Next Article in Journal
Longitudinal Variations in Antibody Responses against SARS-CoV-2 Spike Epitopes upon Serial Vaccinations
Next Article in Special Issue
Immunomodulatory Mechanism and Potential Application of Dental Pulp-Derived Stem Cells in Immune-Mediated Diseases
Previous Article in Journal
Nordalbergin Exerts Anti-Neuroinflammatory Effects by Attenuating MAPK Signaling Pathway, NLRP3 Inflammasome Activation and ROS Production in LPS-Stimulated BV2 Microglia
Previous Article in Special Issue
The Efficiency of Neurospheres Derived from Human Wharton’s Jelly Mesenchymal Stem Cells for Spinal Cord Injury Regeneration in Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

PGE2 Produced by Exogenous MSCs Promotes Immunoregulation in ARDS Induced by Highly Pathogenic Influenza A through Activation of the Wnt-β-Catenin Signaling Pathway

by
Resti Yudhawati
1,2,* and
Kazufumi Shimizu
2,3
1
Department of Pulmonology and Respiratory Medicine, Faculty of Medicine, Universitas Airlangga—Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
2
Indonesia-Japan Collaborative Research Center for Emerging and Re-Emerging Infectious Diseases, Institute of Tropical Disease, Airlangga University, Surabaya 60286, Indonesia
3
Center for Infectious Diseases, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(8), 7299; https://doi.org/10.3390/ijms24087299
Submission received: 6 February 2023 / Revised: 12 April 2023 / Accepted: 13 April 2023 / Published: 14 April 2023

Abstract

:
Acute respiratory distress syndrome is an acute respiratory failure caused by cytokine storms; highly pathogenic influenza A virus infection can induce cytokine storms. The innate immune response is vital in this cytokine storm, acting by activating the transcription factor NF-κB. Tissue injury releases a danger-associated molecular pattern that provides positive feedback for NF-κB activation. Exogenous mesenchymal stem cells can also modulate immune responses by producing potent immunosuppressive substances, such as prostaglandin E2. Prostaglandin E2 is a critical mediator that regulates various physiological and pathological processes through autocrine or paracrine mechanisms. Activation of prostaglandin E2 results in the accumulation of unphosphorylated β-catenin in the cytoplasm, which subsequently reaches the nucleus to inhibit the transcription factor NF-κB. The inhibition of NF-κB by β-catenin is a mechanism that reduces inflammation.

1. Introduction

Acute respiratory distress syndrome (ARDS) is an acute respiratory failure that arises due to acute lung injury which causes a complication [1,2]. ARDS is characterized by diffuse alveolar damage, which leads to many complications, ultimately severe hypoxemia and respiratory failure [1,3]. The etiology of ARDS is quite complex; one factor is bacterial- and viral-induced pneumonia [4,5]. Influenza A virus is one of the reasons for the high incidence of ARDS and fulminant ARDS (FARDS), which includes the highly pathogenic avian influenza (HPAI) H5N1 [6,7]. On 23 February 2023, the Cambodia International Health Regulations (IHR) National Focal Point (NFP) notified WHO of a confirmed case of death of a girl with severe pneumonia induced by avian influenza A (H5N1) virus [8]. In addition, SARS-CoV-2 is another infectious virus reported to induce ARDS [9,10,11].
The mortality of ARDS remains high, although there has been an overall improvement in diagnosis and supportive care. However, no pharmacological therapy is available [12]. According to a meta-analysis study, antiviral medicine is effective for the earliest infection but has little beneficial effect on severe influenza [13]. This medicine cannot repair the inflammation related to associated tissue injury induced by the influenza virus [14]. Developing treatments prioritizing immunomodulation and inhibiting the intracellular signaling cascade, as well as limiting viral replication, is essential because any patient infected with the highly pathogenic influenza virus already has severe complications at admission. Cell-based therapy is a potential therapeutical approach. Allotransplantation therapy utilizing mesenchymal stem cells (MSCs) has shown beneficial effects on ARDS [15]. Furthermore, it has negligible immunogenicity features [16]. Other studies on MSCs have demonstrated their therapeutic potency on multiple organ injuries, including myocardial infarction [17], diabetes [18], sepsis [19], liver diseases [20], and acute kidney failure [21]. In an experimental model of acute lung injury, either due to chemical, viral, or bacterial infection, MSCs were found to remain beneficial [22,23,24]. In addition, studies on MSCs administration to acute lung-injury-virus-induced ARDS are still limited. Studies on MSCs’ effects on lung injury induced by the H1N1 virus have shown conflicting results [25,26]. In contrast, studies of highly pathogenic influenza A virus subtypes H9N2 and H5N1 have indicated that MSCs are effective on lung-injury-induced ARDS [27,28]. However, the mechanism that leads to MSCs mobilization to the damaged tissue site as an immunomodulator remains unclear.
Most studies at present focus on investigations of the regeneration effect of MSCs and its application in treatments that aim to improve tissue injury and regeneration. Knowledge about the crosstalk between regulatory factors in MSCs niches is essential, as it functions in the homeostasis of normal and injured tissue (such as that affected by infection). This pathway is extensively involved in the regulation of adult MSCs’ fate in various tissues. Through the canonical pathway, this Wnt/β-catenin pathway helps the maintenance of MSCs and improves their proliferation ability as a function of regeneration [29,30,31]. However, the role of Wnt signaling in the immunoregulatory function has not drawn much attention as yet. In this review, we seek to address the mechanisms of MSCs as immunomodulatory agents through the canonical Wnt/β catenin signaling pathway.

2. Host Immune Response and Cellular Signaling Pathway Activation Induced by Influenza Virus

The influenza virus is a member of the Orthomyxoviridae family, an enveloped corona virus with a diameter of approximately 80–120 nm. The genome is single-stranded ribonucleic acid (RNA) with a negative sense, consisting of eight (type A and B) or seven (type C) segments. Hundreds of spikes protrude from the surface of the type A virus’s envelope, e.g., hemagglutinin (HA) and neuraminidase (NA). HA is an essential glycoprotein antigen, acting as the site for viral binding and fusion between viral and cell membranes. NA possesses an enzymatic activity, acting to destroy the receptor molecules preventing virus aggregation and releasing the newly assembled virion of the infected cell [32].
The characteristic innate immune response is the primary determinant of the outcome of influenza infection [33]. Three main classes of pattern-recognition receptors (PRRs) recognize the pathogen-associated molecular patterns (PAMPs) of the influenza A virus, e.g., cytosolic retinoic acid inducible gene 1 (RIG-1) sensor [34], toll-like receptor (TLR) 7 in the endosome [35], and nucleotide-binding domain-leucine-rich repeat-containing (NLR) molecules [36]. These three pathways can induce pro-inflammatory cytokines and chemokines, which leads to pathological lung damage [33,36]. The host signaling pathway is directed towards the influenza virus after the virus binds with the host through HA and NA proteins. The HA protein can directly induce the expression of several host cytokines, including TNFα and IL-1 [37]. Host receptors can recognize ribonucleocapsid protein (RNP) and initiate several signaling transductions. The resulting transduction signaling pathways are complicated and overlap through the PKR, ASK1, TLR, RIG, phosphoinositide 3-kinase (PI3K), and PKC pathways. PKR plays roles in the apoptosis processes of cells infected by the influenza A virus through Fas and ligand induction. PKR activation can also activate IKK, which can lead to the phosphorylation and degradation of Iκβ (inhibitory kappa beta), and then cause the active transcription factor, nuclear factor kappa B (NF-κB) subunit p65/p50, to release. NF-κB subunit p65/p50 can induce pro-inflammatory cytokines, chemokines, and proapoptotic molecules, such as INFβ [38].
The single-stranded RNA (ssRNA) of the influenza A virus is recognized by TLR 7 and 8 via the TLR pathway; it then induces several adaptor molecules and transcription factors, including MyD88, TAK1, TAB1, TAB2, and TRAF6, and activates NF-κB. The interaction between viral protein and TLR activates IKK, which can lead to the phosphorylation and degradation of Iκβ, then release the NF-κB transcription factor subunit p65/p50. Through TKR7/8, TANK binding kinase (TBK) 1 and IKK may also be activated, which then induces the activation of the NF-κB transcription factor and interferon regulatory factor (IRF) 3/7, which expresses the interferon β gene and apoptosis regulation. Influenza A can also be recognized by RIG 1, MDA 5 (melanoma differentiation-associated gene 5), and LGP2. RIG1 and MDA5 act as cytoplasmic viral RNA sensors that play an essential role in innate antiviral immunity [37,38].
Viral proteins synthesized in the infected cells partially degrade into smaller peptides in the endosomes/lysosomes, and they form a complex with the MHC class I or II peptides at the cell surface. CD4+ helper T cells recognize the complex with MHC class II [32,33], whereas CD8+ cytotoxic T cells recognize the complex with MHC class I. Such antigen presentations and recognition by T cells activate the adaptive immune response.

3. Cytokine Storm and ALI/ARDS Induced by Influenza Virus Infection

Pro-inflammatory cytokines and chemokines induced by influenza virus infection cause pathological lung damage [33,36]. Highly pathogenic influenza A virus infection causes the release of excessive cytokines (cytokine storm). The release of this large amount of cytokines initially suppresses viral replication. However, this cytokine storm causes local cell death and severe damage to lung tissue, and eventually results in fibrosis. In the early stages, viral pneumonia occurs in interstitial pneumonitis. This process continues with exudation, intra-alveolar edema, inflammatory cells, and erythrocytes mobilization from the surrounding capillaries, as well as hyalin membrane formation and fibroblast accumulation. The inflammatory cells then produce many inflammatory mediator cells, which are clinically referred to as ARDS. As a result, the disruption of oxygen diffusion occurs, so hypoxia/anoxia can damage other organs (anoxic multiorgan dysfunction) [39].
The mechanisms of cytokine storm damage proceed through multiple stages. When the highly pathogenic influenza A virus infects alveolar epithelial cells, these cells release various pro-inflammatory cytokines and chemoattractants. HPAI and their infected cells can be recognized by pattern recognition receptors (PRRs) on innate immune cells, such as airway epithelial cells, inflammatory DCs, macrophages, monocytes, and neutrophils. There are several types of cellular PRRs, as follows: membrane-associated toll-like receptors (TLRs), C-type lectin receptors, cytoplasmic nucleotide-binding oligomerization domain-like receptors, and retinoic acid induced gene-like receptors [40]. The viral RNA in the endosome and cytoplasm is predominantly recognized by membrane-associated TLRs and RIG-I-like receptors, respectively. PRRs recognize pathogen-associated molecular patterns (PAMPs) on different types of microorganisms [41,42]. The engagement of PRRs by PAMPs on HPAI activates PPR-related signaling and induces the expression of various pro-inflammatory cytokines and chemokines to recruit innate immune cells (such as macrophages, neutrophils, and DCs) into the infected airway [43]. Macrophages present viral peptides on the cell surface and activate lymphocyte T cells. Activated macrophages as well as lymphocyte T cells then release various pro-inflammatory cytokines and chemoattractants. All chemokines and cytokines can recruit more macrophages and neutrophils to the lung and, thus, cause more extensive damage [44].
The TLRs and RIG-I-like receptor on the cellular membrane can interact with their specific extra-cellular and endosomal PAMPs [43]. TLR7 signaling, via the adaptor protein MyD88, activates IRF7 and NF-κB, while RIG-I signaling via the adaptor molecule IPD-I located in the mitochondria can activate IRF3 and NF-κB [36,45]. The activation of IRF-7 and IRF-3 may cause IRF-7 and IRF-3 translocation into the nucleus and produce interferon type I, while NF-κB acts as a transcription factor to induce pro-inflammatory cytokine production, including IL-6, TNFα, and pro-IL-1β [36,45].
In addition to PAMP, DAMP (danger-associated molecular pattern) is a host molecule that functions to maintain PRRs’ activation. High-mobility group box 1 (HMGB1) is a DAMP released from damaged cells due to ARDS [46,47]. HMGB1 has high affinity in the RAGE ligand, and its regulation is increased in mice with pneumonia induced by influenza virus infection [48]. The binding of HMGB1 with RAGE produces a signal that activates NF-κB then induces adhesion molecules and pro-inflammatory cytokines [48,49]. Figure 1 illustrates the host immune response and cellular signaling pathway activation induced by the influenza virus, as well as the mechanisms of the cytokine storm.

4. Mesenchymal Stem Cells

MSCs are non-hematopoietic stem cells with the ability to differentiate into mesenchymal and non-mesenchymal lineages, and they originate from mesodermal tissues. These cells can be found in all human organs, specifically in the regions of cell populations found in the perivascular area, mainly in bone marrow, the umbilical cord (blood or Whartons jelly), and fatty tissues. MSCs in the lung can differentiate into the epithelial phenotype and structure lung cells [50]. Currently, autologous and allogeneic MSCs are being investigated in clinical research for the treatment of various diseases, including diabetes mellitus, multiple sclerosis, Crohn’s disease, and end-stage liver disease, as well as to restore left ventricular function in congestive heart failure patients and prevent transplant rejection.
MSCs, sometimes also referred to as marrow stromal stem cells, were first identified in the guinea pig by Friedenstein in 1968 [51]. The first human MSCs were isolated by Haynesworth and Caplan in 1991 [52]. Specific cell surface markers for MSCs are unavailable; hence, the International Society of Cellular Therapy in 2006 defined that MSCs must meet the following three criteria: (1) Under standard tissue culture conditions, MSCs must grow while attached. (2) They must express specific cell surfaces markers, such as CD73, CD90, and CD105, but not express the hematopoietic markers CD11b, CD14, CD34, or CD45. 3) They must have the capacity to differentiate under in vitro conditions into mesenchymal lineages, including adipocytes, osteoblasts, and chondroblasts [53]. These characteristics of isolated human MSCs were all originally described by Pittenger et al. [54].
MSCs are multipotent cells that can differentiate into mesodermal, endodermal, and ectodermal cells [55]. MSCs also induce hematopoietic cells to release several regulatory factors, such as growth factors and anti-inflammatory cytokines, which can modulate the immune response [56]. MSCs were first introduced as an adherent to a clonogenic, non-phagocytic, fibroblastic cell population. Furthermore, these cells can be isolated from various other tissues, including adipose tissue, umbilical cord, placenta, and amniotic fluid [55].

