Next Article in Journal
Oncotherapeutic Strategies in Early Onset Colorectal Cancer
Next Article in Special Issue
Hepatocellular Carcinoma in Patients with Nonalcoholic Fatty Liver Disease: The Prognostic Role of Liver Stiffness Measurement
Previous Article in Journal
Synergistic Combination of Luteolin and Asiatic Acid on Cervical Cancer In Vitro and In Vivo
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

P-21 Activated Kinases in Liver Disorders

1
Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
2
Zhejiang University School of Medicine, Hangzhou 310058, China
3
Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
4
Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou 310006, China
5
Department of Surgery, University of Melbourne, Austin Health, 145 Studley Rd., Heidelberg, VIC 3084, Australia
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2023, 15(2), 551; https://doi.org/10.3390/cancers15020551
Submission received: 14 December 2022 / Revised: 11 January 2023 / Accepted: 13 January 2023 / Published: 16 January 2023

Abstract

:

Simple Summary

Liver disorders, particularly liver cancer and hepatitis, are considered major public health challenges. Nearly 90% of patients with liver cancer are diagnosed at late stages of the disease, which reduces the treatment options, having a significant negative impact on patient survival. Hepatitis is one of major risk factors of liver cancer. The p21 Activated Kinases (PAKs) play important roles in various diseases, and thus become targets for therapeutical purpose. Their roles in liver disorders are emerging. This review highlights the importance of alteration of PAKs within liver disorders, emphasizes their effects on liver cancer and hepatitis, and discusses the potential therapeutical effect of PAKs in liver disorders. We aim to provide an overview on current progress of PAKs in liver diseases, and a useful guide to research into PAK inhibitors and the potential application.

Abstract

The p21 Activated Kinases (PAKs) are serine threonine kinases and play important roles in many biological processes, including cell growth, survival, cytoskeletal organization, migration, and morphology. Recently, PAKs have emerged in the process of liver disorders, including liver cancer, hepatic ischemia-reperfusion injury, hepatitis, and liver fibrosis, owing to their effects in multiple signaling pathways in various cell types. Activation of PAKs promotes liver cancer growth and metastasis and contributes to the resistance of liver cancer to radiotherapy and chemotherapy, leading to poor survival of patients. PAKs also play important roles in the development and progression of hepatitis and other pathological processes of the liver such as fibrosis and ischemia-reperfusion injury. In this review, we have summarized the currently available studies about the role of PAKs in liver disorders and the mechanisms involved, and further explored the potential therapeutic application of PAK inhibitors in liver disorders, with the aim to provide a comprehensive overview on current progress and perspectives of PAKs in liver disorders.

1. Introduction

The p21 Activated Kinases (PAKs) are serine-threonine kinases containing six members, and play important roles in a variety of biological processes such as cell growth, survival, cytoskeletal organization, migration, and morphology [1]. These kinases act downstream of the Ras-related Rho GTPases cell division control protein 42 (CDC42) and RAC, and are involved in multiple signaling pathways such as phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) and Wnt/β-catenin pathways [2,3]. PAKs can be divided into two groups based on structural and biological properties. Group I PAKs include PAK1-3, and group II PAKs contain PAK4-6. PAKs have been involved in many physiological and pathological processes. Their roles in cancer have been extensively investigated. Increasing evidence implicates PAK effects in liver disorders including liver cancer, hepatitis, liver fibrosis, and hepatic ischemia-reperfusion injury (IRI) [4,5,6,7]. In this review, we will discuss the role of PAKs in liver disorders with a focus on liver cancer and hepatitis and the underlying mechanisms, and we will explore the potential therapeutic effects of PAKs in liver disorders. The upstream regulators of PAKs and the roles of PAKs in liver disorders are summarized in Figure 1 and Table 1, respectively.

2. Structure and Activation of PAKs Family

PAK proteins contain a kinase domain at the C-terminal, and an autoinhibitory domain (AID) and a GTPase-binding domain (GBD) at the N-terminus [8]. The AID of Group I PAKs overlaps with the GBD and binds in trans to the kinase domain of another PAK molecule to form a dimer, preventing the formation of an activation loop. The binding of Rho GTPases or other proteins to the GBD dissociates the binding of AID with the kinase domain and causes a conformational change, which in turn triggers autophosphorylation of Ser 144 and Thr 423 and activation of the kinase. Group II PAKs function as monomers and they have AID-like domains different from the AID in group I PAKs [9]. There are two proposed models to explain the activation of group II PAKs. One model is the AID-like domain, which binds to the kinase domain in cis, forming an inactive conformation. When CDC42 or RAC binds to GBD, this conformation will change and release the kinase domain from auto-inhibition [10]. The other model contains two phases for activation. In addition to the binding of Rho GTPases, it also requires the involvement of proteins that contain SH3 domains, which interact with the AID-like domains [11] (Figure 2).
PAKs act in multiple signaling pathways that are crucial for physiological and pathological processes. Particularly, overexpression of PAKs in many types of cancers is correlated with cancer progression and therapeutical resistance. Both PAK1 and PAK4 can activate the PI3K/AKT pathway, one of the key pathways for cell growth and survival. PAKs promote chemoresistance of cancer cells through activating the PI3K/AKT pathway [12]. PAK1 stimulates cell migration by activation of AKT [13]. PAK4 bind to p85, the regulatory subunit of PI3K, which in turn increases the activity of PI3K and phosphorylation of AKT, thereby promoting the migration of cancer cells [14]. On the other hand, PI3K/AKT can also regulate the activation of PAKs. It has been suggested that a positive feedback loop may exist between PAKs and PI3K/AKT in the progression of cancers [15,16,17]. RAS/RAF/mitogen-activated protein kinase (MEK)/extracellular-regulated kinase (ERK) axis is a well-known pathway contributing to the growth, survival, and metabolism of cancer cells. PAKs can phosphorylate MEK and RAF at Ser298 and at Ser338, respectively, promoting the activation of RAS-mediated RAF/MEK/ERK signaling [18]. In IκB/nuclear factor-κB (NF-κB) signaling, PAK1 promoted the phosphorylation and degradation of IκB, leading to the activation and nuclear translocation of NF-κB [19]. Blockading PAK1-activated NF-κB inhibited the growth of HCC and alleviated the inflammatory response in acute pancreatitis [20,21]. PAKs can activate β-catenin signaling and stabilize the activation of β-catenin, contributing to the chemotherapy resistance of cancer [22,23,24]. PAKs can form a complex with β-catenin in the nucleus, which can further stabilize β-catenin. PAK4 can phosphorylate β-catenin at Ser 675, preventing the ubiquitination and degradation of β-catenin [25,26]. PAKs regulate the activation of Lin-11/Isl-1/Mec-3 kinase and matrix metalloproteinases, which are associated with cancer cell metastasis [27,28].
Except for their roles in cancers, alterations of PAKs are also implicated in other human diseases. By regulation of cytoskeleton dynamics, PAKs play key roles in brain development and thus neurological disorders [29]. Mutations in PAK1 are identified in intellectual disability associated with seizures and speech delay whereas mutations in PAK2 and PAK3 are found in autism and X-linked mental retardation. PAK1 regulates the phosphorylation of the proteins controlling heart rate and thus plays an important role in cardiac diseases [30]. PAK2 and PAK4 have been implicated in the dysfunction of pancreatic acinar [31]. PAK-mediated activation of Wnt and G-protein pathways have been reported in type 2 diabetes [32].

3. PAKs in Liver Cancers

Liver cancer is the sixth most common cancer worldwide and the second most common cause of cancer-related death. Hepatocellular carcinoma (HCC) accounts for 85% of liver cancer [33,34]. The major risk factors for liver cancer are chronic infection with hepatitis B virus (HBV) or hepatitis C virus (HCV) [34,35,36]. PAKs have been reported to be upregulated in many cancers including breast cancer, glioblastoma, pancreatic cancer, and HCC, promoting tumor growth and metastasis [7,37,38]. Increased PAKs expression endowed tumors with chemoresistance, and thus PAKs become attractive therapeutical targets for cancer treatment and biomarkers to predict prognosis [39,40,41]. The roles of PAKs in liver cancer, especially HCC, are discussed below (Figure 3).

3.1. Roles of Group I PAKs in Liver Cancer

PAK1 has been the most investigated among the I PAKs group. Overexpression of PAK1 was most frequently observed in HCC and associated with the growth and metastasis of HCC. PAK1 affects HCC progression via multiple pathways, PAK1 stimulated proliferation and inhibited apoptosis of HCC cells by activation of β-catenin signaling, and facilitated migration and invasion by upregulation of Snail [42]. Genetic inhibition of PAK1 suppressed xenograft tumor growth through Snail-, β-catenin-, and p53/p21-mediated pathways [42,43]. Treatment of phorbol ester increased PAK1, which in turn promoted HCC metastasis by activation of paxillin and c-Jun N-terminal kinase (JNK) [44]. PAK1 also promoted HCC metastasis by regulating the formation and degradation of actin stress fibers [45]. PAK1 enhanced vasculogenic mimicry in HCC through the stabilization of hypoxia-inducible factor-1α and phosphorylation of Vimentin [46]. Activation of PAK1-mediated epidermal growth factor receptor (EGFR) and vascular endothelial growth factor receptor (VEGFR) pathways inhibited anoikis, a form of programmed cell death, contributing to the anoikis resistance of HCC cells [47,48].
A natural product, resveratrol, inhibited HCC cell proliferation and survival by downregulation of PAK1, cyclin D1, mitogen-activated protein kinase (MAPK), and AKT [49]. In a liver carcinogenesis animal model, diethylnitrosamine-induced HCC progression was correlated with increased levels of PAK1, cyclin D1, MAPK, and AKT [35]. Myricetin induces apoptosis of HCC cells by abrogating RAS-activated PAK1 and inhibiting MAPK/ERK and PI3K/AKT signaling [50]. KRAS or BRAF mutations have been shown to promote hepatic vascular cavernomas [51]. Although PAK1 has been reported to affect the activation level of RAS/MEK/ERK signaling [52,53], the role of PAK1 in hepatic vascular cavernomas is unknown. The Smad4-MYO18A-PP1A complex has been reported to inhibit the proliferation, migration, invasion, and resistance to pemigatinib of cholangiocarcinoma through dephosphorylating PAK1 and suppressing subsequent intranuclear localization of β-catenin [22].
MicroRNAs (miRNAs) are small, non-coding RNAs that bind to specific mRNA targets to regulate gene expression post-transcriptionally. Altered expression of miRNAs is associated with liver metabolic dysfunction and tumor development. LINC00460, a long intergenic non-protein coding RNA, promoted HCC progression by stimulation of PAK1 through suppressing miR-485-5p [54]. Zhou and colleagues also reported that miR-15b was targeted by PAK1 to promote the proliferation, migration, and invasion of HCC cells [55].
HBV infection is a well-recognized risk factor for liver cancer. Induction of hepatitis B virus X protein to HCC cells promoted anchorage-independent growth and anoikis resistance by upregulation of PAK1, and knockdown of PAK1 inhibited tumor growth of HCC cells and their resistance to anoikis. More importantly, PAK1 expression is positively correlated to HBV infection, worse prognosis, and portal vein tumor thrombosis in patients [56].
PAK2 played a critical role in HCC metastasis. MiR-26a inhibited metastasis of HCC through downregulation of PAK2 [57]. Sato et al. reported that PAK2 phosphorylation was associated with AKT activation in transforming growth factor-β (TGF-β)-induced migration of HCC cells [58]. PAK3 is mainly expressed in the central nervous system for regulation of neuronal plasticity and spinogenesis [59]. However, Gao and colleagues recently found that PAK3 was upregulated in HCC and stimulated the epithelial mesenchymal transition (EMT) of tumor cells through activating the TGF-β/Smad pathway [60].

