922
views
0
recommends
+1 Recommend
1 collections
    0
    shares

      Acta Materia Medica now indexed by SCOPUS from May 2024. Interested in becoming an AMM published author?

      • Platinum Open Access with no APCs.
      • Fast peer review/Fast publication online after article acceptance.

      Check out the call for papers on our website https://amm-journal.org/index.php/2023/04/26/acta-materia-medica-call-for-papers-2/

      scite_
      0
      0
      0
      0
      Smart Citations
      0
      0
      0
      0
      Citing PublicationsSupportingMentioningContrasting
      View Citations

      See how this article has been cited at scite.ai

      scite shows how a scientific paper has been cited by providing the context of the citation, a classification describing whether it supports, mentions, or contrasts the cited claim, and a label indicating in which section the citation was made.

       
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Polyphyllin II suppresses cell migration, invasion, and metastasis by inducing cytoskeletal rearrangement through the ROCK1/LIMK/CFL1 pathway in bladder cancer cells

      Published
      research-article
      Bookmark

            Abstract

            This study was aimed at determining the antimetastatic effect of polyphyllin II (PPII) in bladder cancer (BC) and the underlying mechanisms in vitro and in vivo. Wound healing, Transwell assays, and phalloidin staining were performed to determine the effects of PPII on BC cell migration, invasion, and cytoskeletal formation. Gene transcription and expression changes were detected via RNA sequencing and western blotting. The subchronic toxicity and antimetastatic effects of PPII were evaluated in Nu/Nu nude mice. PPII inhibited the migration and invasion of BC cells. Bioinformatics analysis indicated that cytoskeletal regulation was a potentially regulated process. PPII restrained cytoskeletal formation, as confirmed by phalloidin staining. Mechanistically, PPII was found to decrease p-LIMK1/2 and p-CFL1 expression through ROCK1, and to inhibit increased p-CFL1 levels and invasion and migration abilities of BC cells induced by constitutively active RHOA. Subchronic toxicity evaluation revealed that 3.0 mg/kg PPII had limited effects on tissue morphology, alkaline phosphatase, alanine aminotransferase, and aspartate aminotransferase. Furthermore, PPII treatment effectively inhibited the formation of pulmonary metastatic nodules, as well as the expression of ROCK1, p-LIMK1, and p-CFL1 in the lungs. Thus, PPII inhibits BC cell invasion, migration, and metastasis through the RHOA-ROCK1-LIMK1/2-CFL1 axis, and is a potential candidate for antimetastatic drug development.

            Main article text

            1. INTRODUCTION

            Bladder cancer (BC) is a common malignant tumor of the urinary system. Approximately 573,000 new cases of BC and 213,000 related deaths occur worldwide every year [1]. Occupational exposure to industrial chemicals (such as amines) and tobacco smoking contribute to BC risk [2]. BC is generally classified into non-muscle-invasive bladder cancer (NMIBC), which accounts for approximately 75% of newly diagnosed cases and is characterized by isolation to the lamina propria and urothelium, and muscle-invasive bladder cancer (MIBC), which accounts for the remaining 25% of cases, and invades muscles and additional tissues [3]. Although NMIBC has better prognosis than MIBC, its long-term management is costly, and approximately 20% of cases recur and progress to MIBC [4]. In contrast, MIBC has a high risk of recurrence and frequently metastasizes to the lungs, bones, and liver [5, 6]. Although surgery, chemotherapy, radiotherapy, and immunotherapy are among the therapeutic methods available for invasive BC, metastatic disease has a high mortality rate. Consequently, antimetastatic drugs are urgently needed.

            Research and development of new drugs largely rely on natural products. Traditional Chinese medicines (TCMs) have a long and widespread history of use in China and regions of Asia. In recent years, TCMs and their active ingredients have been reported to possess antitumor, anti-inflammatory, and analgesic properties [7]. We screened various TCMs classified as heat-clearing and detoxifying herbs, and found that the ethanol extract of Paris polyphylla (chonglou) has outstanding antitumor activity, including inhibition of cell viability, induction of cell cycle arrest, and suppression of tumor growth [8, 9]. P. polyphylla is traditionally used to treat headaches, fevers, burns, wounds, and snake poisoning, and its ingredients are used in more than 70 types of proprietary medicines, such as Jidesheng sheyao tablets, Chonglou jieduding tablets, and Gongxuening capsules [10]. In the Chinese Pharmacopoeia (2020 version), P. polyphylla is identified by the content of polyphyllins (PPs, including PPI, PPII, and PPVII). We further found that PPII, which has excellent antitumor activity, inhibits epithelial-mesenchymal transition (EMT) and the expression of matrix metalloproteinases (MMPs)—proteins that play essential roles in tumor metastasis [8]. However, the detailed mechanism through which PPII mediates antimetastatic functions in BC cells and in vivo conditions remains to be clarified.

            Metastasis is a common cause of cancer-related death [11]. Motility and elongation changes triggered by cytoskeletal rearrangement are key factors in tumor cell migration, invasion, and metastasis [11, 12]. The RHO GTPase family, a prominent branch of the Ras superfamily, participates in multiple tumor processes, from initiation to development, including EMT and metastasis [13, 14]. RHOA, CDC42, and RAC1 are well-characterized RHO GTPases that regulate the assembly of actin and the organization of the cytoskeleton through LIMK and CFL1 phosphorylation in various tumor cell types [13, 15]. Our bioinformatics results indicated that PPII affects cytoskeletal rearrangement in T24 cells; however, the underlying mechanisms through which RHO GTPases-associated signaling contribute to inhibiting PPII-induced BC cell migration, invasion, and metastasis remain poorly understood. Herein, we used a variety of in vitro and in vivo methods to elucidate the roles of ROCK1/LIMK/CFL1 pathway-mediated cytoskeletal rearrangement in PPII-mediated suppression of BC migration, invasion, and metastasis; in addition, we evaluated the toxicity of PPII in mice.

            2. MATERIALS AND METHODS

            2.1 Cell culture

            The T24 and 5637 BC cell lines and the SV-HUC-1 (normal bladder) cell line were kindly provided by Professor Shengtian Zhao (Binzhou Medical University, Yantai, Shandong) and grown in RPMI-1640 medium (CM10041; Macgene Technology) (for T24 and 5637 cells) or F12K medium (CM10025; Macgene Technology) (for SV-HUC-1 cells) with 10% fetal bovine serum (10270-106, Gibco). The cell lines were incubated at 37°C in a humidified atmosphere containing 5% CO2.

            2.2 Cell viability assays

            We assessed the sensitivity of BC cells to PPII (A0387, Chengdu Must Bio-Technology) and determined drug concentrations via CCK-8 assays (E-CK-A362, Elabscience). T24 cells (3 × 103 cells/well) and 5637 cells (6 × 103 cells/well) were seeded in 96-well plates and incubated overnight. The cells were subsequently exposed to cisplatin (0–48 μM) or PPII (0–3.2 μM for T24 cells and 0–8 μM for 5637 and SV-HUC-1 cells) at varying concentrations for 48 h. After treatment, the cells were incubated with CCK-8 reagent at 37°C for 2 h, and the optical density at 450 nm was measured via a spectrophotometer (type 1510, Thermo Fisher Scientific).

