Detailed Description
The present invention is further illustrated by the following examples, which are intended to be purely exemplary of the invention and are not intended to be limiting.
Unless defined otherwise, terms related to technology and science in this specification have the same meaning as commonly understood by one of ordinary skill in the art. Although methods and materials similar or equivalent to those described herein can be used in experimental or practical applications, the materials and methods are described below. In case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.
Cell lines and human clinical samples. Human ovarian cancer cell lines A2780cp (provided by professor Benjamin Tsang of University of Ottawa), OVCA433 (provided by professor George SW Tsao of School of Biological Sciences, HKU), HEY, HEYA8 (provided by professor AWS Wong of School of Biological Sciences, HKU), ES2 cells and HEK293 cells of the human embryonic kidney line were purchased from the American Type Culture Collection (American Type Culture Collection, ATCC, Rockville, Md.). Immortalized Human Ovarian Surface Epithelial (HOSE) cell line HOSE 96-9-18 (provided by professor G.S.W. Tsao of School of biological Sciences, HKU). Human retinal tissue collected from surgical procedures performed at Mary Hospital is approved by the institute of University of Hong Kong Board of the University of Hong Kong/Hospital island network of medicine (HKU/HA HKW IRB) (IRB reference: UW 11-298).
Example 1 purification of native MAP30 and preparation of recombinant MAP30
Chinese dried balsam pear seeds were purchased from local Chinese herbal medicine shops. To purify the dayHowever, MAP30, 200g of dried seeds of Momordica charantia were soaked in double distilled water (ddH)2O) overnight. Seeds were placed in 1.5L ddH using Waring blender2Homogenization in O. The slurry was centrifuged at 30000g twice at 4 ℃ for 20 minutes each. The supernatant was filtered using a double-layer filter paper, and the volume of the filtrate was adjusted to 2L with 20mM Tris-HCl (pH7.6) buffer.
The supernatant was loaded onto an 18cm x 5cm Affi-gel blue gel column (1537301, Bio-Rad) (bed volume 350mL) equilibrated with the same buffer. After loading the sample, the column was washed with 3L of buffer to remove unadsorbed material. The adsorbed material was eluted with 1L of 20mM Tris-HCl (pH7.6) buffer containing 1M NaCl. The adsorbed fraction comprising native MAP30 was purified by ddH in a dialysis tube with a molecular weight cut-off of 8kDa (132665, Spectra/Por) at 4 deg.C2O dialyzed thoroughly overnight.
The dialyzed fraction was adjusted to 20mM ammonium acetate (NH)4OAc) (ph4.5) and loaded onto an 18cm x 5cm SP-agarose column (17-0729-01, GE Healthcare) equilibrated with the same buffer (bed volume: 350 mL). After removal of unadsorbed material with the same buffer, the column was loaded with 1.5L of 20mM NH containing 0.2M NaCl4OAc (pH4.5) buffer to remove loosely bound material. Thereafter, 1L of 20mM NH containing 1M NaCl was used4The OAc (pH4.5) buffer eluted the column, yielding a fraction containing native MAP 30. The fractions were dialyzed (132665, Spectra/Por) in a dialysis tube at 4 ℃ with ddH2O was dialyzed thoroughly overnight, frozen in liquid nitrogen, and lyophilized to powder form with a lyophilizer (Thermo Fisher Scientific).
The dried powder was resuspended to 15mg/mL with 50mM Tris-HCl (pH7.6) buffer containing 0.1M NaCl. FPLC-gel filtration was performed using an AKTA purifier (GE Healthcare) on a Superdex 75HR 10/300 column (17-5174-01, GE Healthcare) pre-equilibrated with the same buffer. By size exclusion, the elution profile exhibited two major absorption peaks at OD 280nm, the first peak being purified native MAP 30. The first peak fraction was collected, dialyzed against centricon (VS15T92, Sartorious) with a molecular weight cut-off of 3kDa and lyophilized to powder form ready for further experiments. SDS-PAGE results with samples from different elution volumes of Superdex 75 showed that the fraction of peak 1 gave a single 30kDa protein band belonging to MAP 30. 40mg of MAP30 could be isolated from 100g of Momordica charantia seeds with this purification method.
Recombinant MAP30 was provided by professor JB Zhan, university of zhejiang. Briefly, the MAP30 cDNA was cloned into the pET-28a expression plasmid (Novagen) and introduced into E.coli BL21(DE3) for expression. Following IPTG (1mM) induction, the bacteria were harvested by centrifugation, followed by sonication, washing and affinity purification using His-tag, recombinant MAP30 could be isolated from the soluble fraction according to the manufacturer's (Novagen) instructions.
Example 2 pharmacological Activity test of MAP30 against ovarian cancer
CRISPR/Cas 9-mediated gene knockout
AMPK α 1 and α 2 gene knockouts were performed by transfection of pL-crispr. efs. gfp plasmid (gifted by professor DG Hardie of Dundee University, UK) carrying sgRNA oligonucleotides for AMPK α 1 and α 2. After transfection, puromycin was used for 24 hours, followed by selection with replacement of fresh medium. Western blot analysis was used to detect positive knockout clones for AMPK α 1 and α 2.
Western blot analysis
Protein lysates were isolated from cells using Cell lysis buffer (Cell Signaling Technology) containing protease inhibitor cocktail (Roche) and phenylmethylsulfonyl fluoride (Sigma Chemical Co.). After separation by 10% SDS-PAGE, the resulting mixture was transferred to a polyvinylidene fluoride (PVDF) membrane. Using enhanced chemiluminescent reagent solution (Amersham)TMECL) were run and visualized by X-ray film.
3. Cell cycle and apoptosis assays
Ovarian cancer cells were stained with 250 μ L of 50 μ g/mL propidium iodide (PI (P1304MP, Invitrogen) in PBS in the dark for 15 min. flow cytometry was performed using BD FACSCAntoTM II System (BD Biosciences.) the proportion of cells per phase of the cell cycle was analyzed using Modfit LT Software (Verity Software House.) to detect apoptosis, treated cells were stained according to the manufacturer's protocol (BD Biosciences) as described in annexin V FITC apoptosis detection kit TDSAfter analysis, the mixture passes through BD FACSVerseseTMSystem (BD biosciences) the percentage of apoptotic cells was analyzed.
Fluo-4 NW calcium and calcineurin cell Activity assay
To assess the amount of intracellular calcium, a Fluo-4 NW calcium assay kit (F36206, Thermo) was used according to the manufacturer's protocol. To determine the phosphatase activity of cellular calcineurin (PP2B), a complete colorimetric assay was performed using the calcineurin cellular activity assay kit (BML-AK816, Enzo).
5. Determination of ATP levels
The amount of ATP in ovarian cancer cells was determined using a luminescent ATP detection assay kit (ab113849, Abcam).
6. Glucose uptake and lactate production
The glucose uptake levels of ovarian cancer cells were determined using a glucose uptake assay kit (colorimetric method) (ab136955, Abcam). To determine lactate production and secretion in ovarian cancer cells, an L-lactate test kit (colorimetric method) (ab65331, Abcam) was used according to the manufacturer's protocol.
7. Determination of hexokinase and Lactate Dehydrogenase (LDH) Activity
The level of hexokinase activity was determined using the hexokinase activity test kit (colorimetry) (ab136957, Abcam) and the level of LDH activity was determined using the LDH test kit (colorimetry) (ab102526, Abcam) according to the manufacturer's instructions.
8. Cell proliferation and lesion formation assay
Cell proliferation was assessed by XTT cell proliferation kit (Roche, Basel, Switzerland). For lesion formation assays, approximately 1000 cells were cultured in each well of a six-well plate and incubated with different treatments. Colonies were stained with crystal violet and counted.
9. Basal Membrane matrix cell migration and invasion assay
The migration and invasion capacity of the cells was examined by transwell cell migration and invasion kit (Corning, NY, USA) according to the manufacturer's instructions. Migrated/invaded cells were stained and counted by microscope.
10. In vivo tumorigenicity test
To investigate the effect of MAP30 on tumor growth in vivo, 2X 10 was used6Each ovarian cancer cell was transplanted intraperitoneally into five female BALB/cAnN-nu nude mice per group, which were four weeks old. When a significant tumor developed after about one week, the experimental group received intraperitoneal injections of MAP30(62.6, 125, 250, and 500. mu.g/kg) once every 2 days. For the control group, PBS was injected intraperitoneally only. Whole animal studies were conducted according to guidelines approved by The Committee of Use of Live Animals (The Committee on The Use of Live Animals in Teaching and Research of The University of Hong Kong) (CULATR No.: 2560-11).
11. Data analysis
All experiments were independently repeated at least three times unless otherwise indicated. Values are expressed as mean ± standard error of mean, and compared using the two-tailed Student's t test. Fisher's exact test (for parametric data) and Mann-Whitney test (for nonparametric data) were used, and P.ltoreq.0.05 was considered statistically significant.
