Abstract
P21-activated protein kinases (PAKs) are thought to be at the center of tumor signaling pathways. As a representative member of the group II PAK family, P21-activated protein kinase 4 (PAK4) plays an important role in the development of tumors, with several biological functions such as participating in oncogenic transformation, promoting cell division, resisting aging and apoptosis, regulating cytoskeleton and adhesion, as well as suppressing antitumor immune responses. PAK4 is also crucial in biological processes, including the occurrence, proliferation, survival, migration, invasion, drug resistance, and immune escape of tumor cells. It is closely related to poor prognosis and tumor-related pathological indicators, which have significant clinical and pathological significance. Therefore, this article offers a review of the structure, activation, and biological functions of PAK4 and its clinical and pathological importance. This overview should be of assistance for future research on PAK4 and tumors and provide new ideas for tumor treatment and prognostic evaluation of patients.
1 Introduction
The occurrence and development of tumors involve complex processes and multiple mechanisms. Ongoing research on tumors has led to increased recognition of the important role of P21-activated protein kinases (PAKs) in the occurrence and development of tumors. P21-activated protein kinase 4 (PAK4) is a representative member of the group II PAK family and mediates almost all biological behaviors of tumors at the cell level, from oncogenic transformation [1] to continuous proliferation [2] and from the tenacity of cancer cells [3] to their metastasis [2], drug resistance [4], and immune escape [5]. As a potential tumor biomarker, PAK4 has great clinical and pathological significance in the evaluation of prognostic and pathological indicators [6]. Therefore, it has become a popular research topic in tumor signal transduction and treatment in recent years. Here, we review the structure, activation, and biological function of PAK4 as well as its clinical and pathological importance to provide context for future preclinical and clinical research on PAK4 and tumors.
2 Structure of PAK4
The PAK serine/threonine kinase family consists of six members. These members are divided into two groups according to their structural and activation characteristics: group I (PAK1–3) and group II (PAK4–6). Both groups of PAKs share a similar structure: an amino-terminal p21-binding domain (PBD) which can bind Rho GTPases (Cdc42/Rac), and a highly conserved serine/threonine kinase domain at the carboxy-terminal. The group I PAKs have two proline-rich regions (PXXP) in front of the PBD. Moreover, an autoinhibitory domain (AID) behind the PBD acts with the PBD as a dimer. There is also a PAK-interacting exchange factor (PIX) binding domain that binds PIX between the AID and the serine/threonine kinase domain and a Gβγ binding domain behind the kinase domain. Group II PAKs also have a p21-binding domain and a kinase domain, which are slightly different from group I. The group II PAKs were believed not to contain an AID, but later studies have found that they have a sequence similar to the AID. However, this sequence does not overlap with PBD but is present on its own behind the PBD. Meanwhile, some models suggest that the AID-like domain beside the PBD, which keeps PAK4 inactive, is an autoinhibitory pseudosubstrate domain (PSD) [7,8,9,10].
PAK4 is the first member found in the group Ⅱ PAKs and consists of 591 amino acids (aa). In PAK4, PBD is located at 10–35aa. As in other group II PAKs, the AID-like sequence of PAK4 does not overlap with the PBD as the AID of group I PAKs does but plays its role independently of the PBD sequence. Moreover, a region behind the PBD interacts with the ribonucleoprotein [11]. The serine/threonine kinase domain is located at 323–574aa in PAK4. An integrin-binding domain (IBD) is located in the 505–530aa of PAK4, in the posterior part of the serine/threonine kinase domain, which can bind to some integrins [12,13]. Although this structure is thought to exist in other kinds of PAKs, it has not yet been reported [9]. The kinase domain of PAK4 forms an αC helix, a glycine-rich loop, and some activated fragments spatially, but most of these structures are disordered and very flexible [14]. In addition to the three main structures (the PBD, the AID-like sequence, and the serine/threonine kinase domains), there are several PXXP between the AID-like sequence and the kinase domain, as well as a guanosine exchange factor (GEF) interacting domains behind the PXXP (Figure 1) [8,9,10].

Structure of group I and group II PAKs. All PAKs have PXXP, an amino-terminal PBD, and a carboxy-terminal kinase domain. Group I PAKs have an extra PIX binding domain, Gβγ binding domain, and AID behind the PBD, which acts with the PBD as a dimer. Group II PAKs have an additional AID-like domain (some models suggest it is PSD) that exists alone rather than forms a complex with PBD. PAK4 structure is highlighted. The length of PAK4 is 591aa. Besides its PBD(10–35aa), AID-like domain/PSD(35–68aa), kinase domain (323–574aa), and PXXP, PAK4 also has a RNP binding domain (65–175aa), GEF interacting domain, and IBD (505–530aa). PXXP: proline-rich regions; PBD: p21-binding domain; PIX: PAK interacting exchange factor; AID: autoinhibitory domain; PSD: pseudosubstrate domain; RNP: ribonucleoprotein; GEF: guanosine exchange factor; IBD: integrin-binding domain; aa: amino acid.
3 Regulation of PAK4 kinase activity
Compared to research on group II PAKs, that on the group I PAKs has been more thorough. The regulation mechanism of group I PAKs is trans-auto-inhibition [15]. In the inactive state, two group I PAKs monomers fold into a homodimer in a head-to-tail manner [16]. The AID of one monomer is inserted into the kinase domain of the other [17]. This interaction inhibits autophosphorylation of the PAKs and activation of the kinase domain. Activation of group I PAKs mainly depends on the combination of PBD and CDC42/Rac. A series of conformational changes occur after their binding, and the homodimer state is no longer maintained, so the inhibitory effect of the AID on the kinase domain is reduced. The PAKs are autophosphorylated at multiple sites, switching it from an inactive state to an active state [18,19] (Figure 2a). Group I PAKs can also be regulated by phosphatidylinositol, proteins that contain src homology 3 (SH3) domains, 3-phospho-inositide-dependent kinase-1, PIX, and other mechanisms [20,21,22,23].

Activation of group Ⅰ and group Ⅱ PAKs. (a) The activation of group I PAKs: Two group I PAKs monomers constitute a homodimer. The AID of one monomer is inserted into the kinase domain of the other and inactivates it. The combination of PBD and CDC42/RAC can relieve the auto-inhibition state, leading to the autophosphorylation and activation of the PAKs. (b) Model 1 for the activation of PAK4: The PAK4 monomer remains inactive due to the binding of the kinase domain and the AID-like sequence, but can be activated by binding with CDC42. (c) Model 2 for the activation of PAK4: The interaction of PSD with the kinase domain can inhibit PAK4 activity. The activation of PAK4 involves two steps. CDC42 binds to PBD first to relocate PAK4 in cells. Subsequently, SH3 proteins bind to PSD, resulting in the activation of PAK4. P: phosphorylation; AID: autoinhibitory domain; PBD: p21-binding domain; PSD: pseudosubstrate domain; SH3: src homology 3.
The activation mechanism of group II PAKs is different from that of group I PAKs. For group I PAKs and most protein kinases, phosphorylation of the activation loop is related to kinase activity [24]. However, Ser474 on the activation loop of PAK4 is constitutively autophosphorylated [25]. The phosphorylation of the activation loop is not related to kinase activity, so the change in activity may depend on conformational changes [26]. Presently, there are two clear activation mechanisms of PAK4. One study showed that activation is dependent on the conformational changes of PAK4 mediated by CDC42. PAK4 exists as a monomer in the inactive state and remains inactive due to the binding of the kinase domain and the AID-like sequence. Also, PAK4 is activated when active Cdc42 binds to it and causes conformational change [24] (Figure 2b). Another study showed that activation of PAK4 is dependent on the reduction of PSD autoinhibition mediated by SH3 proteins. The interaction of PSD with the kinase domain can keep PAK4 in an inactive state [14]. During the activation process of PAK4, CDC42/RAC binds to it first, not activating the kinase, but reorienting it [27]. Subsequently, PSD binds to SH3 proteins, resulting in the reduction of auto-inhibition and kinase activation [14] (Figure 2c). Group II PAKs can also be activated through other mechanisms. For example, hepatocyte growth factor (HGF) can enhance the kinase activity of PAK4 in MDK cells [28].
4 PAK4 and tumor occurrence or development
4.1 PAK4 and oncogenic transformation
PAK4 is closely related to the occurrence of tumors. It participates in oncogenic transformation and mediates pre-tumor biological effects such as anchorage-independent growth [29], epithelial-mesenchymal transition (EMT) [30], and polarization loss [31], and thus is a key transduction factor of oncogenic signals. PAK4 undergoes gene amplification in a variety of tumor cells. Its gene is located on chromosome 19q13 (which is associated with tumorigenesis and frequently amplified) and is regarded as an amplification target in a variety of tumors such as pancreatic cancer (PC) and oral squamous cell carcinoma (OSCC) [29,32,33]. Amplification of the PAK4 gene leads to overexpression of PAK4, which is often consistent with oncogenic transformation. Anchorage-independent growth is an important feature of cell oncogenic transformation [34]. One study showed that activated PAK4 induces anchorage-independent growth of mouse fibroblasts and plays a key role in Ras-driven cell transformation [29]. Parvin Beta (ParvB) inhibits integrin-linked kinase, which is associated with anchorage-independent growth [35]. Through next-generation sequencing, it was found that overexpression of PAK4 in mouse mammary epithelial cells (iMMECs) showed a downregulation of ParvB gene expression [30], which also suggests a positive role for PAK4 in anchorage-independent growth. EMT gives cells the ability to metastasize and invade and is considered to be a key step in the oncogenic transformation of cells [36]. Forkhead Box C2 (FoxC2) is a transcription factor that regulates cell migration, and its expression increases during EMT [37]. Overexpression of PAK4 in iMMECs was correlated with an increase in FoxC2 gene expression, suggesting that PAK4 promotes EMT [30]. Loss of polarization is closely related to oncogenic transformation, and the loss of apical polarization of epithelial cells is usually followed by cancer [38]. The overexpression of PAK4 leads to disordered cell apical and basal structures, which destroys cell polarization, especially apical polarization [31]. In premalignant cells, transforming growth factor-beta (TGF-β) plays a growth inhibition role. So, it is the loss of the tumor inhibition mediated by TGF-β that is an important characteristic of many malignantly transformed cells [39]. PAK4 can weaken the growth inhibition mediated by the TGF-β1/small mother against the decapentaplegic (Smad) pathway, which may cause gastric cancer (GC). Through kinase-independent interaction with Smad2/3, PAK4 blocks the phosphorylation of Smad2 at Ser465/467 or Smad3 at Ser423/425 induced by TGF. Under HGF stimulation, PAK4 phosphorylates Smad2 at Ser465, leading to the degradation of Smad2 through the ubiquitin-proteasome pathway [40]. The oncogenic properties of PAK4 have also been confirmed directly. When iMMECs stably expressing PAK4 were implanted into the breast fat pads of athymic mice, breast tumors were formed in the mice [31].
4.2 PAK4 and proliferation
In the process of tumor cell proliferation, PAK4 shows positive significance in regulating cell mitosis [41] and controlling the cell cycle [42]. Inhibition of endogenous PAK4 delays the process of mitosis, and after PAK4 is depleted, the cell even shows mitotic defects such as a lack of centrosome splitting, multipolar spindles, and chromosome lagging [41]. Moreover, PAK4 phosphorylates GTPase Ran at Ser135. The phosphorylation of Ran prevents its binding to Ran guanylate exchange factor (RCC1) and Ran GTPase activating protein (RanGAP1) and inhibits the loading and hydrolysis of GTP at different stages. The result is prevention or maintenance of Ran activation, respectively, which regulates the assembly of Ran-dependent complexes on the mitotic spindle. PAK4 is also closely linked to the regulation of the cell cycle, and it plays a vital role in both the G1 phase and the G2/M transition phase. One study showed that the loss of PAK4 leads to a decrease of G1 cells and induces a block in the G2/M transition, which suggests that PAK4 plays a crucial part in the control of G1 and G2 checkpoints. The level of PAK4 increases transiently in the early stage of G1, and the increase of PAK4 leads to a decrease in P21, which can be a cyclin-dependent kinase inhibitor [42,43]. Similarly, overexpression of PAK4 inhibits the expression of cyclin-dependent kinase inhibitor P57 Kip2 and promotes the degradation of P57 Kip2 through the ubiquitin-proteasome pathway [44]. In addition to the regulation of mitosis and the cell cycle, PAK4 promotes cell proliferation via multiple pathways. Glycometabolism provides energy for tumor cells to support the rapid proliferation of tumors [45]. One of the ways PAK4 promotes proliferation is by promoting glycometabolism. In colon cancer (colon adenocarcinoma, COAD) cells, PAK4 boosts glucose intake and NADPH production, thereby mediating COAD cell proliferation. PAK4 accomplishes this by increasing the activity of glucose-6-phosphate dehydrogenase (G6PD). Interestingly, rather than mediating P53 ubiquitination and degradation directly, PAK4 promotes these processes by enhancing the binding of murine double minute 2 to P53, thereby increasing G6PD activity [46]. The phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) pathway is crucial in tumor proliferation. In breast cancer (BC) cells, PAK4 activates the PI3K/AKT pathway to promote proliferation [47]. In endometrial cancer (EC) cells, the PI3K/AKT pathway activates PAK4 under the influence of estrogen. Activated PAK4 increases the level of estrogen receptor alpha (ERα), and the higher level of ERα promotes the activation of estrogen-mediated PAK4. The two promote each other to form a positive feedback loop, thus increasing the expression of CyclinD1 and promoting cell cycle progression, prompting the proliferation of EC cells [48]. Pathways such as PAK4/c-Src/epidermal growth factor receptor (EGFR), the interaction of PAK4 with β-catenin, and PAK4/LIM domain kinase 1 (LIMK1)/Cofilin–1 are also important in proliferation. In ovarian cancer (OC) cells, PAK4 controls the expression of CyclinD1 and cell division cycle 25A (CDC25A) by activating the c-Src/EGFR pathway, thereby inducing cell proliferation [49]. Similarly, PAK4 binds and phosphorylates β-catenin, inhibiting the degradation of β-catenin and promoting T-cell factor (TCF)/lymphoid enhancer factor (LEF) transcriptional activity induced by β-catenin. Also, PAK4 nuclear accumulation enhances β-catenin nuclear import, promoting TCF/LEF activity, stimulating the expression of CyclinD1 and c-myc, and leading to cell proliferation [50]. The expression of PAK4/LIMK1/Cofilin–1 increases in osteosarcoma (OS) cells, promoting the proliferation of OS cells [51]. Unlike the two cyclin-dependent pathways mentioned above, Cofilin–1 is an actin-binding protein that may participate in mitosis by regulating the cytoskeleton to promote proliferation itself [52].