            2.3 Wound healing assays

            For wound healing assays, T24 (6 × 105 cells/well) and 5637 (8 × 105 cells/well) cells were seeded in six-well plates to form complete monolayers. Three sets of linear scratch wounds were generated on the cell monolayers with a sterile 200-μl pipette tip. The cells were cultured in RPMI-1640 medium supplemented with 1% FBS. Intercellular distances were measured at 0, 24, and 48 hours. The migration rate was determined as follows: migration distance/primary intercellular distance × 100%.

            2.4 Transwell invasion and migration assays

            Transwell assays were conducted with an 8.0 μm pore size upper chamber (REF353097, BD Biosciences) with or without precoated Matrigel (354234, BD Biosciences), to assess invasion and migration, respectively. T24 (1 × 105 cells per well) or 5637 (2 × 105 cells per well) cells subjected to various treatments were added to the chambers and incubated 48 h. After incubation, the cells were fixed with 4% paraformaldehyde (PFA) solution (P0099, Beyotime) for 10 minutes. Subsequently, the cells were stained for 30 minutes with 0.1% crystal violet solution (G1061, Solarbio) and washed three times with PBS. Finally, a cotton swab was used to carefully remove the cells and Matrigel from the upper chamber. The invasive cells present on the lower surface were then quantified via an inverted microscope (Vert. A1, Zeiss). For each group, three fields were observed and captured at a magnification of ×100.

            2.5 Western blotting and antibodies

            Total protein was extracted from T24 and 5637 cells treated with PPII alone or cotransfected with plasmids (pcDNA3.1 or pcDNA3.1-RHOA L63) with jetPRIME transfection reagent (101000027, Polyplus Transfection). Western blot analysis was conducted as previously described [8]. Primary antibodies to the following were used: p-LIMK1/2 (1:1000, #3841S), LIMK1 (1:1000, #3842S), and p-CFL1 (1:1000, #3313S) from Cell Signaling Technology; and RHOA (1:2000, 10749-1-AP), ROCK1 (1:2000, 21850-1-AP), CFL1 (1:5000, 66057-1-Ig), and GAPDH (1:10000, 60004-1-Ig) from Proteintech. Protein band intensities were quantified in ImageJ software (version 1.53q; National Institutes of Health).

            2.6 RNA sequencing and data analysis

            Total RNA was extracted with an MJzol Animal RNA Extraction Kit according to the manufacturer’s protocol. RNA quality was assessed with a NanoPhotometer® spectrophotometer and Agilent 2100 Bioanalyzer. The RNA-seq libraries were prepared with an Illumina TruSeq RNA Sample Prep Kit and sequenced on the NovaSeq 6000 system. Differential expression analysis was conducted with the DESeq2 package [16], to identify significantly differentially expressed genes (DEGs) on the basis of P < 0.05 and a |log2FoldChange| > 1.25. The TPM values of the DEGs were used for c-means clustering with the Mfuzz package in R to identify dynamic expression patterns [17]. Functional enrichment analysis for Gene Ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways was performed with the clusterProfiler package [18], with a significance threshold of P < 0.05.

            qPCR was used to detect the mRNA levels of the genes whose expression was most altered in the groups treated with 0.4 μM PPII versus 0 μM PPII. RNA extraction (AC0205, SparkJade) and cDNA template preparation (AG11728, Accurate Biology) were performed according to the manufacturer’s instructions. qPCR was performed with a reagent kit (AG11701, Accurate Biology) and the primers described in Table 1 .

            Table 1 |

            Primer sequences.

            GenePrimer (5′ to 3′)Length (bp)
            HNRNPC-FGCCAAAAGTGAACCGAGGAAAAGC24
            HNRNPC-RCCGAGCAATAGGAGGAGGAGGAG23
            RPL17-FAGAACACTCGTGAAACTGCTCAGG24
            RPL17-RACCTGCCAACTCCACCATTGTAAC24
            NAA40-FTGTAAGCGAGTGTCTGGACTGGAG24
            NAA40-RGGCTCGGTCATCTGTCATTTCCTC24
            ZNF619-FTGGTTCAGCATCAGCGAGTTCAC23
            ZNF619-RGTTGGGGAGGCAGAGGAGAGAG22
            2.7 Phalloidin staining assays

            T24 and 5637 cells were treated with a concentration gradient of PPII (0, 0.2, or 0.4 μM) for 48 hours. The cells were then reseeded onto microscope slides in 24-well plates and incubated overnight. The cells were subsequently fixed with 4% PFA (P1110, Solarbio) for 10 minutes, permeabilized with 0.5% Triton X-100 (T8200, Solarbio) for 5 minutes, and incubated with 150 nM TRITC-phalloidin (CA1610, Solarbio) for 1 hour. After nuclei were stained with DAPI (C0065, Solarbio), the cells were washed three times with PBS. Representative images were captured with an Invitrogen EVOS M7000 Imaging System, and ImageJ software was used to assess the average F-actin intensity.

            2.8 Immunohistochemistry

            Lung samples were fixed overnight with PFA solution (G1101, Servicebio). The samples were subsequently dehydrated, embedded in paraffin, and sectioned. Antigen retrieval was performed by boiling the sections in sodium citrate buffer (EE0005, SparkJade) for 20 minutes. To suppress endogenous peroxidase activity, the sections were immersed in blocking solution (EE0007, SparkJade) at room temperature for 10 minutes, then blocked with 10% serum at 37°C for 30 minutes to minimize nonspecific staining. Subsequently, the sections were incubated overnight at 4°C with primary antibodies. After being rinsed, the sections were treated with HRP-conjugated secondary antibody (1:200, EF0002, SparkJade) and incubated for 40 minutes at 37°C. Immunoreaction sites were visualized with DAB, and this was followed by counterstaining with hematoxylin and mounting on glass slides. The mean integrated optical density (IOD) of the staining signal (indicating positive expression) was quantified for five samples per group in Image-Pro Plus software (version 6.0, Media Cybernetics). Primary antibodies to the following were used for immunohistochemistry (IHC): Ki-67 at 1:500, GB111141, Servicebio; ROCK1 at 1:800, 21850-1-AP, Proteintech; p-LIMK1 at 1:200, AF3345, Affinity; and p-CFL1 at 1:100, AP0178, ABclonal Technology.

            2.9 Hematoxylin and eosin staining

            The tissue sections were deparaffinized and rehydrated. The sections were stained with hematoxylin solution for 5 minutes, washed, treated with differentiation medium for 0.5 minutes, rinsed with distilled water, and stained with eosin solution for 2 minutes. The sections were subsequently dehydrated with a graded alcohol series, soaked in xylene, and sealed on slides. Imaging was performed with a scanning imaging system (WS-10, Wisleap).

