The underlying molecular mechanism driving clear cell renal cell carcinoma (ccRCC) progression is still to be explored. The significant downregulation of protein tyrosine phosphatase nonreceptor type 3 (PTPN3) expression in the tumor tissues suggested its protective role in ccRCC progression. IHC analysis of PTPN3 protein in 172 ccRCC tissue revealed that PTPN3 was an independently favorable prognostic factor for progression-free survival (P = 0.0166) and overall survival (P = 0.0343) of patients. The ccRCC cell lines SN12C, 1932, ACHN, and Caki-1 were used to evaluate, both in vitro and in vivo, the biological roles of PTPN3. We observed that overexpression of PTPN3 significantly inhibited the proliferation, migration, and invasion of ccRCC cells. In contrast, the knocking down of PTPN3 elicited opposite effects. Overexpressing PTPN3 inhibited xenograft tumor growth and lung metastasis displayed by the in vivo mice models. PTPN3 inhibited tumor cell motility by suppressing the phosphorylation of AKT, and subsequently inactivating the PI3K/AKT signaling pathway of renal cell carcinoma cells. Furthermore, the inhibition of phospho-AKTThr308 and phospho-AKTSer473 reversed PTPN3-induced silencing in tumor cell migration. Our work revealed that the overexpression of PTPN3 could suppress kidney cancer progression by negatively regulating the AKT signaling pathway, and served as a favorable prognostic factor in patients with ccRCC. Our findings provided insight that PTPN3 could be a potential target for therapy aiming to inhibit the malignant behaviors of ccRCC.

Implications:

PTPN3 is an independent favorable prognostic factor for patients with ccRCC and could be a potential target for therapy aiming to inhibit the malignant behaviors of ccRCC.

Kidney cancer represents about 4% of all new cancer diagnoses (1). The most common form of kidney cancer arises from renal epithelium, termed as renal cell carcinoma (RCC; ref. 2). As the most common histologic subtype, clear cell renal cell carcinoma (ccRCC) accounts for 70%–75% of all RCC cases (3). The worldwide incidence and mortality rate of ccRCC has been estimated rising by 2%–3% per decade (4). In spite of the fact that the small renal masses have been more frequently detected in recent year, one third of patients with ccRCC will suffer from metastatic diseases after the diagnosis (5). The treating of metastatic ccRCC still remains a challenge as the underlying mechanism(s) is yet to be elucidated (6, 7).

The PI3K/protein kinase B (also named AKT) signaling pathway has been shown to regulate a series of cellular functions in various types of cancer, including proliferation, autophagy, apoptosis, cell cycle, and metastasis (8, 9). Consequently, several potential therapeutic targets of this signaling pathway have been proposed (10), which have also been found to be activated and playing important roles in ccRCC progression (11, 12).

Protein tyrosine kinases and protein tyrosine phosphatases (PTP) reversibly regulate the phosphorylation of tyrosine proteins, acting as a molecular switch for the regulation of various biological processes (13). Aa a membrane-associated nonreceptor of PTP, PTPN3 contains a C-terminal phosphatase domain, a middle PDZ domain and an N-terminal FERM domain (14). These three domains involve in the regulation of cellular proliferation and growth (15). As a tyrosine phosphatase, PTPN3 could dephosphorylate several substrates, for example, the EGFR pathway substrate 15 (13). However, its role as a nonreceptor protein tyrosine kinase is still unclear.

Accumulating evidence has suggested an equivocal role of PTPN3 in the progression of a variety of human cancers (13, 14, 16). Li and colleagues found that the ectopic expression of PTPN3 in non–small cell lung cancer cells suppressed cellular motility, proliferation, and tumor growth via the regulation of EGFR endocytic trafficking, degradation, and signaling (13). On the contrary, Hou and colleagues showed that PTPN3 may increase malignant growth by cooperating with its substrate, p38γ (16). Gao and colleagues also found that numerous activating mutations and high expression levels of PTPN3 were associated with intrahepatic cholangiocarcinoma recurrence (14). Thereby, the exact role of PTPN3 in ccRCC remains undetermined.

In this study, through the findings of various investigations, we reported that PTPN3 could suppress ccRCC progression by negatively regulating the AKT signaling pathway, providing insight that PTPN3 could be a potential target for therapy aiming to inhibit the malignant behaviors of ccRCC.

Cell lines and culture

Originally bought from ATCC, ccRCC cell lines SN12C, 1932, Caki-1, ACHN were routinely maintained in liquid nitrogen in our laboratory (17, 18). The cell lines were cultured in DMEM or 1640 (Gibco) supplemented with 10% FBS (Gibco) with 100 IU/mL of streptomycin and 100 μg/mL of penicillin in a humidified atmosphere of 5% CO2 at 37°C as described previously (19). We used the short tandem repeat (STR) testing to authenticate all cells and make sure that they were maintained Mycoplasma-free.

Patient tissue samples

Twelve ccRCC tissues and matched paracancerous tissues were retrieved from the Department of Urology, Sun Yat-sen University Cancer Center (SYSUCC, Guangzhou, China) and stored at − 80°C before use for quantitative real-time reverse-transcription PCR (qRT-PCR) analysis. Another cohort of 172 formalin-fixed, paraffin-embedded primary ccRCC specimens collected from the Department of Pathology, SYSUCC was used for IHC evaluation. Written informed consent was obtained from all patients and the study was approved by the ethics committees of SYSUCC as described previously (19).

RNA isolation and qRT-PCR

We used TRIzol reagent (Invitrogen) to isolate total RNA. Complementary DNA (cDNA) was synthesized using a reverse transcription kit (K1622, Thermo Fisher Scientific) following the manufacturer's instructions as described previously (19). qRT-PCR analysis was performed using the SYBR Green PCR Kit (Novazym), and β-actin was used as the internal control. The sequences of real-time PCR primers for β-actin and PTPN3 were as follows: forward: 5′-CACCATTGGCAATGAGCGGTTC-3′ and reverse: 5′-AGGTCTTTGCGGATGTCCACGT-3′, and forward 5′-GGACATCTCAGAACACACGCATG-3′ and reverse 5′-GAAGTCAGCAAATGAGCGGACAG-3′, respectively.