4.1. Allogeneic MSCs

Allogeneic MSCs are exogenous stem cells from the same species as MSCs, but they are not genetically identical. The advantages of allogeneic MSCs (allo-MSCs) in therapeutic applications are manifold compared with autologous MSCs (auto-MSCs). Moreover, auto-MSCs have several associated limitations, including the difficulty of obtaining adequate auto-MSCs from some patients, such as light-weight patients or patients with myelofibrosis. Auto-MSCs isolated from donors of advanced age show decreased biological activity, including activity regarding their differentiation and regenerative potential [57]. Some systemic illnesses, such as diabetes, can affect the inherent features of auto-MSCs, hence reducing their protective effect. Furthermore, it is challenging to collect sufficient amounts of healthy auto-MSCs with high activity from individuals with this condition [58]. The first study seeking to mix MSCs with isolated immune cells—T cells, B cells, and NK cells—and show that each responded to MSCs was undertaken by Aggarwar and Pitternger [59], and this was also the first example of the identification of PGE2 as an effector released from MSCs.
Some other limitations of using auto-MSCs include the very time-consuming nature of auto-MSCs extraction, meaning it is hard to use them quickly against acute diseases such as stroke and myocardial infarction. Meanwhile, allo-MSCs are easily obtainable, allowing for immediate administration. As a result, using allo-MSCs represents a promising alternative therapy to auto-MSCs, with advantages in terms of time and cost efficiency, as well as quality assurance [60]. The activity of various immune cells, including dendritic cells, NK cells, B cells, and T cells, through cell-to-cell contact and soluble factors can be inhibited by MSCs [61]. Allo-MSCs also contribute to lowering the immune response after implantation, owing to their low immunosuppressive and immunogenicity properties.
MSCs exhibit low levels of MHC class I molecules on their surface and lack expressions of MHC class II as well as the co-stimulatory molecules CD80 (B7-1), CD86 (B7-2), and CD40, which may limit immune recognition. However, under infection conditions during which pro-inflammatory factors are released and inflammation occurs, the expression of MHC-II can be up-regulated in antigen-presenting cells (APCs) [62]. Klyushnenkova et al. supported this statement after demonstrating that MSCs treated with IFN-γ showed up-regulated MHC-II molecules [63]. Additionally, MSCs under inflammatory circumstances show an up-regulation of the co-stimulatory molecule CD40. However, T cells are not fully activated because CD40 expression is counterbalanced by the inhibitory molecule CD274 (also known as PD-L1), which is also expressed by MSCs [64]. The hypothesis that allo-MSCs have the same efficacy as auto-MSCs has been proven. However, in vivo studies have revealed that allo-MSCs are not completely immunogenic, thus possibly eliciting an immune response despite their low immunosuppressive and immunogenicity. In vivo and in vitro studies have both reported these findings. However, until recently, this has been a point of contention, with inconclusive results [65,66].

4.2. MSCs Mode and Delivery

There are several techniques for MSCs administration, depending on the underlying clinical indications and pathological abnormalities. The proper selection of the mode of delivering MSCs provides optimal therapeutic efficacy [67]. There are two main methods used to deliver cells into the body: local delivery into the tissue, and systemic delivery. Local delivery is via injection, for example intraperitoneal (IP), intramuscular (IM), or intracardiac, whereas systemic delivery methods involve the administration of cells intravenously (IV) or via intra-arterial (IA) paths [68]. The alternative route of delivery is direct injection (DI) into the tissue or organ targeted in order to escalate the engraftment and/or local differentiation [69].
Another method commonly used to deliver MSCs is via intravascular (IV) infusion, owing to its ease of utilization and low risk [70]. However, cells administered through IV have to pass through the lungs first before being distributed to the rest of the body. This condition leads to a substantial problem called the pulmonary “first-pass” effect, which produces high numbers of entrapment cells [71]. This situation occurs because MSCs are approximately 20–30 µm in diameter, and most particles of this size are filtered out by the lungs [71]. Nevertheless, MSCs entrapment in the lungs provides therapeutic effects on the targeted tissues. A concise review study concluded that MSCs isolated from bone marrow alter the tissue microenvironment by secreting soluble factors (autocrine–paracrine). This makes a major contribution to tissue improvement via the trans-differentiation capacity [72]. Several studies have proven that engraftment plays an important role in MSCs therapy aimed at injury; however, this engraftment ability may not play a key therapeutic role in the lungs. Because lungs’ engraftment ability is less than 5%, the paracrine mechanisms play greater roles [73,74]. The beneficial effects of MSCs are derived from their capacity to secrete paracrine factors that modulate immune response and alter the response of the epithelium, endothelium, and inflammation cells to injury. Administration via IV has been extensively reported to confer therapeutic effects through mechanisms involving secondary signaling effector cell systems and interactions with the host immune systems [69].
Theoretically, the intra-arterial (IA) delivery route enables cells to pass through the lungs at least once, thus avoiding the pulmonary “first-pass” effect, and consequently more cells are able to reach the targeted tissues [68].
Several studies have reported the positive therapeutic effects of MSCs; however, there are still a lot of questions and challenges related to the application of MSCs. Cell therapy can induce cell emboli, which may potentially increase mortality. This type of embolism depends on the concentrations and rates of cell administration [69,75]. Other risks may arise, such as MSCs transformation, potential adverse inflammatory effects, tumor formation, or thrombosis associated with the intravenous infusion of MSCs [70]. Until recently, the safety of MSCs therapy has been under question, but the efficacy and the resulting interaction in the microenvironment of the host remain controversial.

4.3. The Mechanisms of Paracrine, Endocrine, and Cell Engraftment of MSCs

The mechanisms of MSCs as immunomodulatory, improving and regenerating cells, still need to be explored. Some articles have mentioned that paracrine mechanisms are enacted through the secretion of soluble mediators and cell-to-cell contact. Cell-to-cell contact depends on paracrine mechanisms in cells with limited differentiation potential, such as MSCs [76]. The soluble factors include interleukin-1 receptor antagonist [77], interleukin-10 [78,79], prostaglandin E2 (PGE2) [78], LL-37 [80], keratinocyte growth factor [81], and angiopoietin-1 [82].
A study conducted by Ortiz et al. [83] suggested the activity of paracrine mechanisms enacted via soluble factors rather than trans-differentiation mechanisms. Other studies have stated that the role of trans-differentiation is less significant than that of paracrine effects caused by the release of soluble factors in response to acute lung damage [84]. Moreover, the capacity for lung engraftment is less than 5%, and, thus, the paracrine mechanism plays a significant role [73,74].
Krause et al. [85] proved that bone-marrow-derived cells can produce cells from several different organs, including the lungs. Preliminary studies suggest that engraftment plays a vital role in treating injury by MSCs. However, the engraftment properties of the lung do not play a significant therapeutic role. Instead, the capacity of MSCs to secrete paracrine factors is beneficial in modulating immune responses and in altering the responses of the endothelium or epithelia and inflammation cells toward injury [62,86,87].

4.4. MSCs and Niche Inflammation

In addition to the repair and regeneration activities of MSCs in microenvironments where inflammation occurs (inflammation niche), evidence suggests that MSCs have immunomodulatory properties which help in maintaining the immature state of dendritic cells (DCs) through the inhibition of MHC class II, CD1a, CD40, CD80, and CD86 expression and the suppression of pro-inflammatory cytokines’ production [88]. An in vitro study showed that the ability of MSCs to secrete PGE can be increased by altering their environmental conditions. The study showed that exposure to hypoxia can stimulate angiogenic and antiapoptotic factors, such as VEGF, FGF-2, HGF, and IGF-1. MSCs’ exposure to IFγ and TNf-α in culture increased the expressions of PGE2, IDO, TGFβ, and HGF [89].
The immunosuppressive effect produced by MSCs is influenced by several conditions, such as the source of the isolated MSCs, the number of passages in the culture prior to use, the MSC dosages, and the recipient’s specific pathological conditions; however, the immunomodulatory roles of MSCs have attracted great interest on the part of researchers and clinicians [90].

4.5. The Immunobiology and Communication between MSCs and Damaged Tissue

MSCs in situ play an essential role in the cellular homeostasis of host tissue by replacing dead or dysfunctional cells. The capacity of MSCs to modulate immune responses has been shown in an in vitro study [91]. Tissue injury is often accompanied by inflammation, and the inflammation factors may cue a signal that mobilizes MSCs to the damaged tissue. Before carrying out the repair function, the initial preparation of the microenvironment is required.
Tissue damage is commonly accompanied by the activation of immunological or inflammatory cells. The factors produced by cells undergoing apoptosis, necrosis, microvasculature damage, and stroma recruit not only macrophages and neutrophils but also adaptive immune cells, such as CD4 T lymphocytes, CD8 T lymphocytes, and B cells [91,92]. Meanwhile, phagocytic cells release inflammatory mediators, such as TNFα, IL-1β, chemokines, free radicals, and leukotrienes, in response to cell damage. Thus, inflammatory chemicals and immune cells, fibroblasts, and endothelial cells regulate changes in the microenvironment, and as a result, MSCs mobilize and develop, stimulating the replacement of damaged tissue cells. MSCs may originate either from tissue-resident cells or bone marrow. However, the processes of the mobilization of MSCs to damaged tissue sites are poorly understood. Moreover, the mechanisms of how MSCs survive and differentiate into different cell types remain unclear [90].
Cytokines, such as TNFα, IL-1β, and IFNγ, toxins from infectious agents, or hypoxia can induce the release of many MSCs growth factors, such as fibroblast growth factor (FGF), epidermal growth factor (EGF), platelet-derived growth factor (PDGF), keratinocyte growth factor (KGF), vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), transforming growth factor-β (TGF-β), insulin growth factor-1 (IGF-1), angiopoietin-1 (Ang-1), and stromal-cell-derived factor-1 (SDF-1), all of which are induced once the MScs enter the microenvironment [90,93]. The development of fibroblasts, endothelial cells, and tissue progenitor cells is then promoted by these growth factors, which is essential to tissue regeneration and repair [90].

4.6. The Immunosuppression Property of MSCs

The inflammatory niche determines the ability of MSCs to inhibit immune cell activity. IFNγ, together with pro-inflammatory cytokines, such as IL-1α, TNFα and IL-1β, the occurrence of a chemokine burst, and the expression of adhesion molecules, including intercellular adhesion molecule-1 (ICAM-1), CCR5 ligand, CXCR3 ligand, and vascular cell adhesion molecule-1 (VCAM-1), can stimulate MSCs to release large amounts of immunosuppressive factors [94]. The co-activation of these inflammatory molecules induces the accumulation of immune cells near to MSCs; these microenvironmental conditions stimulate MSCs into potent immunosuppressive behavior. Immunosuppressive factors, such as PGE2, IL-10, IL-6, LIF, TSG6, HO-1, and CCL2, can also affect immune cell activation [90]. The communication between the MSCs in damaged tissue and the immunosuppressive property of MSCs can be seen in Figure 2. PGE2 is an immunosuppressive factor with roles and mechanisms that should be investigated comprehensively in the future.

4.7. The Immunomodulation Roles of Exogenous MSCs in Lung Injury

MSCs have unique immunomodulatory and paracrine properties that are advantageous in treating diseases associated with inflammation and organ injury [95]. Several studies have proven that MSCs enact immunosuppressive effects by inhibiting the activity of innate and adaptive immune cells. Cell-to-cell contact mechanisms mediate the immunosuppressive effects. In vitro studies have shown that the PGE2 derived from murine and human MSCs acts as an immunosuppressant by inducing Foxp3þTregs [96], inhibiting NK cells [97], inducing M2 cells [98], and inhibiting dendritic cell maturation [99]. Research on sepsis models has proven that MSCs derived from bone marrow cells can improve sepsis via immunosuppression mechanisms through PGE2. PGE2 also reprograms the host macrophages to increase the production of IL-10, an anti-inflammatory cytokine [78]. Research by Sun et al. [100] administered intratracheal MSCs 4 h after lung injury on mice models with lipopolysaccharide (LPS)-induced ALI and demonstrated that alveolar levels of CD4+CD25+Foxp3+ Treg were reduced and the balance of anti-inflammatory and pro-inflammatory cytokines was maintained. The study by Mei et al. [101] proved that the administration of MSCs intravenously 6 h and 24 h after polymicrobial lung injury in mice models reduced acute inflammatory responses and increased phagocytosis via bacterial clearance.
The study of Ortiz et al. [77] proved that the intravenous administration of MSCs immediately after lung injury in a rat model due to the intratracheal administration of bleomycin could induce the secretion of IL-1 receptor antagonists, prevent macrophages from producing TNFα, and inhibit IL-1α secretion by T lymphocyte cells. In contrast, Rojas et al. [74] proved that the intravenous administration of bone-marrow-derived MSCs in mice models 6 h after lung injury caused by intratracheal administration of bleomycin increased G-CSF and GM-CST levels in circulation and reduced inflammatory cytokines. Furthermore, Maron-Gutierrez et al. [102] demonstrated that intravenous MSC administration to a murine model with ARDS one day after LPS induction reduced inflammation, inhibited fibrogenesis, increased MMP-8 expression, decreased TIMP-1 expression, and caused a shift in macrophage phenotype from M1 to M2.
Several studies used MSCs that were not derived from bone marrow, including Shin et al. [103], with outcomes suggesting that the intravenous administration of human adult stem cells from adipose tissue 30 min after the induction of LPS in rat models with endotoxemia decreased the level of inflammatory cytokines in serum and lungs and prevented renal apoptosis and repaired multiorgan injury. Zhang et al. [104] proved that administering human stem cells and mouse stem cells derived from adipose tissue in mouse models with LPS-induced ARDS increased IL-10 levels and reduced neutrophil levels, lung permeability, and pro-inflammatory cytokine production. Furthermore, Li et al. [28] proved that the intravenous administration of MSCs derived from the human umbilical cord 1 h after LPS induction in rat models with ARDS could reduce mortality, as well as serum TNFα, IL-1β, and IL-6, without reducing IL-10 levels. Chang et al. [105] demonstrated that the intratracheal administration of stem cells derived from the umbilical cord in rat models on postnatal Days 3 and 10 showed a significant protective effect only at the beginning of the inflammation. Several publications evaluating the success of non-mesenchymal-cell-based therapy in ARDS, including that by Ornellas et al. [106], have proven that the administration of BMDMC reduced inflammation and remodeling in mouse models with ARDS due to sepsis. Nandra et al. [107] also suggested that the intravenous administration of BMDMCs in rat models with hemorrhagic shock could reduce kidney and liver damage through the enhancement of PKB and glycogen synthase kinase (GSK)-3 phosphorylation, the weakening of NF-κB activation, and ERK1/2 and ICAM-1 phosphorylation.
A study by Gorman et al. [108] demonstrated that in mechanically ventilated patients with moderate to severe ARDS, increasing doses (100, 200, or 400 × 106 cells) of a single intravenous infusion of human umbilical-cord-derived MSCs (ORBCEL-C) were well-tolerated, supporting the safety of the intravenous administration of ORBCEL-C. However, no important trends over time (from baseline to Days 4, 7, and 14) were identified, and there were no apparent dose-dependent effects of MSCs on any of the plasma markers of systemic inflammation (IL-6, IL-8, or IL-18), epithelial cell injury (SP-D), or endothelial injury/activation (Ang-2/ICAM-1).
Dilogo et al. [23] investigated the intravenous administration of umbilical cord mesenchymal stromal cells in critical patients with COVID-19. They revealed reductions in IL-6 in the MSCs group and an IL-10 increment on Day 7 after MSC application in comparison to baseline. The study also showed the increased expression of LIF (leukemia inhibitory factor) in 80% of recovered patients who were treated with MSCs. LIF shows the ability to repair and regenerate through stem cell niches of type II alveolar epithelial cells. They also saw the suppression of the populations of CD8-CXCR3 and CD56-CXCR3 after MSC application. VEGF also showed an increasing trend in the MSCs group, compared with a decreasing trend in the control group on Day 7. VEGF is an angiogenic factor that is essential in the recovery of damaged lungs, which must be introduced into circulation so that the regeneration of lung capillaries can occur.
Adas et al. [109] suggested that administering MSCs to critically ill COVID-19 patients decreased serum ferritin, IL-6, fibrinogen, and CRP levels significantly. The levels of other pro-inflammatory cytokines (IFNγ, IL-2, IL-12, and IL-17A) appeared to steadily decrease in the MSC transplanted group. The anti-inflammatory cytokines IL-10, IL-13, and IL-1ra appeared to increase steadily and statistically significantly from baseline.

