Antibody to human α-fetoprotein inhibits cell growth of human hepatocellular carcinoma cells by resuscitating the PTEN molecule: in vitro experiments

  • Authors:
    • Kiyoshi Ohkawa
    • Tadashi Asakura
    • Yutaka Tsukada
    • Tomokazu Matsuura
  • View Affiliations

  • Published online on: May 3, 2017     https://doi.org/10.3892/ijo.2017.3982
  • Pages: 2180-2190
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

It has been proposed that α-fetoprotein (AFP) is a new member of the intracellular signaling molecule family of the phosphoinositide 3-kinase (PI3K)/AKT signaling pathway via interaction with the phosphatase and tensin homolog (PTEN). In this study, the effects of anti-human AFP antibody on the functions of PTEN were examined using an AFP-producing human hepatoma cell line. The antibody caused significant inhibition of cell growth, compared to a normal IgG control, with the accumulation of intracellular immune complexes followed by significant reduction of cytosolic functional AFP. Decrease in the amount of AKT phosphorylated on serine (S) 473 indicated that PI3K/AKT signaling was suppressed in the cells. S380-phosphorylated PTEN increased markedly by the second day after antibody treatment, with slight but significant increase in the PTEN protein level. Since phosphorylation at S380 is critical for PTEN stability, the increase in S380-phosphorylated PTEN indicated maintenance of the number of PTEN molecules and the related potential to control PI3K/AKT signaling. p53 protein (P53) significantly, but slightly increased during antibody treatment, because PTEN expression increased the stability and function of P53 via both molecular interactions. P53 phosphorylated at S20 or at S392 dramatically increased, suggesting an increase in the stability, accumulation and activation of P53. Glucose transporter 1 (GLUT1) increased immediately after antibody treatment, pointing to a deficiency of glucose in the cells. Immunofluorescence cytology revealed that antibody-treatment re-distributed GLUT1 molecules throughout the cytoplasm with a reduction of their patchy localization on the cell surface. This suggested that translocation of GLUT1 depends on the PI3K/AKT pathway, in particular on PTEN expression. Antibody therapy targeted at AFP-producing tumor cells showed an inhibitory effect on the PI3K/AKT pathway via the liberation, restoration and functional stabilization of PTEN. PTEN simultaneously induced both P53 activation and intracellular translocation of GLUT1, since these are closely associated with PTEN.

Introduction

More than forty years ago, Mizejewski et al (14), Tsukada et al (5) and others (68) demonstrated in mouse, rat, and human cultured cell systems in vitro and mouse and rat experiments in vivo, that treatment with anti-α-fetoprotein (AFP) serum or its purified antibody had a growth inhibitory effect on AFP-producing tumor cells, such as hepatocellular carcinoma and yolk sac tumor. Ohkawa et al focused on the finding that the administered anti-AFP antibody led to remarkable inhibition of sugar uptake by the neoplastic cells (8), suggesting that one of the main mechanisms of the antitumor effect of the antibody was impeding the transportation of nourishment across the cell membrane. However, most of the mechanisms of action of the antibody-derived antitumor effect remained obscure, because the main research focus shifted to the application of the specific antibodies as carriers of anticancer drugs or toxins in drug delivery systems (917). In addition, antibody-mediated immuno-radioimaging experiments aimed at tumor diagnosis and/or therapeutic modalities using the antibody directed against tumor markers coupled to radioisotopes was promulgated (7,1823) and the mechanism of action underlying the antitumor effect of anti-AFP antibody was overlooked.

The possibility that AFP is a cell growth factor has been discussed recently (2431), and its effect in promoting cell proliferation has been reported in a number of cell types. There have also been many reports that the cell growth-promoting effects of intracellular AFP are based on the close relationship with the phosphoinositide 3-kinase (PI3K)/AKT (known as protein kinase B) signaling pathway (3036). Interactions of intracytoplasmic membrane-bound AFP with several kinds of intracellular functional and essential proteins have been also reviewed (31). These findings suggest that AFP must be a new member of the intracellular molecule family of the PI3K/AKT signaling pathway. It has been reported that AFP in the cell strongly interacts with the phosphatase and tensin homolog deleted on chromosome 10 (PTEN), which is a tumor suppressor molecule that strictly regulates PI3K/AKT signaling (3036), and the interaction leads to a functional decline of PTEN. As a result, the PI3K/AKT signaling pathway might be activated as a tumor survival system. Conversely, experiments on the suppression of AFP protein expression using the RNA interference technology or microRNA network demonstrated that a severe decrease in AFP within the cell liberated intracellular PTEN molecules from the cytoplasmic AFP molecules, and led to restoration of the phosphatase-dependent and/or -independent activities with a regulatory function to the PI3K/AKT signaling pathway (3234). Innovative research with a breakthrough result was recently reported and entailed adding a novel antibody with single-chain variable fragment (scFv) to AFP, thereby blocking AFP and inhibiting hepatoma cell growth via the PI3K/AKT/PTEN signaling pathway (37). However, possible inhibitory mechanism of action of anti-AFP antibody against tumor cell growth did remain obscure in the report.

In the present experiments, to determine the antitumor efficacy of the antibody, the effect of antibody against human AFP on the AFP denaturation and/or degradation caused by antigen-antibody immune reactions, as well as on the biological behavior of PTEN molecules and on the PI3K/AKT signaling pathway were investigated using the AFP-producing human hepatocellular carcinoma cell line, FLC7. FLC7 cells are able to grow in chemically defined synthetic medium without any addition of peptide growth factors or animal proteins. Use of this cell line in the study is convenient and essential in order to determine the effects on cells of an antibody under a culture environment in which the trace effects of known and unknown growth-related materials are absent. This study was conducted to elucidate part of the mechanism underlying the antitumor effect of anti-AFP antibody.

Materials and methods

Cell culture

The FLC7 cell line (previously known as JHH-7), initially established from Japanese patient with hepatocellular carcinoma (38), has been adapted to grow under serum-free condition with differentiated liver functions. In brief, FLC7 cells were initially cultivated in ASF104N medium (Ajinomoto Healthy Supply Co., Inc., Tokyo, Japan), which is a chemically defined serum-free medium containing only recombinant human transferrin and insulin as growth factors. Then the media were replaced with chemically defined completely synthetic media (IS-RPMI, RPMI-1640, 5 mM HEPES, 3×10−8 M Na2SeO3, 1×10−8 M NH4VO3, 3×10−9 M (NH4)6Mo7O24-4H2O, 1×10−7 M FeSO4-7H2O, 3×10−9 M linoleic acid, 3×10−9 M oleic acid and 1.55×10−2 M NaHCO3) without any addition of peptides or animal materials (39) and then the cells were continuously cultivated under the conventional culture conditions in a CO2 incubator. The spent culture media of IS-RPMI (CM) of FLC7 cells culturing were stocked as supplement for sub-culture. FLC7 cells were routinely plated and sub-cultured with 50% CM-containing IS-RPMI and the media were replaced with fresh IS-RPMI after 2 days of culture.

Cytotoxicity

FLC7 cells (5×104) were initially cultured in 24-well culture plates (Greiner Bio-One, Tokyo, Japan) with 1 ml of IS-RPMI containing 0.5% of ASF104N, instead of CM in which AFP and other growth factor related various materials with known and unknown types were probably contained. After 12 h of culture, the cells, attached to the plates, were twice washed with 2 ml of warmed IS-RPMI and then media were replaced completely with 1 ml of fresh warmed IS-RPMI, which contained various concentrations of rabbit immunoglobulin (Ig) G of anti-human AFP antibody or rabbit pre-immune (normal) IgG. The cells were cultured continuously for 96 h with each IgG. After the incubation, viable cells were determined with the colorimetric assay using WST-8 (Cell Counting kit-8, Dojindo Lab., Kumamoto, Japan) and the results were expressed by the following equation: survival rate (%) = 100 × (absorbance at 450 nm of the each IgG-exposed cells) / (absorbance at 450 nm of the non-treated control cells). Additionally another type of cytotoxic assay was also carried out using each rabbit anti-human AFP antibody IgG or rabbit normal IgG at the concentration of the 50% growth inhibitory concentration value (IC50), determined with the above results. In brief, to determine the IC50 in this assay, the cell viability assessed by WST-8 was expressed as the fraction of surviving cells in anti-human AFP antibody IgG-treated cells relative to those in the normal IgG-treated controls, instead of IgG non-treated cells and the IC50 was calculated. At the concentration of IC50, the cells were cultured for indicated periods of time in the IS-RPMI. Results were calculated as mean ± SD of triplicate determinations of two independent experiments.

