
Liver kinase B1 (LKB1), also known as the serine/threonine kinase 11 (STK11) gene, is a widely expressed tumor suppressor gene that is commonly associated with Peutz-Jeghers syndrome (PJS) (Avilés-Salas
AMP-activated protein kinase (AMPK) is a critical enzyme involved in the regulation of various cellular processes including cell growth, homeostasis, and metabolism, as well as autophagy control (Rho
Malic enzymes (MEs) are a family of enzymes that are present in a wide range of species, from bacteria to humans, and are involved in various biological processes, such as metabolism, photosynthesis, and fatty acid biosynthesis (Dmitriev, 2001). In mammals, three isoforms of ME have been identified: ME1, ME2, and ME3 (Chang and Tong, 2003; Hasan
Several studies have reported on the roles of malic enzyme isoforms in tumorigenesis. For example, ME1 suppression has been shown to reduce metastasis of hepatocellular carcinoma by repressing the epithelial-mesenchymal transition in reactive oxygen species-induced signaling cascades (Wen
In this study, we aimed to investigate the functional importance and molecular mechanism of ME3 in LKB1-induced apoptotic cell death in an
Human OVCAR-3 ovarian carcinoma cells and human embryonic kidney cells 293 (HEK293T) were obtained from the American Type Culture Collection (ATCC, Manassas, VA, USA), and were maintained in Dulbecco’s Modified Eagle Medium (DMEM; Life Technologies, Gaithersburg, MD, USA) supplemented with 10% heat-inactivated fetal bovine serum (FBS) and penicillin/streptomycin. Wortmannin and LY294002 were purchased from Sigma (St. Louis, MO, USA). The primary antibodies used in this study were anti-LKB1, anti-ME3, anti-NF-κB, anti-Bcl-2, anti-p53, anti-PI3K, anti-phospho-PI3K, anti-Akt, anti-phospho-Akt (Ser473 and Thr308), anti-PDK-1, anti-phospho-PDK1 (Ser241), anti-mTOR, anti-phospho-mTOR (Ser2448), anti-TSC2, and anti-phospho-TSC2 (Ser1462), anti-p70S6K, anti-phospho-p70S6K (Thr421), anti-GSK-3β, anti-phospho-GSK-3β (Ser9), anti-4E-BP1, anti-phospho-4E-BP1 (Thr70) (Cell Signaling, Beverly, MA), anti-Flag, anti-cyclin D1, anti-CDK4, anti-p21 (Santa Cruz Biotechnology, Santa Cruz, CA, USA), and β-actin (Sigma).
The cDNA encoding the full-length and truncation mutants (Met1–Ala200, Leu201–Asp350, and Glu351–Gln433) of LKB1 were subcloned in the pGilda/LexA yeast shuttle vector at the
The co-immunoprecipitation (co-IP) assays were performed as previously described (Kang
Cells were grown in chamber slides at a density of 4.5×104 cells per well, transfected, incubated with fluorescein isothiocyanate (FITC)-labeled annexin V and propidium iodide for 15 min according to the manufacturer’s instructions (BD PharMingen, Mississauga, ON, USA), and analyzed with a FACS Vantage (BD FACS Calibur Flow Cytometer, BD, Andover, MA, USA). Cell viability was estimated with the MTT assay. Cells were maintained at a density of 4.5×103 cells per well in 96-well plates. Fresh medium including 10% FBS and 20 µL of 3-(4,5-dimethylthiazol-2-yl)-2.5-diphenyl-2H-tetrazolium bromide (MTT) solution (5 µg/mL; Sigma) was added to each well 3 d following transfection, and each well was incubated for an additional 4 h at 37°C. Following centrifugation at 500×
Caspase-3 activity was measured as previously described (Rho
Luciferase activity
Data values were expressed as the mean ± standard deviation (SD) and were estimated by Student’s
The study utilized a yeast two-hybrid (Y2H) protein-protein interaction (PPI) system and co-immunoprecipitation (co-IP) to investigate the molecular mechanism underlying the tumor suppressive effect of LKB1 in ovarian tumorigenesis.
ME3 was identified as a putative binding partner of LKB1 in the human ovary cDNA library through Y2H screening (accession number: BC022472.2; Fig. 1A).
The relative binding of LKB1 to ME3 was confirmed by the activity of β-galactosidase in yeast cells co-expressing LKB1 and ME3. As shown in Fig. 1B, β-galactosidase was fully active with LKB1 and ME3 (82.08 ± 1.22), but failed in the absence of LKB1 (1.84 ± 0.51).