12. Results
(1) MAP30 acts as a natural AMPK activator
Studies of the present invention have shown that natural AMPK activators such as BME and pharmacological AMPK activators a23187, AICAR and metformin inhibit the growth of human ovarian and cervical cancer cells caused by AKT/ERK/FOXM1 and AKT/FOXO3a/FOXM1 signaling, respectively. It has been widely reported that recombinant MAP30 can mediate anticancer effects against human cancers. To test whether there was any difference between recombinant MAP30 and native MAP30 in AMPK activation, Western blot analysis was first performed in the present invention, and the results showed that both types of MAP30(40 μ g/mL) could mediate increased phosphorylation at Thr172 of AMPK and at Ser79 of ACC while decreasing the expression level of FOXM1 and phosphorylation of AKT (Ser473) and ERK in a time-dependent manner (fig. 1A and 1B), indicating that native MAP30 could mediate anticancer effects by inhibiting AKT/ERK/FOXM1 and AKT/FOXO3a/FOXM1 signaling instead of recombinant MAP 30. Furthermore, CRISPR/Cas 9-mediated knockout of AMPK α 1 and α 2 genes in ovarian cancer cell lines such as ES2 cells is robustReduced MAP 30-mediated inhibition of AKT/ERK/FOXM1 signaling activity (FIG. 1C). In addition, knock-out AMPK α 1 α 2 in ES2 cells compared to ES2 control cells treated with MAP30(10 and 40 μ g/mL)-/-Resulting in a 5-fold decrease in cell migration and invasion capacity (fig. 1D).
Unlike common AMPK activators, the present inventors found that MAP30 is similar to BME, amplifying AMPK phosphorylation in WT and R531G cells in a dose and time dependent manner (fig. 1E), suggesting that MAP30 mediated AMPK activation may constitute an AMP independent mechanism. In addition, phosphorylation of AMPK was significantly impaired in response to MAP30 in WT and R531G cells treated with the CaMKK β inhibitor STO-609(10 μ M) (fig. 1F). To further confirm the calcium-dependent pattern of MAP 30-induced AMPK activation in ovarian cancer cell lines, a Fluo-4 NW calcium assay was performed to monitor the cellular calcium concentration. Cellular calcium concentrations in OVCA433 cells were increased in a dose-dependent manner by treatment with MAP30(5, 10, 20, and 40 μ G/mL) compared to controls without MAP30 treatment (fig. 1G). In contrast, knock-out of AMPK α 1 and α 2 significantly reduced the sharp rise in calcium (fig. 1H), confirming a key role for CaMKK β in MAP 30-mediated AMPK activation.
These data support the notion that MAP30, like BME, activates AMPK in inhibiting cell growth, migration/invasion of ovarian cancer cells via CaMKK β signaling, while inhibiting AKT/ERK/FOXM1 transduction.
(2) MAP30 is one of the important components of fructus Momordicae Charantiae extract (BME)
The present invention evaluated the amount of MAP30 in each extract of momordica charantia. Western blot analysis demonstrated the highest level of MAP30 (. about.12 ng/. mu.L) in Thailand (Thai) BME, followed by Chinese BME (. about.9.6 ng/. mu.L) and Taiwan (TW) BME (6 ng/. mu.L) (FIGS. 2A and 2B).
To determine the role of native MAP30 in cell growth, three human ovarian cancer cell lines were treated with different dilutions of native MAP30(5, 10, and 20 μ g/mL). XTT cell proliferation assay functionally demonstrated that the effect of native MAP30 is similar to recombinant MAP30, which can reduce cell viability of ovarian cancer cells by 40% to 80% in a dose-dependent manner compared to its control (fig. 2C). Notably, there was no significant effect on the proliferation of immortalized human ovarian surface epithelial (HOSE 11-12) cells (FIG. 2C). Consistently, the lesion formation assay confirmed that the number of colonies was significantly reduced by about 50% to 90% in a2780cp, OVCA433 and ES2 cells treated with native MAP30(5 and 10 μ g/mL) (fig. 2D). These results indicate that native MAP30 is one of the major bioactive components of BME that exerts a growth-delaying effect on ovarian cancer cells.
(3) MAP30 induces apoptosis and inhibits cancer cell migration and invasion
Similar to AICAR, ovarian cancer cells were treated with MAP30(15 and 45 μ g/mL, 24h), using annexin V-propidium iodide/flow cytometry analysis, which showed a 65% and 150% increase in apoptosis in a dose-dependent manner (fig. 2E). Western blot analysis demonstrated that treatment with MAP30 (45. mu.g/mL) increased cleavage of PARP and caspase 3 in OVCA433 and ES2 cells (FIG. 2F), suggesting that MAP30 induces apoptosis in human ovarian cancer cells through an intrinsic apoptotic mechanism.
Furthermore, the Transwell cell migration assay showed that the cell migration ability of ES2 cells was greatly hindered by MAP30 in a time and concentration dependent manner. After treatment with MAP30(10 and 40. mu.g/mL), there was an approximately 40% to 60% reduction in cell migration (FIG. 2G). Furthermore, the Transwell invasion assay showed that in ES2 cells treated with MAP30(10 and 40 μ G/mL), the number of cells invading through the basement membrane matrix was also reduced by 40% to 60% (fig. 2G).
(4) MAP30 inhibits tumor growth in vivo without causing side effects
In this study, a highly metastatic human ovarian cancer cell line ES2 with a GFP marker was inoculated intraperitoneally into female nude mice. When a significant tumor developed about one week, MAP30(250 μ g/kg) was administered intraperitoneally every 2 days into tumor-bearing mice for a total of 5 injections, and then all mice were sacrificed. The results revealed that tumor-bearing mice treated with MAP30(250 μ g/kg) exhibited about 1.5-fold significant tumor growth and tumor nodule number inhibition compared to the control group (fig. 3A, 3B and 3C). These findings show that MAP30 has a strong in vivo efficacy against human ovarian cancer growth without any effect on mouse body weight (fig. 3D). Interestingly, MAP30 (250. mu.g/kg) treated and control groups both had similar amounts of ascites volume (FIG. 3E), suggesting that there may be a change in the biological composition of ascites between the two groups. Indeed, GC-MS analysis of the target metabolites in ascites was therefore carried out and it was found that the MAP30 treated group showed about a 50% reduction in total fatty acids compared to the control group. Most of them are unsaturated fatty acids, such as C16: 1. c18: 1. c18: 2. c22: 1. c22: 2. c24:1, etc., are important for the biosynthesis and bioenergy requirements of tumor cells (FIGS. 3F and 3G).
Although the studies of the present invention indicate that the MAP30 protein is non-toxic, there is interest in examining the toxicity of the protein in mouse models. Mice treated with an excessively high dose of MAP30(1000 μ g/kg) were found to cause significant weight loss and died even after 1 month (fig. 3H and 3I). However, for mice treated with low dose MAP30(62.5 and 125 μ g/kg) or treated for more than 1 month, no significant changes in body weight were observed, even no death (fig. 3H and 3I). However, biochemical analysis revealed that no changes in the levels of serum creatinine, alanine, transaminase (ALT), aspartate transaminase (AST) and alkaline phosphatase were observed in mice treated with multiple doses (62.5, 125 and 500 μ g/kg) of MAP30 compared to the vehicle control group (fig. 3J), indicating no adverse effects on kidney and liver function, except for a small deviation of AST in the MAP30(500 μ g/kg) treated group, which may have some small liver damage (fig. 3J). However, histopathological studies showed that liver, kidney and spleen structures of mice treated with multiple doses of MAP30(62.5 and 500 μ g/kg) appeared normal compared to the control group (fig. 3K), representing that there was no serious toxic effect of in vivo administration of MAP30, but some unforeseen side effects may occur at higher doses of MAP 30. Therefore, it is necessary to observe the health of mice in the use of MAP30 for a long time.
(5) MAP30 sensitizes ovarian cancer cells to cisplatin-induced cytotoxicity
Given that MAP30 is the major bioactive component of BME, it should sensitize ovarian cancer cells to cisplatin/paclitaxel-induced cytotoxicity. Cell proliferation of ovarian cancer cells was only inhibited by about 50% and 20% by treatment with MAP30 or cisplatin alone (FIG. 4A). However, co-treatment with MAP30 and cisplatin (0.5. mu.g/mL) resulted in at least 75% increase in cytostatic in ovarian cancer cells (FIG. 4A). Furthermore, flow cytometry analysis showed that HEYA8 cells not only caused a significant, dramatic increase in the population of apoptotic cell death in a dose-dependent manner, but also increased the sensitivity of HEYA8 cells to cisplatin-mediated cytotoxicity by 160% and 445% in a dose-dependent manner (fig. 4B).
To further investigate whether MAP30 enhanced cisplatin-induced cytotoxicity in vivo, ES2 tumor-bearing nude mice were injected intraperitoneally with MAP30(125 μ g/kg) and cisplatin (2mg/kg) once every three days for a total of five injections, and then all mice were sacrificed. Consistent with the in vitro data, co-treatment of MAP30 and cisplatin resulted in a significant tumor growth delay in the abdominal cavity of mice, as was a 4-fold reduction in tumor weight and a 2-fold reduction in tumor nodules compared to cisplatin alone (fig. 4C, 4D, and 4G). In addition, mean ascites volume was significantly reduced by cisplatin alone or by co-treatment with MAP30 and cisplatin (fig. 4E), while the body weight of the mice was not severely affected at the end of the experiment (fig. 4F). These findings indicate that MAP30 is not only harmless in vivo, but also helps to inhibit tumor growth by enhancing cisplatin-induced cytotoxicity.
