1. Introduction
The migration of cells is vital in various biological phenomena, including embryogenesis, immune response, inflammation, morphogenesis, and wound healing. In particular, cancer cell migration is a critical step in metastasis, which is the main cause of cancer-related deaths. Epithelial–mesenchymal transition (EMT) has been related to the metastasis of cancer cells by promoting motility and invasion [
1]. During the EMT process, the expression of proteins for adhering adjacent cells, including E-cadherin, Occludin, and Claudin, decreased, while N-cadherin, Vimentin, and EMT transcription factors such as Zeb1, Snail 1/2, and Twist are upregulated [
1]. Here, our focus was on exploring the potential role of inflammation in EMT process within cancer cells. Accordingly, a typical inflammatory agent, LPS, was utilized for inducting inflammation in cancer cells.
Lipopolysaccharide (LPS), a distinctive constituent found in the cell wall of Gram-negative bacteria, serves as a well-known activator of the inflammasome. Within intestinal epithelial cells, the activation of the inflammasome is triggered by LPS through the initiation of Toll-like receptor 4 (TLR4)-mediated signal transduction pathways [
2]. Numerous reports found that LPS stimulates inflammatory responses, causing epithelial cells to change into cancerous forms. For example, it has been shown that LPS promotes the invasion and migration of prostate cancer (PCa) cells [
3] and promotes epithelial–mesenchymal transition (EMT) through TLR4 stimulated by LPS [
4].
LPS has been documented to trigger the activation of NADPH oxidase [
5]. NADPH oxidase is comprised of membrane components, such as gp91phox and p22phox, as well as cytosolic components, including p40phox, p47phox, and p67phox. Upon cellular stimulation, cytosolic components, especially p47phox, undergo phosphorylation by several kinases. Subsequently, phosphorylated p47phox at the Ser345 residue is recruited to the membrane components, initiating the activation of NADPH oxidase. Simultaneously, Rac1/2-GTP, a member of the Rho GTPase subfamily, forms a complex with NADPH oxidase, significantly enhancing its activity [
6].
Typically, Rho GTPase is involved in regulating a range of cellular processes, including the modulation of cytoskeletal proteins, cellular morphology, migration, and cell proliferation. The activities of Rho GTPases are regulated by specific factors such as guanine nucleotide exchange factors (GEFs), GTPase-activating proteins (GAPs), and the guanine nucleotide dissociation factor (GDI) [
7]. The cell migration system encompasses extensively researched members of the Rho-GTPase family, including RhoA, Rac1, and Cdc42, which each play significant roles in regulating cell motility. RhoA specifically contributes to focal adhesion formation, actomyosin contraction, stress fiber formation, and retraction of the cell’s tail, thereby facilitating cell migration [
8]. Studies have indicated that LPS can trigger the disruption of tight junctions in brain microvascular endothelial cells (BMECs) through the activation of the RhoA/ROCK signaling pathway [
9].
In mammals, the intracellular enzyme phospholipase D (PLD) responds to external signals by hydrolyzing phosphatidylcholine (PC), the most abundant phospholipid in cell membranes, to generate the lipid second-messenger phosphatidic acid (PA). The resulting PA, produced by PLD, serves as a regulator for a wide range of cellular processes including vesicular trafficking, membrane fission and fusion, as well as cell growth, proliferation, and migration [
10,
11]. Two mammalian PLD, PLD1 and PLD2, have been identified, both of which include splice variants [
12]. In both in vitro and in vivo contexts, the activity of PLD1 is significantly boosted by protein kinase C (PKC) as well as small GTPases such as the ADP-ribosylation factor (Arf) and Rho families [
13]. In particular, the C-terminal domain of PLD1 interacts with the dominant active mutant RhoA G14V [
14], and it was recently reported that the crystal structure of PLD1 provided an activation mechanism by RhoA and PI(4,5)P
2 [
15]. Notably, G protein-coupled receptors (GPCRs) activated nuclear PLD1 in vascular smooth muscle cells in response to lysophosphatidic acid (LPA) [
16]. It is also reported that PLD1, but not PLD2, is upregulated in non-small cell lung cancer tissue [
17]. Our previous study revealed that p-Tyr42 RhoA contributes to amplifying superoxide production and induces the cell migration of A549 lung cancer cell line in response to PMA [
18]. Based on these reports, we investigated the function of p-Tyr42 RhoA in PLD1 activation, and its downstream signaling pathway.
