Andrographis paniculata methanol extract suppresses the phosphorylation of ETV6‑NTRK3

  • Authors:
    • Hoang Thanh Chi
    • Vo Ngoc Tram
    • Nguyen Trung Quan
    • Bui Thi Kim Ly
  • View Affiliations

  • Published online on: June 8, 2023     https://doi.org/10.3892/br.2023.1630
  • Article Number: 47
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

ETS variant transcription factor 6 (ETV6)-neurotrophic receptor tyrosine kinase 3 (NTRK3) (EN) fusions are typically found in rare diseases, such as primary renal fibrosarcoma (only six cases have been reported), secretory carcinoma of the breast and salivary gland (1 case), and AML (4 cases). Few cases have been reported, and expression of the EN gene fusion requires additional clinical data and fundamental research to be supported. The aim of the present study was to determine the inhibitory effect of Andrographis paniculata methanol extract (MeAP) on EN‑related cell lines, IMS‑M2 and BaF3/EN, as well as evaluate the mechanism of action. Vero cells were used as control cells. Trypan blue staining and MTT were used to evaluate the inhibitory effect of MeAP on tested cells. Western blotting and immunoprecipitation were used to detect the activation of EN after MeAP treatment. The IC50 values of MeAP were found to be 12.38±0.57 µg/ml (IMS‑M2) and 13.06±0.49 µg/ml (BaF3/EN). MeAP was observed to inhibit cell proliferation in a time, dose, and cell density‑dependent manner. The IC50 value for MeAP in Vero cells was markedly higher, at 109.97±4.24 (µg/ml), indicating a much less sensitive effect. Furthermore, MeAP treatment inhibited EN phosphorylation and induced apoptosis in these cells. Collectively, the present study demonstrated that MeAP has an oncogenic effect on EN fusion‑positive cell lines, in particular.

Introduction

The medicinal plant Andrographis paniculata (Burm. f.) Wall. ex Nees (A. paniculata) is indigenous to Southeast Asia, China, North America, and the West Indies (1). This herb is widely used in herbal remedies to treat malaria, viral hepatitis and liver cancer (2). Numerous natural compounds have been identified in A. paniculata, including andrographolide and its derivatives. These compounds are well-known for their biological activities, including anti-SARS CoV-2 activity (3,4). Anticancer studies on this plant have begun a good many years ago, using B16F0 melanoma syngenic and HT-29 xenograft models (2003), colon cancer cell line HT-29 (2004), human lymphocytes and albino mice (2014), the Caco-2 model (2017), and acute myeloid leukemia (AML) cell line U937 (2018) (5-9). The anticancer effect of A. paniculata has been demonstrated in a variety of ways, including cell cycle arrest, induction of apoptosis, anti-angiogenic behavior, and suppression of IL-6 expression (10). Andrographolide was reported to inhibit PI3K/AKT signaling in the A549 cell model (11).

The neurotrophic receptor tyrosine kinase (NTRK)1, NTRK2, and NTRK3 genes code for members of the tropomyosin receptor kinase (Trk) family, an upstream molecule in the PI3K/AKT signaling pathway (12). Native Trk must be stimulated by an extracellular signal, such as nerve growth factor, brain-derived growth factor, or neurotrophin 3, in order to become active (12). However, due to the fusion of NTRK3 and ETS variant transcription factor 6 (ETV6), ETV6-NTRK3 (EN) can self-phosphorylate (13). The EN fusion gene has been identified in congenital fibrosarcoma (14), secretory breast carcinoma (15), AML (16,17), mammary analog secretory carcinoma of the salivary gland (18), chronic eosinophilic leukemia (19) congenital mesoblastic nephroma (20), and thyroid cancer associated with 131I radiation exposure (21). Due to the already low prevalence of NTRK fusions in most tumours (<1%) EN cases are even more uncommon (22). To date, TRK inhibitors are a viable treatment option for patients whose tumours test positive for the EN fusion. Both larotrectinib and entrectinib, which target the NTRK fusion protein, have shown promising results in recent clinical trials against locally advanced and metastatic solid tumours (23,24). EN fusions are typically found in rare diseases, such as primary renal fibrosarcoma (only six cases have been reported), secretory carcinoma of the breast and salivary gland (1 case), and AML (4 cases). Few cases have been reported, and thus EN gene fusion expression requires additional clinical data and fundamental research to be supported.

