Next Article in Journal
G Protein-Coupled Receptor–Ligand Pose and Functional Class Prediction
Previous Article in Journal
LPA-Induced Thromboxane A2-Mediated Vasoconstriction Is Limited to Poly-Unsaturated Molecular Species in Mouse Aortas
Previous Article in Special Issue
Topology and Dynamics of Transcriptome (Dys)Regulation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Interferon Regulatory Factors in Liver Diseases

Department of Gastroenterology, Renmin Hospital of Wuhan University, No. 99 Zhang Zhidong Road, Wuhan 430060, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(13), 6874; https://doi.org/10.3390/ijms25136874
Submission received: 7 May 2024 / Revised: 12 June 2024 / Accepted: 20 June 2024 / Published: 22 June 2024

Abstract

:
The interferon regulatory factors (IRFs) family comprises 11 members that are involved in various biological processes such as antiviral defense, cell proliferation regulation, differentiation, and apoptosis. Recent studies have highlighted the roles of IRF1-9 in a range of liver diseases, including hepatic ischemia–reperfusion injury (IRI), alcohol-induced liver injury, Con A-induced liver injury, nonalcoholic fatty liver disease (NAFLD), cirrhosis, and hepatocellular carcinoma (HCC). IRF1 is involved in the progression of hepatic IRI through signaling pathways such as PIAS1/NFATc1/HDAC1/IRF1/p38 MAPK and IRF1/JNK. The regulation of downstream IL-12, IL-15, p21, p38, HMGB1, JNK, Beclin1, β-catenin, caspase 3, caspase 8, IFN-γ, IFN-β and other genes are involved in the progression of hepatic IRI, and in the development of HCC through the regulation of PD-L1, IL-6, IL-8, CXCL1, CXCL10, and CXCR3. In addition, IRF3-PPP2R1B and IRF4-FSTL1-DIP2A/CD14 pathways are involved in the development of NAFLD. Other members of the IRF family also play moderately important functions in different liver diseases. Therefore, given the significance of IRFs in liver diseases and the lack of a comprehensive compilation of their molecular mechanisms in different liver diseases, this review is dedicated to exploring the molecular mechanisms of IRFs in various liver diseases.

1. Introduction

Interferon regulatory factors (IRFs) are a family of transcription factors that play crucial roles in various aspects of the immune response, including the development and differentiation of immune cells, as well as the regulation of responses to pathogens [1]. IRF1, the first identified IRF that induced type I interferons, exhibited strong antiviral activity against a broad spectrum of viral infections, both in an interferon-dependent and interferon-independent manner [2]. IRF2 was linked to the development of various cancers like colorectal, pancreatic, and hepatocellular carcinoma (HCC), by controlling the transcription of genes such as TP53, CXCL3, Bcl-2, and Bax [3,4,5]. IRF3, IRF5, and IRF7 were essential for the production of type I interferons (IFNs) upon pathogen recognition by receptors, impacting viral and bacterial infections, inflammatory responses and autoimmune diseases, cancer growth, and metastasis, and changes in the tumor microenvironment [6,7,8,9]. IRF4 and IRF8 contributed to tumor immunity by modulating the functions of T cells, B cells, and NK cells [10,11]. IRF6, with its unique helix-turn-helix DNA-binding motif, primarily governs processes in limb and stomatofacial development, with limited documentation on its role in regulating IFNs in higher vertebrates [12]. IRF9 regulated the expression of interferon-stimulated genes (ISGs), which served as markers for type I and III interferon activation [13,14]. IRF10 was only observed in fish and birds, while IRF11 seemed to be a fish-specific member of the IRF family and will not be discussed in this review [15].
Recent research has shown that IRFs play a crucial role in the advancement and exacerbation of various liver diseases, such as hepatic ischemia-reperfusion injury (IRI), alcohol-induced liver injury, Con A-induced liver injury, nonalcoholic fatty liver disease (NALFD), cirrhosis, and HCC. IRF1, a nuclear transcription factor, was involved in various liver diseases, particularly hepatic IRI and HCC. It played a crucial role in regulating gene expression during inflammation and was up-regulated in both cold and warm liver IRI. IRF1 exerted deleterious effects by inducing the expression of inflammatory mediators, but liver grafts deficient in IRF1 showed significantly reduced liver injury [16,17]. Specifically, in the early post-transplantation period, IRF1 overexpression in donor livers led to elevated caspase 3 expression, whereas knockdown of IRF1 reduced caspase 8 activity, resulting in a significant reduction in hepatocyte apoptosis and liver injury. In addition, IRF1 was involved in the hepatic IRI process through the regulation of various immune cell activities (NK and T cells), cytokines (IL-6, IL-12, IL-15, IL-23), and death ligands, as well as oxidative stress [18,19,20,21]. IRF1 also served as a key transcription factor that controlled gene expression in both innate and adaptive immunity, responding to type I (IFN-α/β) and type II (IFN-γ) interferons [22,23]. It was involved in IFN-γ-induced apoptosis of tumor cells by triggering downstream signals like inducible nitric oxide synthase (iNOS), p21, p53, p53-upregulated modulator of apoptosis (PUMA), and Fas-related death domain [24,25,26,27,28]. Yan, Y. et al. showed that IRF1 promotes the migration of CD8+ T cells and NK and NKT cells and stimulates IFN-γ secretion in NK and NKT cells, ultimately leading to tumor cell apoptosis via the CXCL10/CXCR3 pathway [29]. However, Wang, R. et al. found that gamma interferons can upregulate IRF1 expression in HCC cells, which in turn activates human endogenous retrovirus-H long terminal repeat-associating 2 (HHLA2) expression and promotes M2 polarization and macrophage chemotaxis, potentially facilitating immune evasion and HCC progression [30]. IRF1 may have a dual role in HCC, which requires further experimental verification. In addition to IRF1, other members of the IRF family have also been discovered to play significant roles in various liver diseases, demonstrating diverse functions in either promoting or inhibiting these diseases. Consequently, a comprehensive understanding of the role of IRFs in liver disease can offer valuable insights into disease mechanisms and potential therapeutic targets.

Materials and Methods

A systematic search was conducted in the PubMed electronic database to gather relevant literature. Additionally, the reference lists of the primary studies included were reviewed to identify potentially eligible articles. Only articles published in English were considered, with no restrictions on publication year. The index terms encompassed various topics such as ‘interferon regulatory factor’, ‘IRF’, ‘ischemia reperfusion injury’, ‘hepatic ischemia reperfusion injury’, ‘liver injury’, ‘hepatic injury’, ‘NAFLD’, ‘nonalcoholic fatty liver disease’, ‘cirrhosis’, ‘liver fibrosis’, ‘HCC’, and ‘hepatocellular carcinoma’. A critical evaluation was performed on all studies included in this paper.

2. The Structure and Function of IRFs

The eleven members of the IRF family, IRF1 to IRF11, have been identified, with their primary roles centered around transcriptional regulation in the immune system and cell growth [31,32,33]. All IRFs share common domains, including an N-terminal helical DNA-binding domain (DBD) with five conserved tryptophan repeats, and a C-terminal IRF-associated domain (IAD) that facilitates protein interactions and modulates various transcriptional activities [31,34,35]. Each member of the IRF family plays a distinct role in various biological processes, including pathogen response, cytokine signaling, cell growth regulation, hematopoietic development, immune cell development, differentiation, and apoptosis [36]. In-depth study of the functions of IRFs is crucial for comprehending the mechanisms underlying disease occurrence and progression.
IRF1, a pleiotropic transcription factor, exhibited high responsiveness to various stimuli such as IFN, NF-κB, TBK-1, and IKK-ɛ, undergoing rapid and dynamic regulation upon infection [37,38,39,40]. Despite the short life cycle of IRF1 mRNA and protein, it consistently expressed a variety of host defense genes and effectively drove the expression of genes related to innate immunity [31,41,42,43]. Type I/II IFN stimulated hepatocytes to release IL-7, which in turn limited IRF1, enhanced the secretion of pro-inflammatory cytokines induced by lipopolysaccharide (LPS), and reduced macrophage tolerance to LPS [44]. Similar to IRF1, IRF2 competed for the same cis-elements and inhibited the function of IRF1, thereby reducing the production of IRF1-dependent pro-inflammatory genes such as IL-12, IFNβ, and iNOS, which ultimately helped in reducing hepatic IRI [45]. IRF3, a crucial component of the innate immune response, was responsible for detecting and reacting to foreign antigens, thereby helping to prevent viral infections [46]. Besides its role in gene transcription, IRF3 also initiated non-transcriptional and pro-apoptotic signaling pathways. Although the pro-apoptotic activity of IRF3 was beneficial in fighting viral infections, it could also lead to liver cell death and exacerbate detrimental immune responses in liver disease [47]. IRF4 was an oncogenic master transcription factor that induced cancer transcriptional programs by forming unique regulatory circuits that interacted with upstream pathways and binding partners [10]. The expression of IRF4 and PD-1 was positively correlated, and the overexpression of IRF4 was found to impede the proliferation and migration of HCC by suppressing the JAK2/STAT3 signaling pathway and epithelial-mesenchymal transition [48]. Furthermore, IRF4 served as a crucial transcriptional regulator of lipid processing in adipocytes and acted as a suppressor of inflammation in diet-induced obesity. Reduced IRF4 expression was associated with the exacerbation of NAFLD, leading to heightened levels of inflammation and insulin resistance [49]. IRF5 played a central role in inflammation and was recruited to promote inflammatory genes with the assistance of the NF-kB p65 sub-unit RelA, inducing pro-inflammatory cytokines such as IL-6, IL-12, IL-23, and TNF-a [50,51,52]. Highly expressed in monocytes, macrophages, B-cells, and dendritic cells, IRF5 was implicated in various inflammatory and autoimmune diseases [51,53,54,55]. Alzaid, F. et al. showed a significant increase in IRF5 expression in hepatic macrophages of individuals with non-alcoholic fatty liver disease or hepatitis C virus infection, contributing to liver fibrosis. Interestingly, mice lacking IRF5 in myeloid cells were found to be protected from stress-induced hepatic fibrosis, suggesting the crucial role of IRF5 in hepatocellular cell death and liver fibrosis in both mice and humans [56]. The tumor suppressor IRF6 was not implicated in IFN gene regulation and was notably decreased during epithelial-mesenchymal transition (EMT) in gastric and pancreatic ductal adenocarcinomas. This downregulation was controlled by the transcription factor ZEB1 [57,58]. In hepatocytes, IRF6 directly interacted with the promoter of the peroxisome proliferator-activated receptor γ (PPARγ) gene, leading to the inhibition of PPARγ transcription and its associated target genes involved in lipogenesis and fatty acid uptake [59]. Moreover, lower IRF6 expression was observed in colorectal cancer tissues and liver metastases compared to normal tissues. Mechanistically, IRF6 may enhance cisplatin-mediated cell proliferation, migration, invasion, and apoptosis sensitivity to chemotherapy by regulating E-calmodulin and Ki67 [60]. IRF7, initially recognized as a key transcription factor in the production of type I IFNs and regulation of the innate immune response, was situated on chromosome 11p15.5 in humans and chromosome 7 F5 in mice [7,61]. With a diverse array of functions, IRF7 was a pivotal component in the type I/III IFNs induced signaling pathway, contributing significantly to viral infections, autoimmune diseases, and the maintenance of homeostasis [7]. IRF8 played a crucial role in regulating immune cell differentiation and inducing innate immunity. It collaborated with IRF1, IRF2, IRF4, and other transcriptional regulators like TEL, PU.1, MIZ1, and E47 to modulate gene transcription by influencing the formation of DNA-binding compounds and controlling the expression of downstream target genes [62,63,64]. IRF8 knockout mice showed a reduction in inflammatory cell infiltration and cytokine release, leading to improved outcomes after liver IRI. Conversely, overexpression of IRF8 was linked to worsened liver damage and functional abnormalities [65]. Interestingly, in HCC cells, IRF8 overexpression demonstrated enhanced antitumor effects by potentially regulating tumor-associated macrophages and T cell levels in the tumor microenvironment [66]. IRF9 interacted with peroxisome proliferator-activated receptor α (PPARα) and activated PPARα target genes to attenuate inflammation, hepatic steatosis, and insulin resistance [67]. However, IRF9 also suppressed SIRT1 expression, resulting in increased p53 protein acetylation, ultimately exacerbating hepatic IRI [68]. These findings suggest that the role of IRFs may vary depending on the specific liver disease being studied.

3. IRFs and Liver Diseases

IRFs are a group of multifunctional transcription factors that target IFN promoters and interferon-stimulated response elements (ISREs) in ISGs, activating the expression of target genes. This indicates the essential role of IRFs in a range of biological processes including antiviral defense, innate immunity, adaptive immunomodulation, cell growth, apoptosis, and homeostasis maintenance [69]. Despite their known functions, the involvement of IRFs in the regulation of liver diseases has yet to be explored (Table 1).

3.1. IRFs and Liver Ischemia-Reperfusion Injury

IRI was a common complication of liver transplantation, partial hepatectomy, and hypovolemic shock. This phenomenon resulted in complex hepatocellular damage, early graft dysfunction, and an increased risk of acute and chronic rejection, ultimately resulting in poor prognosis and low patient survival rates [114,115]. Treatment for IRI was primarily supportive, as there were no specific drugs or methods available. Various strategies have been explored to address liver IRI, such as ischemic preconditioning, pharmacological and surgical interventions, and gene therapy [116]. Despite advancements in graft management targeting autophagy, oxidative stress, sterile inflammation, and apoptosis, the intricate pathological mechanisms of IRI remained inadequately understood.