Co-IP experiments were performed to further confirm the direct interaction between LKB1 and ME3 in OVCAR-3 cells. LKB1 (pcDNA3.1/LKB1) and ME3 (pcDNA3.1/Flag-ME3) or ME3 (pcDNA3.1/Flag-ME3) and the expression vector (pcDNA3.1) only were co-transfected into OVCAR-3 cells, and immunoprecipitation of the lysates was performed with anti-Flag antibody. The precipitated proteins were immunoblotted with the anti-LKB1 or anti-ME3 primary antibodies, and the results indicated that Flag-ME3 co-immunoprecipitated with LKB1 (lane 2 in upper panel) (Fig. 1C).
The
In this study, we aimed to investigate the effect of LKB1 and ME3 overexpression on ovarian carcinoma cell proliferation and apoptosis. To assess cell viability, we overexpressed LKB1, ME3, or both in OVCAR-3 cells and performed an MTT assay after 48 hours. Our results showed that overexpression of LKB1 or ME3 alone reduced cell viability to approximately 45% and 42%, respectively, compared to control cells. Co-transfection of both plasmids resulted in a further decrease in cell viability (Fig. 2A, left panel). We also investigated the effects of different ratios of LKB1 (0.5 µg) and ME3 (0-0.5 µg) on cell apoptosis, and our data showed that co-transfection at ratios greater than 1:0 and 1:1 was associated with increased rates of apoptosis (Fig. 2A, right panel).
To further investigate the mutual regulation of LKB1 and ME3, we evaluated their expression levels after transient transfection of one another. Our results demonstrated that the expression of LKB1 increased with higher amounts of ME3, and a similar trend was observed for the expression of ME3 after transient transfection of LKB1 (Fig. 2B, left panel). We confirmed that the decrease in cell viability was due to apoptosis using fluorescence-activated cell sorting, and our data showed that overexpression of LKB1 or ME3 increased the apoptotic cell population compared to control cells (Fig. 2C). Co-transfection with both plasmids was more effective than transfection of either plasmid alone, indicating that LKB1 and ME3 additively repressed cell proliferation.
Finally, we investigated the activation of caspase-3 as a potential mechanism for the mutual regulation of LKB1 and ME3. Using a caspase-3 activity assay, we found that cells transfected with LKB1, ME3, or both exhibited a significant increase in caspase-3 activation. Co-transfection with both plasmids resulted in a greater increase in caspase-3 activity compared to transfection of either plasmid alone (Fig. 2D). These findings suggest that ME3 suppresses tumor cell proliferation by augmenting LKB1-mediated apoptosis through direct interaction.
To identify the specific regions of LKB1 and ME3 that are involved in their interaction, three LKB1 truncation mutants and three ME3 truncation mutants were constructed and transformed into yeast cells along with full-length human ME3 or LKB1, respectively (Fig. 3A). Yeast cells containing the Leu201-Asp350 truncation mutant of LKB1 grew on selective media, indicating that this region is capable of binding to ME3. However, yeast cells transformed with the Met1-Ala200 or Glu351-Gln433 truncation mutants failed to grow, indicating that these regions are not sufficient for binding to ME3 (Fig. 3A, right panel). This result was confirmed by a β-galactosidase assay, which measures the activity of the reporter gene that is activated when LKB1 and ME3 bind (Fig. 3A).
To identify the ME3 region that binds to LKB1, three ME3 truncation mutants were constructed and transformed into yeast cells along with full-length human LKB1 (Fig. 3B, left panel, full). The β-galactosidase assay revealed that the region of ME3 that binds to LKB1 is within Met1-Tyr200 (Fig. 3B, right panel). This result was confirmed by co-transforming the Leu201-Asp350 mutant of LKB1 with one of the three ME3 mutants. Yeast cells containing the Leu201-Asp350 mutant of LKB1 with the Met1-Tyr200 ME3 mutant grew on selective media (Fig. 3C) and showed positive β-galactosidase activity, indicating that these two regions can bind to each other. However, yeast cells transformed with the Met1-Ala200 or Glu351-Gln433 ME3 mutants failed to grow (Fig. 3C) and showed no β-galactosidase activity, confirming that these regions are not sufficient for binding to LKB1.
These experiments provide evidence for the specific regions of LKB1 and ME3 that are involved in their interaction.