In this study, we observed that LPS promoted the p-Tyr42 RhoA levels, which activated PLD1 through the interaction between p-Tyr42 RhoA and PLD1 in A549 cells. LPS stimulated cell migration through the expression of EMT marker proteins, and particularly the promoter of which ZEB1 were associated with p-Tyr42 RhoA and PLD1 in the nucleus in response to LPS. Moreover, we discovered that MYH9, also known as Myosin IIA and NMIIA, was a PA-binding protein.
2. Materials and Methods
2.1. Materials
The Tat-C3 recombinant fusion protein, composed of Tat-peptide and C3 toxin, was purified from E. coli using a His-Bind resin column (EMD Milliporre, #70239-3, Darmstadt, Germany). Y27632 (SCM075) was obtained from Millipore Sigma (Burlington, Burlington, NJ, USA). LPS (E. coli 055: B5), N-acetyl-L-cysteine (NAC, A7250), apocynin (A10809), Nonidet P-40 (NP-40), bovine serum albumin (BSA), and isopropyl β-D-thiogalactoside (IPTG) were procured from Sigma-Aldrich (St. Louis, MO, USA). Fetal bovine serum (FBS, 12484010), Dulbecco’s modified Eagle’s medium (DMEM, 11965092), and penicillin–streptomycin antibiotics (15140122) were obtained from GibcoBRL (New York, NY, USA). Protease inhibitor cocktail was purchased from ApexBio (Boston, MA, USA). Fetal bovine serum (FBS), penicillin, and streptomycin were purchased from Cambrex (Verviers, Belgium). LB Broth High Salt (MB-L4488) and skim milk powder (MB-S1667) were sourced from MBcell (SeoCho-Gu, Seoul, Republic of Korea). 4′6-diamidino-2-phenylindole (DAPI) was purchased from Invitrogen (Carlsbad, CA, USA). Alexa fluor-568 and Alexa fluor-594 reagents were obtained from Molecular Probes (Eugene, OR, USA). Protein A/G-agarose beads were purchased from Amersham Biosciences (Piscataway, NJ, USA). The JetPRIME DNA/si-RNA transfection reagent was obtained from Polyplus-transfection (Seoul, Korea). si-PLD1 (sc-44000), si-RhoA (sc-29471), and control si-RNA (sc-37007) were procured from Santa Cruz Biotechnology. si-MYH9 (Bioneer, #4627-1) was obtained from Bioneer (Daejeon, Republic of Korea).
The antibodies utilized in this study were obtained from various providers. An antibody against phosphorylated Tyr42 Rho was generated through immunization with a peptide corresponding to p-Tyr42 Rho (epitope peptide T37VFEN (phospho-) Y42VADIE47). This peptide was synthesized from the phospho-Tyr42 precursor obtained from Young-In Frontier, Seoul, Korea. Normal IgG (sc:2025), actin (sc:58673), PLD1 (sc-28314), Lamin B (sc:965962), Tubulin (sc:32293), ROCK I (sc-17794), ROCK II, (sc:398519), RhoA (sc-418), N-cadherin (sc:393933), and Snail (sc:271977) were purchased from Santa Cruz Biotechnology (Dallas, TX, USA). MYH9 (14844-1-AP) was purchased from Proteintech (Rosemont, IL, USA). HA Tag (3724), p-Lamin A/C (2026) were purchased from Cell Signaling Technology. E-cadherin (ABP0083, Abbkine) and ZEB1 (ABP60963) were purchased from Abbkine (Wuhan, China). p-p47phox (#PA5-37806) was procured from Invitrogen (Waltham, MA, USA). Goat anti-rabbit and goat anti-mouse IgG conjugated with HRP were sourced from Enzo Life Sciences (Farmingdale, NY, USA).
2.2. Cell Culture
A549 cells, derived from human lung adenocarcinoma, were obtained from the Korean Cell Line Bank (Seoul, Republic of Korea). Subsequently, these cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 5% heat-inactivated fetal bovine serum (FBS) and antibiotics (100 U/mL of penicillin and 100 μg/mL of streptomycin) in a humidified environment with 5% CO2 at 37 °C.
2.3. Superoxide Measurement
The levels of superoxide were assessed utilizing the fluorescent probe 2′,7′-dichlorofluorescin diacetate (DCFDA) cellular ROS assay kit (ab113851). A549 cells (2 × 105) were stimulated with the specific treatment in serum-free media, followed by fixation using 4% formaldehyde for 15 min at room temperature. To induce fluorescence, the cells were treated with a solution of 5 μM DCFDA in 1 × PBS (500 μL) for 30 min at 37 °C in the dark. Fluorescence images were recorded using a fluorescence microscope (Axiovert 200, Zeiss; Göttingen, Germany) with filters configured for an excitation wavelength of 485 nm and an emission wavelength of 530 nm.