4.3 PAK4 and survival
Under natural conditions, tumor cells often have tenacious vitality, attributed to their resistance to apoptosis [53] and escape from cell senescence [54]. The resistance of tumor cells to apoptosis is based on inhibiting the apoptotic pathway [55] and activating the survival pathway [56]. In the process of apoptosis inhibition, inhibition of the caspase cascade is key [57]. Overexpression of PAK4 can protect cells from apoptosis induced by serum withdrawal, tumor necrosis factor-alpha (TNFα) treatment, and UV irradiation. It inhibits the proapoptotic protein Bad by enhancing phosphorylation at Ser112 and Ser136, which likely prevents the release of cytochrome C in mitochondria and inhibits activation of caspase–3, thereby inhibiting apoptosis [58]. Also, PAK4 can prevent the caspase cascade by inhibiting the recruitment of caspase–8 to TNFα receptor 1 (TNFR1) [59]. Similarly, PAK4 is essential in the nuclear factor-κB (NF-κB)-mediated survival pathway. PAK4 plays an active role in the TNF-α-induced and NF-κB-mediated survival pathways by promoting TRADD and TNFR1 [60]. PAK4 can also enhance the nuclear accumulation and transcriptional activity of NF-κB by activating Akt and extracellular-signal-regulated protein kinase (ERK) pathways, thereby mediating the activation of cell survival pathways [61]. Cell senescence arrests growth, preventing the development of cancer [62]. Therefore, extended tumor survival cannot be separated from the suppression of senescence. One group found that PAK4 can relieve the senescence-like growth arrest of BC cells by inhibiting the RELB subunit of the NF-κB (RELB) – CCAAT/enhancer-binding protein beta (CEBPB) pathway [63]. Moreover, in primary fibroblasts, PAK4 induces premature cell senescence by activating the ERK pathway and the cell cycle inhibitors P16 INK4 and P19 ARF [64]. It is worth noting that this conflicts with findings that PAK4 resists apoptosis or relieves senescence-like growth inhibition. The effect of PAK4 on cell senescence is still controversial and remains to be explored. Whether PAK4 causes these two contradictory effects due to the difference in the intracellular environments of apoptotic cells, senescent cells, and normal cells is worthy of investigation.
4.4 PAK4 and metastasis
Malignant tumors often metastasize, so treatment results are frequently unsatisfactory [65]. At the cellular level, the essence of tumor metastasis lies in the migration and invasion of tumor cells, which are attributed to the weakening of cell adhesion and reconstruction of the actin skeleton [66]. Integrin mediates the adhesion of cells to the extracellular matrix (ECM), which is central to the process of migration and invasion. PAK4 can bind to the proximal cytoplasmic domain of the integrin β5 subunit through the IBD. However, there is no simple physical combination between them. PAK4 binds to and phosphorylates the integrin β5 subunit at Ser759 and Ser762 in a kinase-dependent manner, inducing cell migration [67]. PAK4 mediates cell migration, and integrin mediates cell adhesion. Interestingly, there is a negative feedback loop between PAK4 and integrin instead of an antagonistic effect. The connection between integrin and vitronectin in the ECM activates PAK4, but activated PAK4, in turn, inhibits integrin clustering and integrin-F-actin connection, which destroys the stability of the integrin-mediated adhesion structure, thereby promoting cell metastasis [68]. The focal adhesions connect the ECM and the cytoskeleton [69], and PAK4 can act on Rho GTPase to regulate the decomposition and formation of focal adhesions. One group showed that under HGF stimulation, PAK4 phosphorylates GEF-H1 at Ser885 and inhibits the activity of RhoA mediated by GEF-H1, regulating adhesion. Also, PAK4 promotes the decomposition of focal adhesions by paxillin at Ser272 [70]. Unlike the inhibitory effect of PAK4 on RhoA, PAK4 protects RhoU from ubiquitination in a kinase-independent manner, simultaneously driving adhesion turnover and promoting cell migration [71]. PAK4 plays a crucial part in the regulation of the actin cytoskeleton in addition to the process of adhesion. PAK4 is an effector molecule of Cdc42Hs. Under the induction of the activated form of Cdc42Hs, PAK4 redistributes in the Golgi apparatus, induces filopodia, and initiates local actin polymerization in the Golgi apparatus [27]. PAK4 also interacts with the neural Wiskott-Aldrich syndrome protein (N–WASP) Verprolin Homology/Cofilin/Acidic (VCA) domain. On this basis, PAK4 phosphorylates the N–WASP VCA domain at Ser484/Ser485, increasing the polymerization rate of actin at the forefront and promoting cell migration [72]. Cell invasion requires degradation of ECM, and the actin-rich invasive pseudopodia participate in this process [73]. In the process of invadopodia formation, PAK4 has also shown a promotion effect. PAK4 mediates invadopodia maturation in melanoma cells by inhibiting postsynaptic density protein 95/disc-large/zonula occludens-RhoGEF (PDZ–RhoGEF) [74]. The underlying mechanism may be that Rho has an inhibitory effect on invadopodia, and PDZ–RhoGEF has a Rho activation effect, so the inhibition of PDZ–RhoGEF leads to a decrease in Rho activity, which leads to the formation of invadopodia [75]. In addition to regulating the internal activities of cell metastasis, PAK4 promotes tumor cell migration and invasion via multiple pathways. Of these, the PAK4/LIMK1/Cofilin–1 pathway has been researched relatively extensively. In OS, the PAK4/LIMK1/Cofilin–1 pathway is upregulated, promoting its migration and invasion. In contrast, silencing PAK4 causes pathway inhibition, weakening the migration and invasion ability of OS cells [51]. In GC cells, DiGeorge critical region 6 L (DGCR6L) interacts with PAK4 and enhances the phosphorylation level of LIMK1 and cofilin in a dose-dependent manner, promoting the migration of GC cells [76]. Similarly, HGF stimulates cell migration in prostate cancer cells via the PAK4/LIMK1/Cofilin–1 pathway [77]. Also, the following factors can all mediate migration and invasion: the PAK4/PI3K/Akt pathway, the PAK4/CEBPB/Claudin 4 (CLDN4) pathway, inhibition of P53, activation of superior cervical ganglia 10, enhancement of c-Src and ERK1/2 activity, induction of Matrix metallopeptidase 2 expression, and binding to eukaryotic elongation factor 1 alpha in a kinase-independent manner [49,78,79,80,81,82].
4.5 PAK4 and drug resistance
At present, chemotherapy is still the dominant treatment option for cancer, although there are many ways to treat tumors. Unfortunately, during chemotherapy, drug resistance often leads to disappointing therapeutic outcomes [83]. Therefore, research on the mechanism of drug resistance has become very relevant. PAK4 is prominently involved in generating tumor drug resistance via multiple pathways. It can confer cis-diamminedichloroplatinum (CDDP) resistance via activation of the mitogen-activated protein kinase kinase (MEK)/ERK and PI3K/Akt pathways in GC cells [4]. Similarly, PAK4 contributes to CDDP resistance via the PI3K/AKT pathway in cervical cancer (CC) cells [84]. As a predictive marker of gemcitabine (Gem) sensitivity, PAK4 is overexpressed in Gem drug-resistant PC cells, while silencing PAK4 restores Gem sensitivity [85]. In addition to the high expression of PAK4, p-Bad protein content was also shown to increase in drug-resistant PC cells. Importantly, silencing PAK4 by PAK4 and nicotinamide phosphoribosyl-transferase (NAMPT) dual inhibitor KPT–9274 reduces p-Bad, leading to the resensitization of Gem-drug-resistant PC cells. This suggests that the PAK4/p-Bad pathway is a positive factor in developing Gem resistance [86]. A recent study has also demonstrated that the novel PAK4 inhibitor, PAKib, significantly enhances the inhibition of Gem on PC [87].
4.6 PAK4 and antitumor immune responses
As a new therapy for tumors, immunotherapy shows great potential. At the present stage, cancer immunotherapy mainly includes programmed death 1 (PD–1)/programmed death-ligand 1(PD–L1) and chimeric antigen receptor T cell-related therapy [88]. PAK4, a target of tumor immune escape, is of high importance to regulating antitumor immunity. The multi-omics analysis of bioinformatics revealed that PAK4 is rich in the “T cell receptor signaling pathway” regulates macrophage polarization through heat shock protein 105 kDa (HSPH1), and significantly impacts neoantigen production and tumor immune regulation [89]. PD–1 is expressed on T cells, whereas PD–L1 is expressed on antigen-presenting cells and cancer cells. The combination of the two can inhibit the antitumor immune-mediated by T cells [5]. Thus, the PD–1/PD–L1 pathway is considered a vital cause of antitumor immunosuppression. Activation of PAK4 improves tumor resistance to PD–1 blockade. PAK4 is overexpressed in melanoma with poor T cell infiltration and facilitates the activation of the WNT/β-catenin pathway [90]. The activated intranuclear β-catenin interacts with TCF/LEF to upregulate the expression of PD–L1 [5]. Furthermore, tumors with genetic PAK4 deletion are more sensitive to PD–1 blockade, and the tumor can completely subside under the action of anti-PD–1 drugs. Likewise, the efficacy of anti-PD–1 treatment was improved as impacted by PAK4 and NAMPT dual inhibitor KPT–9274 [90]. There has been remarkable results of CAR-T therapy for the treatment of hematological tumors (e.g., refractory B cell malignancy) [91]. However, since the tumor microenvironment with abnormal blood vessels inhibits T cell infiltration and activation, CAR-T has no significant effect on solid tumors [92]. PAK4, as a selective regulator of genetic reprogramming and abnormal tumor blood vessels, plays a vital role in tumor resistance to CAR-T. PAK4 improves myocyte enhancer factor 2D to be combined with Zinc finger E-box binding homeobox 1 (ZEB1) promoter and induces the expression of ZEB1, which inhibits claudin-14’s transcription, thus improving vascular permeability. Moreover, PAK4 can induce SLUG expression, downregulate the expression of intercellular adhesion molecule–1/Vascular Cell Adhesion Molecule–1, and reduce the adhesion ability of T cells. Accordingly, in glioblastoma, the inhibition of PAK4 can repair the microenvironment of abnormal tumor blood vessels and increase the efficacy of CAR-T [93].