Cell lysate

Either rabbit anti-human AFP antibody IgG- or rabbit normal IgG-treated cultured cells with various time intervals and concentrations were rapidly washed twice with 2 ml of ice-cold Dulbecco's PBS (−) in wells, then added 200 μl of ice-cold cell lysis buffer (50 mM Tris-HCl, pH 7.5, 0.15 M NaCl, 1 mM EDTA, 0.1% sodium dodecyl sulfate (SDS), 1 mM phenylmethyl sulphonyl fluoride, 1% Triton X-100, 1% deoxycholate, 1 mM Na3VO4, 1 mM NaF, 0.02% NaN3 and protease inhibitor cocktail and phosphatase inhibitor cocktail 2 and 3 (Sigma-Aldrich Japan, Tokyo, Japan). After incubation for 20 min on ice, collected lysates were centrifuged at 23,000 × g for 20 min at 4°C. The supernatants were kept frozen at −80°C until use. The protein concentration was measured by the DC protein assay kit (Bio-Rad, Richmond, CA, USA). Bovine serum albumin (BSA) was used as the standard.

AFP-enzyme-linked immunosorbent assay (ELISA)

AFP concentrations in the cultured media and the cell lysates were determined using the sandwich ELISA system (AFP ELISA kit; Abnova, Taipei, Taiwan), according to the manufacturer's instructions. Intracellular AFP concentrations were expressed as (ng)/(mg of cell lysate). Data were expressed as mean ± SD of two independent experiments.

SDS polyacrylamide gel electrophoresis/immunoblotting (SDS-PAGE/WB)

After SDS-PAGE/WB, the polyvinylidene fluoride filters (Abcam, Tokyo, Japan) were blocked and then incubated with the corresponding primary antibodies, followed by horseradish peroxidase (HRP)-labeled second antibodies directed against rabbit, mouse, or goat IgG (Abcam), as described previously (40). Enhanced chemiluminescence (ECL) signals (ImmunoStar, Wako Pure Chemical Industries, Ltd., Tokyo, Japan) were detected by a cooled CCD camera system (ATTO light-capture II type AE-6981; ATTO Co., Tokyo, Japan). Primary antibodies used were AKT, AKT phosphorylated at serine (S) 473 (pAKT), p53 protein (P53) phosphorylated at S20 (pP53S20), MDM2 phosphorylated at S166 (pMDM2) (Cell Signaling Technology Inc., Tokyo, Japan), extracellular signal regulated kinase 1/2 (ERK1/2), PTEN, β-catenin (BD Biosciences, Tokyo, Japan), ERK1/2 diphosphorylated at T183 and Y185 (pERK1/2), β-actin, α-tubulin (Sigma-Aldrich Japan), PTEN phosphorylated at S380 (pPTEN), glycogen synthase kinase 3β phosphorylated at S9 (pGSK3β), glucose transporter isoform 1 (GLUT1), P53, P53 phosphorylated at S392 (pP53S392), P53 phosphorylated at S46 (pP53S46) and AFP (Abcam). Densitometric analysis of bands was performed using ImageJ software (1.50i, NIH). Data were expressed in arbitrary units as average of at least three independent experiments. The data of densitometric analyses from all bands were corrected by densities of each α-tubulin band, which was used as a loading control, and this provided the exact changes of the protein levels after antibody treatment. The data were calculated according to the following formula: the relative intensity = (the band intensity of the indicated day) / (the band intensity at day 7 of rabbit normal IgG-treated cells, instead of IgG non-treated control cells at day 0 or day 7).

Dot blot analysis

Cell lysates prepared from cells treated with either rabbit normal IgG or anti-human AFP antibody IgG for 7 days at 50.0 μg/ml were spotted (each 1 μl of 10, 5, 2.5 and 1 μg/μl) on nitrocellulose filters (Bio-Rad), dried and blocked. For detection of rabbit IgG, such as antibody administered and probably internalized in the cells, HRP-labeled goat anti-rabbit IgG was employed. For detection of intracellular AFP, the same above filters were used after procedure of de-probing the antibodies with signal stripping buffer (Restore™ PLUS Western Blot Stripping Buffer; Thermo Fisher Scientific Inc., Yokohama, Japan). After blocking, the filters were incubated with goat anti-human AFP antibody (Abcam) followed by HRP-labeled donkey anti-goat IgG (Abcam). Signals of each spot were obtained by ECL capture (ATTO light-capture II). Densitometric data were expressed as mean ± SD of three independent experiments.

Immunofluorescence cytology of subcellular localization of GLUT1

FLC7 cells were cultured with media containing either rabbit normal IgG or rabbit anti-human AFP antibody IgG, at 75 μg/ml concentration using a glass bottom culture dish (Iwaki Glass Base Dish, Asahi Glass Co., Ltd., Tokyo, Japan). After 5 days of culture, the cells were gently rinsed twice with cold Dulbecco's PBS (−), fixed with 3.3% formaldehyde in Dulbecco's PBS (−) for 10 min at 4°C. After rinsing with Dulbecco's PBS (−), the cells were permeabilized with 0.1% Triton X-100 in Dulbecco's PBS (−) for 10 min at 4°C and then blocked with 5% BSA and donkey normal IgG (5 mg/ml) in Dulbecco's PBS (−) containing 0.1% Tween-20 (TPBS) for 4 h. The cells were incubated with goat anti-GLUT 1 antibody (C-20; Santa Cruz Biotechnology, Inc., Dallas, TX, USA) diluted with 5% BSA in TPBS for 17 h at 4°C. After gentle washing, the cells were further incubated with Alexa Fluor® 488-labeled donkey anti-goat IgG (Abcam) in 5% BSA containing TPBS for 1 h at 37°C. After rinsing with Dulbecco's PBS (−), immunofluorescence images were captured using a Keyence BZ-8000 microscope (Keyence Corp., Inc., Osaka, Japan).

Reagents

For investigation on cell growth inhibition tests, rabbit polyclonal anti-human AFP antibody IgG (#PA-012) was obtained from Nippon Bio-Test Lab. Inc. (Tokyo, Japan). Rabbit normal IgG was purchased from Sigma-Aldrich Japan. For cell treatments, both IgG solutions were extensively dialyzed against Dulbecco's PBS (−) followed by IS-RPMI at 4°C. The solutions were filter-sterilized before use. BSA and donkey normal serum were from Sigma-Aldrich Japan. Donkey normal IgG fraction was prepared from its normal serum by the salting out method using 33% ammonium sulfate, followed by dialyzed extensively against 0.9% NaCl at 4°C. Protein concentration was measured by the DC protein assay kit (Bio-Rad) using the bovine serum IgG as the standard. RPMI-1640 medium was from Nissui Pharmaceutical Co., Ltd. (Tokyo, Japan). All other chemicals were of reagent grade.

Statistical analysis

Values of p<0.05 were considered statistically significant using Fisher's exact test.

Results and Discussion

Growth inhibition by anti-AFP antibody

Continuous exposure of the cells for 96 h to rabbit anti-human AFP antibody dose-dependently inhibited cell growth of the human hepatocellular carcinoma FLC7, as compared with the normal IgG control (Fig. 1A). The concentration of IgG of anti-human AFP antibody at IC50 was calculated to be ~50.0 μg/ml. As a result of treating cells for up to 7 days with rabbit anti-human AFP antibody at the concentration of 50.0 μg/ml, the antibody exhibited significant and time-dependent growth inhibition compared to the rabbit normal IgG used as the control (Fig. 1B).