2.4. Transfection of Plasmid DNA and si-RNA
The A549 cells were transfected with plasmid DNA and small interfering RNA (si-RNA) using the JetPRIME reagent (polyplus transfection, Illkirch, France). Plasmid constructs HA-RhoA WT, HA-RhoA Y42E, HA-RhoA-Y42F and si-RNAs targeting RhoA, PLD1, MYH9, and control si-RNA were utilized in this study. In all si-RNA transfection experiments, A549 cells were initially seeded to attain 30–40% confluence in six-well culture dishes and subsequently transfected with si-RNAs (50–100 nM) using the JetPRIME transfection reagent by following the manufacturer’s instructions. When conducting both DNA and si-RNA transfection in the same culture dishes, si-RNA was transfected initially, followed by the transfection of DNA (2 µg plasmid DNA) after 24 h using fresh media containing 5% serum. After 48 h for DNA transfection and 72 h for si-RNA transfection, the cells were harvested, and protein expression levels were evaluated through Western blot analysis.
2.5. Western Blotting
Western blot analysis was conducted on A549 cells. The cells were washed with 1 × PBS and lysed in RIPA buffer containing 20 mM Tris-HCl pH 7.4, 1 mM MgCl2, 1% (v/v) Nonidet-P40, and 125 mM NaCl. The lysates were supplemented with a phosphatase/protease inhibitor cocktail. After centrifugation for 15 min at 13,000× g at 4 °C, the protein concentration in the supernatants was determined using a BCA protein quantification assay kit (Abbkine, GA, USA). Subsequently, the cell lysates were separated using 10–14% SDS-PAGE and transferred to a PVDF membrane. Non-specific binding was mitigated by incubating the membrane with non-fat dried milk and 0.05% Tween-20 in a Tris-buffered saline solution. Specific antibodies were employed to probe the target proteins, followed by washing and incubation with peroxidase-conjugated secondary antibodies. Visualization of the bound antibodies was accomplished using an ECL system.
2.6. Immunoprecipitation (IP)
Immunoprecipitation was carried out in A549 cells. The cells were rinsed with 1 × PBS and lysed in a buffer containing 20 mM Tris pH 7.4, 120 mM NaCl, 1 mM MgCl2, and 1% Nonidet P-40. A protease/phosphatase inhibitor mixture was added to the lysates. The lysates were clarified through centrifugation and subsequently subjected to a preliminary clearance step using protein A/G-agarose beads for 1 h. The cleared supernatant was incubated with a specific antibody (1:1000 dilutions) overnight at 4 °C. Protein A/G-agarose beads (30 μL) were added to the lysate and the mixture was incubated with shaking. After the beads settled, mixtures were washed three times with a lysis buffer. The proteins bound to the beads were released and analyzed using immunoblotting with specific primary antibodies.
2.7. Cell Fractionation
To separate cytosolic and nuclear fractions, NE-PER nuclear and cytoplasmic extraction reagents (CER: Thermo Scientific #78833, Waltham, MA, USA) were employed. Upon completion of the treatment, A549 cells were collected in ice-cold 1 × PBS and centrifuged at 13,000× g for 15 min. A portion of the resulting cell pellet (20 μL) was mixed with ice-cold CER I (200 μL), CER II (11 μL), and protease inhibitors, and then vortexed and centrifuged to obtain the cytoplasmic protein extract (supernatant). The remaining pellet, containing the nuclei, was re-suspended in ice-cold NER, vortexed, and centrifuged to obtain the nuclear extract. Using the appropriate antibodies, the resulting fractions were subjected to immunoblotting analysis. The tubulin protein and lamin B protein were used as markers for the cytosol and nucleus, respectively.
2.8. Monolayer Wound Healing Assay
A549 cells were seeded in 6-well plates at a density of 1.5 × 105 cells per well and incubated for 24 h. Subsequently, the cells underwent various treatments, including plasmid DNA transfection, siRNA transfection, or LPS (5 μg/mL) stimulation for 24 h. To inhibit cell proliferation during migration, serum starvation was implemented for 12 h before creating the “wound” scratch. Once the cell monolayer reached confluence, a 200 µL sterile plastic pipette tip was used to create scratches. The wounds were regularly photographed (0, 24, and 48 h) and the area of cell-free wounds was measured using a microscope (Axiovert 200, Zeiss, Baden-Wurttemberg, Germany). The assay was performed in at least three independent experiments.