5 Clinical and pathological significance
Although tumor surgery and chemotherapy technologies have developed tremendously, patient prognosis is still unsatisfactory due to tumor metastasis and spread [94,95]. Therefore, identifying tumor biomarkers to evaluate and improve the prognostic effect has become a hot research area. As a popular tumor biomarker, PAK4 plays a vital role in the prognosis of patients with various tumors in the digestive system [33], reproductive system [47], and respiratory system [96]. Exhibiting significant clinical and pathological effects, PAK4 is not only highly expressed in a variety of tumor cells as a possible diagnostic and therapeutic target but also related to clinicopathological indicators such as depth of invasion or tumor size, lymph node metastasis, distant metastasis, tumor stage, histological features, and prognosis (Table 1). Among the digestive system tumors, PAK4 plays a vital role in evaluating clinical and pathological indicators. One study demonstrated that patients with high PAK4 expression in OSCC tend to have poor overall survival (OS) rates and that tumors with high PAK4 expression have a larger volume and deeper invasion depth [33]. In GC, high PAK4 expression is not only associated with deeper invasion depth but also with more severe lymph node metastasis, distant metastasis, advanced tumor stage, and recurrence. Therefore, it is not difficult to understand why the disease-specific survival rate and relapse-free survival (RFS) rate of patients with high PAK4 expression are poor [97]. In colorectal cancer (CRC), high PAK4 expression is closely related to serous layer infiltration and advanced tumor stage but has no connection with lymph node metastasis and disease-free survival (DFS) [98]. Another study showed that high expression of PAK4 in COAD is associated with advanced tumor stage and histological grade, but not with the depth of invasion, lymph node metastasis, or distant metastasis [46]. The fact that high expression of PAK4 is not related to the depth of invasion seems to be inconsistent with the fact that high expression of PAK4 is related to serous layer infiltration; so whether the difference in the scope of inclusion of the two samples (the former was CRC and the latter COAD) or the limited number of samples caused the differences remains to be explored. It is also worth noting that contrary to the poor prognosis associated with high PAK4 expression, for PC patients, PAK4-negativeness is closely related to poor OS and DFS. PAK4-negative PC exhibits larger tumor volume and poor histological differentiation. However, the samples for this study came from patients who may have already been cured, and the number of samples was limited. For the general PC population, the question of whether low PAK4 expression indicates a poor prognosis still needs further investigation [99]. PAK4 has also shown its clinical and pathological importance in reproductive system tumors. In BC, high PAK4 expression usually leads to poor OS and DFS and is closely related to larger tumor volume, more lymph node metastasis, and advanced AJCC stage, but it is not associated with histological grade [47]. Another study showed that PAK4 gradually increased with the progression of BC (advanced invasiveness > early invasiveness > noninvasiveness) [100]. For patients with OC, the activation and overexpression of PAK4 are often associated with poor prognosis. The OS and DFS period of patients with overexpressed PAK4 of OC are shorter. Not only that, PAK4 is linked to the advanced FIGO stage and poor histological grade [49]. Similarly, in CC, high expression of PAK4 is also related to poor OS, lymph node metastasis, distant metastasis, advanced FIGO stage, and poor histological grade [84]. It is interesting to note that low PAK4 expression predicts a poor prognosis for patients with EC. Low PAK4 expression is also closely related to deeper myometrial invasion, advanced FIGO grade, and histological grade. These findings contradict those of the abovementioned studies, namely that high expression of PAK4 is linked to poor prognosis in other reproductive system tumors. The endometrium is a periodic proliferation tissue sensitive to hormone regulation. Whether the contradictory result is related to the unique biological environment of the endometrium is a question worthy of further study [101]. In addition, in non-small cell lung cancer (NSCLC), a respiratory tumor, PAK4 is considered to be an independent predictor of poor survival. NSCLC patients with upregulated PAK4 tend to have a shorter OS period, and high PAK4 expression is closely related to lymph node metastasis, distant metastasis, and advanced tumor stage [96].
Association between the expression of PAK4 and clinicopathological factors
Tumor | PAK4 | T (depth of invasion or tumor size) | N (regional lymph node metastasis) | M (distant metastasis) | Tumor stage | Survival | Histological feature | References |
---|---|---|---|---|---|---|---|---|
OSCC | + | T3–4, P = 0.0112 | P = 0.126 | P = 0.1052 | P = 0.1048 | Poor OS, P = 0.0164 | Differentiated degree, P = 0.1486 | [33] |
GC | + | T2–4, P < 0.001 | N1–3, P < 0.001 | M1, P < 0.001 | II–IV, P < 0.001 | Poor DSS, P < 0.001; Poor RFS, P < 0.001 | Differentiated degree, P = 0.215 | [97] |
CRC | + | T3–4, P = 0.012 | P = 0.753 | III–IV, P = 0.004 | Poor DFS, P = 0.234 | Differentiated degree, P = 0.307 | [98] | |
COAD | + | P = 0.656 | P = 0.425 | P = 0.543 | III–IV (pStage), P = 0.02 | Histological grade, P = 0.003 | [46] | |
BC | + | T2–3, P = 0.007 | N2–3(pN), P = 0.027 | III (AJCC stage), P = 0.012 | Poor OS, P = 0.003; Poor DFS, P = 0.001 | Histological grade, P = 0.230 | [47] | |
OC | + | III–IV (FIGO Stage), P < 0.05 | Poor OS, P < 0.05; Poor DFS, P < 0.05 | G3 (Histological grade), P < 0.05 | [49] | |||
CC | + | P = 0.59763 | N1, P = 0.00525 | M1, P = 0.02105 | IIb–IIIb (FIGO Stage), P = 0.00822 | Poor OS, P = 0.0014 | G3 (Histological grade), P = 0.03464 | [84] |
NSCLC | + | P = 0.757 | N2–3, P = 0.034 | M1, P = 0.022 | III–IV, P = 0.042 | Poor OS, P = 0.002 | Differentiated degree, P = 0.020 | [96] |
PC | - | All T3–4 | Poor OS, P = 0.003; Poor DFS, P = 0.016 | Differentiated degree, P < 0.001 | [99] | |||
EC | - | III–IV (FIGO Stage), P = 0.017 | Poor OS, P = 0.026 | G3 (Histological grade), P < 0.05 | [101] |
OSCC: oral squamous cell carcinoma; GC: gastric cancer; CRC: colorectal cancer; COAD: colon adenocarcinoma (colon cancer); BC: breast cancer; OC: ovarian cancer; CC: cervical cancer. NSCLC: non-small cell lung cancer; PC: pancreatic cancer; EC: endometrial cancer; OS: overall survival; DSS: disease-specific survival; RFS: relapse-free survival; DFS: disease-free survival.
All p values in bold mean “<0.05”, indicating significant differences.
6 Conclusions
The influence of PAK4 runs throughout the genesis and development of tumors. From preneoplastic cells losing homeostasis to tumor cells with continuous proliferation and from migration and invasion to treatment-resistant tumor cells, complex biological processes and rich signaling involve PAK4 (Figure 3). Activated PAK4 promotes the proliferation [47], migration, invasion [51], and treatment resistance [86,93] of tumor cells through the activation of various signaling pathways. Afterward, these cell activities and characteristics can lead to volume increase, diffusion, metastasis, and reduced sensitivity to chemotherapy and immunotherapy of tumor tissues, because of which the prognosis of tumor patients with high PAK4 expression is often poor. PAK4 is also minimally expressed in normal tissues, while it is overexpressed in various tumors and thus regarded as a tumor marker and treatment target [61]. Also, PAK4 is closely related to tumor infiltration depth, lymph node metastasis, stage classification, and other pathological indicators [97], whose clinical application prospects are broad.

PAK4 signaling pathways. PAK4 promotes cellular proliferation (top). PAK4 regulates cell mitosis to facilitate proliferation by phosphorylating Ran and activating the LIMK1/cofilin–1 pathway. PAK4 improves the binding of Mdm2 with P53 to facilitate the ubiquitination and degradation of P53 and increases G6PD activity. Furthermore, PAK4 promotes ubiquitination degradation of P57 kip2 and P21 degradation. PAK4 and ERα activate each other, thus forming a positive feedback loop to upregulate the expression of CyclinD1. PAK4 activates the c-Src/EGFR pathway and upregulates the expressions of CyclinD1 and CDC25A. In addition, PAK4 phosphorylates β-catenin and enhances its nuclear import. Endonuclear β-catenin and TCF/LEF work jointly to upregulate the expressions of c-myc and CyclinD1. PAK4 promotes cellular metastasis (right). PAK4 interacts with DGCR6L to improve the phosphorylation level of LIMK1 and cofilin. PAK4 facilitates metastasis by activating CEBPB/CLDN4 and PI3K/Akt pathways, inhibiting P53, activating SCG10 and N–WASP, and inhibiting RhoA exchange activity mediated by GEF–H1 in a kinase-dependent manner. Furthermore, PAK4 protects RhoU from ubiquitination while binding to and activating eEF1A1 in a kinase-independent manner. PAK4 phosphorylates the integrin β5 subunit while inhibiting the activity of integrin. The connection between integrin and VN activates PAK4, thus regulating the cell movement through the negative feedback loop. PAK4 phosphorylates paxillin and facilitates the decomposition of adhesive spots. Induced by activated CDC42, PAK4 induces filamentous pseudopodia formation. Moreover, PAK4 inhibits pseudopodia maturation mediated by PDZ-RhoGEF. PAK4 promotes drug resistance and regulates cellular aging (bottom right). Phosphorylation of Bad by PAK4 endows tumors with Gem resistance. Meanwhile, CDDP resistance is imparted via PI3K/Akt and MEK/ERK pathways. In addition, PAK4 inhibits the senescence-like growth arrest of cells mediated by the RELB-C/EBPβ pathway. PAK4 mediates cell senescence by activating the MEK/ERK pathway and the P16 INK4/P19 ARF. PAK4 inhibits cellular apoptosis (bottom). PAK4 inhibits the activity of Bad by phosphorylation. It can prevent the release of cytochrome C in mitochondria and inhibit the activation of caspase–9/Caspase–3. In addition, PAK4 can inhibit apoptosis by inhibiting caspase–8. PAK4 facilitates the binding of TRADD to TNFR1 while inhibiting apoptosis under TNF-α inducement and NF-κB mediation. Moreover, PAK4 activates both PI3K/Akt and MEK/ERK pathways, thus having an effect on NF-κB so as to inhibit apoptosis. PAK4 promotes oncogenic transformation (bottom left). PAK4 interacts with Smad2/3 in a kinase-independent manner to block TGF-β induced phosphorylation of Smad2/3. Besides, PAK4 phosphorylates Smad2 in response to HGF, thus mediating the ubiquitination of Smad2. PAK4 upregulates FoxC2 expression and downregulates ParvB expression. PAK4 inhibits antitumor immunity (left). PAK4 induces the expressions of ZEB1 and SLUG, thus inhibiting the expressions of Claudin–14 and ICAM–1/VCAM–1, respectively. PAK4 upregulates the expression of PD–L1 via the β-catenin pathway. Activation and inhibition of proteins are indicated by arrows and blocked lines, respectively. P: phosphorylation; Ub: ubiquitination; LIMK1: LIM domain kinase 1; Mdm2: murine double minute 2; G6PD: glucose–6-phosphate dehydrogenase; ERα: estrogen receptor alpha; EGFR: epidermal growth factor receptor; CDC25A: cell division cycle 25A; TCF: T-cell factor; LEF: lymphoid enhancer factor; DGCR6L: DiGeorge critical region 6L; CEBPB: CCAAT/enhancer-binding protein beta; CLDN4: Claudin 4; PI3K: phosphoinositide 3-kinase; AKT: protein kinase B; SCG10: superior cervical ganglia 10; N-WASP: neural Wiskott-Aldrich syndrome protein; GEF: guanosine exchange factor; eEF1A1: eukaryotic elongation factor 1 alpha; VN: vitronectin; PDZ-RhoGEF: postsynaptic density protein 95/disc-large/zonula occludens-RhoGEF; Gem: gemcitabine; CDDP: cis-diamminedichloroplatinum; MEK: mitogen-activated protein kinase; ERK: extracellular-signal regulated protein kinase; TNFR1: tumor necrosis factor alpha receptor 1; NF-κB: nuclear factor-κB; TGF-β: transforming growth factor beta; HGF: hepatocyte growth factor; Smad: small mother against decapentaplegic; FoxC2: Forkhead Box C2; ParvB: Parvin Beta; ZEB1: Zinc finger E-box binding homeobox 1; ICAM–1: intercellular adhesion molecule–1; VCAM–1: vascular cell adhesion molecule–1; PD–L1: programmed death ligand 1.
At the moment, PAK4 has been studied extensively in relation to tumor biology function, but there is still conflicting information regarding its role in cell aging. It is contradictory that PAK4 inhibits aging by inhibiting RELB and mediates premature cell aging by activating the ERK pathway and cell cycle inhibitors P16 INK4 and P19 ARF [63,64]. Drug resistance and immune escape of tumors have been reported as the major challenges of chemotherapy and immunotherapy [83,102]. PAK4 resists drugs, and antitumor immune response based on multiple mechanisms, which has been found as a novel target to solve the low efficiency of chemotherapy and immunotherapy [5]. KPT–9274 stands out in targeting PAK4 to improve the efficacy of tumor therapy. KPT–9274 is recognized as a dual inhibitor of PAK4 and NAMPT, which is capable of effectively inhibiting the growth of mantle cell lymphoma, follicular lymphoma, and diffuse large B-cell lymphoma [103]. Moreover, it can significantly improve the efficacy of chemotherapy and immunotherapy in cooperation with GEM or anti-PD-1 treatment [86,90], which can be considered a novel direction in the combined treatment of tumors. However, PAK4 is not the specific target of KPT–9274, and the effect of the inhibition of PAK4 should be studied in depth. Ideal tumor markers need high sensitivity and good specificity. They are also related to tumor size, stage, and severity and can be used to detect tumor localization and efficacy [104,105,106]. Although PAK4 is related to tumor size, stage, and severity, it is overexpressed in various tumors with poor specificity [33,84,96]. Meanwhile, its sensitivity for detecting tumor positioning and tumor efficacy has not been determined. We expect high expression of PAK4 to indicate poor patient prognosis in tumor cases, but in PC [99] and EC [101], contrary to most findings, low PAK4 expression seems to be closely linked with poor prognosis. However, this result was not conclusive and should be confirmed by further research.
To summarize, PAK4 plays an important role in tumor occurrence and development and is important in prognosis and as a tumor biomarker. This article summarizes the role of PAK4 in tumor cytogenesis, proliferation, survival, migration invasion, and drug resistance. We also highlight its clinical pathological significance to benefit preclinical and clinical research on PAK4 and tumors.
-
Funding information: This work is supported by the National Natural Science Foundation of China (81230077) and the Science Foundation supported by the Educational Department of Liaoning Province (JC2019030).