            2.10 Detection of serum biochemical parameters

            Serum biochemical parameters, including alkaline phosphatase (ALP), alanine aminotransferase (ALT), and aspartate aminotransferase (AST) levels, were measured with specific assay kits (BC2145 for ALP, BC1555 for ALT, and BC1565 for AST, Beijing Solarbio Science & Technology) according to the manufacturer’s instructions.

            2.11 Animal studies

            Male Nu/Nu nude mice (5 weeks old) were obtained from Beijing Vital River Laboratory Animal Technology (license number: SCXK (Beijing) 2021-0006). The mice were housed under pathogen-free conditions with a 12-hour light/dark cycle, and given ad libitum access to food and water. All animal experiments were approved by the Ethics Committee of the Affiliated Hospital of Shandong University of Traditional Chinese Medicine (approval No. AWE-2022-016).

            For subchronic toxicity testing of PPII in vivo, the mice received intraperitoneal (IP) injections of 0, 3, or 6 mg/kg PPII every 2 days. After 63 days of treatment, the mice were euthanized, and blood samples were collected. Heart, liver, spleen, lung, kidney, and stomach tissues were harvested and fixed in tissue fixative (G1101, Servicebio). Hematoxylin and eosin (HE) staining was performed to examine morphological changes in the tissues, and the serum concentrations of ALP, ALT, and AST were determined as described above.

            For analysis of lung metastasis, T24 cells (2.0 × 106) resuspended in 100 μl PBS were injected into the tail vein of each Nu/Nu nude mouse. The mice received IP injections of PPII at doses of 0, 1.5, or 3.0 mg/kg every 2 days for 63 days. After treatment was completed, the mouse lungs were dissected and embedded in paraffin for HE and IHC analysis. The weights of the lungs and livers were recorded for each group.

            2.12 Statistical analysis

            Statistical analysis was performed via analysis of variance. The data are presented as mean ± standard deviation. The levels of statistical significance are indicated as follows: *P < 0.05, **P < 0.01, and ***P < 0.001. Nonsignificant differences (P > 0.05) are denoted with NS.

            3. RESULTS

            3.1 PPII inhibits BC cell migration and invasion

            The sensitivity of BC cells to PPII was evaluated by treatment of T24, 5637, and SV-HUC-1 cells with a concentration gradient of cisplatin (0–48 μM for cells; Figure 1A–C ) as a control or increasing concentrations of PPII (0–3.2 μM for T24 cells and 0–8 μM for 5637 and SV-HUC-1 cells; Figure 1D–F ) for 48 h. The IC50 values of cisplatin and PPII were then calculated for the three cell lines. PPII showed significant inhibitory effects on the viability of both T24 and 5637 cells, with IC50 values of 1.00 ± 0.04 μM and 1.87 ± 0.17 μM, respectively, thus indicating that PPII had greater inhibitory effects than cisplatin (17.95 ± 1.08 μM for T24 and 20.57 ± 0.27 μM for 5637). Moreover, SV-HUC-1 cells were less sensitive to cisplatin (29.53 ± 1.71 μM) and PPII (3.89 ± 0.42 μM) than T24 and 5637 cells.

            Next follows the figure caption
            Figure 1 |

            Effects of PPII on T24 and 5637 cell viability.

            Cytotoxicity of cisplatin as a positive control against T24 (A), 5637 (B), and SV-HUC-1 (C) cells after 48 h treatment. Cytotoxicity of PPII in T24 (D), 5637 (E), and SV-HUC-1 (F) cells after 48 h treatment. Screening for PPII concentrations not affecting cell viability in T24 (G) and 5637 (H) cells. NS, no significant difference; ***, P < 0.001; PPII, polyphyllin II.

            Considering the potential interference of drug-induced toxicity on the invasion and migration of tumor cells, we treated BC cells with low concentrations of PPII (0–0.8 μM) to identify noncytotoxic ranges. CCK-8 assays suggested that cell viability remained unaffected under 0–0.4 μM PPII treatment, whereas significant inhibition of viability was observed under 0.8 μM PPII treatment (P < 0.001, Figure 1G, H ) in both T24 and 5637 cells.

            The effect of PPII on BC migration was investigated with wound healing assays in T24 and 5637 cells, which revealed that 0.4 μM PPII significantly inhibited wound closure in both cell lines (P < 0.01, Figure 2A–D ). To assess the inhibitory effect of PPII on invasion, we subjected T24 and 5637 cells to Transwell invasion assays after treatment with PPII (0, 0.2, or 0.4 μM) for 48 h. PPII, even at low concentrations, significantly restrained the invasion of both T24 and 5637 cells (P < 0.05, Figure 2E–H ). Therefore, PPII suppressed BC cells’ migratory and invasive ability.

            Next follows the figure caption
            Figure 2 |

            PPII inhibits migration and invasion of BC cells.

            Wound healing assays of T24 (A) and 5637 (C) cells treated with PPII (0, 0.2, or 0.4 μM) for 48 h, and statistical analysis of the migration rates are presented in B and D, respectively. Transwell invasion assays of T24 (E) and 5637 (G) cells treated with PPII (0, 0.2, or 0.4 μM) for 48 h. Cells that crossed the membrane were counted and are presented in F and H. Scale bar: 50 μm; NS, no significant difference; *, P < 0.05, **, P < 0.01, ***, P < 0.001.

            3.2 RNA sequencing analysis of T24 cells treated with PPII

            For bioinformatics analysis of the molecular mechanism underlying the PPII-induced inhibition of BC cell migration and invasion, we treated T24 cells with PPII (0, 0.2, or 0.4 μM) for 48 h, and subsequently performed RNA sequencing. We identified 120 upregulated genes and 117 downregulated genes in the 0.2 μM PPII-treated group with respect to the control group (0 μM PPII, Figure 3A ), and 230 upregulated genes and 367 downregulated genes in the 0.4 μM PPII-treated group with respect to the control group ( Figure 3B ) with a fold change value > 1.25 of DEGs. The qPCR results confirmed that the mRNA levels of HNRNPC and RPL17 were increased, whereas the levels of NAA40 and ZNF619 were decreased in 0.4 μM PPII-treated group compared to control group ( Figure 3C ), thus confirming the reliability of the sequencing results.

            Next follows the figure caption
            Figure 3 |

            RNA sequencing analysis of T24 cells treated with PPII.

            Volcano plot showing the distribution of upregulated and downregulated genes in the 0.2 μM PPII-treated group versus the 0 μM PPII-treated group (A) and the 0.4 μM PPII-treated group versus the 0 μM PPII-treated group (B). qPCR detection of the mRNA levels of the top DEGs in the 0.4 μM PPII-treated group versus the 0 μM PPII-treated group (C). Gene expression heatmap showing a targeted cluster of genes whose expression levels decreased with increasing drug concentration (D). GO (E) and KEGG (F) analyses of the targeted cluster genes.