3.2. Roles of Group II PAKs in Liver Cancer

PAK4 enhanced HCC cell survival by modulating caspase-8 and NF-κB pathways [61]. In an HCC transgenic mouse model, miR-199-3p inhibited HCC growth by targeting the PAK4/RAF/MEK/ERK pathway [62,63]. On the contrary, an increased cyclin-dependent kinase 5 (CDK5) regulatory subunit-associated protein 3 in HCC accelerated tumor metastasis bound to and activated PAK4 [64]. PAK4 promoted migration and invasion of HCC cells through direct phosphorylation of p53 at S215 [65]. MiR-1271 suppressed HCC growth and metastasis through downregulation of RAF/MEK/ERK signaling, achieved by inhibiting Zic2/PAK4. PAK4 was overexpressed in metastatic tumor tissues compared with primary tumor and normal tissues, and PAK4 expression was correlated with worse survival of HCC patients. The results from multivariate analyses of 615 HCC patients showed that PAK4 was an independent indicator of poor prognosis [66]. MiR-433 was reduced in HCC tissues. MiR-433 repressed the viability of HCC cells through directly binding to the sequence at 3’-UTR of PAK4 mRNA, which in turn inhibited PAK4 expression [67]. MiRNAs are key upstream regulators that directly modulate the translation of multiple oncogenic proteins including PAK4. A miRNA cocktail therapy targeting PAK4, mechanistic target of rapamycin (mTOR), and RAS homolog gene family member C (RHOC), showed remarkable anti-HCC effects in patient-derived xenografts [68].
Immunohistochemical results from tumor and corresponding para-tumor tissues of 273 HCC patients showed that PAK5 was overexpressed in tumor tissues and strongly correlated with tumor progression and worse prognosis of HCC patients [69]. MiR-129 and miR-526a, which were downregulated in HCC tissues and cell lines, have been shown to suppress the proliferation and invasion of HCC cells through interfering the translation of PAK5 [70,71]. Gene silencing of PAK5 attenuated the proliferation and colony formation of HepG2 cells and inhibited the tumor progression in vivo [72]. Overexpression of PAK5 reduced the apoptosis induced by cisplatin in HCC cells [73]. PAK5 conferred HCC with chemoresistance to adriamycin and 5-fluorouracil by phosphorylating β-catenin and promoting its nuclear translocation, leading to the increased transcription of adenosine triphosphate-binding cassette subfamily B member 1 (ABCB1), a gene responsible for multidrug resistance [69]. Recently, it has been reported that miR-138-1-3p enhanced the sensitivity of HCC to sorafenib by inhibiting PAK5/β-catenin/ABCB1 axis [24]. Inhibition of PAK5 also sensitized HCC to radiotherapy by inducing cell apoptosis, cell cycle arrest, and DNA damage [74].
Elevated PAK6 was also found in HCC tissues compared with para-tumor tissues, and PAK6 expression was positively associated with the high proliferative ability of HCC and poor prognosis of patients [75]. A recent study indicated PAK6-regulated mitosis, through downregulation of Eg5, could repress PAK6 expression and in turn, inhibit PAK6 in hepatocarcinogenesis [76]. Contrarily, Liu et al. reported that PAK6 played a tumor suppressive role by showing that HCC patients with lower PAK6 had a higher tumor node metastasis stage and worse survival [77]. Further studies are necessary to explore the role of PAK6 in HCC.

4. PAKs in Hepatic Ischemia/Reperfusion Injury

Hepatic IRI commonly occurs in the process of hemorrhagic shock, liver surgery, and liver transplantation [78,79]. The underlying mechanisms of hepatic IRI relate to the elevated oxidative stress and activation of immune-metabolic responses, which are harmful to normal cellular structures and functions, and result in liver damage [80]. PAKs participate in the modulation of immune responses and inflammation [81,82], and PAKs have been reported to play a critical role in hepatic IRI [6,83].
Neuregulin-1/PAK1 axis was found to decrease IRI of liver grafts with or without steatosis through increasing vascular endothelial growth factor-α and insulin growth factor-1 levels, respectively [83]. PAK4 was upregulated in hepatic IRI in mice and humans to promote hepatic hypoxia-reoxygenation-induced damage through phosphorylating nuclear factor erythroid 2-related factor 2 and suppressing its transcriptional activity [6]. Genetic knockout or pharmacological inhibition of PAK4 reduced the inflammation and necrosis of hepatocytes [6]. These results suggest that PAK4 inhibition may protect the liver from IRI-induced damage. The roles of other PAK members in liver IRI have not been investigated.

5. PAKs in Hepatitis

Hepatitis is defined as inflammation of the liver, and its etiologies include virus infection, parasitic invasion, alcohol abuse, autoimmunity imbalance, and metabolic disorder. The main characteristic of hepatitis is represented by the infiltration of inflammatory cells, which induces apoptosis and necrosis of hepatocytes leading to liver damage and dysfunctions [84,85,86,87]. The role of PAKs in hepatitis, especially virus-related ones, are discussed here.
In hepatitis B, phosphorylation of PAK1 induced the translocation of RAF-1 to mitochondrial, which in turn facilitated the anti-apoptotic effect of HBV X protein in hepatocyte [88]. In fact, HBV X protein can activate PAK1, which protected HCC from anoikis and promoted the metastasis of HCC [56]. Results from the PCR showed that the PAK3 gene was the preferential integration site of HBV DNA and the integration potentially changed the expression of PAK3 [5]. Yu and colleagues conducted a prospective nested case-control study and analyzed the peripheral blood samples of enrolled 240 HBeAg seropositive chronic hepatitis B patients treated with interferon-α (IFN-α). They have found that patients with genotype TT of rs9676717 in PAK4 gene were more sensitive to IFN-α therapy (p = 0.015) [89]. However, there were few associations between polymorphisms in PAK4 gene and susceptibility to HBV-related HCC [90].
Different from the promoting role of PAK1 in HBV, PAK1 had potentially inhibited HCV replication, and its antiviral effects were independent of interferon regulatory factor 3 but dependent on the mTOR-activated PI3K/AKT and ERK [91]. HCV protected the infected hepatocytes from cell death by suppressing apoptosis and inflammatory reaction partially through the upregulation of PAK2 [92]. It was reported that quasi-enveloped mammalian hepatitis E virus infection relied on the regulation of the CDC42-associated PAK1/non-myosin IIA/Cofilin pathway, which regulated F-actin polymerization and membrane remodelling [93,94].
In a mouse model infected with Schistosoma japonicum, PAK1 in Kupffer cells was elevated and facilitated the differentiation of CD4+ T cells to T helper 17 cells via the NF-κB/interferon regulatory factor 1/interleukin-6 pathway, leading to aggravated hepatic inflammation [95]. Increased PAK1 expression in Kupffer cells was also noticed in patients with autoimmune hepatitis, and the PAK1 expression was found to be associated with disease progression [96]. The increased expression of PAK6 found in alcoholic hepatitis may contribute to tumorigenesis [97].

6. PAKs in Liver Fibrosis

Liver fibrosis is implicated in the interaction between injured hepatocytes, inflammatory cells, and hepatic myofibroblasts. In response to injury, hepatocytes will express more profibrogenic miRNAs and proteins such as TGFβ and Notch, thus initiating fibrosis [98]. The transformation from hepatic stellate cells to myofibroblasts that produce extracellular matrix (ECM) provides a protection to liver tissue from injury, and there is a balance between ECM production and degradation. However, the excessive accumulation of ECM produced by consistently activated myofibroblasts will impair the normal physiological structure and function of the liver, resulting in the occurrence of liver fibrosis [99].
PAK1 and PAK3 were upregulated in activated hepatic myofibroblasts and promoted fibrotic effects by enhancing the expression of integrin β-1, which is essential for myofibroblasts activation and ECM production [100]. Chen et al. reported that Kolotho expression was significantly associated with liver fibrosis in HCC patients, contributing to the resistance of HCC cells to anoikis by activating VEGFR2/PAK1 signaling [47]. Using a co-culture system and a CCl4-induced fibrosis mouse model, Zhou et al. revealed that human umbilical cord mesenchymal stem cells inactivated LX-2 cells and consequently reduced liver fibrosis through increasing miR-455-3p-mediated inhibition on PAK2 translation [4]. It was also reported that Atg7-deficient mice exhibited perisinusoidal/pericellular fibrosis in liver with age, and PAK4 may act as a crucial mediator in this pathological process [101].