-
Author contributions: X.Y. designed the study and performed the writing of the full text. C.H., J.L., and X.S. participated in the collection of literature. X.L. critically revised the work.
-
Conflict of interest: Authors state no conflict of interest.
-
Data availability statement: Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.
References
[1] Wong LE, Chen N, Karantza V, Minden A. The Pak4 protein kinase is required for oncogenic transformation of MDA-MB-231 breast cancer cells. Oncogenesis. 2013;2(6):e50.10.1038/oncsis.2013.13Search in Google Scholar
[2] Wang Y, Shao F, Chen L. miR-425 regulates ovarian cancer proliferation, metastasis, and apoptosis by repressing PAK4 expression. Asia Pac J Clin Oncol. 2022;18(1):76–83.10.1111/ajco.13519Search in Google Scholar
[3] Fu Y, Fang L, Yin Q, Wu Q, Sui W, Sun Y, et al. Interfering with pak4 protein expression affects osteosarcoma cell proliferation and migration. Biomed Res Int. 2021;2021:9977001.10.1155/2021/9977001Search in Google Scholar
[4] 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(2):e00094.10.1042/BSR20130102Search in Google Scholar
[5] Naїja A, Merhi M, Inchakalody V, Fernandes Q, Mestiri S, Prabhu KS, et al. The role of PAK4 in the immune system and its potential implication in cancer immunotherapy. Cell Immunol. 2021;367:104408.10.1016/j.cellimm.2021.104408Search in Google Scholar
[6] Thillai K, Sarker D, Wells C. PAK4 pathway as a potential therapeutic target in pancreatic cancer. Future Oncol. 2018;14(7):579–82.10.2217/fon-2017-0458Search in Google Scholar
[7] Rane CK, Minden A. P21 activated kinases: structure, regulation, and functions. Small GTPases. 2014;5:e28003.10.4161/sgtp.28003Search in Google Scholar
[8] Kumar R, Sanawar R, Li X, Li F. Structure, biochemistry, and biology of PAK kinases. Gene. 2017;605:20–31.10.1016/j.gene.2016.12.014Search in Google Scholar
[9] Wells CM, Jones GE. The emerging importance of group II PAKs. Biochem J. 2010;425(3):465–73.10.1042/BJ20091173Search in Google Scholar
[10] Jaffer ZM, Chernoff J. p21-activated kinases: three more join the Pak. Int J Biochem Cell Biol. 2002;34(7):713–7.10.1016/S1357-2725(01)00158-3Search in Google Scholar
[11] Baldassa S, Calogero AM, Colombo G, Zippel R, Gnesutta N. N-terminal interaction domain implicates PAK4 in translational regulation and reveals novel cellular localization signals. J Cell Physiol. 2010;224(3):722–33.10.1002/jcp.22172Search in Google Scholar
[12] Zhang H, Li Z, Viklund EK, Strömblad S. P21-activated kinase 4 interacts with integrin alpha v beta 5 and regulates alpha v beta 5-mediated cell migration. J Cell Biol. 2002;158(7):1287–97.10.1083/jcb.200207008Search in Google Scholar
[13] Ha BH, Davis MJ, Chen C, Lou HJ, Gao J, Zhang R, et al. Type II p21-activated kinases (PAKs) are regulated by an autoinhibitory pseudosubstrate. Proc Natl Acad Sci U S A. 2012;109(40):16107–12.10.1073/pnas.1214447109Search in Google Scholar
[14] Eswaran J, Lee WH, Debreczeni JE, Filippakopoulos P, Turnbull A, Fedorov O, et al. Crystal Structures of the p21-activated kinases PAK4, PAK5, and PAK6 reveal catalytic domain plasticity of active group II PAKs. Structure. 2007;15(2):201–13.10.1016/j.str.2007.01.001Search in Google Scholar
[15] Lei M, Lu W, Meng W, Parrini MC, Eck MJ, Mayer BJ, et al. Structure of PAK1 in an autoinhibited conformation reveals a multistage activation switch. Cell. 2000;102(3):387–97.10.1016/S0092-8674(00)00043-XSearch in Google Scholar
[16] Pirruccello M, Sondermann H, Pelton JG, Pellicena P, Hoelz A, Chernoff J, et al. A dimeric kinase assembly underlying autophosphorylation in the p21 activated kinases. J Mol Biol. 2006;361(2):312–26.10.1016/j.jmb.2006.06.017Search in Google Scholar
[17] Arias-Romero LE, Chernoff J. A tale of two PAKs. Biol Cell. 2008;100(2):97–108.10.1042/BC20070109Search in Google Scholar
[18] Buchwald G, Hostinova E, Rudolph MG, Kraemer A, Sickmann A, Meyer HE, et al. Conformational switch and role of phosphorylation in PAK activation. Mol Cell Biol. 2001;21(15):5179–89.10.1128/MCB.21.15.5179-5189.2001Search in Google Scholar
[19] Parrini MC, Lei M, Harrison SC, Mayer BJ. Pak1 kinase homodimers are autoinhibited in trans and dissociated upon activation by Cdc42 and Rac1. Mol Cell. 2002;9(1):73–83.10.1016/S1097-2765(01)00428-2Search in Google Scholar
[20] Strochlic TI, Viaud J, Rennefahrt UE, Anastassiadis T, Peterson JR. Phosphoinositides are essential coactivators for p21-activated kinase 1. Mol Cell. 2010;40(3):493–500.10.1016/j.molcel.2010.10.015Search in Google Scholar PubMed PubMed Central
[21] Galisteo ML, Chernoff J, Su YC, Skolnik EY, Schlessinger J. The adaptor protein Nck links receptor tyrosine kinases with the serine-threonine kinase Pak1. J Biol Chem. 1996;271(35):20997–1000.10.1074/jbc.271.35.20997Search in Google Scholar
[22] King CC, Gardiner EM, Zenke FT, Bohl BP, Newton AC, Hemmings BA, et al. p21-activated kinase (PAK1) is phosphorylated and activated by 3-phosphoinositide-dependent kinase1 (PDK1). J Biol Chem. 2000;275(52):41201–9.10.1074/jbc.M006553200Search in Google Scholar
[23] Manser E, Loo TH, Koh CG, Zhao ZS, Chen XQ, Tan L, et al. PAK kinases are directly coupled to the PIX family of nucleotide exchange factors. Mol Cell. 1998;1(2):183–92.10.1016/S1097-2765(00)80019-2Search in Google Scholar
[24] Baskaran Y, Ng YW, Selamat W, Ling FT, Manser E. Group I and II mammalian PAKs have different modes of activation by Cdc42. EMBO Rep. 2012;13(7):653–9.10.1038/embor.2012.75Search in Google Scholar
[25] Chong C, Tan L, Lim L, Manser E. The mechanism of PAK activation. Autophosphorylation events in both regulatory and kinase domains control activity. J Biol Chem. 2001;276(20):17347–53.10.1074/jbc.M009316200Search in Google Scholar
[26] Johnson LN, Noble ME, Owen DJ. Active and inactive protein kinases: structural basis for regulation. Cell. 1996;85(2):149–58.10.1016/S0092-8674(00)81092-2Search in Google Scholar
[27] Abo A, Qu J, Cammarano MS, Dan C, Fritsch A, Baud V, et al. PAK4, a novel effector for Cdc42Hs, is implicated in the reorganization of the actin cytoskeleton and in the formation of filopodia. EMBO J. 1998;17(22):6527–40.10.1093/emboj/17.22.6527Search in Google Scholar PubMed PubMed Central
[28] Wells CM, Abo A, Ridley AJ. PAK4 is activated via PI3K in HGF-stimulated epithelial cells. J Cell Sci. 2002;115(Pt 20):3947–56.10.1242/jcs.00080Search in Google Scholar PubMed
[29] Callow MG, Clairvoyant F, Zhu S, Schryver B, Whyte DB, Bischoff JR, et al. Requirement for PAK4 in the anchorage-independent growth of human cancer cell lines. J Biol Chem. 2002;277(1):550–8.10.1074/jbc.M105732200Search in Google Scholar PubMed
[30] Rane CK, Patel M, Cai L, Senapedis W, Baloglu E, Minden A. Decrypting the PAK4 transcriptome profile in mammary tumor forming cells using next generation sequencing. Genomics. 2017;S0888–7543(17):30128–3.10.1016/j.ygeno.2017.10.004Search in Google Scholar PubMed
[31] Liu Y, Chen N, Cui X, Zheng X, Deng L, Price S, et al. The protein kinase Pak4 disrupts mammary acinar architecture and promotes mammary tumorigenesis. Oncogene. 2010;29(44):5883–94.10.1038/onc.2010.329Search in Google Scholar PubMed PubMed Central
[32] Chen S, Auletta T, Dovirak O, Hutter C, Kuntz K, El-ftesi S, et al. Copy number alterations in pancreatic cancer identify recurrent PAK4 amplification. Cancer Biol Ther. 2008;7(11):1793–802.10.4161/cbt.7.11.6840Search in Google Scholar PubMed PubMed Central
[33] Begum A, Imoto I, Kozaki K, Tsuda H, Suzuki E, Amagasa T, et al. Identification of PAK4 as a putative target gene for amplification within 19q13.12–q13.2 in oral squamous-cell carcinoma. Cancer Sci. 2009;100(10):1908–16.10.1111/j.1349-7006.2009.01252.xSearch in Google Scholar PubMed
[34] Thullberg M, Strömblad S. Anchorage-independent cytokinesis as part of oncogenic transformation? Cell Cycle. 2008;7(8):984–8.10.4161/cc.7.8.5674Search in Google Scholar PubMed
[35] Sepulveda JL, Wu C. The parvins. Cell Mol Life Sci. 2006;63(1):25–35.10.1007/s00018-005-5355-1Search in Google Scholar PubMed PubMed Central
[36] Puisieux A, Brabletz T, Caramel J. Oncogenic roles of EMT-inducing transcription factors. Nat Cell Biol. 2014;16(6):488–94.10.1038/ncb2976Search in Google Scholar PubMed
[37] Li C, Ding H, Tian J, Wu L, Wang Y, Xing Y, et al. Forkhead box protein C2 promotes epithelial-mesenchymal transition, migration and invasion in cisplatin-resistant human ovarian cancer cell line (SKOV3/CDDP). Cell Physiol Biochem. 2016;39(3):1098–110.10.1159/000447818Search in Google Scholar PubMed
[38] Macara IG, McCaffrey L. Cell polarity in morphogenesis and metastasis. Philos Trans R Soc Lond B Biol Sci. 2013;368(1629):20130012.10.1098/rstb.2013.0012Search in Google Scholar PubMed PubMed Central
[39] Yang L, Moses HL. Transforming growth factor beta: tumor suppressor or promoter? Are host immune cells the answer? Cancer Res. 2008;68(22):9107–11.10.1158/0008-5472.CAN-08-2556Search in Google Scholar PubMed PubMed Central
[40] Wang C, Li Y, Zhang H, Liu F, Cheng Z, Wang D, et al. Oncogenic PAK4 regulates Smad2/3 axis involving gastric tumorigenesis. Oncogene. 2014;33(26):3473–84.10.1038/onc.2013.300Search in Google Scholar PubMed
[41] Bompard G, Rabeharivelo G, Cau J, Abrieu A, Delsert C, Morin N. P21-activated kinase 4 (PAK4) is required for metaphase spindle positioning and anchoring. Oncogene. 2013;32(7):910–9.10.1038/onc.2012.98Search in Google Scholar PubMed
[42] Nekrasova T, Minden A. PAK4 is required for regulation of the cell-cycle regulatory protein p21, and for control of cell-cycle progression. J Cell Biochem. 2011;112(7):1795–806.10.1002/jcb.23092Search in Google Scholar PubMed
[43] Bompard G, Rabeharivelo G, Frank M, Cau J, Delsert C, Morin N. Subgroup II PAK-mediated phosphorylation regulates Ran activity during mitosis. J Cell Biol. 2010;190(5):807–22.10.1083/jcb.200912056Search in Google Scholar PubMed PubMed Central
[44] Li Y, Wang D, Zhang H, Wang C, Dai W, Cheng Z, et al. P21-activated kinase 4 regulates the cyclin-dependent kinase inhibitor p57(kip2) in human breast cancer. Anat Rec (Hoboken). 2013;296(10):1561–7.10.1002/ar.22754Search in Google Scholar PubMed
[45] Cao L, Wu J, Qu X, Sheng J, Cui M, Liu S, et al. Glycometabolic rearrangements-aerobic glycolysis in pancreatic cancer: causes, characteristics and clinical applications. J Exp Clin Cancer Res. 2020;39(1):267.10.1186/s13046-020-01765-xSearch in Google Scholar PubMed PubMed Central
[46] Zhang X, Zhang X, Li Y, Shao Y, Xiao J, Zhu G, et al. PAK4 regulates G6PD activity by p53 degradation involving colon cancer cell growth. Cell Death Dis. 2017;8(5):e2820.10.1038/cddis.2017.85Search in Google Scholar PubMed PubMed Central
[47] He LF, Xu HW, Chen M, Xian ZR, Wen XF, Chen MN, et al. Activated-PAK4 predicts worse prognosis in breast cancer and promotes tumorigenesis through activation of PI3K/AKT signaling. Oncotarget. 2017;8(11):17573–85.10.18632/oncotarget.7466Search in Google Scholar PubMed PubMed Central
[48] Su T, Qu JJ, Wang K, Li BL, Zhao D, Zhu YP, et al. Cross-talk between p21-activated kinase 4 and ERα signaling triggers endometrial cancer cell proliferation. Oncotarget. 2017;8(40):68083–94.10.18632/oncotarget.19188Search in Google Scholar PubMed PubMed Central
[49] Siu MK, Chan HY, Kong DS, Wong ES, Wong OG, Ngan HY, et al. p21-activated kinase 4 regulates ovarian cancer cell proliferation, migration, and invasion and contributes to poor prognosis in patients. Proc Natl Acad Sci U S A. 2010;107(43):18622–7.10.1073/pnas.0907481107Search in Google Scholar PubMed PubMed Central
[50] Li Y, Shao Y, Tong Y, Shen T, Zhang J, Li Y, et al. Nucleo-cytoplasmic shuttling of PAK4 modulates β-catenin intracellular translocation and signaling. Biochim Biophys Acta. 2012;1823(2):465–75.10.1016/j.bbamcr.2011.11.013Search in Google Scholar PubMed
[51] Li ZF, Yao YD, Zhao YY, Liu Y, Liu ZH, Hu P, et al. Effects of PAK4/LIMK1/Cofilin–1 signaling pathway on proliferation, invasion, and migration of human osteosarcoma cells. J Clin Lab Anal. 2020;34(9):e23362.10.1002/jcla.23362Search in Google Scholar PubMed PubMed Central
[52] Giardino E, Catalano R, Barbieri AM, Treppiedi D, Mangili F, Spada A, et al. Cofilin is a mediator of RET-promoted medullary thyroid carcinoma cell migration, invasion and proliferation. Mol Cell Endocrinol. 2019;495:110519.10.1016/j.mce.2019.110519Search in Google Scholar PubMed
[53] Manning FC, Patierno SR. Apoptosis: inhibitor or instigator of carcinogenesis? Cancer Invest. 1996;14(5):455–65.10.3109/07357909609018903Search in Google Scholar PubMed
[54] Provinciali M, Cardelli M, Marchegiani F, Pierpaoli E. Impact of cellular senescence in aging and cancer. Curr Pharm Des. 2013;19(9):1699–709.10.2174/1381612811319090017Search in Google Scholar
