossRef] [Green Version]
  58. Prasanna, S.J.; Gopalakrishnan, D.; Shankar, S.R.; Vasandan, A.B. Pro-Inflammatory Cytokines, Ifngamma and Tnfalpha, Influence Immune Properties of Human Bone Marrow and Wharton Jelly Mesenchymal Stem Cells Differentially. PLoS ONE 2010, 5, e9016. [Google Scholar] [CrossRef]
  59. Sato, K.; Ozaki, K.; Oh, I.; Meguro, A.; Hatanaka, K.; Nagai, T.; Muroi, K.; Ozawa, K. Nitric Oxide Plays a Critical Role in Suppression of T-Cell Proliferation by Mesenchymal Stem Cells. Blood 2007, 109, 228–234. [Google Scholar] [CrossRef]
  60. Ding, Y.; Xu, D.; Feng, G.; Bushell, A.; Muschel, R.J.; Wood, K.J. Mesenchymal Stem Cells Prevent the Rejection of Fully Allogenic Islet Grafts by the Immunosuppressive Activity of Matrix Metalloproteinase-2 and -9. Diabetes 2009, 58, 1797–1806. [Google Scholar] [CrossRef] [Green Version]
  61. Glennie, S.; Soeiro, I.; Dyson, P.J.; Lam, E.W.; Dazzi, F. Bone Marrow Mesenchymal Stem Cells Induce Division Arrest Anergy of Activated T Cells. Blood 2005, 105, 2821–2827. [Google Scholar] [CrossRef] [PubMed]
  62. Rasmusson, I.; Le Blanc, K.; Sundberg, B.; Ringden, O. Mesenchymal Stem Cells Stimulate Antibody Secretion in Human B Cells. Scand. J. Immunol. 2007, 65, 336–343. [Google Scholar] [CrossRef] [PubMed]
  63. Nitzsche, F.; Muller, C.; Lukomska, B.; Jolkkonen, J.; Deten, A.; Boltze, J. Concise Review: Msc Adhesion Cascade-Insights into Homing and Transendothelial Migration. Stem Cells 2017, 35, 1446–1460. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Sotiropoulou, P.A.; Perez, S.A.; Gritzapis, A.D.; Baxevanis, C.N.; Papamichail, M. Interactions between Human Mesenchymal Stem Cells and Natural Killer Cells. Stem Cells 2006, 24, 74–85. [Google Scholar] [CrossRef]
  65. Lawrence, T.; Natoli, G. Transcriptional Regulation of Macrophage Polarization: Enabling Diversity with Identity. Nat. Rev. Immunol. 2011, 11, 750–761. [Google Scholar] [CrossRef]
  66. Lee, K.C.; Lin, H.C.; Huang, Y.H.; Hung, S.C. Allo-Transplantation of Mesenchymal Stem Cells Attenuates Hepatic Injury through Il1ra Dependent Macrophage Switch in a Mouse Model of Liver Disease. J. Hepatol. 2015, 63, 1405–1412. [Google Scholar] [CrossRef]
  67. Melief, S.M.; Schrama, E.; Brugman, M.H.; Tiemessen, M.M.; Hoogduijn, M.J.; Fibbe, W.E.; Roelofs, H. Multipotent Stromal Cells Induce Human Regulatory T Cells through a Novel Pathway Involving Skewing of Monocytes toward Anti-Inflammatory Macrophages. Stem Cells 2013, 31, 1980–1991. [Google Scholar] [CrossRef]
  68. Beyth, S.; Borovsky, Z.; Mevorach, D.; Liebergall, M.; Gazit, Z.; Aslan, H.; Galun, E.; Rachmilewitz, J. Human Mesenchymal Stem Cells Alter Antigen-Presenting Cell Maturation and Induce T-Cell Unresponsiveness. Blood 2005, 105, 2214–2219. [Google Scholar] [CrossRef] [Green Version]