            Among all sequenced genes, one cluster (denoted the target cluster) of genes ( Figure 3D ) notably showed changes in gene expression consistent with the changes in drug concentration. That is, the expression levels of genes in this cluster, as compared with those in the control group, changed slightly after treatment with low concentrations of PPII (0.2 μM) but clearly decreased after treatment with high concentrations of PPII (0.4 μM).

            GO and KEGG analyses of the target cluster suggested enrichment in a series of terms associated with cell migration, invasion, and metastasis, such as positive regulation of cell adhesion, extracellular structure organization, basement membrane organization, and ECM-receptor interaction ( Figure 3E, F ). GO analysis further revealed that genes associated with cytoskeletal regulation, such as the regulation of actin filament-based process, regulation of actin cytoskeleton organization, and actin filament organization, were enriched in the target cluster ( Figure 3E ), whereas regulation of actin cytoskeleton was enriched in the KEGG analysis ( Figure 3F ). However, no specific cytoskeletal regulatory pathway was identified. Sorting of genes associated with cytoskeletal regulation in the GO ( Suppl. Table S1 ) and KEGG ( Suppl. Table S2 ) enrichment analyses revealed involvement of RHO GTPase-associated effectors, including RHO, RAC, and LIMK. Our findings indicated that the ability of PPII to inhibit BC cell motility was closely associated with cytoskeletal rearrangement, and that RHO GTPase-associated pathways might play important roles in PPII-mediated cytoskeletal regulation in BC cells.

            In summary, the treatment of T24 cells with PPII resulted in enrichment in gene expression associated with cell motility and cytoskeletal regulation. Consequently, we focused on the changes in the cytoskeleton induced by PPII.

            3.3 PPII represses cytoskeletal formation and CFL1 phosphorylation in BC cells

            Given that cytoskeletal rearrangement plays a crucial role in cell migration, invasion, and metastasis, we conducted F-actin staining assays with phalloidin to assess whether PPII treatment might induce cytoskeletal changes in BC cells. Representative images for each group revealed noticeably diminished cell protrusion and staining intensity under treatment with high concentrations of PPII (0.4 μM, Figure 4A ). Statistical analysis of the average F-actin intensity suggested a significant decrease in staining intensity, particularly in the 0.4 μM groups of T24 and 5637 cells compared to 0 μM groups (P < 0.05, Figure 4B, C ).

            Next follows the figure caption
            Figure 4 |

            PPII suppresses cytoskeletal formation in BC cells.

            T24 and 5637 cells were treated with PPII for 48 h, after which actin filaments were stained with TRITC-phalloidin (red), and nuclei were stained with DAPI (blue). Scale bar: 75 μm (A). Average calculated F-actin intensity of T24 (B) and 5637 (C) cells subjected to TRITC-phalloidin staining. T24 cells were treated with a PPII concentration gradient, as indicated, for 48 h, and the expression levels of cytoskeletal regulation-associated ROCK1, p-LIMK1/2, and p-CFL1 were detected by western blotting. Relative expression levels are displayed under the corresponding lanes (D). Bar chart showing relative expression levels of ROCK1 to GAPDH; p-LIMK1/2 to LIMK1; and p-CFL1 to CFL1 (E). The 5637 cells were treated similarly to T24 cells, and western blotting results and relative protein expression levels are presented in D and F, respectively. NS, no significant difference; *, P < 0.05, **, P < 0.01, ***, P < 0.001.

            The RHO GTPase pathway is widely recognized to play an essential role in cytoskeletal regulation. Our RNA sequencing results revealed the involvement of RHO GTPase-associated pathways in cytoskeletal changes after PPII treatment. CFL, which can sever actin filaments, is a common effector of multiple RHO GTPase pathways that regulates cytoskeletal polymerization and depolymerization; therefore, we detected the phosphorylation levels of CFL1 and its kinase LIMK1/2 via western blotting. Comparing to the control group (0 μM PPII), PPII decreased the levels of p-LIMK1/2 and p-CFL1 ( Figure 4D ). Further grayscale analysis revealed that p-LIMK1/2/LIMK1 and p-CFL1/CFL1 levels significantly decreased in the groups after treatment with 0.4 μM PPII (P < 0.05, Figure 4E, F ). The relative expression level of ROCK1, an upstream regulator of LIMK1/2, also decreased after 48 h treatment with 0.2 μM and 0.4 μM PPII in T24 and 5637 cells (P < 0.05, Figure 4D–F ). Thus, PPII decreased the expression levels of ROCK1 and suppressed the phosphorylation of effectors, including LIMK1/2 and CFL1.

            3.4 PPII inhibits RHOA-mediated migration, invasion, and CFL1 phosphorylation

            Given that ROCK1 acts as an effector of RHOA instead of CDC42 and RAC1, and that RHOA is abnormally activated in BC, we further assessed the changes in cell migration, invasion, and CFL1 phosphorylation under RHOA activation in BC cells.

            The effects of PPII on the invasion and migration of BC cells transfected with constitutively active RHOA (RHOA L63) were detected through Transwell assays. Comparing to the control group, migration assays without Matrigel revealed that RHOA L63 overexpression increased the numbers of T24 and 5637 cells that crossed the Transwell membrane, whereas PPII treatment inhibited the increase induced by the activation of RHOA ( Figure 5A, B ). Statistical analysis of cell count data confirmed that RHOA L63 overexpression significantly increased the numbers of BC cells that crossed the Transwell membrane; this number was further suppressed by the addition of 0.4 μM PPII ( Figure 5C, D ). Invasion assays with Matrigel were also performed with T24 ( Figure 5E, G ) and 5637 ( Figure 5F, H ) cells and yielded results consistent with those of the migration analysis. Briefly, RHOA L63 transfection increased the invasive ability of BC cells, whereas this increase was inhibited by the addition of 0.4 μM PPII.

            Next follows the figure caption
            Figure 5 |

            PPII inhibits cell migration and invasion, and CFL1 phosphorylation mediated by RHOA.

            Transwell migration assays of T24 (A) and 5637 (B) cells transfected with constitutively active RHOA mutant (RHOA L63) and treated with PPII for 48 h. Cells that crossed the membrane were counted and are presented in C and D, respectively. Transwell invasion assays of T24 (E) and 5637 (F) cells transfected with RHOA L63 and treated with PPII for 48 h. Cells that crossed the membrane were counted and are presented in G and H. Cells were treated as described above, and the expression levels of RHOA, ROCK1, and p-CFL1 were detected by western blotting. Relative expression levels are displayed under the corresponding lanes (I). Relative expression levels of ROCK1 to GAPDH, and p-CFL1 to CFL1 are shown in J and K, respectively. Scale bar: 50 μm; Statistical results are presented as means ± standard deviations of three independent experiments. *, P < 0.05, **, P < 0.01, ***, P < 0.001.