[55] Abraha AM, Ketema EB. Apoptotic pathways as a therapeutic target for colorectal cancer treatment. World J Gastrointest Oncol. 2016;8(8):583–91.10.4251/wjgo.v8.i8.583Search in Google Scholar PubMed PubMed Central
[56] Megison ML, Gillory LA, Beierle EA. Cell survival signaling in neuroblastoma. Anticancer Agents Med Chem. 2013;13(4):563–75.10.2174/1871520611313040005Search in Google Scholar PubMed PubMed Central
[57] Parrish AB, Freel CD, Kornbluth S. Cellular mechanisms controlling caspase activation and function. Cold Spring Harb Perspect Biol. 2013;5(6):a008672.10.1101/cshperspect.a008672Search in Google Scholar PubMed PubMed Central
[58] Gnesutta N, Qu J, Minden A. The serine/threonine kinase PAK4 prevents caspase activation and protects cells from apoptosis. J Biol Chem. 2001;276(17):14414–9.10.1074/jbc.M011046200Search in Google Scholar PubMed
[59] Gnesutta N, Minden A. Death receptor-induced activation of initiator caspase 8 is antagonized by serine/threonine kinase PAK4. Mol Cell Biol. 2003;23(21):7838–48.10.1128/MCB.23.21.7838-7848.2003Search in Google Scholar PubMed PubMed Central
[60] Li X, Minden A. PAK4 functions in tumor necrosis factor (TNF) alpha-induced survival pathways by facilitating TRADD binding to the TNF receptor. J Biol Chem. 2005;280(50):41192–200.10.1074/jbc.M506884200Search in Google Scholar PubMed
[61] Tyagi N, Bhardwaj A, Singh AP, McClellan S, Carter JE, Singh S. p-21 activated kinase 4 promotes proliferation and survival of pancreatic cancer cells through AKT- and ERK-dependent activation of NF-κB pathway. Oncotarget. 2014;5(18):8778–89.10.18632/oncotarget.2398Search in Google Scholar PubMed PubMed Central
[62] Vargas J, Feltes BC, Poloni Jde F, Lenz G, Bonatto D. Senescence; an endogenous anticancer mechanism. Front Biosci (Landmark Ed). 2012;17:2616–43.10.2741/4074Search in Google Scholar PubMed
[63] Costa TDF, Zhuang T, Lorent J, Turco E, Olofsson H, Masia-Balague M, et al. PAK4 suppresses RELB to prevent senescence-like growth arrest in breast cancer. Nat Commun. 2019;10(1):3589.10.1038/s41467-019-11510-4Search in Google Scholar PubMed PubMed Central
[64] Cammarano MS, Nekrasova T, Noel B, Minden A. Pak4 induces premature senescence via a pathway requiring p16INK4/p19ARF and mitogen-activated protein kinase signaling. Mol Cell Biol. 2005;25(21):9532–42.10.1128/MCB.25.21.9532-9542.2005Search in Google Scholar PubMed PubMed Central
[65] Lambert AW, Pattabiraman DR, Weinberg RA. Emerging biological principles of metastasis. Cell. 2017;168(4):670–91.10.1016/j.cell.2016.11.037Search in Google Scholar PubMed PubMed Central
[66] Krakhmal NV, Zavyalova MV, Denisov EV, Vtorushin SV, Perelmuter VM. Cancer invasion: patterns and mechanisms. Acta Naturae. 2015;7(2):17–28.10.32607/20758251-2015-7-2-17-28Search in Google Scholar
[67] Li Z, Zhang H, Lundin L, Thullberg M, Liu Y, Wang Y, et al. p21-activated kinase 4 phosphorylation of integrin beta5 Ser-759 and Ser-762 regulates cell migration. J Biol Chem. 2010;285(31):23699–710.10.1074/jbc.M110.123497Search in Google Scholar PubMed PubMed Central
[68] Li Z, Lock JG, Olofsson H, Kowalewski JM, Teller S, Liu Y, et al. Integrin-mediated cell attachment induces a PAK4-dependent feedback loop regulating cell adhesion through modified integrin alpha v beta 5 clustering and turnover. Mol Biol Cell. 2010;21(19):3317–29.10.1091/mbc.e10-03-0245Search in Google Scholar PubMed PubMed Central
[69] Yam JW, Tse EY, Ng IO. Role and significance of focal adhesion proteins in hepatocellular carcinoma. J Gastroenterol Hepatol. 2009;24(4):520–30.10.1111/j.1440-1746.2009.05813.xSearch in Google Scholar PubMed
[70] Wells CM, Whale AD, Parsons M, Masters JR, Jones GE. PAK4: a pluripotent kinase that regulates prostate cancer cell adhesion. J Cell Sci. 2010;123(Pt 10):1663–73.10.1242/jcs.055707Search in Google Scholar PubMed PubMed Central
[71] Dart AE, Box GM, Court W, Gale ME, Brown JP, Pinder SE, et al. PAK4 promotes kinase-independent stabilization of RhoU to modulate cell adhesion. J Cell Biol. 2015;211(4):863–79.10.1083/jcb.201501072Search in Google Scholar PubMed PubMed Central
[72] Zhao M, Spiess M, Johansson HJ, Olofsson H, Hu J, Lehtiö J, et al. Identification of the PAK4 interactome reveals PAK4 phosphorylation of N-WASP and promotion of Arp2/3-dependent actin polymerization. Oncotarget. 2017;8(44):77061–74.10.18632/oncotarget.20352Search in Google Scholar PubMed PubMed Central
[73] McNiven MA, Baldassarre M, Buccione R. The role of dynamin in the assembly and function of podosomes and invadopodia. Front Biosci. 2004;9:1944–53.10.2741/1348Search in Google Scholar PubMed
[74] Nicholas NS, Pipili A, Lesjak MS, Ameer-Beg SM, Geh JL, Healy C, et al. PAK4 suppresses PDZ-RhoGEF activity to drive invadopodia maturation in melanoma cells. Oncotarget. 2016;7(43):70881–97.10.18632/oncotarget.12282Search in Google Scholar PubMed PubMed Central
[75] Tsai WC, Chen CL, Chen HC. Protein tyrosine phosphatase SHP2 promotes invadopodia formation through suppression of Rho signaling. Oncotarget. 2015;6(27):23845–56.10.18632/oncotarget.4313Search in Google Scholar PubMed PubMed Central
[76] Li X, Ke Q, Li Y, Liu F, Zhu G, Li F. DGCR6L, a novel PAK4 interaction protein, regulates PAK4-mediated migration of human gastric cancer cell via LIMK1. Int J Biochem Cell Biol. 2010;42(1):70–9.10.1016/j.biocel.2009.09.008Search in Google Scholar PubMed
[77] Ahmed T, Shea K, Masters JR, Jones GE, Wells CM. A PAK4-LIMK1 pathway drives prostate cancer cell migration downstream of HGF. Cell Signal. 2008;20(7):1320–8.10.1016/j.cellsig.2008.02.021Search in Google Scholar PubMed
[78] King H, Thillai K, Whale A, Arumugam P, Eldaly H, Kocher HM, et al. PAK4 interacts with p85 alpha: implications for pancreatic cancer cell migration. Sci Rep. 2017;7:42575.10.1038/srep42575Search in Google Scholar PubMed PubMed Central
[79] Wang F, Gao Y, Tang L, Ning K, Geng N, Zhang H, et al. A novel PAK4-CEBPB-CLDN4 axis involving in breast cancer cell migration and invasion. Biochem Biophys Res Commun. 2019;511(2):404–8.10.1016/j.bbrc.2019.02.070Search in Google Scholar PubMed
[80] Xu HT, Lai WL, Liu HF, Wong LL, Ng IO, Ching YP. PAK4 phosphorylates p53 at serine 215 to promote liver cancer metastasis. Cancer Res. 2016;76(19):5732–42.10.1158/0008-5472.CAN-15-3373Search in Google Scholar PubMed
[81] Guo Q, Su N, Zhang J, Li X, Miao Z, Wang G, et al. PAK4 kinase-mediated SCG10 phosphorylation involved in gastric cancer metastasis. Oncogene. 2014;33(25):3277–87.10.1038/onc.2013.296Search in Google Scholar PubMed
[82] Li X, Li J, Li F. P21 activated kinase 4 binds translation elongation factor eEF1A1 to promote gastric cancer cell migration and invasion. Oncol Rep. 2017;37(5):2857–64.10.3892/or.2017.5543Search in Google Scholar PubMed
[83] Kartal-Yandim M, Adan-Gokbulut A, Baran Y. Molecular mechanisms of drug resistance and its reversal in cancer. Crit Rev Biotechnol. 2016;36(4):716–26.10.3109/07388551.2015.1015957Search in Google Scholar PubMed
[84] Shu XR, Wu J, Sun H, Chi LQ, Wang JH. PAK4 confers the malignance of cervical cancers and contributes to the cisplatin-resistance in cervical cancer cells via PI3K/AKT pathway. Diagn Pathol. 2015;10:177.10.1186/s13000-015-0404-zSearch in Google Scholar PubMed PubMed Central
[85] Moon SU, Kim JW, Sung JH, Kang MH, Kim SH, Chang H, et al. p21-Activated kinase 4 (PAK4) as a predictive marker of gemcitabine sensitivity in pancreatic cancer cell lines. Cancer Res Treat. 2015;47(3):501–8.10.4143/crt.2014.054Search in Google Scholar PubMed PubMed Central
[86] Mohammad RM, Li Y, Muqbil I, Aboukameel A, Senapedis W, Baloglu E, et al. Targeting Rho GTPase effector p21 activated kinase 4 (PAK4) suppresses p-Bad-microRNA drug resistance axis leading to inhibition of pancreatic ductal adenocarcinoma proliferation. Small GTPases. 2019;10(5):367–77.10.1080/21541248.2017.1329694Search in Google Scholar PubMed PubMed Central
[87] He H, Dumesny C, Ang CS, Dong L, Ma Y, Zeng J, et al. A novel PAK4 inhibitor suppresses pancreatic cancer growth and enhances the inhibitory effect of gemcitabine. Transl Oncol. 2022;16:101329.10.1016/j.tranon.2021.101329Search in Google Scholar PubMed PubMed Central
[88] Nouri Rouzbahani F, Shirkhoda M, Memari F, Dana H, Mahmoodi Chalbatani G, Mahmoodzadeh H, et al. Immunotherapy a new hope for cancer treatment: a review. Pak J Biol Sci. 2018;21(3):135–50.10.3923/pjbs.2018.135.150Search in Google Scholar PubMed
[89] Li LB, Yang L, Xie GQ, Zhou XC, Shen XB, Xu QL, et al. The modulation relationship of genomic pattern of intratumor heterogeneity and immunity microenvironment heterogeneity in hepatocellular carcinoma. Oncol Lett. 2020;20(5):233.10.3892/ol.2020.12096Search in Google Scholar PubMed PubMed Central
[90] Abril-Rodriguez G, Torrejon DY, Liu W, Zaretsky JM, Nowicki TS, Tsoi J, et al. PAK4 inhibition improves PD-1 blockade immunotherapy. Nat Cancer. 2020;1(1):46–58.10.1038/s43018-019-0003-0Search in Google Scholar PubMed PubMed Central
[91] Porter DL, Hwang WT, Frey NV, Lacey SF, Shaw PA, Loren AW, et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci Transl Med. 2015;7(303):303ra139.10.1126/scitranslmed.aac5415Search in Google Scholar PubMed PubMed Central
[92] Xie YJ, Dougan M, Jailkhani N, Ingram J, Fang T, Kummer L, et al. Nanobody-based CAR T cells that target the tumor microenvironment inhibit the growth of solid tumors in immunocompetent mice. Proc Natl Acad Sci U S A. 2019;116(16):7624–31.10.1073/pnas.1817147116Search in Google Scholar PubMed PubMed Central
[93] Ma W, Wang Y, Zhang R, Yang F, Zhang D, Huang M, et al. Targeting PAK4 to reprogram the vascular microenvironment and improve CAR-T immunotherapy for glioblastoma. Nat Cancer. 2021;2(1):83–97.10.1038/s43018-020-00147-8Search in Google Scholar PubMed
[94] Ling Z, Cheng B, Tao X. Epithelial-to-mesenchymal transition in oral squamous cell carcinoma: Challenges and opportunities. Int J Cancer. 2021;148(7):1548–61.10.1002/ijc.33352Search in Google Scholar PubMed
[95] Wang J, Li R, Li M, Wang C. Fibronectin and colorectal cancer: signaling pathways and clinical implications. J Recept Signal Transduct Res. 2021;41(4):313–20.10.1080/10799893.2020.1817074Search in Google Scholar PubMed
[96] Cai S, Ye Z, Wang X, Pan Y, Weng Y, Lao S, et al. Overexpression of P21-activated kinase 4 is associated with poor prognosis in non-small cell lung cancer and promotes migration and invasion. J Exp Clin Cancer Res. 2015;34(1):48.10.1186/s13046-015-0165-2Search in Google Scholar PubMed PubMed Central
[97] Kobayashi K, Inokuchi M, Takagi Y, Otsuki S, Fujimori Y, Sato Y, et al. Prognostic significance of PAK4 expression in gastric cancer. J Clin Pathol. 2016;69(7):580–5.10.1136/jclinpath-2015-203330Search in Google Scholar PubMed
[98] Song B, Wang W, Zheng Y, Yang J, Xu Z. P21-activated kinase 1 and 4 were associated with colorectal cancer metastasis and infiltration. J Surg Res. 2015;196(1):130–5.10.1016/j.jss.2015.02.035Search in Google Scholar PubMed
[99] Park S, Kim JW, Kim H, Kim JW, Kim YJ, Lee KW, et al. Prognostic value of p21-activated kinase 4 in resected pancreatic cancer. APMIS. 2017;125(8):699–707.10.1111/apm.12705Search in Google Scholar PubMed
[100] Bi Y, Tian M, Le J, Wang L, Liu X, Qu J, et al. Study on the expression of PAK4 and P54 protein in breast cancer. World J Surg Oncol. 2016;14(1):160.10.1186/s12957-016-0913-6Search in Google Scholar PubMed PubMed Central
[101] Siu MK, Kong DS, Ngai SY, Chan HY, Jiang L, Wong ES, et al. p21-activated kinases 1, 2 and 4 in endometrial cancers: effects on clinical outcomes and cell proliferation. PLoS One. 2015;10(7):e0133467.10.1371/journal.pone.0133467Search in Google Scholar PubMed PubMed Central
[102] Draghi A, Chamberlain CA, Furness A, Donia M. Acquired resistance to cancer immunotherapy. Semin Immunopathol. 2019;41(1):31–40.10.1007/s00281-018-0692-ySearch in Google Scholar PubMed
[103] Khan HY, Uddin MH, Balasubramanian SK, Sulaiman N, Iqbal M, Chaker M, et al. PAK4 and NAMPT as novel therapeutic targets in diffuse large B-cell lymphoma, follicular lymphoma, and mantle cell lymphoma. Cancers (Basel). 2021;14(1):160.10.3390/cancers14010160Search in Google Scholar PubMed PubMed Central
[104] Lai LC, Cheong SK, Goh KL, Leong CF, Loh CS, Lopez JB, et al. Clinical practice guidelines committee on tumour markers of the academy of medicine of Malaysia. Clinical usefulness of tumour markers. Malays J Pathol. 2003;25(2):83–105.Search in Google Scholar
[105] Bendardaf R, Lamlum H, Pyrhönen S. Prognostic and predictive molecular markers in colorectal carcinoma. Anticancer Res. 2004;24(4):2519–30.Search in Google Scholar
[106] Amiri FS. Serum tumor markers in chronic kidney disease: as clinical tool in diagnosis, treatment and prognosis of cancers. Ren Fail. 2016;38(4):530–44.10.3109/0886022X.2016.1148523Search in Google Scholar PubMed