7. Therapeutic Effects of PAK Inhibitors in Liver Disorders

The main strategy for treating HBV or HCV-related liver diseases is long-term administration of antiviral drugs such as entecavir, disoproxil, and IFN-α. Different from HCV, HBV has the characteristic of escaping innate immunity, thus immune therapy is also a vital treatment for HBV [102,103]. For metabolism-related liver diseases, limiting alcohol consumption, changing eating patterns and diet composition, and reducing insulin resistance and improving lipid metabolism are effective regimens [104,105].
Liver resection and transplantation are considered as curative treatments for HCC. However, only a small proportion of patients can have these surgical operations at the diagnosis stage and the recurrence after surgery is high. Most HCC patients are diagnosed at advanced stages of the disease and can only have non-surgical treatments, including chemo-, radio-, and immune-therapies. The first-line therapy for advanced HCC includes broad-spectrum tyrosine kinase inhibitors, such as sorafenib and lenvatinib, and combinational therapies of immunotherapy and anti-angiogenesis therapy [106]. Sorafenib and the newly approved lenvatinib are oral multi-tyrosine kinase inhibitors, and there was no significant difference between the two inhibitors in improving the overall survival of unresectable HCC patients (13.6 versus 12.3 months) [107]. The combination of immunotherapy of programmed cell death-ligand 1 (PD-L1) blockade and anti-angiogenesis therapy of VEGF blockade has been approved as one of best available first-line treatments [106]. Although these strategies provide an extension in survival, they cause broad spectrum toxic side effects, and patients eventually develop therapy resistance. To overcome therapy resistance and in searching for alternative treatment, gene therapy emerges as a promising remedy for HCC, especially with gene mutation [108].
PAKs play key roles in liver disorders and thus are considered useful targets for the treatment of liver diseases. PAK inhibitors have been developed and can be divided into two categories depending on the binding sites within PAK molecules, ATP-competitive inhibitors, and allosteric PAK inhibitors [109]. ATP-competitive PAK inhibitors block PAK phosphorylation by targeting the ATP-binding pocket within the kinase domain. This type of PAK inhibitors is further classified into aminopyrazole-based inhibitors, aminopyrimidine-based inhibitors, indolocarbazole-based inhibitors, 2-amino pyrido[2,3-d] pyrimidine-7(8H)-one-based inhibitors, and other ATP-competitive inhibitors. Compared with ATP-competitive PAK inhibitors, allosteric PAK inhibitors have the potential to be more selective and display discriminative inhibitory activity among PAK family proteins [110,111].
The studies of the effects of PAK inhibitors in liver disorders are limited (Figure 4). PAK4 was upregulated and played a critical role in the process of hepatic IRI. Application of an ATP-competitive PAK4 inhibitor ND201651 reduced hepatic IRI [6]. Another novel specific PAK4 inhibitor, SPA7012, a pyrazolo [3,4-d] pyrimidine derivative, also showed an anti-inflammatory effect on the liver during hepatic IRI [112]. More experimental evidence is necessary for its clinical application.
IPA-3 is a non-ATP-competitive allosteric inhibitor targeting PAK1 phosphorylation. Since group I PAKs share the same inhibitory domain in the N-terminal sequence, IPA-3 also suppresses the activation of PAK2 and PAK3 [113]. Administration of IPA-3 remarkably improved the bile duct ligation and CCl4-induced fibrosis in mice without hepatotoxicity [100]. IPA-3 suppressed HCC growth by inhibition of PAK1-mediated JNK phosphorylation and NF-κB intranuclear localization [21]. HCC patients with high Klotho expression showed a poor overall survival. IPA-3 could sensitize HCC cells to anoikis by blocking the Klotho/VEGFR2/PAK1 axis, thus inhibiting HCC progression [47].
Many PAK inhibitors have been developed because of the critical roles of PAKs in various disease conditions. The effects and mechanisms of these inhibitors have been reviewed elsewhere. Although the efficacy of PAK inhibitors has been tested in many preclinical models, none has been successfully tested through a clinical trial. KPT-9274, a PAK4 and nicotinamide phosphoribosyltransferase (NAMPT) dual inhibitor, is the only one under phase I clinical trials (NCT02702492 and NCT04281420) for solid tumors and non-Hodgkin’s lymphoma [114,115,116]. Recently, CP734, a PAK1 inhibitor, was reported to suppress pancreatic cancer growth in mouse [117]. PAKib, a newly identified PAK4 inhibitor, was also found to suppress pancreatic cancer in a mouse model [118]. The efficacy of these newly developed PAK inhibitors in liver disorders are to be tested.

8. Discussion

The initiation and progression of liver disorders are accompanied by dysregulation of glucolipid metabolism, as the liver plays a central role in glucose, lipid, and energy metabolism [119]. Therefore, it is necessary to discuss the potential role of PAKs in glucolipid metabolism of the liver to further explore the mechanisms involved in the roles of PAKs in liver disorders.
Immunotherapy in cancer treatment has achieved great success. The application of immune checkpoint inhibitors has brought significant changes in the management of HCC, which is one of the major disorders in the liver and the 16th leading cause of death worldwide [120]. Thus, it is important to extend our discussion here to the possible effects of PAKs on the immunotherapy of liver disorders.

8.1. Potential Role of PAKs in Glucolipid Metabolism of Liver

Compared with normal hepatocytes, glucose transporters are highly expressed in HCC cells for the increased requirement of glucose for sustaining the rapid growth of tumors. PAK1 plays a critical role in glucose production. Aged Pak1−/− mice showed impaired pyruvate tolerance and glucagon-like peptide-1 secretion response, resulting increased glucose disposal in the liver [121]. Fatty acid is another major energy source for HCC cells, especially under poor nutrient supply, and the proteins responsible for fatty acid uptake, transport, and synthesis are upregulated in HCC [122]. AMP-activated protein kinase and mTOR signaling are vital for maintaining the balance of glucolipid metabolism in the liver [123,124]. The activation of the PI3K/AKT/mTOR pathway enhanced lipid synthesis, thereby exacerbating the liver steatosis and promoting HCC progression [125]. The potential role of PAKs in the lipid metabolism of the liver has also been implicated. PAK1 and PAK2 in the liver can be affected by fatty acid and phospholipids. For example, the unsaturated fatty acid oleate was found to stimulate PAK1 activity, and a low-dose cardiolipin also promoted the autophosphorylation of PAK1 and PAK2 [126,127]. The autophosphorylation of PAK2 further facilitated the phosphorylation of histone H2B [127]. PAK4 in fetal hepatocytes was implicated in the sexual dimorphism of metabolic disease through the regulation of de novo lipogenesis [128]. The promoting role of PAK4 in glucose intake and lipid biosynthesis in cancer cells has also been reported [129].
Nonalcoholic fatty liver disease (NAFLD) is characterized by abnormal lipid accumulation and comprises fatty liver and non-alcoholic steatohepatitis. NAFLD is the most prevalent liver disorder and recognized as a metabolic syndrome in the liver [130]. NAFLD is one of the primary risk factors for end-stage liver diseases including liver fibrosis/cirrhosis and HCC. Global prevalence of NAFLD is 25.24% and still on the rise, and the affected populations tend to be younger [130,131,132]. Given the role of PAKs in liver fibrosis and HCC, more closed studies are necessary to explore the association between PAKs and NAFLD and the mechanisms involved. Patients with NAFLD are more intolerant to IRI because of the overproduction of reactive oxygen species and disorder of scavenging mechanisms in the liver, thus present with more complications after surgery or liver transplantation [133,134]. IRI occurred in hepatectomy and increased the risk of HCC recurrence in the fatty liver [135]. The utilization of fatty donor liver during transplantation was associated with worse prognosis of patients [136]. Therefore, it is becoming important to examine the role of PAKs in IRI of NAFLD as well as the efficacy of PAKs intervention.

8.2. Potential Role of PAKs in Immunotherapy for Liver Disorders

The tumor microenvironment plays key roles in the regulation of immune activation or evasion driven by the opposing actions of antitumor effectors and immunosuppressive components. It is worth noticing that the key cells with immunosuppressive roles implicated in HCC immune evasion include Kupffer cells, monocyte-derived macrophages, regulatory T cells and myeloid-derived suppressive cells [137]. Alteration of PAK1 in Kupffer cells in autoimmune hepatitis implicates the potential immune regulation of PAK in liver diseases [96]. Recently, PAK effects on tumor microenvironment and immune response have been reported. Depletion of PAK1 enhanced the immune system and inhibited intestinal tumorigenesis in a mouse model of intestinal cancer [138]. Inhibition of PAK1 also stimulated the intra-tumoral CD4+ and CD8+ T cells and suppressed pancreatic cancer [82]. More recently, it has been reported in melanoma that PAK4 expression was elevated in the patients’ samples with poor infiltrated immune cells and thus decreased the response to a PD-1 blockade, and that knockout of PAK4 enhanced the infiltration of CD8+ T cells, possibly by downregulation of PD-L1 [139]. These reports indicated the important role of PAKs in tumor immune response and the tumor microenvironment. Although the effects of PAKs on liver cancer have been widely studied, the impact of PAKs on immune response in the tumor microenvironment of liver cancer is rarely reported. The role of PAKs in liver cancer immune microenvironment deserves more investigation.

9. Conclusions

Increasing evidence has shown that PAKs play pivotal roles in the progression of liver disorders including liver cancer, hepatic IRI, hepatitis, and liver fibrosis. Elevated PAKs expression in liver cancer stimulates tumor growth and metastasis, contributes to therapeutical resistance, and is associated with the poor prognosis of patients. The studies of the effects of PAK1 and PAK4 in hepatic IRI have provided a useful guide for the potential application of PAK inhibitors in clinical settings. PAK1, the most extensively studied PAKs, plays important roles in the occurrence of hepatitis and liver fibrosis through affecting hepatocytes, immune cells, and myofibroblasts. It is worth noting that PAK1 promotes the development of HBV, hepatitis E virus, parasite, and autoimmune-related hepatitis while inhibiting HCV-induced hepatitis. With the increased occurrence of metabolism-related liver disease such as NAFLD, it is becoming important to explore the association between PAKs and liver metabolism and mechanisms involved. As our knowledge of PAKs and liver disorders accumulates, the efficacy of PAK inhibitors in liver disorders will be explored, which will provide a useful guide in clinical applications.

Author Contributions

Conceptualization, H.H., X.Q. and K.W.; investigation, H.H., X.Q. and H.X.; writing—original draft preparation, X.Q., H.X. and F.G.; writing—review and editing, H.H., X.Q. and K.W.; supervision, H.H. and X.X.; project administration, H.H. and X.X.; funding acquisition, K.W., H.H. and X.X. All authors have read and agreed to the published version of the manuscript.

Funding

This study was funded by Zhejiang Provincial Natural Science Foundation, Grant LY22H160048; the Youth Program of the National Natural Science Foundation of China, Grant 82003248; the Key Research & Development Plan of Zhejiang Province, Grant 2019C03050; and the Major Research Plan of the National Natural Science Foundation of China, Grant 92159202.