  69. Desmouliere, A.; Geinoz, A.; Gabbiani, F.; Gabbiani, G. Transforming Growth Factor-Beta 1 Induces Alpha-Smooth Muscle Actin Expression in Granulation Tissue Myofibroblasts and in Quiescent and Growing Cultured Fibroblasts. J. Cell Biol. 1993, 122, 103–111. [Google Scholar] [CrossRef] [Green Version]
  70. Kim, G.; Kim, M.Y.; Baik, S.K. Transient Elastography Versus Hepatic Venous Pressure Gradient for Diagnosing Portal Hypertension: A Systematic Review and Meta-Analysis. Clin. Mol. Hepatol. 2017, 23, 34–41. [Google Scholar] [CrossRef] [Green Version]
  71. Parekkadan, B.; Van Poll, D.; Megeed, Z.; Kobayashi, N.; Tilles, A.W.; Berthiaume, F.; Yarmush, M.L. Immunomodulation of Activated Hepatic Stellate Cells by Mesenchymal Stem Cells. Biochem. Biophys. Res. Commun. 2007, 363, 247–252. [Google Scholar] [CrossRef] [Green Version]
  72. Wynn, T.A. Fibrotic Disease and the T(H)1/T(H)2 Paradigm. Nat. Rev. Immunol. 2004, 4, 583–594. [Google Scholar] [CrossRef] [Green Version]
  73. Eom, Y.W.; Yoon, Y.; Baik, S.K. Mesenchymal Stem Cell Therapy for Liver Disease: Current Status and Future Perspectives. Curr. Opin. Gastroenterol. 2021, 37, 216–223. [Google Scholar] [CrossRef]
  74. Sharma, R.R.; Pollock, K.; Hubel, A.; Mckenna, D. Mesenchymal Stem or Stromal Cells: A Review of Clinical Applications and Manufacturing Practices. Transfusion 2014, 54, 1418–1437. [Google Scholar] [CrossRef]
  75. Ali, G.; Mohsin, S.; Khan, M.; Nasir, G.A.; Shams, S.; Khan, S.N.; Riazuddin, S. Nitric Oxide Augments Mesenchymal Stem Cell Ability to Repair Liver Fibrosis. J. Transl. Med. 2012, 10, 75. [Google Scholar] [CrossRef] [Green Version]
  76. Lange, C.; Bassler, P.; Lioznov, M.V.; Bruns, H.; Kluth, D.; Zander, A.R.; Fiegel, H.C. Liver-Specific Gene Expression in Mesenchymal Stem Cells Is Induced by Liver Cells. World J. Gastroenterol. 2005, 11, 4497–4504. [Google Scholar] [CrossRef]
  77. Banas, A.; Teratani, T.; Yamamoto, Y.; Tokuhara, M.; Takeshita, F.; Quinn, G.; Okochi, H.; Ochiya, T. Adipose Tissue-Derived Mesenchymal Stem Cells as a Source of Human Hepatocytes. Hepatology 2007, 46, 219–228. [Google Scholar] [CrossRef]
  78. Shu, S.N.; Wei, L.; Wang, J.H.; Zhan, Y.T.; Chen, H.S.; Wang, Y. Hepatic Differentiation Capability of Rat Bone Marrow-Derived Mesenchymal Stem Cells and Hematopoietic Stem Cells. World J. Gastroenterol. 2004, 10, 2818–2822. [Google Scholar] [CrossRef]
  79. Herrera, M.B.; Bruno, S.; Buttiglieri, S.; Tetta, C.; Gatti, S.; Deregibus, M.C.; Bussolati, B.; Camussi, G. Isolation and Characterization of a Stem Cell Population from Adult Human Liver. Stem Cells 2006, 24, 2840–2850. [Google Scholar] [CrossRef]