            BC cells were treated with 0.4 μM PPII, and RHOA L63 was simultaneously overexpressed. Western blotting revealed that RHOA L63 overexpression increased the expression levels of ROCK1 and p-CFL1 compared to the control group, whereas treatment with 0.4 μM PPII inhibited this increase ( Figure 5I ). Grayscale analysis also showed that the levels of ROCK1/GAPDH and p-CFL1/CFL1 increased with overexpression of RHOA L63 but decreased after PPII treatment (P < 0.05, Figure 5J, K ).

            In summary, the invasion and migration of T24 and 5637 cells were inhibited by PPII under RHOA activation. Mechanistically, PPII not only suppressed the expression of ROCK1 and p-CFL1 but also attenuated their expression in BC cells overexpressing RHOA L63.

            3.5 Subchronic toxicity of PPII in mice

            PPII treatment at 15 mg/kg and 25 mg/kg has been described in an in vivo study [19]; nevertheless, PPI and PPVI were used at a concentration of 5 mg/kg [20, 21]. Given the major differences in the concentrations used in animal experiments with different PPs and the lack of subchronic toxicity evaluation of PPII, we administered PPII (0, 3, or 6 mg/kg) to Nu/Nu nude mice via the IP route every 2 days.

            After 63 days of PPII treatment, abdominal distension was observed in the 6 mg/kg treatment group ( Suppl. Figure S1A–C ; weight curve in Figure 6A ). Mice were euthanized to obtain heart, liver, spleen, lung, kidney, and stomach tissues, which were weighed separately. Increases in the dimensions of the liver ( Figure 6B ) and spleen ( Suppl. Figure S1D ) were observed in 6 mg/kg PPII-treated group compared to control group. Furthermore, the weights of heart, liver, spleen, and lung tissues in the 6 mg/kg treatment group were significantly greater than those in the control group (0 mg/kg; P < 0.05, Figure 6C and Suppl. Figure S1E ). Moreover, no significant differences were observed in the weights of kidney and stomach tissues between the PPII-treated groups and the control group ( Figure 6C and Suppl. Figure S1E ).

            Next follows the figure caption
            Figure 6 |

            PPII induced weight increases and morphological changes in the livers and kidneys of Nu/Nu mice.

            Mice were euthanized after 63 days of PPII administration (0, 3, and 6 mg/kg; five mice per group) every 2 days. Weight curves of the treated groups are shown (A). Livers from the groups treated with a concentration gradient of PPII are presented (B). Bar charts showing the differences in the weights of the liver, lungs, and kidneys between groups (C). HE staining indicating morphological changes in the liver, lungs, and kidneys of groups treated with PPII. Scale bar: 100 μm (D). ALP, ALT, AST, and the AST/ALT ratio are displayed in bar charts (E). NS, no significant difference; **, P < 0.01, ***, P < 0.001.

            Subsequently, we conducted HE staining analysis on the above tissues. Compared with the control group, the group treated with 6 mg/kg PPII showed mild granular degeneration and visible focal necrosis in liver cells. In addition, aggregation of foam cells in the lungs, mild hypertrophy of myocardial cells, and extramedullary hematopoiesis in the spleen were found in the 6 mg/kg PPII treatment group, whereas the morphology of the kidney and stomach tissues did not change at the macroscopic level between treatments ( Figure 6D and Suppl. Figure S1F ). Furthermore, we detected liver serological indicators. Although no significant difference was observed in ALP, ALT, or AST between the PPII-treated groups and the control group, the AST/ALT ratio was significantly elevated in the 6 mg/kg treatment group (P < 0.01, Figure 6E ), thus indicating negative effects on liver function.

            The subchronic toxicity assay of PPII in Nu/Nu nude mice suggested that IP administration of 6 mg/kg PPII led to alterations in the weight and morphology of multiple tissues, including those of the heart, liver, spleen, and lungs, along with a significant increase in the AST/ALT ratio. In contrast, these effects were not observed at the 3 mg/kg dose. On the basis of these findings, we conducted subsequent animal experiments with drug concentrations of 0, 1.5, or 3.0 mg/kg.

            3.6 PPII inhibits BC cell metastasis in vivo

            On the basis of a previous report verifying successful lung colonization of T24 cells after injection into the caudal vein in mice [22], we further examined the effects of PPII on lung colonization and the formation of metastatic nodules by T24 cells, according to previously described methods. Nu/Nu nude mice were subjected to IP administration of PPII for 63 days after tail vein injection of T24 cells (2.0 × 106 cells per mouse, Suppl. Figure S2A ), and the weights of mice in the three groups (0, 1.5 and 3.0 mg/kg PPII) were plotted as weight curves ( Suppl. Figure S2B ). The lung tissues obtained from euthanized mice and the surface nodules on the lung tissue of each group are shown in Figure 7A . In agreement with the subchronic toxicity assessment results, PPII at concentrations of 1.5 or 3.0 mg/kg did not lead result in weights of the lungs ( Figure 7B ) or livers ( Suppl. Figure S2C ) differing from those in the control group.

            Next follows the figure caption
            Figure 7 |

            PPII inhibits lung metastasis of BC cells.

            Mice were injected with T24 cells via the tail vein and euthanized after 63 days of PPII administration (0, 1.5, or 3.0 mg/kg; five mice per group) every 2 days. Representative pulmonary metastatic nodules are shown. Scale bar: 5 mm (A). Differences in the weights of lung tissue (B) and the numbers of nodules (C) among the three treatment groups are shown in bar charts. HE staining revealing representative pulmonary metastatic nodules in the indicated groups (D). IHC showing the expression levels of ROCK1 (E) and p-CFL1 (F) in the indicated PPII treatment groups (scale bar: 100 μm). Differences in the mean IOD between groups were calculated. Arrows represent representative metastatic nodules. IOD, integrated optical density; NS, no significant difference; *, P < 0.05, **, P < 0.01, ***, P < 0.001.

            Statistical analysis of the numbers of tumor nodules among treatment groups revealed a decreasing trend in the group treated with 1.5 mg/kg PPII (IP) compared to the control group; however, no significant difference was observed. In contrast, the 3.0 mg/kg PPII group exhibited a significant decrease compared to 0 mg/kg PPII-treated group ( Figure 7C ). Representative tumor nodules detected by HE staining in each group are shown in Figure 7D . Furthermore, PPII (3.0 mg/kg) treatment significantly inhibited F-actin intensity at lung metastasis sites ( Suppl. Figure S2D and E ). The IHC results suggested significant downregulation of cell proliferation-associated Ki-67 expression levels in the 3.0 mg/kg PPII treatment group (P < 0.001, Suppl. Figure S2F, G ). Moreover, the protein levels of ROCK1 (P < 0.001, Figure 7E ), p-LIMK1 (P < 0.05, Suppl. Figure S2H, I ), and p-CFL1 (P < 0.05, Figure 7F ), which are associated with cytoskeletal regulation, were also significantly lower in the treatment group than the control group.

            In conclusion, PPII significantly inhibited the expression of the cytoskeletal regulatory proteins ROCK1, p-LIMK1, and p-CFL1 in vivo, and consequently impeded the formation of metastatic nodules after tail vein injection of T24 cells.