Possibility of the formation of immune complexes

ELISA-determination of AFP showed that addition of anti-human AFP antibody decreased the AFP concentration in the medium below the detection limit (data not shown). At the same time, anti-human AFP antibody significantly reduced intracellular AFP levels up to 3.67–15.7% of the AFP concentration in rabbit normal IgG-treated control cells (Fig. 2A). Interestingly, however, SDS-PAGE/WB analysis of cell lysates using a goat antibody against human AFP and an HRP-labeled donkey anti-goat IgG antibody without cross-reactions to rabbit IgG revealed that the amount of AFP in the anti-AFP antibody-treated cells did not decrease substantially compared with that in IgG-treated control cells (Fig. 2B and C). Additionally, by anti-AFP antibody treatment of the cells, three detectable immunoreactive bands were found and they might be degradation products of the AFP molecules. Typical result is shown in Fig. 2C. Dot blot analyses demonstrated that the presence of anti-rabbit IgG specific immunoreactive substances, presumably derived from administered rabbit anti-AFP antibody to the cells, was sufficiently detectable with higher significance compared to normal IgG-administered cells (Fig. 2D and Table I). By contrast, the AFP reactive densitometric signals in both cell lysates were nearly the same. In each concentration of cell lysates applied on nitrocellulose filters, rabbit IgG derived signals were ~2.4–3.4 times higher in the anti-AFP antibody-treated cell lysates as compared to normal IgG-treated corresponding cell lysates (Table I). In these results, treatment of the cells with anti-AFP antibody might have led to the presence of free antibody due to an excess of antibody and the presence of immune complex (IC) generated from the administered antibody and AFP in the cell. When intracellular AFP is measured by an immunochemical technique such as ELISA, it can be predicted that both the excess antibody and the IC competitively inhibit the assay system. Analysis of the amount of intracellular AFP molecules by two different methods showed an obvious discrepancy between the results of each method, pointing to the generation and accumulation of IC in the cells. These results suggest that the amount of AFP was not reduced in antibody-treated cells, but rather, the reaction products of AFP and polyclonal anti-AFP antibody administered, the so-called ICs, had accumulated. Generation and accumulation of ICs consisting of multimeric aggregates of antibody and AFP is expected to inhibit AFP function without a large fluctuation in the amount of intracellular AFP. In other words, a large amount of AFP exists in the cytoplasm, but it is possible that its functions have been lost. As shown for the first time in this experiment the existence of intracellular IC derived from the antibody added extracellularly is sufficiently suspected.

Figure 2

Reduction of intracellular functional AFP concentration. Cells were treated with either rabbit anti-AFP antibody IgG (aAFP) or rabbit normal IgG (nIgG) at 50.0 μg/ml for 7 days. Cell lysates were prepared. (A) AFP concentration was assayed with ELISA. Columns, mean of triplicate determinations of two independent examinations; bars, SD. nIgG, mean ± SD of six selected samples obtained from cells treated with rabbit normal IgG on days 0 and 7. *Significant difference (p<0.05) compared to cells treated with rabbit either aAFP at day 0 or nIgG at days 0 and 7. **Non-significant difference (p>0.05) compared to cells treated with aAFP at day 0. Details are seen in Materials and methods. (B) Detection of anti-AFP antibody-reactive bands using SDS-PAGE/WB. Densitometric intensities of bands were analyzed by NIH ImageJ software and were corrected by densities of each α-tubulin band. The data were calculated according to the following formula: the relative intensity (ratio) = (the band intensity of the indicated day) / (the band intensity of the nIgG-treated cells on day 7). Details are seen in Materials and methods. (C) Typical SDS-PAGE/WB pattern of intracellular anti-AFP antibody reactive bands (a) and α-tubulin (b). (D) Dot reactivities of cell lysates to anti-AFP antibody (AFP), or anti-rabbit IgG antibody (rabbit IgG). Immunoreactivities were determined in the lysates from cells treated with either aAFP or nIgG for 7 days at 50.0 μg/ml, using ECL-densitometric analyses. Details are seen in Materials and methods.

Table I

Increased densitometric intensities of intracellular rabbit IgG immunoreactivity in cell lysates treated with rabbit anti-AFP antibody IgG (dot blot analysis).

Table I

Increased densitometric intensities of intracellular rabbit IgG immunoreactivity in cell lysates treated with rabbit anti-AFP antibody IgG (dot blot analysis).

Antibody toCell lysate (µg)Integrated density
aAFP treated cells
nIgG treated cells
MeanSDMeanSD
Rabbit IgG10205936a34735.367785.728598.6
5142118a31881.851623.717121.1
2.5101126a13536.744261.020816.0
163133.7a30589.218683.37903.6
AFP1040879851751.834124058997.5
531585352217.129163149265.0
2.523780531213.622115650002.1
114543449046.112324640601.6

a Significantly higher (p<0.05) density compared to lysate from cells treated with normal rabbit IgG (nIgG). Details are seen in Materials and methods. aAFP, rabbit anti-AFP antibody IgG.

Intracellular uptake of IgG molecules such as antibodies is known to occur by pinocytosis, macropinocytosis or endocytosis across the cell membrane. However, the mechanism for maintaining the function of antibody internalized in the cytoplasm is not well known (4143). In recent years, McEwan et al and Watkinson et al and their co-workers proposed the concept of intracellular antibody immunity (44,45) and many investigators reported methods for screening cell-internalizing antibodies, which possess high cell-internalizing activity, from polyclonal antibodies (46,47). It is possible that the cell-internalized antibody exerts its own unique function in the cell. Accordingly, there are many unknown factors to elucidate in this interesting field.

It is well -known that the cell growth promoting effect of AFP is exerted by interaction with many other molecules (3237). Furthermore, in principle, intermolecular interaction is based on the three-dimensional structure of related molecules. As a result of antibody binding followed by IC generation, it is highly possible that AFP molecules with a conformational change may lose their ability to interact with other related molecules and become so-called non-functional AFP. Even though abundant non-functional AFP is present in the cell, AFP functions such as growth promotion must be suppressed. Conversely, it is reasonable to infer that the intracellular AFP concentration measured by ELISA may correspond to the amount of AFP, which is not affected by IC generation and remains functional. Based on these results, it can be deduced that most of the AFP molecules detected by SDS - PAGE/WB in antibody-treated cells are non-functional AFP, and the cellular environment resulting from anti-AFP antibody treatment in this study was very similar to the environment of intracellular AFP elimination caused by experiments on RNA interference and microRNA technology, as reported previously (3234,37). Therefore, we confirmed the importance of further analyzing the changes in some major molecules interacting with the AFP molecule in the cell induced by anti-AFP antibody treatment.

Additionally in this experiment, the amount of AFP in the antibody-treated cells fluctuated slightly (Fig. 2B). The possible cause of this variation is considered to be the result of intracellular degradation of AFP derived from ICs, based on the anti-AFP antibody reactive bands in the low molecular weight region recognized by SDS-PAGE/WB analysis of the cell lysate treated with anti-AFP antibody (Fig. 2C). It is also another possibility on AFP-fragmentation that anti-AFP antibody in the cytoplasm binds to fragments of AFP molecule on messenger RNA being translated at the ribosomes, as reported in the purification experiment for messenger RNA of AFP by Miura et al (48). If this phenomenon occurs in the cell, it can be predicted that AFP protein synthesis would be inhibited and the protein fragments with incomplete length would be generated.

Anti-AFP antibody-induced suppression of the P13K/AKT pathway via PTEN stabilization

In this study, phosphorylation on S473 of the AKT molecule which is the protagonist of the PI3K/AKT signaling pathway was reduced by anti-human AFP antibody treatment (Fig. 3A). This result indicated that the PI3K/AKT signaling pathway was suppressed in anti-AFP antibody-treated FLC7 cells. It has been reported that in AFP-producing tumor cells, intracellular AFP molecules bind and interact closely with PTEN molecules, thereby suppressing the PTEN functions (both phosphatase-dependent and -independent activities) and consequently upregulating the PI3K/AKT signaling pathway (30,3235). Interestingly, the phosphorylated PTEN molecules at S380 were increased markedly by antibody treatment, with an extremely slight but significant variation in the intracellular protein level of PTEN (Fig. 3). At the same time, phosphorylation of ERK1/2 (T183 and Y185) was also reduced, suggesting an inhibition state of the MAP kinase signaling systems in the cells (Fig. 3A). There have been some reports that each PTEN molecule, which localizes in the nucleus or cytoplasm, exhibited different effects on various signaling pathways (4953). According to these reports, PTEN localizing in the cytoplasm decreases the level of phosphorylated AKT, upregulates P27kip1 and is required for apoptosis, whereas PTEN localizing in the nucleus downregulates phosphorylated MAP kinase and cyclin D1 and is crucial for cell cycle arrest, respectively. A recent study demonstrated that the anti-proliferative effect of synthetic anti-AFP scFv antibody on hepatic tumor cells was based on induction of G1 cell cycle arrest and apoptosis, probably related to the PI3K/AKT/PTEN signaling network (37), but a possible mechanism of action of anti-AFP scFv on cell-growth inhibition was not discussed in their report. As a result, the PTEN molecule can be expected to participate in the mechanism underlying the antitumor effect manifested by anti-AFP antibody.