MTS assay

The cells were plated into 96-well plates at a density of 2,000 cells/well in 200 μL normal culture medium. Cell growth was determined using MTS (Cell Titer 96 Aqueous One Solution Cell Proliferation Assay solution; Promega), as described previously (19). Every day at the same time, 10 μL of the MTS reagents were added to 100 μL culture medium per well and incubated for 2–4 hours at 37°C. The microplate reader (Bio-Tek EPOCH2) was used to read the OD490 of these cells seeded into 96-well plates. All experiments were repeated three times independently.

RNA interference

For transient knockdown of PTPN3, by using the Lipofectamine RNAiMAX Reagent (Invitrogen), siRNAs targeting human PTPN3 were transfected into cells. Gene-Pharma Biological Technology made the siRNAs whose sequences target human PTPN3:5′-GCGUGGUACAGACCUUUAATT-3′(si1#) and 5′-GCUGAAUCCAGGGAACAUATT-3′(si2#), and 5′-CATTAATGTCGGACAAC-3′ for the negative control sequence.

Clonogenic assay

Equal numbers of cells were seeded at a density of 500 cells/mL into 6-well plates and incubated for 10 days at 37°C. At the end of the experiments, the cells were stained with 0.1% crystal violet for 15 minutes. We used the microscope (NIKON ECLIPSE 80i) to count the number of positive colonies, those with >50 cells.

Lentiviral transduction studies

PTPN3-overexpressing plasmid or vector were purchased from Genecopoeia Co. Ltd. as described previously (19). Lentiviruses were produced in 293 T cells cotransfected with lentiplasmid and packing plasmids using the X-treme GENE DNA transfection reagents (Roche). The cells were first incubated with infectious lentiviruses for 12 hours and then exchanged to incubation with fresh medium for another 12 hours, after which they were selected with puromycin (2 ng/L) for 3 days and validated by qRT-PCR and immunoblotting (19).

Western blot analysis

The primary antibodies used were PTPN3 (#GTX54572, GeneTex), GAPDH (#60004-1-Ig, ProteinTech), Phospho-AKT (Thr308; #13038, Cell Signaling Technology), Phospho-AKT (Ser473; #4060P, Cell Signaling Technology), AKT (#60203-2-Ig ProteinTech). We purchased the horseradish peroxidase–conjugated goat anti-rabbit or anti-mouse secondary antibodies from Vazyme Biotech. Proteins were visualized with an enhanced chemiluminescence detection system (Bio-Rad) as described previously (19).

Migration and invasion assays

A total of 5 × 104 cells of ACHN, 3 × 104 cells of Caki-1, 4 × 104 cells of SN12C, or 2 × 104 cells of 1932 were seeded in a serum-free medium in the top chambers (Corning) with 700 μL of DMEM with 10% FBS in the bottom chambers. After culturing for 24 hours, the cells were fixed with methanol and then they were stained with 0.1% crystal violet for 30 minutes at room temperature. For invasion assays, the top chamber membranes were coated with Matrigel (Corning, Life Sciences). Three random fields per well were observed, and cells were counted under a microscope (NIKON ECLIPSE 80i) as described previously (19). The inhibitor of AKT–MK-2206 (1 μmol/L) was purchased from Selleck Chemicals. Both experiments were performed in triplicates.

Wound healing assays

Cells were seeded into 6-well plates (Corning) and cultured to a 90% confluent followed by 24-hour starvation in a serum-free medium. We used the sterile 200-μL tip to create artificial wounds in the cell monolayer and the floating cells were removed by washing with PBS as described previously (19). We used an inverted microscope (OLYMPUS IX73) to capture respective images at 0, 6, 12, 24, 48, 72, and 96 hours.

IHC staining

Rabbit anti-PTPN3 antibody (1:100 dilution, #GTX54572, GeneTex) was used, and the IHC staining results were assigned a mean score considering both the intensity of staining and the proportion of tumor cells with an unequivocal positive reaction as described previously (19). Two pathologists without prior knowledge of patient data assessed each section independently. The percentage of stained cells was scored as follows: 0% as 0; 1%–10% as 1; 11%–50% as 2; 51%–80% as 3; 81%–100% as 4, and the intensity of staining: no staining as 0; weak staining as 1; moderate staining as 2; strong staining as 3. Then multiplying the two was the score of each tissue (score range: 0–12).

Animal experiments

Female athymic mice between 4 and 6 weeks of age were obtained from Beijing Vital River Laboratory Animal Center. Mice were cared in accordance with the principles and procedures outlined in the Institutional Animal Care and Use Committee of SYSUCC as described previously (19). A total of 8 × 106/0.15 mL SN12C cells stably expressing scrambled shRNA control or shPTPN3 were injected subcutaneously into the mice. Ten days after the injection, the mice were sacrificed and the tumors were excised and measured. We injected SN12C kidney tumor cells persistently expressing scrambled shRNA control or shPTPN3 (1.5 × 106 cells/0.1 mL PBS) into the lateral tail vein of nude mice to get lung metastases. After eight weeks, the lungs were resected, weighed, and photographed. We counted the number of metastatic nodules on the surface of the lungs visually after staining with picric acid. Paraformaldehyde was used to fix the lungs for paraffin sections and then hematoxylin and eosin were used to stain the lungs with a standard histologic protocol.