4.8. The Effect of Exogenous Mesenchymal Stem Cells on Influenza-Virus-Induced Lung Injury

MSCs’ administration to ARDS induced by influenza A viral infection has yet to be widely studied; however, investigation on humans is still limited. Several studies focusing on different sub-types of the influenza virus have shown beneficial results. Table 1 summarizes the research and the viruses used, the sources of the MSCs administered, and the results.

5. Prostaglandin E2 Acts as an Immunosuppressive Properties of Mesenchymal Stromal Cells

Depending on the stimulation in the microenvironment, MSCs can act in both immune activation and immune suppression. These mechanisms are also known as sensory functions and “switchers” of the body’s immune system [116]. PGE2 plays a key role in the immunosuppressive properties of MSCs. An in vitro study demonstrated that MSCs exposed to IFN-γ and TNF-α in a culture of MSCs increased the expression of PGE2 [89]. MSCs constitutively secrete the soluble factors of PGE2 without the activation signal. In addition, PGE2 is easily detected in the culture very quickly in only 4 h [117].
Li et al. suggested that MSCs transfected to overexpress cyclooxygenase (COX)-2 is a potent immunomodulator [118]. Daniel et al. [117] also confirmed that PGE2 indicates the therapeutic efficacy of mesenchymal stem cells; they proved that exogenous PGE2 is sufficient to inhibit the secretion of TNF-α and IFN-γ. PGE2 not only represents the crucial immunomodulator factor expressed by MSCs but also functions as a marker to assess the efficacy of MSC therapy. A study by Yañez et al. [119] suggested that myeloid dendritic cells (m-DCs) co-cultured with Ad-MSCs or BM-MSCs notably increased the level of PGE2 detected in the culture of MSCs.
As an essential mediator, PGE2 modulates many biological functions, including immune responses, blood pressure, gastrointestinal integrity, and fertility. PGE2 dysregulation or degradation has been linked to a number of pathological conditions [120]. PGE2 is a prostaglandin produced from arachidonic acid (AA) by COX. Many cell types, including macrophages and dendritic cells, can produce COX-derived PGE2 [121]. PGE2 is an essential mediator that modulates various physiological and pathophysiological processes through autocrine or paracrine mechanisms. PGE2 is mediated by four G-protein-coupled receptors (EP1-EP4), each of which has a unique physiological and pathophysiological effect [121]. PGE2 is involved in the proliferation and migration of several cell types, in addition to its role in the inflammatory response [122,123].
Some evidence suggests the beneficial effects of PGE in the lung, and its role in several physiological processes, such as the maintenance of barrier integrity [124,125]. Several studies have shown that PGE2 may trigger cell proliferation and inhibit apoptosis in lung epithelial cells [124,125]. COX is a key enzyme in arachidonic acid’s (AA) conversion into prostaglandin. COX can maintain and regulate the physiological functions of blood vessels, kidneys, and other organs by producing substances such as PGE2. Some studies have found that PGE/COX-2 is closely correlated with the Wnt/β-catenin signaling pathway. An in vitro study proved that PGE2 activation enables unphosphorylated β-catenin to accumulate in the cytoplasm, then get into the nucleus and activate genes involved in regeneration functions [122,126,127,128]. Another study by Zheng et al. [129] demonstrated that PGE2/COX-2 was involved in fracture healing by promoting the transcription and translation of the key gene of β-catenin in the Wnt/β-catenin signaling pathway; this pathway facilitates the formation of calluses and osteoblast proliferation and accelerates fracture healing.
The activation of the β-catenin pathway not only functions in the regeneration process but also is active in immunoregulation mechanisms. According to several studies, the mechanisms of immunoregulation act via the cross-regulation between the Wnt/β-catenin and NF-κB signaling pathways [130,131,132,133,134].

6. PGE2 Promotes Immunoregulation through Activation of Wnt-β-Catenin Signaling

PGE2, produced via the COX pathway, has three identified COX isoforms. One is COX-2, which is undetectable in most tissues but is activated in response to various inflammatory mediators, including pro-inflammatory cytokines. COX-2 is associated with the pathogenesis of several diseases [135,136]. The literature mentions that in damaged tissue, PGE2 is increased as a function of the homeostasis mechanism [124].
The β-catenin protein is a subunit of the cadherin protein complex that acts as an intracellular signal transducer, which is also involved in the regeneration and immunoregulation process. β-catenin is a double-function protein that regulates the coordination of cell adhesion and the transcription of genes. β-catenin is homologue with γ-catenin, also known as plakoglobin, and is expressed in various tissues [137]. The Wnt/β-catenin signaling pathway is a crucial pathway for several biological processes. The aberration of this signaling can be found in several human diseases [138].
Wnt/β-catenin signaling also acts as the regulation pathway of the stem cells niche in several tissues. However, the role of Wnt signaling in the immunoregulatory function has not been extensively reported, although it is broadly involved in the regulation of adult MSCs’s fate in various tissues. Through the canonical pathway, this Wnt/β-catenin pathway helps the maintenance of MSCs and improves their proliferation ability as a function of regeneration [29].
The available studies suggest that one of the main roles of Wnt signaling is determining the fate of and controlling stem cells [139]. Gehart and Clevers [140] reported one of the main communicative roles in the stem cells niche is played by the Wnt/β-catenin pathway. The activation of the canonical Wnt pathway, which is dependent on β-catenin, is essential to the biology of MSCs and induces differentiation and proliferation [139].
The elevation of β-catenin under certain tissue damage conditions is indispensable. During viral infection, different transduction signaling pathways control various gene expressions that regulate several molecular and cellular activities required for the eradication of microorganisms and the regulation of inflammation [134]. The inflammation response must be regulated tightly because uncontrolled inflammation can lead to tissue damage. Hence, the β-catenin signaling pathway is crucial due to its role in inhibiting the expression of several inflammation molecules during pathogen infection. In addition, β-catenin signaling also plays an essential role in the processes of cell differentiation and stem cell self-renewal maintenance [141].

6.1. β-Catenin Protein Activates Wnt-β-Catenin Signaling

β-catenin is a CTNNB1 protein encoded by the human CTNNB1 gene that regulates the coordination of adhesion between cells and gene transcription. This protein acts as the main signal transmitter in the Wnt signaling pathway. β-catenin is also detected in adherent junctions, forming a complex with E cadherin and actin filaments from the cytoskeleton [142].
Wnt/β-catenin is a fundamental pathway essential for embryogenesis, cell differentiation, and the maintenance of stem cell self-renewal in humans, lime molds, and worms. Because of the destruction of the complex controlling the level of free β-catenin (which refers to the form unbound to the E-cadherin complex), β-catenin is undetectable in the cytoplasm, nucleus, or unstimulated cells. This complex is made up mainly of the tumor suppressor adenomatous polyposis coli (APC) and Axin, which serves as a protein scaffold for the Ser/Thr protein kinase (casein kinase 1α (CK1α)) and GSK-3β to phosphorylate β-catenin at the N-terminal residues Ser45, Ser33, Ser37, and Thr41. This phosphorylation labels β-catenin, enabling it to be polyubiquitinated by the Skp1-Cdc53-F-Box E3 ubiquitin ligase complex (SCFβ–TRCP) and degraded by the 26S proteasome. GSK-3α is able to regulate β-catenin phosphorylation both in vitro and in vivo, suggesting that both isoforms of GSK-3 contribute to maintaining low levels of β-catenin under baseline conditions [134].

6.1.1. Wnt Signaling and Regulation

Axin molecules oligomerize with each other in resting cells via the C-terminal DIX domain, which has two interfaces that bind together, forming linear oligomers or even polymers in the cell’s cytoplasm. The only other protein known to have a DIX domain is Dishevelled, which has unique characteristics. A single Dsh protein from Drosophila is related to three paralogous genes in mammals, namely Dvl1, Dvl2, and Dvl3. It corresponds to the cytoplasmic region of the receptor frizzled with its domains, namely PDZ and DEP. When the Wnt molecule binds to the frizzled domain, it starts a cascade that exposes Dishevelled’s DIX domain and creates a site that can completely hook Axin. The oligomer of axin is titrated away, and the β-catenin complex is destroyed by Dsh [143].
Frizzled has essential cytoplasmatic segments containing a GSK-3 pseudo-substrate sequence (Pro-Pro-Pro-Ser-Pro-x-Ser) that is pre-phosphorylated by CKI, namely LRP5 and LRP6 proteins [144]. Once bound to the receptor complex, Axin binds to β-catenin and activates GSK-3. In this way, the Axin-bound receptor prevents β-catenin-mediated phosphorylation. Therefore, the β-catenin is damaged yet continues to be produced up to an increased concentration. Once the level of β-catenin rises sufficiently to saturate all sites in the cytoplasm, β-catenin is translocated into the cell nucleus. After involving the transcription factors of LEF1, TCF1, TCF2, or TCF3, β-catenin releases other transcription factors, binds to transcriptional activators, and then switches to target genes [144].

6.1.2. Canonical Signaling Pathway

Canonical Wnt signal activates when the Wnt ligand binds to the Fz receptor and the LRP coreceptor, activating Dvl and inhibiting GSK-3β. Without a Wnt ligand, the activation of Wnt signaling does not occur; GSK-3β, axin, and APC bind β-catenin. This binding stimulates the phosphorylation of β-catenin by GSK-3β and is followed by the ubiquitinylation of β-catenin, which is further degraded by proteasomes. β-catenin accumulates in the nucleus and binds to the T cell factor and lymphoid enhancer factors (TCF/LEF) to regulate gene transcription [145]. Figure 3A,B illustrate the canonical signaling pathway.

6.2. PGE2 Elevates the Accumulation of β-Catenin in Lung Injury

PGE2 levels in the lung compared with other organs tend to be higher, as evidenced by the isolation of alveolar epithelial cells that release high amounts of PGE2 [146]. According to in vitro studies using cell lines performed by Fujino et al. [126] and Fang et al. [147], PGE2 was shown to stimulate a double signaling cascade, comprising the activation of PI3K and protein kinase Akt by the free G-protein βγ subunit and the direct binding of the G-protein αs subunit to Axin. Although the activity of GSK-3β is regulated by these two pathways independently, the activation of GSK-3β reduces β-catenin signaling by promoting the phosphorylation of β-catenin, which is subsequently degraded by the 26S proteasome. Akt phosphorylation via PI3K signaling inhibits the activation of GSK-3β and APC in the protein subunits of the G-protein-linked PGE2 receptor EP2, as well as associating the activated α subunit of Gs with the RGS domain of Axin, so that the axin–GSK–3β complex releases β-catenin [122,126,147]. When PGE2 is activated in these two pathways, unphosphorylated β-catenin accumulates in the cytoplasm. T cell factor-4 (TCF4) and hypoxia-inducible factor-1 (HIF1) are two genes that β-catenin activates once it enters the nucleus, and this promotes cell survival, proliferation, and angiogenesis [122,126,127,128]. An in vitro study using human colon cancer cells conducted by Shao et al. [148] demonstrated that PGE2 activation escalates GSK-3 phosphorylation.
Consequently, β-catenin is accumulated, and the expression of T cell-4 transcription factor is induced, which forms a transcriptionally active complex with β-catenin. Furthermore, similar to the Wnt/β-catenin pathway, the translocation of β-catenin into the cell nucleus releases transcription factors, which then bind to transcriptional activators and switch to target genes [144]. Figure 3C illustrates the mechanisms by which PGE2 promotes immunoregulation through the activation of Wnt-β-catenin signaling.

6.3. Cross-Regulation between β-Catenin and NF-κB Signaling Pathways

The cross-regulation between the Wnt/β-catenin and NF-κB signaling pathways is essential to the regulation of diverse genes and active pathways, e.g., many physiological and pathological effects, regulations associated with the areas of growth, immune function, inflammation, tumorigenesis, tumor invasion, and metastasis, as well as cardiovascular and bone disease. However, the activity and signaling consequences are also regulated by direct interactions between the two pathways, resulting in diversity and complexity [130]. β-catenin and NF-κB can activate the expression of the gene-inducible nitric oxide synthase (iNOS), yet β-catenin also has an inhibiting effect on NF-κB. The functional cross-regulatory mechanisms between these two pathways play a complex role in Wnt/β catenin and NF-κB signaling in the pathogenesis of certain cancers and other diseases [130]. β-catenin plays an essential role in the development and function of homeostasis. Deng et al. [131] showed that catenin application in colon cancer decreased the transactivation activity and the expression of NF-κB target genes and inhibited the expression of NF-κB target genes, including Fas and TRAF1.
A colonic epithelial cell infection study with Salmonella typhimurium demonstrated the activity of β-catenin, which is able to inhibit NF-κB via two possible mechanisms: by stabilizing IκBα or by binding to NF-κB subunit p50, which prevents its transport from the cytoplasm to the nucleus and also occurs in the nucleus. Both mechanisms can cause a decrease in the transcriptional activity of NF-κB. The inhibition of NF-κB by β-catenin activation reduces inflammation. The study found that the infection of colonic epithelial cells with Salmonella typhimurium caused an elevation in GSK-3β-dependent β-catenin phosphorylation, leading to the up-regulation of Wnt2, IL-6, and IL-8 via TLR5, and thus activating NF-κB [132]. Liu et al. [133] proved that the ectopic over-expression of Wnt11 and Wnt2 decreased the transcriptional activity of NF-κB and AP1. This evidence suggests that Wnt 11 and Wnt2 activity triggers a negative feedback mechanism that controls NF-κB through β-catenin activation. Figure 3D illustrates the cross-regulation between the β-catenin and NF-κB signaling pathways.