3.1.1. Autophagy and Oxidative Stress

IRF1 and IRF5 expression were found to be significantly increased in the liver following exposure to IRI [75,81]. IRF1 had been shown to have a detrimental impact on hepatic IRI by regulating the expression of various inflammatory mediators. Overexpression of IRF1 in donor livers led to elevated expression of caspase 3 during the early post-transplantation period. Conversely, knockdown of IRF1 resulted in reduced mRNA levels of death ligands and receptors in hepatocytes, as well as decreased caspase 8 activity, leading to a notable decrease in hepatocyte apoptosis and liver injury [16,17,20]. Yu, Y. et al. demonstrated that IRF1 played a critical role in promoting P62 expression by activating autophagy through P38 phosphorylation, leading to hepatocyte death [77]. Additionally, Yan, B. et al. showed that IRF1 overexpression induces autophagy, suppresses β-catenin expression, and worsens hepatic IRI [73]. Furthermore, IRF1 exacerbated hepatic IRI via JNK-mediated autophagy, resulting in increased Beclin1 levels. Conversely, silencing IRF1 reduces high mobility group box 1 (HMGB1) expression and release in the liver, decreases LC3II and Beclin1 levels, and mitigates hepatic IRI [72,75]. Reactive oxygen species (ROS) also played a crucial role in IRI of the liver, primarily originating from Kupfer cells and mitochondria during IRI [117]. Elevated hepatic ROS levels at the onset of IRI triggered the expression of IRF1, promoting the transcription of HMGB1. Acetylated HMGB1 then activated the downstream TLR-4/NF-κB signaling pathway, resulting in the release of inflammatory mediators like TNF-α and IL-1β, worsening hepatic IRI damage [70]. The inhibition of IRF1-mediated HMGB1 release and subsequent TLR activation or p38 MAPK signaling pathway inactivation was shown to prevent hepatic IRI [71,79]. In addition, IRI upregulated iNOS and promoted the transcriptional activity of IRF1 through HDAC2-mediated histone H3 acetylation, which led to cell death and tissue injury [74]. Moreover, IRF1 regulated the transcription of Rab27a (a guanosine triphosphatase) and extracellular vesicle secretion, leading to oxidized phospholipids activation in neutrophils and subsequent hepatic IRI [76]. Thus, autophagy and oxidative stress play an important role in hepatic IRI, but it has not been clearly elucidated, and more studies are still needed.

3.1.2. Inflammatory Cytokines

IRI first triggers pro-inflammatory signaling cascades like TNF-α, IL-6, IFN, and NF-kB. As liver cells perish, cytokines and chemokines are produced, leading to a chemical storm that attracts neutrophils and other immune cells to the liver. This recruitment further stimulates the release of CXCL1, CXCL2, and complement, exacerbating the IRI [118]. Moreover, IRI activates the innate immune system, which drives the overall development of inflammatory hepatocellular injury. Further exploration of the roles played by immune cells and inflammatory cytokines in IRI is crucial for us to design safe and effective therapeutic strategies to ameliorate IRI in patients [119]. In recent years, it has been found that multiple IRF family members can promote the progression of IRI by regulating interleukin production and immune cell infiltration. Yokota, S. et al. discovered that knocking down IRF1 led to a decrease in the population of NK, NKT, and CD8+ T cells in the liver. Additionally, they observed that IRF1 increases cytotoxic effects and systemic inflammatory responses, worsening hepatic injury by controlling the expression of IL-15 and IL-15Rα mRNA in mouse hepatocytes and hepatic dendritic cells [78]. Another study by Nakano, R. et al. demonstrated that overexpression of IRF1 in ApoE-/- mice exacerbated hepatic IRI injury by activating hepatic NK and T cells through IL-15 production [18]. Additionally, early induction of IL-23 in hepatic IRI activated the IFN-γ/IRF1 pathway, leading to increased apoptosis and necrosis [21]. Type I IFNs were found to up-regulate hepatic IRF1 expression, which in turn regulated apoptosis and induced hepatic injury after IRI. Deprivation of plasmacytoid dendritic cells (pDC) in mice after IRI resulted in milder hepatic injury, reduced levels of hepatic IL-6, TNF-α, and apoptosis, and impaired expression of IRF1 and pro-apoptotic molecules such as Fas ligand, Fas, and death receptor 5 [19]. Therefore, IRF1 played a crucial role in hepatic IRI by regulating immune cells and their cytokines, making it a potential target for mitigating IRI. In addition, endogenous IRF2 was typically expressed in the liver and can be slightly upregulated by liver IRI. Overexpression of IRF2 was shown to protect against hepatic IRI and also restricted the production of IRF1-dependent proinflammatory factors such as IL-12, IFNβ [45]. Loi, P. et al. found that IRF3-deficient mice exhibited heightened hepatic necrosis and increased neutrophil infiltration due to elevated expression of IL-12/IL-23p40, IL-23p19, and IL-17A mRNA, along with reduced expression of IL-27p28 mRNA [80]. Wang, P.-X. et al. also found that IRF9 decreased the expression of SIRT1 and increased the level of acetyl p53, leading to a significant increase in immune cell infiltration, inflammatory cytokine levels and hepatocyte apoptosis [68]. Therefore, an in-depth study of the role of IRFs in IRI can help us better understand the mechanisms of disease development and develop feasible therapeutic measures.

3.2. IRFs and Alcohol-Induced Liver Injury/Alcoholic Liver Disease

Alcoholic-induced liver injury (ALI) is typically identified by disrupted liver function, inflammatory cell accumulation, and oxidative stress. Clinically, alcoholic liver disease (ALD) is the predominant form of ALI. The abuse of alcohol leads to liver injury through various mechanisms, such as oxidative and non-oxidative ethanol metabolism, the production of oxidative stress, damage and dysfunction of mitochondria and lysosomes, endoplasmic reticulum stress, inflammation, cytokine release, and the triggering of cell death [120]. Approximately 90% of individuals with alcoholism develop hepatic steatosis, with 35% of those individuals progressing to alcoholic hepatitis. Unfortunately, 40% of patients diagnosed with severe alcoholic hepatitis do not survive beyond 6 months despite receiving treatment [121]. Regrettably, corticosteroids continue to be the conventional treatment for severe alcoholic hepatitis, showing no advancements in the past forty years [121,122]. It is imperative to enhance our comprehension of the pathogenic mechanisms underlying ALD, particularly the liver injury it induces, to formulate more efficient treatment approaches. Luther, J. et al. discovered that mice fed alcohol showed heightened hepatic expression of the cGAS-IRF3 pathway. And they observed that the downregulation of connexin 32 (the predominant hepatic gap junction) led to a decrease in IRF3 expression, ultimately leading to a decrease in liver injury as evidenced by a notable reduction in ALT/AST levels [89]. Furthermore, IRF1-mediated caspase 1 inflammatory vesicles and NOX2-dependent ROS pathways were found to exacerbate ALI and steatosis [87]. In addition, excessive alcohol consumption led to dysbiosis of the gut microbiota, compromising the integrity of the intestinal epithelium and facilitating the transport of gut microbial products (e.g., LPS) to the liver, which was recognized by Toll-like receptor-4 (TLR4), ultimately leading to liver injury and subsequent ALD [88,123]. Liang, S. et al. demonstrated that chronic ethanol intake and LPS injection resulted in elevated serum ALT and IL-1 levels, along with enhanced hepatic CCL5 and CXCL10 expression. In normal conditions, macrophages could facilitate IRF1 degradation through autophagy, eliminate damaged mitochondria, and restrict macrophage activation and inflammation. However, following p62 silencing or myeloid cell-specific autophagy-related 7 knockout, IRF1 accumulation occurred in autophagy-deficient macrophages and translocated into the nucleus, leading to increased expression of CCL5 and CXCL10 [88]. Petrasek, J. et al. showed that systemic IRF3 knockout mice were protective against ALI, steatosis, and inflammation, but knockout of IRF3 only in parenchymal cells exacerbated ALI. Hepatic parenchymal cells were further found to be a major source of type I IFNs, whose action was dependent on TLR4/IRF3. Meanwhile, type I IFNs potentiated LPS-induced IL-10 and inhibited inflammatory cytokine production in mouse macrophages and human leukocytes. Thus, IRF3 activation in liver parenchymal cells and the resulting type I IFNs are protective against ALD by modulating the inflammatory function of macrophages [124]. However, Sanz-Garcia, C. et al. found that Gao-binge ethanol exposure activated IRF3 signaling and led to liver injury. IRF3 was further found to have a non-transcriptional function and could be induced to bind to Bax in mitochondria and activate caspases 3 and 9, which in turn activated the apoptotic pathway and limited NF-κB activity [125]. Therefore, IRF3 could regulate the innate immune environment of the liver and alleviate ALI/ALD by increasing apoptosis of immune cells.

3.3. IRFs and Con A-Induced Liver Injury

Conjugin A (Con A), a plant lectin isolated from the miller bean (Canavalia ensiformis), was recognized for its role as a T-lymphocyte activator in the immune response to allograft rejection, viral infections or autoimmune disorders in mammals [126]. Con A stimulated the release of a variety of cytokines from immune cells (such as TNF-α, IFN-γ, GM-CSF, IL-2, IL-1β), induced oxidative stress, activated multiple signaling pathways (NF-κB, MAPK, PI3K/PDK1/mTOR, STAT3, and STAT5), and altered the Bax/Bcl-2 ratio, ultimately resulting in severe liver inflammation and hepatocyte apoptosis/necrosis [83,127,128,129,130]. However, the precise cellular mechanisms underlying liver dysfunction induced by Con A activation remained incompletely understood. Recent studies have revealed the involvement of IRFs in Con A-induced liver injury. Binding of Con A to the mannose 6-phosphate receptor of HSCs induced JAK2/STAT-1 phosphorylation and promoted IRF1 transcription, which in turn inhibited superoxide dismutase (SOD) expression and promoted JNK and caspase 3 activation, leading to oxidative stress and apoptosis in hepatocytes [82]. In a study involving mice injected with Con A, elevated levels of IL-28A were observed. Deficiency in IL-28A was found to limit M1 macrophage polarization by modulating a signaling pathway that inhibits IRF5, consequently reducing the release of pro-inflammatory cytokines such as TNF-α, IL-12, IL-6, and IL-1β from M1 macrophages [83]. Corilagin was shown to effectively inhibit the release of pro-inflammatory cytokines in M1 macrophages by restraining the activation of the IRF5 signaling pathway, providing protection against Con A-induced immune-mediated liver injury. This treatment also resulted in reduced expression of M1 macrophage-associated pro-inflammatory cytokines and genes, including IL-6, IL-12, and iNOS [84]. Additionally, in Con A-induced hepatitis, the absence of liver X receptor α (LXRα) resulted in an increase in MDSCs in the liver, which in turn attenuated liver injury. Mechanistically, MDSCs from LXRα-/- mice exhibited significantly lower expression of IRF8, facilitating the expansion of MDSCs and effectively reducing immune injury in the liver [85].

3.4. IRFs and Post-Transplantation/Other Modes of Liver Injury

Studies have shown that increased IRF1 and IRF4 expression in patients undergoing acute rejection post-liver transplantation contributes to liver inflammation and injury [91,131,132]. Moreover, treatment with tacrolimus (TCA) suppresses IRF4 expression, thereby alleviating acute rejection after liver transplantation. This effect may be mediated through the TAC-NFAT-IRF4 and BATF/JUN/IRF4 complex-IL-21 axes, with the latter inhibiting IL-12-producing Tfh cells and consequently reducing liver injury [91,92]. Zhao, W. et al. also demonstrated that silencing IRF4 resulted in decreased levels of inflammatory factors such as TNF-α, IL-6, and IFN-γ and induced anti-inflammatory IL-10 levels, thereby attenuating acute liver transplant rejection in mice [133]. In adult patients with acute hepatitis A caused by HAV infection, severe liver injury was observed with elevated levels of chemokines such as CXCL10, CCL4, and CCL5. However, inhibiting IRF3 expression was found to decrease CXCL10 production, thus alleviating liver injury [90]. Similarly, in patients diagnosed with primary sclerosing cholangitis and primary biliary cirrhosis, bile acids promoted IRF3 phosphorylation, resulting in elevated expression of the target gene ZBP1. This ultimately exacerbated hepatic injury and fibrosis through interaction with RIP1, RIP3, and NLRP3 [86]. Additionally, in an LPS-induced liver injury model in mice, IRF3 expression was increased, a transcription factor was linked to systemic inflammation, while B-HA was shown to attenuate LPS-stimulated inflammatory responses by inhibiting the activation of the TLR4 signaling pathway through the phosphorylation of IRF3 [134,135]. In summary, IRFs play an important role in liver injury caused by a variety of etiological factors, and in-depth study of the molecular mechanisms of IRFs can help to understand the occurrence of liver injury and develop appropriate therapeutic strategies.

3.5. IRFs and Nonalcoholic Fatty Liver Disease

Inflammation has significant implications for metabolism, particularly in the context of obesity and NAFLD. Studies on NAFLD mice induced with a high-fat, high-fructose diet revealed the progression of simple steatosis, steatohepatitis, and hepatic fibrosis over 4, 8, and 16 weeks, respectively. The expression of IRF3 and IRF7 showed an increase at week 4, peaked at week 8, and returned to basal levels by week 16 [136]. However, research on liver tissues from 11 patients with NAFLD and 11 controls did not show a significant difference in IRF3 expression [137]. In mice on an HFD diet, systemic knockdown of IRF3 prevented steatosis and glycemic abnormalities, while hepatocyte-specific knockdown of IRF3 only impacted glycemic abnormalities. Mechanistically, IRF3-mediated Ppp2r1b induced an increase in PP2A activity, leading to AMPKα and AKT dephosphorylation [93]. Knockdown of IRF3 in mouse livers with HFD and FFA-induced L-O2 cells resulted in reduced hepatic inflammation and apoptosis, potentially through the regulation of the NF-κB signaling pathway, inflammatory cytokines, and apoptotic signaling [94]. Moreover, obese NAFLD patients show heightened hepatic IRF3 activation, which can be reversed with bariatric surgical treatment [93]. In a NASH mouse model, skeletal muscle-specific IRF4 knockout mice displayed improvements in hepatic steatosis, inflammation, and fibrosis without affecting body weight. IRF4 plays a role in transcriptionally regulating FSTL1, establishing a connection between muscle and liver [93]. In the HFD-induced NAFLD model, hepatic IRF6 was inhibited by promoter hypermethylation, and hepatocyte-specific transgenic mice overexpressing IRF6 exhibited attenuated steatosis and metabolic disease. Mechanistically, hepatocyte IRF6 bound directly to the promoter of the PPARγ gene and subsequently stopped transcription of PPARγ and its target genes (regulating adipogenesis and fatty acid uptake), resulting in amelioration of NAFLD progression [59].