Statistical analysis

The SPSS V.14 software package was used to perform statistical data analyses. P < 0.05 was considered significant. Data are presented as the mean ± SD. Multivariate analyses were performed using the Cox proportional hazards model. Survival curves were constructed using the Kaplan–Meier method and compared using the log-rank test as described previously (19). We used t test to compare PTPN3 mRNA levels between normal and tumor samples from the kidney renal clear cell carcinoma dataset of The Cancer Genome Atlas (TCGA). We have uploaded the key raw data onto the Research Data Deposit (RDD) platform (www.researchdata.org.cn) to validate the authenticity of this article, with the approval number as RDDB2019000767.

Reduced expression of PTPN3 correlate with poor prognosis in patients with ccRCC

We investigated the expression of PTPN3 in ccRCC tissues. Quantitative real-time PCR analysis showed that PTPN3 mRNA was dramatically lower in tumoral tissues as compared with their corresponding adjacent normal renal tissues in the 12 matched human ccRCC tissues (Fig. 1A). We also performed IHC staining in 172 human ccRCC samples (Fig. 1B) to evaluate the expression level of PTPN3 in ccRCC tissue. PTPN3 mainly expressed in the cytoplasm of tumor cells in those IHC-stained human ccRCC tissue samples. The association between PTPN3 expression and clinicopathologic features to survival results are summarized in Tables 1 and 2. Patients with tumors having high PTPN3 expression (cut-off score > 6) had significantly longer progression-free survival (PFS) and overall survival (OS) than those with low PTPN3 expression tumors (Fig. 1C; P = 0.0343 vs. P = 0.0166). Multivariate analyses revealed that high PTPN3 expression was an independent and favorable prognostic indicator for PFS (P = 0.047) in all patients (Table 2).

Figure 1.

PTPN3 mRNA and protein expression levels in ccRCC tissues and cell lines. A, qRT-PCR analysis of PTPN3 mRNA expression levels in ccRCC matched tissues (n = 12). The mRNA levels of PTPN3 in ccRCC tissues were lower than the paired normal tissues (P = 0.0006, paired t test). B, Levels of PTPN3 protein expression in ccRCC tissue chip are shown under both low and high magnifications of a light microscope; low PTPN3 level correlated with shorter overall survival and progression-free survival in patients with ccRCC. C, In ccRCC tissue chip from our hospital, the overall survival (OS) and progression-free survival (PFS) rate were significantly higher in the higher PTPN3 group rate. D, Similarly, using data from the TCGA database, we observed that the overall survival and progression-free survival rate was significantly higher in the higher PTPN3 group.

Figure 1.

PTPN3 mRNA and protein expression levels in ccRCC tissues and cell lines. A, qRT-PCR analysis of PTPN3 mRNA expression levels in ccRCC matched tissues (n = 12). The mRNA levels of PTPN3 in ccRCC tissues were lower than the paired normal tissues (P = 0.0006, paired t test). B, Levels of PTPN3 protein expression in ccRCC tissue chip are shown under both low and high magnifications of a light microscope; low PTPN3 level correlated with shorter overall survival and progression-free survival in patients with ccRCC. C, In ccRCC tissue chip from our hospital, the overall survival (OS) and progression-free survival (PFS) rate were significantly higher in the higher PTPN3 group rate. D, Similarly, using data from the TCGA database, we observed that the overall survival and progression-free survival rate was significantly higher in the higher PTPN3 group.

Close modal
Table 1.

Association between expression of PTPN3 and clinicopathologic characteristics in 172 patients with ccRCC.

CasesPTPN3 Expression
Clinical factors(n = 172)HighLowP
Gender 
 Female 47 37 10 0.428 
 Male 125 91 34  
Age 
 ≤50 86 65 21 0.727 
 >50 86 63 23  
T stage 
 T1 131 99 32 0.535 
 T2 41 29 12  
Fuhrman score 
 1 33 23 10 0.243 
 2 104 81 23  
 3 30 22  
 4  
Sarcomatoid differentiation score 
 0 168 126 42 0.257 
 1  
Necrosis score 
 0 136 103 33 0.442 
 1 36 25 11  
Vascular invasion score 
 0 164 123 41 0.429 
 1  
Nerve invasion score 
 0 172 128 44 — 
 1  
Fibrillar collagen score 
 0 49 40 0.172 
 1 123 88 35  
Lymph node invasion score 
 0 172 128 44 — 
 1  
CasesPTPN3 Expression
Clinical factors(n = 172)HighLowP
Gender 
 Female 47 37 10 0.428 
 Male 125 91 34  
Age 
 ≤50 86 65 21 0.727 
 >50 86 63 23  
T stage 
 T1 131 99 32 0.535 
 T2 41 29 12  
Fuhrman score 
 1 33 23 10 0.243 
 2 104 81 23  
 3 30 22  
 4  
Sarcomatoid differentiation score 
 0 168 126 42 0.257 
 1  
Necrosis score 
 0 136 103 33 0.442 
 1 36 25 11  
Vascular invasion score 
 0 164 123 41 0.429 
 1  
Nerve invasion score 
 0 172 128 44 — 
 1  
Fibrillar collagen score 
 0 49 40 0.172 
 1 123 88 35  
Lymph node invasion score 
 0 172 128 44 — 
 1  
Table 2.

Univariate and multivariate analyses of different parameters for overall survival (OS) and progression-free survival (PFS) of patients with ccRCC.