7. The Mechanisms of Immunoregulation after Exogenous MSCs Administration on Virus-Induced Acute Lung Injury

Based on the available literature, the administration of MSCs could enhance endogenous PGE2, escalate the number of cells expressing β-catenin, and decrease the NF-κB transcription factor and TNFα pro-inflammatory cytokine expression, as well as IL-1β.
As previously explained, the characteristics of the innate immune response are the primary determinant of the outcome of influenza infection. PAMPs of the influenza A virus are recognized by three main classes of PRRs, including the cytosolic sensor RIG-I [34], TLR7 in endosomes [35], and NLR molecules [36]. These three pathways can induce the production of pro-inflammatory cytokines via NF-κB activation, which causes damage to lung pathology [33,36]. The inhibition of the NF-κB transcription factor inhibits the production of excessive pro-inflammatory cytokines due to exposure to the highly pathogenic influenza A virus, thereby preventing further tissue damage. MSCs exposure to pro-inflammatory cytokines, such as IFγ and TNfα in MSCs cultures, can increase the expression of PGE2 and other molecules [89]. According to the previously mentioned literature, the repair mechanisms of acute lung damage induced by the highly pathogenic influenza A virus begin with the release of excessive pro-inflammatory cytokines (cytokines storm), which stimulates allotransplanted MSCs to produce large amounts of active biomolecules. One of the active biomolecules is transforming growth factor-β (TGF-β), which can increase endogenous PGE2 expression by inducing COX2. A study by Fang et al. [149] suggested that TGF-β regulates COX-2 expression and PGE2 production in human granulosa cells.
The production of active biomolecules of PGE2 increases the accumulation of β-catenin in the cytoplasm. As previously mentioned, PGE2 has been proven to stimulate a double signaling cascade involving the activation of PI3K and protein kinase Akt by the free G-protein βγ subunit and the direct association of the G-protein αs subunit to Axin. The activation of GSK-3β promotes the phosphorylation of β-catenin and subsequent degradation by the 26S proteasome, reducing β-catenin signaling. On the other hand, Akt phosphorylation via PI3K signaling inhibits the activation of GSK-3β and APC in the protein subunits of G-protein-linked PGE2 receptor EP2, as well as the association of the activated α subunit of Gs with the RGS domain of Axin, so that the Axin–GSK–3β complex releases β-catenin [122,126,147].
In the further course of the disease, other pathways are needed to increase the activation of β-catenin, which serves to maintain homeostatic function in severe lung injury. The increase in β-catenin through the trans-differentiation process of the canonical Wnt/β-catenin pathway, which is usually involved in cell development through frizzled receptors, has a similar effect on PGE2, which functions to decrease the arrangement of the Axin-GSK-3β-APC complex, thereby stabilizing the accumulation of β-catenin in the cytoplasm [126,128,147]. The trans-differentiation process is characterized by the increased expressions of several Wnt ligands, the increased phosphorylation of Wnt signaling intermediates (i.e., LRP6 and DVL3), increased transcriptionally active β-catenin, and the increased expression of β-catenin-driven target genes that may initially be beneficial in lung injury [150,151].
The β-catenin protein plays a vital role in the process of immunoregulation. The inhibition of NF-κB transcription factor activity is a β-catenin pathway that plays a role in the immunoregulation mechanism. According to previous studies [84,131,148], β-catenin plays a vital role in homeostatic function by inhibiting NF-κB through cross-regulatory mechanisms. This cross-regulation is essential for regulating various genes and their active pathway, which involves controlling many physiological and pathological effects related to immune and inflammation functions.
Decreased NF-κB activity also has a significant direct effect on RAGE expression. The activation of the NF-κB pathway through RAGE can provide positive feedback. Furthermore, NF-κB can regulate the activation of RAGE, which causes an elevation of RAGE expression. This positive feedback cycle can further increase inflammation and tissue injury. These conditions occur mainly in diseases with elevated levels of the RAGE ligand [152,153]. Other studies have stated that HMGB1 signaling results in NF-κB expression. This activation promotes inflammation through a positive feedback loop because NF-κB can elevate the activation of various receptors, including RAGE [154,155]. Figure 4 illustrates the mechanisms of immunoregulation after exogenous MSCs administration on virus-induced acute lung injury.

8. Conclusions

The characteristics of innate immune response are essential to tissue damage caused by highly pathogenic influenza A virus infection. ARDS involves damage in the alveolar–epithelial–endothelial barrier of the lung, which causes the leakage of proteins of intravascular origin into the alveolar lumen. In addition to PAMPs, DAMPs are host molecules that regulate the activation of PRRs during influenza A virus infection. Activation of this molecule, in turn, triggers a signaling cascade that promotes the transcription factor NF-κB, which encodes pro-inflammatory cytokines and chemokines. Allotransplanted MSCs may exert a beneficial effect on tissue injury by inhibiting the transcription factor NF-κB. The effect of allotransplanted MSCs against inhibitory transcription factor NF-κB is known to operate through several related mechanisms, via the release of PGE2 and the accumulation of β-catenin.
The β-catenin protein plays a crucial role in the process of immunoregulation. Inhibition of the activity of transcription factor NF-κB is one of the β-catenin pathways that play a role in the mechanism of immunoregulation. The cross-regulatory mechanism of β-catenin in the cytoplasm activates Iκβ, thereby inhibiting NF-κB activity. However, there are still many other pathways that are involved in the mechanism of immunoregulation through inhibiting the innate immune system in addition to inhibiting NF-κB activation. Thus, the utilization of these approaches in acute lung damage caused by the highly pathogenic influenza A virus requires further research both in animal models and humans. Furthermore, the innate immune system is likely to activate its adaptive function to perpetuate the process. Therefore, it is necessary to evaluate the effects of MSCs on the adaptive immune system. Hopefully, a better understanding can be formed of the mechanisms of pro-inflammatory pathways that contribute to lung injury and host defenses, which can inhibit inflammatory damage while maintaining the host defense system. The mechanism of immunoregulation through β-catenin activation requires further research into the development of methods that can modify MSCs in vitro before transplantation, one of which involves genetic modification through the transfection method, such that MSCs can be made to carry the β-catenin gene. Another alternative approach is to provide combination therapy with β-catenin and MSCs.

Author Contributions

R.Y. and K.S. conceived the topic; R.Y. reviewed the literature and prepared the manuscript; K.S. reviewed the literature and wrote some sections of the manuscript; R.Y. designed figures and tables; K.S. edited and revised the manuscript critically for intellectual content. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We would like to express our gratitude to the members of the Department of Pulmonology and Respiratory Medicine, Medical Faculty of Airlangga University, for their helpful discussions and contributions. We are also immensely grateful to Nimas Roro Gayatri for helping us in the editing and proofreading process. Figures were created in Biorender© at the Dr. Soetomo General Hospital, Department of Pulmonology and Respiratory Medicine, Surabaya, Indonesia.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ALIAcute lung injury
Allo-MSCsAllogeneic MSCs
Ang-1Angiopoietin-1
AP1Activator protein 1
APCAdenomatous polyposis coli
Aqp5Aquaporin 5
ARDSAcute respiratory distress syndrome
Auto-MSCsAutologous MSCs
BASCsBronchioalveolar stem cells
BM-MSCsBone-marrow-derived mesenchymal stem cells
CK1αCasein kinase 1α
COX-2Cyclooxygenase-2
CXCR4+Chemokine receptor 4+
DAMPsDanger-associated molecular patterns
DCsDendritic cells
ECMExtracellular matrix
EGFEpidermal growth factor
FARDSFulminant ARDS
FGFFibroblast growth factor
G-CSFGranulocyte colony-stimulating factor
GM-CSFGranulocyte macrophage colony-stimulating factor
GSK-3βGlycogen synthase kinase-3β
HAHemagglutinin
HGFHepatocyte growth factor
HMCsHuman mesangial cells
HSCsHematopoietic stem cells
ICAM-1Intercellular adhesion molecule-1
IGF-1Insulin growth factor-1
IκβInhibitory kappa beta
iNOSInducible nitric oxide synthase
IRFInterferon regulatory factor
JNKJun N kinase
KGFKeratinocyte growth factor
LEFLymphoid enhancer factors
LPSLipopolysaccharide
MDA 5Melanoma differentiation-associated gene 5
MSCsMesenchymal stem cells
NANeuraminidase
NLRNucleotide-binding domain-leucine-rich repeat-containing
PAMPsPathogen-associated molecular patterns
PDGFPlatelet-derived growth factor
PGE2Prostaglandin E2
PRRsPattern-recognition receptors
PI3KPhosophoinositide 3-kinase
RAGEReceptor for advanced glycation endproducts
RIG-1Retinol acid inducible gene 1
RNARibonucleic acid
RNPRibonucleate protein
SARSSevere acute respiratory syndrome
SCA 1Stem cell antigen 1
SCFStem cell factor
SDF-1Stromal-derived factor-1
SSEA-1Stage-specific embryonic antigen 1
ssRNASingle-stranded RNA
TBKTANK binding kinase
TCFT-cell factor
TGF-βTransforming growth factor-β
TLRToll-like receptor
VCAM-1Vascular cell adhesion molecule-1
VEGFVascular endothelial growth factor