The purpose of the present study was to ascertain the inhibitory effect of A. paniculata methanol extract (MeAP) on EN-associated cell lines (IMS-M2 and BaF3/EN) as well as to identify the mechanism of action.

Materials and methods

Materials and plant extraction preparation

Andrographis paniculata (Burm. f.) Wall. ex Nees (A. paniculata) was collected in August 2017 and verified by Dr Dang Van My (Traditional Medicine Center, Tinh Bien, Vietnam) (voucher no. BNAG-2017-0115). The leaves were cleaned and dried at 40˚C in a dry oven after collection. Dry samples were blended into a fine powder and mixed with methanol (1:10 w/v). The mixture was spun at room temperature for 4 days before filtering with Whatman filter paper. The obtained extract was evaporated at 40˚C under vacuum. Subsequently, the crude extract continued to be placed in the drying oven at 40˚C for solvent evaporation and elevated drying. The extraction efficacy was 4.18%. The 200-mg/ml stock solution was produced by weighing and dissolving the dry crude extract in dimethylsulfoxide (DMSO). The stock solution was stored at -20˚C.

The inhibitor of EN, PKC412 (Sigma-Aldrich; Merck KGaA) was dissolved in DMSO and was used as a positive reagent control (13). Control cells were grown with the same amount of carrier DMSO as in the highest reagent concentration. In all of the experiments, the amount of DMSO was kept below 0.1% to keep it from killing cells.

Cell lines and culture conditions

In the present study, the EN-positive human AML cell line, IMS-M2, provided by Professor Yuko Sato (University of Tokyo, Tokyo, Japan), the stable transfection with EN, BaF3/EN (established by HTC), and the Vero (ATCC-CCL-81) cells were used (13,25). Vero cells were used as negative control cells to specify the selective effect of MeAP on cells with EN. The cells were cultured at 37˚C in a humidified incubator with 5% CO2 in Roswell Park Memorial Institute 1640 medium (Sigma-Aldrich; Merck KGaA) supplemented with 10% fetal bovine serum (Thermo Fisher Scientific, Inc.), 100 IU/ml penicillin, and 0.1 mg/ml streptomycin (both from Sigma-Aldrich; Merck KGaA).

Cell viability

The cells (1x105 cells/ml) were seeded in 6-well plates with or without MeAP treatment. Following treatment of the cells, 10 µl of cell suspension and 10 µl of 0.4% Trypan blue (product no. T8154; Sigma Aldrich; Merck KGaA) were mixed together. The cells were manually counted with a hemocytometer (26). After 48 h of incubation, the cytotoxicity of MeAP was determined dose-dependently, and the half maximal inhibitory concentration (IC50) was then calculated.

For dose and cell-density dependent tests, cells were treated with or without MeAP (12.5 µg/ml) for 48 h. For the time-dependent test, cells were treated with or without MeAP (12.5 µg/ml) at 24, 48, 72 and 96 h.

The cytotoxic activity of MeAP was determined using the 3-(4,5-dimethylthiazol)-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) method. DMSO (100 µl) was used to used to dissolve the purple formazan and the absorbance was read at 570 nm using a universal microplate reader (27) for the remaining adherent cell lines. The methods cited were modified to accommodate the research conditions of the laboratory.

To identify the selective effects of MeAP on leukemia cells, the Selective Index (SI) values were calculated. SI values >3 were considered to be highly selective for cells of interest (28).

Morphological changes in MeAP-treated cells

IMS-M2 and BaF3/EN cells were seeded overnight at a density of 1x105 cells/well in six-well culture plates. The cells were then treated with varying concentrations of MeAP (6.25, 12.5, 25, and 50 µg/ml) or PKC412 (60 nM) and maintained at 37˚C and 5% CO2 for 72 h. The untreated cells served as the control. To detect the morphological changes in the cells, an inverted light microscope was used at a magnification of x10.