PTEN was modified through strong phosphorylation at S380 of the carboxyl-terminal region (C-terminal tail domain) on the second day and thereafter due to antibody treatment. Phosphorylation of the S380, S385, T382, and T383 residues, but not T366, in the C-terminal region of PTEN molecules is known to not only increase the stability of the PTEN molecule but also decrease its phosphatase activity (5460). The C-terminal tail domain of PTEN has been shown to be important in regulating the stability and half-life of the molecules. Specifically, it is known that phosphorylation of the S380 residue is critical for PTEN protein stability (6164). Phosphorylation on S380 of PTEN at an early stage after antibody administration indicated that molecular stability was improved, but, on the other hand, excessive phosphatase activity was somewhat inhibited. It is thought that this phenomenon may be related to maintaining the molecular numbers of PTEN and to sustaining the potential for PTEN to control the PI3K/AKT signaling pathway. However, further investigations are necessary to clarify the true action mechanism(s) of each PTEN molecule with phosphorylation at various target residues, because neither change in PI3K enzyme activity nor exact contents of phosphatidylinositol-3,4,5-triphosphate was determined in this study.

As a result of a decrease in phosphorylated AKT (S473), phosphorylation on S9 of the GSK3β molecule, which is one of the target molecules of AKT (65), was relatively reduced (Fig. 3A). Conversely, enhancement of kinase activity of GSK3β is probable. Participation of GSK3β in PTEN-phosphorylation is already well recognized (54,55,58,65,66), and it is thought that an increase in kinase activities of GSK3β might enhance phosphorylation at T366 on the target PTEN molecules (58). T366 phosphorylation promotes destabilization and degradation of PTEN molecules as reported previously (58,67). However, in the present study, PTEN protein levels did show gradual and significant increase during treatment for up to 7 days (Fig. 3B and C). Due to the failure of molecular co-localization and interaction between AFP and PTEN occurring in the cytoplasm of the AFP-producing tumor cells, acute and severe reduction of the intracellular functional, immunoreactive AFP concentration due to IC generation caused by the administration of anti-AFP antibody resulted in a massive release of free and probably functional PTEN molecules from AFP in the cytoplasm. Moreover, intracellular protein levels of PTEN after antibody administration in this experiment were elevated slightly but significantly as shown in Fig. 3B and C. This finding suggests that selective phosphorylation on S380 of the free PTEN molecules induced molecular stabilization with the suppression of enzyme activity, as a mechanism to maintain potential control of the PI3K/AKT signaling pathway without degrading the rapidly increased level of PTEN molecules in the cell. To the contrary, this reaction should maintain the cancer cell property of preferential growth as a cancer cell inactivates excess PTEN molecules, which act as a major suppressive regulator in the PI3K/AKT signaling pathway and are, therefore, an impediment to proliferation reactions of the cancer cells. This mechanism is a self-defense reaction that usually occurs in cancer cells in order to survive. This is an interesting phenomenon in cancer biology and requires further elucidation. It has been reported that phosphorylation at S380 of the PTEN molecule at the C-terminal tail, does not involve casein kinase 2 and GSK3β (5860), but a result of the interaction of other proteins, such as protein interacting with carboxyl terminus-1 (PICT-1) (63,64). Okahara et al suggested that PICT-1 is a PTEN-interacting protein that promotes the phosphorylation and stability of PTEN, and can regulate the phosphorylation of PTEN at S380 (63,64). The authors concluded that the binding of PTEN to PICT-1 governs its turnover via phosphorylation of the C-terminal region. Their result and the findings of our present study provide insight into unknown other molecular mechanism(s) by which PTEN turnover is controlled.

Upregulation of P53 functions by anti-AFP antibody treatment

Protein levels of P53 were slightly and significantly increased during antibody treatment (Fig. 3), because PTEN increased P53 stability and function via interaction of both molecules, which bind the C2 domain of PTEN to the C-terminus of P53 (68,69). Therefore, in cells lacking PTEN, P53 levels are significantly reduced owing to decreased stability as reported previously (68). Expression of wild-type or phosphatase-dead forms of PTEN with a recombinant construct also increases P53 stability in MDM2 (a ubiquitin ligase for P53)-independent manner (68). Phosphorylation reactions of P53 on S20 and on S392 were significantly enhanced after antibody treatment (Fig. 3), suggesting stabilization and activation of the molecule. Post translational modifications of the P53 molecule, such as phosphorylation, acetylation, and ubiquitination, have been recognized to regulate the stability and functions of P53 proteins (6974). It is well-known that DNA damage-induced phosphorylation at S20 of P53 leads to a reduced interaction between P53 and its negative regulator, MDM2. As a result, S20 phosphorylation impairs the ability of MDM2 to bind P53, promoting both accumulation and functional activation of P53 in response to DNA damage (72). MDM2 plays a central role in regulating the stability of P53 and inhibits P53 accumulation by targeting it for ubiquitination and proteasomal degradation (7577). Phosphorylated P53 at S392 is also essential for promoting its tetramerization, stability, and functional activity as well as for S20-phosphorylation reactions (70,71). These findings indicate that the regular functions of P53 recovered after antibody treatment. Phosphorylation of P53 at S46, which regulates the ability of P53 to induce apoptosis (73), also increased slightly, suggesting the triggering of apoptotic signaling (Fig. 3A).

It is of interest that an extreme and significant increase in phosphorylated MDM2 on S166 (pMDM2) was observed in antibody-treated tumor cells within 2 days after the beginning of antibody administration (Fig. 3). As known previously (76,77), the presence of pMDM2 indicates the enhancement of E3 ligase activity to promote ubiquitin-dependent degradation of P53. Additionally, phosphorylation at S166 and at S186 of MDM2 has been shown to depend on AKT enzyme activity (76,77). In the present study, antibody administration elicited a decrease in intracellular AFP, releasing PTEN from AFP, followed by stabilization of the molecular functions of free PTEN. The resultant free but functional PTEN was able to suppress the PI3K/AKT signaling system, which also serves as a survival pathway in cancer cells. PTEN also promoted stabilization and expression of the biological functions of the P53 molecule that interacts with the PTEN molecule. Under such circumstances, it can be deduced that the accumulation and abnormal increase of pMDM2 (S166) in liver cancer cells are needed in order to maintain a steady state of the cancer cells and enhance the degradation of P53, the tumor suppressor molecule, even when AKT enzyme activity is inhibited. There was no difference in the expression level of β-catenin protein (Fig. 3A), a major protein of the Wnt/β-catenin pathway, which is one of the important cell survival regulatory pathways involving the PI3K/AKT/GSK3β axis (66).