3.6. IRFs and Liver Fibrosis

Liver fibrosis is a precursor to cirrhosis and results from extracellular mesenchymal protein deposition, activated hepatic stellate cells (HSC) are essential for the development of liver fibrosis, and liver inflammation triggered by activation of liver resident macrophages and massive leukocyte aggregation are also associated with liver fibrosis-related acute and chronic liver injury. A study revealed a lower frequency of the AG genotype of the IRF3(-925A/G) gene in cirrhotic patients, suggesting a potential protective effect against HCV infection [138]. Yu, J. et al. discovered that Gαs-coupled GPCR signaling increased IRF3 phosphorylation through cAMP-mediated PKA activation, resulting in elevated IL-33 expression, ultimately promoting HSC activation, and subsequently contributing to hepatic fibrosis progression [99]. Iracheta-Vellve, A. et al. demonstrated that in CCL4-treated hepatocytes, endoplasmic reticulum (ER) stress via STING triggered TBK1 phosphorylation, followed by IRF3 phosphorylation, which then interacted with BAX in mitochondria through its BH3-only structural domain, leading to pro-apoptotic caspase 3 activation and hepatocyte apoptosis, ultimately contributing to liver fibrosis [98]. However, Wu, Q. et al. found that STING increased IRF3 phosphorylation via TBK1, subsequently inhibiting CDK4/6-mediated RB hyperphosphorylation and inactivating E2F transcription factors, thereby inducing senescence in HSC cells. Interestingly, total knockdown or conditional deletion of IRF3 in HSC exacerbated liver fibrosis, indicating a dual role for IRF3 in this process that required further investigation [100]. Moreover, INF-γ was found to significantly upregulate indoleamine 2,3-dioxygenase (IDO) expression through STAT1 activation, leading to tryptophan depletion and subsequent G1 cell cycle arrest. Upon release from IFN-γ-induced G1 cell cycle arrest by 1-MT treatment, HSC apoptosis was significantly increased due to enhanced IRF1 expression [97]. Similarly, GRh2 attenuates hepatic inflammation and fibrosis by up-regulating IRF1 expression, which inhibits SLC7A11 and promotes HSC iron death and inactivation [96]. In human hepatocyte macrophages with liver fibrosis from NAFLD or HCV infection, elevated IRF5 expression triggered the release of inflammatory cytokines and death effectors, leading to hepatocyte caspase-dependent apoptosis. Conversely, IRF5-deficient macrophages under hepatocyte stress exhibited immunosuppressive polarization, secreting IL-10 and TGFβ to support BCL2 family member-mediated anti-apoptotic signaling in hepatocytes during metabolic or toxic stress-induced liver fibrosis [56].

3.7. IRFs and Hepatocellular Carcinoma

HCC posed a significant global health challenge due to its high fatality rate [139,140]. Despite undergoing combination therapies such as radiochemotherapy and immunotherapy, the 5-year survival rate for HCC remained notably low [141]. IRF1 and IRF2 were essential in regulating interferon activity, where the absence of IRF1 and the increase in IRF2 expression had been associated with aggressive traits in different types of cancer. IRF1, acting as a tumor suppressor gene, enhanced the migration of CD8+ T-cells, NK cells, and NKT cells, while also stimulating IFN-γ secretion in NK and NKT cells. This activation led to apoptosis in tumor cells through the CXCL10/CXCR3 paracrine axis [29]. Conversely, the downregulation of IRF2 significantly reduced invasive capacity, correlating with the decreased expression of STAT3, p-STAT3 and MMP9 [142]. However, increased IRF1 mRNA expression was observed in patients with highly differentiated or early HCC. In vitro studies demonstrated that IFN-γ induced PD-L1 mRNA and protein expression by enhancing IRF1 levels in both mouse and human HCC cells, whereas IRF2 overexpression down-regulated IFN-γ-induced PD-L1 promoter activity and protein abundance. Additionally, IRF1 was found to antagonize IRF2 binding to the IRE promoter element in PD-L1, leading to the upregulation of PD-L1 in the tumor microenvironment [143]. Furthermore, IRF1 was found to post-transcriptionally suppress and induce apoptosis in HCC cells by facilitating the interaction between miR-195 and the 3′UTR of checkpoint kinase 1 (CHK1). However, IRF1 also enhanced PD-L1 expression by promoting STAT3 phosphorylation [106]. Therefore, elevated IRF-1 expression not only triggered apoptosis in HCC cells but also boosted PD-L1 levels, shedding light on the regulation of the PD-L1/PD-1 pathway in HCC therapy. In both human and mouse HCC cells, IL-33 overexpression was shown to inhibit proliferation and decrease PD-L1 levels at the transcriptional level by enhancing the ubiquitin-dependent degradation of IRF1. This process was disrupted by E3 ligase RanBP2-mediated SUMOylation of IL-33 at Lys54 [102]. Additionally, treatment with cisplatin and CHK1 inhibitors led to the upregulation of major histocompatibility complex (MHC) class I associated chains A (MICA) expression through IRF1-mediated transcriptional effects, resulting in increased infiltration of NK cells and CD8+ T cells in HCC tissues [104]. FOXO1 functioned as a tumor suppressor by promoting macrophage infiltration and antitumor polarization via positive regulation of the IRF1/NO pathway. Polarized macrophages further inhibited HCC proliferation and migration by suppressing IL-6/STAT3 activation [105]. NR4A1 was markedly upregulated in tumor-infiltrating NK cells, which reduced the efficacy of anti-PD-1 therapy through modulation of the IFN-γ/p-STAT1/IRF1 signaling pathway, leading to dysfunctional tumor-infiltrating NK cells [103]. Moreover, high expression levels of miR-23a, miR-31, and miR-301a in HCC enhanced its progression by inhibiting IRF1 expression [107,108,109]. Conversely, miR-345 showed low expression in HCC, counteracting the inhibitory impact of IRF1 through reversible trans-overexpression, thereby influencing the epithelial-mesenchymal transition process and tumor metastasis [101]. Phosphatase and tensin homolog (PTEN) in HBV-associated HCC regulated the nuclear localization of IRF3 by dephosphorylating IRF3 at ser97, leading to the inhibition of the PI3K/AKT pathway and the reduction of oncogenic effects [110]. IRF4 overexpression suppressed the proliferation and migration capabilities of HCC cells by inhibiting the JAK2/STAT3 signaling pathway and epithelial-mesenchymal transition [48]. IRF5, identified as a tumor suppressor, exhibits down-regulated mRNA and protein expression levels in HCV-infected human hepatocytes and cells with autonomous replication of HCV RNA. Conversely, restoration of IRF5 expression hampered HCV protein translation and RNA replication [111]. However, Fang, Y. et al. observed an upregulation of IRF5 expression in HCC, facilitating an oncogenic effect by enhancing glycolysis through upregulation of lactate dehydrogenase A (LDHA) expression [112]. In addition, miR-424-3p reduced the interferon pathway by attenuating the transcriptional activation of STAT1/2 and IRF9 genes by SRF, which in turn enhanced matrix metalloproteinases (MMPs)-mediated ECM remodeling [113]. Therefore, IRF1, IRF3, IRF5, and IRF9 are all involved in the development of HCC, and further research into the molecular mechanisms involving IRFs in carcinogenesis could facilitate the discovery of new targeted therapies.

4. Conclusions and Future Perspectives

In summary, IRF1, IRF2, IRF3, IRF5, IRF8, and IRF9 are involved in hepatic IRI; IRF1, IRF5, and IRF8 are involved in Con A-induced liver injury; IRF3 is involved in cholestasis-induced liver injury and HAV-induced liver injury; IRF4 is involved in liver transplantation-induced liver injury; IRF1 and IRF3 are involved in alcohol-induced liver injury; IRF3, IRF4, and IRF6 are involved in the development of NAFLD; IRF1, IRF3, and IRF5 are involved in the development of liver fibrosis; and IRF1, IRF3, IRF4, IRF5, IRF8, and IRF9 are involved in the development of HCC. Almost all members of the IRF family play a role in different liver diseases, especially IRF1, which has been the most studied. IRF7 is less well-studied in liver diseases, and its expression is only increased in certain disease processes and usually accompanied by IRF3. Although the family of IRFs plays a very important role in liver diseases, there are no targeted drugs against IRFs to treat the disease process. In addition, the same IRFs may play different functions in different liver diseases, promoting or inhibiting the disease. Therefore, further clarification of the molecular mechanisms of IRFs in liver diseases is needed in the future to guide clinical practices and the development of corresponding targeted drugs.

Author Contributions

Literature analysis and conceptualization, C.Z.; original draft preparation and writing, C.Z. and X.Z.; review, and supervision, H.L., Z.H. and M.C. All authors have read and agreed to the published version of the manuscript.