(6) MAP30 blocks the effects of the tumor microenvironment on ovarian cancer cells
Previous studies have shown that the carcinogenic properties of ovarian cancer are enhanced when co-cultured in ascites or Omentum Conditioned Medium (OCM). As expected, the cell growth rate of human ovarian cancer cell lines such as OV2008 and SKOV3 was significantly enhanced by OCM treatment (fig. 5A), suggesting that OCM can stimulate cell growth of ovarian cancer cells and also support oncogenic effects of OCM in the metastatic cascade. However, the XTT cell proliferation assay showed significant reduction in cell proliferation rates of human ovarian cancer cell lines OV2008 (P.ltoreq.0.001) and SKOV3 (P.ltoreq.0.05) cells compared to their OCM control by treatment with MAP30 (10. mu.g/mL) and 1% (v/v) BME (positive control) (FIG. 5A). Moreover, the lesion formation assay showed a significant reduction in the number of colonies in MAP30(10 μ g/mL) treated a2780cp by about 60% compared to OCM control (fig. 5B). Wound healing trials revealed that wound closure rates were significantly accelerated for ovarian cancer cell lines such as SKOV3 and ES2 upon treatment with OCM, resulting in about 30% and 60% increase in cell migration compared to controls, respectively (fig. 5C). Furthermore, Transwell cell migration assays were also performed to better quantify the migration effects of OCM on human ovarian carcinoma cells. Consistent with previous wound healing assays, the data showed an approximately 2.5-fold increase in cell migration rate of SKOV3 cells treated with OCM (OCM control) compared to control medium (DMEM control) (fig. 5D). The Transwell cell invasion assay showed a significant increase in the number of cells invaded by the basement membrane matrix by OCM-cultured ovarian cancer cell line SKOV3 compared to untreated control medium (DMEM control) (fig. 5E). These findings support the notion that OCM has the ability to promote cell migration and cell invasion in human ovarian cancer cells, the stimulatory effects of which are counteracted by the natural AMPK activator MAP 30.
(7) MAP30 inhibits glycolysis and de novo lipogenesis in ovarian cancer cells
In this study, the L-lactate assay consistently demonstrated that lactate production was higher in various ovarian cancer cell lines than in immortalized human ovarian surface epithelial (HOSE 96) cells during two days of culture (fig. 6A). Such enhanced lactate production in ovarian cancer cells is a significant feature that is attached to the treatment of the native AMPK activator MAP30(40 μ g/mL) and the positive controls AICAR (1mM) and a23187(2 μ M) (fig. 6B), suggesting that a reduction in glycolysis occurs in ovarian cancer cells via MAP30 mediated AMPK activation. Such a decrease in glycolysis is limited by the activity of glycolytic enzymes such as hexokinase and LDH, since the hexokinase and LDH activity of OVCA433 cells was significantly inhibited by more than 50% in a time and dose dependent manner upon MAP30 treatment for 24 and 48 hours (fig. 6C and 6D). Importantly, the net reduction in AMPK-activated glycolysis mediated by MAP30 contributes to the reduction in carbon flux. For this purpose, glucose uptake assays were performed, with MAP30(10 and 40 μ g/mL) behaving similarly to the drug AMPK activator AICAR (1mM), inhibiting glucose uptake of approximately 30% to 60% in OVCA433 cells in a dose-dependent manner. Western blot analysis further confirmed that expression of GLUT1 and GLUT3 was significantly decreased in a time-dependent manner in OVCA433 and ES2 cells treated with MAP30(40 μ g/mL) (fig. 6F). This data indicates that MAP30 is able to inhibit glucose uptake by reducing the GLUT-1 and-3 transporters. To further investigate the role of MAP30 in the glucose metabolism kinetics of tumor growth in vivo, 18F-FDG Micro-PET/CT was performed.
Since the omentum is considered to be the preferred site for ovarian cancer metastasis, it is postulated that lipids specifically regulate ovarian cancer cells in the omentum microenvironment to promote malignant cell survival and metastatic cancer cell spread. In the present invention, OCM-cultured ovarian cancer cell line OVCA433 showed increased lipid droplet formation using Nile Red fluorescent staining, while MAP30 co-treatment (5 and 10. mu.g/mL) significantly reduced lipid droplet numbers by 50-70% (FIG. 6G). To further determine whether MAP30 accelerates catabolic lipolysis in human ovarian cancer cells, lipolysis assays were performed showing that MAP30(10 and 40 μ g/mL) induced an approximately 2-4 fold increase in cellular lipolysis in ovarian cancer cells (fig. 6H). Moreover, ATP detection assays revealed a significant reduction in ATP production in ovarian cancer cells due to MAP30(10 and 40 μ g/mL) treatment (fig. 6I). This is understood as the activation of AMPK promotes catabolism and inhibits anabolism to conserve cellular energy. The findings of the present invention together indicate that MAP30 mediated AMPK activation triggers a series of catabolic processes in the metabolic profile of ovarian cancer cells, including decreased lipid synthesis from surrounding sources and enhanced fatty acid oxidation even in the fatty acid-enriched ascites microenvironment.
(8) The natural MAP30 protein is superior to the recombinant MAP30 protein
Although we found that recombinant and native MAP30 were similar in size (-30 kDa) (see fig. 7) and both had a role in AMPK activation and anti-cancer, the post-translational modification of native MAP30 had other functional roles not possessed by recombinant MAP30 protein. Preliminary data have shown that native MAP30 shows about 10-30% greater inhibition of cell proliferation of ovarian cancer cells compared to recombinant MAP30 (see figure 8). Furthermore, the toxicity of the recombinant protein may reduce the safety of applying MAP30 in cancer therapy.
The natural MAP30 protein consists of 286 amino acids, has a molecular weight of 30kDa, and has an amino acid sequence shown in SEQ ID NO. 1. Wherein the N-terminal MVKCLLLSFLIIAIFIGVPTAKG is a signal peptide which is removed upon secretion of the mature MAP30 protein from plant cells (Lee-Huang S.et al. FEBS Lett.272:12-18 (1990)). The recombinant MAP30 protein consists of 284 amino acids, the amino acid sequence of the recombinant MAP30 protein is shown in SEQ ID NO.2 (ACCESSION AAB35194), the calculated molecular weight is 31.9kDa, and the estimated pI is 9.0. In the recombinant MAP30 protein, the above-mentioned signal peptide sequence was replaced by a histidine-tag (His-tag), which did not affect the functional effects of MAP 30. However, native MAP30 showed a stronger AMPK activation effect (see fig. 9), and a stronger cell growth delaying effect. Western Blot analysis showed the difference (. about.2-5 kDa) between native MAP30 protein and recombinant MAP30 protein at the N-terminus (see FIGS. 7 and 10). In addition to the differences in protein length, a glycosylation site was present at amino acid 74 of MAP30 protein. In native MAP30 protein, this site was glycosylated, but in the recombinant MAP30 protein produced by e.coli, it was not glycosylated, as e.coli does not have a protein glycosylation system. Translational modification of the MAP30 protein resulted in a functional difference between the native MAP30 protein and the recombinant MAP30 protein. In conclusion, the differences in protein structure and glycosylation between the native MAP30 protein and the recombinant MAP30 protein resulted in differences in cytotoxicity and AMPK activation in ovarian cancer cells. The differences in protein structure are the main reason why the native MAP30 protein has better AMPK activation and other cytotoxic effects on ovarian cancer cells.
In addition, the rapid production of recombinant MAP30 protein also caused some toxicity due to the accumulation of protein aggregates.
(9) Discussion of the related Art
Acquired chemotherapy resistance is currently a major obstacle in the clinical management of ovarian cancer. This has led to an urgent need to find alternative treatment regimens or new pharmaceutical agents. Chinese medicine has been widely used in ancient China and India for the treatment of human diseases including cancer. It not only targets cancer cells, but also treats the entire body, such as the gas, blood and immune system. Today, traditional Chinese medicine is used for the purpose of supplementing alternative medicines. Bioactive compounds from chinese medicine have interesting functions in anticancer by modulating cancer cells at multiple levels and multiple targets. This makes the traditional Chinese medicine and its bioactive compounds potentially useful applications as ideal adjuvants in modern cancer therapy. Bitter gourd extract (BME) has previously been reported to exert inhibitory effects on ovarian Cancer cells by activating AMPK/mTOR signaling (Yung MM et al. integer Cancer ther. 2016Sep; 15(3): 376-89.). When co-administered with cisplatin, BME may exhibit a synergistic effect that enhances cisplatin-mediated cytotoxicity in ovarian cancer cells. In this study, MAP30 protein was isolated from dry seeds of momordica charantia. Functional and biochemical analysis showed MAP30 to be one of the major bioactive components in BME. The present invention was able to demonstrate that different functional anticancer effects are positively correlated with the amount of MAP30 in each BME.