© 2022 Xinbo Yu et al., published by De Gruyter
This work is licensed under the Creative Commons Attribution 4.0 International License.
Articles in the same Issue
- Biomedical Sciences
- Effects of direct oral anticoagulants dabigatran and rivaroxaban on the blood coagulation function in rabbits
- The mother of all battles: Viruses vs humans. Can humans avoid extinction in 50–100 years?
- Knockdown of G1P3 inhibits cell proliferation and enhances the cytotoxicity of dexamethasone in acute lymphoblastic leukemia
- LINC00665 regulates hepatocellular carcinoma by modulating mRNA via the m6A enzyme
- Association study of CLDN14 variations in patients with kidney stones
- Concanavalin A-induced autoimmune hepatitis model in mice: Mechanisms and future outlook
- Regulation of miR-30b in cancer development, apoptosis, and drug resistance
- Informatic analysis of the pulmonary microecology in non-cystic fibrosis bronchiectasis at three different stages
- Swimming attenuates tumor growth in CT-26 tumor-bearing mice and suppresses angiogenesis by mediating the HIF-1α/VEGFA pathway
- Characterization of intestinal microbiota and serum metabolites in patients with mild hepatic encephalopathy
- Functional conservation and divergence in plant-specific GRF gene family revealed by sequences and expression analysis
- Application of the FLP/LoxP-FRT recombination system to switch the eGFP expression in a model prokaryote
- Biomedical evaluation of antioxidant properties of lamb meat enriched with iodine and selenium
- Intravenous infusion of the exosomes derived from human umbilical cord mesenchymal stem cells enhance neurological recovery after traumatic brain injury via suppressing the NF-κB pathway
- Effect of dietary pattern on pregnant women with gestational diabetes mellitus and its clinical significance
- Potential regulatory mechanism of TNF-α/TNFR1/ANXA1 in glioma cells and its role in glioma cell proliferation
- Effect of the genetic mutant G71R in uridine diphosphate-glucuronosyltransferase 1A1 on the conjugation of bilirubin
- Quercetin inhibits cytotoxicity of PC12 cells induced by amyloid-beta 25–35 via stimulating estrogen receptor α, activating ERK1/2, and inhibiting apoptosis
- Nutrition intervention in the management of novel coronavirus pneumonia patients
- circ-CFH promotes the development of HCC by regulating cell proliferation, apoptosis, migration, invasion, and glycolysis through the miR-377-3p/RNF38 axis
- Bmi-1 directly upregulates glucose transporter 1 in human gastric adenocarcinoma
- Lacunar infarction aggravates the cognitive deficit in the elderly with white matter lesion
- Hydroxysafflor yellow A improved retinopathy via Nrf2/HO-1 pathway in rats
- Comparison of axon extension: PTFE versus PLA formed by a 3D printer
- Elevated IL-35 level and iTr35 subset increase the bacterial burden and lung lesions in Mycobacterium tuberculosis-infected mice
- A case report of CAT gene and HNF1β gene variations in a patient with early-onset diabetes
- Study on the mechanism of inhibiting patulin production by fengycin
- SOX4 promotes high-glucose-induced inflammation and angiogenesis of retinal endothelial cells by activating NF-κB signaling pathway
- Relationship between blood clots and COVID-19 vaccines: A literature review
- Analysis of genetic characteristics of 436 children with dysplasia and detailed analysis of rare karyotype
- Bioinformatics network analyses of growth differentiation factor 11
- NR4A1 inhibits the epithelial–mesenchymal transition of hepatic stellate cells: Involvement of TGF-β–Smad2/3/4–ZEB signaling
- Expression of Zeb1 in the differentiation of mouse embryonic stem cell
- Study on the genetic damage caused by cadmium sulfide quantum dots in human lymphocytes
- Association between single-nucleotide polymorphisms of NKX2.5 and congenital heart disease in Chinese population: A meta-analysis
- Assessment of the anesthetic effect of modified pentothal sodium solution on Sprague-Dawley rats
- Genetic susceptibility to high myopia in Han Chinese population
- Potential biomarkers and molecular mechanisms in preeclampsia progression
- Silencing circular RNA-friend leukemia virus integration 1 restrained malignancy of CC cells and oxaliplatin resistance by disturbing dyskeratosis congenita 1
- Endostar plus pembrolizumab combined with a platinum-based dual chemotherapy regime for advanced pulmonary large-cell neuroendocrine carcinoma as a first-line treatment: A case report
- The significance of PAK4 in signaling and clinicopathology: A review
- Sorafenib inhibits ovarian cancer cell proliferation and mobility and induces radiosensitivity by targeting the tumor cell epithelial–mesenchymal transition
- Characterization of rabbit polyclonal antibody against camel recombinant nanobodies
- Active legumain promotes invasion and migration of neuroblastoma by regulating epithelial-mesenchymal transition
- Effect of cell receptors in the pathogenesis of osteoarthritis: Current insights
- MT-12 inhibits the proliferation of bladder cells in vitro and in vivo by enhancing autophagy through mitochondrial dysfunction
- Study of hsa_circRNA_000121 and hsa_circRNA_004183 in papillary thyroid microcarcinoma
- BuyangHuanwu Decoction attenuates cerebral vasospasm caused by subarachnoid hemorrhage in rats via PI3K/AKT/eNOS axis
- Effects of the interaction of Notch and TLR4 pathways on inflammation and heart function in septic heart
- Monosodium iodoacetate-induced subchondral bone microstructure and inflammatory changes in an animal model of osteoarthritis
- A rare presentation of type II Abernethy malformation and nephrotic syndrome: Case report and review
- Rapid death due to pulmonary epithelioid haemangioendothelioma in several weeks: A case report
- Hepatoprotective role of peroxisome proliferator-activated receptor-α in non-cancerous hepatic tissues following transcatheter arterial embolization
- Correlation between peripheral blood lymphocyte subpopulations and primary systemic lupus erythematosus
- A novel SLC8A1-ALK fusion in lung adenocarcinoma confers sensitivity to alectinib: A case report
- β-Hydroxybutyrate upregulates FGF21 expression through inhibition of histone deacetylases in hepatocytes
- Identification of metabolic genes for the prediction of prognosis and tumor microenvironment infiltration in early-stage non-small cell lung cancer
- BTBD10 inhibits glioma tumorigenesis by downregulating cyclin D1 and p-Akt
- Mucormycosis co-infection in COVID-19 patients: An update
- Metagenomic next-generation sequencing in diagnosing Pneumocystis jirovecii pneumonia: A case report
- Long non-coding RNA HOXB-AS1 is a prognostic marker and promotes hepatocellular carcinoma cells’ proliferation and invasion
- Preparation and evaluation of LA-PEG-SPION, a targeted MRI contrast agent for liver cancer
- Proteomic analysis of the liver regulating lipid metabolism in Chaohu ducks using two-dimensional electrophoresis
- Nasopharyngeal tuberculosis: A case report
- Characterization and evaluation of anti-Salmonella enteritidis activity of indigenous probiotic lactobacilli in mice
- Aberrant pulmonary immune response of obese mice to periodontal infection
- Bacteriospermia – A formidable player in male subfertility
- In silico and in vivo analysis of TIPE1 expression in diffuse large B cell lymphoma
- Effects of KCa channels on biological behavior of trophoblasts
- Interleukin-17A influences the vulnerability rather than the size of established atherosclerotic plaques in apolipoprotein E-deficient mice
- Multiple organ failure and death caused by Staphylococcus aureus hip infection: A case report
- Prognostic signature related to the immune environment of oral squamous cell carcinoma
- Primary and metastatic squamous cell carcinoma of the thyroid gland: Two case reports
- Neuroprotective effects of crocin and crocin-loaded niosomes against the paraquat-induced oxidative brain damage in rats
- Role of MMP-2 and CD147 in kidney fibrosis
- Geometric basis of action potential of skeletal muscle cells and neurons
- Babesia microti-induced fulminant sepsis in an immunocompromised host: A case report and the case-specific literature review
- Role of cerebellar cortex in associative learning and memory in guinea pigs
- Application of metagenomic next-generation sequencing technique for diagnosing a specific case of necrotizing meningoencephalitis caused by human herpesvirus 2
- Case report: Quadruple primary malignant neoplasms including esophageal, ureteral, and lung in an elderly male
- Long non-coding RNA NEAT1 promotes angiogenesis in hepatoma carcinoma via the miR-125a-5p/VEGF pathway
- Osteogenic differentiation of periodontal membrane stem cells in inflammatory environments
- Knockdown of SHMT2 enhances the sensitivity of gastric cancer cells to radiotherapy through the Wnt/β-catenin pathway
- Continuous renal replacement therapy combined with double filtration plasmapheresis in the treatment of severe lupus complicated by serious bacterial infections in children: A case report
- Simultaneous triple primary malignancies, including bladder cancer, lymphoma, and lung cancer, in an elderly male: A case report
- Preclinical immunogenicity assessment of a cell-based inactivated whole-virion H5N1 influenza vaccine
- One case of iodine-125 therapy – A new minimally invasive treatment of intrahepatic cholangiocarcinoma
- S1P promotes corneal trigeminal neuron differentiation and corneal nerve repair via upregulating nerve growth factor expression in a mouse model
- Early cancer detection by a targeted methylation assay of circulating tumor DNA in plasma
- Calcifying nanoparticles initiate the calcification process of mesenchymal stem cells in vitro through the activation of the TGF-β1/Smad signaling pathway and promote the decay of echinococcosis
- Evaluation of prognostic markers in patients infected with SARS-CoV-2
- N6-Methyladenosine-related alternative splicing events play a role in bladder cancer
- Characterization of the structural, oxidative, and immunological features of testis tissue from Zucker diabetic fatty rats
- Effects of glucose and osmotic pressure on the proliferation and cell cycle of human chorionic trophoblast cells
- Investigation of genotype diversity of 7,804 norovirus sequences in humans and animals of China
- Characteristics and karyotype analysis of a patient with turner syndrome complicated with multiple-site tumors: A case report
- Aggravated renal fibrosis is positively associated with the activation of HMGB1-TLR2/4 signaling in STZ-induced diabetic mice
- Distribution characteristics of SARS-CoV-2 IgM/IgG in false-positive results detected by chemiluminescent immunoassay
- SRPX2 attenuated oxygen–glucose deprivation and reperfusion-induced injury in cardiomyocytes via alleviating endoplasmic reticulum stress-induced apoptosis through targeting PI3K/Akt/mTOR axis
- Aquaporin-8 overexpression is involved in vascular structure and function changes in placentas of gestational diabetes mellitus patients
- Relationship between CRP gene polymorphisms and ischemic stroke risk: A systematic review and meta-analysis
- Effects of growth hormone on lipid metabolism and sexual development in pubertal obese male rats
- Cloning and identification of the CTLA-4IgV gene and functional application of vaccine in Xinjiang sheep
- Antitumor activity of RUNX3: Upregulation of E-cadherin and downregulation of the epithelial–mesenchymal transition in clear-cell renal cell carcinoma
- PHF8 promotes osteogenic differentiation of BMSCs in old rat with osteoporosis by regulating Wnt/β-catenin pathway
- A review of the current state of the computer-aided diagnosis (CAD) systems for breast cancer diagnosis
- Bilateral dacryoadenitis in adult-onset Still’s disease: A case report
- A novel association between Bmi-1 protein expression and the SUVmax obtained by 18F-FDG PET/CT in patients with gastric adenocarcinoma
- The role of erythrocytes and erythroid progenitor cells in tumors
- Relationship between platelet activation markers and spontaneous abortion: A meta-analysis
- Abnormal methylation caused by folic acid deficiency in neural tube defects
- Silencing TLR4 using an ultrasound-targeted microbubble destruction-based shRNA system reduces ischemia-induced seizures in hyperglycemic rats
- Plant Sciences
- Seasonal succession of bacterial communities in cultured Caulerpa lentillifera detected by high-throughput sequencing
- Cloning and prokaryotic expression of WRKY48 from Caragana intermedia
- Novel Brassica hybrids with different resistance to Leptosphaeria maculans reveal unbalanced rDNA signal patterns
- Application of exogenous auxin and gibberellin regulates the bolting of lettuce (Lactuca sativa L.)