Univariate analysisMultivariate analysis
VariablesHR (CI)PHR (CI)P
OS 
 Fuhrman score 2.322 (1.519–3.550) <0.001 2.322 (1.519–3.550) <0.001 
 Sarcomatoid differentiation score 6.031 (1.847–19.692) 0.001 1.695 (0.423–6.788) 0.325 
 PTPN3 level 0.504 (0.264–0.962) 0.034 0.568 (0.294–1.099) 0.071 
PFS 
 Fuhrman score 2.13 (1.420–3.194) <0.001 2.017 (1.356–2.999) 0.001 
 Sarcomatoid differentiation score 5.69 (1.753–18.471) 0.001 1.719 (0.435–6.788) 0.435 
 PTPN3 level 0.483 (0.263–0.888) 0.017 0.527 (0.285–0.974) 0.047 
Univariate analysisMultivariate analysis
VariablesHR (CI)PHR (CI)P
OS 
 Fuhrman score 2.322 (1.519–3.550) <0.001 2.322 (1.519–3.550) <0.001 
 Sarcomatoid differentiation score 6.031 (1.847–19.692) 0.001 1.695 (0.423–6.788) 0.325 
 PTPN3 level 0.504 (0.264–0.962) 0.034 0.568 (0.294–1.099) 0.071 
PFS 
 Fuhrman score 2.13 (1.420–3.194) <0.001 2.017 (1.356–2.999) 0.001 
 Sarcomatoid differentiation score 5.69 (1.753–18.471) 0.001 1.719 (0.435–6.788) 0.435 
 PTPN3 level 0.483 (0.263–0.888) 0.017 0.527 (0.285–0.974) 0.047 

Note: Statistical significance (P < 0.05) is shown in bold.

Abbreviation: CI, confidence interval.

We also analyzed the gene expression data of patients with ccRCC in the TCGA database. PTPN3 mRNA was found to be significantly lower in all tumor samples than in paired normal tissues (Supplementary Fig. S1A–S1C; P < 0.05). For separating the cases into low and high PTPN3 expression groups, the mean expression level was used as the cut-off value. Kaplan–Meier survival analyses of TCGA data also showed that lower expression of PTPN3 was associated with poorer survival in ccRCC (Fig 1D; P < 0.0001). These observations suggest that low levels of PTPN3 may act as a novel prognostic factor for patients with ccRCC.

Knocking down PTPN3 promotes ccRCC cell growth in vitro and in vivo

As is known to us, 1932 and SN12C were derived from primary human RCC, Caki-1 was a widespread model line of metastatic ccRCC, and ACHN was model line of metastatic papillary RCC (20). We transfected SN12C and 1932 cells with siRNA for PTPN3 (si1# and si2#) or negative control siRNA (Fig. 2A and B). PTPN3 suppression significantly increased RCC cell colony formation and proliferation (Fig. 2C) abilities of these two cell lines (Supplementary Fig. S2). Furthermore, PTPN3 was stably overexpressed in SN12C, Caki-1, and ACHN cell lines (Fig. 2D) and overexpressing PTPN3 effectively suppressed cell proliferation (Fig. 2E) and decreased colony formation ability (Fig. 2F). In addition, the overexpression of PTPN3 significantly inhibited tumor growth in nude mice (Fig. 2G and F). Taken together, these results suggested that PTPN3 can act as a tumor suppressor in the development of RCC.

Figure 2.

Suppression of PTPN3 promotes RCC cell growth in vitro and in vivo. A, Suppression of PTPN3 in RCC cells were determined by real-time quantitative PCR, normalized to β-actin. B, Suppression of PTPN3 in RCC cells were determined by immunoblotting analysis, GAPDH was used as a loading control. C, Suppression of PTPN3 promotes cells proliferation was determined by the MTS assay; ***, P < 0.001, results of Student t test for each paired samples on day 4 or day 5 from three independent experiments. D, Overexpression of PTPN3 in RCC cells were determined by immunoblotting analysis, GAPDH was used as a loading control. E, Overexpression of PTPN3 inhibits cells proliferation was determined by the MTS assay; ***, P < 0.001, results of Student t test for each paired samples on day 4 or day 5 from three independent experiments. F, Overexpression of PTPN3 decreased cell colony numbers; representative micrographs and quantification of crystal violet–stained cells from three independent experiments. Columns, average of three independent experiments; bars, SD. *, P < 0.05; **, P < 0.01; Student t test. G, Overexpression of PTPN3 in SN12C cells and the vector control cells were subcutaneously injected into nude mice. H, The terminal tumor weights are decreased compared with the control group (***, P < 0.001, result of Student t test).

Figure 2.

Suppression of PTPN3 promotes RCC cell growth in vitro and in vivo. A, Suppression of PTPN3 in RCC cells were determined by real-time quantitative PCR, normalized to β-actin. B, Suppression of PTPN3 in RCC cells were determined by immunoblotting analysis, GAPDH was used as a loading control. C, Suppression of PTPN3 promotes cells proliferation was determined by the MTS assay; ***, P < 0.001, results of Student t test for each paired samples on day 4 or day 5 from three independent experiments. D, Overexpression of PTPN3 in RCC cells were determined by immunoblotting analysis, GAPDH was used as a loading control. E, Overexpression of PTPN3 inhibits cells proliferation was determined by the MTS assay; ***, P < 0.001, results of Student t test for each paired samples on day 4 or day 5 from three independent experiments. F, Overexpression of PTPN3 decreased cell colony numbers; representative micrographs and quantification of crystal violet–stained cells from three independent experiments. Columns, average of three independent experiments; bars, SD. *, P < 0.05; **, P < 0.01; Student t test. G, Overexpression of PTPN3 in SN12C cells and the vector control cells were subcutaneously injected into nude mice. H, The terminal tumor weights are decreased compared with the control group (***, P < 0.001, result of Student t test).

Close modal

PTPN3 inhibits ccRCC cell migration, invasion, and metastasis

We performed wound-healing, transwell migration, and invasion assays to examine whether PTPN3 influences renal cell carcinoma cellular mobility. Findings from these assays revealed that the knockdown of PTPN3 with siPTPN3 increased the migration and invasion of renal cell carcinoma cells transfected (Fig. 3A, B, and E). Accordingly, the overexpression of PTPN3 significantly decreased the migration and invasion ACHN and SN12C cells (Fig. 3C, D, and F). Furthermore, the tail veins and lung metastatic in vivo experiments in nude mice showed that the number of lung metastatic tumors were significantly fewer in the SN12C-PTPN3 group than the control group (P < 0.05; Fig. 3G and H). Taken together, these results showed an important inhibitory role of PTPN3 in the migration, invasion, and metastasis of ccRCC cells.