2.9. Chromatin Immunoprecipitation (ChIP) and PCR
The ChIP experiment was conducted by following the protocol provided by Abcam (Cambridge, UK). In brief, A549 cells were subjected to LPS (5 μg/mL) exposure for 24 h, followed by the addition of formaldehyde as a crosslinker (final concentration: 0.75%) for 1 h. The crosslinking reaction was halted by treating with 125 mM glycine for 20 min. Subsequent to sonication in the ChIP lysis buffer (RIPA buffer), the fragmented chromatin protein complex was exposed to specific antibodies and subsequently precipitated using protein A/G beads. Afterward, the beads underwent washing, and the bound DNAs were eluted using elution buffer (containing 1% SDS and 100 mM NaHCO3). Subsequent removal of RNA and proteins was achieved by incubation with RNAse and proteinase K. Ultimately, DNA purification was carried out through phenol–chloroform extraction. PCR primers for ZEB1 (located on human chromosome 10, positions 31,318,016–31,319,215: forward primer: 5′-CAAACCTGCCCTTCCCCTCA-3′, reverse primer: 5′-CTCTACGGCCGGAACCTTGT-3′) and Snail1 (located on human chromosome 20, positions 48,598,513–48,599,512: forward primer: 5′-GAACGGGTGCTCTTGGCTAGCTG-3′, reverse primer: 5′-TCGAGCGAAGCGAGGCCTC-3′) were synthesized by Bioneer (Daejeon, Republic of Korea).
2.10. Immunofluorescent Staining
A549 cells were cultured and fixed with 4% paraformaldehyde for 10 min. To facilitate membrane permeabilization, a solution of 0.5% TX-100 detergents and PBS (1 × PBS) was applied for 10 min, followed by rinsing with 1 × PBS. The cells were then incubated overnight at 4 °C with primary antibodies targeting specific antibodies (1:100 dilutions). After incubation, the cells underwent washing with 1 × PBS. To visualize the antibodies, an Alexa Fluor 546 (red color)- or Alexa Fluor 488 (green color)-conjugated secondary antibodies were used at a dilution of 1:50 for 2 h at room temperature while keeping the environment free from light. The nuclei were stained with DAPI (4′,6-diamidino-2-phenylindole), which was added 10 min before washing with 1 × PBS at a concentration of 1 μg/mL. Fluorescence images were captured using a conventional fluorescence microscope (Axiovert 200, Zeiss, Oberkochen, Germany) and a confocal microscope (LSM 780NLO).
2.11. Quantitative Real-Time Polymerase Chain Reaction (qPCR)
Total RNA extraction from A549 cells was carried out using TRIzol reagent (Ambion, CA, USA). The concentration and purity of the isolated RNA were assessed spectrophotometrically at 260 nm and 280 nm with NanoDrop (Thermo Scientific). Reverse transcription of RNA into cDNA was performed using M-MLV reverse transcriptase (NEB-UK, Hitchin, UK) according to the manufacturer’s protocol. Subsequently, the generated cDNA was utilized with the Taq SYBR Rox 2X fast Q-PCR master mix (TQ1210, SMOBIO Technology, Inc., Hinchu, Taiwan) for real-time qPCR (RT-qPCR) quantification employing an Applied Biosystems StepOnePlus PCR system. Each sample was analyzed in triplicate, and the relative quantities of specifically amplified cDNA were determined using the comparative threshold cycle (CT) value method. GAPDH served as an endogenous reference gene. Primers for ZEB1 (forward: 5′-GTGCACAAGAAGAGCCACAAGTA-3′, reverse: 5′-GGTTGGCACTTGGTGGGATTAC-3′) and GAPDH (forward: 5′-AGAAGGCTGGGGCTCATTTG-3′, reverse: 5′-AGGGGCCATCCACAGTCTTC-3′) in measuring mRNA levels were synthesized by Bioneer (Daejeon, Republic of Korea).
2.12. Statistical Analysis
Data analysis and visualization were performed using GraphPad Prism 8 (GraphPad Software, San Diego, CA, USA). All experiments were repeated at least three times. The data are presented as mean ± SE (standard error). Statistical comparisons were performed using Student’s t-test with the GraphPad Prism software, and differences between two groups were considered statistically significant if the p values were below the designated threshold (* p < 0.05, ** p < 0.01, *** p < 0.001).