Indeed, BME comprises a variety of bioactive compounds and has been shown to have anti-cancer effects against a variety of human cancers. For example, Kuguacin J (KJ) isolated from an extract of momordica charantia has been reported to sensitize ovarian cancer cells to paclitaxel resistance. In addition, the ribonuclease RNase MC2 can induce apoptotic mediators in apoptosis that cause human breast cancer. Even for biologically inactive compounds, a novel peptide BG-4 from Momordica charantia has been shown to promote apoptosis in human colon cancer. However, these components did not show any relationship to or function of AMPK. It is generally known that momordica charantia has been used in ancient china and india for the treatment of diabetes, similar to metformin isolated from goat beans for use as a first line treatment for diabetes. This suggests that the main function of momordica charantia is AMPK activator. Coincidently, this MAP30 protein has been shown to act as a natural AMPK activator, inducing AMPK activation in an AMP-dependent manner through the CaMKK β pathway and inhibiting mTOR signaling, inhibiting the newly discovered signaling AKT/ERK/FOXM1, showing inhibitory effects on ovarian cancer cells.
AMPK is not only a key energy sensor in normal cells, but is also a crucial factor in tumor development. AMPK is known to be a key sensor for maintaining cellular energy balance regulating multiple cellular metabolic pathways. Emerging evidence has shown that malignant cells require high AMPK activity to initially overcome metabolic stress from the tumor microenvironment, while advanced cancers prefer low AMPK activity to enhance cell growth and progression. This suggests that late stage and more aggressive cancer cells prefer low AMPK activity to reduce metabolic limitations, such as mTOR, for exerting high carcinogenicity. On the other hand, this confers an advantage to AMPK activators in selectively disrupting ovarian cancer cell function. The ability of native MAP30 isolated from non-toxic momordica charantia to increase AMPK activity through the CaMKK β pathway and in an AMP-independent manner suggests that it targets cancer cells from another perspective whose survival and other cellular activities are not affected by AMP stress conditions, such as a nutrient-rich microenvironment. Indeed, the present study shows that the native MAP30 can significantly inhibit tumor growth, particularly tumor colonization in tumor xenograft mouse models, as well as cell growth, cell migration/invasion. mTOR signaling is generally known to be an important pathway in cellular metabolism, growth, proliferation, survival and resistance to drugs. However, mTOR inhibitors are not sufficient to achieve a broad and potent anticancer and anti-chemotherapeutic drug resistance effect. MAP30, however, appears to offset this deficiency. The findings of the present invention show that MAP30 may not only inhibit oncogenic AKT/ERK/FOXM1 signaling, but also regulate cellular metabolism, such as reducing glucose uptake by inhibiting GLUT-1 and GLUT-3 expression, as well as lipid droplet formation, possibly even affecting the resistant lipolysis of ovarian cancer cells in a nutrient-free microenvironment. To further demonstrate these in vivo effects, treatment of tumor-bearing mice with MAP30 will be performed by the 18F-FDG Micro-PET/CT assay used to study the glucose uptake capacity of tumor cells in the mice, as is being done. The discovery of the present invention can provide a scientific basis for its potential use in combating the chemotherapeutic resistance of ovarian cancer cells to existing chemotherapeutic drugs.
And (4) conclusion: the findings of the present invention indicate that MAP30 protein is one of the major bioactive components of momordica charantia, exerts a potent inhibitory effect on the carcinogenic properties of ovarian cancer cells by activating CaMKK β/AMPK signaling, and inhibits AKT/ERK/FOXM1 signaling activity as well as regulates glycolysis and adipogenesis. The bioactive protein has the potential to develop into a chemotherapeutic supplement for ovarian cancer.
Example 3 MAP30 protein from Momordica charantia has therapeutic effects on ovarian cancer in vivo by altering metabolism and inducing apoptosis of iron and has synergistic activity with cisplatin
A comprehensive study was conducted to characterize the effect of native MAP30 isolated from momordica charantia seeds on ovarian cancer cells. The use of Omentum Conditioned Medium (OCM) and mouse tumor xenograft model demonstrated that non-toxic MAP30 in combination with cisplatin exhibited potent anticancer and chemoresistance effects on ovarian cancer cells. These findings indicate that MAP30 can be used as a supplement to augment the therapeutic outcome of existing platinum-based chemotherapy regimens.
The material and the method are as follows:
1. cell lines and human clinical samples
The human ovarian cancer cell line a2780cp was provided by professor Benjamin Tsang at ottawa university, canada. HEY and HEYA8 human ovarian carcinoma cells were provided by professor AWS Wong of the institute of bioscience, hong kong university. ES2 human epithelial ovarian cancer cells and the human embryonic kidney cell line HEK293 were purchased from American type culture Collection (ATCC, Manassas, Va., USA). Additional human ovarian cancer cell lines OVCA433 and SKOV3, as well as two immortalized Human Ovarian Surface Epithelial (HOSE) cell lines HOSE11-12 and HOSE 96-9-18 were provided by professor G.S.W.Tsao of the biomedical college of hong Kong university. Professor DG Hardie, university of england dundi, provided a pair of WT and R531G AMPK γ 2 isogenic HEK293 cells. All cell lines were mycoplasma free. Human omentum tissue was collected from procedures performed in Mary Hospital (IRS reference: UW 11-298) with prior approval by the agency review Committee of the hong Kong university/Hospital administration, hong Kong Western networking (HKU/HA HKW IRS).
2. Purification of native MAP30 from dried seeds of bitter gourd
Natural MAP30 was isolated from dried seeds of Momordica charantia (Momordica charantia L.) Kunth.
CRISPR/Cas 9-mediated gene knockout
Knock-out of AMPK α 1 and α 2 genes was performed by transfection of the pL-crispr. efs. gfp plasmid (provided by professor DG Hardie, university of dungdi, england) carrying sgRNA oligonucleotides for AMPK α 1 and α 2. Following transfection, puromycin was used for selection. Immunoblot analysis was used to detect positive knockout clones for AMPK α 1 and α 2.
4. Immunoblot analysis
Using a mixture containing protease inhibitors (Roche, Basel, Switzerland) and benzylCell lysis buffer (Cell Signaling Technology, Danvers, MA, USA) of sulfonyl fluoride (Sigma-Aldrich corp., st. louis, MO, USA) protein lysates were isolated from cells. Proteins were separated using 10% SDS-PAGE and then transferred to polyvinylidene fluoride (PVDF) membranes. Immunoassays using enhanced chemiluminescent reagent solutions and visualisation on X-ray film, or by
CLx Imaging was performed using Image studio software (Li-Cor Biosciences) for analysis (LI-COR Biosciences, Lincoln, Nebraska, USA).
5. Cell cycle and apoptosis assays
Ovarian cancer cells were stained with 250 μ L of 50 μ g/mL PBS solution of Propidium Iodide (PI) (P1304MP, Invitrogen) in the dark for 15 minutes. Flow cytometry was performed using the BD FACSCAntoTM II system (BD Biosciences, San Jose, Calif., USA). The proportion of cells at each stage of the cell cycle was analyzed using Modfit LT Software (Verity Software House, Topsham, ME, USA). To detect apoptosis, the treated cells were stained using Annexin V FITC apoptosis detection kit TDS according to the manufacturer's protocol (BD Biosciences). After flow cytometry, BD FACSVerse was usedTMThe system (BD Biosciences) analyzed the percentage of apoptotic cells. To examine the level of activated caspase-3 and-7 in ovarian cancer cells after MAP30 treatment, treated cells in 48-well microplates (CLS3548, Corning, NY, USA) were analyzed using CellEventTM caspase 3/7 green detection reagent (C10423, Invitrogen) according to the manufacturer's protocol.
6. Lipid peroxidation assay and determination of GSH/GSSG Activity
To evaluate lipid peroxidation or iron apoptosis (ferroposis) of ovarian cancer cells co-treated for 24 hours with MAP30 (40. mu.g/mL), and either Erastin (5. mu.M) alone (B1524, APExBIO Technology LLC., Boston, MA, USA), or the iron apoptosis inhibitor Ferrostatin-1(Fer-1) (10. mu.M) (SML0583, Sigma-Aldrich, St. Louis, MI, USA) in combination with MAP30 (40. mu.g/mL), C11 BODIPY 581/591C11 dye (lipid DIP 581/591C11 dye (lipid apoptosis) was usedMass peroxidation sensor) (2.5. mu.M) (D3861, ThermoFisher Scientific, Waltham, MA, USA). Using BD FACSLricTMThe flow cytometry system (BD Biosciences, San Jose, CA, USA) performed flow cytometry analysis, analyzing at least 10000 cells per cell sample. Oxidation of C11 BODIPY 581/591 was indicated by an increase in green fluorescence intensity in the FITC channel (which shows oxidized probe). Data were analyzed using FlowJo version 7.6 software. For confocal imaging experiments, each experimental group was examined for treated ovarian cancer cells and imaged using a confocal imaging system (Carl Zeiss LSM 800). Three independent biological replicates were performed for each condition according to the manufacturer's protocol.