Western blot analysis

The IMS-M2 and BaF3/EN cells were plated at a density of 1x105 cells/ml on a 10-cm dish with varying concentrations of MeAP (12.5, 25, or 50 µg/ml). After the indicated time points (4, 8 or 24 h) of incubation, the cells were removed and washed twice with PBS (-) (TBR Technology Corporation). The cells were then lysed in ice-cold protein lysis buffer (10 mM disodium diphosphate, 50 mM sodium fluoride, 5 mM ethylenediaminetetraacetic acid, 1 mM sodium orthovanadate, 5 mM 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid, 1 mM phenylmethylsulfonyl fluoride, 0.01% Triton X-100, 150 mM sodium chloride, and 75 µg/ml aprotinin) (29). After centrifuging the cells at 15,000 x g for 10 min (4˚C), total protein cell lysates were obtained. Protein samples (20 µg, as measured by the BCA protein assay kit) were loaded onto wells, and the proteins were resolved on a 12.5% polyacrylamide gel electrophoresis, and then electroblotted onto a Hypond-P membrane (Amersham; Cytiva). The membrane was then blocked at room temperature for 1 h with 5% skim milk buffer. Following a wash, primary antibodies were used to probe the membrane, and ECL was used to detect antibody binding (Amersham; Cytiva). Anti-TrkC (C-14) (1:500; cat. no. sc-11; Santa Cruz Biotechnology, Inc.), anti-actin (1:1,000; cat. no. A2066; Sigma-Aldrich; Merck KGaA), anti-phosphotyrosine 4G10 (1:1,000; cat. no. 05-321MG; Upstate Biotechnology, Inc.; Merck KGaA), caspase-3 (1:1,000; cat. no. 9662; Cell Signaling Technology, Inc.) and anti-PARP (1:1,000; cat. no. 016-16831; FUJIFILM Wako Pure Chemical Corporation) were used as primary antibodies. The primary antibodies were incubated at room temperature for 1 h, or overnight at 4˚C. The membranes were then washed twice for 15 min each time and incubated with a horseradish peroxidase (HRP)-conjugated secondary antibody for 1 h at room temperature (1:1,000; anti-mouse IgG HRP (cat. no. sc-2031) or anti-rabbit IgG HRP (cat. no. sc-2317; both from Santa Cruz Biotechnology, Inc.).

Immunoprecipitation (IP)

IMS-M2 or BaF3/EN cells were treated with MeAP (12.5, 25 or 50 g/ml) for 8 h and then harvested for IP. The cells were lysed as described above. A total of 500 mg of total cell lysates were immunoprecipitated overnight at 4˚C with anti-TrkC (C-14) (1:500; cat. no. sc-11; Santa Cruz Biotechnology, Inc.). Subsequently, Protein G Sepharose 4 Fast Flow (Amersham Pharmacia Biosciences; Cytiva) was added and all the procedures were carried out in accordance with the manufacturer's instructions. The immunoprecipitates were washed with Tris-buffered saline with Tween-20 three times. Using SDS-PAGE and western blotting, the bound proteins were separated and analysed.

Statistical analysis

Data were compiled from three independent experiments and are presented as the mean ± SEM. Data were compared using a paired Student's t-test or a one-way ANOVA with Tukey's post hoc test in GraphPad Prism version 8.3.0. (GraphPad Software, Inc.). P<0.05 was considered to indicate a statistically significant difference.

Results

Effect of MeAP on EN-positive cells in a dose-dependent manner

The inhibitory potential of MeAP was determined by assessing its cytotoxic effects on IMS-M2 and BaF3/EN cells at varying concentrations. MeAP (0, 6.25, 12.5, 25 and 50 µg/ml) was added to the 6-well plates and results were collected after 48 h. PKC412 at 60 nM (13) was used as a positive control. MeAP was found to inhibit the viability of these cells (Fig. 1A). The ANOVA method was used to determine whether there were any statistically significant differences in the toxic tendency of the extract across these cell lines. The IC50 values for IMS-M2 and BaF3/EN were 12.38±0.57 µg/ml and 13.06±0.49 µg/ml, respectively.