References

  1. Dushianthan, A.; Grocott, M.P.; Postle, A.D.; Cusack, R. Acute respiratory distress syndrome and acute lung injury. Postgrad. Med. J. 2011, 87, 612–622. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Ware, L.B.; Matthay, M.A. The Acute Respiratory Distress Syndrome. N. Engl. J. Med. 2000, 342, 1334–1349. [Google Scholar] [CrossRef] [PubMed]
  3. Fanelli, V.; Vlachou, A.; Ghannadian, S.; Simonetti, U.; Slutsky, A.S.; Zhang, H. Acute respiratory distress syndrome: New definition, current and future therapeutic options. J. Thorac. Dis. 2013, 5, 326–334. [Google Scholar] [PubMed]
  4. Matthay, M.A.; Zemans, R.L. The acute respiratory distress syndrome: Pathogenesis and treatment. Annu. Rev. Pathol. Mech. Dis. 2011, 6, 147–163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Short, K.R.; Kroeze, E.J.B.V.; Fouchier, R.A.M.; Kuiken, T. Pathogenesis of influenza-induced acute respiratory distress syndrome. Lancet Infect. Dis. 2014, 14, 57–69. [Google Scholar] [CrossRef] [PubMed]
  6. Bauer, T.T.; Ewig, S.; Rodloff, A.C.; Müller, E.E. Acute respiratory distress syndrome and pneumonia: A comprehensive review of clinical data. Clin. Infect. Dis. 2006, 43, 748–756. [Google Scholar] [CrossRef]
  7. Sugamata, R.; Dobashi, H.; Nagao, T.; Yamamoto, K.-I.; Nakajima, N.; Sato, Y.; Aratani, Y.; Oshima, M.; Sata, T.; Kobayashi, K.; et al. Contribution of neutrophil-derived myeloperoxidase in the early phase of fulminant acute respiratory distress syndrome induced by influenza virus infection. Microbiol. Immunol. 2012, 56, 171–182. [Google Scholar] [CrossRef]
  8. World Health Organization. Disease Outbreak News; Avian Influenza A (H5N1)—Cambodia. Available online: https://www.who.int/emergencies/disease-outbreak-news/item/2023-DON445 (accessed on 20 March 2023).
  9. Bi, Z.; Hong, W.; Que, H.; He, C.; Ren, W.; Yang, J.; Lu, T.; Chen, L.; Lu, S.; Peng, X.; et al. Inactivated SARS-CoV-2 induces acute respiratory distress syndrome in human ACE2-transgenic mice. Signal Transduct. Target. Ther. 2021, 6, 349–360. [Google Scholar] [CrossRef]
  10. Barbeta, E.; Motos, A.; Torres, A.; Ceccato, A.; Ferrer, M.; Cilloniz, C.; Bueno, L.; Badia, J.R.; Castro, P.; Ferrando, C.; et al. SARS-CoV-2-Induced Acute Respiratory Distress Syndrome: Pulmonary Mechanics and Gas-Exchange Abnormalities. Ann. Am. Thorac. Soc. 2020, 17, 1164–1168. [Google Scholar] [CrossRef]
  11. Pfortmueller, C.A.; Spinetti, T.; Urman, R.D.; Luedi, M.M.; Schefold, J.C. COVID-19-associated acute respiratory distress syndrome (CARDS): Current knowledge on pathophysiology and ICU treatment—A narrative review. Best Pract. Res. Clin. Anaesthesiol. 2021, 35, 351–368. [Google Scholar] [CrossRef]
  12. Esteban, A.; Frutos-Vivar, F.; Muriel, A.; Ferguson, N.D.; Peñuelas, O.; Abraira, V.; Raymondos, K.; Rios, F.; Nin, N.; Apezteguía, C.; et al. Evolution of Mortality over Time in Patients Receiving Mechanical Ventilation. Am. J. Respir. Crit. Care Med. 2013, 188, 220–230. [Google Scholar] [CrossRef] [PubMed]
  13. Hsu, J.; Santesso, N.; Mustafa, R.; Brozek, J.; Chen, Y.L.; Hopkins, J.P.; Cheung, A.; Hovhannisyan, G.; Ivanova, L.; Flottorp, S.A.; et al. Antivirals for treatment of influenza: a systematic review and meta-analysis of observational studies. Ann. Intern. Med. 2012, 156, 512–524. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Thanunchai, M.; Hongeng, S.; Thitithanyanont, A. Mesenchymal Stromal Cells and Viral Infection. Stem Cells Int. 2015, 2015, 860950. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Cao, C.; Zhang, L.; Liu, F.; Shen, J. Therapeutic Benefits of Mesenchymal Stem Cells in Acute Respiratory Distress Syndrome: Potential Mechanisms and Challenges. J. Inflamm. Res. 2022, 15, 5235–5246. [Google Scholar] [CrossRef] [PubMed]
  16. Lohan, P.; Coleman, C.M.; Murphy, J.M.; Griffin, M.D.; Ritter, T.; E Ryan, A. Changes in immunological profile of allogeneic mesenchymal stem cells after differentiation: Should we be concerned? Stem Cell Res. Ther. 2014, 5, 99–109. [Google Scholar] [CrossRef] [Green Version]
  17. Thavapalachandran, S.; Le, T.Y.L.; Romanazzo, S.; Rashid, F.N.; Ogawa, M.; Kilian, K.A.; Brown, P.; Pouliopoulos, J.; Barry, A.M.; Fahmy, P.; et al. Pluripotent stem cell-derived mesenchymal stromal cells improve cardiac function and vascularity after myocardial infarction. Cytotherapy 2021, 23, 1074–1084. [Google Scholar] [CrossRef] [PubMed]
  18. El-Sawah, S.G.; Rashwan, H.M.; Althobaiti, F.; Aldhahrani, A.; Fayad, E.; Shabana, E.S.; El-Hallous, E.I.; Amen, R.M. AD-MSCs and BM-MSCs Ameliorationg Effects on The Metabolic and Hepato-Renal Abnormalities in Type 1 Diabetic Rats. Saudi J. Biol. Sci. 2022, 29, 1053–1060. [Google Scholar] [CrossRef]
  19. Luo, F.; Jiang, W.; Xu, Y.; Liu, X.-M.; Wang, W.; Zhang, W.; Luo, C. The Mechanisms Involved in Mesenchymal Stem Cell Alleviation of Sepsis-Induced Acute Lung Injury in Mice: A Pilot Study. Curr. Ther. Res. 2020, 93, 100593. [Google Scholar] [CrossRef]
  20. Wang, Y.; Wang, J.-L.; Ma, H.-C.; Tang, Z.-T.; Ding, H.-R.; Shi, X.-L. Mesenchymal stem cells increase heme oxygenase 1-activated autophagy in treatment of acute liver failure. Biochem. Biophys. Res. Commun. 2019, 508, 682–689. [Google Scholar] [CrossRef]
  21. Zhou, T.; Liao, C.; Lin, S.; Lin, W.; Zhong, H.; Huang, S. The Efficacy of Mesenchymal Stem Cells in Therapy of Acute Kidney Injury Induced by Ischemia-Reperfusion in Animal Models. Stem Cells Int. 2020, 2020, 1873921. [Google Scholar] [CrossRef]
  22. Gad, E.S.; Salama, A.A.A.; El-Shafie, M.F.; Arafa, H.M.M.; Abdelsalam, R.M.; Khattab, M. The Anti-Fibrotic and Anti-Inflammatory Potential of Bone Marrow-Derived Mesenchymal Stem Cells and Nindetanib in Bleomycin-Induced Lung Fibrosis in Rats. Inflammation 2020, 43, 123–134. [Google Scholar] [CrossRef] [PubMed]
  23. Dilogo, I.H.; Aditianingsih, D.; Sugiarto, A.; Burhan, E.; Damayanti, T.; Sitompul, P.A.; Mariana, N.; Antarianto, R.D.; Liem, I.K.; Kispa, T.; et al. Umbilical cord mesenchymal stromal cells as critical COVID-19 adjuvant therapy: A randomized controlled trial. Stem Cells Transl. Med. 2021, 10, 1279–1287. [Google Scholar] [CrossRef] [PubMed]
  24. Li, L.L.; Zhu, Y.G.; Jia, X.M.; Liu, D.; Qu, J.M. Adipose-Derived Mesenchymal Stem Cells Ameliorating Pseudomonas Aeruginosa-Induced Acute Lung Infection via Inhibition of NLRC4 Inflammasome. Front. Cell. Infect. Microbiol 2021, 10, 581535. [Google Scholar] [CrossRef]
  25. Jankauskaite, L.; Schmoldt, C.; Lohmeyer, J.; Herold, S. Therapeutic potential of murine bone marrow derived mesenchymal stem cells in influenza virus-induced pneumonia. Pneumologie 2014, 68, A25. [Google Scholar] [CrossRef]
  26. Gotts, J.E.; Abbott, J.; Matthay, M.A. Influenza causes prolonged disruption of the alveolar-capillary barrier in mice unresponsive to mesenchymal stem cell therapy. Am. J. Physiol. Lung Cell. Mol. Physiol. 2014, 307, L395–L406. [Google Scholar] [CrossRef] [PubMed]
  27. Chan, M.C.W.; Kuok, D.I.T.; Leung, C.Y.H.; Hui, K.P.Y.; Valkenburg, S.A.; Lau, E.H.Y.; Nicholls, J.M.; Fang, X.; Guan, Y.; Lee, J.W.; et al. Human mesenchymal stromal cells reduce influenza A H5N1-associated acute lung injury in vitro and in vivo. Proc. Natl. Acad. Sci. USA 2016, 113, 3621–3626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Li, J.; Li, D.; Liu, X.; Tang, S.; Wei, F. Human umbilical cord mesenchymal stem cells reduce systemic inflammation and attenuate LPS-induced acute lung injury in rats. J. Inflamm. 2012, 9, 33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Zhang, Q.; Yu, J.; Chen, Q.; Yan, H.; Du, H.; Luo, W. Regulation of pathophysiological and tissue regenerative functions of mediated via the WNT signaling pathway (Review). Mol. Med. Rep. 2021, 24, 648–661. [Google Scholar] [CrossRef] [PubMed]
  30. Liu, J.; Xiao, Q.; Xiao, J.; Niu, C.; Li, Y.; Zhang, X.; Zhou, Z.; Shu, G.; Yin, G. Wnt/β-catenin signalling: Function, biological mechanisms, and therapeutic opportunities. Signal Transduct. Target. Ther. 2022, 7, 3. [Google Scholar] [CrossRef]
  31. Lin, Q.; Wu, L.; Chatla, S.; Chowdhury, F.A.; Atale, N.; Joseph, J.; Du, W. Hematopoietic stem cell regeneration through paracrine regulation of the Wnt5a/Prox1 signaling axis. J. Clin. Investig. 2022, 132, e155914. [Google Scholar] [CrossRef]
  32. Gürtler, L. Virolog of Human Influenza. In Influenza Report 2006; Kamps, B.S., Hoffmann, C., Presier, W., Eds.; Flying Publisher: Paris, France, 2006; pp. 87–91. Available online: https://www.paho.org/hq/dmdocuments/2010/Influenza%20Report%202006_Sebastian_2006.pdf (accessed on 9 December 2022).
  33. Owen, D.M.; Gale, M. Fighting the flu with inflammasome signaling. Immunity 2009, 30, 476–478. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Pichlmair, A.; Schulz, O.; Tan, C.P.; Naslund, T.I.; Liljestrom, P.; Weber, F.; Reis e Sousa, C. RIG-I-mediated antiviral responses to single-stranded RNA bearing 5′-phosphates. Science 2006, 314, 997–1001. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Lund, J.M.; Alexopoulou, L.; Sato, A.; Karow, M.; Adams, N.C.; Gale, N.W.; Iwasaki, A.; Flavell, R.A. Recognition of Single-Stranded RNA Viruses by Toll-Like Receptor 7. Proc. Natl. Acad. Sci. USA 2004, 101, 5598–5603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Pang, I.K.; Iwasaki, A. Inflammasomes as mediators of immunity against influenza virus. Trends Immunol. 2011, 32, 34–41. [Google Scholar] [CrossRef] [Green Version]
  37. Gaur, P.; Munjhal, A.; Lal, S.K. Influenza virus and cell signaling pathways. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2011, 17, RA148–RA154. [Google Scholar] [CrossRef] [Green Version]
  38. Dai, X.; Zhang, L.; Hong, T. Host cellular signaling induced by influenza virus. Sci. China Life Sci. 2011, 54, 68–74. [Google Scholar] [CrossRef] [Green Version]
  39. Osterholm, M.T. Preparing for the next pandemic. N. Engl. J. Med. 2005, 352, 1839–1842. [Google Scholar] [CrossRef] [Green Version]
  40. Pizzolla, A.; Smith, J.M.; Brooks, A.G.; Reading, P.C. Pattern recognition receptor immunomodulation of innate immunity as a strategy to limit the impact of influenza virus. J. Leukoc. Biol. 2017, 101, 851–861. [Google Scholar] [CrossRef]
  41. Betakova, T.; Kostrabova, A.; Lachova, V.; Turianova, L. Cytokines induced during influenza virus infection. Curr. Pharm. Des. 2017, 23, 2616–2622. [Google Scholar] [CrossRef]
  42. Freeman, C.M.; Martinez, F.J.; Han, M.K.; Ames, T.M.; Chensue, S.W.; Todt, J.C.; Arenberg, D.A.; Meldrum, C.A.; Getty, C.; McCloskey, L. Lung dendritic cell expression of maturation molecules increases with worsening chronic obstructive pulmonary disease. Am. J. Respir. Crit. Care Med. 2009, 180, 1179–1188. [Google Scholar] [CrossRef] [Green Version]
  43. Gu, Y.; Hsu, A.C.-Y.; Pang, Z.; Pan, H.; Zuo, X.; Wang, G.; Zheng, J.; Wang, F. Role of the Innate Cytokine Storm Induced by the Influenza A Virus. Viral Immunol. 2019, 32, 244–251. [Google Scholar] [CrossRef] [PubMed]
  44. Maines, T.R.; Szretter, K.J.; Perrone, L.; Belser, J.A.; Bright, R.A.; Zeng, H.; Tumpey, T.M.; Katz, J.M. Pathogenesis of emerging avian influenza viruses in mammals and the host innate immune response. Immunol. Rev. 2008, 225, 68–84. [Google Scholar] [CrossRef] [PubMed]
  45. Allen, I.C.; Scull, M.A.; Moore, C.B.; Holl, E.K.; McElvania-TeKippe, E.; Taxman, D.J.; Guthrie, E.H.; Pickles, R.J.; Ting, J.P.Y. The NLRP3 Inflammasome Mediates In Vivo Innate Immunity to Influenza A Virus through Recognition of Viral RNA. Immunity 2009, 30, 556–565. [Google Scholar] [CrossRef] [Green Version]
  46. Lamkanfi, M.; Dixit, V.M. Mechanisms and functions of inflammasomes. Cell 2014, 157, 1013–1022. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Tolle, L.B.; Standiford, T.J. Danger-associated molecular patterns (DAMPs) in acute lung injury. J. Pathol. 2013, 229, 145–146. [Google Scholar] [CrossRef] [Green Version]
  48. van Zoelen, M.A.D.; van der Sluijs, K.F.; Achouiti, A.; Florquin, S.; Braun-Pater, J.M.; Yang, H.; Nawroth, P.P.; Tracey, K.J.; Bierhaus, A.; van der Poll, T. Receptor for Advanced Glycation end Products Is Detrimental during Influenza A Virus Pneumonia. Virology 2009, 391, 265–273. [Google Scholar] [CrossRef] [Green Version]
  49. van Beijnum, J.R.; Buurman, W.A.; Griffioen, A.W. Convergence and amplification of toll-like receptor (TLR) and receptor for advanced gkycation end products (RAGE) signaling pathways via high mobility group B1 (HMGB1). Angiogenesis 2008, 11, 91–99. [Google Scholar] [CrossRef]
  50. Wang, M.; Yuan, Q.; Xie, L. Mesenchymal Stem Cell-Based Immunomodulation: Properties and Clinical Application. Stem Cells Int. 2018, 2018, 3057624. [Google Scholar] [CrossRef]
  51. 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]
  52. Haynesworth, S.E.; Goshima, J.; Goldberg, V.M.; Caplan, A.I. Characterization of cells with osteogenic potential from human marrow. Bone 1992, 13, 81–88. [Google Scholar] [CrossRef]
  53. Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.C.; Krause, D.S.; Deans, R.J.; Keating, A.; Prockop, D.J.; Horwitz, E.M. Minimal Criteria for Defining Multipotent Mesenchymal Stromal Cells: The International Society for Cellular Therapy Position Statement. Cytotherapy 2006, 8, 315–317. [Google Scholar] [CrossRef] [PubMed]
  54. Pittenger, M.F.; Mackay, A.M.; Beck, S.C.; Jaiswal, R.K.; Douglas, R.; Mosca, J.D.; Moorman, M.A.; Simonetti, D.W.; Craig, S.; Marshak, D.R. Multilineage potential of adult human mesenchymal stem cells. Science 1999, 284, 143–170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Weiss, D.J.; Kolls, J.K.; Ortiz, L.; Panoskaltsis-Mortari, A.; Prockop, D.J. Stem Cells and Cell Therapies in Lung Biology and Lung Diseases. Proc. Am. Thorac. Soc. 2008, 5, 637–667. [Google Scholar] [CrossRef] [PubMed]