Changes in the GSH/GSSG ratio of ovarian cancer cells following co-treatment with MAP30 and/or Erastin were detected using GSH/GSSG ratio detection assay kit II (fluorescent-green) (ab205811, Abcam, Cambridge, MA, USA) according to the manufacturer's instructions.
Fluo-4 NW calcium assay
To assess the amount of intracellular calcium, a Fluo-4 NW calcium assay kit (F36206, Thermo) was used according to the manufacturer's protocol.
Determination of ATP levels and Reactive Oxygen Species (ROS) production
The amount of ATP in ovarian cancer cells was evaluated using a luminescent ATP detection assay kit (ab113849, Abcam). Cellular ROS production was assessed using the DCFDA-Cellular ROS assay (ab113851, Abcam) according to the manufacturer's protocol.
9. Glucose uptake and lactate production
The level of glucose uptake in ovarian cancer cells was measured using a glucose uptake assay kit (colorimetric) (ab136955, Abcam). To measure lactate production in and lactate secretion from ovarian cancer cells, an L-lactate test kit (colorimetric method) (ab65331, Abcam) was used according to the manufacturer's protocol.
10. Determination of hexokinase and Lactate Dehydrogenase (LDH) Activity
Hexokinase activity was measured using the hexokinase activity test kit (colorimetry) (ab136957, Abcam) and LDH activity was measured using the LDH test kit (colorimetry) (ab102526, Abcam) according to the manufacturer's instructions.
11. Relative quantification of target metabolites of fatty acids
Target metabolites in ascites were analyzed using gas chromatography-mass spectrometry (GC-MS) at the LKS medical institute proteomics and metabonomics Core laboratory (CPOS-PM Core) at Hong Kong University (HKU). GC/MS chromatograms were acquired in SCAN and SIM mode using an Agilent 7890B GC-Agilent 7010 triple quadrupole mass spectrometer system. Data analysis was performed using Agilent MassHunter workstation quantitative analysis software (CPOS-PM Core, HKU).
12. Cell proliferation and lesion formation assay
Cell proliferation was assessed using the XTT cell proliferation kit (Roche, Basel, Switzerland). For lesion formation assays, approximately 1000 cells were cultured in each well of a six-well plate and incubated with different treatments. Colonies were stained with crystal violet and counted.
13.CellTiter-
3D cell viability assay
Suspending ovarian cancer cells in a suspension containing 2% Geltrex
TMMedia of matrix (Gibco BRL, Gaithersburg, Md., USA) and seeded in triplicate at a density of about 1000 cells per well in Nunclon with ultra low cell attachment surface
TMSphera
TM96U-microwell plates (Thermo Fisher Scientific). The cells were subsequently grown for one week and then treated for three days with serially diluted concentrations of each drug alone (MAP30 and cisplatin) or a combination at a fixed molar ratio. After drug treatment, CellTiter-
The 3D cell viability assay detects viable cells. IC50 values were determined for each drug using non-linear regression, and drug synergy was determined using the Combenefit algorithm.
14. Matrigel cell migration and invasion assay
The migration and invasion capacity of cells was examined using the Transwell cell migration and invasion kit (Corning, NY, USA) according to the manufacturer's instructions. Migrated/invaded cells were stained and counted using a microscope.
15. In vivo tumorigenicity test
To investigate the effect of MAP30 on tumor growth in vivo, 2X 10 was used6Each ES2 ovarian carcinoma cell was injected intraperitoneally (i.p.) into a group of five four-week-old BALB/cAnN-nu female nude mice. When a significant tumor was formed within about one week, the experimental group was treated every 2 days by injection of MAP30(5 μ g). Control group was intraperitoneally injected with PBS only. Fluorescence stereomicroscopy (Nikon, japan) was used to capture fluorescence and bright field images of tumor nodules scattered within the peritoneal cavity of mice. The entire animal study was conducted according to guidelines approved by the Committee for use of live animals in the university of hong Kong (CULATR No. 3817-15).
16. Data analysis
All experiments were repeated at least three times independently unless otherwise stated. Values are expressed as mean ± SEM and compared using two-tailed student's t-test. Fisher's exact test (for parametric data) and Mann-Whitney test (for nonparametric data) were used, and P.ltoreq.0.05 was considered statistically significant.
As a result:
MAP30 is the main component of Momordica charantia extract (BME)
BME is a natural AMPK activator that inhibits growth, cell migration, and invasion of ovarian cancer cells. It was observed that BME from different sources can activate AMPK to different extents, suggesting a dose-dependent effect of MAP 30. Monoclonal MAP30 antibody was used to demonstrate the highest MAP30 levels (-12 ng/. mu.l) in thailand (Thai) BME, followed by china BME (-9.6 ng/. mu.l) and chinese Taiwan (TW) BME (6 ng/. mu.l) (fig. S1A and S1B), indicating that the different levels of MAP30 from three varieties of momordica charantia are completely consistent with their ability to activate AMPK in ovarian cancer cells.
Functional studies showed that MAP30 inhibited cell viability of ovarian cancer cells (a2780cp, OVCA433 and ES2) by 40% to 80% in a dose-dependent manner compared to controls using XTT cell proliferation assays (fig. 1A). Notably, cell proliferation of immortalized human ovarian surface epithelial (HOSE 11-12) cells was not significantly inhibited (FIG. 11A). Lesion formation analysis showed that the number of colonies in ovarian cancer cells (a2780cp, OVCA433 and ES2) was significantly reduced from 50% to 90% after treatment with MAP30(5 and 10 μ g/mL) compared to controls (fig. 11B). To test whether MAP30 inhibited migration and invasion of ovarian cancer cells, ES2 ovarian cancer cells were treated with MAP30(15 and 40 μ g/mL) for 15 and 24 hours and subjected to a Transwell cell migration/invasion assay. Quantitative analysis showed that MAP30(15 and 40 μ g/mL) treatment for 15 and 24h reduced the cell migration and cell invasion capacity of ES2 cells by 40-70% in a dose and time dependent manner (fig. 11C). These tumor suppressive effects were also found in ovarian cancer cells treated with BME. Based on the functional similarity of BME in ovarian cancer cells and MAP30 content response, MAP30 is the main bioactive component of BME. MAP30 exhibited selective cytotoxicity and inhibition of ovarian cancer cells, but no cytotoxicity to normal ovarian epithelial cells.
MAP30 triggering cell cycle arrest and apoptosis in ovarian cancer cells
To describe the basic molecular mechanism by which MAP30 inhibits tumor growth, the effect of MAP30 on cell cycle progression of ovarian cancer cells was investigated using flow cytometry analysis of DNA content. Cell cycle analysis showed that MAP30 caused a clear arrest in S phase in various ovarian cancer cells with highly variable levels, unlike AICAR-induced cell cycle arrest in G1 phase (fig. S2). MAP30 is the major component of BME and it may have a similar functional role as BME in AMPK-mediated cell growth and apoptosis. Thus, it was examined whether MAP30 functions like other AMPK activators (such as AICAR) in causing apoptosis in ovarian cancer cells. AICAR (1mM, 24h) was used as a positive control. OVCA433 ovarian cancer cells treated with MAP30(10 and 40 μ g/mL, 24h) showed a dose-dependent increase in the rate of apoptosis of 2 to 3 fold compared to the negative control (fig. 11D). The CellEventTM caspase 3/7 green assay revealed that MAP30 induced mitochondria-mediated caspase activation in ES2 following co-treatment with MAP30 and cisplatin (figure 21). Immunoblot analysis further demonstrated that treatment with MAP30(40 μ g/mL) increased cleavage and caspase 3 activity of PARP in OVCA433 and ES2 cells (fig. 11E). These results indicate that MAP30 induces cell cycle S-phase arrest and apoptosis in ovarian cancer cells.
MAP30 acting as a natural AMPK activator
The study shows that BME is a natural AMPK activator, and remarkably inhibits the carcinogenic capacity of ovarian cancer and cervical cancer by inhibiting AKT/ERK/FOXM1 and AKT/FOXO3a/FOXM1 signaling pathways respectively. This study compared the functional differences between native MAP30 (isolated from dried bitter gourd seeds) and recombinant MAP30 protein (produced by e. Immunoblot analysis showed that native MAP30 protein (40 μ g/mL) and recombinant MAP30 protein (30 μ g/mL) significantly increased phosphorylation at Thr172 of AMPK and Ser79 of ACC, but decreased levels of FOXM1 and phosphorylation of AKT and ERK (Ser473) in a time-dependent manner (fig. 12A and 12B). Considering that both recombinant MAP30(30 μ g/mL) and native MAP30(40mg/mL) can activate AMPK, they were also shown to inhibit mTOR signaling activity in another AMPK downstream target, i.e. ovarian cancer cells (fig. 12C). This finding confirms that native and recombinant MAP30 exhibit anti-cancer effects on ovarian cancer cells through negative regulation of the AKT/ERK/FOXM1 and mTOR signaling cascades. Notably, the role of these proteins is consistent with the functional effects of BME on ovarian and cervical cancer cells. To further demonstrate the down-regulation of the AKT/ERK/FOXM1 signaling cascade and its associated oncogenic properties, a CRISPR/Cas9 gene knockout system was used to delete AMPK- α 1 and- α 2(AMPK α 1 α 2) in ovarian cancer cells-/-). As a result, it was confirmed that AMPK-. alpha.1 and-. alpha.2 (AMPK. alpha.1. alpha.2)-/-) The complete depletion significantly attenuated MAP 30-mediated inhibition of the AKT/ERK/FOXM1 signaling cascade (fig. 12D) and resulted in an approximately 2-fold decrease in the cell migration and invasion capacity of ovarian cancer cells (fig. 12E). These results indicate that inhibition of MAP30 occurs in an AMPK-dependent manner.