  80. Safwan, M.; Ramachandran, P.; Vij, M.; Shanmugam, N.; Rela, M. Impact of Ductal Plate Malformation on Survival with Native Liver in Children with Biliary Atresia. Pediatr. Surg. Int. 2015, 31, 837–843. [Google Scholar] [CrossRef]
  81. Livesey, E.; Cortina Borja, M.; Sharif, K.; Alizai, N.; Mcclean, P.; Kelly, D.; Hadzic, N.; Davenport, M. Epidemiology of Biliary Atresia in England and Wales (1999–2006). Arch. Dis. Child Fetal Neonatal Ed. 2009, 94, F451–F455. [Google Scholar] [CrossRef] [PubMed]
  82. Burns, J.; Davenport, M. Adjuvant Treatments for Biliary Atresia. Transl. Pediatr. 2020, 9, 253–265. [Google Scholar] [CrossRef] [PubMed]
  83. Bezerra, J.A. Potential Etiologies of Biliary Atresia. Pediatr. Transpl. 2005, 9, 646–651. [Google Scholar] [CrossRef] [PubMed]
  84. Vij, M.; Rela, M. Biliary Atresia: Pathology, Etiology and Pathogenesis. Future Sci. OA 2020, 6, FSO466. [Google Scholar] [CrossRef] [Green Version]
  85. Hartley, J.; Harnden, A.; Kelly, D. Biliary Atresia. BMJ 2010, 340, c2383. [Google Scholar] [CrossRef]
  86. Davenport, M.; Ong, E.; Sharif, K.; Alizai, N.; Mcclean, P.; Hadzic, N.; Kelly, D.A. Biliary Atresia in England and Wales: Results of Centralization and New Benchmark. J. Pediatr. Surg. 2011, 46, 1689–1694. [Google Scholar] [CrossRef]
  87. Davenport, M.; Stringer, M.D.; Tizzard, S.A.; Mcclean, P.; Mieli-Vergani, G.; Hadzic, N. Randomized, Double-Blind, Placebo-Controlled Trial of Corticosteroids after Kasai Portoenterostomy for Biliary Atresia. Hepatology 2007, 46, 1821–1827. [Google Scholar] [CrossRef]
  88. Mack, C.L.; Spino, C.; Alonso, E.M.; Bezerra, J.A.; Moore, J.; Goodhue, C.; Ng, V.L.; Karpen, S.J.; Venkat, V.; Loomes, K.M.; et al. A Phase I/Iia Trial of Intravenous Immunoglobulin Following Portoenterostomy in Biliary Atresia. J. Pediatr. Gastroenterol. Nutr. 2019, 68, 495–501. [Google Scholar] [CrossRef]
  89. Rasmusson, I. Immune Modulation by Mesenchymal Stem Cells. Exp. Cell Res. 2006, 312, 2169–2179. [Google Scholar] [CrossRef] [Green Version]
  90. Lei, J.; Chai, Y.; Xiao, J.; Hu, H.; Liu, Z.; Xiao, Y.; Yi, L.; Huang, J.; Xiang, T.; Zhang, S. Antifibrotic Potential of Bone Marrowderived Mesenchymal Stem Cells in Biliary Atresia Mice. Mol. Med. Rep. 2018, 18, 3983–3988. [Google Scholar] [CrossRef] [Green Version]
  91. Christ, B.; Bruckner, S.; Winkler, S. The Therapeutic Promise of Mesenchymal Stem Cells for Liver Restoration. Trends Mol. Med. 2015, 21, 673–686. [Google Scholar] [CrossRef]
  92. Liu, Z.; Meng, F.; Li, C.; Zhou, X.; Zeng, X.; He, Y.; Mrsny, R.J.; Liu, M.; Hu, X.; Hu, J.F.; et al. Human Umbilical Cord Mesenchymal Stromal Cells Rescue Mice from Acetaminophen-Induced Acute Liver Failure. Cytotherapy 2014, 16, 1207–1219. [Google Scholar] [CrossRef]