Funding

The work was supported by the Key Research and Development Program of Hubei Province to Mingkai Chen (2020BCB007), and Renmin Hospital of Wuhan University Cross-Innovation Talent Project to Mingkai Chen (JCRCFZ-2022-017).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new experimental data were created.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Jefferies, C.A. Regulating IRFs in IFN Driven Disease. Front. Immunol. 2019, 10, 441432. [Google Scholar] [CrossRef] [PubMed]
  2. Zhou, H.; Tang, Y.D.; Zheng, C. Revisiting IRF1-mediated antiviral innate immunity. Cytokine Growth Factor Rev. 2022, 64, 1–6. [Google Scholar] [CrossRef] [PubMed]
  3. Wang, X.; Zhang, X.; Wang, G.; Wang, L.; Lin, Y.; Sun, F. Hsa-miR-513b-5p suppresses cell proliferation and promotes P53 expression by targeting IRF2 in testicular embryonal carcinoma cells. Gene 2017, 626, 344–353. [Google Scholar] [CrossRef] [PubMed]
  4. Liao, W.; Overman, M.J.; Boutin, A.T.; Shang, X.; Zhao, D.; Dey, P.; Li, J.; Wang, G.; Lan, Z.; Li, J.; et al. KRAS-IRF2 Axis Drives Immune Suppression and Immune Therapy Resistance in Colorectal Cancer. Cancer Cell 2019, 35, 559–572.e7. [Google Scholar] [CrossRef] [PubMed]
  5. Guo, Y.; Xu, J.; Du, Q.; Yan, Y.; Geller, D.A. IRF2 regulates cellular survival and Lenvatinib-sensitivity of hepatocellular carcinoma (HCC) through regulating β-catenin. Transl. Oncol. 2021, 14, 101059. [Google Scholar] [CrossRef] [PubMed]
  6. Qing, F.; Liu, Z. Interferon regulatory factor 7 in inflammation, cancer and infection. Front. Immunol. 2023, 14, 1190841. [Google Scholar] [CrossRef]
  7. Ma, W.; Huang, G.; Wang, Z.; Wang, L.; Gao, Q. IRF7: Role and regulation in immunity and autoimmunity. Front. Immunol. 2023, 14, 1236923. [Google Scholar] [CrossRef] [PubMed]
  8. Al Hamrashdi, M.; Brady, G. Regulation of IRF3 activation in human antiviral signaling pathways. Biochem. Pharmacol. 2022, 200, 115026. [Google Scholar] [CrossRef]
  9. Kaur, A.; Lee, L.H.; Chow, S.C.; Fang, C.M. IRF5-mediated immune responses and its implications in immunological disorders. Int. Rev. Immunol. 2018, 37, 229–248. [Google Scholar] [CrossRef]
  10. Wong, R.W.J.; Ong, J.Z.L.; Theardy, M.S.; Sanda, T. IRF4 as an Oncogenic Master Transcription Factor. Cancers 2022, 14, 4314. [Google Scholar] [CrossRef]
  11. Moorman, H.R.; Reategui, Y.; Poschel, D.B.; Liu, K. IRF8: Mechanism of Action and Health Implications. Cells 2022, 11, 2630. [Google Scholar] [CrossRef] [PubMed]
  12. Schutte, B.C.; Saal, H.M.; Goudy, S.; Leslie, E.J. IRF6-Related Disorders. In GeneReviews(®); Adam, M.P., Feldman, J., Mirzaa, G.M., Pagon, R.A., Wallace, S.E., Bean, L.J.H., Gripp, K.W., Amemiya, A., Eds.; University of Washington: Seattle, WA, USA, 1993. [Google Scholar]
  13. Fink, K.; Grandvaux, N. STAT2 and IRF9: Beyond ISGF3. Jakstat 2013, 2, e27521. [Google Scholar] [CrossRef] [PubMed]
  14. Paul, A.; Ismail, M.N.; Tang, T.H.; Ng, S.K. Phosphorylation of interferon regulatory factor 9 (IRF9). Mol. Biol. Rep. 2023, 50, 3909–3917. [Google Scholar] [CrossRef] [PubMed]
  15. Li, L.; Chen, S.N.; Nie, P. IRF11 regulates positively type I IFN transcription and antiviral response in mandarin fish, Siniperca chuatsi. Dev. Comp. Immunol. 2021, 114, 103846. [Google Scholar] [CrossRef] [PubMed]
  16. Tsung, A.; Stang, M.T.; Ikeda, A.; Critchlow, N.D.; Izuishi, K.; Nakao, A.; Chan, M.H.; Jeyabalan, G.; Yim, J.H.; Geller, D.A. The transcription factor interferon regulatory factor-1 mediates liver damage during ischemia-reperfusion injury. Am. J. Physiol.-Gastrointest. Liver Physiol. 2006, 290, G1261–G1268. [Google Scholar] [CrossRef] [PubMed]
  17. Ueki, S.; Dhupar, R.; Cardinal, J.; Tsung, A.; Yoshida, J.; Ozaki, K.S.; Klune, J.R.; Murase, N.; Geller, D.A. Critical role of interferon regulatory factor-1 in murine liver transplant ischemia reperfusion injury. Hepatology 2010, 51, 1692–1701. [Google Scholar] [CrossRef] [PubMed]
  18. Nakano, R.; Chogahara, I.; Ohira, M.; Imaoka, K.; Sato, S.; Bekki, T.; Sato, K.; Imaoka, Y.; Marlen, D.; Tanaka, Y.; et al. Atherosclerosis Deteriorates Liver Ischemia/Reperfusion Injury Via Interferon Regulatory Factor-1 Overexpression in a Murine Model. Transplant. Proc. 2024, 56, 678–685. [Google Scholar] [CrossRef] [PubMed]
  19. Castellaneta, A.; Yoshida, O.; Kimura, S.; Yokota, S.; Geller, D.A.; Murase, N.; Thomson, A.W. Plasmacytoid dendritic cell-derived IFN-α promotes murine liver ischemia/reperfusion injury by induction of hepatocyte IRF-1. Hepatology 2014, 60, 267–277. [Google Scholar] [CrossRef] [PubMed]
  20. Kim, K.-H.; Dhupar, R.; Ueki, S.; Cardinal, J.; Pan, P.; Cao, Z.; Cho, S.W.; Murase, N.; Tsung, A.; Geller, D.A. Donor graft interferon regulatory factor-1 gene transfer worsens liver transplant ischemia/reperfusion injury. Surgery 2009, 146, 181–189. [Google Scholar] [CrossRef]
  21. Klune, J.R.; Bartels, C.; Luo, J.; Yokota, S.; Du, Q.; Geller, D.A. IL-23 mediates murine liver transplantation ischemia-reperfusion injury via IFN-γ/IRF-1 pathway. Am. J. Physiol.-Gastrointest. Liver Physiol. 2018, 315, G991–G1002. [Google Scholar] [CrossRef]
  22. Honda, K.; Taniguchi, T. IRFs: Master regulators of signalling by Toll-like receptors and cytosolic pattern-recognition receptors. Nat. Rev. Immunol. 2006, 6, 644–658. [Google Scholar] [CrossRef] [PubMed]
  23. Ghazarian, M.; Revelo, X.S.; Nøhr, M.K.; Luck, H.; Zeng, K.; Lei, H.; Tsai, S.; Schroer, S.A.; Park, Y.J.; Chng, M.H.Y.; et al. Type I Interferon Responses Drive Intrahepatic T cells to Promote Metabolic Syndrome. Sci. Immunol. 2017, 2, eaai7616. [Google Scholar] [CrossRef] [PubMed]
  24. Vadrot, N.; Legrand, A.; Nello, E.; Bringuier, A.F.; Guillot, R.; Feldmann, G. Inducible nitric oxide synthase (iNOS) activity could be responsible for resistance or sensitivity to IFN-gamma-induced apoptosis in several human hepatoma cell lines. J. Interferon Cytokine Res. 2006, 26, 901–913. [Google Scholar] [CrossRef] [PubMed]
  25. Detjen, K.M.; Murphy, D.; Welzel, M.; Farwig, K.; Wiedenmann, B.; Rosewicz, S. Downregulation of p21(waf/cip-1) mediates apoptosis of human hepatocellular carcinoma cells in response to interferon-gamma. Exp. Cell Res. 2003, 282, 78–89. [Google Scholar] [CrossRef] [PubMed]
  26. Gao, J.; Senthil, M.; Ren, B.; Yan, J.; Xing, Q.; Yu, J.; Zhang, L.; Yim, J.H. IRF-1 transcriptionally upregulates PUMA, which mediates the mitochondrial apoptotic pathway in IRF-1-induced apoptosis in cancer cells. Cell Death Differ. 2010, 17, 699–709. [Google Scholar] [CrossRef] [PubMed]
  27. Zhang, X.N.; Liu, J.X.; Hu, Y.W.; Chen, H.; Yuan, Z.H. Hyper-activated IRF-1 and STAT1 contribute to enhanced interferon stimulated gene (ISG) expression by interferon alpha and gamma co-treatment in human hepatoma cells. Biochim. Biophys. Acta 2006, 1759, 417–425. [Google Scholar] [CrossRef] [PubMed]
  28. Tada, S.; Saito, H.; Tsunematsu, S.; Ebinuma, H.; Wakabayashi, K.; Masuda, T.; Ishii, H. Interferon regulatory factor-1 gene abnormality and loss of growth inhibitory effect of interferon-alpha in human hepatoma cell lines. Int. J. Oncol. 1998, 13, 1207–1216. [Google Scholar] [CrossRef]
  29. Yan, Y.; Zheng, L.; Du, Q.; Yazdani, H.; Dong, K.; Guo, Y.; Geller, D.A. Interferon regulatory factor 1(IRF-1) activates anti-tumor immunity via CXCL10/CXCR3 axis in hepatocellular carcinoma (HCC). Cancer Lett. 2021, 506, 95–106. [Google Scholar] [CrossRef]
  30. Wang, R.; Guo, H.; Tang, X.; Zhang, T.; Liu, Y.; Zhang, C.; Yu, H.; Li, Y. Interferon Gamma-Induced Interferon Regulatory Factor 1 Activates Transcription of HHLA2 and Induces Immune Escape of Hepatocellular Carcinoma Cells. Inflammation 2022, 45, 308–330. [Google Scholar] [CrossRef]
  31. Taniguchi, T.; Ogasawara, K.; Takaoka, A.; Tanaka, N. IRF family of transcription factors as regulators of host defense. Annu. Rev. Immunol. 2001, 19, 623–655. [Google Scholar] [CrossRef]
  32. Barnes, B.; Lubyova, B.; Pitha, P.M. On the role of IRF in host defense. J. Interferon Cytokine Res. 2002, 22, 59–71. [Google Scholar] [CrossRef] [PubMed]
  33. Nguyen, H.; Hiscott, J.; Pitha, P.M. The growing family of interferon regulatory factors. Cytokine Growth Factor. Rev. 1997, 8, 293–312. [Google Scholar] [CrossRef] [PubMed]
  34. Chen, W.; Lam, S.S.; Srinath, H.; Jiang, Z.; Correia, J.J.; Schiffer, C.A.; Fitzgerald, K.A.; Lin, K.; Royer, W.E., Jr. Insights into interferon regulatory factor activation from the crystal structure of dimeric IRF5. Nat. Struct. Mol. Biol. 2008, 15, 1213–1220. [Google Scholar] [CrossRef] [PubMed]
  35. Ikushima, H.; Negishi, H.; Taniguchi, T. The IRF Family Transcription Factors at the Interface of Innate and Adaptive Immune Responses. Cold Spring Harb. Symp. Quant. Biol. 2013, 78, 105–116. [Google Scholar] [CrossRef] [PubMed]
  36. Chen, Y.-J.; Li, J.; Lu, N.; Shen, X.-Z. Interferon regulatory factors: A key to tumour immunity. Int. Immunopharmacol. 2017, 49, 1–5. [Google Scholar] [CrossRef] [PubMed]
  37. Storchi, L.; Remoli, A.L.; Marsili, G.; Acchioni, C.; Acchioni, M.; Battistini, A.; Sgarbanti, M.; Marrone, A. A model of the three-dimensional structure of human interferon responsive factor 1 and its modifications upon phosphorylation or phosphorylation-mimicking mutations. J. Biomol. Struct. Dyn. 2019, 37, 4632–4643. [Google Scholar] [CrossRef] [PubMed]
  38. Forero, A.; Ozarkar, S.; Li, H.; Lee, C.H.; Hemann, E.A.; Nadjsombati, M.S.; Hendricks, M.R.; So, L.; Green, R.; Roy, C.N.; et al. Differential Activation of the Transcription Factor IRF1 Underlies the Distinct Immune Responses Elicited by Type I and Type III Interferons. Immunity 2019, 51, 451–464.e6. [Google Scholar] [CrossRef] [PubMed]
  39. Schoggins, J.W.; Wilson, S.J.; Panis, M.; Murphy, M.Y.; Jones, C.T.; Bieniasz, P.; Rice, C.M. A diverse range of gene products are effectors of the type I interferon antiviral response. Nature 2011, 472, 481–485. [Google Scholar] [CrossRef] [PubMed]
  40. Venkatesh, D.; Ernandez, T.; Rosetti, F.; Batal, I.; Cullere, X.; Luscinskas, F.W.; Zhang, Y.; Stavrakis, G.; García-Cardeña, G.; Horwitz, B.H.; et al. Endothelial TNF Receptor 2 Induces IRF1 Transcription Factor-Dependent Interferon-β Autocrine Signaling to Promote Monocyte Recruitment. Immunity 2013, 38, 1025–1037. [Google Scholar] [CrossRef]
  41. Yamane, D.; Feng, H.; Rivera-Serrano, E.E.; Selitsky, S.R.; Hirai-Yuki, A.; Das, A.; McKnight, K.L.; Misumi, I.; Hensley, L.; Lovell, W.; et al. Basal expression of interferon regulatory factor 1 drives intrinsic hepatocyte resistance to multiple RNA viruses. Nat. Microbiol. 2019, 4, 1096–1104. [Google Scholar] [CrossRef]
  42. Feng, H.; Lenarcic, E.M.; Yamane, D.; Wauthier, E.; Mo, J.; Guo, H.; McGivern, D.R.; González-López, O.; Misumi, I.; Reid, L.M.; et al. NLRX1 promotes immediate IRF1-directed antiviral responses by limiting dsRNA-activated translational inhibition mediated by PKR. Nat. Immunol. 2017, 18, 1299–1309. [Google Scholar] [CrossRef]
  43. Blumenthal, A.; Feng, H.; Zhang, Y.-B.; Gui, J.-F.; Lemon, S.M.; Yamane, D. Interferon regulatory factor 1 (IRF1) and anti-pathogen innate immune responses. PLoS Pathog. 2021, 17, e1009220. [Google Scholar] [CrossRef]
  44. Rueschenbaum, S.; Cai, C.; Schmidt, M.; Schwarzkopf, K.; Dittmer, U.; Zeuzem, S.; Welsch, C.; Lange, C.M. Translation of IRF-1 Restricts Hepatic Interleukin-7 Production to Types I and II Interferons: Implications for Hepatic Immunity. Front. Immunol. 2021, 11, 581352. [Google Scholar] [CrossRef] [PubMed]
  45. Klune, J.R.; Dhupar, R.; Kimura, S.; Ueki, S.; Cardinal, J.; Nakao, A.; Nace, G.; Evankovich, J.; Murase, N.; Tsung, A.; et al. Interferon regulatory factor-2 is protective against hepatic ischemia-reperfusion injury. Am. J. Physiol.-Gastrointest. Liver Physiol. 2012, 303, G666–G673. [Google Scholar] [CrossRef] [PubMed]
  46. Wang, J.; Li, H.; Xue, B.; Deng, R.; Huang, X.; Xu, Y.; Chen, S.; Tian, R.; Wang, X.; Xun, Z.; et al. IRF1 Promotes the Innate Immune Response to Viral Infection by Enhancing the Activation of IRF3. J. Virol. 2020, 94, 10-1128. [Google Scholar] [CrossRef]
  47. Glanz, A.; Chakravarty, S.; Fan, S.; Chawla, K.; Subramanian, G.; Rahman, T.; Walters, D.; Chakravarti, R.; Chattopadhyay, S. Autophagic degradation of IRF3 induced by the small-molecule auranofin inhibits its transcriptional and proapoptotic activities. J. Biol. Chem. 2021, 297, 101274. [Google Scholar] [CrossRef]