Figure 3.

PTPN3 influences RCC cell migration, invasion and overexpressing PTPN3 impairs RCC metastasis in vivo. A, Suppression of PTPN3 in RCC cells could promote cell migration and cell invasion in SN12C and 1932 cells. B, Quantification of the effects of PTPN3 suppression on the migratory abilities and invasion ability in SN12C and 1932 cells as determined by transwell assays. All of the experiments were performed at least three times. Columns, average of three independent experiments; bars, SD. *, P < 0.05; **, P < 0.01, ***, P < 0.001; result of Student t test. C, Overexpression of PTPN3 in RCC cells could inhibit cell migration and cell invasion in ACHN and SN12C cells compared with vector control cells. D, Quantification of the effects of PTPN3 overexpression on the migratory and invasion abilities in ACHN and SN12C cells as determined by transwell assays. All experiments were performed at least three times. Columns, average of three independent experiments; bars, SD. *, P < 0.05; ***, P < 0.001; result of Student t test. E, Representative images of the effects of PTPN3 suppression on the migratory abilities of SN12C and 1932 cells as determined by wound-healing assays. All experiments were performed at least three times. F, Representative images of the effects of PTPN3 overexpression on the migratory abilities of ACHN and SN12C cells as determined by wound-healing assays. The experiments were performed at least three times. G, Overexpression of PTPN3 decreased in vivo metastatic rate of SN12C cells. Histologic image of lung metastasis in nude mice after tail vein injection of SN12C cells. Left, representative picture of lungs after picric acid staining; right, representative H&E staining of lungs; scale bar, 500 μm. H, The numbers of lung metastases (mean ± SD) were counted and summarized.

Figure 3.

PTPN3 influences RCC cell migration, invasion and overexpressing PTPN3 impairs RCC metastasis in vivo. A, Suppression of PTPN3 in RCC cells could promote cell migration and cell invasion in SN12C and 1932 cells. B, Quantification of the effects of PTPN3 suppression on the migratory abilities and invasion ability in SN12C and 1932 cells as determined by transwell assays. All of the experiments were performed at least three times. Columns, average of three independent experiments; bars, SD. *, P < 0.05; **, P < 0.01, ***, P < 0.001; result of Student t test. C, Overexpression of PTPN3 in RCC cells could inhibit cell migration and cell invasion in ACHN and SN12C cells compared with vector control cells. D, Quantification of the effects of PTPN3 overexpression on the migratory and invasion abilities in ACHN and SN12C cells as determined by transwell assays. All experiments were performed at least three times. Columns, average of three independent experiments; bars, SD. *, P < 0.05; ***, P < 0.001; result of Student t test. E, Representative images of the effects of PTPN3 suppression on the migratory abilities of SN12C and 1932 cells as determined by wound-healing assays. All experiments were performed at least three times. F, Representative images of the effects of PTPN3 overexpression on the migratory abilities of ACHN and SN12C cells as determined by wound-healing assays. The experiments were performed at least three times. G, Overexpression of PTPN3 decreased in vivo metastatic rate of SN12C cells. Histologic image of lung metastasis in nude mice after tail vein injection of SN12C cells. Left, representative picture of lungs after picric acid staining; right, representative H&E staining of lungs; scale bar, 500 μm. H, The numbers of lung metastases (mean ± SD) were counted and summarized.

Close modal

PTPN3 inhibits ccRCC cellular motility through suppression of PI3K/AKT signaling

Protein kinase B (also named AKT) is an important mediator of cell proliferation, apoptosis, autophagy, and metastasis. Here, we found that silencing PTPN3 in the 1932 and SN12C cells could enhance the expression of phospho-AKTThr308 and phospho-AKTSer473 (Fig. 4A). Conversely, the expression of phospho-AKTThr308 and phospho-AKTSer473 in 1932 and SN12C cells were decreased by overexpressing PTPN3 (Fig. 4A). Furthermore, we found that treatment with MK-2206 decreased phospho-AKTThr308 and phospho-AKTSer473 in SN12C-siPTPN3 and 1932-1-siPTPN3 cells (Fig. 4B). These results suggest that PTPN3 had some negative effects on the PI3K/AKT signaling pathway in RCC cells. PTPN3 seemed increased by the treatment of AKT inhibition for 1932 cell line, while this experimental phenomenon could not be repeated for SN12C cell line. In addition, we found that AKT inhibition by MK-2206 impaired the migration and invasion of SN12C-siPTPN3 and 1932-siPTPN3 cells (Fig. 5A and B), as well as the proliferation of SN12C-siPTPN3 and 1932-siAKT cells (Fig. 5C and D). These results illustrate that the knockdown of PTPN3 stimulated RCC cells' growth, migration, and invasion through the activation of the AKT pathway.

Figure 4.

PTPN3 inhibits RCC cellular growth, migration, and invasion through suppression of the PI3K/AKT signaling. A, Western blot analysis of AKT, P-AKTThr308, and P-AKTSer473 expression levels after overexpression of PTPN3 and silencing of PTPN3 from independent experiments. B, The expression of the PI3K/AKT signaling in PTPN3 silencing 1932 and SN12C cells after MK-2206 treatment was detected by immunoblotting from independent experiments. After treatment with 1 μmol/L of MK-2206 or PBS for 24 hours, cell lysates were harvested for Western blot detection. Relative gradation corrected by GAPDH is shown below each band.

Figure 4.

PTPN3 inhibits RCC cellular growth, migration, and invasion through suppression of the PI3K/AKT signaling. A, Western blot analysis of AKT, P-AKTThr308, and P-AKTSer473 expression levels after overexpression of PTPN3 and silencing of PTPN3 from independent experiments. B, The expression of the PI3K/AKT signaling in PTPN3 silencing 1932 and SN12C cells after MK-2206 treatment was detected by immunoblotting from independent experiments. After treatment with 1 μmol/L of MK-2206 or PBS for 24 hours, cell lysates were harvested for Western blot detection. Relative gradation corrected by GAPDH is shown below each band.