  93. Chen, L.; Zhang, J.; Yang, L.; Zhang, G.; Wang, Y.; Zhang, S. The Effects of Conditioned Medium Derived from Mesenchymal Stem Cells Cocultured with Hepatocytes on Damaged Hepatocytes and Acute Liver Failure in Rats. Stem Cells Int. 2018, 2018, 9156560. [Google Scholar] [CrossRef]
  94. Guo, G.; Zhuang, X.; Xu, Q.; Wu, Z.; Zhu, Y.; Zhou, Y.; Li, Y.; Lu, Y.; Zhang, B.; Talbot, P.; et al. Peripheral Infusion of Human Umbilical Cord Mesenchymal Stem Cells Rescues Acute Liver Failure Lethality in Monkeys. Stem Cell Res. Ther. 2019, 10, 84. [Google Scholar] [CrossRef] [Green Version]
  95. Jang, Y.O.; Kim, Y.J.; Baik, S.K.; Kim, M.Y.; Eom, Y.W.; Cho, M.Y.; Park, H.J.; Park, S.Y.; Kim, B.R.; Kim, J.W.; et al. Histological Improvement Following Administration of Autologous Bone Marrow-Derived Mesenchymal Stem Cells for Alcoholic Cirrhosis: A Pilot Study. Liver Int. 2014, 34, 33–41. [Google Scholar] [CrossRef]
  96. Suk, K.T.; Yoon, J.H.; Kim, M.Y.; Kim, C.W.; Kim, J.K.; Park, H.; Hwang, S.G.; Kim, D.J.; Lee, B.S.; Lee, S.H.; et al. Transplantation with Autologous Bone Marrow-Derived Mesenchymal Stem Cells for Alcoholic Cirrhosis: Phase 2 Trial. Hepatology 2016, 64, 2185–2197. [Google Scholar] [CrossRef]
  97. Zhang, Z.; Lin, H.; Shi, M.; Xu, R.; Fu, J.; Lv, J.; Chen, L.; Lv, S.; Li, Y.; Yu, S.; et al. Human Umbilical Cord Mesenchymal Stem Cells Improve Liver Function and Ascites in Decompensated Liver Cirrhosis Patients. J. Gastroenterol. Hepatol. 2012, 27 (Suppl. S2), 112–120. [Google Scholar] [CrossRef]
  98. Mohamadnejad, M.; Alimoghaddam, K.; Bagheri, M.; Ashrafi, M.; Abdollahzadeh, L.; Akhlaghpoor, S.; Bashtar, M.; Ghavamzadeh, A.; Malekzadeh, R. Randomized Placebo-Controlled Trial of Mesenchymal Stem Cell Transplantation in Decompensated Cirrhosis. Liver Int. 2013, 33, 1490–1496. [Google Scholar] [CrossRef]
  99. Kantarcioglu, M.; Demirci, H.; Avcu, F.; Karslioglu, Y.; Babayigit, M.A.; Karaman, B.; Ozturk, K.; Gurel, H.; Akdogan Kayhan, M.; Kacar, S.; et al. Efficacy of Autologous Mesenchymal Stem Cell Transplantation in Patients with Liver Cirrhosis. Turk. J. Gastroenterol. 2015, 26, 244–250. [Google Scholar] [CrossRef] [Green Version]
  100. Mohamadnejad, M.; Alimoghaddam, K.; Mohyeddin-Bonab, M.; Bagheri, M.; Bashtar, M.; Ghanaati, H.; Baharvand, H.; Ghavamzadeh, A.; Malekzadeh, R. Phase 1 Trial of Autologous Bone Marrow Mesenchymal Stem Cell Transplantation in Patients with Decompensated Liver Cirrhosis. Arch. Iran Med. 2007, 10, 459–466. [Google Scholar]
  101. Kharaziha, P.; Hellstrom, P.M.; Noorinayer, B.; Farzaneh, F.; Aghajani, K.; Jafari, F.; Telkabadi, M.; Atashi, A.; Honardoost, M.; Zali, M.R.; et al. Improvement of Liver Function in Liver Cirrhosis Patients after Autologous Mesenchymal Stem Cell Injection: A Phase I-Ii Clinical Trial. Eur. J. Gastroenterol. Hepatol. 2009, 21, 1199–1205. [Google Scholar] [CrossRef]