Unlike most drug AMPK activators, MAP30 functions similarly to BME, in that both factors are dose-sumTime-dependent increases phosphorylation of AMPK in WT and R531G AMPK γ 2 isogenic HEK293 cells (fig. 12F). These results indicate that MAP 30-mediated AMPK activation may be involved in AMP-independent mechanisms. Treatment with the CaMKK β inhibitor STO-609(10 μ M) significantly attenuated phosphorylation of AMPK in WT and R531G HEK293 cells in response to MAP30 (fig. 12G). To further confirm the calcium-dependent pattern of MAP 30-induced AMPK activation in ovarian cancer cells, a Fluo-4 NW calcium assay was used. The results show calcium ion (Ca) in OVCA433 cells compared to controls co-treated with MAP302) Was increased dose-dependently (fig. 12H). In contrast, CRISPR/Cas 9-mediated gene knock-out of AMPK α 1 and α 2(AMPK α 1 α 2)-/-) Reduce intracellular Ca2+Increase (fig. 12I), which confirms a key role of CaMKK β in MAP 30-mediated AMPK activation.
Taken together, these data support that MAP30 is the major component of BME, activating AMPK and inhibiting the AKT/ERK/FOXM1 and mTOR pathways via the CaMKK β pathway, as well as the associated cell growth, cell migration/invasion of ovarian cancer cells.
MAP30 Induction of iron apoptosis in ovarian cancer cells
MAP30 causes intracellular Ca in ovarian cancer cells2+Increase in (FIG. 12G), and Ca2+May increase cytoplasmic oxidative stress and mitochondrial dysfunction. Excessive cytosolic Reactive Oxygen Species (ROS) may lead to lipid peroxidation and iron apoptosis, which are alternatives to programmed cell death. To investigate whether MAP30 caused iron apoptosis, it was investigated whether MAP30 enhanced ROS production in ovarian cancer cells. MAP30 was compared to the iron apoptosis activator Erastin and it was observed that MAP30 increased intracellular ROS production in ovarian cancer cells in a dose-dependent manner (fig. 13A). Notably, MAP30 is at AMPK α 1 α 2-/-Less ROS production was induced in ovarian cancer cells (fig. 13A). The reduced ratio of glutathione to oxidized glutathione (GSH/GSSG) is the central cellular antioxidant system that prevents oxidative damage. To investigate whether MAP30 reduced the GSH/GSSG ratio during ROS elevation like Erastin, G was measured in ovarian cancer cells co-treated with MAP30 and ErastinSH/GSSG ratio. MAP30 significantly reduced the GSH/GSSG ratio in a dose-dependent manner and was shown to be negatively correlated with ROS levels (fig. 13B). In AMPK alpha 1 alpha 2-/-In ovarian cancer cells, the effect of MAP30 on GSH/GSSG ratio was reduced, but there was no difference in the effect of Erastin (fig. 13B). Studies have shown that Erastin prevents cellular cysteine uptake by inhibiting the cystine/glutamate reverse transporter (xCT) and glutathione peroxidase 4(GPX4), while GSH depletion induces lipid peroxidation and iron apoptosis. Thus, the effect of MAP30 on GPX4 and Erastin was compared. Immunoblot analysis showed that MAP30 was present in wild type and AMPK α 1 α 2-/-GPX4 protein was reduced in a dose-dependent manner in ovarian cancer cells similar to Erastin. By using BODIPYTM581/591C11 as a lipid peroxidation probe demonstrated that MAP30 induces lipid peroxidation in ovarian cancer cells and that a potent inhibitor of apoptosis, Ferrostatin-1(Fer-1), inhibited this effect (FIGS. 13D and 13E). Because of the MAP30 vs. AMPK α 1 α 2-/-The effect of ROS production and GSH/GSSG ratio depletion in ovarian cancer cells was minor, so MAP 30-mediated lipid peroxidation in these cells was studied. Flow cytometry analysis revealed that MAP30 significantly induced lipid peroxidation, which was counteracted by Fer-1 in ovarian cancer cells (fig. 13F). However, MAP30 is only present in AMPK α 1 α 2-/-Lipid peroxidation was slightly induced in ovarian cancer cells (fig. 13F). These findings confirm that MAP30 is an inducer of iron apoptosis in ovarian cancer cells. However, the mechanism by which loss of AMPK activity reduces the efficiency of MAP30 in inducing iron apoptosis is not clear. Further studies were necessary to elucidate the functional role of AMPK in MAP 30-induced iron apoptosis.
MAP30 inhibits tumor growth in vivo without adverse effects
A highly metastatic human ovarian cancer cell line ES2, labeled with Green Fluorescent Protein (GFP), was inoculated intraperitoneally into female nude mice. After a significant tumor occurred within about one week, MAP30 (250. mu.g/kg) was injected intraperitoneally into tumor-bearing mice every other day for a total of 5 injections, and then sacrificed. Tumor-bearing mice treated with MAP30 showed significant tumor growth inhibition with an approximately 1.5-fold reduction in tumor nodule numbers compared to the control group (fig. 14A, 14B and 14C). These findings indicate that MAP30 has a potent in vivo efficacy against human ovarian cancer growth without any adverse effect on mouse body weight (fig. 14D). Notably, similar ascites volumes were found in the MAP30(250 μ g/kg) treated group and the control group (fig. 14E), indicating a change in the biological composition of ascites between the two groups. Thus, GC-MS analysis of the target metabolites in ascites was performed, showing about 50% reduction in total fatty acids for the MAP30 treated group compared to the control group. Most fatty acids were unsaturated fatty acids, such as C16:1, C18:1, C18:2, C22:1, C22:2, and C24:1, which meet the basic biosynthetic and bioenergy requirements of tumor cells (fig. 14F and 14G).
Although the study showed that the MAP30 protein is non-toxic, the toxicity of the protein was examined in a mouse model. Mice treated with high dose of MAP30 (500. mu.g/kg) showed approximately 10% weight loss (FIGS. 14H and 14I). However, mice treated with lower doses of MAP30(62.5 and 125 μ g/kg) for more than 1 month showed no significant change in body weight, and no death was observed (fig. 14H and 14I). Biochemical analysis revealed no observable changes in serum creatinine, alanine Aminotransferase (ALT), aspartate Aminotransferase (AST) or alkaline phosphatase levels in mice treated with 62.5 and 125 μ g/kg MAP30 compared to vehicle control (fig. 14J), indicating no adverse effects on kidney and liver function. A small deviation in AST was observed with high dose MAP30(500 μ g/kg) treatment, indicating some mild liver injury (fig. 14J). However, histopathology showed that liver, kidney and spleen structures of mice treated with low and high doses of MAP30(62.5 and 500 μ g/kg, respectively) were similar to the control group (fig. 14K), indicating that there was no serious toxic effect of MAP30 administration in vivo. However, at high doses MAP30 may have some unexpected side effects. Therefore, it is necessary to study the long-term toxicity of MAP30 in mice.
Synergistic enhancement of cisplatin-mediated cytotoxicity of MAP30 in vitro
Since MAP30 induced apoptosis and iron apoptosis in ovarian cancer cells, it was examined whether MAP30 sensitizes ovarian cancer cells to cisplatin/paclitaxel-induced cytotoxicity. Three advanced ovarian cancer cell lines were co-treated with different concentrations of cisplatin and/or MAP30 using a 2D culture system. Treatment with MAP30 alone or cisplatin alone inhibited cell proliferation in ovarian cancer cells by 50% and about 20%, respectively (fig. 15A). However, co-treatment with MAP30 and cisplatin (0.5. mu.g/mL) increased the cytostatic rate of ovarian cancer cells to at least 75% (FIG. 15A). Flow cytometry analysis showed a significant and dose-dependent increase in HEYA8 cell population undergoing apoptotic death, with MAP30 increasing cisplatin-mediated cytotoxicity by about 1.6-4.5 fold in a dose-dependent manner compared to controls (fig. 15B).