4. Discussion
This study was initiated with the question of whether p-Tyr42 RhoA plays a regulatory role in PLD1 enzyme activity, given the well-established role of RhoA-GTP in activating PLD1. Recently, researchers elucidated the crystal structure of PLD1 in association with RhoA-GTP. In this study, they discovered that the C-terminal domain of PLD1 binds to the switch I domain of RhoA-GTP [
15]. Notably, the Tyr42 residue of RhoA is situated in switch I, although it is located in the marginal region of switch I (see
Supplementary Figure S2). Both RhoA and p-Tyr42 RhoA were found to interact with PLD1 (
Figure 2C). However, it is worth mentioning that RhoA Y42F, which serves as a dephospho-mimic form, did not enhance PLD1 enzyme activity (
Figure 2D). These findings substantiate our hypothesis that the p-Tyr42 residue of RhoA plays a critical role in the activation of effector proteins, such as PLD1, which leads to the generation of PA.
Numerous proteins have been previously documented to bind to PA [
22,
23,
24,
25]. The PX domain of p47phox features two distinct basic pockets on its membrane-binding surface, each designed for specific phospholipids, including PI(3,4)P
2/PI(3)P and PA/PS (phosphatidylserine). It is noteworthy that PA has been shown to enhance NADPH oxidase activity [
26]. Similar findings were reported by another researcher, indicating that both PLD1 and PA stimulate ROS production, with PA binding and promoting NADPH oxidase activity [
27]. Consequently, we can deduce that p-Tyr42 RhoA/PLD1/PA collectively play a critical role in superoxide generation. Furthermore, our previous research has unveiled a reciprocal relationship between superoxide and Tyr42 phosphorylation in RhoA, specifically involving the pathway: superoxide-Src–p-Tyr42 RhoA [
20]. Additionally, we have found that p-Tyr42 RhoA activates ROCK2, which further phosphorylates and activates p47phox, resulting in the activation of NADPH oxidase and generation of superoxide: p-Tyr42 RhoA–ROCK2–p-p47phox–NADPH oxidase-superoxide [
18]. Based on these findings, we propose that superoxide and p-Tyr42 RhoA mutually regulate each other in a positive feedback loop manner. Notably, we hypothesized the existence of another positive feedback loop pathway, involving the signaling cascade: PA-NADPH oxidase-superoxide-Src-p-Tyr42 RhoA-PLD1-PA (
Figure 4C).
PA serves as a pivotal signaling molecule, contributing to a diverse range of regulatory functions within cellar processes. These functions encompass cell metabolism and growth, cell death, cytoskeletal remodeling, exocytosis, receptor endocytosis, membrane trafficking, and organelle dynamics [
23]. Remarkably, alterations in PA levels have been linked to changes in actin filament dynamics. The previous studies have reported that exogenous PA and PLD can induce stress fibers and increase F-actin levels [
28,
29]. An increase in the PA level is correlated with enhanced actin filament formation, while a decrease is associated with the disassembly of actin filaments [
30]. We further verified that PA beads exhibit a robust binding affinity to actin (
Figure 5A). Moreover, F-actin has been documented to stimulate PLD activity, whereas monomeric G-actin exerts an inhibitory effect on PLD activity [
30].
To the best of our knowledge, this study represents the first observation of the binding between PA and MYH9 (Myosin IIA). As a result, we postulate that PA plays a critical role in stimulating the interaction between F-actin and MYH9 (Myosin IIA). MYH9 (Myosin IIA) is composed of a heavy chain (230 kDa) and two regulatory light chains (20 kDa) that control the myosin activity, along with two essential light chains (17 kDa) that stabilize the heavy-chain structure [
31]. Phosphorylation of the C-terminal region of MYH9 by PKC, casein kinase II (CKII), and transient receptor potential melastatin 7 (TRPM7) plays a pivotal role in its assembly–disassembly process [
31]. Given that we identified the binding of the MYH9 heavy chain, excluding the light chains, to PA-conjugated beads (
Figure 5A), we postulate that the globular N-terminal region of MYH9 may contain the binding site for PA. Incidentally, it is well established that myosin light-chain kinase (MLCK) and ROCK phosphorylate regulatory subunit in MYH9 (Myosin IIA), thereby activating it [
31]. Expanding on these discoveries, we suggest that MYH9 (Myosin IIA) binding to PA is activated simultaneously by p-Tyr42 RhoA/ROCK in both the cytosol and the nucleus. Actually, MYH9 and F-actin were co-localized in response to LPS, suggesting that MYH9 and F-actin interact with each other (
Figure 5G).