Acknowledgments

The authors would like to thank the Henry Baldwin Cancer Research Trust Fund and the Construction Fund of Key Medical Disciplines of Hangzhou (OO20200093) for the financial support. Figures are created with BioRender.com.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ABCB1, adenosine triphosphate-binding cassette subfamily B member 1; AID, autoinhibitory domain; AKT, protein kinase B; CDC42, cell division control protein 42; CDK5RAP3, cyclin-dependent kinase 5 kinase regulatory subunit-associated protein 3; ECM, extracellular matrix; EGFR, epidermal growth factor receptor; ERK, extracellular-regulated kinase; GBD, GTPase-binding domain; HBV, hepatitis B virus; HCC, hepatocellular carcinoma; HCV, hepatitis C virus; IFN-α, interferon-α; IRI, ischemia-reperfusion injury; JNK, c-Jun N-terminal kinase; MAPK, mitogen-activated protein kinase; MEK, mitogen-activated protein kinase; Mgat5, N-acetylglucosaminyltransferase V; mTOR, mechanistic target of rapamycin; NAFLD, nonalcoholic fatty liver disease; NF-κB, nuclear factor-κB; NRG-1, neuregulin-1; PAKs, p21 Activated Kinases; PD-L1, programmed cell death-ligand 1; PI3K, phosphoinositide 3-kinase; ROS, reactive oxygen species; TGF-β, transforming growth factor-β; VEGFR, vascular endothelial growth factor receptor.