            4. DISCUSSION

            Our results indicated that PPII inhibits the invasion and migration of T24 and 5637 cells. Bioinformatics analysis revealed that PPII affects cytoskeletal rearrangement in BC cells, as indicated by diminished TRITC-phalloidin staining. Therefore, the inhibitory effect of PPII on BC cell invasion and migration might be achieved through induction of cytoskeletal rearrangement. Moreover, PPII treatment not only decreased the levels of ROCK1 and p-LIMK1, thereby inhibiting CFL1 phosphorylation, but also decreased the level of p-CFL1 when constitutively active RHOA L63 was overexpressed, thereby indicating that PPII suppressed RHOA-mediated CFL1 phosphorylation in BC cells. Moreover, the enhanced invasion and migration of BC cells induced by RHOA L63 was inhibited by PPII. Furthermore, subchronic toxicity detection demonstrated that IP administration of 6 mg/kg PPII in Nu/Nu nude mice led to morphological changes in multiple organs, including the liver and spleen, as well as dysregulation of serological indicators, such as the AST/ALT ratio. However, the 3 mg/kg concentration had limited adverse effects and was used in subsequent experiments. PPII was found to inhibit the formation of metastatic pulmonary nodules induced by tail vein injection of T24 cells, as well as the expression of ROCK1, p-LIMK1, and p-CFL1 associated with cytoskeletal regulation in vivo.

            We previously reported that P. polyphylla ethanol extract upregulates the expression of CDKN1A; inhibits the expression of CCNB1 and CDK1; induces G2/M phase arrest in J82 cells; and inhibits tumor growth in vivo [9]. In addition, we previously demonstrated that PPII, a main component of P. polyphylla, regulates EMT-associated factors, and decreases levels of MMP2 and MMP9 [8]. In the present study, we further revealed the molecular mechanism through which PPII inhibits the invasion and migration of T24 and 5637 cells through the ROCK1/LIMK/CFL1 pathway and cytoskeletal rearrangement; evaluated the subchronic toxicity of PPII; and finally determined the role of PPII in inhibiting metastasis in vivo ( Figure 8 ).

            Next follows the figure caption
            Figure 8 |

            PPII inhibits the metastasis signaling cascade through cytoskeletal rearrangement in BC cells (created in Figdraw; https://www.figdraw.com/#/).

            PPs from P. polyphylla have been widely used in antitumor experiments in vivo. For example, 1–3 mg/kg PPI has been used in animal models of lung cancer and ovarian cancer [2325], and glioma models established in BALB/c nude mice have been treated with 5 mg/kg PPVI [21]. However, PPII has also been administered at concentrations of 15 mg/kg and 25 mg/kg via IP injection into BALB/c nude mice with ovarian cancer xenograft tumors [19]. In addition, most of the animal studies on PPs described above used the IP administration route. The concentration of PPII used for lung metastasis of BC in Nu/Nu nude mice and its toxicity to the body had not been reported; therefore, we used IP administration to detect the effects of a concentration gradient of PPII (0, 3, and 6 mg/kg) on multiple organs in nude mice. Our results suggested increased liver, spleen, lung, and heart tissue weights; changes in tissue morphology revealed by HE staining; and an elevated AST/ALT ratio in the 6 mg/kg PPII treatment group, whereas 3 mg/kg PPII had relatively mild effects and was used in subsequent antimetastatic analyses. In subchronic toxicity evaluation, the 6 mg/kg treatment group also had ascites (four of five mice); therefore, the ratio of tissue to body weight was not used in the statistical analysis to minimize bias.

            The inactive form of CFL is phosphorylated (Ser3), whereas its active form is nonphosphorylated; its cleavage activity increases the efficiency of G-actin dissociation from F-actin, thus leading to F-actin depolymerization [26]. However, our conclusions did not consider the reuse of F-actin, although the depolymerized F-actin end can be reused for actin polymerization [26]. Therefore, CFL plays a dual regulatory role in both actin polymerization and depolymerization. Evidence suggests that CFL is involved in tumor cell invasion and migration [27, 28]; that diminished CFL phosphorylation inhibits invasion and migration [29]; and the decrease of p-CFL level also reported to promote cell motility [30]. Our results indicated that PPII inhibits the invasion, migration, and metastasis of BC cells by inhibiting CFL1 phosphorylation mediated by the ROCK1/LIMK axis, thus inhibiting tumor cell motility.

            RHO GTPases, an important branch of the Ras superfamily, regulate cytoskeletal rearrangement and are abnormally expressed in tumors. Moreover, these GTPases are considered potential biomarkers or therapeutic targets for cancer [13]. As important RHO GTPases, RHOA [31], RAC1 [32], and CDC42 [33] have been extensively studied in BC, and are closely associated with tumor progression, particularly migration, invasion, and metastasis [3436]. In the present study, PPII treatment was found to inhibit the expression of ROCK1, and the phosphorylation of its downstream effector factors LIMK1/2 and CFL1. Subsequently, the upstream regulator of ROCK1, the constitutively active mutant form of RHOA (RHOA L63), was overexpressed. Western blot and Transwell assays revealed that PPII inhibited RHOA-mediated CFL1 phosphorylation, as well as BC cell invasion and migration. Nevertheless, we cannot rule out the possibility RAC1 and CDC42 might play roles in the above process. Whether RAC1 and CDC42 participate in the regulatory effects of PPII on cytoskeletal rearrangement in BC cells must be verified in further studies.

            5. CONCLUSION

            In conclusion, this study revealed that PPII inhibits the phosphorylation of CFL1, and the invasion and migration of T24 and 5637 cells, via the ROCK1/LIMK axis in BC cells. Subchronic toxicity evaluation revealed that a drug concentration of 3.0 mg/kg had limited adverse effects on tissue morphology and the AST/ALT ratio in Nu/Nu mice. This concentration was confirmed to have inhibitory effects on BC lung metastasis after tail vein injection. Because of its potential benefits, PPII may be a valuable candidate for the development of future treatments aimed at managing BC metastasis.

            LIST OF ABBREVIATIONS

            ALP, Alkaline phosphatase; ALT, Alanine aminotransferase; AST, Aspartate aminotransferase; BC, Bladder cancer; DEGs, Differentially expressed genes; EMT, Epithelial-mesenchymal transition; GO, Gene ontology; HE, Hematoxylin and eosin; IHC, Immunohistochemistry; IOD, Integrated optical density; IP, Intraperitoneal; KEGG, Kyoto encyclopedia of genes and genomes; MIBC, Muscle-invasive bladder cancer; MMPs, Matrix metalloproteinase; NMIBC, Nonmuscle-invasive bladder cancer; PFA, Paraformaldehyde; PPs, Polyphyllins; TCMs, Traditional Chinese medicines.