Induction of subcellular translocation of GLUT1 by anti-AFP antibody

In the present study, protein levels of a universal molecule in the glucose transporter species, GLUT1, showed a significant increase immediately after antibody treatment and continued for 7 days (Fig. 3). The increase in GLUT1 protein levels observed early during antibody treatment clearly pointed to a deficiency of and need for glucose in the tumor cells. It is known that a proliferating tumor cell vigorously consumes sugar, especially glucose, irrespective of the presence or absence of oxygen. The fact that the PTEN function in FLC7 cells with a high level of AFP production was strongly suppressed by the AFP is compatible with the above-mentioned results. Under the condition of malfunction of PTEN, immunofluorescence cytology revealed a change in the subcellular localization of GLUT1 molecules (Fig. 4). In control cells with malfunction of PTEN, treated with rabbit normal IgG, many GLUT1 molecules were localized patchily on the surface of the cell membrane. In contrast, in cells treated with rabbit anti-AFP antibody IgG, the distribution of the weak fluorescence indicating GLUT1 was found throughout the cytoplasm, and localization of GLUT1 on the cell surface membranes was reduced. Morani et al reported that in genetically manipulated human thyroid cancer cell lines, the loss of PTEN expression was associated with increased expression of GLUT1 on the cell surface plasma membrane and enhanced the translocation of GLUT1 proteins onto the surface of plasma membrane from the cytoplasm (78). They concluded that the PTEN protein regulated plasma membrane expression of GLUT1 and that the loss of function of PTEN increased the probability of cancer detection by 18F-fluorodeoxyglucose positron emission tomography or other glucose-based imaging diagnosis in a clinical setting. This result was similar to the reported finding that the plasma membrane translocation of GLUT1 was dependent on the PI3K/AKT signaling pathway (7981). Immunocytohistologic studies have also shown that enhanced glucose uptake in cancer cells correlates with overexpression of GLUTs, especially of GLUT1 (78,81). Our results fully supported the previous findings that the restored PTEN molecules liberated from AFP closely participated in the process of intracellular distribution of GLUT1. To clarify the relationship between PTEN function and the translocation of GLUT1, it may be important to investigate other unknown function(s) of the PTEN molecule involved in regulating subcellular migration of the target molecules. As reported previously in our study (8), the inhibition of glucose uptake by the treatment of AFP-producing rat hepatoma AH66 cells with horse anti-rat AFP antibody occurred within a few hours after the treatment. There was no remarkable change in the Km value in the kinetic analysis of the uptake, while a decrease in the Vmax was observed indicating that non-competitive inhibition was the main cause of glucose uptake failure. At that time, it was assumed that no qualitative change in the transport carrier occurred, but there was a quantitative change and a decrease in the number of unknown sugar transporters. Currently, one of the effective treatments of hepatocellular carcinoma in the clinical setting is inhibition of angiogenesis in liver cancer tissue to induce malnutrition, such as deficiency of sugar, other nutrients and oxygen (8285). The induction of such malnutrition in cancer cells in the present study by administration of anti-AFP antibody is a significant finding.

There have been several reports that small but detectable amounts of autoantibodies against human AFP were produced by an unknown factor in vivo, including in the presence of cancer (8690). Generation of autoantibodies to carcinoembryonic antigen (CEA) has been also reported in several kinds of cancers and the expression of these anti-CEA autoantibodies depleted the serum CEA antigen levels (9194). It can be postulated that by a similar unknown mechanism, excess amounts of autoantibodies raised against AFP in the serum or in the cells are generated, and then bind to AFP, thereby decreasing the AFP concentration. However, there are no reports yet of any findings related to AFP that might compare with the rapid deletion of serum CEA due to increased expression of anti-CEA autoantibody. The phenomenon of anti-AFP antibody eliminating abundant functional AFP from the cytoplasm as well as from the extracellular matrices, with severe accumulation of ICs as a result of some unknown phenomenon cannot be expected for liver cancer cells. Liver cells, parenchymal and non-parenchymal interstitial cells, as well as liver cancer cells exhibit a confusing biological reaction in response to the cellular environment resulting from anti-AFP antibody treatment.

As shown in this study, the administration of anti-AFP antibody had an inhibitory effect on cell growth via suppression of the PI3K/AKT signaling pathway. Because of a decrease in intracellular functional AFP resulting from the generation of ICs consisting of AFP and anti-AFP antibody, the production and accumulation of ICs derived from the AFP-anti AFP antibody reaction exerted the same cell growth inhibitory effect as inhibition of intracellular AFP expression. Unfortunately, the cytotoxicity and antitumor efficacy of the specific antibody alone were weaker in comparison to so-called anticancer agents. However, a more recent report indicated that intracellular AFP expression is closely related to hepatocarcinogenesis, as well as liver cancer progression (95). For such reasons, elucidating the mechanism of action of cytotoxicity of anti-AFP antibody on AFP-producing tumor cells serves as a sound basis not only for use in combination with other effective liver cancer therapy but also for developing strategic measures for liver cancer prevention.

Acknowledgments

This study was supported in part by MEXT-Supported Program for the Strategic Research Foundation at Private Universities (2011–2015) from the Ministry of Education, Culture, Sports, Science and Technology.

References

1 

Mizejewski GJ and Allen RP: Immunotherapeutic suppression in transplantable solid tumours. Nature. 250:50–52. 1974. View Article : Google Scholar : PubMed/NCBI

2 

Mizejewski GJ, Young SR and Allen RP: α fetoprotein: Effect of heterologous antiserum on hepatoma cells in vitro. J Natl Cancer Inst. 54:1361–1367. 1975. View Article : Google Scholar : PubMed/NCBI

3 

Mizejewski GJ and Allen RP: α-fetoprotein: Studies of tumor-associated antigen cytotoxicity in mouse hepatoma BW7756. Clin Immunol Immunopathol. 11:307–317. 1978. View Article : Google Scholar : PubMed/NCBI

4 

Mizejewski GJ and Dillon WR: Immunobiologic studies in hepatoma-bearing mice passively immunized to α-fetoprotein. Arch Immunol Ther Exp (Warsz). 27:655–662. 1979.

5 

Tsukada Y, Mikuni M, Watabe H, Nishi S and Hirai H: Effect of anti-alpha-fetoprotein serum on some cultured tumor cells. Int J Cancer. 13:187–195. 1974. View Article : Google Scholar : PubMed/NCBI

6 

Wepsic HT, Tsukada Y, Takeichi N, Nishi S and Hirai H: Effect of horse antibody to rat alpha-fetoprotein upon the growth of AH-66 in Donryu rats. Int J Cancer. 25:655–661. 1980. View Article : Google Scholar : PubMed/NCBI

7 

Koji T, Ishii N, Munehisa T, Kusumoto Y, Nakamura S, Tamenishi A, Hara A, Kobayashi K, Tsukada Y, Nishi S, et al: Localization of radioiodinated antibody to alpha-fetoprotein in hepatoma transplanted in rats and a case report of alpha-fetoprotein antibody treatment of a hepatoma patient. Cancer Res. 40:3013–3015. 1980.PubMed/NCBI

8 

Ohkawa K, Tsukada Y, Hibi N and Hirai H: The inhibitory effects of horse anti-rat AFP antiserum on the uptake of 2-deoxy-D-glucose by AFP-producing rat hepatoma cells. Int J Cancer. 33:497–502. 1984. View Article : Google Scholar : PubMed/NCBI

9 

Tsukada Y, Bischof WK, Hibi N, Hirai H, Hurwitz E and Sela M: Effect of a conjugate of daunomycin and antibodies to rat alpha-fetoprotein on the growth of alpha-fetoprotein-producing tumor cells. Proc Natl Acad Sci USA. 79:621–625. 1982. View Article : Google Scholar : PubMed/NCBI

10 

Tsukada Y, Kato Y, Umemoto N, Takeda Y, Hara T and Hirai H: An anti-alpha-fetoprotein antibody-daunorubicin conjugate with a novel poly-L-glutamic acid derivative as intermediate drug carrier. J Natl Cancer Inst. 73:721–729. 1984.PubMed/NCBI

11 

Tsukada Y, Hurwitz E, Kashi R, Sela M, Hibi N, Hara A and Hirai H: Chemotherapy by intravenous administration of conjugates of daunomycin with monoclonal and conventional anti-rat alpha-fetoprotein antibodies. Proc Natl Acad Sci USA. 79:7896–7899. 1982. View Article : Google Scholar : PubMed/NCBI

12 

Tsukada Y, Hurwitz E, Kashi R, Sela M, Hibi N, Hara A and Hirai H: Effect of a conjugate of daunomycin and purified polyclonal or monoclonal antibodies to rat alpha-fetoprotein on the growth of alpha-fetoprotein-producing tumor cells. Ann NY Acad Sci. 417:262–269. 1983. View Article : Google Scholar : PubMed/NCBI

13 

Kato Y, Tsukada Y, Hara T and Hirai H: Enhanced antitumor activity of mitomycin C conjugated with anti-alpha-fetoprotein antibody by a novel method of conjugation. J Appl Biochem. 5:313–319. 1983.PubMed/NCBI

14 

Tsukada Y, Ohkawa K and Hibi N: Suppression of human alpha-foetoprotein-producing hepatocellular carcinoma growth in nude mice by an anti alpha-foetoprotein antibody-daunorubicin conjugate with a poly-L-glutamic acid derivative as intermediate drug carrier. Br J Cancer. 52:111–116. 1985. View Article : Google Scholar : PubMed/NCBI