- Phytoremediation of pollutants from wastewater: A concise review
- Genome-wide identification and characterization of NBS-encoding genes in the sweet potato wild ancestor Ipomoea trifida (H.B.K.)
- Alleviative effects of magnetic Fe3O4 nanoparticles on the physiological toxicity of 3-nitrophenol to rice (Oryza sativa L.) seedlings
- Selection and functional identification of Dof genes expressed in response to nitrogen in Populus simonii × Populus nigra
- Study on pecan seed germination influenced by seed endocarp
- Identification of active compounds in Ophiopogonis Radix from different geographical origins by UPLC-Q/TOF-MS combined with GC-MS approaches
- The entire chloroplast genome sequence of Asparagus cochinchinensis and genetic comparison to Asparagus species
- Genome-wide identification of MAPK family genes and their response to abiotic stresses in tea plant (Camellia sinensis)
- Selection and validation of reference genes for RT-qPCR analysis of different organs at various development stages in Caragana intermedia
- Cloning and expression analysis of SERK1 gene in Diospyros lotus
- Integrated metabolomic and transcriptomic profiling revealed coping mechanisms of the edible and medicinal homologous plant Plantago asiatica L. cadmium resistance
- A missense variant in NCF1 is associated with susceptibility to unexplained recurrent spontaneous abortion
- Assessment of drought tolerance indices in faba bean genotypes under different irrigation regimes
- The entire chloroplast genome sequence of Asparagus setaceus (Kunth) Jessop: Genome structure, gene composition, and phylogenetic analysis in Asparagaceae
- Food Science
- Dietary food additive monosodium glutamate with or without high-lipid diet induces spleen anomaly: A mechanistic approach on rat model
- Binge eating disorder during COVID-19
- Potential of honey against the onset of autoimmune diabetes and its associated nephropathy, pancreatitis, and retinopathy in type 1 diabetic animal model
- FTO gene expression in diet-induced obesity is downregulated by Solanum fruit supplementation
- Physical activity enhances fecal lactobacilli in rats chronically drinking sweetened cola beverage
- Supercritical CO2 extraction, chemical composition, and antioxidant effects of Coreopsis tinctoria Nutt. oleoresin
- Functional constituents of plant-based foods boost immunity against acute and chronic disorders
- Effect of selenium and methods of protein extraction on the proteomic profile of Saccharomyces yeast
- Microbial diversity of milk ghee in southern Gansu and its effect on the formation of ghee flavor compounds
- Ecology and Environmental Sciences
- Effects of heavy metals on bacterial community surrounding Bijiashan mining area located in northwest China
- Microorganism community composition analysis coupling with 15N tracer experiments reveals the nitrification rate and N2O emissions in low pH soils in Southern China
- Genetic diversity and population structure of Cinnamomum balansae Lecomte inferred by microsatellites
- Preliminary screening of microplastic contamination in different marine fish species of Taif market, Saudi Arabia
- Plant volatile organic compounds attractive to Lygus pratensis
- Effects of organic materials on soil bacterial community structure in long-term continuous cropping of tomato in greenhouse
- Effects of soil treated fungicide fluopimomide on tomato (Solanum lycopersicum L.) disease control and plant growth
- Prevalence of Yersinia pestis among rodents captured in a semi-arid tropical ecosystem of south-western Zimbabwe
- Effects of irrigation and nitrogen fertilization on mitigating salt-induced Na+ toxicity and sustaining sea rice growth
- Bioengineering and Biotechnology
- Poly-l-lysine-caused cell adhesion induces pyroptosis in THP-1 monocytes
- Development of alkaline phosphatase-scFv and its use for one-step enzyme-linked immunosorbent assay for His-tagged protein detection
- Development and validation of a predictive model for immune-related genes in patients with tongue squamous cell carcinoma
- Agriculture
- Effects of chemical-based fertilizer replacement with biochar-based fertilizer on albic soil nutrient content and maize yield
- Genome-wide identification and expression analysis of CPP-like gene family in Triticum aestivum L. under different hormone and stress conditions
- Agronomic and economic performance of mung bean (Vigna radiata L.) varieties in response to rates of blended NPS fertilizer in Kindo Koysha district, Southern Ethiopia
- Influence of furrow irrigation regime on the yield and water consumption indicators of winter wheat based on a multi-level fuzzy comprehensive evaluation
- Discovery of exercise-related genes and pathway analysis based on comparative genomes of Mongolian originated Abaga and Wushen horse
- Lessons from integrated seasonal forecast-crop modelling in Africa: A systematic review
- Evolution trend of soil fertility in tobacco-planting area of Chenzhou, Hunan Province, China
- Animal Sciences
- Morphological and molecular characterization of Tatera indica Hardwicke 1807 (Rodentia: Muridae) from Pothwar, Pakistan
- Research on meat quality of Qianhua Mutton Merino sheep and Small-tail Han sheep
- SI: A Scientific Memoir
- Suggestions on leading an academic research laboratory group
- My scientific genealogy and the Toronto ACDC Laboratory, 1988–2022
- Erratum
- Erratum to “Changes of immune cells in patients with hepatocellular carcinoma treated by radiofrequency ablation and hepatectomy, a pilot study”
- Erratum to “A two-microRNA signature predicts the progression of male thyroid cancer”
- Retraction
- Retraction of “Lidocaine has antitumor effect on hepatocellular carcinoma via the circ_DYNC1H1/miR-520a-3p/USP14 axis”
Articles in the same Issue
- Biomedical Sciences
- Effects of direct oral anticoagulants dabigatran and rivaroxaban on the blood coagulation function in rabbits
- The mother of all battles: Viruses vs humans. Can humans avoid extinction in 50–100 years?