            ETHICS APPROVAL AND CONSENT TO PARTICIPATE

            All experiments involving animals were approved by the Ethics Committee of the Affiliated Hospital of Shandong University of Traditional Chinese Medicine (No. AWE-2022-016) and conformed to National Institutes of Health (NIH) guidelines for the ethical treatment and use of laboratory animals in scientific research.

            CONSENT FOR PUBLICATION

            All authors agreed to publish this study.

            AVAILABILITY OF DATA AND MATERIALS

            The raw sequence data reported in this paper have been deposited in the Genome Sequence Archive (Genomics, Proteomics & Bioinformatics 2021) in National Genomics Data Center (Nucleic Acids Res 2022), China National Center for Bioinformation / Beijing Institute of Genomics, Chinese Academy of Sciences (GSA-Human: HRA005259) that are publicly accessible at https://ngdc.cncb.ac.cn/gsa-human. The datasets produced and analyzed in the present study can be obtained from the corresponding author upon a reasonable request.

            CONFLICTS OF INTEREST

            All authors declare no conflicts of interest.

            SUPPLEMENTARY MATERIAL

            REFERENCES

            1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al.. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: A Cancer Journal for Clinicians. 2021. Vol. 71:209–249

            2. Yang L, Huang W, Bai X, Wang H, Wang X, Xiao H, et al.. Androgen Dihydrotestosterone Promotes Bladder Cancer Cell Proliferation and Invasion via EPPK1-Mediated MAPK/JUP Signalling. Cell Death & Disease. 2023. Vol. 14:363

            3. Lenis AT, Lec PM, Chamie K, Mshs MD. Bladder Cancer: A Review. JAMA. 2020. Vol. 324:1980–1991

            4. Guneri-Sozeri PY, Ozden-Yilmaz G, Kisim A, Cakiroglu E, Eray A, Uzuner H, et al.. FLI1 and FRA1 Transcription Factors Drive the Transcriptional Regulatory Networks Characterizing Muscle Invasive Bladder Cancer. Communications Biology. 2023. Vol. 6:199

            5. Fu J, Zhang W, Jiang T. Immunogenic Cell Death Mediation Patterns Reveal Novel Paradigm for Characterizing the Immune Microenvironment and Immunotherapeutic Responses in Bladder Cancer. Frontiers in Genetics. 2022. Vol. 13:1035484

            6. Martins-Lima C, Chianese U, Benedetti R, Altucci L, Jeronimo C, Correia MP. Tumor Microenvironment and Epithelial-Mesenchymal Transition in Bladder Cancer: Cytokines in the Game? Frontiers in Molecular Biosciences. 2022. Vol. 9:1070383

            7. Wang Q, Mo J, Zhao C, Huang K, Feng M, He W, et al.. Raddeanin A Suppresses Breast Cancer-Associated Osteolysis Through Inhibiting Osteoclasts and Breast Cancer Cells. Cell Death & Disease. 2018. Vol. 9:376

            8. Niu W, Xu L, Li J, Zhai Y, Sun Z, Shi W, et al.. Polyphyllin II Inhibits Human Bladder Cancer Migration and Invasion by Regulating EMT-Associated Factors and MMPs. Oncology Letters. 2020. Vol. 20:2928–2936

            9. Liu Z, Sun Z, Zhang D, Ma C, Jiang Y, Cao G, et al.. Paris Polyphylla Ethanol Extract Induces G2/M Arrest and Suppresses Migration and Invasion in Bladder Cancer. Translational Cancer Research. 2020. Vol. 9:5994–6004

            10. Qin XJ, Ni W, Chen CX, Liu HY. Seeing the Light: Shifting from Wild Rhizomes to Extraction of Active Ingredients from Above-Ground Parts of Paris Polyphylla var. Yunnanensis. Journal of Ethnopharmacology. 2018. Vol. 224:134–139

            11. Liu L, Cui J, Zhao Y, Liu X, Chen L, Xia Y, et al.. KDM6A-ARHGDIB Axis Blocks Metastasis of Bladder Cancer by Inhibiting Rac1. Molecular Cancer. 2021. Vol. 20:77

            12. Zeng RJ, Zheng CW, Chen WX, Xu LY, Li EM. Rho GTPases in Cancer Radiotherapy and Metastasis. Cancer and Metastasis Reviews. 2020. Vol. 39:1245–1262

            13. Wang T, Rao D, Yu C, Sheng J, Luo Y, Xia L, et al.. RHO GTPase Family in Hepatocellular Carcinoma. Experimental Hematology & Oncology. 2022. Vol. 11:91

            14. Ungefroren H, Wellner UF, Keck T, Lehnert H, Marquardt JU. The Small GTPase RAC1B: A Potent Negative Regulator of-and Useful Tool to Study-TGFbeta Signaling. Cancers. 2020. Vol. 12:3475

            15. Hall A. The Cytoskeleton and Cancer. Cancer Metastasis Reviews. 2009. Vol. 28:5–14

            16. Love MI, Huber W, Anders S. Moderated Estimation of Fold Change and Dispersion for RNA-Seq Data with DESeq2. Genome Biology. 2014. Vol. 15:550

            17. Kumar L, Futschik ME. Mfuzz: A Software Package for Soft Clustering of Microarray Data. Bioinformation. 2007. Vol. 2:5–7

            18. Wu T, Hu E, Xu S, Chen M, Guo P, Dai Z, et al.. ClusterProfiler 4.0: A Universal Enrichment Tool for Interpreting Omics Data. Innovation (Camb). 2021. Vol. 2:100141

            19. Yang M, Zou J, Zhu H, Liu S, Wang H, Bai P, et al.. Paris Saponin II Inhibits Human Ovarian Cancer Cell-Induced Angiogenesis by Modulating NF-KappaB Ssignaling. Oncology Reports. 2015. Vol. 33:2190–2198

            20. Long J, Pi X. Polyphyllin I Promoted Melanoma Cells Autophagy and Apoptosis via PI3K/Akt/mTOR Signaling Pathway. Biomed Research International. 2020. Vol. 2020:5149417

            21. Liu W, Chai Y, Hu L, Wang J, Pan X, Yuan H, et al.. Polyphyllin VI Induces Apoptosis and Autophagy via Reactive Oxygen Species Mediated JNK and P38 Activation in Glioma. OncoTargets and Therapy. 2020. Vol. 13:2275–2288

            22. Liu X, Cui J, Gong L, Tian F, Shen Y, Chen L, et al.. The CUL4B-miR-372/373-PIK3CA-AKT Axis Regulates Metastasis in Bladder Cancer. Oncogene. 2020. Vol. 39:3588–3603

            23. Gu L, Feng J, Zheng Z, Xu H, Yu W. Polyphyllin I Inhibits the Growth of Ovarian Cancer Cells in Nude Mice. Oncology Letters. 2016. Vol. 12:4969–4974

            24. Li L, Wu J, Zheng F, Tang Q, Wu W, Hann SS. Inhibition of EZH2 via Activation of SAPK/JNK and Reduction of p65 and DNMT1 as a Novel Mechanism in Inhibition of Human Lung Cancer Cells by Polyphyllin I. Journal of Experimental & Clinical Cancer Research: CR. 2016. Vol. 35:112