15 

Ohkawa K, Hibi N and Tsukada Y: Evaluation of a conjugate of purified antibodies against human AFP-dextran-daunorubicin to human AFP-producing yolk sac tumor cell lines. Cancer Immunol Immunother. 22:81–86. 1986. View Article : Google Scholar : PubMed/NCBI

16 

Tsukada Y, Ohkawa K and Hibi N: Therapeutic effect of treatment with polyclonal or monoclonal antibodies to alpha-fetoprotein that have been conjugated to daunomycin via a dextran bridge: Studies with an alpha-fetoprotein-producing rat hepatoma tumor model. Cancer Res. 47:4293–4295. 1987.PubMed/NCBI

17 

Ohkawa K, Tsukada Y, Hibi N, Umemoto N and Hara T: Selective in vitro and in vivo growth inhibition against human yolk sac tumor cell lines by purified antibody against human alpha-fetoprotein conjugated with mitomycin C via human serum albumin. Cancer Immunol Immunother. 23:81–86. 1986. View Article : Google Scholar : PubMed/NCBI

18 

Kim EE, DeLand FH, Nelson MO, Bennett S, Simmons G, Alpert E and Goldenberg DM: Radioimmunodetection of cancer with radiolabeled antibodies to alpha-fetoprotein. Cancer Res. 40:3008–3012. 1980.PubMed/NCBI

19 

Kim EE, Deland FH, Casper S, Corgan RL, Primus FJ and Goldenberg DM: Radioimmunodetection of colorectal cancer. Cancer. 45(Suppl): 1243–1247. 1980. View Article : Google Scholar : PubMed/NCBI

20 

Uriel J, Villacampa MJ, Moro R, Naval J and Failly-Crépin C: Uptake of radiolabeled a-fetoprotein by mouse mammary carcinomas and its usefulness in tumor scintigraphy. Cancer Res. 44:5314–5319. 1984.PubMed/NCBI

21 

Goldenberg DM: Cancer imaging with CEA antibodies: Historical and current perspectives. Int J Biol Markers. 7:183–188. 1992.PubMed/NCBI

22 

Behr TM, Liersch T, Greiner-Bechert L, Griesinger F, Béhé M, Markus PM, Gratz S, Angerstein C, Brittinger G, Becker H, et al: Radioimmunotherapy of small-volume disease of metastatic colorectal cancer. Cancer. 94(Suppl): 1373–1381. 2002. View Article : Google Scholar : PubMed/NCBI

23 

Aarts F, Boerman OC, Sharkey RM, Hendriks T, Chang CH, McBride WJ, Bleichrodt RP, Oyen WJ and Goldenberg DM: Pretargeted radioimmunoscintigraphy in patients with primary colorectal cancer using a bispecific anticarcinoembryonic antigen CEA X anti-di-diethylenetriaminepentaacetic acid F(ab′)2 antibody. Cancer. 116(Suppl): 1111–1117. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Mizejewski GJ: Biological role of alpha-fetoprotein in cancer: Prospects for anticancer therapy. Expert Rev Anticancer Ther. 2:709–735. 2002. View Article : Google Scholar : PubMed/NCBI

25 

Li MS, Li PF, Yang FY, He SP, Du GG and Li G: The intracellular mechanism of alpha-fetoprotein promoting the proliferation of NIH 3T3 cells. Cell Res. 12:151–156. 2002. View Article : Google Scholar : PubMed/NCBI

26 

Li MS, Li PF, He SP, Du GG and Li G: The promoting molecular mechanism of alpha-fetoprotein on the growth of human hepatoma Bel7402 cell line. World J Gastroenterol. 8:469–475. 2002. View Article : Google Scholar : PubMed/NCBI

27 

Li P, Wang SS, Liu H, Li N, McNutt MA, Li G and Ding HG: Elevated serum alpha fetoprotein levels promote pathological progression of hepatocellular carcinoma. World J Gastroenterol. 17:4563–4571. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Moro R, Gulyaeva-Tcherkassova J and Stieber P: Increased alpha-fetoprotein receptor in the serum of patients with early-stage breast cancer. Curr Oncol. 19:e1–e8. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Wang S, Jiang W, Chen X, Zhang C, Li H, Hou W, Liu Z, McNutt MA, Lu F and Li G: Alpha-fetoprotein acts as a novel signal molecule and mediates transcription of Fn14 in human hepatocellular carcinoma. J Hepatol. 57:322–329. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Zhu M, Lin B, Zhou P and Li M: Molecular analysis of AFP and HSA interactions with PTEN potein. BioMed Res Int. 2015:2569162015. View Article : Google Scholar

31 

Mizejewski GJ: Nonsecreted cytoplasmic alpha-fetoprotein: A newly discovered role in intracellular signaling and regulation. An update and commentary. Tumour Biol. 36:9857–9864. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Li M, Li H, Li C, Wang S, Jiang W, Liu Z, Zhou S, Liu X, McNutt MA and Li G: Alpha-fetoprotein: A new member of intracellular signal molecules in regulation of the PI3K/AKT signaling in human hepatoma cell lines. Int J Cancer. 128:524–532. 2011. View Article : Google Scholar

33 

Gao R, Cai C, Gan J, Yang X, Shuang Z, Liu M, Li S and Tang H: miR-1236 down-regulates alpha-fetoprotein, thus causing PTEN accumulation, which inhibits the PI3K/Akt pathway and malignant phenotype in hepatoma cells. Oncotarget. 6:6014–6028. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Zhu M, Guo J, Li W, Lu Y, Fu S, Xie X, Xia H, Dong X, Chen Y, Quan M, et al: Hepatitis B virus X protein induces expression of alpha-fetoprotein and activates PI3K/mTOR signaling pathway in liver cells. Oncotarget. 6:12196–12208. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Su R, Nan H, Guo H, Ruan Z, Jiang L, Song Y and Nan K: Associations of components of PTEN/AKT/mTOR pathway with cancer stem cell markers and prognostic value of these biomarkers in hepatocellular carcinoma. Hepatol Res. 46:1380–1391. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Zhu M, Guo J, Xia H, Li W, Lu Y, Dong X, Chen Y, Xie X, Fu S and Li M: Alpha-fetoprotein activates AKT/mTOR signaling to promote CXCR4 expression and migration of hepatoma cells. Oncoscience. 2:59–70. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Ji X, Shen Y, Sun H and Gao X: A novel anti-alpha-fetoprotein single-chain variable fragment displays anti-tumor effects in HepG2 cells as a single agent or in combination with paclitaxel. Tumour Biol. 37:10085–10096. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Matsumoto M, Matsuura T, Aoki K, Maehashi H, Iwamoto T, Ohkawa K, Yoshida K, Yanaga K and Takada K: An efficient system for secretory production of fibrinogen using a hepatocellular carcinoma cell line. Hepatol Res. 45:315–325. 2015. View Article : Google Scholar

39 

Nakabayashi H, Taketa K, Miyano K, Yamane T and Sato J: Growth of human hepatoma cells lines with differentiated functions in chemically defined medium. Cancer Res. 42:3858–3863. 1982.PubMed/NCBI

40 

Ohkawa K, Tsukada Y, Murae M, Kimura E, Takada K, Abe T, Terashima Y and Mitani K: Serum levels and biochemical characteristics of human ovarian carcinoma-associated antigen defined by murine monoclonal antibody, CF511. Br J Cancer. 60:953–960. 1989. View Article : Google Scholar : PubMed/NCBI

41 

Baumann H and Doyle D: Metabolic fate of cell surface glycoproteins during immunoglobulin-induced internalization. Cell. 21:897–907. 1980. View Article : Google Scholar : PubMed/NCBI

42 

Press OW, Hansen JA, Farr A and Martin PJ: Endocytosis and degradation of murine anti-human CD3 monoclonal antibodies by normal and malignant T-lymphocytes. Cancer Res. 48:2249–2257. 1988.PubMed/NCBI

43 

Kyriakos RJ, Shih LB, Ong GL, Patel K, Goldenberg DM and Mattes MJ: The fate of antibodies bound to the surface of tumor cells in vitro. Cancer Res. 52:835–842. 1992.PubMed/NCBI

44 

McEwan WA, Tam JC, Watkinson RE, Bidgood SR, Mallery DL and James LC: Intracellular antibody-bound pathogens stimulate immune signaling via the Fc receptor TRIM21. Nat Immunol. 14:327–336. 2013. View Article : Google Scholar : PubMed/NCBI