References

  1. Rane, C.K.; Minden, A. P21 activated kinase signaling in cancer. Semin. Cancer Biol. 2019, 54, 40–49. [Google Scholar] [CrossRef] [PubMed]
  2. Fruman, D.A.; Chiu, H.; Hopkins, B.D.; Bagrodia, S.; Cantley, L.C.; Abraham, R.T. The PI3K Pathway in Human Disease. Cell 2017, 170, 605–635. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Li, Q.; Sun, M.; Wang, M.; Feng, M.; Yang, F.; Li, L.; Zhao, J.; Chang, C.; Dong, H.; Xie, T.; et al. Dysregulation of Wnt/β-catenin signaling by protein kinases in hepatocellular carcinoma and its therapeutic application. Cancer Sci. 2021, 112, 1695–1706. [Google Scholar] [CrossRef]
  4. Zhou, Q.; Gu, T.; Zhang, Y.; Li, H.; Zhuansun, X.; Xu, S.; Kuang, Y. Human Umbilical Cord Mesenchymal Stem Cells Ameliorate Hepatic Stellate Cell Activation and Liver Fibrosis by Upregulating MicroRNA-455-3p through Suppression of p21-Activated Kinase-2. BioMed Res. Int. 2021, 2021, 6685605. [Google Scholar] [CrossRef] [PubMed]
  5. Ruan, P.; Dai, X.; Sun, J.; He, C.; Huang, C.; Zhou, R.; Chemin, I. Integration of hepatitis B virus DNA into p21-activated kinase 3 (PAK3) gene in HepG2.2.15 cells. Virus Genes 2020, 56, 168–173. [Google Scholar] [CrossRef] [PubMed]
  6. Mao, Y.; Han, C.Y.; Hao, L.; Lee, Y.; Son, J.B.; Choi, H.; Lee, M.R.; Yang, J.D.; Hong, S.K.; Suh, K.S.; et al. p21-activated kinase 4 inhibition protects against liver ischemia/reperfusion injury: Role of nuclear factor erythroid 2-related factor 2 phosphorylation. Hepatology 2022, 76, 345–356. [Google Scholar] [CrossRef] [PubMed]
  7. Tse, E.Y.; Ching, Y.P. The role of p21-activated kinases in hepatocellular carcinoma metastasis. J. Mol. Signal. 2014, 9, 7. [Google Scholar] [CrossRef] [Green Version]
  8. Rane, C.K.; Minden, A. P21 activated kinases: Structure, regulation, and functions. Small GTPases 2014, 5, e28003. [Google Scholar] [CrossRef] [Green Version]
  9. Liu, H.; Liu, K.; Dong, Z. The Role of p21-Activated Kinases in Cancer and Beyond: Where Are We Heading? Front. Cell Dev. Biol. 2021, 9, 41381. [Google Scholar] [CrossRef]
  10. Baskaran, Y.; Ng, Y.; Selamat, W.; Ling, F.T.P.; Manser, E. Group I and II mammalian PAKs have different modes of activation by Cdc42. EMBO Rep. 2012, 13, 653–659. [Google Scholar] [CrossRef]
  11. Ha, B.H.; Davis, M.J.; Chen, C.; Lou, H.J.; Gao, J.; Zhang, R.; Krauthammer, M.; Halaban, R.; Schlessinger, J.; Turk, B.E.; et al. Type II p21-activated kinases (PAKs) are regulated by an autoinhibitory pseudosubstrate. Proc. Natl. Acad. Sci. USA 2012, 109, 16107–16112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Chan, C.H.; Chiou, L.W.; Lee, T.Y.; Liu, Y.R.; Hsieh, T.H.; Yang, C.Y.; Jeng, Y.M. PAK and PI3K pathway activation confers resistance to KRAS(G12C) inhibitor sotorasib. Br. J. Cancer 2023, 128, 148–159. [Google Scholar] [CrossRef] [PubMed]
  13. Higuchi, M.; Onishi, K.; Kikuchi, C.; Gotoh, Y. Scaffolding function of PAK in the PDK1–Akt pathway. Nature 2008, 10, 1356–1364. [Google Scholar] [CrossRef] [PubMed]
  14. King, H.; Thillai, K.; Whale, A.; Arumugam, P.; Eldaly, H.; Kocher, H.M.; Wells, C.M. PAK4 interacts with p85 alpha: Implications for pancreatic cancer cell migration. Sci. Rep. 2017, 7, 2575. [Google Scholar] [CrossRef] [Green Version]
  15. Park, S.Y.; An, J.M.; Seo, J.T.; Seo, S.R. Y-27632 Induces Neurite Outgrowth by Activating the NOX1-Mediated AKT and PAK1 Phosphorylation Cascades in PC12 Cells. Int. J. Mol. Sci. 2020, 21, 7679. [Google Scholar] [CrossRef]
  16. Fu, X.; Feng, J.; Zeng, D.; Ding, Y.; Yu, C.; Yang, B. PAK4 confers cisplatin resistance in gastric cancer cells via PI3K/Akt- and MEK/ERK-dependent pathways. Biosci. Rep. 2014, 34, e00094. [Google Scholar] [CrossRef]
  17. Huang, N.; Li, S.; Xie, Y.; Han, Q.; Xu, X.M.; Sheng, Z.H. Reprogramming an energetic AKT-PAK5 axis boosts axon energy supply and facilitates neuron survival and regeneration after injury and ischemia. Curr. Biol. 2021, 31, 3098–3114. [Google Scholar] [CrossRef]
  18. Beeser, A.; Jaffer, Z.M.; Hofmann, C.; Chernoff, J. Role of Group A p21-activated Kinases in Activation of Extracellular-regulated Kinase by Growth Factors. J. Biol. Chem. 2005, 280, 36609–36615. [Google Scholar] [CrossRef] [Green Version]
  19. Frost, J.A.; Swantek, J.L.; Stippec, S.; Yin, M.J.; Gaynor, R.; Cobb, M.H. Stimulation of NFkappa B activity by multiple signaling pathways requires PAK1. J. Biol. Chem. 2000, 275, 19693–19699. [Google Scholar] [CrossRef] [Green Version]
  20. Zhu, M.; Xu, Y.; Zhang, W.; Gu, T.; Wang, D. Inhibition of PAK1 alleviates cerulein-induced acute pancreatitis via p38 and NF-κB pathways. Biosci. Rep. 2019, 39, BSR20182221. [Google Scholar] [CrossRef]
  21. Wong, L.L.; Lam, I.P.; Wong, T.Y.; Lai, W.L.; Liu, H.F.; Yeung, L.L.; Ching, Y.P. IPA-3 inhibits the growth of liver cancer cells by suppressing PAK1 and NF-κB activation. PLoS ONE 2013, 8, e68843. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Liu, J.; Ren, G.; Li, K.; Liu, Z.; Wang, Y.; Chen, T.; Mu, W.; Yang, X.; Li, X.; Shi, A.; et al. The Smad4-MYO18A-PP1A complex regulates β-catenin phosphorylation and pemigatinib resistance by inhibiting PAK1 in cholangiocarcinoma. Cell Death Differ. 2022, 29, 818–831. [Google Scholar] [CrossRef]
  23. Yi, Y.; Li, P.; Huang, Y.; Chen, D.; Fan, S.; Wang, J.; Yang, M.; Zeng, S.; Deng, J.; Lv, X.; et al. P21-activated kinase 2-mediated β-catenin signaling promotes cancer stemness and osimertinib resistance in EGFR-mutant non-small-cell lung cancer. Oncogene 2022, 41, 4318–4329. [Google Scholar] [CrossRef] [PubMed]
  24. Li, T.T.; Mou, J.; Pan, Y.J.; Huo, F.C.; Du, W.Q.; Liang, J.; Wang, Y.; Zhang, L.S.; Pei, D.S. MicroRNA-138-1-3p sensitizes sorafenib to hepatocellular carcinoma by targeting PAK5 mediated β-catenin/ABCB1 signaling pathway. J. Biomed. Sci. 2021, 28, 56. [Google Scholar] [CrossRef]
  25. Li, Y.; Shao, Y.; Tong, Y.; Shen, T.; Zhang, J.; Li, Y.; Gu, H.; Li, F. Nucleo-cytoplasmic shuttling of PAK4 modulates β-catenin intracellular translocation and signaling. Biochim. Biophys. Acta (BBA)—Mol. Cell Res. 2012, 1823, 465–475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Vershinin, Z.; Feldman, M.; Chen, A.; Levy, D. PAK4 Methylation by SETD6 Promotes the Activation of the Wnt/β-Catenin Pathway. J. Biol. Chem. 2016, 291, 6786–6795. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Bastea, L.I.; Döppler, H.; Pearce, S.E.; Durand, N.; Spratley, S.J.; Storz, P. Protein kinase D-mediated phosphorylation at Ser99 regulates localization of p21-activated kinase 4. Biochem. J. 2013, 455, 251–260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Wang, J.; Hirose, H.; Du, G.; Chong, K.; Kiyohara, E.; Witz, I.P.; Hoon, D.S.B. P-REX1 amplification promotes progression of cutaneous melanoma via the PAK1/P38/MMP-2 pathway. Cancer Lett. 2017, 407, 6–75. [Google Scholar] [CrossRef]
  29. Zhang, K.; Wang, Y.; Fan, T.; Zeng, C.; Sun, Z.S. The p21-activated kinases in neural cytoskeletal remodeling and related neurological disorders. Protein Cell 2022, 13, 6–25. [Google Scholar] [CrossRef]
  30. Wang, Y.; Wang, S.; Lei, M.; Boyett, M.; Tsui, H.; Liu, W.; Wang, X. The p21-activated kinase 1 (Pak1) signalling pathway in cardiac disease: From mechanistic study to therapeutic exploration. Br. J. Pharmacol. 2017, 175, 1362–1374. [Google Scholar] [CrossRef]
  31. Ramos-Alvarez, I.; Jensen, R.T. P21-activated kinase 4 in pancreatic acinar cells is activated by numerous gastrointestinal hormones/neurotransmitters and growth factors by novel signaling, and its activation stimulates secretory/growth cascades. Am. J. Physiol. Gastrointest. Liver Physiol. 2018, 315, G302–G317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Dammann, K.; Khare, V.; Coleman, C.; Berdel, H.; Gasche, C. p-21 Activated Kinase as a Molecular Target for Chemoprevention in Diabetes. Geriatrics 2018, 3, 73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. El-Serag, H.B.; Rudolph, K.L. Hepatocellular carcinoma: Epidemiology and molecular carcinogenesis. Gastroenterology 2007, 132, 2557–2576. [Google Scholar] [CrossRef]
  34. McGlynn, K.A.; London, W.T. The global epidemiology of hepatocellular carcinoma: Present and future. Clin. Liver Dis. 2011, 15, 223–243. [Google Scholar] [CrossRef] [Green Version]
  35. Parekh, P.; Rao, K.V. Overexpression of cyclin D1 is associated with elevated levels of MAP kinases, Akt and Pak1 during diethylnitrosamine-induced progressive liver carcinogenesis. Cell Biol. Int. 2007, 31, 35–43. [Google Scholar] [CrossRef]
  36. Razavi, H.; Waked, I.; Sarrazin, C.; Myers, R.P.; Idilman, R.; Calinas, F.; Vogel, W.; Mendes Correa, M.C.; Hézode, C.; Lázaro, P.; et al. The present and future disease burden of hepatitis C virus (HCV) infection with today’s treatment paradigm. J. Viral Hepat. 2014, 21 (Suppl. 1), 4–59. [Google Scholar] [CrossRef]
  37. Best, M.; Gale, M.E.; Wells, C.M. PAK-dependent regulation of actin dynamics in breast cancer cells. Int. J. Biochem. Cell Biol. 2022, 146, 06207. [Google Scholar] [CrossRef] [PubMed]
  38. Wang, M.; Zhang, C.; Zheng, Q.; Ma, Z.; Qi, M.; Di, G.; Ling, S.; Xu, H.; Qi, B.; Yao, C.; et al. RhoJ facilitates angiogenesis in glioblastoma via JNK/VEGFR2 mediated activation of PAK and ERK signaling pathways. Int. J. Biol. Sci. 2022, 18, 942–955. [Google Scholar] [CrossRef] [PubMed]
  39. Lei, K.; Luo, M.; Tu, Z.; Lv, S.; Liu, J.; Gong, C.; Ye, M.; Wu, M.; Sheng, Y.; Long, X.; et al. Comprehensive analysis of the prognostic implications and functional exploration of PAK gene family in human cancer. Cancer Cell Int. 2022, 22, 275. [Google Scholar] [CrossRef]
  40. Yu, X.; Huang, C.; Liu, J.; Shi, X.; Li, X. The significance of PAK4 in signaling and clinicopathology: A review. Open Life Sci. 2022, 17, 586–598. [Google Scholar] [CrossRef]