- Knockdown of G1P3 inhibits cell proliferation and enhances the cytotoxicity of dexamethasone in acute lymphoblastic leukemia
- LINC00665 regulates hepatocellular carcinoma by modulating mRNA via the m6A enzyme
- Association study of CLDN14 variations in patients with kidney stones
- Concanavalin A-induced autoimmune hepatitis model in mice: Mechanisms and future outlook
- Regulation of miR-30b in cancer development, apoptosis, and drug resistance
- Informatic analysis of the pulmonary microecology in non-cystic fibrosis bronchiectasis at three different stages
- Swimming attenuates tumor growth in CT-26 tumor-bearing mice and suppresses angiogenesis by mediating the HIF-1α/VEGFA pathway
- Characterization of intestinal microbiota and serum metabolites in patients with mild hepatic encephalopathy
- Functional conservation and divergence in plant-specific GRF gene family revealed by sequences and expression analysis
- Application of the FLP/LoxP-FRT recombination system to switch the eGFP expression in a model prokaryote
- Biomedical evaluation of antioxidant properties of lamb meat enriched with iodine and selenium
- Intravenous infusion of the exosomes derived from human umbilical cord mesenchymal stem cells enhance neurological recovery after traumatic brain injury via suppressing the NF-κB pathway
- Effect of dietary pattern on pregnant women with gestational diabetes mellitus and its clinical significance
- Potential regulatory mechanism of TNF-α/TNFR1/ANXA1 in glioma cells and its role in glioma cell proliferation
- Effect of the genetic mutant G71R in uridine diphosphate-glucuronosyltransferase 1A1 on the conjugation of bilirubin
- Quercetin inhibits cytotoxicity of PC12 cells induced by amyloid-beta 25–35 via stimulating estrogen receptor α, activating ERK1/2, and inhibiting apoptosis
- Nutrition intervention in the management of novel coronavirus pneumonia patients
- circ-CFH promotes the development of HCC by regulating cell proliferation, apoptosis, migration, invasion, and glycolysis through the miR-377-3p/RNF38 axis
- Bmi-1 directly upregulates glucose transporter 1 in human gastric adenocarcinoma
- Lacunar infarction aggravates the cognitive deficit in the elderly with white matter lesion
- Hydroxysafflor yellow A improved retinopathy via Nrf2/HO-1 pathway in rats
- Comparison of axon extension: PTFE versus PLA formed by a 3D printer
- Elevated IL-35 level and iTr35 subset increase the bacterial burden and lung lesions in Mycobacterium tuberculosis-infected mice
- A case report of CAT gene and HNF1β gene variations in a patient with early-onset diabetes
- Study on the mechanism of inhibiting patulin production by fengycin
- SOX4 promotes high-glucose-induced inflammation and angiogenesis of retinal endothelial cells by activating NF-κB signaling pathway
- Relationship between blood clots and COVID-19 vaccines: A literature review
- Analysis of genetic characteristics of 436 children with dysplasia and detailed analysis of rare karyotype
- Bioinformatics network analyses of growth differentiation factor 11
- NR4A1 inhibits the epithelial–mesenchymal transition of hepatic stellate cells: Involvement of TGF-β–Smad2/3/4–ZEB signaling
- Expression of Zeb1 in the differentiation of mouse embryonic stem cell
- Study on the genetic damage caused by cadmium sulfide quantum dots in human lymphocytes
- Association between single-nucleotide polymorphisms of NKX2.5 and congenital heart disease in Chinese population: A meta-analysis
- Assessment of the anesthetic effect of modified pentothal sodium solution on Sprague-Dawley rats
- Genetic susceptibility to high myopia in Han Chinese population
- Potential biomarkers and molecular mechanisms in preeclampsia progression
- Silencing circular RNA-friend leukemia virus integration 1 restrained malignancy of CC cells and oxaliplatin resistance by disturbing dyskeratosis congenita 1
- Endostar plus pembrolizumab combined with a platinum-based dual chemotherapy regime for advanced pulmonary large-cell neuroendocrine carcinoma as a first-line treatment: A case report
- The significance of PAK4 in signaling and clinicopathology: A review
- Sorafenib inhibits ovarian cancer cell proliferation and mobility and induces radiosensitivity by targeting the tumor cell epithelial–mesenchymal transition
- Characterization of rabbit polyclonal antibody against camel recombinant nanobodies
- Active legumain promotes invasion and migration of neuroblastoma by regulating epithelial-mesenchymal transition
- Effect of cell receptors in the pathogenesis of osteoarthritis: Current insights
- MT-12 inhibits the proliferation of bladder cells in vitro and in vivo by enhancing autophagy through mitochondrial dysfunction
- Study of hsa_circRNA_000121 and hsa_circRNA_004183 in papillary thyroid microcarcinoma
- BuyangHuanwu Decoction attenuates cerebral vasospasm caused by subarachnoid hemorrhage in rats via PI3K/AKT/eNOS axis
- Effects of the interaction of Notch and TLR4 pathways on inflammation and heart function in septic heart
- Monosodium iodoacetate-induced subchondral bone microstructure and inflammatory changes in an animal model of osteoarthritis
- A rare presentation of type II Abernethy malformation and nephrotic syndrome: Case report and review
- Rapid death due to pulmonary epithelioid haemangioendothelioma in several weeks: A case report
- Hepatoprotective role of peroxisome proliferator-activated receptor-α in non-cancerous hepatic tissues following transcatheter arterial embolization
- Correlation between peripheral blood lymphocyte subpopulations and primary systemic lupus erythematosus
- A novel SLC8A1-ALK fusion in lung adenocarcinoma confers sensitivity to alectinib: A case report
- β-Hydroxybutyrate upregulates FGF21 expression through inhibition of histone deacetylases in hepatocytes
- Identification of metabolic genes for the prediction of prognosis and tumor microenvironment infiltration in early-stage non-small cell lung cancer
- BTBD10 inhibits glioma tumorigenesis by downregulating cyclin D1 and p-Akt
- Mucormycosis co-infection in COVID-19 patients: An update
- Metagenomic next-generation sequencing in diagnosing Pneumocystis jirovecii pneumonia: A case report
- Long non-coding RNA HOXB-AS1 is a prognostic marker and promotes hepatocellular carcinoma cells’ proliferation and invasion
- Preparation and evaluation of LA-PEG-SPION, a targeted MRI contrast agent for liver cancer
- Proteomic analysis of the liver regulating lipid metabolism in Chaohu ducks using two-dimensional electrophoresis
- Nasopharyngeal tuberculosis: A case report
- Characterization and evaluation of anti-Salmonella enteritidis activity of indigenous probiotic lactobacilli in mice
- Aberrant pulmonary immune response of obese mice to periodontal infection
- Bacteriospermia – A formidable player in male subfertility
- In silico and in vivo analysis of TIPE1 expression in diffuse large B cell lymphoma
- Effects of KCa channels on biological behavior of trophoblasts
- Interleukin-17A influences the vulnerability rather than the size of established atherosclerotic plaques in apolipoprotein E-deficient mice
- Multiple organ failure and death caused by Staphylococcus aureus hip infection: A case report
- Prognostic signature related to the immune environment of oral squamous cell carcinoma
- Primary and metastatic squamous cell carcinoma of the thyroid gland: Two case reports
- Neuroprotective effects of crocin and crocin-loaded niosomes against the paraquat-induced oxidative brain damage in rats
- Role of MMP-2 and CD147 in kidney fibrosis
- Geometric basis of action potential of skeletal muscle cells and neurons
- Babesia microti-induced fulminant sepsis in an immunocompromised host: A case report and the case-specific literature review
- Role of cerebellar cortex in associative learning and memory in guinea pigs
- Application of metagenomic next-generation sequencing technique for diagnosing a specific case of necrotizing meningoencephalitis caused by human herpesvirus 2
- Case report: Quadruple primary malignant neoplasms including esophageal, ureteral, and lung in an elderly male
- Long non-coding RNA NEAT1 promotes angiogenesis in hepatoma carcinoma via the miR-125a-5p/VEGF pathway
- Osteogenic differentiation of periodontal membrane stem cells in inflammatory environments
- Knockdown of SHMT2 enhances the sensitivity of gastric cancer cells to radiotherapy through the Wnt/β-catenin pathway
- Continuous renal replacement therapy combined with double filtration plasmapheresis in the treatment of severe lupus complicated by serious bacterial infections in children: A case report
- Simultaneous triple primary malignancies, including bladder cancer, lymphoma, and lung cancer, in an elderly male: A case report
- Preclinical immunogenicity assessment of a cell-based inactivated whole-virion H5N1 influenza vaccine
- One case of iodine-125 therapy – A new minimally invasive treatment of intrahepatic cholangiocarcinoma
- S1P promotes corneal trigeminal neuron differentiation and corneal nerve repair via upregulating nerve growth factor expression in a mouse model
- Early cancer detection by a targeted methylation assay of circulating tumor DNA in plasma
- Calcifying nanoparticles initiate the calcification process of mesenchymal stem cells in vitro through the activation of the TGF-β1/Smad signaling pathway and promote the decay of echinococcosis
- Evaluation of prognostic markers in patients infected with SARS-CoV-2
- N6-Methyladenosine-related alternative splicing events play a role in bladder cancer
- Characterization of the structural, oxidative, and immunological features of testis tissue from Zucker diabetic fatty rats
- Effects of glucose and osmotic pressure on the proliferation and cell cycle of human chorionic trophoblast cells
- Investigation of genotype diversity of 7,804 norovirus sequences in humans and animals of China
- Characteristics and karyotype analysis of a patient with turner syndrome complicated with multiple-site tumors: A case report
- Aggravated renal fibrosis is positively associated with the activation of HMGB1-TLR2/4 signaling in STZ-induced diabetic mice
- Distribution characteristics of SARS-CoV-2 IgM/IgG in false-positive results detected by chemiluminescent immunoassay
- SRPX2 attenuated oxygen–glucose deprivation and reperfusion-induced injury in cardiomyocytes via alleviating endoplasmic reticulum stress-induced apoptosis through targeting PI3K/Akt/mTOR axis
- Aquaporin-8 overexpression is involved in vascular structure and function changes in placentas of gestational diabetes mellitus patients
- Relationship between CRP gene polymorphisms and ischemic stroke risk: A systematic review and meta-analysis
- Effects of growth hormone on lipid metabolism and sexual development in pubertal obese male rats
- Cloning and identification of the CTLA-4IgV gene and functional application of vaccine in Xinjiang sheep
- Antitumor activity of RUNX3: Upregulation of E-cadherin and downregulation of the epithelial–mesenchymal transition in clear-cell renal cell carcinoma
- PHF8 promotes osteogenic differentiation of BMSCs in old rat with osteoporosis by regulating Wnt/β-catenin pathway
- A review of the current state of the computer-aided diagnosis (CAD) systems for breast cancer diagnosis
- Bilateral dacryoadenitis in adult-onset Still’s disease: A case report
- A novel association between Bmi-1 protein expression and the SUVmax obtained by 18F-FDG PET/CT in patients with gastric adenocarcinoma
- The role of erythrocytes and erythroid progenitor cells in tumors
- Relationship between platelet activation markers and spontaneous abortion: A meta-analysis
- Abnormal methylation caused by folic acid deficiency in neural tube defects
- Silencing TLR4 using an ultrasound-targeted microbubble destruction-based shRNA system reduces ischemia-induced seizures in hyperglycemic rats
- Plant Sciences
- Seasonal succession of bacterial communities in cultured Caulerpa lentillifera detected by high-throughput sequencing
- Cloning and prokaryotic expression of WRKY48 from Caragana intermedia
- Novel Brassica hybrids with different resistance to Leptosphaeria maculans reveal unbalanced rDNA signal patterns
- Application of exogenous auxin and gibberellin regulates the bolting of lettuce (Lactuca sativa L.)
- Phytoremediation of pollutants from wastewater: A concise review
- Genome-wide identification and characterization of NBS-encoding genes in the sweet potato wild ancestor Ipomoea trifida (H.B.K.)
- Alleviative effects of magnetic Fe3O4 nanoparticles on the physiological toxicity of 3-nitrophenol to rice (Oryza sativa L.) seedlings
- Selection and functional identification of Dof genes expressed in response to nitrogen in Populus simonii × Populus nigra
- Study on pecan seed germination influenced by seed endocarp
- Identification of active compounds in Ophiopogonis Radix from different geographical origins by UPLC-Q/TOF-MS combined with GC-MS approaches
- The entire chloroplast genome sequence of Asparagus cochinchinensis and genetic comparison to Asparagus species
- Genome-wide identification of MAPK family genes and their response to abiotic stresses in tea plant (Camellia sinensis)
- Selection and validation of reference genes for RT-qPCR analysis of different organs at various development stages in Caragana intermedia
- Cloning and expression analysis of SERK1 gene in Diospyros lotus
- Integrated metabolomic and transcriptomic profiling revealed coping mechanisms of the edible and medicinal homologous plant Plantago asiatica L. cadmium resistance
- A missense variant in NCF1 is associated with susceptibility to unexplained recurrent spontaneous abortion
- Assessment of drought tolerance indices in faba bean genotypes under different irrigation regimes
- The entire chloroplast genome sequence of Asparagus setaceus (Kunth) Jessop: Genome structure, gene composition, and phylogenetic analysis in Asparagaceae
- Food Science
- Dietary food additive monosodium glutamate with or without high-lipid diet induces spleen anomaly: A mechanistic approach on rat model
- Binge eating disorder during COVID-19
- Potential of honey against the onset of autoimmune diabetes and its associated nephropathy, pancreatitis, and retinopathy in type 1 diabetic animal model
- FTO gene expression in diet-induced obesity is downregulated by Solanum fruit supplementation
- Physical activity enhances fecal lactobacilli in rats chronically drinking sweetened cola beverage
- Supercritical CO2 extraction, chemical composition, and antioxidant effects of Coreopsis tinctoria Nutt. oleoresin
- Functional constituents of plant-based foods boost immunity against acute and chronic disorders
- Effect of selenium and methods of protein extraction on the proteomic profile of Saccharomyces yeast
- Microbial diversity of milk ghee in southern Gansu and its effect on the formation of ghee flavor compounds
- Ecology and Environmental Sciences
- Effects of heavy metals on bacterial community surrounding Bijiashan mining area located in northwest China
- Microorganism community composition analysis coupling with 15N tracer experiments reveals the nitrification rate and N2O emissions in low pH soils in Southern China
- Genetic diversity and population structure of Cinnamomum balansae Lecomte inferred by microsatellites
- Preliminary screening of microplastic contamination in different marine fish species of Taif market, Saudi Arabia
- Plant volatile organic compounds attractive to Lygus pratensis
- Effects of organic materials on soil bacterial community structure in long-term continuous cropping of tomato in greenhouse
- Effects of soil treated fungicide fluopimomide on tomato (Solanum lycopersicum L.) disease control and plant growth
- Prevalence of Yersinia pestis among rodents captured in a semi-arid tropical ecosystem of south-western Zimbabwe
- Effects of irrigation and nitrogen fertilization on mitigating salt-induced Na+ toxicity and sustaining sea rice growth
- Bioengineering and Biotechnology
- Poly-l-lysine-caused cell adhesion induces pyroptosis in THP-1 monocytes
- Development of alkaline phosphatase-scFv and its use for one-step enzyme-linked immunosorbent assay for His-tagged protein detection
- Development and validation of a predictive model for immune-related genes in patients with tongue squamous cell carcinoma
- Agriculture
- Effects of chemical-based fertilizer replacement with biochar-based fertilizer on albic soil nutrient content and maize yield
- Genome-wide identification and expression analysis of CPP-like gene family in Triticum aestivum L. under different hormone and stress conditions
- Agronomic and economic performance of mung bean (Vigna radiata L.) varieties in response to rates of blended NPS fertilizer in Kindo Koysha district, Southern Ethiopia
- Influence of furrow irrigation regime on the yield and water consumption indicators of winter wheat based on a multi-level fuzzy comprehensive evaluation
- Discovery of exercise-related genes and pathway analysis based on comparative genomes of Mongolian originated Abaga and Wushen horse
- Lessons from integrated seasonal forecast-crop modelling in Africa: A systematic review
- Evolution trend of soil fertility in tobacco-planting area of Chenzhou, Hunan Province, China
- Animal Sciences
- Morphological and molecular characterization of Tatera indica Hardwicke 1807 (Rodentia: Muridae) from Pothwar, Pakistan
- Research on meat quality of Qianhua Mutton Merino sheep and Small-tail Han sheep
- SI: A Scientific Memoir
- Suggestions on leading an academic research laboratory group
- My scientific genealogy and the Toronto ACDC Laboratory, 1988–2022
- Erratum
- Erratum to “Changes of immune cells in patients with hepatocellular carcinoma treated by radiofrequency ablation and hepatectomy, a pilot study”
- Erratum to “A two-microRNA signature predicts the progression of male thyroid cancer”
- Retraction
- Retraction of “Lidocaine has antitumor effect on hepatocellular carcinoma via the circ_DYNC1H1/miR-520a-3p/USP14 axis”