45 

Watkinson RE, McEwan WA and James LC: Intracellular antibody immunity. J Clin Immunol. 34(Suppl 1): S30–S34. 2014. View Article : Google Scholar : PubMed/NCBI

46 

Yoshikawa M, Mukai Y, Okada Y, Tsumori Y, Tsunoda S, Tsutsumi Y, Aird WC, Yoshioka Y, Okada N, Doi T, et al: Robo4 is an effective tumor endothelial marker for antibody-drug conjugates based on the rapid isolation of the anti-Robo4 cell-internalizing antibody. Blood. 121:2804–2813. 2013. View Article : Google Scholar : PubMed/NCBI

47 

Ha KD, Bidlingmaier SM, Su Y, Lee NK and Liu B: Identification of novel macropinocytosing human antibodies by phage display and high-content analysis. Methods Enzymol. 585:91–110. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Miura K, Law SW, Nishi S and Tamaoki T: Isolation of alphafetoprotein messenger RNA from mouse yolk sac. J Biol Chem. 254:5515–5521. 1979.PubMed/NCBI

49 

Zhou BP, Liao Y, Xia W, Spohn B, Lee MH and Hung MC: Cytoplasmic localization of p21Cip1/WAF1 by Akt-induced phosphorylation in HER-2/neu-overexpressing cells. Nat Cell Biol. 3:245–252. 2001. View Article : Google Scholar : PubMed/NCBI

50 

Chung JH and Eng C: Nuclear-cytoplasmic partitioning of phosphatase and tensin homologue deleted on chromosome 10 (PTEN) differentially regulates the cell cycle and apoptosis. Cancer Res. 65:8096–8100. 2005. View Article : Google Scholar : PubMed/NCBI

51 

Chung JH, Ginn-Pease ME and Eng C: Phosphatase and tensin homologue deleted on chromosome 10 (PTEN) has nuclear localization signal-like sequences for nuclear import mediated by major vault protein. Cancer Res. 65:4108–4116. 2005. View Article : Google Scholar : PubMed/NCBI

52 

Chung JH, Ostrowski MC, Romigh T, Minaguchi T, Waite KA and Eng C: The ERK1/2 pathway modulates nuclear PTEN-mediated cell cycle arrest by cyclin D1 transcriptional regulation. Hum Mol Genet. 15:2553–2559. 2006. View Article : Google Scholar : PubMed/NCBI

53 

Gil A, Andrés-Pons A, Fernández E, Valiente M, Torres J, Cervera J and Pulido R: Nuclear localization of PTEN by a Ran-dependent mechanism enhances apoptosis: Involvement of an N-terminal nuclear localization domain and multiple nuclear exclusion motifs. Mol Biol Cell. 17:4002–4013. 2006. View Article : Google Scholar : PubMed/NCBI

54 

Al-Khouri AM, Ma Y, Togo SH, Williams S and Mustelin T: Cooperative phosphorylation of the tumor suppressor phosphatase and tensin homologue (PTEN) by casein kinases and glycogen synthase kinase 3beta. J Biol Chem. 280:35195–35202. 2005. View Article : Google Scholar : PubMed/NCBI

55 

Tamguney T and Stokoe D: New insights into PTEN. J Cell Sci. 120:4071–4079. 2007. View Article : Google Scholar : PubMed/NCBI

56 

Georgescu MM, Kirsch KH, Akagi T, Shishido T and Hanafusa H: The tumor-suppressor activity of PTEN is regulated by its carboxyl-terminal region. Proc Natl Acad Sci USA. 96:10182–10187. 1999. View Article : Google Scholar : PubMed/NCBI

57 

Tolkacheva T and Chan AM: Inhibition of H-Ras transformation by the PTEN/MMAC1/TEP1 tumor suppressor gene. Oncogene. 19:680–689. 2000. View Article : Google Scholar : PubMed/NCBI

58 

Maccario H, Perera NM, Davidson L, Downes CP and Leslie NR: PTEN is destabilized by phosphorylation on Thr366. Biochem J. 405:439–444. 2007. View Article : Google Scholar : PubMed/NCBI

59 

Torres J and Pulido R: The tumor suppressor PTEN is phosphorylated by the protein kinase CK2 at its C terminus. Implications for PTEN stability to proteasome-mediated degradation. J Biol Chem. 276:993–998. 2001. View Article : Google Scholar

60 

Milella M, Falcone I, Conciatori F, Cesta Incani U, Del Curatolo A, Inzerilli N, Nuzzo CM, Vaccaro V, Vari S, Cognetti F, et al: PTEN: Multiple functions in human malignant tumors. Front Oncol. 5:242015. View Article : Google Scholar : PubMed/NCBI

61 

Vazquez F, Ramaswamy S, Nakamura N and Sellers WR: Phosphorylation of the PTEN tail regulates protein stability and function. Mol Cell Biol. 20:5010–5018. 2000. View Article : Google Scholar : PubMed/NCBI

62 

Birle D, Bottini N, Williams S, Huynh H, deBelle I, Adamson E and Mustelin T: Negative feedback regulation of the tumor suppressor PTEN by phosphoinositide-induced serine phosphorylation. J Immunol. 169:286–291. 2002. View Article : Google Scholar : PubMed/NCBI

63 

Okahara F, Ikawa H, Kanaho Y and Maehama T: Regulation of PTEN phosphorylation and stability by a tumor suppressor candidate protein. J Biol Chem. 279:45300–45303. 2004. View Article : Google Scholar : PubMed/NCBI

64 

Okahara F, Itoh K, Nakagawara A, Murakami M, Kanaho Y and Maehama T: Critical role of PICT-1, a tumor suppressor candidate, in phosphatidylinositol 3,4,5-trisphosphate signals and tumorigenic transformation. Mol Biol Cell. 17:4888–4895. 2006. View Article : Google Scholar : PubMed/NCBI

65 

Doble BW and Woodgett JR: GSK-3: Tricks of the trade for a multi-tasking kinase. J Cell Sci. 116:1175–1186. 2003. View Article : Google Scholar : PubMed/NCBI

66 

Saini MK and Sanyal SN: PTEN regulates apoptotic cell death through PI3-K/Akt/GSK3p signaling pathway in DMH induced early colon carcinogenesis in rat. Exp Mol Pathol. 93:135–146. 2012. View Article : Google Scholar : PubMed/NCBI

67 

Tibarewal P, Zilidis G, Spinelli L, Schurch N, Maccario H, Gray A, Perera NM, Davidson L, Barton GJ and Leslie NR: PTEN protein phosphatase activity correlates with control of gene expression and invasion, a tumor-suppressing phenotype, but not with AKT activity. Sci Signal. 5:ra182012. View Article : Google Scholar : PubMed/NCBI

68 

Freeman DJ, Li AG, Wei G, Li HH, Kertesz N, Lesche R, Whale AD, Martinez-Diaz H, Rozengurt N, Cardiff RD, et al: PTEN tumor suppressor regulates p53 protein levels and activity through phosphatase-dependent and -independent mechanisms. Cancer Cell. 3:117–130. 2003. View Article : Google Scholar : PubMed/NCBI

69 

Li AG, Piluso LG, Cai X, Wei G, Sellers WR and Liu X: Mechanistic insights into maintenance of high p53 acetylation by PTEN. Mol Cell. 23:575–587. 2006. View Article : Google Scholar : PubMed/NCBI

70 

Hupp TR, Meek DW, Midgley CA and Lane DP: Regulation of the specific DNA binding function of p53. Cell. 71:875–886. 1992. View Article : Google Scholar : PubMed/NCBI

71 

Sakaguchi K, Sakamoto H, Lewis MS, Anderson CW, Erickson JW, Appella E and Xie D: Phosphorylation of serine 392 stabilizes the tetramer formation of tumor suppressor protein p53. Biochemistry. 36:10117–10124. 1997. View Article : Google Scholar : PubMed/NCBI

72 

Shieh SY, Taya Y and Prives C: DNA damage-inducible phosphorylation of p53 at N-terminal sites including a novel site, Ser20, requires tetramerization. EMBO J. 18:1815–1823. 1999. View Article : Google Scholar : PubMed/NCBI