In order to better understand the biological effects of LKB1 and ME3 in inducing apoptosis, we conducted an experiment to investigate the expression of proteins involved in cell cycle progression in OVCAR-3 cells. We estimated the transcriptional activity of these proteins using a luciferase activity assay. After transfecting OVCAR-3 cells with LKB1 or ME3, we measured the expression of cyclin D1 and cyclin-dependent kinase 4 (CDK4). The expression levels of cyclin D1 and CDK4 were decreased upon LKB1 or ME3 transfection in the cells. However, upon transfection of LKB1 or ME3 small interfering RNA (siRNA) into the cells, the expression levels of cyclin D1 and CDK4 were restored, as shown in Fig. 4A and 4B (Fig. 4A, 4B). Additionally, the expression of p53 and p21, both involved in cell cycle regulation and apoptosis, was increased in cells transfected with LKB1 or ME3, while the expression of Bcl-2 and NF-κB, two anti-apoptotic proteins, was reduced (Fig. 4A, 4B).
These findings were confirmed through a luciferase reporter gene activity assay, which demonstrated that LKB1 and/or ME3 overexpression significantly increased the transcription of p53 and p21, while decreasing the transcription of Bcl-2 and NF-κB (Fig. 4C, 4D).
These results suggest that LKB1 and ME3 overexpression may induce apoptosis in OVCAR-3 cells by regulating the transcription of key proteins involved in cell cycle progression and apoptosis.
The experiment aimed to investigate the impact of LKB1 and ME3 on the PI3K/mTOR/4E-BP1 signaling pathways in OVCAR-3 tumor cells, which regulate cell proliferation, protein synthesis, and angiogenesis (Guertin and Sabatini, 2005; Ma and Blenis, 2009; Jeon, 2016; Saxton and Sabatini, 2017; Sun and Song, 2021).
We transfected OVCAR-3 cells with LKB1, ME3, or both, and observed a significant decrease in the phosphorylation of PI3K, which was similar to the effect of PI3K inhibitors wortmannin and LY294002 (Fig. 5A). The inhibition of PI3K activity led to a reduction in the phosphorylation and overall levels of Akt, a downstream target of PI3K (Fig. 5B). Additionally, we investigated the involvement of upstream and downstream components of Akt/mTOR, including PDK1, TSC-2, p70S6K, GSK-3β, and 4E-BP1. The results showed that the phosphorylation of mTOR/4E-BP1 was suppressed by the expression of LKB1 and ME3 (Fig. 5B).
These results suggest that LKB1 and ME3 repressed tumor cell growth via PI3K/mTOR/4E-BP1-dependent signaling pathways in ovarian tumorigenesis.
This study investigated the role and mechanisms of LKB1 and ME3 in regulating apoptosis and cell cycle progression in ovarian tumorigenesis. The findings suggest that LKB1 and ME3 play critical roles in these processes and dysregulation of these proteins may contribute to tumorigenesis.
The study identified a direct interaction between LKB1 and ME3 and found that LKB1 and ME3 target the PI3K/mTOR/4E-BP1 signaling pathway to trigger apoptosis in ovarian tumorigenesis, highlighting the potential therapeutic value of targeting this pathway in the treatment of ovarian cancer.
The study also provides insight into the role of LKB1 in gynecological cancers. Loss of LKB1 expression has been identified as an early event in high-grade serous ovarian tumor development, and LKB1 mutations have been found in cervical tumors, suggesting that dysregulation of LKB1 may be a contributing factor in the development of various types of gynecological cancers (Wingo
Our study is consistent with recent research indicating that miRNAs may play a role in the development of ovarian tumors. In particular, mature miR-17 appears to target the LKB1-p53-p21/WAF1 pathway, which can result in epigenetic alterations that promote tumorigenesis (Liu
LKB1 interacts with PTEN to inhibit cell proliferation and survival and represses anti-apoptotic factors to inhibit cell survival (Mehenni
The identification of LKB1 as a tumor suppressor gene adds to the growing body of literature on the genetic aberrations underlying ovarian tumor development (Tanwar
Furthermore, our study demonstrated that LKB1 and ME3 are involved in suppressing PI3K/Akt signaling pathway components (Fig. 5), which are crucial regulators of malignant tumors (Ishigami
In summary, our study provides important new insights into the role of LKB1 and ME3 in tumor suppression and their potential as therapeutic targets in ovarian tumorigenesis. Future studies can focus on further elucidating the molecular mechanisms underlying LKB1-mediated signaling pathways through its interaction with ME3.
This work was supported by a grant from the National Cancer Center, Korea (NCC-2112500-1 and 2210450-1) and the Basic Science Research Program and the BK21 FOUR program through the NRF (NRF-2018R1A5A2023127, and NRF-2020R1A2C3004973).
The authors have declared that no competing interest exists.
![]() |
![]() |