  41. Kumar, R.; Li, D.Q. PAKs in Human Cancer Progression: From Inception to Cancer Therapeutic to Future Oncobiology. Adv. Cancer Res. 2016, 130, 37–209. [Google Scholar] [CrossRef]
  42. Cao, F.; Yin, L.X. PAK1 promotes proliferation, migration and invasion of hepatocellular carcinoma by facilitating EMT via directly up-regulating Snail. Genomics 2020, 112, 694–702. [Google Scholar] [CrossRef] [PubMed]
  43. Zhang, Z.-L.; Liu, G.-C.; Peng, L.; Zhang, C.; Jia, Y.-M.; Yang, W.-H.; Mao, L. Effect of PAK1 gene silencing on proliferation and apoptosis in hepatocellular carcinoma cell lines MHCC97-H and HepG2 and cells in xenograft tumor. Gene Ther. 2018, 25, 284–296. [Google Scholar] [CrossRef]
  44. Ching, Y.P.; Leong, V.Y.; Lee, M.F.; Xu, H.T.; Jin, D.Y.; Ng, I.O. P21-activated protein kinase is overexpressed in hepatocellular carcinoma and enhances cancer metastasis involving c-Jun NH2-terminal kinase activation and paxillin phosphorylation. Cancer Res. 2007, 67, 3601–3608. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Gujdár, A.; Sipeki, S.; Bander, E.; Buday, L.; Faragó, A. Phorbol ester-induced migration of HepG2 cells is accompanied by intensive stress fibre formation, enhanced integrin expression and transient down-regulation of p21-activated kinase 1. Cell. Signal. 2003, 15, 307–318. [Google Scholar] [CrossRef]
  46. Zhang, J.G.; Zhou, H.M.; Zhang, X.; Mu, W.; Hu, J.N.; Liu, G.L.; Li, Q. Hypoxic induction of vasculogenic mimicry in hepatocellular carcinoma: Role of HIF-1 α, RhoA/ROCK and Rac1/PAK signaling. BMC Cancer 2020, 20, 32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Chen, L.; Liu, H.; Liu, J.; Zhu, Y.; Xu, L.; He, H.; Zhang, H.; Wang, S.; Wu, Q.; Liu, W.; et al. Klotho endows hepatoma cells with resistance to anoikis via VEGFR2/PAK1 activation in hepatocellular carcinoma. PLoS ONE 2013, 8, e58413. [Google Scholar] [CrossRef] [Green Version]
  48. Liu, J.; Liu, H.; Zhang, W.; Wu, Q.; Liu, W.; Liu, Y.; Pan, D.; Xu, J.; Gu, J. N-acetylglucosaminyltransferase V confers hepatoma cells with resistance to anoikis through EGFR/PAK1 activation. Glycobiology 2013, 23, 1097–1109. [Google Scholar] [CrossRef]
  49. Parekh, P.; Motiwale, L.; Naik, N.; Rao, K.V. Downregulation of cyclin D1 is associated with decreased levels of p38 MAP kinases, Akt/PKB and Pak1 during chemopreventive effects of resveratrol in liver cancer cells. Exp. Toxicol. Pathol. 2011, 63, 67–173. [Google Scholar] [CrossRef]
  50. Iyer, S.C.; Gopal, A.; Halagowder, D. Myricetin induces apoptosis by inhibiting P21 activated kinase 1 (PAK1) signaling cascade in hepatocellular carcinoma. Mol. Cell. Biochem. 2015, 407, 223–237. [Google Scholar] [CrossRef]
  51. Janardhan, H.P.; Meng, X.; Dresser, K.; Hutchinson, L.; Trivedi, C.M. KRAS or BRAF mutations cause hepatic vascular cavernomas treatable with MAP2K-MAPK1 inhibition. J. Exp. Med. 2020, 217, e20192205. [Google Scholar] [CrossRef]
  52. Chen, L.; Bi, S.; Hou, J.; Zhao, Z.; Wang, C.; Xie, S. Targeting p21-activated kinase 1 inhibits growth and metastasis via Raf1/MEK1/ERK signaling in esophageal squamous cell carcinoma cells. Cell Commun. Signal. 2019, 17, 31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Cheng, C.; Kong, X.; Wang, H.; Gan, H.; Hao, Y.; Zou, W.; Wu, J.; Chi, Y.; Yang, J.; Hong, Y.; et al. Trihydrophobin 1 Interacts with PAK1 and Regulates ERK/MAPK Activation and Cell Migration. J. Biol. Chem. 2009, 284, 8786–8796. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Tu, J.; Zhao, Z.; Xu, M.; Chen, M.; Weng, Q.; Ji, J. LINC00460 promotes hepatocellular carcinoma development through sponging miR-485-5p to up-regulate PAK1. Biomed. Pharmacother. 2019, 118, 109213. [Google Scholar] [CrossRef] [PubMed]
  55. Zhou, Y.; Fan, R.G.; Qin, C.L.; Jia, J.; Wu, X.D.; Zha, W.Z. LncRNA-H19 activates CDC42/PAK1 pathway to promote cell proliferation, migration and invasion by targeting miR-15b in hepatocellular carcinoma. Genomics 2019, 111, 1862–1872. [Google Scholar] [CrossRef]
  56. Xu, J.; Liu, H.; Chen, L.; Wang, S.; Zhou, L.; Yun, X.; Sun, L.; Wen, Y.; Gu, J. Hepatitis B Virus X Protein Confers Resistance of Hepatoma Cells to Anoikis by Up-regulating and Activating p21-Activated Kinase 1. Gastroenterology 2012, 143, 199–212.e4. [Google Scholar] [CrossRef]
  57. Zhang, X.; Zhang, X.; Wang, T.; Wang, L.; Tan, Z.; Wei, W.; Yan, B.; Zhao, J.; Wu, K.; Yang, A.; et al. MicroRNA-26a is a key regulon that inhibits progression and metastasis of c-Myc/EZH2 double high advanced hepatocellular carcinoma. Cancer Lett. 2018, 426, 8–108. [Google Scholar] [CrossRef]
  58. Sato, M.; Matsuda, Y.; Wakai, T.; Kubota, M.; Osawa, M.; Fujimaki, S.; Sanpei, A.; Takamura, M.; Yamagiwa, S.; Aoyagi, Y. p21-activated kinase-2 is a critical mediator of transforming growth factor-β-induced hepatoma cell migration. J. Gastroenterol. Hepatol. 2013, 28, 1047–1055. [Google Scholar] [CrossRef]
  59. Koth, A.P.; Oliveira, B.R.; Parfitt, G.M.; Buonocore Jde, Q.; Barros, D.M. Participation of group I p21-activated kinases in neuroplasticity. J. Physiol. 2014, 108, 70–277. [Google Scholar] [CrossRef]
  60. Gao, Z.; Zhong, M.; Ye, Z.; Wu, Z.; Xiong, Y.; Ma, J.; Chen, H.; Zhu, Y.; Yang, Y.; Zhao, Y.; et al. PAK3 promotes the metastasis of hepatocellular carcinoma by regulating EMT process. J. Cancer 2022, 13, 153–161. [Google Scholar] [CrossRef]
  61. Li, Q.; Zhang, X.; Wei, N.; Liu, S.; Ling, Y.; Wang, H. p21-activated kinase 4 as a switch between caspase-8 apoptosis and NF-κB survival signals in response to TNF-α in hepatocarcinoma cells. Biochem. Biophys. Res. Commun. 2018, 503, 3003–3010. [Google Scholar] [CrossRef] [PubMed]
  62. Callegari, E.; D’Abundo, L.; Guerriero, P.; Simioni, C.; Elamin, B.K.; Russo, M.; Cani, A.; Bassi, C.; Zagatti, B.; Giacomelli, L.; et al. miR-199a-3p Modulates MTOR and PAK4 Pathways and Inhibits Tumor Growth in a Hepatocellular Carcinoma Transgenic Mouse Model. Mol. Ther. Nucleic Acids 2018, 11, 85–493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Hou, J.; Lin, L.; Zhou, W.; Wang, Z.; Ding, G.; Dong, Q.; Qin, L.; Wu, X.; Zheng, Y.; Yang, Y.; et al. Identification of miRNomes in human liver and hepatocellular carcinoma reveals miR-199a/b-3p as therapeutic target for hepatocellular carcinoma. Cancer Cell 2011, 19, 232–243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Mak, G.W.; Chan, M.M.; Leong, V.Y.; Lee, J.M.; Yau, T.O.; Ng, I.O.; Ching, Y.P. Overexpression of a novel activator of PAK4, the CDK5 kinase-associated protein CDK5RAP3, promotes hepatocellular carcinoma metastasis. Cancer Res. 2011, 71, 2949–2958. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Xu, H.T.; Lai, W.L.; Liu, H.F.; Wong, L.L.; Ng, I.O.; Ching, Y.P. PAK4 Phosphorylates p53 at Serine 215 to Promote Liver Cancer Metastasis. Cancer Res. 2016, 76, 5732–5742. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Lu, S.X.; Zhang, C.Z.; Luo, R.Z.; Wang, C.H.; Liu, L.L.; Fu, J.; Zhang, L.; Wang, H.; Xie, D.; Yun, J.P. Zic2 promotes tumor growth and metastasis via PAK4 in hepatocellular carcinoma. Cancer Lett. 2017, 402, 1–80. [Google Scholar] [CrossRef] [PubMed]
  67. Xue, J.; Chen, L.Z.; Li, Z.Z.; Hu, Y.Y.; Yan, S.P.; Liu, L.Y. MicroRNA-433 inhibits cell proliferation in hepatocellular carcinoma by targeting p21 activated kinase (PAK4). Mol. Cell. Biochem. 2015, 399, 7–86. [Google Scholar] [CrossRef]
  68. Shao, S.; Hu, Q.; Wu, W.; Wang, M.; Huang, J.; Zhao, X.; Tang, G.; Liang, T. Tumor-triggered personalized microRNA cocktail therapy for hepatocellular carcinoma. Biomater. Sci. 2020, 8, 6579–6591. [Google Scholar] [CrossRef]
  69. Zhang, D.G.; Gong, C.C.; Wu, X.J.; Ren, X.; Sedaka, R.S.; Chen, W.C.; Huo, F.C.; Chen, C.; Du, W.Q.; Pei, D.S. P21-activated kinase 5 potentiates the chemoresistant phenotype of liver cancer. Signal Transduct. Target. Ther. 2021, 6, 47. [Google Scholar] [CrossRef]
  70. Zhai, J.; Qu, S.; Li, X.; Zhong, J.; Chen, X.; Qu, Z.; Wu, D. miR-129 suppresses tumor cell growth and invasion by targeting PAK5 in hepatocellular carcinoma. Biochem. Biophys. Res. Commun. 2015, 464, 161–167. [Google Scholar] [CrossRef]
  71. Zhan, L.; Pan, Y.; Chen, L.; Chen, Z.; Zhang, H.; Sun, C. MicroRNA-526a targets p21-activated kinase 7 to inhibit tumorigenesis in hepatocellular carcinoma. Mol. Med. Rep. 2017, 16, 837–844. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Fang, Z.P.; Jiang, B.G.; Gu, X.F.; Zhao, B.; Ge, R.L.; Zhang, F.B. P21-activated kinase 5 plays essential roles in the proliferation and tumorigenicity of human hepatocellular carcinoma. Acta Pharmacol. Sin. 2014, 35, 82–88. [Google Scholar] [CrossRef] [PubMed]
  73. Zhang, D.G.; Zhang, J.; Mao, L.L.; Wu, J.X.; Cao, W.J.; Zheng, J.N.; Pei, D.S. p21-Activated kinase 5 affects cisplatin-induced apoptosis and proliferation in hepatocellular carcinoma cells. Tumour. Biol. 2015, 36, 3685–3691. [Google Scholar] [CrossRef] [PubMed]
  74. Gu, Y.F.; Kong, L.T. Inhibiting p21-activated kinase (PAK7) enhances radiosensitivity in hepatocellular carcinoma. Hum. Exp. Toxicol. 2021, 40, 2202–2214. [Google Scholar] [CrossRef]
  75. Chen, H.; Miao, J.; Li, H.; Wang, C.; Li, J.; Zhu, Y.; Wang, J.; Wu, X.; Qiao, H. Expression and prognostic significance of p21-activated kinase 6 in hepatocellular carcinoma. J. Surg. Res. 2014, 189, 81–88. [Google Scholar] [CrossRef]
  76. Zheng, J.; Zhang, C.; Li, Y.; Jiang, Y.; Xing, B.; Du, X. p21-activated kinase 6 controls mitosis and hepatocellular carcinoma progression by regulating Eg5. Biochim. Biophys. Acta (BBA)—Mol. Cell Res. 2020, 1868, 118888. [Google Scholar] [CrossRef]