  48. Yuan, J.; Liu, Z.; Wu, Z.; Yang, J.; Yang, J. Construction and validation of an IRF4 risk score to predict prognosis and response to immunotherapy in hepatocellular carcinoma. Int. Immunopharmacol. 2022, 113, 109411. [Google Scholar] [CrossRef] [PubMed]
  49. Eguchi, J.; Kong, X.; Tenta, M.; Wang, X.; Kang, S.; Rosen, E.D. Interferon Regulatory Factor 4 Regulates Obesity-Induced Inflammation Through Regulation of Adipose Tissue Macrophage Polarization. Diabetes 2013, 62, 3394–3403. [Google Scholar] [CrossRef]
  50. Takaoka, A.; Yanai, H.; Kondo, S.; Duncan, G.; Negishi, H.; Mizutani, T.; Kano, S.; Honda, K.; Ohba, Y.; Mak, T.W.; et al. Integral role of IRF-5 in the gene induction programme activated by Toll-like receptors. Nature 2005, 434, 243–249. [Google Scholar] [CrossRef]
  51. Krausgruber, T.; Blazek, K.; Smallie, T.; Alzabin, S.; Lockstone, H.; Sahgal, N.; Hussell, T.; Feldmann, M.; Udalova, I.A. IRF5 promotes inflammatory macrophage polarization and TH1-TH17 responses. Nat. Immunol. 2011, 12, 231–238. [Google Scholar] [CrossRef]
  52. Saliba, D.G.; Heger, A.; Eames, H.L.; Oikonomopoulos, S.; Teixeira, A.; Blazek, K.; Androulidaki, A.; Wong, D.; Goh, F.G.; Weiss, M.; et al. IRF5:RelA interaction targets inflammatory genes in macrophages. Cell Rep. 2014, 8, 1308–1317. [Google Scholar] [CrossRef]
  53. Weiss, M.; Blazek, K.; Byrne, A.J.; Perocheau, D.P.; Udalova, I.A. IRF5 is a specific marker of inflammatory macrophages in vivo. Mediat. Inflamm. 2013, 2013, 245804. [Google Scholar] [CrossRef]
  54. Almuttaqi, H.; Udalova, I.A. Advances and challenges in targeting IRF5, a key regulator of inflammation. FEBS J. 2018, 286, 1624–1637. [Google Scholar] [CrossRef]
  55. Khoyratty, T.E.; Udalova, I.A. Diverse mechanisms of IRF5 action in inflammatory responses. Int. J. Biochem. Cell Biol. 2018, 99, 38–42. [Google Scholar] [CrossRef] [PubMed]
  56. Alzaid, F.; Lagadec, F.; Albuquerque, M.; Ballaire, R.; Orliaguet, L.; Hainault, I.; Blugeon, C.; Lemoine, S.; Lehuen, A.; Saliba, D.G.; et al. IRF5 governs liver macrophage activation that promotes hepatic fibrosis in mice and humans. JCI Insight 2016, 1, e88689. [Google Scholar] [CrossRef] [PubMed]
  57. Kim, I.K.; Diamond, M.S.; Yuan, S.; Kemp, S.B.; Kahn, B.M.; Li, Q.; Lin, J.H.; Li, J.; Norgard, R.J.; Thomas, S.K.; et al. Plasticity-induced repression of Irf6 underlies acquired resistance to cancer immunotherapy in pancreatic ductal adenocarcinoma. Nat. Commun. 2024, 15, 1532. [Google Scholar] [CrossRef] [PubMed]
  58. Li, D.; Cheng, P.; Wang, J.; Qiu, X.; Zhang, X.; Xu, L.; Liu, Y.; Qin, S. IRF6 Is Directly Regulated by ZEB1 and ELF3, and Predicts a Favorable Prognosis in Gastric Cancer. Front. Oncol. 2019, 9, 220. [Google Scholar] [CrossRef]
  59. Tong, J.; Han, C.J.; Zhang, J.Z.; He, W.Z.; Zhao, G.J.; Cheng, X.; Zhang, L.; Deng, K.Q.; Liu, Y.; Fan, H.F.; et al. Hepatic Interferon Regulatory Factor 6 Alleviates Liver Steatosis and Metabolic Disorder by Transcriptionally Suppressing Peroxisome Proliferator-Activated Receptor γ in Mice. Hepatology 2019, 69, 2471–2488. [Google Scholar] [CrossRef] [PubMed]
  60. Tan, L.; Qu, W.; Wu, D.; Liu, M.; Ai, Q.; Hu, H.; Wang, Q.; Chen, W.; Zhou, H. The interferon regulatory factor 6 promotes cisplatin sensitivity in colorectal cancer. Bioengineered 2022, 13, 10504–10517. [Google Scholar] [CrossRef]
  61. Ning, S.; Pagano, J.S.; Barber, G.N. IRF7: Activation, regulation, modification and function. Genes. Immun. 2011, 12, 399–414. [Google Scholar] [CrossRef]
  62. Luo, W.W.; Tong, Z.; Cao, P.; Wang, F.B.; Liu, Y.; Zheng, Z.Q.; Wang, S.Y.; Li, S.; Wang, Y.Y. Transcription-independent regulation of STING activation and innate immune responses by IRF8 in monocytes. Nat. Commun. 2022, 13, 4822. [Google Scholar] [CrossRef] [PubMed]
  63. Salem, S.; Salem, D.; Gros, P. Role of IRF8 in immune cells functions, protection against infections, and susceptibility to inflammatory diseases. Hum. Genet. 2020, 139, 707–721. [Google Scholar] [CrossRef] [PubMed]
  64. Jiang, D.S.; Wei, X.; Zhang, X.F.; Liu, Y.; Zhang, Y.; Chen, K.; Gao, L.; Zhou, H.; Zhu, X.H.; Liu, P.P.; et al. IRF8 suppresses pathological cardiac remodelling by inhibiting calcineurin signalling. Nat. Commun. 2014, 5, 3303. [Google Scholar] [CrossRef] [PubMed]
  65. Shi, G.; Zhang, Z.; Ma, S.; Li, Y.; Du, S.; Chu, Y.; Li, Y.; Tang, X.; Yang, Y.; Chen, Z.; et al. Hepatic interferon regulatory factor 8 expression mediates liver ischemia/reperfusion injury in mice. Biochem. Pharmacol. 2021, 192, 114728. [Google Scholar] [CrossRef] [PubMed]
  66. Wu, H.; Li, Y.; Shi, G.; Du, S.; Wang, X.; Ye, W.; Zhang, Z.; Chu, Y.; Ma, S.; Wang, D.; et al. Hepatic interferon regulatory factor 8 expression suppresses hepatocellular carcinoma progression and enhances the response to anti–programmed cell death protein-1 therapy. Hepatology 2022, 76, 1602–1616. [Google Scholar] [CrossRef] [PubMed]
  67. Wang, X.-A.; Zhang, R.; Jiang, D.; Deng, W.; Zhang, S.; Deng, S.; Zhong, J.; Wang, T.; Zhu, L.-H.; Yang, L.; et al. Interferon regulatory factor 9 protects against hepatic insulin resistance and steatosis in male mice. Hepatology 2013, 58, 603–616. [Google Scholar] [CrossRef] [PubMed]
  68. Wang, P.-X.; Zhang, R.; Huang, L.; Zhu, L.-H.; Jiang, D.-S.; Chen, H.-Z.; Zhang, Y.; Tian, S.; Zhang, X.-F.; Zhang, X.-D.; et al. Interferon regulatory factor 9 is a key mediator of hepatic ischemia/reperfusion injury. J. Hepatol. 2015, 62, 111–120. [Google Scholar] [CrossRef]
  69. Dhawan, T.; Zahoor, M.A.; Heryani, N.; Workenhe, S.T.; Nazli, A.; Kaushic, C. TRIM26 Facilitates HSV-2 Infection by Downregulating Antiviral Responses through the IRF3 Pathway. Viruses 2021, 13, 70. [Google Scholar] [CrossRef]
  70. Li, K.; Feng, Z.; Wang, L.; Ma, X.; Wang, L.; Liu, K.; Geng, X.; Peng, C. Chlorogenic Acid Alleviates Hepatic Ischemia–Reperfusion Injury by Inhibiting Oxidative Stress, Inflammation, and Mitochondria-Mediated Apoptosis In Vivo and In Vitro. Inflammation 2023, 46, 1061–1076. [Google Scholar] [CrossRef]
  71. Luo, J.; Li, J.; Li, T.; Zhang, Z.; Chen, G.; Li, Q.; Qi, H.; Si, Z.; Fan, Z. PIAS1 Alleviates Hepatic Ischemia-Reperfusion Injury in Mice through a Mechanism Involving NFATc1 SUMOylation. Dis. Markers 2022, 2022, 4988539. [Google Scholar] [CrossRef]
  72. Li, S.; He, J.; Xu, H.; Yang, J.; Luo, Y.; Song, W.; Qiao, B.; Zhang, H. Autophagic activation of IRF-1 aggravates hepatic ischemia–reperfusion injury via JNK signaling. MedComm 2021, 2, 91–100. [Google Scholar] [CrossRef]
  73. Yan, B.; Luo, J.; Kaltenmeier, C.; Du, Q.; Stolz, D.B.; Loughran, P.; Yan, Y.; Cui, X.; Geller, D.A. Interferon Regulatory Factor-1 (IRF1) activates autophagy to promote liver ischemia/reperfusion injury by inhibiting β-catenin in mice. PLoS ONE 2020, 15, e0239119. [Google Scholar] [CrossRef]
  74. Du, Q.; Luo, J.; Yang, M.-Q.; Liu, Q.; Heres, C.; Yan, Y.-H.; Stolz, D.; Geller, D.A. iNOS/NO is required for IRF1 activation in response to liver ischemia-reperfusion in mice. Mol. Med. 2020, 26, 56. [Google Scholar] [CrossRef]
  75. Cui, Z.; Li, S.; Liu, Z.; Zhang, Y.; Zhang, H. Interferon Regulatory Factor 1 Activates Autophagy to Aggravate Hepatic Ischemia-Reperfusion Injury by Increasing High Mobility Group Box 1 Release. Cell. Physiol. Biochem. 2018, 48, 328–338. [Google Scholar] [CrossRef]
  76. Yang, M.q.; Du, Q.; Goswami, J.; Varley, P.R.; Chen, B.; Wang, R.h.; Morelli, A.E.; Stolz, D.B.; Billiar, T.R.; Li, J.; et al. Interferon regulatory factor 1–Rab27a regulated extracellular vesicles promote liver ischemia/reperfusion injury. Hepatology 2018, 67, 1056–1070. [Google Scholar] [CrossRef] [PubMed]
  77. Yu, Y.; Li, S.; Wang, Z.; He, J.; Ding, Y.; Zhang, H.; Yu, W.; Shi, Y.; Cui, Z.; Wang, X.; et al. Interferon regulatory factor-1 activates autophagy to aggravate hepatic ischemia-reperfusion injury via the P38/P62 pathway in mice. Sci. Rep. 2017, 7, 43684. [Google Scholar] [CrossRef] [PubMed]
  78. Yokota, S.; Yoshida, O.; Dou, L.; Spadaro, A.V.; Isse, K.; Ross, M.A.; Stolz, D.B.; Kimura, S.; Du, Q.; Demetris, A.J.; et al. IRF-1 Promotes Liver Transplant Ischemia/Reperfusion Injury via Hepatocyte IL-15/IL-15Rα Production. J. Immunol. 2015, 194, 6045–6056. [Google Scholar] [CrossRef]
  79. Cho, H.I.; Kim, K.M.; Kwak, J.H.; Lee, S.K.; Lee, S.M. Protective mechanism of anethole on hepatic ischemia/reperfusion injury in mice. J. Nat. Prod. 2013, 76, 1717–1723. [Google Scholar] [CrossRef] [PubMed]
  80. Loi, P.; Yuan, Q.; Torres, D.; Delbauve, S.; Laute, M.-A.; Lalmand, M.-C.; Pétein, M.; Goriely, S.; Goldman, M.; Flamand, V. Interferon regulatory factor 3 deficiency leads to interleukin-17-mediated liver ischemia-reperfusion injury. Hepatology 2013, 57, 351–361. [Google Scholar] [CrossRef]
  81. Nasiri, M.; Saadat, M.; Karimi, M.H.; Azarpira, N.; Saadat, I. Evaluating mRNA Expression Levels of the TLR4/IRF5 Signaling Axis during Hepatic Ischemia-Reperfusion Injuries. Exp. Clin. Transpl. Transplant. 2019, 17, 648–652. [Google Scholar] [CrossRef]
  82. Rani, R.; Kumar, S.; Sharma, A.; Mohanty, S.K.; Donnelly, B.; Tiao, G.M.; Gandhi, C.R. Mechanisms of concanavalin A-induced cytokine synthesis by hepatic stellate cells: Distinct roles of interferon regulatory factor-1 in liver injury. J. Biol. Chem. 2018, 293, 18466–18476. [Google Scholar] [CrossRef] [PubMed]
  83. Zhang, J.; Cheng, D.; Zhang, H.; Liu, Z.; Gao, M.; Wei, L.; Yan, F.; Li, C.; Wang, L.; Dong, G.; et al. Interleukin 28A aggravates Con A-induced acute liver injury by promoting the recruitment of M1 macrophages. FASEB J. 2024, 38, e23443. [Google Scholar] [CrossRef] [PubMed]
  84. Yan, F.; Cheng, D.; Wang, H.; Gao, M.; Zhang, J.; Cheng, H.; Wang, C.; Zhang, H.; Xiong, H. Corilagin Ameliorates Con A-Induced Hepatic Injury by Restricting M1 Macrophage Polarization. Front. Immunol. 2022, 12, 807509. [Google Scholar] [CrossRef] [PubMed]
  85. Li, B.; Lian, M.; Li, Y.; Qian, Q.; Zhang, J.; Liu, Q.; Tang, R.; Ma, X. Myeloid-Derived Suppressive Cells Deficient in Liver X Receptor α Protected from Autoimmune Hepatitis. Front. Immunol. 2021, 12, 732102. [Google Scholar] [CrossRef] [PubMed]
  86. Zhuang, Y.; Ortega-Ribera, M.; Thevkar Nagesh, P.; Joshi, R.; Huang, H.; Wang, Y.; Zivny, A.; Mehta, J.; Parikh, S.M.; Szabo, G. Bile acid-induced IRF3 phosphorylation mediates cell death, inflammatory responses, and fibrosis in cholestasis-induced liver and kidney injury via regulation of ZBP1. Hepatology 2024, 79, 752–767. [Google Scholar] [CrossRef] [PubMed]
  87. Li, H.; Chen, X.; Xu, J.; Zhu, L.; Li, C.; Sun, X.; Li, X.; Guo, J.; Li, J.; Wang, S.; et al. GRP/GRPR enhances alcohol-associated liver injury through the IRF1-mediated Caspase-1 inflammasome and NOX2-dependent ROS pathway. Hepatology 2024, 79, 392–408. [Google Scholar] [CrossRef] [PubMed]
  88. Liang, S.; Zhong, Z.; Kim, S.Y.; Uchiyama, R.; Roh, Y.S.; Matsushita, H.; Gottlieb, R.A.; Seki, E. Murine macrophage autophagy protects against alcohol-induced liver injury by degrading interferon regulatory factor 1 (IRF1) and removing damaged mitochondria. J. Biol. Chem. 2019, 294, 12359–12369. [Google Scholar] [CrossRef] [PubMed]
  89. Luther, J.; Khan, S.; Gala, M.K.; Kedrin, D.; Sridharan, G.; Goodman, R.P.; Garber, J.J.; Masia, R.; Diagacomo, E.; Adams, D.; et al. Hepatic gap junctions amplify alcohol liver injury by propagating cGAS-mediated IRF3 activation. Proc. Natl. Acad. Sci. USA 2020, 117, 11667–11673. [Google Scholar] [CrossRef]
  90. Sung, P.S.; Hong, S.-H.; Lee, J.; Park, S.-H.; Yoon, S.K.; Chung, W.J.; Shin, E.-C. CXCL10 is produced in hepatitis A virus-infected cells in an IRF3-dependent but IFN-independent manner. Sci. Rep. 2017, 7, 6387. [Google Scholar] [CrossRef]
  91. Tang, T.; Xu, T.; Liu, X.; Yang, T.; Zhang, L.; Yang, Z. Roles of BATF/JUN/IRF4 complex in tacrolimus mediated immunosuppression on Tfh cells in acute rejection after liver transplantation. J. Cell. Physiol. 2020, 236, 1776–1786. [Google Scholar] [CrossRef]
  92. Tang, T.; Lu, Q.; Yang, X.; Liu, X.; Liao, R.; Zhang, Y.; Yang, Z. Roles of the tacrolimus-dependent transcription factor IRF4 in acute rejection after liver transplantation. Int. Immunopharmacol. 2015, 28, 257–263. [Google Scholar] [CrossRef] [PubMed]