Close modal
Figure 5.

Inhibitors of P-AKTThr308 and P-AKTSer473 impair the migration, invasion, and growth of RCC cells induced by PTPN3 silencing. A and B, Cells were treated with 1 μmol/L MK-2206 or PBS for 24 hours, then MK-2206 was removed and cells were subjected to migration and invasion. Representative images and quantification of MK-2206 show the attenuating effects of PTPN3 silencing on the migratory abilities of SN12C and 1932 cells as determined by migration and invasion assays. Columns, average of three independent experiments; bars, SD. **, P < 0.01; ***, P < 0.001; Student t test. C and D, Cells were treated with 1 μmol/L MK-2206 or PBS for 24 hours, then MK-2206 was removed, and cells were subjected to and MTS assays. MK-2206 attenuate the accelerated effects of PTPN3 silencing on the growth abilities of SN12C and 1932 cells as determined by MTS assays. The difference is statistically significant in both the control and drug groups. Both experiments were repeated independently three times.

Figure 5.

Inhibitors of P-AKTThr308 and P-AKTSer473 impair the migration, invasion, and growth of RCC cells induced by PTPN3 silencing. A and B, Cells were treated with 1 μmol/L MK-2206 or PBS for 24 hours, then MK-2206 was removed and cells were subjected to migration and invasion. Representative images and quantification of MK-2206 show the attenuating effects of PTPN3 silencing on the migratory abilities of SN12C and 1932 cells as determined by migration and invasion assays. Columns, average of three independent experiments; bars, SD. **, P < 0.01; ***, P < 0.001; Student t test. C and D, Cells were treated with 1 μmol/L MK-2206 or PBS for 24 hours, then MK-2206 was removed, and cells were subjected to and MTS assays. MK-2206 attenuate the accelerated effects of PTPN3 silencing on the growth abilities of SN12C and 1932 cells as determined by MTS assays. The difference is statistically significant in both the control and drug groups. Both experiments were repeated independently three times.

Close modal

Although metastases cause the most death in patients with ccRCC, the detailed mechanisms of metastasis remain unclear (6, 7). In this study, we revealed that the expression of PTPN3 was downregulated in ccRCC cell lines and tissue samples. Also, the overexpression of PTPN3 inhibited renal cell carcinoma cell growth, migration, and invasion, and resulted smaller subcutaneous tumors and fewer pulmonary metastatic tumors in nude mice. Furthermore, lower expression of PTPN3 protein in ccRCC tissues could independently predict poor overall survival and progression-free survival in ccRCC. From this preliminary study, we found that PTPN3 could be a powerful conservator for antimetastasis in ccRCC.

The PI3K/AKT pathway has been shown to play important roles in a variety of biological processes, and the dysfunction of the AKT signaling was associated with diseases such as cancer, diabetes, and autoimmune (21, 22). PIP3 recruits AKT and PDK1 and other pleckstrin homology domain–containing proteins to the membrane of cells. Phosphorylating at Thr308 by PDK1 and at Ser473 by the mTOR complex 2 (mTORC2) increases the kinase activity of AKT (23). Directly and indirectly, AKT phosphorylates many downstream proteins to enhance cell survival and growth including the GSKs, p27KIP1, FoxO transcription factors, MDM2, and BAD (24). In this study, we discovered that PTPN3 attenuated AKT activity in RCC by downregulating both phospho-AKTThr308 and phospho-AKTSer473.

Because AKT was found to be a crucial regulator of cell proliferation, growth, and survival in different kinds of tumors (25), our findings may be applicable to not only RCC, but also other cancers that are driven by the increase of AKT signaling. Targeting AKT pathways through PTPN3 may be a major goal to develop therapeutics. Although our results suggested that PTPN3 could inhibit tumor progression in RCC, in other cancers, different mechanisms could be involved (26–28). What was more, Li and colleagues found that tyrosine-phosphorylated Src and DAAM1 could be dephosphorylated by the catalyzing of PTPN3 and thus suppresses lung cancer cell invasiveness (29). However, Yuan and colleagues found that PTPN3 profoundly potentiates TGFβ signaling independent of its phosphatase activity and thus acts as a tumor suppressor in liver cancer (30). In our study, we found that PTPN3 might inhibit the progression of RCC via the PI3K/AKT signaling pathway because PTPN3 could decrease the expression of both phospho-AKTThr308 and phospho-AKTSer473 in RCC cells. However, whether PTPN3 was able to dephosphorylate and directly act on AKT was still to be validated in further researches.

The mechanism between AKT and cancer has been reported in many other studies. For instance, some researchers found that m6A methylation could affect AKT activity in adult T-cell leukemia (AML; ref. 31) and endometrial cancer (26); CITED2 could activate the nucleolin–AKT pathway and thus promoted prostate cancer metastasis (32); SMAD and AKT pathways interacted to confer prooncogenic responses to TGFβ and drove breast cancer metastasis (33). Some studies further reported the potential mechanisms between AKT pathways and renal cell cancer. AKT was found to positively regulate rasfonin-enhanced autophagy and caspase-dependent apoptosis primarily through affecting the glycolytic pathway in renal cancer cells (34). However, the upstream potential mechanism of AKT was rarely reported. Some researchers found that downregulating PIK3R1 in RCC promoted the migration, propagation, stem-like phenotype, and EMT in RCC cells via the AKT/GSK3β/CTNNB1 pathway and may lead to the progression of RCC (35). In this study, we found that PTPN3 was actually an inhibitor for AKT pathway in ccRCC upon controlling cellular proliferation and motility.

Conclusions

PTPN3 inhibits RCC cells' growth, migration, and invasion via downregulation of the AKT pathway.

No potential conflicts of interest were disclosed.