  56. Newman, R.E.; Yoo, D.; Leroux, M.A.; Danilkovitch-Miagkova, A. Treatment of inflammatory diseases with mesenchymal stem cells. Inflamm. Allergy Drug Targets 2009, 8, 110–123. [Google Scholar] [CrossRef]
  57. Jung, H.G.; Ahn, E.-K.; Lee, J.-H.; Kim, Y.-H.; Leem, S.-H.; Heo, J.; Kim, H. Effects of harvesting sites and ages on adipose tissue-derived stem cells in rat. Tissue Eng. Regen. Med. 2014, 11, 137–142. [Google Scholar] [CrossRef]
  58. Cianfarani, F.; Toietta, G.; Di Rocco, G.; Cesareo, E.; Zambruno, G.; Odorisio, T. Diabetes impairs adipose tissue—Derived stem cell function and efficiency in promoting wound healing. Wound Repair Regen. 2013, 21, 545–553. [Google Scholar] [CrossRef]
  59. Aggarwal, S.; Pittenger, M.F. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood 2005, 105, 1815–1822. [Google Scholar] [CrossRef] [Green Version]
  60. Zhang, J.; Huang, X.; Wang, H.; Liu, X.; Zhang, T.; Wang, Y.; Hu, D. The challenges and promises of allogeneic mesenchymal stem cells for use as a cell-based therapy. Stem Cell Res. Ther. 2015, 6, 234. [Google Scholar] [CrossRef] [Green Version]
  61. Griffin, M.D.; Ryan, A.E.; Alagesan, S.; Lohan, P.; Treacy, O.; Ritter, T. Anti-donor immune responses elicited by allogeneic mesenchymal stem cells: What have we learned so far? Immunol. Cell Biol. 2013, 91, 40–51. [Google Scholar] [CrossRef]
  62. Sun, Y.Q.; Zhang, Y.; Li, X.; Deng, M.-X.; Gao, W.-X.; Yao, Y.; Chiu, S.-M.; Liang, X.; Gao, F.; Chan, C.W.; et al. Insensitivity of Human iPS Cells-Derived Mesenchymal Stem Cells to Interferon-γ-Induced HLA Expression Potentiates Repair Efficiency of Hind Limb Ischemia in Immune Humanized NOD Scid Gamma Mice. Stem Cells 2015, 33, 3452–3467. [Google Scholar] [CrossRef] [Green Version]
  63. 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 supression. J. Biomed. Sci. 2005, 12, 47–57. [Google Scholar] [CrossRef] [PubMed]
  64. Haddad, R.; Saldanha-Araujo, F. Mechanisms of T-Cell Immunosuppression by Mesenchymal Stromal Cells: What Do We Know So Far? Biomed Res. Int. 2014, 2014, 216806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Kurtzberg, J.; Prockop, S.; Teira, P.; Bittencourt, H.; Lewis, V.; Chan, K.W.; Horn, B.; Yu, L.; Talano, J.A.; Nemecek, E.; et al. Allogeneic Human Mesenchymal Stem Cell Therapy (Remestemcel-L, Prochymal) as a Rescue Agent for Severe Refractory Acute Graft-Versus-Host Disease in Pediatric Patients. Biol. Blood Marrow Transplant. 2014, 20, 229–235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Sbano, P.; Cuccia, A.; Mazzanti, B.; Urbani, S.; Giusti, B.; Lapini, I.; Rossi, L.; Abbate, R.; Marseglia, G.; Nannetti, G.; et al. Use of donor bone marrow mesenchymal stem cells for treatment of skin allograft rejection in a preclinical rat model. Arch. Dermatol. Res. 2008, 300, 115–124. [Google Scholar] [CrossRef] [Green Version]
  67. Urban, V.S.; Kiss, J.; Kovács, J.; Gócza, E.; Vas, V.; Monostori, E.; Uher, F. Mesenchymal stem cells cooperate with bone marrow cells in therapy of diabetes. Stem Cells 2008, 26, 244–253. [Google Scholar] [CrossRef] [PubMed]
  68. Kean, J.T.; Lin, P.; Caplan, A.I.; Dennis, J.E. MSCs: Delivery Routes and Engraftment, Cell-Targeting Strategies, and Immune Modulation. Stem Cells Int. 2013, 2013, 732742. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Caplan, H.; Olson, S.D.; Kumar, A.; George, M.; Prabhakara, K.S.; Wenzel, P.; Bedi, S.; Toledano-Furman, N.E.; Triolo, F.; Kamhieh-Milz, J.; et al. Mesenchymal Stromal Cell Therapeutic Delivery: Translational Challenge to Clinical Application. Front. Immunol. 2019, 10, 1645. [Google Scholar] [CrossRef]
  70. Moll, G.; Ankrum, J.A.; Kamhieh-Milz, J.; Bieback, K.; Ringdén, O.; Volk, H.D.; Geissler, S.; Reinke, P. Intravascular mesenchymal stromal/stemcell therapy product diversification: Time dor new clinical guidelines. Trends Mol. Med. 2019, 25, 149–163. [Google Scholar] [CrossRef] [Green Version]
  71. Fischer, U.M.; Harting, M.T.; Jimenez, F.; Monzon-Posadas, W.O.; Xue, H.; Savitz, S.I.; Laine, G.A.; Cox, C.S., Jr. Pulmonary passage is a major obstacle for intravenous stem cell delivery: The pulmonary first-pass effect. Stem Cells Dev. 2009, 18, 683–691. [Google Scholar] [CrossRef]
  72. Phinney, D.G.; Prockop, D.J. Concise review: Mesenchymal stem/multipotent stromal cells: The state of transdifferentiation and modes of tissue repair current views. Stem Cells 2007, 225, 2896–2902. [Google Scholar] [CrossRef]
  73. Kotton, D.N.; Fabian, A.J.; Mulligan, R.C. Failure of bone marrow to reconstitute lung epithelium. Am. J. Respir. Cell Mol. Biol. 2005, 33, 328–334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Rojas, M.; Xu, J.; Woods, C.R.; Mora, A.L.; Spears, W.; Roman, J.; Brigham, K.L. Bone marrow–derived mesenchymal stem cells in repair of the injured lung. Am. J. Respir. Cell Mol. Biol. 2005, 33, 145–152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Janowski, M.; Lyczek, A.; Engels, C.; Xu, J.; Lukomska, B.; Bulte, J.W.; Walczak, P. Cell size and velocity of injection are major determinants of the safety of intracarotid stem cell transplantation. J. Cereb. Blood Flow Metab. 2013, 33, 921–927. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Lee, J.W.; Fang, X.; Krasnodembskaya, A.; Howard, J.P.; Matthay, M.A. Concise review: Mesenchymal stem cells for acute lung injury: Role of paracrine soluble factors. Stem Cells 2011, 29, 913–919. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Ortiz, L.A.; DuTreil, M.; Fattman, C.; Pandey, A.C.; Torres, G.; Go, K.; Phinney, D.G. Interleukin 1 receptor antagonist mediates the antiinflammatory and antifibrotic effect of mesenchymal stem cells during lung injury. Proc. Natl. Acad. Sci. USA 2007, 104, 11002–11007. [Google Scholar] [CrossRef] [Green Version]
  78. Németh, K.; Leelahavanichkul, A.; Yuen, P.S.; Mayer, B.; Parmelee, A.; Doi, K.; Robey, P.G.; Leelahavanichkul, K.; Koller, B.H.; Brown, J.; et al. Bone marrow stromal cells attenuate sepsis via prostaglandin E2—Dependent reprogramming of host macrophages to increase their interleukin-10 production. Nat. Med. 2009, 15, 42–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Gupta, N.; Su, X.; Popov, B.; Lee, J.W.; Serikov, V.; Matthay, M.A. Intrapulmonary delivery of bone marrow-derived mesenchymal stem cells improves survival and attenuates endotoxin-induced acute lung injury in mice. J. Immunol. 2007, 179, 1855–1863. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Krasnodembskaya, A.; Song, Y.; Fang, X.; Gupta, N.; Serikov, V.; Lee, J.-W.; Matthay, M.A. Antibacterial effect of human mesenchymal stem cells is mediated in part from secretion of the antimicrobial peptide LL-37. Stem Cells 2010, 28, 2229–2238. [Google Scholar] [CrossRef] [Green Version]
  81. Lee, J.W.; Fang, X.; Gupta, N.; Serikov, V.; Matthay, M.A. Allogeneic human mesenchymal stem cells for treatment of E. coli endotoxin-induced acute lung injury in the ex vivo perfused human lung. Proc. Natl. Acad. Sci. USA 2009, 106, 16357–16362. [Google Scholar] [CrossRef] [Green Version]
  82. Mei, S.H.J.; McCarter, S.D.; Deng, Y.; Parker, C.H.; Liles, W.C.; Stewart, D.J. Prevention of LPS-induced acute lung injury in mice by mesenchymal stem cells overexpressing angiopoietin 1. PLoS Med. 2007, 4, e269. [Google Scholar] [CrossRef]
  83. Ortiz, L.A.; Gambelli, F.; McBride, C.; Gaupp, D.; Baddoo, M.; Kaminski, N.; Phinney, D.G. Mesenchymal stem cell engraftment in lung is enhanced in response to bleomycin exposure and ameliorates its fibrotic effects. Proc. Natl. Acad. Sci. USA 2003, 100, 8407–8411. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Prockop, D.J.; Kota, D.J.; Bazhanov, N.; Reger, R.L. Evolving paradigms for repair of tissues by adult stem/progenitor cells (MSCs). J. Cell. Mol. Med. 2010, 14, 2190–2199. [Google Scholar] [CrossRef] [PubMed]
  85. Krause, D.S.; Theise, N.D.; Collector, M.I.; Henegariu, O.; Hwang, S.; Gardner, R.; Neutzel, S.; Sharkis, S.J. Multi-organ, multi-lineage engraftment by a single bone marrow-derived stem cell. Cell 2001, 105, 369–377. [Google Scholar] [CrossRef] [Green Version]
  86. Glennie, S.; Soeiro, I.; Dyson, P.J.; Lam, E.; Dazzi, F.; Lutsiak, M.E.C.; Semnani, R.T.; De Pascalis, R.; Kashmiri, S.V.S.; Schlom, J.; et al. Bone marrow mesenchymal stem cells induce division arrest anergy of activated T cells. Blood 2005, 105, 2821–2827. [Google Scholar] [CrossRef] [PubMed]
  87. Rasmusson, I.; Ringdén, O.; Sundberg, B.; Le Blanc, K. Mesenchymal stem cells inhibit the formation of cytotoxic T lymphocytes, but not activated cytotoxic T lymphocytes or natural killer cells. Transplantation 2003, 76, 1208–1213. [Google Scholar] [CrossRef] [PubMed]
  88. Jiang, X.X.; Zhang, Y.; Liu, B.; Zhang, S.-X.; Wu, Y.; Yu, X.-D.; Mao, N. Human mesenchymal stem cells inhibit differentiation and function of monocyte-derived dendritic cells. Blood 2005, 105, 4120–4126. [Google Scholar] [CrossRef] [Green Version]
  89. Madrigal, M.; Rao, K.S.; Riordan, N.H. A review of therapeutic effects of mesenchymal stem cell secretions and induction of secretory modification by different culture methods. J. Transl. Med. 2014, 12, 260. [Google Scholar] [CrossRef] [Green Version]
  90. Ma, S.; Xie, N.; Li, W.; Yuan, B.; Shi, Y.; Wang, Y. Immunobiology of mesenchymal stem cells. Cell Death Differ. 2014, 21, 216–225. [Google Scholar] [CrossRef] [Green Version]
  91. Eming, S.A.; Krieg, T.; Davidson, J.M. Inflammation in wound repair: Molecular and cellular mechanisms. J. Investig. Dermatol. 2007, 127, 514–525. [Google Scholar] [CrossRef] [Green Version]
  92. Luster, A.D.; Alon, R.; von Andrian, U.H. Immune cell migration in inflammation: Present and future therapeutic targets. Nat. Immunol. 2005, 6, 1182–1190. [Google Scholar] [CrossRef]
  93. Shi, Y.; Su, J.; Roberts, A.I.; Shou, P.; Rabson, A.B.; Ren, G. How mesenchymal stem cells interact with tissue immune responses. Trends Immunol. 2012, 33, 136–143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Ren, G.; Zhao, X.; Zhang, L.; Zhang, J.; L’Huillier, A.; Ling, W.; Roberts, A.I.; Le, A.D.; Shi, S.; Shao, C.; et al. Inflammatory cytokine-induced intercellular adhesion molecule-1 and vascular cell adhesion molecule-1 in mesenchymal stem cells are critical for immunosuppression. J. Immunol. 2010, 184, 2321–2328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Baksh, D.; Song, L.; Tuan, R.S. Adult mesenchymal stem cells: Characterization, differentiation, and application in cell and gene therapy. J. Cell. Mol. Med. 2004, 8, 301–316. [Google Scholar] [CrossRef]
  96. English, K.; Ryan, J.M.; Tobin, L.; Murphy, M.J.; Barry, F.P.; Mahon, B.P. Cell contact, prostaglandin E(2) and transforming growth factor beta 1 play non-redundant roles in human mesenchymal stem cell induction of CD4 þ CD25(High) forkhead box P3 þ regulatory T cells. Clin. Exp. Immunol. 2009, 156, 149–160. [Google Scholar] [CrossRef] [Green Version]
  97. Spaggiari, G.M.; Capobianco, A.; Abdelrazik, H.; Becchetti, F.; Mingari, M.C.; Moretta, L. Mesenchymal stem cells inhibit natural killer-cell proliferation, cytotoxicity, and cytokine production: Role of indoleamine 2,3-dioxygenase and prostaglandin E2. Blood 2008, 111, 1327–1333. [Google Scholar] [CrossRef]
  98. Ylöstalo, J.H.; Bartosh, T.J.; Coble, K.; Prockop, D.J. Human mesenchymal stem/stromal cells cultured as spheroids are self-activated to produce prostaglandin E2 that directs stimulated macrophages into an anti-inflammatory phenotype. Stem Cells 2012, 30, 2283–2296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Spaggiari, G.M.; Abdelrazik, H.; Becchetti, F.; Moretta, L. MSCs inhibit monocyte-derived DC maturation and function by selectively interfering with the generation of immature DCs: Central role of MSC-derived prostaglandin E2. Blood 2009, 113, 6576–6583. [Google Scholar] [CrossRef] [PubMed]
  100. Sun, J.; Han, Z.-B.; Liao, W.; Yang, S.G.; Yang, Z.; Yu, J.; Meng, L.; Wu, R.; Han, Z.C. Intrapulmonary delivery of human umbilical cord mesenchymal stem cells attenuates acute lung injury by expanding CD4 + CD25+ Forkhead Boxp3 (FOXP3) + regulatory T cells and balancing anti- and pro-inflammatory factors. Cell. Physiol. Biochem. 2011, 27, 587–596. [Google Scholar] [CrossRef]
  101. Mei, S.H.; Haitsma, J.J.; Dos Santos, C.C.; Deng, Y.; Lai, P.F.; Slutsky, A.S.; Liles, W.C.; Stewart, D.J. Mesenchymal stem cells reduce inflammation while enhancing bacterial clearance and improving survival in sepsis. Am. J. Respir. Crit. Care Med. 2010, 182, 1047–1057. [Google Scholar] [CrossRef]
  102. Maron-Gutierrez, T.; Silva, J.D.; Asensi, K.D.; Bakker-Abreu, I.; Shan, Y.; Diaz, B.L.; Goldenberg, R.C.S.; Mei, S.H.J.; Stewart, D.J.; Morales, M.M.M.; et al. Effects of mesenchymal stem & cell therapy on the time course of pulmonary remodeling depend on the etiology of lung injury in mice. Crit. Care Med. 2013, 41, e319–e333. [Google Scholar]