  93. Patel, S.J.; Liu, N.; Piaker, S.; Gulko, A.; Andrade, M.L.; Heyward, F.D.; Sermersheim, T.; Edinger, N.; Srinivasan, H.; Emont, M.P.; et al. Hepatic IRF3 fuels dysglycemia in obesity through direct regulation of Ppp2r1b. Sci. Transl. Med. 2022, 14, eabh3831. [Google Scholar] [CrossRef] [PubMed]
  94. Qiao, J.T.; Cui, C.; Qing, L.; Wang, L.S.; He, T.Y.; Yan, F.; Liu, F.Q.; Shen, Y.H.; Hou, X.G.; Chen, L. Activation of the STING-IRF3 pathway promotes hepatocyte inflammation, apoptosis and induces metabolic disorders in nonalcoholic fatty liver disease. Metabolism 2018, 81, 13–24. [Google Scholar] [CrossRef] [PubMed]
  95. Guo, S.; Feng, Y.; Zhu, X.; Zhang, X.; Wang, H.; Wang, R.; Zhang, Q.; Li, Y.; Ren, Y.; Gao, X.; et al. Metabolic crosstalk between skeletal muscle cells and liver through IRF4-FSTL1 in nonalcoholic steatohepatitis. Nat. Commun. 2023, 14, 6047. [Google Scholar] [CrossRef] [PubMed]
  96. Lang, Z.; Yu, S.; Hu, Y.; Tao, Q.; Zhang, J.; Wang, H.; Zheng, L.; Yu, Z.; Zheng, J. Ginsenoside Rh2 promotes hepatic stellate cell ferroptosis and inactivation via regulation of IRF1-inhibited SLC7A11. Phytomedicine 2023, 118, 154950. [Google Scholar] [CrossRef] [PubMed]
  97. Oh, J.E.; Shim, K.Y.; Lee, J.I.; Choi, S.I.; Baik, S.K.; Eom, Y.W. 1-Methyl-L-tryptophan promotes the apoptosis of hepatic stellate cells arrested by interferon-γ by increasing the expression of IFN-γRβ, IRF-1 and FAS. Int. J. Mol. Med. 2017, 40, 576–582. [Google Scholar] [CrossRef] [PubMed]
  98. Iracheta-Vellve, A.; Petrasek, J.; Gyongyosi, B.; Satishchandran, A.; Lowe, P.; Kodys, K.; Catalano, D.; Calenda, C.D.; Kurt-Jones, E.A.; Fitzgerald, K.A.; et al. Endoplasmic Reticulum Stress-induced Hepatocellular Death Pathways Mediate Liver Injury and Fibrosis via Stimulator of Interferon Genes. J. Biol. Chem. 2016, 291, 26794–26805. [Google Scholar] [CrossRef] [PubMed]
  99. Yu, J.-H.; Choi, M.G.; Lee, N.Y.; Kwon, A.; Lee, E.; Koo, J.H. Hepatocyte GPCR signaling regulates IRF3 to control hepatic stellate cell transdifferentiation. Cell Commun. Signal. 2024, 22, 48. [Google Scholar] [CrossRef] [PubMed]
  100. Wu, Q.; Leng, X.; Zhang, Q.; Zhu, Y.Z.; Zhou, R.; Liu, Y.; Mei, C.; Zhang, D.; Liu, S.; Chen, S.; et al. IRF3 activates RB to authorize cGAS-STING-induced senescence and mitigate liver fibrosis. Sci. Adv. 2024, 10, eadj2102. [Google Scholar] [CrossRef]
  101. Yu, M.; Xue, H.; Wang, Y.; Shen, Q.; Jiang, Q.; Zhang, X.; Li, K.; Jia, M.; Jia, J.; Xu, J.; et al. miR-345 inhibits tumor metastasis and EMT by targeting IRF1-mediated mTOR/STAT3/AKT pathway in hepatocellular carcinoma. Int. J. Oncol. 2017, 50, 975–983. [Google Scholar] [CrossRef]
  102. Wang, Z.; Pan, B.; Qiu, J.; Zhang, X.; Ke, X.; Shen, S.; Wu, X.; Yao, Y.; Tang, N. SUMOylated IL-33 in the nucleus stabilizes the transcription factor IRF1 in hepatocellular carcinoma cells to promote immune escape. Sci. Signal 2023, 16, eabq3362. [Google Scholar] [CrossRef]
  103. Yu, W.; He, J.; Wang, F.; He, Q.; Shi, Y.; Tao, X.; Sun, B. NR4A1 mediates NK-cell dysfunction in hepatocellular carcinoma via the IFN-γ/p-STAT1/IRF1 pathway. Immunology 2022, 169, 69–82. [Google Scholar] [CrossRef]
  104. Li, X.; Huang, J.; Wu, Q.; Du, Q.; Wang, Y.; Huang, Y.; Cai, X.; Geller, D.A.; Yan, Y. Inhibition of Checkpoint Kinase 1 (CHK1) Upregulates Interferon Regulatory Factor 1 (IRF1) to Promote Apoptosis and Activate Anti-Tumor Immunity via MICA in Hepatocellular Carcinoma (HCC). Cancers 2023, 15, 850. [Google Scholar] [CrossRef]
  105. Cui, X.; Zhao, H.; Wei, S.; Du, Q.; Dong, K.; Yan, Y.; Geller, D.A. Hepatocellular carcinoma-derived FOXO1 inhibits tumor progression by suppressing IL-6 secretion from macrophages. Neoplasia 2023, 40, 100900. [Google Scholar] [CrossRef]
  106. Yan, Y.; Zheng, L.; Du, Q.; Cui, X.; Dong, K.; Guo, Y.; Geller, D.A. Interferon regulatory factor 1 (IRF-1) downregulates Checkpoint kinase 1 (CHK1) through miR-195 to upregulate apoptosis and PD-L1 expression in Hepatocellular carcinoma (HCC) cells. Br. J. Cancer 2021, 125, 101–111. [Google Scholar] [CrossRef]
  107. Dong, K.; Du, Q.; Cui, X.; Wan, P.; Kaltenmeier, C.; Luo, J.; Yan, B.; Yan, Y.; Geller, D.A. MicroRNA-301a (miR-301a) is induced in hepatocellular carcinoma (HCC) and down- regulates the expression of interferon regulatory factor-1. Biochem. Biophys. Res. Commun. 2020, 524, 273–279. [Google Scholar] [CrossRef]
  108. Wan, P.Q.; Zhang, J.H.; Du, Q.; Dong, K.; Luo, J.; Heres, C.; Geller, D.A. Analysis of the relationship between microRNA-31 and interferon regulatory factor-1 in hepatocellular carcinoma cells. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 647–654. [Google Scholar] [CrossRef] [PubMed]
  109. Yan, Y.; Liang, Z.; Du, Q.; Yang, M.; Geller, D.A. MicroRNA-23a downregulates the expression of interferon regulatory factor-1 in hepatocellular carcinoma cells. Oncol. Rep. 2016, 36, 633–640. [Google Scholar] [CrossRef]
  110. Kim, G.W.; Imam, H.; Khan, M.; Mir, S.A.; Kim, S.J.; Yoon, S.K.; Hur, W.; Siddiqui, A. HBV-Induced Increased N6 Methyladenosine Modification of PTEN RNA Affects Innate Immunity and Contributes to HCC. Hepatology 2020, 73, 533–547. [Google Scholar] [CrossRef]
  111. Cevik, O.; Li, D.; Baljinnyam, E.; Manvar, D.; Pimenta, E.M.; Waris, G.; Barnes, B.J.; Kaushik-Basu, N. Interferon regulatory factor 5 (IRF5) suppresses hepatitis C virus (HCV) replication and HCV-associated hepatocellular carcinoma. J. Biol. Chem. 2017, 292, 21676–21689. [Google Scholar] [CrossRef]
  112. Fang, Y.; Lu, Z.H.; Liu, B.Z.; Li, N.; Yang, M.Z.; Wang, P. IRF5 promotes glycolysis in the progression of hepatocellular carcinoma and is regulated by TRIM35. J. Dig. Dis. 2023, 24, 480–490. [Google Scholar] [CrossRef] [PubMed]
  113. Feng, L.; Chen, X.; Li, P.; Li, Y.; Zhai, Y.; Liu, X.; Jin, Q.; Zhang, H.; Yu, C.; Xing, B.; et al. miR-424-3p promotes metastasis of hepatocellular carcinoma via targeting the SRF-STAT1/2 axis. Carcinogenesis 2023, 44, 610–625. [Google Scholar] [CrossRef] [PubMed]
  114. Zhang, Y.; Zhu, J.; Guo, L.; Zou, Y.; Wang, F.; Shao, H.; Li, J.; Deng, X. Cholecystokinin protects mouse liver against ischemia and reperfusion injury. Int. Immunopharmacol. 2017, 48, 180–186. [Google Scholar] [CrossRef] [PubMed]
  115. Jiménez-Castro, M.B.; Cornide-Petronio, M.E.; Gracia-Sancho, J.; Peralta, C. Inflammasome-Mediated Inflammation in Liver Ischemia-Reperfusion Injury. Cells 2019, 8, 1131. [Google Scholar] [CrossRef] [PubMed]
  116. Gazia, C.; Lenci, I.; Manzia, T.M.; Martina, M.; Tisone, G.; Angelico, R.; Abenavoli, L.; Grassi, G.; Signorello, A.; Baiocchi, L. Current Strategies to Minimize Ischemia-Reperfusion Injury in Liver Transplantation: A Systematic Review. Rev. Recent. Clin. Trials 2021, 16, 372–380. [Google Scholar] [CrossRef] [PubMed]
  117. Elias-Miró, M.; Jiménez-Castro, M.B.; Rodés, J.; Peralta, C. Current knowledge on oxidative stress in hepatic ischemia/reperfusion. Free Radic. Res. 2013, 47, 555–568. [Google Scholar] [CrossRef] [PubMed]
  118. Kaltenmeier, C.; Wang, R.; Popp, B.; Geller, D.; Tohme, S.; Yazdani, H.O. Role of Immuno-Inflammatory Signals in Liver Ischemia-Reperfusion Injury. Cells 2022, 11, 2222. [Google Scholar] [CrossRef] [PubMed]
  119. Lu, L.; Zhou, H.; Ni, M.; Wang, X.; Busuttil, R.; Kupiec-Weglinski, J.; Zhai, Y. Innate Immune Regulations and Liver Ischemia-Reperfusion Injury. Transplantation 2016, 100, 2601–2610. [Google Scholar] [CrossRef] [PubMed]
  120. Williams, J.A.; Ding, W.-X. Role of autophagy in alcohol and drug-induced liver injury. Food Chem. Toxicol. 2020, 136, 111075. [Google Scholar] [CrossRef]
  121. Lucey, M.R.; Mathurin, P.; Morgan, T.R. Alcoholic hepatitis. N. Engl. J. Med. 2009, 360, 2758–2769. [Google Scholar] [CrossRef]
  122. Gao, B.; Bataller, R. Alcoholic liver disease: Pathogenesis and new therapeutic targets. Gastroenterology 2011, 141, 1572–1585. [Google Scholar] [CrossRef]
  123. Gao, B.; Seki, E.; Brenner, D.A.; Friedman, S.; Cohen, J.I.; Nagy, L.; Szabo, G.; Zakhari, S. Innate immunity in alcoholic liver disease. Am. J. Physiol. Gastrointest. Liver Physiol. 2011, 300, G516–G525. [Google Scholar] [CrossRef]
  124. Petrasek, J.; Dolganiuc, A.; Csak, T.; Nath, B.; Hritz, I.; Kodys, K.; Catalano, D.; Kurt-Jones, E.; Mandrekar, P.; Szabo, G. Interferon regulatory factor 3 and type I interferons are protective in alcoholic liver injury in mice by way of crosstalk of parenchymal and myeloid cells. Hepatology 2011, 53, 649–660. [Google Scholar] [CrossRef]
  125. Sanz-Garcia, C.; Poulsen, K.L.; Bellos, D.; Wang, H.; McMullen, M.R.; Li, X.; Chattopadhyay, S.; Sen, G.; Nagy, L.E. The non-transcriptional activity of IRF3 modulates hepatic immune cell populations in acute-on-chronic ethanol administration in mice. J. Hepatol. 2019, 70, 974–984. [Google Scholar] [CrossRef] [PubMed]
  126. Gantner, F.; Leist, M.; Lohse, A.W.; Germann, P.G.; Tiegs, G. Concanavalin A-induced T-cell-mediated hepatic injury in mice: The role of tumor necrosis factor. Hepatology 1995, 21, 190–198. [Google Scholar] [CrossRef] [PubMed]
  127. Ibrahim, S.R.M.; Sirwi, A.; Eid, B.G.; Mohamed, S.G.A.; Mohamed, G.A. Summary of Natural Products Ameliorate Concanavalin A-Induced Liver Injury: Structures, Sources, Pharmacological Effects, and Mechanisms of Action. Plants 2021, 10, 228. [Google Scholar] [CrossRef]
  128. Wang, Y.; Meng, J.; Men, L.; An, B.; Jin, X.; He, W.; Lu, S.; Li, N. Rosmarinic Acid Protects Mice from Concanavalin A-Induced Hepatic Injury through AMPK Signaling. Biol. Pharm. Bull. 2020, 43, 1749–1759. [Google Scholar] [CrossRef]
  129. Sang, R.; Yu, Y.; Ge, B.; Xu, L.; Wang, Z.; Zhang, X. Taraxasterol from Taraxacum prevents concanavalin A-induced acute hepatic injury in mice via modulating TLRs/NF-κB and Bax/Bc1-2 signalling pathways. Artif. Cells Nanomed. Biotechnol. 2019, 47, 3929–3937. [Google Scholar] [CrossRef]
  130. Ye, X.J.; Xu, R.; Liu, S.Y.; Hu, B.; Shi, Z.J.; Shi, F.L.; Zeng, B.; Xu, L.H.; Huang, Y.T.; Chen, M.Y.; et al. Taraxasterol mitigates Con A-induced hepatitis in mice by suppressing interleukin-2 expression and its signaling in T lymphocytes. Int. Immunopharmacol. 2022, 102, 108380. [Google Scholar] [CrossRef]
  131. Shen, K.; Zheng, S.S.; Park, O.; Wang, H.; Sun, Z.; Gao, B. Activation of innate immunity (NK/IFN-gamma) in rat allogeneic liver transplantation: Contribution to liver injury and suppression of hepatocyte proliferation. Am. J. Physiol. Gastrointest. Liver Physiol. 2008, 294, G1070–G1077. [Google Scholar] [CrossRef]
  132. Janfeshan, S.; Yaghobi, R.; Eidi, A.; Karimi, M.H.; Geramizadeh, B.; Malekhosseini, S.A.; Kafilzadeh, F. Expression Profile of Interferon Regulatory Factor 1 in Chronic Hepatitis B Virus-Infected Liver Transplant Patients. Exp. Clin. Transpl. Transplant. 2017, 15, 669–675. [Google Scholar] [CrossRef]
  133. Zhao, W.; Zhang, Z.; Zhao, Q.; Liu, M.; Wang, Y. Inhibition of Interferon Regulatory Factor 4 Attenuates Acute Liver Allograft Rejection in Mice. Scand. J. Immunol. 2015, 82, 262–268. [Google Scholar] [CrossRef] [PubMed]
  134. Nehme, A.; Ghahramanpouri, M.; Ahmed, I.; Golsorkhi, M.; Thomas, N.; Munoz, K.; Abdipour, A.; Tang, X.; Wilson, S.M.; Wasnik, S.; et al. Combination therapy of insulin-like growth factor I and BTP-2 markedly improves lipopolysaccharide-induced liver injury in mice. FASEB J. 2022, 36, e22444. [Google Scholar] [CrossRef]
  135. You, N.; Chu, S.; Cai, B.; Gao, Y.; Hui, M.; Zhu, J.; Wang, M. Bioactive hyaluronic acid fragments inhibit lipopolysaccharide-induced inflammatory responses via the Toll-like receptor 4 signaling pathway. Front. Med. 2021, 15, 292–301. [Google Scholar] [CrossRef] [PubMed]
  136. Liu, J.; Zhuang, Z.j.; Bian, D.x.; Ma, X.j.; Xun, Y.h.; Yang, W.j.; Luo, Y.; Liu, Y.l.; Jia, L.; Wang, Y.; et al. Toll-like receptor-4 signalling in the progression of non-alcoholic fatty liver disease induced by high-fat and high-fructose diet in mice. Clin. Exp. Pharmacol. Physiol. 2014, 41, 482–488. [Google Scholar] [CrossRef] [PubMed]
  137. Kanuri, G.; Ladurner, R.; Skibovskaya, J.; Spruss, A.; Königsrainer, A.; Bischoff, S.C.; Bergheim, I. Expression of toll-like receptors 1–5 but not TLR 6–10 is elevated in livers of patients with non-alcoholic fatty liver disease. Liver Int. 2014, 35, 562–568. [Google Scholar] [CrossRef] [PubMed]