  77. Liu, W.; Liu, Y.; Liu, H.; Zhang, W.; Fu, Q.; Xu, J.; Gu, J. Tumor Suppressive Function of p21-activated Kinase 6 in Hepatocellular Carcinoma. J. Biol. Chem. 2015, 290, 28489–28501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Wang, M.; Zhang, J.; Gong, N. Role of the PI3K/Akt signaling pathway in liver ischemia reperfusion injury: A narrative review. Ann. Palliat. Med. 2022, 11, 806–817. [Google Scholar] [CrossRef]
  79. Dery, K.J.; Kupiec-Weglinski, J.W. New insights into ischemia-reperfusion injury signaling pathways in organ transplantation. Curr. Opin. Organ Transplant. 2022, 27, 424–433. [Google Scholar] [CrossRef]
  80. Gao, F.; Qiu, X.; Wang, K.; Shao, C.; Jin, W.; Zhang, Z.; Xu, X. Targeting the Hepatic Microenvironment to Improve Ischemia/Reperfusion Injury: New Insights into the Immune and Metabolic Compartments. Aging Dis. 2022, 13, 1196–1214. [Google Scholar] [CrossRef]
  81. Ma, Y.; Nikfarjam, M.; He, H. The trilogy of P21 activated kinase, autophagy and immune evasion in pancreatic ductal adenocarcinoma. Cancer Lett. 2022, 24, 215868. [Google Scholar] [CrossRef] [PubMed]
  82. Wang, K.; Zhan, Y.; Huynh, N.; Dumesny, C.; Wang, X.; Asadi, K.; Herrmann, D.; Timpson, P.; Yang, Y.; Walsh, K.; et al. Inhibition of PAK1 suppresses pancreatic cancer by stimulation of anti-tumour immunity through down-regulation of PD-L1. Cancer Lett. 2020, 472, 8–18. [Google Scholar] [CrossRef] [PubMed]
  83. Micó-Carnero, M.; Casillas-Ramírez, A.; Sánchez-González, A.; Rojano-Alfonso, C.; Peralta, C. The Role of Neuregulin-1 in Steatotic and Non-Steatotic Liver Transplantation from Brain-Dead Donors. Biomedicines 2022, 10, 978. [Google Scholar] [CrossRef]
  84. Castaneda, D.; Gonzalez, A.J.; Alomari, M.; Tandon, K.; Zervos, X.B. From hepatitis A to E: A critical review of viral hepatitis. World J. Gastroenterol. 2021, 27, 1691–1715. [Google Scholar] [CrossRef]
  85. Hosseini, N.; Shor, J.; Szabo, G. Alcoholic Hepatitis: A Review. Alcohol Alcohol. 2019, 54, 408–416. [Google Scholar] [CrossRef]
  86. Sucher, E.; Sucher, R.; Gradistanac, T.; Brandacher, G.; Schneeberger, S.; Berg, T. Autoimmune Hepatitis—Immunologically Triggered Liver Pathogenesis—Diagnostic and Therapeutic Strategies. J. Immunol. Res. 2019, 2019, 9437043. [Google Scholar] [CrossRef] [Green Version]
  87. Targher, G.; Tilg, H.; Byrne, C.D. Non-alcoholic fatty liver disease: A multisystem disease requiring a multidisciplinary and holistic approach. Lancet Gastroenterol. Hepatol. 2021, 6, 578–588. [Google Scholar] [CrossRef] [PubMed]
  88. Chen, J.; Siddiqui, A. Hepatitis B Virus X Protein Stimulates the Mitochondrial Translocation of Raf-1 via Oxidative Stress. J. Virol. 2007, 81, 6757–6760. [Google Scholar] [CrossRef] [Green Version]
  89. Yu, F.X.; Zhang, X.L.; Wang, Y.P.; Ma, N.; Du, H.; Ma, J.M.; Liu, D.W. Gene polymorphisms of interleukin-28, p21-activated protein kinases 4, and response to interferon-α based therapy in Chinese patients with chronic hepatitis B. Chin. Med. J. (Engl.) 2013, 126, 1726–1731. [Google Scholar]
  90. Ma, N.; Zhang, X.; Yu, F.; Gao, P.; Fan, Q.; Liu, L.; Liu, D. Role of IFN-ks, IFN-ks related genes and the DEPDC5 gene in Hepatitis B virus-related liver disease. J. Viral Hepat. 2014, 21, e29–e38. [Google Scholar] [CrossRef]
  91. Ishida, H.; Li, K.; Yi, M.; Lemon, S.M. p21-activated kinase 1 is activated through the mammalian target of rapamycin/p70 S6 kinase pathway and regulates the replication of hepatitis C virus in human hepatoma cells. J. Biol. Chem. 2007, 282, 11836–11848. [Google Scholar] [CrossRef] [PubMed]
  92. Nguyen, H.; Sankaran, S.; Dandekar, S. Hepatitis C virus core protein induces expression of genes regulating immune evasion and anti-apoptosis in hepatocytes. Virology 2006, 354, 58–68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Fan, M.; Luo, Y.; Zhang, B.; Wang, J.; Chen, T.; Liu, B.; Sun, Y.; Nan, Y.; Hiscox, J.A.; Zhao, Q.; et al. Cell Division Control Protein 42 Interacts With Hepatitis E Virus Capsid Protein and Participates in Hepatitis E Virus Infection. Front. Microbiol. 2021, 12, 75083. [Google Scholar] [CrossRef] [PubMed]
  94. Zhang, B.; Fan, M.; Fan, J.; Luo, Y.; Wang, J.; Wang, Y.; Liu, B.; Sun, Y.; Zhao, Q.; Hiscox, J.A.; et al. Avian Hepatitis E Virus ORF2 Protein Interacts with Rap1b to Induce Cytoskeleton Rearrangement That Facilitates Virus Internalization. Microbiol. Spectr. 2022, 10, e0226521. [Google Scholar] [CrossRef]
  95. Chang, H.; He, K.Y.; Li, C.; Ni, Y.Y.; Li, M.N.; Chen, L.; Hou, M.; Zhou, Z.; Xu, Z.P.; Ji, M.J. P21 activated kinase-1 (PAK1) in macrophages is required for promotion of Th17 cell response during helminth infection. J. Cell. Mol. Med. 2020, 24, 14325–14338. [Google Scholar] [CrossRef]
  96. Lin, R.; Zhang, J.; Zhou, L.; Wang, B. Altered function of monocytes/macrophages in patients with autoimmune hepatitis. Mol. Med. Rep. 2016, 13, 3874–3880. [Google Scholar] [CrossRef] [Green Version]
  97. Afifiyan, N.; Tillman, B.; French, B.A.; Sweeny, O.; Masouminia, M.; Samadzadeh, S.; French, S.W. The role of Tec kinase signaling pathways in the development of Mallory Denk Bodies in balloon cells in alcoholic hepatitis. Exp. Mol. Pathol. 2017, 103, 191–199. [Google Scholar] [CrossRef]
  98. Kisseleva, T.; Brenner, D. Molecular and cellular mechanisms of liver fibrosis and its regression. Nat. Rev. Gastroenterol. Hepatol. 2020, 18, 151–166. [Google Scholar] [CrossRef]
  99. Roehlen, N.; Crouchet, E.; Baumert, T.F. Liver Fibrosis: Mechanistic Concepts and Therapeutic Perspectives. Cells 2020, 9, 875. [Google Scholar] [CrossRef] [Green Version]
  100. Martin, K.; Pritchett, J.; Llewellyn, J.; Mullan, A.F.; Athwal, V.S.; Dobie, R.; Harvey, E.; Zeef, L.; Farrow, S.; Streuli, C.; et al. PAK proteins and YAP-1 signalling downstream of integrin beta-1 in myofibroblasts promote liver fibrosis. Nat. Commun. 2016, 7, 2502. [Google Scholar] [CrossRef] [Green Version]
  101. Romermann, D.; Ansari, N.; Schultz-Moreira, A.R.; Michael, A.; Marhenke, S.; Hardtke-Wolenski, M.; Longerich, T.; Manns, M.P.; Wedemeyer, H.; Vogel, A.; et al. Absence of Atg7 in the liver disturbed hepatic regeneration after liver injury. Liver Int. 2020, 40, 1225–1238. [Google Scholar] [CrossRef] [PubMed]
  102. Odenwald, M.A.; Paul, S. Viral hepatitis: Past, present, and future. World J. Gastroenterol. 2022, 28, 1405–1429. [Google Scholar] [CrossRef] [PubMed]
  103. Fanning, G.C.; Zoulim, F.; Hou, J.; Bertoletti, A. Therapeutic strategies for hepatitis B virus infection: Towards a cure. Nat. Rev. Drug Discov. 2019, 18, 827–844. [Google Scholar] [CrossRef]
  104. Powell, E.E.; Wong, V.W.; Rinella, M. Non-alcoholic fatty liver disease. Lancet 2021, 397, 2212–2224. [Google Scholar] [CrossRef] [PubMed]
  105. Ganne-Carrié, N.; Nahon, P. Hepatocellular carcinoma in the setting of alcohol-related liver disease. J. Hepatol. 2019, 70, 284–293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Fan, Y.; Xue, H.; Zheng, H. Systemic Therapy for Hepatocellular Carcinoma: Current Updates and Outlook. J. Hepatocell. Carcinoma 2022, 9, 233–263. [Google Scholar] [CrossRef] [PubMed]
  107. Kudo, M.; Finn, R.S.; Qin, S.; Han, K.H.; Ikeda, K.; Piscaglia, F.; Baron, A.; Park, J.W.; Han, G.; Jassem, J.; et al. Lenvatinib versus sorafenib in first-line treatment of patients with unresectable hepatocellular carcinoma: A randomised phase 3 non-inferiority trial. Lancet 2018, 391, 1163–1173. [Google Scholar] [CrossRef] [Green Version]
  108. Chakraborty, E.; Sarkar, D. Emerging Therapies for Hepatocellular Carcinoma (HCC). Cancers 2022, 14, 2798. [Google Scholar] [CrossRef]
  109. Yao, D.; Li, C.; Rajoka, M.S.R.; He, Z.; Huang, J.; Wang, J.; Zhang, J. P21-Activated Kinase 1: Emerging biological functions and potential therapeutic targets in Cancer. Theranostics 2020, 10, 9741–9766. [Google Scholar] [CrossRef]
  110. Rudolph, J.; Crawford, J.J.; Hoeflich, K.P.; Chernoff, J. p21-activated kinase inhibitors. Enzymes 2013, 34, 57–180. [Google Scholar]
  111. Amirthalingam, M.; Palanisamy, S.; Tawata, S. p21-Activated kinase 1 (PAK1) in aging and longevity: An overview. Ageing Res. Rev. 2021, 71, 101443. [Google Scholar] [CrossRef]
  112. Mao, Y.; Lee, E.; Yang, X.; Bae, E.J.; Jeon, R.; Park, B.H. Targeting p21-activated kinase 4 (PAK4) with pyrazolo[3,4-d]pyrimidine derivative SPA7012 attenuates hepatic ischaemia-reperfusion injury in mice. J. Enzyme Inhib. Med. Chem. 2022, 37, 2133–2146. [Google Scholar] [CrossRef] [PubMed]
  113. Deacon, S.W.; Beeser, A.; Fukui, J.A.; Rennefahrt, U.E.; Myers, C.; Chernoff, J.; Peterson, J.R. An isoform-selective, small-molecule inhibitor targets the autoregulatory mechanism of p21-activated kinase. Chem. Biol. 2008, 15, 322–331. [Google Scholar] [CrossRef] [Green Version]
  114. Abu Aboud, O.; Chen, C.H.; Senapedis, W.; Baloglu, E.; Argueta, C.; Weiss, R.H. Dual and Specific Inhibition of NAMPT and PAK4 By KPT-9274 Decreases Kidney Cancer Growth. Mol. Cancer Ther. 2016, 15, 2119–2129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Li, N.; Lopez, M.A.; Linares, M.; Kumar, S.; Oliva, S.; Martinez-Lopez, J.; Xu, L.; Xu, Y.; Perini, T.; Senapedis, W.; et al. Dual PAK4-NAMPT Inhibition Impacts Growth and Survival, and Increases Sensitivity to DNA-Damaging Agents in Waldenström Macroglobulinemia. Clin. Cancer Res. 2019, 25, 369–377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Cordover, E.; Wei, J.; Patel, C.; Shan, N.L.; Gionco, J.; Sargsyan, D.; Wu, R.; Cai, L.; Kong, A.-N.; Jacinto, E.; et al. KPT-9274, an Inhibitor of PAK4 and NAMPT, Leads to Downregulation of mTORC2 in Triple Negative Breast Cancer Cells. Chem. Res. Toxicol. 2020, 33, 482–491. [Google Scholar] [CrossRef]
  117. Wang, J.; Zhu, Y.; Chen, J.; Yang, Y.; Zhu, L.; Zhao, J.; Yang, Y.; Cai, X.; Hu, C.; Rosell, R.; et al. Identification of a novel PAK1 inhibitor to treat pancreatic cancer. Acta Pharm. Sin. B 2020, 10, 603–614. [Google Scholar] [CrossRef]