  103. Shin, S.; Kim, Y.; Jeong, S.; Hong, S.; Kim, I.; Lee, W.; Choi, S. The therapeutic effect of human adult stem cells derived from adipose tissue in endotoxemic rat model. Int. J. Med. Sci. 2013, 10, 8–18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Zhang, S.; Danchuk, S.D.; Imhof, K.M.; A Semon, J.; A Scruggs, B.; Bonvillain, R.W.; Strong, A.L.; Gimble, J.M.; Betancourt, A.M.; E Sullivan, D.; et al. Comparison of the therapeutic effects of human and mouse adipose-derived stem cells in a murine model of lipopolysaccharide-induced acute lung injury. Stem Cell Res. Ther. 2013, 4, 13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Chang, Y.S.; Choi, S.J.; Ahn, S.Y.; Sung, D.K.; Sung, S.I.; Yoo, H.S.; Oh, W.I.; Park, W.S. Timing of umbilical cord blood derived mesenchymal stem cells transplantation determines therapeutic efficacy in the neonatal hyperoxic lung injury. PLoS ONE 2013, 8, e52419. [Google Scholar] [CrossRef] [Green Version]
  106. Ornellas, D.S.; Maron-Gutierrez, T.; Ornellas, F.M.; Cruz, F.F.; Oliveira, G.P.; Lucas, I.H.; Fujisaki, L.; Oliveira, M.G.; Teodoro, W.R.; Capelozzi, V.L.; et al. Early and late effects of bone marrow-derived mononuclear cell therapy on lung and distal organs in experimental sepsis. Respir. Physiol. Neurobiol. 2011, 178, 304–314. [Google Scholar] [CrossRef]
  107. Nandra, K.K.; Takahashi, K.; Collino, M.; Benetti, E.; Wong, W.S.F.; Goh, F.Y.; Suzuki, K.; Patel, N.S.A.; Thiemermann, C. Acute treatment with bone marrow-derived mononuclear cells attenuates the organ injury/dysfunction induced by hemorrhagic shock in the rat. Shock 2012, 37, 592–598. [Google Scholar] [CrossRef]
  108. Gorman, E.; Shankar-Hari, M.; Hopkins, P.; Tunnicliffe, W.S.; Perkins, G.D.; Silversides, J.; McGuigan, P.; Krasnodembskaya, A.; Jackson, C.; Boyle, R.; et al. Repair of acute respiratory distress syndrome by stromal cell administration (REALIST) trial: A phase 1 trial. EClinicalMedicine 2021, 41, 101167. [Google Scholar] [CrossRef] [PubMed]
  109. Adas, G.; Cukurova, Z.; Yasar, K.K.; Yilmaz, R.; Isiksacan, N.; Kasapoglu, P.; Yesilbag, Z.; Koyuncu, I.; Karaoz, E. The Systematic Effect of Mesenchymal Stem Cell Therapy in Critical COVID-19 Patients: A Prospective Double Controlled Trial. Cell Transplant. 2021, 30, 9636897211024942. [Google Scholar] [CrossRef]
  110. Chen, J.; Hu, C.; Chen, L.; Tang, L.; Zhu, Y.; Xu, X.; Chen, L.; Gao, H.; Lu, X.; Yu, L.; et al. Clinical Study of Mesenchymal Stem Cell Treatment for Acute Respiratory Distress Syndrome Induced by Epidemic Influenza A (H7N9) Infection: A Hint for COVID-19 Treatment. Engineering 2020, 6, 1153–1161. [Google Scholar] [CrossRef]
  111. Yudhawati, R.; Amin, M.; Rantam, F.A.; Prasetya, R.R.; Dewantari, J.R.; Nastri, A.M.; Poetranto, E.D.; Wulandari, L.; Lusida, M.I.; Koesnowidagdo, S.; et al. Bone marrow-derived mesenchymal stem cells attenuate plmonary inflammation and lung damage caused by highly pathofenic avian influenza A/H5N1 virus in BALB/c mice. BMC Infect. Dis. 2020, 20, 823–838. [Google Scholar] [CrossRef]
  112. Bogatcheva, N.C.; Coleman, M.E. Concentrated Scretome of Adipose Stromal Cells Limits Influenza A Virus-Induced Lung Injury in Mice. Cells 2021, 10, 720. [Google Scholar] [CrossRef]
  113. Khatri, M.; Richardson, L.A.; Meulia, T. Mesenchymal stem cell-derived extracellular vesicles attenuate influenza virus-induced acute lung injury in a pig model. Stem Cell Res. Ther. 2018, 9, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Li, Y.; Xu, J.; Shi, W.; Chen, C.; Shao, Y.; Zhu, L.; Lu, W.; Han, X. Mesenchymal stromal cell treatment prevents H9N2 avian influenza virus-induced acute lung injury in mice. Stem Cell Res. Ther. 2016, 7, 159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Loy, H.; Kuok, D.I.; Hui, K.P.; Choi, M.H.; Yuen, W.; Nicholls, J.M.; Peiris, J.M.; Chan, M.C. Therapeutic Implications of Human Umbilical Cord Mesenchymal Stromal Cells in Attenuating Influenza A(H5N1) Virus–Associated Acute Lung Injury. J. Infect. Dis. 2019, 219, 189–196. [Google Scholar] [CrossRef]
  116. Mallis, P.; Michalopoulos, E.; Chatzistamatiou, T.; Giokas, C.S. Interplay between mesenchymal stromal cells and immune system: Clinical applications in immune-related diseases. Explor. Immunol. 2021, 1, 112–139. [Google Scholar] [CrossRef]
  117. Kota, D.J.; Prabhakara, K.S.; Toledano-Furman, N.; Bhattarai, D.; Chen, Q.; DiCarlo, B.; Smith, P.; Triolo, F.; Wenzel, P.L.; Cox, C.S.; et al. Prostaglandin E2 Indicates Therapeutic Efficacy of Mesenchymal Stem Cells in Experimental Traumatic Brain Injury. Stem Cells 2017, 35, 1416–1430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Li, D.; Han, Y.; Zhuang, Y.; Fu, J.; Liu, H.; Shi, Q.; Ju, X. Overexpression of COX-2 but not indoleamine 2,3-dioxygenase-1 enhances the immunosuppressive ability of human umbilical cord-derived mesenchymal stem cells. Int. J. Mol. Med. 2015, 35, 1309–1316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Yañez, R.; Oviedo, A.; Aldea, M.; Bueren, J.A.; Lamana, M.L. Prostaglandin E2 plays a key role in the immunosuppressive properties of adipose and bone marrow tissue-derived mesenchymal stromal cells. Exp. Cell Res. 2010, 316, 3109–3123. [Google Scholar] [CrossRef]
  120. Legler, D.F.; Bruckne, M.; Uetz-von Allmen, E.; Krause, P. Prostaglandin E2 at new glance: Novel insights in functional diversity offer therapeutic chances. Int. J. Biochem. Cell Biol. 2010, 42, 198–201. [Google Scholar] [CrossRef] [Green Version]
  121. Lu, X.; Han, J.; Xu, X.; Xu, J.; Liu, L.; Huang, Y.; Yang, Y.; Qiu, H. PGE2 Promotes the migration of mesenchymal stem cells through the activation of FAK and ERK1/2 pathway. Stem Cells Int. 2017, 2017, 8178643. [Google Scholar] [CrossRef] [Green Version]
  122. Jiang, J.; Dingledine, R. Role of prostaglandin receptor EP2 in the regulations of cancer cell proliferation, invasion, and inflammation. J. Pharmacol. Exp. Ther. 2013, 344, 360–367. [Google Scholar] [CrossRef] [Green Version]
  123. Lee, B.C.; Kim, H.S.; Shin, T.H.; Kang, I.; Lee, J.Y.; Kim, J.-J.; Kang, H.K.; Seo, Y.; Lee, S.; Yu, K.-R.; et al. PGE2 maintains self-renewal of human adult stem cells via EP2-mediated autocrine signaling and its production is regulated by cell-to-cell contact. Sci. Rep. 2016, 6, 26298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Vancheri, C.; Mastruzzo, C.; Sortino, M.A.; Crimi, N. The lung as a privileged site for the beneficial actions of PGE2. Trends Immunol. 2004, 25, 40–46. [Google Scholar] [CrossRef]
  125. Konya, V.; Maric, J.; Jandl, K.; Luschnig, P.; Aringer, I.; Lanz, I.; Platzer, W.; Theiler, A.; Bärnthaler, T.; Frei, R.; et al. Activation of EP4 receptors prevents endotoxin-induced neutrophil infiltration into the airways and enhances microvascular barrier function. Br. J. Pharmacol. 2015, 172, 4454–4468. [Google Scholar] [CrossRef] [Green Version]
  126. Fujino, H.; West, K.A.; Regan, J.W. Phosphorylation of glycogen synthase kinase-3 and stimulation of T-cell factor signaling following activation of EP2 and EP4 prostanoid receptors by prostaglandin E2. J. Biol. Chem. 2002, 277, 2614–2619. [Google Scholar] [CrossRef] [Green Version]
  127. Castellone, M.D.; De Falco, V.; Rao, D.M.; Bellelli, R.; Muthu, M.; Basolo, F.; Fusco, A.; Gutkind, J.S.; Santoro, M. The β catenin axis integrates multiple signals downstrem foe RET/Papillary thyroid carcinoma leading to cell proliferation. Cancer Res. 2009, 69, 1867–1876. [Google Scholar] [CrossRef] [Green Version]
  128. Shevtsov, S.P.; Haq, S.; Force, T. Activation of β-catenin Signaling Pathways by Classical G-Protein-Coupled Receptors: Mechanisms and Consequences in Cycling and Non-cycling Cells. Cell Cycle 2006, 5, 2295–2300. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Zheng, C.; Qu, Y.X.; Wang, B.; Shen, P.F.; Xu, J.D.; Chen, Y.X. COX-2/PGE2 facilitates fracture healing by activating the Wnt/β-catenin signaling pathway. Eur. Rev. Med. Pharmalogical Sci. 2019, 23, 9721–9728. [Google Scholar]
  130. Du, Q.; Geller, D.A. Cross-Regulation between Wnt and NF-β B Signaling Pathways. Immunopathol. Dis. Ther. 2010, 1, 151–181. [Google Scholar]
  131. Deng, J.; Miller, S.A.; Wang, H.Y.; Xia, W.; Wen, Y.; Zhou, B.P.; Li, Y.; Lin, S.Y.; Hung, M.C. Beta-catenin interacts with and inhibits NF-kappa B in human colon and breast cancer. Cancer cell 2002, 2, 323–334. [Google Scholar] [CrossRef] [Green Version]
  132. Sun, J.; Robert, M.E.; Duan, Y.; Rao, A.S.; He, T.C.; Chang, E.B.; Madara, J.L. Crosstalk between NFkB and β catenin pathways in bacterial colonized intestinal epithelial cell. Am. J. Physiol. Gastrointest. Liver Physiol. 2005, 289, G129–G137. [Google Scholar] [CrossRef] [Green Version]
  133. Liu, X.; Lu, R.; Wu, S.; Zhang, Y.-G.; Xia, Y.; Sartor, B.R.; Sun, J. Wnt2 inhibits enteric bacterial induce inflammation in intestinal epithelial cell. Inflamm. Bowel Dis. 2012, 18, 418–429. [Google Scholar] [CrossRef] [PubMed]
  134. Silva-García, O.; Valdez-Alarcón, J.J.; Baizabal-Aguirre, V.M. The Wnt/β-Catenin Signaling Pathway Controls the Inflammatory Response in Infections Caused by Pathogenic Bacteria. Mediat. Inflamm. 2014, 2014, 310183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Herschman, H.R. Prostaglandin synthase 2. Biochim. Biophys. Acta BBA Lipids Lipid Metab. 1996, 1299, 125–140. [Google Scholar] [CrossRef]
  136. Hinz, B.; Brune, K. Cyclooxygenase-2–10 years later. J. Pharmacol. Exp. Ther. 2002, 300, 367–375. [Google Scholar] [CrossRef]
  137. MacDonald, B.T.; Tamai, K.; He, X. Wnt/beta-catenin signaling: Components, mechanisms, and diseases. Dev. Cell 2009, 17, 9–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Suryawanshi, A.; Tadagavadi, R.K.; Swafford, D.; Manicassamy, S. Modulation of Inflammatory Responses by Wnt/β-Catenin Signaling in Dendritic Cells: A Novel Immunotherapy Target for Autoimmunity and Cancer. Front. Immunol. 2016, 7, 460. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. de Winter, T.J.J.; Nusse, R. Running Against the Wnt: How Wnt/β-Catenin Suppresses Adipogenesis. Front. Cell Dev. Biol. 2021, 9, 627429. [Google Scholar] [CrossRef] [PubMed]
  140. Gehart, H.; Clevers, H. Tales fromthe crypt: New insights into intestinal stem cells. Nat. Rev. 2019, 16, 19–34. [Google Scholar] [CrossRef]
  141. Silva-Garcia, O.; Valdez-Alarcόn, J.J.; Baizabal-Aguirre, V.M. Wnt/β-Catenin Signaling as a Molecular Target by Pathogenic Bacteria. Front. Immunol. 2019, 10, 2135. [Google Scholar] [CrossRef] [Green Version]
  142. Pokutta, S.; Weis, W.I. Structure of thr dimerization and β—Catenin binding region of α catenin. Mol. Cell 2000, 5, 533–543. [Google Scholar] [CrossRef]
  143. Fiedler, M.; Mendoza-Topaz, C.; Rutherford, T.J.; Mieszczanek, J.; Bienz, M. Dishevelled interacts with the DIX domain polymerization interface of Axin to interfere with its function in down-regulating β-catenin. Proc. Natl. Acad. Sci. USA 2011, 108, 1937–1942. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Metcalfe, C.; Bienz, M. Inhibition of GSK3 by Wnt signalling—Two contrasting models. J. Cell Sci. 2011, 124, 3537–3544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Etheridge, S.L.; Spencer, G.J.; Heath, D.J.; Genever, P.G. Expression Profiling and Functional Analysis of Wnt Signaling Mechanisms in Mesenchymal Stem Cells. Stem Cells 2014, 22, 849–860. [Google Scholar] [CrossRef]
  146. Bärnthaler, T.; Maric, J.; Platzer, W.; Konya, V.; Theiler, A.; Hasenöhrl, C.; Gottschalk, B.; Trautmann, S.; Schreiber, Y.; Graier, W.F.; et al. The Role of PGE2 in Alveolar Epithelial and Lung Microvascular Endothelial Crosstalk. Sci. Rep. 2017, 7, 7923. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Fang, X.; Yu, S.X.; Lu, Y.; Bast, R.C.; Woodgett, J.R.; Mills, G.B. Phosphorylation and inactivation of glycogen synthase kinase 3 by protein kinase A. Proc. Natl. Acad. Sci. USA 2000, 97, 11960–11965. [Google Scholar] [CrossRef] [Green Version]
  148. Shao, J.; Jung, C.; Liu, C.; Sheng, H. Prostaglandin E2 Stimulates the B-Catenin/T Cell Factor-Dependent Transcription in Colon Cancer. J. Biol. Chem. 2005, 280, 26565–26572. [Google Scholar] [CrossRef] [Green Version]
  149. Fang, L.; Chang, H.-M.; Cheng, J.-C.; Leung, P.C.K.; Sun, Y.-P. TGF-β1 induces COX-2 expression and pge2 production in human granulosa cells through smad signaling pathways. J. Clin. Endocrinol. Metab. 2014, 99, E1217–E1226. [Google Scholar] [CrossRef] [Green Version]
  150. Flozak, A.S.; Lam, A.P.; Russell, S.; Jain, M.; Peled, O.N.; Sheppard, K.A.; Beri, R.; Mutlu, G.M.; Budinger, G.S.; Gottardi, C.J. Beta-catenin/T-cell factor signaling is activated during lung injury and promotes the survival and migration of alveolar epithelial cells. J. Biol. Chem. 2010, 285, 3157–3167. [Google Scholar] [CrossRef] [Green Version]
  151. Mutze, K.; Vierkotten, S.; Milosevic, J.; Eickelberg, O.; Königshoff, M. Enolase 1 (ENO1) and protein disulfide-isomerase associated 3 (PDIA3) regulate Wnt/β-catenin-driven transdifferentiation of murine alveolar epithelial cells. Dis. Model. Mech. 2015, 8, 877–890. [Google Scholar]
  152. Abraham, E.; Arcaroli, J.; Carmody, A.; Wang, H.; Tracey, K.J. HMG-1 as a mediator of acute lung inflammation. J. Immunol. 2000, 165, 2950–2954. [Google Scholar] [CrossRef] [Green Version]
  153. Xie, J.; Méndez, J.D.; Méndez-Valenzuela, V.; Hernandez, M.M.A. Cellular signalling of the receptor for advanced glycation end products (RAGE). Cell. Signal. 2013, 25, 2185–2197. [Google Scholar] [CrossRef] [PubMed]