  138. Talaat, R.M.; Elsayed, S.S.; Abdel-Hakem, N.E.; El-Shenawy, S.Z. Genetic Polymorphism in Toll-Like Receptor 3 and Interferon Regulatory Factor 3 in Hepatitis C Virus-Infected Patients: Correlation with Liver Cirrhosis. Viral Immunol. 2022, 35, 609–615. [Google Scholar] [CrossRef] [PubMed]
  139. Forner, A.; Reig, M.; Bruix, J. Hepatocellular carcinoma. Lancet 2018, 391, 1301–1314. [Google Scholar] [CrossRef] [PubMed]
  140. Craig, A.J.; von Felden, J.; Garcia-Lezana, T.; Sarcognato, S.; Villanueva, A. Tumour evolution in hepatocellular carcinoma. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 139–152. [Google Scholar] [CrossRef]
  141. Huang, A.; Yang, X.-R.; Chung, W.-Y.; Dennison, A.R.; Zhou, J. Targeted therapy for hepatocellular carcinoma. Signal Transduct. Target. Ther. 2020, 5, 146. [Google Scholar] [CrossRef]
  142. Yi, Y.; Wu, H.; Gao, Q.; He, H.W.; Li, Y.W.; Cai, X.Y.; Wang, J.X.; Zhou, J.; Cheng, Y.F.; Jin, J.J.; et al. Interferon regulatory factor (IRF)-1 and IRF-2 are associated with prognosis and tumor invasion in HCC. Ann. Surg. Oncol. 2013, 20, 267–276. [Google Scholar] [CrossRef] [PubMed]
  143. Yan, Y.; Zheng, L.; Du, Q.; Yan, B.; Geller, D.A. Interferon regulatory factor 1 (IRF-1) and IRF-2 regulate PD-L1 expression in hepatocellular carcinoma (HCC) cells. Cancer Immunol. Immunother. 2020, 69, 1891–1903. [Google Scholar] [CrossRef] [PubMed]
Table 1. The function and mechanism of IRFs in liver diseases.
Table 1. The function and mechanism of IRFs in liver diseases.
First
Author
Publication YearDiseaseModelStimulationState of IRFsInvolving
Molecules
Signal
Pathway
Effects
Nakano, R. [18]2024Hepatic Ischemia-Reperfusion InjurymiceApoE-/-upregulation of IRF1IL-15-Atherosclerosis can mirror intrahepatic immunity, particularly activating liver NK and T cells through IL-15 production, thereby exacerbating hepatic damage. The upregulation of IL-15 expression is associated with IRF1 overexpression.
Li, K. [70]2023Hepatic Ischemia-Reperfusion InjurySprague-Dawley ratChlorogenic acid (CGA)deregulation of IRF1HMGB1-CGA pretreatment significantly decreased the levels of reactive oxygen species following HIRI, inhibited HMGB1 release by decreasing IRF1 expression, inhibited the expression of HMGB1, TLR-4, MyD88, P-IκB-α, NF-κB P65, and P-P65, and promoted IκB-α degradation. Thus, CGA appears to inhibit oxidative stress and inflammatory responses during HIRI.
Luo, J. [71]2022Hepatic Ischemia-Reperfusion InjurymicePIAS1deregulation of IRF1p38PIAS1/NFATc1/HDAC1/IRF1/p38 MAPKPIAS1 inactivated p38 MAPK signaling by inhibiting HDAC1-mediated IRF1 through NFATc1 SUMOylation, thereby repressing the inflammatory response and apoptosis of hepatocytes in vitro, and alleviating liver injury in vivo.
Li, S. [72]2021Hepatic Ischemia-Reperfusion Injurymice-upregulation of IRF1JNK, Beclin1IRF1/JNKIRF1 is associated with JNK pathway activation followed by increases in Beclin1 protein levels. This JNK-induced autophagic cell death then leads to cell failure and plays an important role in liver function damage.
Yan, B. [73]2020Hepatic Ischemia-Reperfusion Injurymice-upregulation of IRF1β-catenin-IRF1-induced autophagy aggravates hepatic IR injury in part by inhibiting β-catenin.
Du, Q. [74]2020Hepatic Ischemia-Reperfusion InjurymiceiNOS/NOupregulation of IRF1iNOS, PUMA, p21-iNOS/NO-induced HDAC2 mediated histone H3 deacetylation and promoted IRF1 transcriptional activity.
Klune, J.R. [21]2018Hepatic Ischemia-Reperfusion InjurymiceIL-23upregulation of IRF1--The overexpression of IL-23 in vivo through the use of an adenovirus vector also led to the expression of IL-17, CXCL2, IFN-γ, and IRF1. The increased expression of IL-23 also led to increased apoptosis in the liver. IL-23 is induced early by I/R in the liver. Its signaling leads to the activation of the IL-17/CXCL2 and IFN-γ/IRF1 pathways, resulting in increased apoptosis and necrosis.
Cui, Z. [75]2018Hepatic Ischemia-Reperfusion Injurymice-upregulation of IRF1HMGB1-The levels of hepatic IRF1 mRNA and protein were significantly increased in livers after exposure to IRI, as well as an IRI-induced increase in HMGB1 mRNA and release of HMGB1 in liver tissue. IRF1 activates autophagy to aggravate hepatic IRI by increasing HMGB1 release.
Yang, M.Q. [76]2018Hepatic Ischemia-Reperfusion Injurymice-upregulation of IRF1Rab27a, extracellular vesicle, OxPL-IRF1 regulates Rab27a transcription and EV secretion, leading to OxPL activation of neutrophils and subsequent hepatic IR injury.
Yu, Y. [77]2017Hepatic Ischemia-Reperfusion Injurymice-upregulation of IRF1P38P38/P62IRF1 functioned as a trigger to activate autophagy via P38 activation and P62 was required for this P38-mediated autophagy. IRF1 appears to exert a pivotal role in hepatic IRI, by predisposing hepatocytes to activate an autophagic pathway. Such an effect promotes autophagic cell death through the P38/P62 pathway.
Yokota, S. [78]2015Hepatic Ischemia-Reperfusion Injurymice-upregulation of IRF1IL-15/IL-15Rα-IRF1 promotes liver transplantation (LTx) I/R injury via hepatocyte IL-15/IL-15Rα production which suggests that targeting IRF-1 and IL-15/IL-15Rα may be effective in reducing I/R injury associated with LTx.
Castellaneta, A. [19]2014Hepatic Ischemia-Reperfusion InjurymiceIFN-αupregulation of IRF1Fas ligand, its receptor (Fas) and death receptor 5-IFN-α derived from liver pDC plays a key role in the pathogenesis of liver I/R injury by enhancing apoptosis as a consequence of the induction of hepatocyte IRF1 expression.
Cho, H.I. [79]2013Hepatic Ischemia-Reperfusion Injurymiceanetholederegulation of IRF1HMGB1/TLR-Anethole protects against hepatic I/R injury by the suppression of IRF1-mediated HMGB1 release and subsequent TLR activation.
Ueki, S. [17]2010Hepatic Ischemia-Reperfusion Injurymice-deregulation of IRF1caspase-8, IFN-γ-IRF1 deficiency in liver grafts, but not in recipients, resulted in a significant reduction in hepatocyte apoptosis and liver injury, as well as improved survival. Deficiency of IRF1 signaling in grafts resulted in significantly reduced messenger RNA (mRNA) levels for death ligands and death receptors in hepatocytes, as well as decreased caspase-8 activities, indicating that IRF1 mediates death ligand-induced hepatocyte death.
Kim, K.H. [20]2009Hepatic Ischemia-Reperfusion InjurySprague-Dawley rat-upregulation of IRF1IFN-β, IFN-γ, IL-12, caspase-3-Rats pretreated with AdIRF-1 before transplantation had elevated alanine aminotransferase levels and increased expression of IFN-β, IFN-γ, IL-12, and iNOS in the short-term period (3 h) when compared with donor livers pretreated with Adnull.
Klune, J.R. [45]2012Hepatic Ischemia-Reperfusion Injurymice-upregulation of IRF1IL-12, IFN-β, iNOS-IRF2 overexpression limits the production of IRF1-dependent proinflammatory genes, such as IL-12, IFNβ, and iNOS, even in the presence of IRF1 induction. Additionally, isograft liver transplantation with IRF2 heterozygote knockout (IRF2+/-) donor grafts that have reduced endogenous IRF2 levels results in worse injury following cold I/R during murine orthotopic liver transplantation.
Loi, P. [80]2013Hepatic Ischemia-Reperfusion Injurymice-deregulation of IRF3IL-27p35, IL-27p28IRF3/TLR4/IL-23/IL-17Quantification of cytokine gene expression revealed an increased liver expression of IL-12/IL-23p40, IL-23p19 mRNA, and IL-17A mRNA in IRF3-deficient versus wildtype mice, whereas IL-27p28 mRNA expression was diminished in the absence of IRF3. IRF3-dependent events downstream of TLR4 control the IL-23/IL-17 axis in the liver and this regulatory role of IRF3 is relevant to liver ischemia-reperfusion injury.
Nasiri, M. [81]2019Hepatic Ischemia-Reperfusion InjurymiceN-acetylcysteinederegulation of IRF5--Pretreatment with N-acetylcysteine significantly decreased the mRNA levels of TLR4/IRF5 and its downstream cytokines 3 h after reperfusion and subsequently improved the previously mentioned hepatic damages 168 h after reperfusion.
Shi, G. [65]2021Hepatic Ischemia-Reperfusion Injurymice-upregulation of IRF8CCXCL1, CXCL9, CCRL2-IRF8 is up-regulated in the early hypnotic stage of IR. Upregulated IRF8 activates NF-κВ signaling pathway and promotes the release of autophagy-dependent or -independent chemokines, which in turn recruit massive neutrophils into the damaged liver sites, exacerbating hepatic I/R inflammatory injury.
Wang, P.X. [68]2015Hepatic Ischemia-Reperfusion Injurymice-upregulation of IRF9SIRT1, p53-A deficiency in IRF9 markedly reduced the necrotic area, serum ALT, AST, immune cell infiltration, inflammatory cytokine levels, and hepatocyte apoptosis after liver I/R. IRF9 has a novel function of inducing hepatocyte apoptosis after I/R injury by decreasing SIRT1 expression and increasing acetyl-p53 levels.
Rani, R. [82]2018Con A-Induced Hepatic InjurymiceCon Aupregulation of IRF1SOD, JNK, caspase 3-Con A binding to the mannose 6-phosphate receptor (Man-R) on HSCs induces JAK2 phosphorylation, which then phosphorylates STAT-1. The activated STAT1 translocates into the nucleus and induces IRF1 transcription. IRF1 produced this way stimulates IFNβ transcription. IFNβ protein released by HSCs binds to the IFNαβ receptor on hepatocytes and instigates JAK2/STAT1 activation, followed by IRF1 synthesis. IRF1 (through a currently unidentified mechanism) inhibits SOD expression leading to oxidative stress. Oxidative stress and IFNαβ stimulate JNK and caspase 3 activation, and cause apoptosis of hepatocytes.
Zhang, J. [83]2024Con A-Induced Hepatic InjurymiceIL-28A deletionderegulation of IRF5IL-1β, IL-6, IL-12, TNF-α-IL-28A deletion plays an important protective role in the Con A-induced acute liver injury model and IL-28A deficiency inhibits the activation of M1 macrophages by inhibiting the NF-κB, MAPK, and IRF signaling pathways.
Yan, F. [84]2022Con A-Induced Hepatic InjurymiceCorilaginderegulation of IRF5IL-6, IL-12, TNF-a, iNOS-Corilagin protects mice from Con A-induced immune-mediated hepatic injury by limiting M1 macrophage activation via the MAPK, NF-κB, and IRF signaling pathways, suggesting corilagin as a possible treatment choice for immune-mediated hepatic injury.
Li, B. [85]2021Con A-Induced Hepatic InjurymiceLXRa-/-deregulation of IRF8--Abrogation of LXRα facilitated the expansion of MDSCs by downregulating IRF8, thereby ameliorating hepatic immune injury profoundly.
Zhuang, Y. [86]2024Cholestatic-Induced Hepatic InjurymiceBile acidphosphorylation of IRF3ZBP1IRF3-ZBP1IRF3 knockout (IRF3-/-) mice showed significantly attenuated liver and kidney damage and fibrosis compared to wide-type mice after bile duct ligation. ZBP1 interacted with RIP1, RIP3, and NLRP3, thereby revealing its potential role in the regulation of cell-death and inflammation pathways. Bile acid-induced p-IRF3 and the IRF3-ZBP1 axis play a central role in the pathogenesis of cholestatic liver and kidney injury.
Li, H. [87]2024Alcohol-Induced Hepatic InjurymiceGRPRupregulation of IRF1Caspase-1, NOX2-The pro-inflammatory and oxidative stress roles of GRPR might be dependent on IRF1-mediated the Caspase-1 inflammasome and the NOX2-dependent reactive oxygen species pathway, respectively.
Liang, S. [88]2019Alcohol-Induced Hepatic Injurymicep62 silencing or ATG7 deletionupregulation of IRF1CCL5, CXCL10-Macrophage autophagy protects against ALD by promoting IRF1 degradation and removal of damaged mitochondria, limiting macrophage activation and inflammation. Upon p62 silencing or ATG7 deletion, IRF1 starts to accumulate in autophagy-deficient macrophages and translocates into the nucleus, where it induces CCL5 and CXCL10 expression.
Luther, J. [89]2020Alcohol-Induced Hepatic InjurymiceCx32 deletionderegulation of IRF3IFNβ, IFIT2, IFIT3-Disruption of Cx32 in ALD impaired IRF3-stimulated gene expression, resulting in decreased hepatic injury despite an increase in hepatic steatosis.
Sung, P.S. [90]2017Hepatitis A-Induced Hepatic InjuryHAV-infected cells-deregulation of IRF3CXCL10-CXCL10 production was reduced by silencing the expression of RIG-I-like receptor signal molecules, such as mitochondrial antiviral signaling proteins and IRF3, in HAV-infected cells.