  118. He, H.; Dumesny, C.; Ang, C.-S.; Dong, L.; Ma, Y.; Zeng, J.; Nikfarjam, M. A novel PAK4 inhibitor suppresses pancreatic cancer growth and enhances the inhibitory effect of gemcitabine. Transl. Oncol. 2022, 16, 101329. [Google Scholar] [CrossRef]
  119. Jones, J.G. Hepatic glucose and lipid metabolism. Diabetologia 2016, 59, 1098–1103. [Google Scholar] [CrossRef] [Green Version]
  120. Asrani, S.K.; Devarbhavi, H.; Eaton, J.; Kamath, P.S. Burden of liver diseases in the world. J. Hepatol. 2019, 70, 151–171. [Google Scholar] [CrossRef]
  121. Chiang, Y.-T.A.; Ip, W.; Shao, W.; Song, Z.E.; Chernoff, J.; Jin, T. Activation of cAMP Signaling Attenuates Impaired Hepatic Glucose Disposal in Aged Male p21-Activated Protein Kinase-1 Knockout Mice. Endocrinology 2014, 155, 2122–2132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Du, D.; Liu, C.; Qin, M.; Zhang, X.; Xi, T.; Yuan, S.; Hao, H.; Xiong, J. Metabolic dysregulation and emerging therapeutical targets for hepatocellular carcinoma. Acta Pharm. Sin. B 2021, 12, 558–580. [Google Scholar] [CrossRef] [PubMed]
  123. Ren, Y.; Shen, H.M. Critical role of AMPK in redox regulation under glucose starvation. Redox Biol. 2019, 25, 01154. [Google Scholar] [CrossRef] [PubMed]
  124. Mao, Z.; Zhang, W. Role of mTOR in Glucose and Lipid Metabolism. Int. J. Mol. Sci. 2018, 19, 2043. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Chen, J.; Chen, J.; Huang, J.; Li, Z.; Gong, Y.; Zou, B.; Liu, X.; Ding, L.; Li, P.; Zhu, Z.; et al. HIF-2α upregulation mediated by hypoxia promotes NAFLD-HCC progression by activating lipid synthesis via the PI3K-AKT-mTOR pathway. Aging (Albany NY) 2019, 11, 10839–10860. [Google Scholar] [CrossRef]
  126. Morrice, N.A.; Fecondo, J.; Wettenhall, R.E. Differential effects of fatty acid and phospholipid activators on the catalytic activities of a structurally novel protein kinase from rat liver. FEBS Lett. 1994, 351, 171–175. [Google Scholar] [CrossRef] [Green Version]
  127. Yu, W.; Liu, J.; Morrice, N.A.; Wettenhall, R.E. Isolation and characterization of a structural homologue of human PRK2 from rat liver. Distinguishing substrate and lipid activator specificities. J. Biol. Chem. 1997, 272, 10030–10034. [Google Scholar] [CrossRef] [Green Version]
  128. Joshi, A.; Azuma, R.; Akumuo, R.; Goetzl, L.; Pinney, S.E. Gestational diabetes and maternal obesity are associated with sex-specific changes in miRNA and target gene expression in the fetus. Int. J. Obes. (Lond.) 2020, 44, 1497–1507. [Google Scholar] [CrossRef]
  129. Zhang, X.; Zhang, X.; Li, Y.; Shao, Y.; Xiao, J.; Zhu, G.; Li, F. PAK4 regulates G6PD activity by p53 degradation involving colon cancer cell growth. Cell Death Dis. 2017, 8, e2820. [Google Scholar] [CrossRef] [Green Version]
  130. Zhou, J.; Zhou, F.; Wang, W.; Zhang, X.-J.; Ji, Y.-X.; Zhang, P.; She, Z.G.; Zhu, L.; Cai, J.; Li, H. Epidemiological Features of NAFLD From 1999 to 2018 in China. Hepatology 2020, 71, 1851–1864. [Google Scholar] [CrossRef]
  131. Younossi, Z.M.; Koenig, A.B.; Abdelatif, D.; Fazel, Y.; Henry, L.; Wymer, M. Global epidemiology of nonalcoholic fatty liver disease-Meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology 2016, 64, 73–84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Dewidar, B.; Kahl, S.; Pafili, K.; Roden, M. Metabolic liver disease in diabetes—From mechanisms to clinical trials. Metabolism 2020, 111, 154299. [Google Scholar] [CrossRef] [PubMed]
  133. Tang, S.P.; Mao, X.L.; Chen, Y.H.; Yan, L.L.; Ye, L.P.; Li, S.W. Reactive Oxygen Species Induce Fatty Liver and Ischemia-Reperfusion Injury by Promoting Inflammation and Cell Death. Front. Immunol. 2022, 13, 870239. [Google Scholar] [CrossRef] [PubMed]
  134. Guo, J.; Akahoshi, T.; Mizuta, Y.; Murata, M.; Narahara, S.; Kawano, T.; Nagao, Y.; Zhang, S.; Tomikawa, M.; Kawanaka, H.; et al. Histidine-Rich Glycoprotein Alleviates Liver Ischemia/Reperfusion Injury in Mice With Nonalcoholic Steatohepatitis. Liver Transpl. 2021, 27, 840–853. [Google Scholar] [CrossRef] [PubMed]
  135. Yang, F.; Zhang, Y.; Ren, H.; Wang, J.; Shang, L.; Liu, Y.; Zhu, W.; Shi, X. Ischemia reperfusion injury promotes recurrence of hepatocellular carcinoma in fatty liver via ALOX12-12HETE-GPR31 signaling axis. J. Exp. Clin. Cancer Res. 2019, 38, 1–14. [Google Scholar] [CrossRef] [Green Version]
  136. Czigany, Z.; Lurje, I.; Schmelzle, M.; Schöning, W.; Öllinger, R.; Raschzok, N.; Sauer, I.M.; Tacke, F.; Strnad, P.; Trautwein, C.; et al. Ischemia-Reperfusion Injury in Marginal Liver Grafts and the Role of Hypothermic Machine Perfusion: Molecular Mechanisms and Clinical Implications. J. Clin. Med. 2020, 9, 846. [Google Scholar] [CrossRef] [Green Version]
  137. Ringelhan, M.; Pfister, D.; O’Connor, T.; Pikarsky, E.; Heikenwalder, M. The immunology of hepatocellular carcinoma. Nat. Immunol. 2018, 19, 222–232. [Google Scholar] [CrossRef]
  138. Huynh, N.; Wang, K.; Yim, M.; Dumesny, C.J.; Sandrin, M.S.; Baldwin, G.S.; Nikfarjam, M.; He, H. Depletion of p21-activated kinase 1 up-regulates the immune system of APC(∆14/+) mice and inhibits intestinal tumorigenesis. BMC Cancer 2017, 17, 431. [Google Scholar] [CrossRef]
  139. Abril-Rodriguez, G.; Torrejon, D.Y.; Liu, W.; Zaretsky, J.M.; Nowicki, T.S.; Tsoi, J.; Puig-Saus, C.; Baselga-Carretero, I.; Medina, E.; Quist, M.J.; et al. PAK4 inhibition improves PD-1 blockade immunotherapy. Nat. Cancer 2020, 1, 46–58. [Google Scholar] [CrossRef]
Figure 1. Upstream regulators of PAKs in liver disorders. The upstream regulators of PAKs in liver disorders include exogenous compounds and endogenous molecules (proteins, miRNAs and lncRNAs). CDK5RAP3, CDK5 kinase regulatory subunit-associated protein 3; IRI, ischemia-reperfusion injury; Mgat5, N-acetylglucosaminyltransferase V; PAK, p21 Activated Kinase.
Figure 1. Upstream regulators of PAKs in liver disorders. The upstream regulators of PAKs in liver disorders include exogenous compounds and endogenous molecules (proteins, miRNAs and lncRNAs). CDK5RAP3, CDK5 kinase regulatory subunit-associated protein 3; IRI, ischemia-reperfusion injury; Mgat5, N-acetylglucosaminyltransferase V; PAK, p21 Activated Kinase.
Cancers 15 00551 g001
Figure 2. The structure and activation of PAKs. (A) Two-dimensional diagram of PAKs structure. (B) Different activation mechanisms of Group I PAKs and Group II PAKs.
Figure 2. The structure and activation of PAKs. (A) Two-dimensional diagram of PAKs structure. (B) Different activation mechanisms of Group I PAKs and Group II PAKs.
Cancers 15 00551 g002
Figure 3. The pathways affected by PAKs in liver cancer. PAKs modulate multiple signaling pathways (e.g., RAF/MEK/ERK, β-catenin) to promote the proliferation, invasion/migration, angiogenesis, and therapy resistance of cancer cells while inhibiting apoptosis.
Figure 3. The pathways affected by PAKs in liver cancer. PAKs modulate multiple signaling pathways (e.g., RAF/MEK/ERK, β-catenin) to promote the proliferation, invasion/migration, angiogenesis, and therapy resistance of cancer cells while inhibiting apoptosis.
Cancers 15 00551 g003
Figure 4. PAK inhibitors applied in liver disorders. Abbreviations: HCC, hepatocellular carcinoma; IRI, ischemia-reperfusion injury; PAK, p21 Activated Kinase; ROS, reactive oxygen species.
Figure 4. PAK inhibitors applied in liver disorders. Abbreviations: HCC, hepatocellular carcinoma; IRI, ischemia-reperfusion injury; PAK, p21 Activated Kinase; ROS, reactive oxygen species.
Cancers 15 00551 g004
Table 1. Overview of PAKs in liver disorders.
Table 1. Overview of PAKs in liver disorders.
Liver DisordersGroup I PAKsGroup II PAKs
Liver cancerPAK1:
β-catenin → Proliferation/invasion↑ and apoptosis↓
MAPK/ERK, PI3K/AKT → Apoptosis↓
Snail → Migration and invasion↑
Snail, β-catenin, p53/p21 → Xenograft tumor growth↑
Paxillin, JNK → Actin stress fibers and metastasis↑
Anchorage-independent growth and anoikis resistance ↑
HIF-1α, Vimentin → Vasculogenic mimicry↑
PAK2:
Migration↑
PAK3:
TGF-β/Smad → EMT↑
PAK4:
Caspase-8, NF-κB → Apoptosis↓
RAF/MEK/ERK → Tumor growth and metastasis↑
P53 → Migration and invasion↑
PAK5:
Proliferation and invasion↑
Apoptosis induced by cisplatin↓
β-catenin/ABCB1 → Resistance to adriamycin, 5-fluorouracil and sorafenib↑
Radiotherapy resistance↑
PAK6:
Proliferation↑
Eg5 → mitosis↑
HCC metastasis↑
Liver IRIPAK1:
VEGFα, IGF1 → IRI↓
PAK4:
Nrf2 → Inflammation and necrosis of hepatocytes↑
HepatitisPAK1:
RAF-1 → HBV X protein → Apoptosis↓
HCV replication↓
NMIIA/Cofilin → HEV infection↑
NF-κB/IRF1/IL-6 → The differentiation of CD4+ T cells to Th17 cells and inflammation in Schistosoma japonicum infection↑
Autoimmune hepatitis progression↑
PAK2:
Apoptosis and inflammatory reaction in cells infected with HCV↓
PAK3:
HBV replication↑
PAK6:
Tumorigenesis in alcoholic hepatitis↑
Liver fibrosisPAK1:
Integrin β-1 → Myofibroblasts activation and ECM production↑
PAK2:
Myofibroblast activation and liver fibrosis↑
PAK3:
Integrin β-1 → Myofibroblasts activation and ECM production↑
PAK4:
Perisinusoidal/pericellular fibrosis↑
↑—increment; ↓—decrement.
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

Qiu, X.; Xu, H.; Wang, K.; Gao, F.; Xu, X.; He, H. P-21 Activated Kinases in Liver Disorders. Cancers 2023, 15, 551. https://doi.org/10.3390/cancers15020551

AMA Style

Qiu X, Xu H, Wang K, Gao F, Xu X, He H. P-21 Activated Kinases in Liver Disorders. Cancers. 2023; 15(2):551. https://doi.org/10.3390/cancers15020551

Chicago/Turabian Style

Qiu, Xun, Hanzhi Xu, Kai Wang, Fengqiang Gao, Xiao Xu, and Hong He. 2023. "P-21 Activated Kinases in Liver Disorders" Cancers 15, no. 2: 551. https://doi.org/10.3390/cancers15020551

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