73 

Oda K, Arakawa H, Tanaka T, Matsuda K, Tanikawa C, Mori T, Nishimori H, Tamai K, Tokino T, Nakamura Y, et al: p53AIP1, a potential mediator of p53-dependent apoptosis, and its regulation by Ser-46-phosphorylated p53. Cell. 102:849–862. 2000. View Article : Google Scholar : PubMed/NCBI

74 

Hirao A, Kong YY, Matsuoka S, Wakeham A, Ruland J, Yoshida H, Liu D, Elledge SJ and Mak TW: DNA damage-induced activation of p53 by the checkpoint kinase Chk2. Science. 287:1824–1827. 2000. View Article : Google Scholar : PubMed/NCBI

75 

Haupt Y, Maya R, Kazaz A and Oren M: Mdm2 promotes the rapid degradation of p53. Nature. 387:296–299. 1997. View Article : Google Scholar : PubMed/NCBI

76 

Mayo LD and Donner DB: A phosphatidylinositol 3-kinase/Akt pathway promotes translocation of Mdm2 from the cytoplasm to the nucleus. Proc Natl Acad Sci USA. 98:11598–11603. 2001. View Article : Google Scholar : PubMed/NCBI

77 

Zhou BP, Liao Y, Xia W, Zou Y, Spohn B and Hung MC: HER-2/neu induces p53 ubiquitination via Akt-mediated MDM2 phosphorylation. Nat Cell Biol. 3:973–982. 2001. View Article : Google Scholar : PubMed/NCBI

78 

Morani F, Phadngam S, Follo C, Titone R, Aimaretti G, Galetto A, Alabiso O and Isidoro C: PTEN regulates plasma membrane expression of glucose transporter 1 and glucose uptake in thyroid cancer cells. J Mol Endocrinol. 53:247–258. 2014. View Article : Google Scholar : PubMed/NCBI

79 

Samih N, Hovsepian S, Aouani A, Lombardo D and Fayet G: Glut-1 translocation in FRTL-5 thyroid cells: Role of phosphatidylinositol 3-kinase and N-glycosylation. Endocrinology. 141:4146–4155. 2000. View Article : Google Scholar : PubMed/NCBI

80 

Hajduch E, Litherland GJ and Hundal HS: Protein kinase B (PKB/Akt) - a key regulator of glucose transport? FEBS Lett. 492:199–203. 2001. View Article : Google Scholar : PubMed/NCBI

81 

Ciampi R, Vivaldi A, Romei C, Del Guerra A, Salvadori P, Cosci B, Pinchera A and Elisei R: Expression analysis of facilitative glucose transporters (GLUTs) in human thyroid carcinoma cell lines and primary tumors. Mol Cell Endocrinol. 291:57–62. 2008. View Article : Google Scholar : PubMed/NCBI

82 

Wang CH, Wey KC, Mo LR, Chang KK, Lin RC and Kuo JJ: Current trends and recent advances in diagnosis, therapy, and prevention of hepatocellular carcinoma. Asian Pac J Cancer Prev. 16:3595–3604. 2015. View Article : Google Scholar : PubMed/NCBI

83 

Taketomi A: Clinical trials of antiangiogenic therapy for hepatocellular carcinoma. Int J Clin Oncol. 21:213–218. 2016. View Article : Google Scholar : PubMed/NCBI

84 

Dhir M, Melin AA, Douaiher J, Lin C, Zhen WK, Hussain SM, Geschwind JF, Doyle MB, Abou-Alfa GK and Are C: A review and update of treatment options and controversies in the management of hepatocellular carcinoma. Ann Surg. 263:1112–1125. 2016. View Article : Google Scholar : PubMed/NCBI

85 

Lin J, Wu L, Bai X, Xie Y, Wang A, Zhang H, Yang X, Wan X, Lu X, Sang X, et al: Combination treatment including targeted therapy for advanced hepatocellular carcinoma. Oncotarget. 7:71036–71051. 2016.PubMed/NCBI

86 

Nakata K, Muro T, Furukawa R, Kono K, Kusumoto Y, Ishii N, Munehisa T, Koji T and Nagataki S: Presence of immunoglobulin G in human sera binding to alphafetoprotein. Oncodev Biol Med. 4:C101–C104. 1983.PubMed/NCBI

87 

Asano T, Yamada N, Ochiai T, Sato H and Fukao T: Presence of anti-AFP-antibody producing B cells in peripheral blood lymphocyte of hepatocellular carcinoma patient. Nihon Shokakibyo Gakkai Zasshi. 81:2781984.In Japanese.

88 

Sassi F, Ayed K, el Gaied A and Dellagi K: Presence of antialphafetoprotein immunoglobulin G in serum of a patient with hepatocellular carcinoma. Gastroenterol Clin Biol. 15:661–662. 1991.in French.

89 

Liu H, Zhang J, Wang S, Pang Z, Wang Z, Zhou W and Wu M: Screening of autoantibodies as potential biomarkers for hepatocellular carcinoma by using T7 phase display system. Cancer Epidemiol. 36:82–88. 2012. View Article : Google Scholar

90 

Negm OH, Hamed MR, Schoen RE, Whelan RL, Steele RJ, Scholefield J, Dilnot EM, Shantha Kumara HM, Robertson JF and Sewell HF: Human blood autoantibodies in the detection of colorectal cancer. PLoS One. 11:e01569712016. View Article : Google Scholar : PubMed/NCBI

91 

Ura Y, Ochi Y, Hamazu M, Ishida M, Nakajima K and Watanabe T: Studies on circulating antibody against carcinoembryonic antigen (CEA) and CEA-like antigen in cancer patients. Cancer Lett. 25:283–295. 1985. View Article : Google Scholar : PubMed/NCBI

92 

Konstadoulakis MM, Syrigos KN, Albanopoulos C, Mayers G and Golematis B: The presence of anti-carcinoembryonic antigen (CEA) antibodies in the sera of patients with gastrointestinal malignancies. J Clin Immunol. 14:310–313. 1994. View Article : Google Scholar : PubMed/NCBI

93 

Haidopoulos D, Konstadoulakis MM, Antonakis PT, Alexiou DG, Manouras AM, Katsaragakis SM and Androulakis GF: Circulating anti-CEA antibodies in the sera of patients with breast cancer. Eur J Surg Oncol. 26:742–746. 2000. View Article : Google Scholar : PubMed/NCBI

94 

Ladd J, Lu H, Taylor AD, Goodell V, Disis ML and Jiang S: Direct detection of carcinoembryonic antigen autoantibodies in clinical human serum samples using a surface plasmon resonance sensor. Colloids Surf B Biointerfaces. 70:1–6. 2009. View Article : Google Scholar : PubMed/NCBI

95 

Zhu M, Li W, Lu Y, Dong X, Lin B, Chen Y, Zhang X, Guo J and Li M: HBx drives alpha fetoprotein expression to promote initiation of liver cancer stem cells through activating PI3K/AKT signal pathway. Int J Cancer. 140:1346–1355. 2017. View Article : Google Scholar

Related Articles

Journal Cover

June-2017
Volume 50 Issue 6

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ohkawa K, Asakura T, Tsukada Y and Matsuura T: Antibody to human α-fetoprotein inhibits cell growth of human hepatocellular carcinoma cells by resuscitating the PTEN molecule: in vitro experiments. Int J Oncol 50: 2180-2190, 2017
APA
Ohkawa, K., Asakura, T., Tsukada, Y., & Matsuura, T. (2017). Antibody to human α-fetoprotein inhibits cell growth of human hepatocellular carcinoma cells by resuscitating the PTEN molecule: in vitro experiments. International Journal of Oncology, 50, 2180-2190. https://doi.org/10.3892/ijo.2017.3982
MLA
Ohkawa, K., Asakura, T., Tsukada, Y., Matsuura, T."Antibody to human α-fetoprotein inhibits cell growth of human hepatocellular carcinoma cells by resuscitating the PTEN molecule: in vitro experiments". International Journal of Oncology 50.6 (2017): 2180-2190.
Chicago
Ohkawa, K., Asakura, T., Tsukada, Y., Matsuura, T."Antibody to human α-fetoprotein inhibits cell growth of human hepatocellular carcinoma cells by resuscitating the PTEN molecule: in vitro experiments". International Journal of Oncology 50, no. 6 (2017): 2180-2190. https://doi.org/10.3892/ijo.2017.3982