Tang, T. [91]2021Liver Transplantation-Induced Hepatic InjuryratsTacrolimus (TAC)deregulation of IRF4IL-21BATF/JUN/IRF4 complex-IL-21TAC inhibited the expression of the BATF/JUN/IRF4 complex and interacted with the promoter of BATF and IRF4. The BATF/JUN/IRF4 complex participated in the inhibition of IL-21-producing Tfh cells after treatment with TAC.
Tang, T. [92]2015Liver Transplantation-Induced Hepatic InjuryratsTACderegulation of IRF4-TAC-NFAT-IRF4TAC treatment prolonged the survival of liver allografts in recipients, significantly attenuating hepatic tissue injury and improving liver function. IRF4 expression in grafts was downregulated after TAC treatment.
Patel, S.J. [93]2022Nonalcoholic Fatty Liver DiseasemiceHFDupregulation of IRF3Ppp2r1bIRF3-PPP2R1BGlobal ablation of IRF3 in mice on a high-fat diet protects against both steatosis and dysglycemia, whereas hepatocyte-specific loss of IRF3 affects only dysglycemia. Integration of the IRF3-dependent transcriptome and cistrome in mouse hepatocytes identifies Ppp2r1b as a direct IRF3 target responsible for mediating its metabolic actions on glucose homeostasis. IRF3-mediated induction of Ppp2r1b amplified PP2A activity, with subsequent dephosphorylation of AMPKα and AKT.
Qiao, J.T. [94]2018Nonalcoholic Fatty Liver DiseaseL-O2 cellKnocking down STINGderegulation of IRF3p-p65/p65, TNF-α, IL-6, and IL-1β-STING and IRF3 were upregulated in livers of HFD-fed mice and in FFA-induced L-O2 cells. Knocking down either STING or IRF3 led to a significant reduction in FFA-induced hepatic inflammation and apoptosis, as evidenced by the modulation of the NF-κB signaling pathway, inflammatory cytokines, and apoptotic signaling
Guo, S. [95]2023Nonalcoholic Fatty Liver DiseaseIRF4 knockout (F4MKO) mice-deregulation of IRF4FSTL1IRF4-FSTL1-DIP2A/CD14Skeletal muscle-specific IRF4 knockout (F4MKO) mice exhibited ameliorated hepatic steatosis, inflammation, and fibrosis, without changes in body weight, when put on a NASH diet. Dual luciferase assays showed that IRF4 can transcriptionally regulate FSTL1. Further, inducing FSTL1 expression in the muscles of F4MKO mice is sufficient to restore liver pathology.
Tong, J. [59]2019Nonalcoholic Fatty Liver DiseasemiceHFDderegulation of IRF6PPARγ-IRF6 is downregulated by the promoter, hypermethylation, upon metabolic stimulus exposure, which fails to inhibit Pparγ and its targets, driving abnormalities in lipid metabolism.
Lang, Z. [96]2023Liver FibrosismiceGRh2upregulation of IRF1SLC7A11-GRh2 up-regulates IRF1 expression, resulting in the suppression of SLC7A11, which contributes to HSC ferroptosis and inactivation. GRh2 ameliorates liver fibrosis by enhancing HSC ferroptosis and inhibiting liver inflammation. GRh2 may be a promising drug for treating liver fibrosis.
Oh, J.E. [97]2017Liver FibrosisHSCsIFN-γ and 1-methyl-L-tryptophan (1-MT)upregulation of IRF1--IDO expression was markedly increased by IFN-γ through STAT1 activation and resulted in the depletion of tryptophan. This depletion induced G1 cell cycle arrest. When the cells were released from an IFN-γ-mediated G1 cell cycle arrest by treatment with 1-MT, the apoptosis of the HSCs was markedly increased through the induction of IFN-γRβ, IRF1, and FAS.
Iracheta-Vellve, A. [98]2016Liver FibrosismiceSTINGphosphorylation of IRF3Caspase 3-In CCl4-treated hepatocytes, ER stresses results of the phosphorylation of TBK1 via STING, followed by phosphorylation of IRF3. IRF3 associates with BAX in the mitochondria through its BH3-only domain, leading to pro-apoptotic caspase 3 activation and hepatocyte apoptosis. After chronic CCl4 administration, hepatocyte apoptosis is associated with secondary necrosis, which results in liver fibrosis.
Yu, J.H. [99]2024Liver FibrosismiceGαs-coupled GPCR signalingIRF3 phosphorylationIL-33GPCR-IRF3-IL-33Gαs-coupled GPCR signaling increases IRF3 phosphorylation through cAMP-mediated activation of PKA. This leads to an increase in IL-33 expression, which further contributes to HSC activation. Hepatocyte GPCR signaling regulates IRF3 to control HSC trans-differentiation and provides insight for understanding the complex intercellular communication during liver fibrosis progression and suggests therapeutic opportunities for the disease.
Wu, Q. [100]2024Liver FibrosismiceSTINGIRF3 phosphorylationCDK4/6STING-IRF3-RBThe IRF3-RB interaction attenuates cyclin-dependent kinase 4/6 (CDK4/6)-mediated RB hyperphosphorylation that mobilizes RB to deactivate E2 family (E2F) transcription factors, thereby driving cells into senescence. STING-IRF3-RB signaling plays a notable role in HSCs within various murine models, pushing activated HSCs toward senescence. IRF3 global knockout or conditional deletion in HSCs aggravates liver fibrosis, a process mitigated by the CDK4/6 inhibitor.
Alzaid, F. [56]2016Liver Fibrosismicehepatocellular stressupregulation of IRF5FasL, TNF, MHC II, IL6, and IL1β-IRF5-competent liver macrophages undergo proinflammatory activation in response to hepatocellular stress. Proinflammatory activation (M1) induces inflammatory a cytokine and death effector release that induces hepatocyte caspase (Casp)-dependent apoptosis. Activated factors include Fas ligand (FasL), TNF, MHC II, IL6, and IL1β. Proinflammatory activation induces type 1 and type 17 responses from CD4+T cells. Type 1 and 17 cytokines include IL17 and IFNγ. In IRF5-deficient macrophages, hepatocellular stress leads to immunosuppressive (MReg) polarization and secretion of IL10 and TGFβ. TGFβ expression induces differentiation of CD4+T cells into CD4+ FoxP3+ T cells, which contributes to IL-10 release. Secretion of IL-10 in IRF5 deficiency promotes anti-apoptotic signaling in hepatocytes mediated by B cell lymphoma 2 (BCL2) family members. This process maintains cell survival under stress.
Yu, M. [101]2017Hepatocellular CarcinomaHCC cellsmiR-345deregulation of IRF1Slug, Snail and TwistmTOR/STAT3/AKTOver-expression of IRF1 mRNA was inversely correlated with the level of miR-345 in HCC specimens. Restoration of IRF1 resulted in promoted EMT and cell mobility in miR-345 overexpressing HCCLM3 cells. miR-345 acts as an inhibitor of the EMT process in HCC cells by targeting IRF1 and this study highlights the potential effects of miR-345 on prognosis and treatment of HCC.
Wang, Z. [102]2023Hepatocellular CarcinomaHCC cellsSUMOylated IL-33stabilized IRF1PD-L1, IL-8, CXCL1-An increase in SUMOylated IL-33 in HCC cells in cocultures and in vivo-stabilized IRF1 and increased PD-L1 abundance and chemokine IL-8 secretion, which prevented the activation of cytotoxic T cells and promoted the M2 polarization of macrophages, respectively.
Yu, W. [103]2023Hepatocellular CarcinomaNK cellNR4A1deregulation of IRF1-IFN-γ/p-STAT1/IRF1NR4A1 was significantly highly expressed in tumor-infiltrating NK cells, which mediated the dysfunction of tumor-infiltrating NK cells by regulating the IFN-γ/p-STAT1/IRF1 signaling pathway, attenuated the anti-tumor function of NK cells, and reduced the efficacy of anti-PD-1 therapy.
Li, X. [104]2023Hepatocellular CarcinomaHCC cellsCHK1deregulation of IRF1MICAIRF1-MICAOverexpressed CHK1 suppresses IRF1 expression through proteolysis. Cisplatin and CHK1 inhibition upregulate MICA expression through IRF1-mediated transcriptional effects. DNA damage regulates the interaction of CHK1 and IRF1 to activate anti-tumor immunity via the IRF1-MICA pathway in HCC.
Cui, X. [105]2023Hepatocellular CarcinomamiceFOXO1upregulation of IRF1CD206, IL-6, NO-These effects may be partially dependent on FOXO1 transcriptionally modulating the IRF-1/NO axis, exerting effects on macrophages and decreasing IL-6 release from macrophages in the tumor microenvironment indirectly. This feedback suppressed the progression of HCC by inactivating IL-6/STAT3 in HCC.
Yan, Y. [106]2021Hepatocellular CarcinomaHCC cellsIFN-γupregulation of IRF4miR-195, CHK1-IRF1 induces miR-195 to suppress CHK1 protein expression, which induces apoptosis of HCC cells. IRF1 expression or CHK1 inhibition also upregulates PD-L1 expression via increased STAT3 phosphorylation.
Yan, Y. [29]2021Hepatocellular Carcinomamice-upregulation of IRF1CXCL10/CXCR3IRF1/CXCL10/CXCR3IRF1 increased CD8+ T cells, NK and NKT cells migration, and activated IFN-γ secretion in NK and NKT cells to induce tumor apoptosis through the CXCL10/CXCR3 paracrine axis.
Dong, K. [107]2020Hepatocellular CarcinomaHCC cellsmiR-301aderegulation of IRF1--Chronic hypoxia induces miR-301a in HCC in vitro and decreases IRF1 expression. Finally, miR-301a inhibition increases apoptosis and decreases HCC cell proliferation.
Wan, P.Q. [108]2020Hepatocellular CarcinomaHCC cellsmiR-31deregulation of IRF1--IRF1 was negatively correlated with miR-31 in HCC tissues and paired adjacent tissues. The expression level of miR-31 was inversely correlated with IRF1. MiR-31 inhibitor up-regulated the expression levels of IRF-1 in HuH7 cells, whereas the miR-31 mimic down-regulated the expression levels of IRF1.
Yan, Y. [109]2016Hepatocellular CarcinomaHCC cellsmiR-23aderegulation of IRF1--MiR-23a mimics down-regulated IFNγ-induced IRF1 protein expression, while the miR-23a inhibitor increased IRF1. MiR-23a promotes HCC growth by downregulating IRF1.
Kim, G.W. [110]2021Hepatocellular CarcinomaHCC cellsPTENnegative phosphorylation of IRF3PI3K/AKT-PTEN controlled IRF3 nuclear localization by negative phosphorylation of IRF3 at Ser97, and PTEN reduced carcinogenesis by inhibiting the PI3K/AKT pathway.
Yuan, J. [48]2022Hepatocellular CarcinomaHCC cells-upregulation of IRF4JAK2/STAT3-In vitro experiments demonstrated that the overexpression of IRF4 inhibited the proliferation and migration capacity of HCC cells by restricting the JAK2/STAT3 signaling pathway and epithelial-mesenchymal transition.
Cevik, O. [111]2017Hepatocellular CarcinomaHCC cells-upregulation of IRF5HCV protein translation and RNA replication-IRF5 re-expression inhibited HCV protein translation and RNA replication. IRF5 re-expression induced apoptosis via loss in mitochondrial membrane potential, down-regulated autophagy, and inhibited hepatocyte cell migration/invasion.
Fang, Y. [112]2023Hepatocellular CarcinomaHCC cellsTRIM35deregulation of IRF5lactate dehydrogenase A (LDHA)-IRF5 was found to upregulate the expression of lactate dehydrogenase A (LDHA) and promote glycolysis. Tripartite motif containing 35 (TRIM35) interacted with IRF5, promoting its ubiquitination and degradation. These findings reveal the oncogenic function of IRF5 in the progression of HCC by enhancing glycolysis, further supporting the potential of IRF5 as a viable target for HCC therapy.
Wu, H. [66]2022Hepatocellular Carcinomamice-upregulation of IRF8CCL20-Overexpression of IRF8 in HCC cells significantly enhanced antitumor effects in immune-competent mice, modulating infiltration of tumor-associated macrophages (TAMs) and T cell exhaustion in tumor microenvironment. IRF8 regulated recruitment of TAMs by inhibiting the expression of CCL20. Adeno-associated virus 8-mediated hepatic IRF8 rescue significantly suppressed HCC progression and enhanced the response to anti-PD-1 therapy.
Feng, L. [113]2023Hepatocellular CarcinomaHCC cellsmiR-424-3pderegulation of IRF9ISG15, IFITM1, OASL, TRIM21SRF-STAT1/2/IRF9miR-424-3p reduces the interferon pathway by attenuating the transactivation of SRF on STAT1/2 and IRF9 genes, which in turn enhances the matrix metalloproteinases (MMPs)-mediated ECM remodeling.
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

Zeng, C.; Zhu, X.; Li, H.; Huang, Z.; Chen, M. The Role of Interferon Regulatory Factors in Liver Diseases. Int. J. Mol. Sci. 2024, 25, 6874. https://doi.org/10.3390/ijms25136874

AMA Style

Zeng C, Zhu X, Li H, Huang Z, Chen M. The Role of Interferon Regulatory Factors in Liver Diseases. International Journal of Molecular Sciences. 2024; 25(13):6874. https://doi.org/10.3390/ijms25136874

Chicago/Turabian Style

Zeng, Chuanfei, Xiaoqin Zhu, Huan Li, Ziyin Huang, and Mingkai Chen. 2024. "The Role of Interferon Regulatory Factors in Liver Diseases" International Journal of Molecular Sciences 25, no. 13: 6874. https://doi.org/10.3390/ijms25136874

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