Conception and design: X.-S. Peng, C.-N. Qian

Development of methodology: X.-S. Peng, J.-P. Yang, Y.-Y. Qiang, M.-D. Wang, C.-N. Qian

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): X.-S. Peng, J.-P. Yang, Y.-Y. Qiang, L.-S. Zheng, L.-X. Peng, Y. Mei, D.-F. Meng, Z.-J. Liu, F.-J. Zhou, C.-N. Qian

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): X.-S. Peng, J.-P. Yang, Y.-Y. Qiang, R. Sun, L.-S. Zheng, L.-X. Peng, Y.-H. Lang, Y. Mei, D.-F. Meng, Z.-J. Liu, B.-J. Huang, C.-N. Qian

Writing, review, and/or revision of the manuscript: X.-S. Peng, J.-P. Yang, Y. Mei, D.-F. Meng, C.-N. Qian

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): X.-S. Peng, J.-P. Yang, Y.-Y. Qiang, R. Sun, Y. Cao, L.-S. Zheng, L.-X. Peng, Y.-H. Lang, Y. Mei, D.-F. Meng, C.-N. Qian

Study supervision: X.-S. Peng, B.-J. Huang, C.-N. Qian

Other (contributed reagents/materials/analysis tools): C.-Z. Li

Some of the results here are based upon data generated by the TCGA Research Network. This work was supported by grants from the National Natural Science Foundation of China (nos. 81872384, 81672872, and 81472386, to C.-N. Qian, nos. 81972785, 81773162, and 81572901, to B.-J. Huang), the Science and Technology Planning Project of Guangdong Province, China (no. 2013B021800065, to R. Sun), the Provincial Natural Science Foundation of Guangdong, China (nos. 2016A030311011, to C.-N. Qian; no. 2017A030313866, to B.-J. Huang), and a research program from Sun Yat-sen University (no. 84000-18843409 to C.-N. Qian).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Siegel
R
,
Naishadham
D
,
Jemal
A
. 
Cancer statistics, 2013
.
CA Cancer J Clin
2013
;
63
:
11
30
.
2.
Siegel
RL
,
Miller
KD
,
Jemal
A
. 
Cancer statistics, 2017
.
CA Cancer J Clin
2017
;
67
:
7
30
.
3.
Shuch
B
,
Amin
A
,
Armstrong
AJ
,
Eble
JN
,
Ficarra
V
,
Lopez-Beltran
A
, et al
Understanding pathologic variants of renal cell carcinoma: distilling therapeutic opportunities from biologic complexity
.
Eur Urol
2015
;
67
:
85
97
.
4.
Comperat
E
,
Camparo
P
. 
Histological classification of malignant renal tumours at a time of major diagnostic and therapeutic changes
.
Diagn Interv Imaging
2012
;
93
:
221
31
.
5.
Stadler
WM
. 
Targeted agents for the treatment of advanced renal cell carcinoma
.
Cancer
2005
;
104
:
2323
33
.
6.
Huang
Q
,
Sun
Y
,
Ma
X
,
Gao
Y
,
Li
X
,
Niu
Y
, et al
Androgen receptor increases hematogenous metastasis yet decreases lymphatic metastasis of renal cell carcinoma
.
Nat Commun
2017
;
8
:
918
.
7.
Lin
YW
,
Lee
LM
,
Lee
WJ
,
Chu
CY
,
Tan
P
,
Yang
YC
, et al
Melatonin inhibits MMP-9 transactivation and renal cell carcinoma metastasis by suppressing Akt-MAPKs pathway and NF-kappaB DNA-binding activity
.
J Pineal Res
2016
;
60
:
277
90
.
8.
Roy
B
,
Pattanaik
AK
,
Das
J
,
Bhutia
SK
,
Behera
B
,
Singh
P
, et al
Role of PI3K/Akt/mTOR and MEK/ERK pathway in Concanavalin A induced autophagy in HeLa cells
.
Chem Biol Interact
2014
;
210
:
96
102
.
9.
Luo
C
,
Cen
S
,
Ding
G
,
Wu
W
. 
Mucinous colorectal adenocarcinoma: clinical pathology and treatment options
.
Cancer Commun
2019
;
39
:
13
.
10.
Polivka
J
 Jr
,
Janku
F
. 
Molecular targets for cancer therapy in the PI3K/AKT/mTOR pathway
.
Pharmacol Ther
2014
;
142
:
164
75
.
11.
Guo
H
,
German
P
,
Bai
S
,
Barnes
S
,
Guo
W
,
Qi
X
, et al
The PI3K/AKT pathway and renal cell carcinoma
.
J Genet Genomics
2015
;
42
:
343
53
.
12.
Wu
F
,
Wu
S
,
Gou
X
. 
Identification of biomarkers and potential molecular mechanisms of clear cell renal cell carcinoma
.
Neoplasma
2018
;
65
:
242
52
.
13.
Li
MY
,
Lai
PL
,
Chou
YT
,
Chi
AP
,
Mi
YZ
,
Khoo
KH
, et al
Protein tyrosine phosphatase PTPN3 inhibits lung cancer cell proliferation and migration by promoting EGFR endocytic degradation
.
Oncogene
2015
;
34
:
3791
803
.
14.
Gao
Q
,
Zhao
YJ
,
Wang
XY
,
Guo
WJ
,
Gao
S
,
Wei
L
, et al
Activating mutations in PTPN3 promote cholangiocarcinoma cell proliferation and migration and are associated with tumor recurrence in patients
.
Gastroenterology
2014
;
146
:
1397
407
.
15.
Zhang
SH
,
Liu
J
,
Kobayashi
R
,
Tonks
NK
. 