Increasing evidence notes various limitations of the 2D system in testing cancer cells for chemoresistance, including its poor mimic in vivo on the tumor microenvironment. Recently, in vitro three-dimensional culture systems (3D systems) have been developed that replicate the in vivo tumor microenvironment and more accurately represent drug responses. Therefore, it was further investigated whether MAP30 synergistically enhanced cisplatin cytotoxicity using a 3D culture system. CellTiter-
3D cell viability analysis for assessment at 2% Geltrex
TMSensitivity of 3D spherical ovarian cancer cells cultured in stroma (Gibco BRL) to native MAP30 and/or cisplatin treatment. MAP30 or cisplatin treatment alone produced a dose-dependent inhibitory effect on cell viability of OVCA433 (fig. 15C) and HEY spheroids (fig. 22). Next, it was determined whether co-treatment with MAP30 and cisplatin enhanced the therapeutic effect compared to each drug alone. Ovarian cancer cell lines, such as OVCA433 and HEY cells, were treated with MAP30 and cisplatin in a fixed molar ratio at serially diluted concentrations. The combination of the two drugs induced significantly more significant slowing of cell viability compared to the individual treatments in each of OVCA433 (fig. 15C) and HEY cells (fig. 22). The effect of the drug combination was determined using the Combenefit algorithm and the synergy matrix showed that the dual inhibition produced excellent synergistic anti-cancer effects on OVCA433 (fig. 15D) and HEY cells (fig. 22). These data indicate that MAP30 synergistically enhances cisplatin cytotoxicity in ovarian cancer cells.
MAP30 enhances cisplatin-induced cytotoxicity in vivo
To further investigate whether MAP30 enhances cisplatin-induced cytotoxicity in vivo, ES2 tumor-bearing nude mice were injected intraperitoneally with MAP30 (125. mu.g/kg) and cisplatin (2mg/kg) once every three days for five total injections, and then sacrificed. Consistent with the results of the in vitro study, co-treatment with MAP30 and cisplatin resulted in a significant reduction in tumor growth, as indicated by a 4-fold reduction in tumor weight (fig. 16A and 16B) and a 2-fold reduction in tumor nodules (fig. 16A and 16B) compared to cisplatin alone treatment. At the end of the experiment, treatment with cisplatin alone or with MAP30 in combination with cisplatin significantly reduced the mean ascites volume (fig. 16D) without significantly affecting the body weight of the mice (fig. 16E). These findings confirm that MAP30 is harmless in vivo and helps to inhibit tumor growth by enhancing cisplatin-induced cytotoxicity.
MAP30 inhibits glycolysis and de novo fat synthesis in ovarian cancer cells
There is increasing evidence that AMPK is a metabolic tumor suppressor that controls tumor development and progression. This study investigated whether MAP30 alters tumor metabolism to achieve its inhibitory effect on the oncogenic properties of ovarian cancer. L-lactic acid assay analysis showed that ovarian cancer cells typically produced relatively higher levels of lactic acid than immortalized human ovarian surface epithelial control cells throughout two days of culture (FIG. 17A). This increased lactate production in ovarian cancer cells was clearly offset by positive control treatment with the native AMPK activator MAP30(40 μ g/mL) and with drug AMPK activators such as AICAR (1mM) and a23187(2 μ M) (fig. 17B), indicating that the reduction in glycolysis in ovarian cancer cells occurs through MAP30 mediated AMPK activation. Notably, the decrease in glycolysis was positively correlated with a decrease in glycolytic enzyme activity such as hexokinase and LDH, which was significantly inhibited by 30-80% and 50-90% after MAP30(10-40 μ g/mL) treatment for 24 to 48 hours, respectively (fig. 17C and 17D). Notably, it is hypothesized that the net reduction in glycolysis through MAP 30-mediated AMPK activation is due to a drop in carbon flux. Thus, glucose uptake assays were performed. MAP30(10-40 μ g/mL) behaved similarly to the drug AMPK activator AICAR (1mM) and inhibited glucose uptake in OVCA433 cells by about 30% to 60% in a dose-dependent manner (fig. 17E). Immunoblot analysis further confirmed that expression of GLUT1 and GLUT3 glucose transporters significantly decreased in a time-dependent manner after treatment with MAP30(40 μ g/mL), but that another glucose transporter GLUT4 remained unchanged (fig. 17F). These data indicate that MAP30 inhibits glucose uptake by reducing the GLUT-1 and-3 transporters.
Omentum is the preferential site for metastasis of ovarian cancer. Thus, it is hypothesized that free fatty acids enriched in the omentum microenvironment specifically modulate ovarian tumor cells, thereby promoting malignant cell survival and metastatic cancer cell spread. Lipid droplet formation was increased using nile red fluorescent staining for OCM-cultured ovarian cancer cell line OVCA433, while co-treatment with MAP30(5 and 10 μ G/mL) reduced the lipid droplet number by 50-70% (fig. 17G). To further determine whether MAP30 accelerates catabolic lipolysis in human ovarian cancer cells, lipolysis assays were performed. The results showed that MAP30(10 and 40. mu.g/mL) induced an approximately 2-4 fold increase in cellular lipolysis in ovarian cancer cells (FIG. 17H). ATP detection assay showed a significant reduction in ATP production in ovarian cancer cells due to MAP30(10 and 40 μ g/mL) treatment (fig. 17I). Overall, the findings of the present invention indicate that MAP30 interrupts glucose uptake and lipid droplet formation in ovarian cancer cells, indicating that MAP30 will inhibit the invasiveness and metastasis of ovarian cancer in the ascites microenvironment.
Discussion:
traditional Chinese medicines target cancer cells and treat the entire body, including the qi, blood, organs and immune system. Traditional Chinese medicine is used as a supplementary substitute drug. Bioactive compounds from chinese medicine have significant anti-cancer activity by modulating multiple levels and targets in cancer cells. Therefore, the traditional Chinese medicine and the bioactive compound thereof can be used as ideal auxiliary agents for modern cancer treatment. In china and india, bitter melon has been used medically to improve digestion and treat diabetes and obesity and is known for its "plant insulin" activity. Momordica charantia extract (BME) exerts an inhibitory effect on ovarian cancer cells by activating AMPK/mTOR signaling. The present study isolated MAP30 protein from dried bitter gourd seeds. Functional and biochemical analysis showed that MAP30 is the major bioactive component in BME.
BME comprises a variety of bioactive compounds, and it exhibits anti-cancer effects in a variety of human cancers. For example, Kukukocin J (KJ) isolated from extract of momordica charantia reduces resistance of ovarian cancer cells to paclitaxel. Rnase MC2 induces apoptotic mediators and apoptosis in human breast cancer. A novel peptide BG-4 from Momordica charantia can promote apoptosis of human colon cancer. However, these studies did not show any relationship or function of AMPK. Bitter gourd has been used in china and india for the treatment of diabetes, as it is used for the same purpose as the first line treatment for diabetes, metformin. This activity indicates that bitter gourd has a functional effect on AMPK activation. The balsam pear seeds are used for treating diabetes and obesity in traditional Chinese medicine. This use may be due to the abundance of bioactive metabolites in bitter melon seeds, which regulate cellular metabolism, as recently discovered using metabolomic profiling studies. MAP30 protein isolated from dried bitter gourd seeds was reported to promote apoptosis in prostate cancer cells. This result prompted us to isolate MAP30 protein from dried bitter gourd seeds. The yield of MAP30 protein was 40mg per 100g of balsam pear seeds. Notably, MAP30 proved to be the major component of BME. MAP30 activated AMPK via CaMKK β pathway in an AMP-independent manner and inhibited mTOR and the novel AKT/ERK/FOXM1 signaling pathway, thereby producing an inhibitory effect on ovarian cancer.
AMPK is a key energy sensor in healthy cells and a key factor in tumor development. AMPK is a key sensor for maintaining cellular energy balance that regulates multiple cellular metabolic pathways. Emerging evidence suggests that malignant cells require higher AMPK activity to initially overcome metabolic stresses from the tumor microenvironment, but relatively lower AMPK activity is beneficial in promoting cell growth and development in advanced cancers. These data indicate that advanced and more aggressive cancer cells prefer low AMPK activity to reduce metabolic limitations, such as mTOR, to exert high carcinogenicity. These data also provide an advantage for AMPK activators in selectively impairing ovarian cancer cell function without affecting normal cells. Native MAP30 isolated from non-toxic momordica charantia increases AMPK activity through the CaMKK β pathway and in an AMP-independent manner, which provides another perspective for targeting cancer cells whose survival and other cellular activities are not affected by AMP stress conditions, such as a nutrient-rich microenvironment. The present study shows that native MAP30 significantly inhibited cell growth by S-phase cell cycle arrest, cell migration/invasion, and tumor growth or colonization in a mouse tumor xenograft model. mTOR signaling is an important pathway for cellular metabolism, growth, proliferation, survival, and resistance. However, mTOR inhibitors do not exhibit a broad and potent anti-cancer and anti-chemo-resistant effect. MAP30 does not exhibit this drawback.