  102. El-Ansary, M.; Abdel-Aziz, I.; Mogawer, S.; Abdel-Hamid, S.; Hammam, O.; Teaema, S.; Wahdan, M. Phase Ii Trial: Undifferentiated Versus Differentiated Autologous Mesenchymal Stem Cells Transplantation in Egyptian Patients with Hcv Induced Liver Cirrhosis. Stem Cell Rev. Rep. 2012, 8, 972–981. [Google Scholar] [CrossRef]
  103. Amin, M.A.; Sabry, D.; Rashed, L.A.; Aref, W.M.; El-Ghobary, M.A.; Farhan, M.S.; Fouad, H.A.; Youssef, Y.A. Short-Term Evaluation of Autologous Transplantation of Bone Marrow-Derived Mesenchymal Stem Cells in Patients with Cirrhosis: Egyptian Study. Clin. Transpl. 2013, 27, 607–612. [Google Scholar] [CrossRef]
  104. 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]
  105. Salama, H.; Zekri, A.R.; Medhat, E.; Al Alim, S.A.; Ahmed, O.S.; Bahnassy, A.A.; Lotfy, M.M.; Ahmed, R.; Musa, S. Peripheral Vein Infusion of Autologous Mesenchymal Stem Cells in Egyptian Hcv-Positive Patients with End-Stage Liver Disease. Stem Cell Res. Ther. 2014, 5, 70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Xu, L.; Gong, Y.; Wang, B.; Shi, K.; Hou, Y.; Wang, L.; Lin, Z.; Han, Y.; Lu, L.; Chen, D.; et al. Randomized Trial of Autologous Bone Marrow Mesenchymal Stem Cells Transplantation for Hepatitis B Virus Cirrhosis: Regulation of Treg/Th17 Cells. J. Gastroenterol. Hepatol. 2014, 29, 1620–1628. [Google Scholar] [CrossRef]
  107. Sakai, Y.; Fukunishi, S.; Takamura, M.; Kawaguchi, K.; Inoue, O.; Usui, S.; Takashima, S.; Seki, A.; Asai, A.; Tsuchimoto, Y.; et al. Clinical Trial of Autologous Adipose Tissue-Derived Regenerative (Stem) Cells Therapy for Exploration of Its Safety and Efficacy. Regen. Ther. 2021, 18, 97–101. [Google Scholar] [CrossRef]
  108. Liang, J.; Zhang, H.; Zhao, C.; Wang, D.; Ma, X.; Zhao, S.; Wang, S.; Niu, L.; Sun, L. Effects of Allogeneic Mesenchymal Stem Cell Transplantation in the Treatment of Liver Cirrhosis Caused by Autoimmune Diseases. Int. J. Rheum. Dis. 2017, 20, 1219–1226. [Google Scholar] [CrossRef]
  109. Fang, X.; Liu, L.; Dong, J.; Zhang, J.; Song, H.; Song, Y.; Huang, Y.; Cui, X.; Lin, J.; Chen, C.; et al. A Study About Immunomodulatory Effect and Efficacy and Prognosis of Human Umbilical Cord Mesenchymal Stem Cells in Patients with Chronic Hepatitis B-Induced Decompensated Liver Cirrhosis. J. Gastroenterol. Hepatol. 2018, 33, 774–780. [Google Scholar] [CrossRef]
  110. Peng, L.; Xie, D.Y.; Lin, B.L.; Liu, J.; Zhu, H.P.; Xie, C.; Zheng, Y.B.; Gao, Z.L. Autologous Bone Marrow Mesenchymal Stem Cell Transplantation in Liver Failure Patients Caused by Hepatitis B: Short-Term and Long-Term Outcomes. Hepatology 2011, 54, 820–828. [Google Scholar] [CrossRef]