If PLD1 produces PA within the nucleus and binds to MYH9 (Myosin IIA), it prompts the question of the function of MYH9 (Myosin IIA) within the nucleus. Remarkably, the phosphorylation of Ser1916 in the α-helical rod and Ser1943 in the non-helical tail in MYH9 has been observed to increase during the TGF-β-induced EMT process [
32]. Notably, PA can stimulate a variety of kinases, including Raf-1, PKCs, PKN, mTOR, mTORC2, Akt, PAK1, p70S6K1, Fer, GRK, LATS1, and KSR1 [
33]. Based on these findings, we suggest that the C-terminal phosphorylation of MYH9 may be implicated in the EMT process. During the EMT process, key EMT transcription factors such as SNAI1 (Snail 1), SLUG (Snail 2), TWIST, and ZEB1/2 bind directly to the promoter region of E-cadherin, inhibiting its transcription. This results in a decrease in E-cadherin levels, which play a crucial role in the formation of adherence junctions, a critical factor for cell–cell interaction. As a consequence, the decrease in E-cadherin prompts cell migration and the initiation of metastasis. Notably, the downregulation of E-cadherin and concurrent upregulation of N-cadherin during EMT have been observed in various cancers. This cadherin ‘switch’ is associated with an enhancement in cell migration and invasion, contributing to the progression of cancer [
34].
In this study, we investigated the localization of both p-Tyr42 RhoA and PLD1 in the nucleus (
Figure 4I). Moreover, we found that p-Tyr42 RhoA and PLD1 possess the ability to bind to the promoter region of ZEB1 (
Figure 4J). Furthermore, through co-immunoprecipitation (
Figure 5J) and PA-conjugated beads, we observed an interaction between p-Tyr42 RhoA, PLD1 and MYH9 (
Figure 5I). Additionally, it is worth noting that ROCK2 is present in the nucleus and has the capability to phosphorylate and activate p300 acetyltransferase enzyme activity. This suggests that ROCK2 may potentially play a role in the regulation of histone acetylation and influence specific gene expression [
35]. Moreover, MYH9 (Myosin IIA) was found to be present in the nucleus (
Figure 5B,C,E) and to regulate the expression of EMT marker proteins (
Figure 5F). In addition to its conventional role as a motor protein, myosin exhibits increasing evidence of involvement in the regulation of the chromatin structure, chromosomal translocation, transcription regulation, and DNA repair [
36]. Nuclear myosin I (NMI) and actin influence the chromatin dynamics and function in the interphase nucleus [
37]. NMI interacts with transcription factors, while actin engages with RNA polymerase. Furthermore, NM1 contributes to the activation of RNA polymerase II transcription [
38]. Additionally, NMI–actin interactions contribute to the transition of the initiation complex into the elongation complex [
39]. Moreover, Myosin VI and Myosin V are implicated in the regulation of gene transcription. Notably, non-muscle Myosin II is present in the nucleus and facilitates the regulation of gene expression [
40,
41]. Building upon these prior findings, our hypothesis suggests that the p-Tyr42 RhoA/PLD1/MYH9 complex may possibly regulate ZEB1 expression in the nucleus (
Figure 4J and
Figure 5I,J). Moreover, we theorize that the function of MYH9 in the cytosol is to enhance its interaction with F-actin through p-Tyr42 RhoA/PLD1/PA, ultimately promoting cell migration.
Nevertheless, several questions remain unanswered. The most significant among them is whether p-Tyr42 RhoA, PLD1, and PA generated by PLD1, in conjunction with MYH9 and actin, which interact with PA, play a role in regulating the cytoskeletal dynamics within the nucleus to influence the chromatin structure and gene expression. This particular aspect has not been investigated in the current paper and should be considered for future research endeavors. Furthermore, it is particular intriguing to identify the specific genes that are regulated by MYH9, p-Tyr42 RhoA, and PLD1 through transcriptome analyses using si-MYH9, si-PLD1, and RhoA Y42F transfection techniques. Overall, we have presented compelling evidence that p-Tyr42 RhoA plays a significant role in the transcriptional regulation of specific genes in conjunction with other transcription factors [
7,
19,
20].