Emerging evidence suggests that the ascites microenvironment provides growth factors and reprograms the metabolism of cancer cells to promote the progression of ovarian cancer through peritoneal metastasis. Thus, targeting cancer cell metabolism is a promising approach to compromise ovarian cancer aggressiveness in the tumor microenvironment. The findings of the present invention indicate that MAP30 inhibits the oncogenic AKT/ERK/FOXM1 signaling cascade and regulates cellular metabolism, e.g. by inhibiting the expression of the major glucose transporters GLUT1 and GLUT3, lipid droplet formation, and even lipolysis to reduce glucose uptake. In the murine ovarian cancer model of the invention, glycolysis and inhibition of lipid metabolism result in a significant reduction in tumor spread. The findings of the present invention demonstrate that MAP30 activates the AMPK pathway via CaMKK beta, resulting in Ca in ovarian cancer cells2+Is increased. Unconstrained Ca2+The signal may produce mitochondrial damage and increase ROS stress. Excess ROS induce caspase-dependent apoptotic cell death pathway activation in ovarian cancer cells.
In addition to mediating caspase activation and apoptosis, excess ROS may induce lipid peroxidation of cell membranes and lead to iron-dependent cell death, i.e., iron apoptosis. Studies have shown that ovarian cancer cells take up Free Fatty Acids (FFA) from the ascites microenvironment to support their metastatic progression in peritoneal metastasis. These metastatic ovarian cancer cells typically have a higher ratio of unsaturated FFA to saturated FFA in the ascites microenvironment. Due to multiple unsaturationFatty Acids (PUFAs) are excellent substrates for lipid peroxidation and are increased in metastatic ovarian cancer cells, and therefore whether MAP30 induces iron apoptosis to inhibit metastatic colonization in ovarian cancer was investigated. Erastin is a synthetic lethal compound that triggers iron apoptosis in cancer cells through xCT/GPX4 and GSH depletion. Using Erastin as a positive control, MAP30 was found to inhibit GPX4 as well, decreasing the GSH/GSSG ratio, and increasing the amount of ROS, enhancing lipid peroxidation. However, Ferrostatin-1(Fer-1) prevented the induction of lipid peroxidation, confirming that MAP30 is an inducer of iron apoptosis in ovarian cancer cells. Erastin is a class 1 iron apoptosis inducer (FIN) that induces iron apoptosis through the inactivation of GPX and depletion of intracellular GSH. Similar to Erastin, MAP30 depletes the intracellular antioxidant system, leading to accumulation of lipid peroxides, enhancing the susceptibility of ovarian cancer cells to free radical oxidative damage and cell death. On the other hand, since MAP30 is a natural AMPK activator, it is necessary to investigate whether AMPK activity is associated with iron apoptosis. Indeed, several recent studies have shown that AMPK activity mediates iron apoptosis, but the results in different cell types remain controversial. For example, Xong x. et al demonstrate that AMPK phosphorylates BECN1, which blocks system xCT and enhances iron apoptosis through Erastin-type induction in colon adenocarcinoma cells. In contrast, Lee H et al showed that stress-activated AMPK inhibited iron apoptosis in ischemic tissues through RSL 3-driven induction. In this study, it was found that depletion of AMPK reduced the MAP 30-induced Ca2+Influx and ROS, but result in a lesser reduction in GSH/GSSG ratio and lipid peroxidation in ovarian cancer cells. These results indicate that the presence or absence of AMPK has some effect on MAP 30-induced iron apoptosis in ovarian cancer cells, but its intrinsic mechanism remains to be elucidated.
And (4) conclusion:
MAP30 is a natural AMPK activator and modulates multiple levels and targets simultaneously. Notably, these activities of MAP30 enhanced its anti-cancer response and anti-chemoresistance in ovarian cancer, supporting its use as a supplement in advanced ovarian cancer to improve the efficacy of current chemotherapy regimens in a synergistic manner.
Although the present invention has been described to a certain degree, it is apparent that appropriate changes in the respective conditions may be made without departing from the spirit and scope of the present invention. It is to be understood that the invention is not limited to the described embodiments, but is to be accorded the scope consistent with the claims, including equivalents of each element described.
Sequence listing
<110> university of hong Kong
<120> application of momordica charantia protein MAP30 in preparation of drugs or chemotherapy supplements for preventing and treating ovarian cancer
<130> FIC20210062
<160> 2
<170> SIPOSequenceListing 1.0
<210> 1
<211> 286
<212> PRT
<213> Momordica charantia
<220>
<221> SIGNAL
<222> (1)..(23)
<223> Signal peptide
<220>
<221> CARBOHYD
<222> (74)..(74)
<223> glycosylation site
<400> 1
Met Val Lys Cys Leu Leu Leu Ser Phe Leu Ile Ile Ala Ile Phe Ile
1 5 10 15
Gly Val Pro Thr Ala Lys Gly Asp Val Asn Phe Asp Leu Ser Thr Ala
20 25 30
Thr Ala Lys Thr Tyr Thr Lys Phe Ile Glu Asp Phe Arg Ala Thr Leu
35 40 45
Pro Phe Ser His Lys Val Tyr Asp Ile Pro Leu Leu Tyr Ser Thr Ile
50 55 60
Ser Asp Ser Arg Arg Phe Ile Leu Leu Asn Leu Thr Ser Tyr Ala Tyr
65 70 75 80
Glu Thr Ile Ser Val Ala Ile Asp Val Thr Asn Val Tyr Val Val Ala
85 90 95
Tyr Arg Thr Arg Asp Val Ser Tyr Phe Phe Lys Glu Ser Pro Pro Glu
100 105 110
Ala Tyr Asn Ile Leu Phe Lys Gly Thr Arg Lys Ile Thr Leu Pro Tyr
115 120 125
Thr Gly Asn Tyr Glu Asn Leu Gln Thr Ala Ala His Lys Ile Arg Glu
130 135 140
Asn Ile Asp Leu Gly Leu Pro Ala Leu Ser Ser Ala Ile Thr Thr Leu
145 150 155 160
Phe Tyr Tyr Asn Ala Gln Ser Ala Pro Ser Ala Leu Leu Val Leu Ile
165 170 175
Gln Thr Thr Ala Glu Ala Ala Arg Phe Lys Tyr Thr Glu Arg His Val
180 185 190
Ala Lys Tyr Val Ala Thr Asn Phe Lys Pro Asn Leu Ala Ile Ile Ser
195 200 205
Leu Glu Asn Gln Trp Ser Ala Leu Ser Lys Gln Ile Phe Leu Ala Gln
210 215 220
Asn Gln Gly Gly Lys Phe Arg Asn Pro Val Asp Leu Ile Lys Pro Thr
225 230 235 240
Gly Glu Arg Phe Gln Val Thr Asn Val Asp Ser Asp Val Val Lys Gly
245 250 255
Asn Ile Lys Leu Leu Leu Asn Ser Arg Ala Ser Thr Ala Asp Glu Asn
260 265 270
Phe Ile Thr Thr Met Thr Leu Leu Gly Glu Ser Val Val Asn
275 280 285
<210> 2
<211> 284
<212> PRT
<213> Momordica charantia
<220>
<221> MUTAGEN
<222> (1)..(21)
<223> histidine tag
<400> 2
Met Gly Ser Ser His His His His His His Ser Ser Gly Leu Val Pro
1 5 10 15
Arg Gly Ser His Met Asp Val Asn Phe Asp Leu Ser Thr Ala Thr Ala
20 25 30
Lys Thr Tyr Thr Lys Phe Ile Glu Asp Phe Arg Ala Thr Leu Pro Phe
35 40 45
Ser His Lys Val Tyr Asp Ile Pro Leu Leu Tyr Ser Thr Ile Ser Asp
50 55 60
Ser Arg Arg Phe Ile Leu Leu Asn Leu Thr Ser Tyr Ala Tyr Glu Thr
65 70 75 80
Ile Ser Val Ala Ile Asp Val Thr Asn Val Tyr Val Val Ala Tyr Arg
85 90 95
Thr Arg Asp Val Ser Tyr Phe Phe Lys Glu Ser Pro Pro Glu Ala Tyr
100 105 110
Asn Ile Leu Phe Lys Gly Thr Arg Lys Ile Thr Leu Pro Tyr Thr Gly
115 120 125
Asn Tyr Glu Asn Leu Gln Thr Ala Ala His Lys Ile Arg Glu Asn Ile
130 135 140
Asp Leu Gly Leu Pro Ala Leu Ser Ser Ala Ile Thr Thr Leu Phe Tyr
145 150 155 160
Tyr Asn Ala Gln Ser Ala Pro Ser Ala Leu Leu Val Leu Ile Gln Thr
165 170 175
Thr Ala Glu Ala Ala Arg Phe Lys Tyr Thr Glu Arg His Val Ala Lys
180 185 190
Tyr Val Ala Thr Asn Phe Lys Pro Asn Leu Ala Ile Ile Ser Leu Glu
195 200 205
Asn Gln Trp Ser Ala Leu Ser Lys Gln Ile Phe Leu Ala Gln Asn Gln
210 215 220
Gly Gly Lys Phe Arg Asn Pro Val Asp Leu Ile Lys Pro Thr Gly Glu
225 230 235 240
Arg Phe Gln Val Thr Asn Val Asp Ser Asp Val Val Lys Gly Asn Ile
245 250 255
Lys Leu Leu Leu Asn Ser Arg Ala Ser Thr Ala Asp Glu Asn Phe Ile
260 265 270
Thr Thr Met Thr Leu Leu Gly Glu Ser Val Val Asn
275 280