  111. Amer, M.E.; El-Sayed, S.Z.; El-Kheir, W.A.; Gabr, H.; Gomaa, A.A.; El-Noomani, N.; Hegazy, M. Clinical and Laboratory Evaluation of Patients with End-Stage Liver Cell Failure Injected with Bone Marrow-Derived Hepatocyte-Like Cells. Eur. J. Gastroenterol. Hepatol. 2011, 23, 936–941. [Google Scholar] [CrossRef] [PubMed]
  112. 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]
  113. Lin, B.L.; Chen, J.F.; Qiu, W.H.; Wang, K.W.; Xie, D.Y.; Chen, X.Y.; Liu, Q.L.; Peng, L.; Li, J.G.; Mei, Y.Y.; et al. Allogeneic Bone Marrow-Derived Mesenchymal Stromal Cells for Hepatitis B Virus-Related Acute-on-Chronic Liver Failure: A Randomized Controlled Trial. Hepatology 2017, 66, 209–219. [Google Scholar] [CrossRef] [Green Version]
  114. Shi, M.; Liu, Z.; Wang, Y.; Xu, R.; Sun, Y.; Zhang, M.; Yu, X.; Wang, H.; Meng, L.; Su, H.; et al. A Pilot Study of Mesenchymal Stem Cell Therapy for Acute Liver Allograft Rejection. Stem Cells Transl. Med. 2017, 6, 2053–2061. [Google Scholar] [CrossRef] [Green Version]
  115. Detry, O.; Vandermeulen, M.; Delbouille, M.H.; Somja, J.; Bletard, N.; Briquet, A.; Lechanteur, C.; Giet, O.; Baudoux, E.; Hannon, M.; et al. Infusion of Mesenchymal Stromal Cells after Deceased Liver Transplantation: A Phase I-Ii, Open-Label, Clinical Study. J. Hepatol. 2017, 67, 47–55. [Google Scholar] [CrossRef] [Green Version]
  116. Zhang, Y.C.; Liu, W.; Fu, B.S.; Wang, G.Y.; Li, H.B.; Yi, H.M.; Jiang, N.; Wang, G.; Zhang, J.; Yi, S.H.; et al. Therapeutic Potentials of Umbilical Cord-Derived Mesenchymal Stromal Cells for Ischemic-Type Biliary Lesions Following Liver Transplantation. Cytotherapy 2017, 19, 194–199. [Google Scholar] [CrossRef]
  117. Ullah, M.; Liu, D.D.; Thakor, A.S. Mesenchymal Stromal Cell Homing: Mechanisms and Strategies for Improvement. iScience 2019, 15, 421–438. [Google Scholar] [CrossRef] [Green Version]
  118. Yuan, M.; Hu, X.; Yao, L.; Jiang, Y.; Li, L. Mesenchymal Stem Cell Homing to Improve Therapeutic Efficacy in Liver Disease. Stem Cell Res. Ther. 2022, 13, 179. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Vimalesvaran, S.; Nulty, J.; Dhawan, A. Cellular Therapies in Pediatric Liver Diseases. Cells 2022, 11, 2483. https://doi.org/10.3390/cells11162483

AMA Style

Vimalesvaran S, Nulty J, Dhawan A. Cellular Therapies in Pediatric Liver Diseases. Cells. 2022; 11(16):2483. https://doi.org/10.3390/cells11162483

Chicago/Turabian Style

Vimalesvaran, Sunitha, Jessica Nulty, and Anil Dhawan. 2022. "Cellular Therapies in Pediatric Liver Diseases" Cells 11, no. 16: 2483. https://doi.org/10.3390/cells11162483

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