  154. Mukhopadhyay, S.; Hoidal, J.R.; Tapan, K. Role of TNFα in pulmonary pathophysiology. Respir. Res. 2006, 7, 125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Sparvero, L.J.; Asafu-Adjei, D.; Kang, R.; Tang, D.; Amin, N.; Im, J.; Rutledge, R.; Lin, B.; A Amoscato, A.; Zeh, H.J.; et al. RAGE (Receptor for Advanced Glycation Endproducts), RAGE Ligands, and their Role in Cancer and Inflammation. J. Transl. Med. 2009, 7, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Cytokine Storm and ALI/ARDS Induced by Influenza Virus Infection. When a highly pathogenic influenza A virus infects alveolar epithelial cells, macrophages recognize this virus using TLR 7, which involves several adaptor molecules and transcription factors, such as MyD88, and activates IRF7 and NF-κB. Influenza A virus can also be recognized by RIG 1 via the adaptor molecule IPD-I and activate IRF3 and NF-κB. The activation of IRF-7 and IRF-3 causes their translocation into the nucleus. This produces interferon type I, while NF-κB acts as a transcription factor to induce pro-inflammatory cytokines’ production, including IL-6, TNFα, and pro-IL-1β. Macrophages also present virus peptides on the cell surface through MHC class II and activate lymphocyte CD4 T cells, which differentiate into Th1. Activated lymphocyte T cells then release various pro-inflammatory cytokines and chemoattractants. All of these mechanisms result in a cytokine storm. The cytokine storm then leads to ARDS, characterized by necrosis, tissue damage, an influx of leucocytes, and blood vessel dilation, causing edema fluids to accumulate in the alveolar, characterized by tissue hypoxia and hypoxemia. The damaged tissue releases HMGB1 and binds with RAGE, activating the intracellular signaling pathway and resulting in NF-κB activation and pro-inflammatory gene expression induction; thus, the cytokine storm continues. We created this figure using the BioRender online app and license.
Figure 1. Cytokine Storm and ALI/ARDS Induced by Influenza Virus Infection. When a highly pathogenic influenza A virus infects alveolar epithelial cells, macrophages recognize this virus using TLR 7, which involves several adaptor molecules and transcription factors, such as MyD88, and activates IRF7 and NF-κB. Influenza A virus can also be recognized by RIG 1 via the adaptor molecule IPD-I and activate IRF3 and NF-κB. The activation of IRF-7 and IRF-3 causes their translocation into the nucleus. This produces interferon type I, while NF-κB acts as a transcription factor to induce pro-inflammatory cytokines’ production, including IL-6, TNFα, and pro-IL-1β. Macrophages also present virus peptides on the cell surface through MHC class II and activate lymphocyte CD4 T cells, which differentiate into Th1. Activated lymphocyte T cells then release various pro-inflammatory cytokines and chemoattractants. All of these mechanisms result in a cytokine storm. The cytokine storm then leads to ARDS, characterized by necrosis, tissue damage, an influx of leucocytes, and blood vessel dilation, causing edema fluids to accumulate in the alveolar, characterized by tissue hypoxia and hypoxemia. The damaged tissue releases HMGB1 and binds with RAGE, activating the intracellular signaling pathway and resulting in NF-κB activation and pro-inflammatory gene expression induction; thus, the cytokine storm continues. We created this figure using the BioRender online app and license.
Ijms 24 07299 g001
Figure 2. Communication of MSCs in Damaged Tissue and the Immunosuppression Property of MSCs. Tissue injury, inflammation, and hypoxia factors cue a signal that mobilizes MSCs to the damaged tissue. Once MSCs enter the microenvironment of the injured tissue, IFNγ, together with pro-inflammatory cytokines, such as TNFα, IL-1α, and IL-1β, the occurrence of a burst of chemokine, and the expression of adhesion molecules, stimulate MSCs to release large amounts of immunosuppressive factors. These immunosuppressive factors then promote the activation of immune cells, which induce regulatory cells and inhibit cytokine production. This figure was created using the BioRender online app and license.
Figure 2. Communication of MSCs in Damaged Tissue and the Immunosuppression Property of MSCs. Tissue injury, inflammation, and hypoxia factors cue a signal that mobilizes MSCs to the damaged tissue. Once MSCs enter the microenvironment of the injured tissue, IFNγ, together with pro-inflammatory cytokines, such as TNFα, IL-1α, and IL-1β, the occurrence of a burst of chemokine, and the expression of adhesion molecules, stimulate MSCs to release large amounts of immunosuppressive factors. These immunosuppressive factors then promote the activation of immune cells, which induce regulatory cells and inhibit cytokine production. This figure was created using the BioRender online app and license.
Ijms 24 07299 g002
Figure 3. PGE2 Promotes Immunoregulation through Cross-Regulation between β-catenin and NF-κB by Activating the Wnt-β-catenin Signaling Pathways. (A) Without the Wnt ligand, β-catenin is bound by GSK-3β, Axin, and APC. This binding-stimulated phosphorylation of β-catenin by GSK-3β leads to the ubiquitination of β-catenin, which is further degraded by proteasomes. (B) When the Wnt ligand binds to the Fz receptor and the LRP co-receptor, activation of the canonical Wnt signal occurs, followed by activation of Dvl and inhibition of GSK-3β. To regulate gene transcription, β-catenin accumulates in the nucleus and binds to T cell factor and lymphoid enhancer factors (TCF/LEF). (C) PGE2 stimulates a double signaling cascade through the EP2 receptor via PI3K and protein kinase Akt activation involving the free G-protein βγ subunit and the direct association of the G-protein αs subunit to Axin. Following PGE2 activation in these two pathways, unphosphorylated β-catenin accumulates in the cytoplasm, then enters the nucleus and inhibits the transcription factor of NF-κB (D). The activity of β-catenin can inhibit NF-κB via two possible mechanisms—by stabilizing IκBα or by binding to NF-κB subunit p50—which prevents its transport from the cytoplasm to the nucleus and also occurs in the nucleus. Both mechanisms can cause a decrease in the transcriptional activity of NF-κB. The inhibition of NF-κB by β-catenin activation reduces inflammation. This figure was created using the BioRender online app and license.
Figure 3. PGE2 Promotes Immunoregulation through Cross-Regulation between β-catenin and NF-κB by Activating the Wnt-β-catenin Signaling Pathways. (A) Without the Wnt ligand, β-catenin is bound by GSK-3β, Axin, and APC. This binding-stimulated phosphorylation of β-catenin by GSK-3β leads to the ubiquitination of β-catenin, which is further degraded by proteasomes. (B) When the Wnt ligand binds to the Fz receptor and the LRP co-receptor, activation of the canonical Wnt signal occurs, followed by activation of Dvl and inhibition of GSK-3β. To regulate gene transcription, β-catenin accumulates in the nucleus and binds to T cell factor and lymphoid enhancer factors (TCF/LEF). (C) PGE2 stimulates a double signaling cascade through the EP2 receptor via PI3K and protein kinase Akt activation involving the free G-protein βγ subunit and the direct association of the G-protein αs subunit to Axin. Following PGE2 activation in these two pathways, unphosphorylated β-catenin accumulates in the cytoplasm, then enters the nucleus and inhibits the transcription factor of NF-κB (D). The activity of β-catenin can inhibit NF-κB via two possible mechanisms—by stabilizing IκBα or by binding to NF-κB subunit p50—which prevents its transport from the cytoplasm to the nucleus and also occurs in the nucleus. Both mechanisms can cause a decrease in the transcriptional activity of NF-κB. The inhibition of NF-κB by β-catenin activation reduces inflammation. This figure was created using the BioRender online app and license.
Ijms 24 07299 g003
Figure 4. Immunoregulation Mechanisms of Exogenous MSCs on Virus-Induced Acute Lung Injury. A single-stranded RNA of the highly pathogenic influenza A virus binding with the host initiates several signaling transductions. IKKβ is then activated, which can lead to the phosphorylation and degradation of Iκβ, causing the active transcription factor NF-κB to release. NF-κB encodes pro-inflammatory cytokines and chemokine attractants, which lead to ARDS. The injured cells release HMGB1, which has a high affinity to RAGE ligands. HMGB1 bonds with RAGE, and this cues a signal to activate NF-κB, which releases adhesion molecules and pro-inflammatory cytokines. CXCL12 (SDF-1) regulation increases in the inflammation niche due to pro-inflammatory cytokine stimulation. CXCL12 (SDF-1) interacts with its ligand CXCR4, which is expressed by exogenous MSCs and starts the homing process. Active biomolecules of endogenous PGE2 are then produced in large amounts, which leads to the phosphorylation of GSK-3 such that β-catenin accumulates in the cytoplasm. These accumulations inhibit NF-κB via the activation of Iκβ, and as a consequence, the release of pro-inflammatory cytokines and chemokines is inhibited. This figure was created using the BioRender online app and license.
Figure 4. Immunoregulation Mechanisms of Exogenous MSCs on Virus-Induced Acute Lung Injury. A single-stranded RNA of the highly pathogenic influenza A virus binding with the host initiates several signaling transductions. IKKβ is then activated, which can lead to the phosphorylation and degradation of Iκβ, causing the active transcription factor NF-κB to release. NF-κB encodes pro-inflammatory cytokines and chemokine attractants, which lead to ARDS. The injured cells release HMGB1, which has a high affinity to RAGE ligands. HMGB1 bonds with RAGE, and this cues a signal to activate NF-κB, which releases adhesion molecules and pro-inflammatory cytokines. CXCL12 (SDF-1) regulation increases in the inflammation niche due to pro-inflammatory cytokine stimulation. CXCL12 (SDF-1) interacts with its ligand CXCR4, which is expressed by exogenous MSCs and starts the homing process. Active biomolecules of endogenous PGE2 are then produced in large amounts, which leads to the phosphorylation of GSK-3 such that β-catenin accumulates in the cytoplasm. These accumulations inhibit NF-κB via the activation of Iκβ, and as a consequence, the release of pro-inflammatory cytokines and chemokines is inhibited. This figure was created using the BioRender online app and license.
Ijms 24 07299 g004
Table 1. The study of MSCs in lung injury induced by different sub-types of influenza virus.
Table 1. The study of MSCs in lung injury induced by different sub-types of influenza virus.
NoAuthorVirusMSCS SourcesOutcome of InfectionStudy DesignReferences
1Chen, J., et al. (2020)Influenza A (H7N9)Allogeneic, menstrual-blood-derived MSCsLower levels of PCT, serum creatinin (sCr), creatine
kinase (CK), prothrombin time (PT), and D-dimer; hemoglobin (Hb) up-regulated; improvement on CCT
Human[110]
2Yudhawati, R., et al. (2020)Avian influenza A/H5N1 of A/turkey/East Java/Av154/2013Bone-marrow-derived mesenchymal stem cells (BM-MSCs)Lower levels of lung alveolar protein, PaO2/FiO2 ratio, and histopathological score; suppressed expressions of NF-κB, RAGE, TNFα, and IL-1β; Sftpc and Aqp5+ enhancedIn vivo (mice)[111]
3Bogatcheva and Coleman (2021)Influenza A Virus H1N1Adipose stromal cellsLimits pulmonary histopathological changes, infiltration of inflammatory cells, protein leak, water accumulation, and arterial oxygen saturation (SpO2) reduction; significant suppression of IL-6
and MCP-1; viral antigen, BAL TNF-α, PDL1, and Angpt2 levels lowered
In vivo (mice)[112]
4Chan et al. (2016)H5N1 influenza A viruses A/Hong Kong/483/97 (483/97), A/Hong Kong/486/97 (486/97), and A/Vietnam/3046/04 (3046/04)Bone-marrow-derived human MSCsTransporter proteins CFTR and α1Na,K-ATPase enhanced; Ang1 in MSCS supernatants, KGF secretion, CFTR protein expression increased; lower CD4+ T cells, NK cells, IP10, MCP-1, MCP-3, M1P-1α, RANTES, IL-4, IL-17, and TNF-α; more macrophages/monocytesIn vitro and in vivo[27]
5Khatri et al. (2018)Influenza virus H1N1, H3N2, H9N5Isolated extracellular vesicles EVs from swine bone-marrow-derived MSCsReduced influenza virus replication; lower levels of TNF-α and CXCL10; cytokine IL-10 were higherIn vivo (pigs)[113]
6Li, Y., et al. (2016)A/Hong Kong/2108/2003 [H9N2 (HK)] H9N2 virusAdipocyte, chondrocytes, and osteocytes murine MSCsHigher PaO2, SaO2 and pH values; PaCO2, GM-CSF, MCP-1, KC, MIP-1α, MIG, IL-1α, IL-6, IL-10, TNF-α, IFN-γ, ERK, JNK, CD14, and TLR4 expression were loweredIn vivo (mice)[114]
7Loy, H., et al. (2019)Influenza A/Hong Kong/483/97(H5N1)Human umbilical cord MSCsIn vitro: reduced IL-β, IFN-λ1, IL-6, IL-1β, IFN-γ-induced protein 10 (IP-10), MCP-1, and RANTES; enhances responses of IL-4, IL-10, IL-11, IL-13, IL-RA, IL-10, IL-11, and IL-RA.
In vivo: greatest significance for IP-10, MCP-1, RANTES, and IL-6; lower concentrations of Evans blue dye, IP-10, RANTES, TNF-α, MCP-1, IL-1β, IL-6, and IL-8
In vitro and in vivo (mice)[115]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Yudhawati, R.; Shimizu, K. PGE2 Produced by Exogenous MSCs Promotes Immunoregulation in ARDS Induced by Highly Pathogenic Influenza A through Activation of the Wnt-β-Catenin Signaling Pathway. Int. J. Mol. Sci. 2023, 24, 7299. https://doi.org/10.3390/ijms24087299

AMA Style

Yudhawati R, Shimizu K. PGE2 Produced by Exogenous MSCs Promotes Immunoregulation in ARDS Induced by Highly Pathogenic Influenza A through Activation of the Wnt-β-Catenin Signaling Pathway. International Journal of Molecular Sciences. 2023; 24(8):7299. https://doi.org/10.3390/ijms24087299

Chicago/Turabian Style

Yudhawati, Resti, and Kazufumi Shimizu. 2023. "PGE2 Produced by Exogenous MSCs Promotes Immunoregulation in ARDS Induced by Highly Pathogenic Influenza A through Activation of the Wnt-β-Catenin Signaling Pathway" International Journal of Molecular Sciences 24, no. 8: 7299. https://doi.org/10.3390/ijms24087299

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