Identification of the cell cycle regulator VCP (p97/CDC48) as a substrate of the band 4.1-related protein-tyrosine phosphatase PTPH1
.
J Biol Chem
1999
;
274
:
17806
12
.
16.
Hou
SW
,
Zhi
HY
,
Pohl
N
,
Loesch
M
,
Qi
XM
,
Li
RS
, et al
PTPH1 dephosphorylates and cooperates with p38gamma MAPK to increase ras oncogenesis through PDZ-mediated interaction
.
Cancer Res
2010
;
70
:
2901
10
.
17.
Zhang
RL
,
Yang
JP
,
Peng
LX
,
Zheng
LS
,
Xie
P
,
Wang
MY
, et al
RNA-binding protein QKI-5 inhibits the proliferation of clear cell renal cell carcinoma via post-transcriptional stabilization of RASA1 mRNA
.
Cell Cycle
2016
;
15
:
3094
104
.
18.
Qian
CN
. 
The rationale for preventing cancer cachexia: targeting excessive fatty acid oxidation
.
Chin J Cancer
2016
;
35
:
67
.
19.
Qiang
YY
,
Li
CZ
,
Sun
R
,
Zheng
LS
,
Peng
LX
,
Yang
JP
, et al
Along with its favorable prognostic role, CLCA2 inhibits growth and metastasis of nasopharyngeal carcinoma cells via inhibition of FAK/ERK signaling
.
J Exp Clin Cancer Res
2018
;
37
:
34
.
20.
Brodaczewska
KK
,
Szczylik
C
,
Fiedorowicz
M
,
Porta
C
,
Czarnecka
AM
. 
Choosing the right cell line for renal cell cancer research
.
Mol Cancer
2016
;
15
:
83
.
21.
Manning
BD
,
Toker
A
. 
AKT/PKB signaling: navigating the network
.
Cell
2017
;
169
:
381
405
.
22.
Vivanco
I
,
Sawyers
CL
. 
The phosphatidylinositol 3-Kinase AKT pathway in human cancer
.
Nat Rev Cancer
2002
;
2
:
489
501
.
23.
Zhao
P
,
Chen
H
,
Wen
D
,
Mou
S
,
Zhang
F
,
Zheng
S
. 
Personalized treatment based on mini patient-derived xenografts and WES/RNA sequencing in a patient with metastatic duodenal adenocarcinoma
.
Cancer Commun
2018
;
38
:
54
.
24.
Manning
BD
,
Cantley
LC
. 
AKT/PKB signaling: navigating downstream
.
Cell
2007
;
129
:
1261
74
.
25.
Thorpe
LM
,
Yuzugullu
H
,
Zhao
JJ
. 
PI3K in cancer: divergent roles of isoforms, modes of activation and therapeutic targeting
.
Nat Rev Cancer
2015
;
15
:
7
24
.
26.
Liu
J
,
Eckert
MA
,
Harada
BT
,
Liu
SM
,
Lu
Z
,
Yu
K
, et al
m(6)A mRNA methylation regulates AKT activity to promote the proliferation and tumorigenicity of endometrial cancer
.
Nat Cell Biol
2018
;
20
:
1074
83
.
27.
Lien
EC
,
Lyssiotis
CA
,
Juvekar
A
,
Hu
H
,
Asara
JM
,
Cantley
LC
, et al
Glutathione biosynthesis is a metabolic vulnerability in PI(3)K/Akt-driven breast cancer
.
Nat Cell Biol
2016
;
18
:
572
8
.
28.
Tenbaum
SP
,
Ordonez-Moran
P
,
Puig
I
,
Chicote
I
,
Arques
O
,
Landolfi
S
, et al
beta-catenin confers resistance to PI3K and AKT inhibitors and subverts FOXO3a to promote metastasis in colon cancer
.
Nat Med
2012
;
18
:
892
901
.
29.
Li
MY
,
Peng
WH
,
Wu
CH
,
Chang
YM
,
Lin
YL
,
Chang
GD
, et al
PTPN3 suppresses lung cancer cell invasiveness by counteracting Src-mediated DAAM1 activation and actin polymerization
.
Oncogene
2019
;
38
:
7002
16
.
30.
Yuan
B
,
Liu
J
,
Cao
J
,
Yu
Y
,
Zhang
H
,
Wang
F
, et al
PTPN3 acts as a tumor suppressor and boosts TGF-beta signaling independent of its phosphatase activity
.
EMBO J
2019
;
38
:
e99945
.
31.
Vu
LP
,
Pickering
BF
,
Cheng
Y
,
Zaccara
S
,
Nguyen
D
,
Minuesa
G
, et al
The N(6)-methyladenosine (m(6)A)-forming enzyme METTL3 controls myeloid differentiation of normal hematopoietic and leukemia cells
.
Nat Med
2017
;
23
:
1369
76
.
32.
Shin
SH
,
Lee
GY
,
Lee
M
,
Kang
J
,
Shin
HW
,
Chun
YS
, et al
Aberrant expression of CITED2 promotes prostate cancer metastasis by activating the nucleolin-AKT pathway
.
Nat Commun
2018
;
9
:
4113
.
33.
Xie
F
,
Jin
K
,
Shao
L
,
Fan
Y
,
Tu
Y
,
Li
Y
, et al
FAF1 phosphorylation by AKT accumulates TGF-beta type II receptor and drives breast cancer metastasis
.
Nat Commun
2017
;
8
:
15021
.
34.
Lu
Q
,
Yan
S
,
Sun
H
,
Wang
W
,
Li
Y
,
Yang
X
, et al
Akt inhibition attenuates rasfonin-induced autophagy and apoptosis through the glycolytic pathway in renal cancer cells
.
Cell Death Dis
2015
;
6
:
e2005
.
35.
Lin
Y
,
Yang
Z
,
Xu
A
,
Dong
P
,
Huang
Y
,
Liu
H
, et al
PIK3R1 negatively regulates the epithelial-mesenchymal transition and stem-like phenotype of renal cancer cells through the AKT/GSK3beta/CTNNB1 signaling pathway
.
Sci Rep
2015
;
5
:
8997
.