            25. Lai L, Shen Q, Wang Y, Chen L, Lai J, Wu Z, et al.. Polyphyllin I Reverses the Resistance of Osimertinib in Non-Small Cell Lung Cancer Cell Through Regulation of PI3K/Akt Signaling. Toxicology Applied Pharmacology. 2021. Vol. 419:115518

            26. Bravo-Cordero JJ, Magalhaes MA, Eddy RJ, Hodgson L, Condeelis J. Functions of Cofilin in Cell Locomotion and Invasion. Nature Reviews Molecular Cell Biology. 2013. Vol. 14:405–415

            27. Zhuang H, Li Q, Zhang X, Ma X, Wang Z, Liu Y, et al.. Downregulation of Glycine Decarboxylase Enhanced Cofilin-Mediated Migration in Hepatocellular Carcinoma Cells. Free Radical Biology Medicine. 2018. Vol. 120:1–12

            28. Yao B, Li Y, Chen T, Niu Y, Wang Y, Yang Y, et al.. Hypoxia-Induced Cofilin 1 Promotes Hepatocellular Carcinoma Progression by Regulating the PLD1/AKT Pathway. Clinical and Translational Medicine. 2021. Vol. 11:e366

            29. Pipatpanyanugoon N, Wareesawetsuwan N, Prasopporn S, Poolex W, Pisitkun T, Kaewkong W, et al.. BAIAP2L1 Enables Cancer Cell Migration and Facilitates Phospho-Cofilin Asymmetry Localization in the Border Cells. Cancer Communication. 2022. Vol. 42:75–79

            30. Peverelli E, Giardino E, Treppiedi D, Locatelli M, Vaira V, Ferrero S, et al.. Dopamine Receptor Type 2 (DRD2) Inhibits Migration and Invasion of Human Tumorous Pituitary Cells Through ROCK-Mediated Cofilin Inactivation. Cancer Letters. 2016. Vol. 381:279–286

            31. Chen H, Yang W, Li Y, Ji Z. PLAGL2 Promotes Bladder Cancer Progression via RACGAP1/RhoA GTPase/YAP1 Signaling. Cell Death & Disease. 2023. Vol. 14:433

            32. Hua X, Xiang D, Guo M, Qian X, Chen R, Li T, et al.. Induction of RAC1 Protein Translation and MKK7/JNK-Dependent Autophagy Through Dicer/miR-145/SOX2/miR-365a Axis Contributes to Isorhapontigenin (ISO) Inhibition of Human Bladder Cancer Invasion. Cell Death & Disease. 2022. Vol. 13:753

            33. Redelman-Sidi G, Iyer G, Solit DB, Glickman MS. Oncogenic Activation of Pak1-Dependent Pathway of Macropinocytosis Determines BCG Entry into Bladder Cancer Cells. Cancer Research. 2013. Vol. 73:1156–1167

            34. Tuntithavornwat S, Shea DJ, Wong BS, Guardia T, Lee SJ, Yankaskas CL, et al.. Giant Obscurin Regulates Migration and Metastasis via RhoA-Dependent Cytoskeletal Remodeling in Pancreatic Cancer. Cancer Letters. 2022. Vol. 526:155–167

            35. Mei C, Liu C, Gao Y, Dai WT, Zhang W, Li X, et al.. eIF3a Regulates Colorectal Cancer Metastasis via Translational Activation of RhoA and Cdc42. Frontiers Cell Developments Biology. 2022. Vol. 10:794329

            36. Zhu Z, Yu Z, Rong Z, Luo Z, Zhang J, Qiu Z, et al.. The Novel GINS4 Axis Promotes Gastric Cancer Growth and Progression by Activating Rac1 and CDC42. Theranostics. 2019. Vol. 9:8294–8311

            Graphical abstract

            Next follows the Graphical Abstract

            Highlights
            • PPII suppresses the ROCK1/LIMK/CFL1 pathway in BC cells.

            • PPII (3.0 mg/kg) has limited effects on tissue morphology and serological indicators.

            • PPII inhibits the formation of metastatic pulmonary nodules.

            • Expression of cytoskeletal regulatory proteins is restrained by PPII in vivo.

            In brief

            This study identified the crucial role of PPII in inhibiting the migration, invasion, and metastasis of BC cells by modulating the ROCK1/LIMK/CFL1 pathway, as demonstrated through both in vivo and in vitro experiments. Additionally, the study also assesses the subchronic toxicity of PPII.

            Author and article information

            Journal
            amm
            Acta Materia Medica
            Compuscript (Ireland )
            2737-7946
            21 November 2024
            : 3
            : 4
            : 405-422
            Affiliations
            [a ]Central Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
            [b ]Institute for Literature and Culture of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
            [c ]Shandong Key Laboratory of Dominant Diseases of Traditional Chinese Medicine, Jinan, Shandong, China
            [d ]Medical Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
            [e ]School of Public Health, North China University of Science and Technology, Tangshan, Hebei, China
            [f ]First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
            [g ]Nephropathy Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
            [h ]Department of Pathology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
            [i ]Neurology Department, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
            [j ]Department of Pharmacy, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
            [k ]Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
            Author notes
            *Correspondence: xuxiangdong0818@ 123456163.com , Tel: 0531-68616761 (X. Xu); songql1025@ 123456126.com , Tel: 0531-68617994 (Q. Song); zhenguow@ 123456vip.126.com , Tel: 0531-68616427 (Z. Wang)
            Author information
            https://orcid.org/0000-0002-5392-9230
            https://orcid.org/0009-0005-5024-8789
            Article
            10.15212/AMM-2024-0043
            35a34185-b264-4ee9-8a3d-c22d45467f99
            2024 The Authors.

            Creative Commons Attribution 4.0 International License

            History
            : 18 July 2024
            : 11 October 2024
            : 19 October 2024
            Page count
            Figures: 8, Tables: 1, References: 36, Pages: 18
            Funding
            Funded by: Natural Science Foundation of Shandong Province, China
            Award ID: ZR2020QH337
            Funded by: China Postdoctoral Science Foundation
            Award ID: 2022M722000
            We thank Professor Shengtian Zhao (Binzhou Medical University, Yantai, Shandong) for providing the T24, 5637, and SV-HUC-1 cell lines, and the Shanghai Biotechnology Corporation for sequencing and preliminary data analysis. The present study was supported by the Natural Science Foundation of Shandong Province, China (grant no. ZR2020QH337), and the China Postdoctoral Science Foundation (grant no. 2022M722000).
            Categories
            Research Article

            Toxicology,Pathology,Biochemistry,Clinical chemistry,Pharmaceutical chemistry,Pharmacology & Pharmaceutical medicine
            Bladder cancer,Metastasis,CFL1 phosphorylation,ROCK1,Cytoskeleton formation,Polyphyllin II

            Comments

            Comment on this article