Morphological assessment was conducted 48 h after treatment (Fig. 1B). The observations indicated that the cells grew normally in the untreated group, with normal morphology such as round shape, uniform size, intact membrane and nucleus (IMS-M2 and BaF3/EN). In comparison to the control group, when the extract concentration was increased, the number and size of the treated cells significantly decreased. The shape of the cells was altered and cells began to shrink. By contrast to organelle dilation caused by early membrane permeability, cell shrinkage is a frequent and prominent morphological property of the apoptotic cell death process (30,31). The observational experiment revealed that the assessed cell lines exhibited apoptotic tendencies.

Effect of MeAP on AML cells in time-dependent manner

For up to 96 h, IMS-M2 and BaF3/EN cells were examined for viability with or without MeAP (12.5 µg/ml) treatment. At five time points (0, 24, 48, 72 and 96 h), the number of viable cells was observed and calculated. As revealed in Fig. 2, MeAP was able to inhibit the viability of cells in time-dependent manner. After 24 h of exposure to the extract, the cells grew very slowly and the number of cells was not significantly altered (Fig. 2, left panels). Between 48 and 72 h, the number of viable cells began to markedly decline. After 72 h of treatment, only 15% of the cells remained viable, and decreased to ~0% 24 h later. The cell density decreased as the culture time was extended. Fig. 2, right panel shows how cell morphology changed after MeAP co-cultured, with cell shrinkage, cell debris, and membrane blebbing.

Effect of MeAP on AML cells in a cell density-dependent manner

Statistical analysis of the effect of MeAP (12.5 µg/ml) revealed statistical differences between the control and experimental groups at the same cell density, as well as differences between the groups (Fig. 3). Experiments with various densities (P<0.0001; <α=0.05) were performed. When treated with MeAP, almost all cells died at a density of 104 cells/ml. When the cell density increased from 5x104 to 5x105 cells/ml, the percentage of viable cells increased from 8.72 to 21.86%. The percentage of viable cells in the experimental group was 23.84±0.58% at a density of 106 cells/ml. The percentage of cells in the MeAP-treated group was significantly lower than that in the control group, decreasing from 100 to 76.15% at a density of 104-106 cells/ml. In summary, the inhibitory effect of MeAP on IMS-M2 and BaF3/EN cell viability at the IC50 concentrations was cell density-dependent.

Selective index

As illustrated in Fig. 4, MeAP exerted a significantly less sensitive effect on Vero cells, with an IC50 value of 109.97±4.24 (µg/ml). It was also demonstrated that MeAP had an SI value >3, which indicated that it had marked cytotoxic potential and was very efficient at killing leukemia cells (Fig. 4, right panel).

MeAP inhibits the activity of EN protein

Although it is unknown how EN leads to cancer, the fusion protein has been considered as a significant target for cancer treatment (9,32). Additionally, previous research using an EN-expressing cell model demonstrated that PKC412 is an effective inhibitor of EN-associated leukemia (13).

It was hypothesized that the viability inhibition observed in both cell lines is due to the fusion protein's phosphorylation being inhibited. To elucidate the mechanism of MeAP-mediated viability inhibition in IMS-M2 and BaF3/EN cells, the phosphorylation status of EN in these cells was examined after treatment with or without MeAP. To determine the status of EN tyrosine phosphorylation, the total protein of EN was immunoprecipitated with TrkC antibody and immunoblotted with 4G10 antibody. As predicted, MeAP treatment inhibited the phosphorylation of EN in a dose-dependent manner, but total EN protein levels were unaffected (Fig. 5).

MeAP induces apoptosis in IMS-M2 and BaF3/EN cells

Cell viability inhibition of IMS-M2 and BaF3/EN cells following MeAP treatment may result in changes in the shape and size of cells, as illustrated in Fig. 1, Fig. 2 and Fig. 3. Next, it was examined whether MeAP treatment could affect the expression of apoptotic markers. These cells were treated with MeAP at a concentration of 50 µg/ml for up to 24 h. The results indicated that MeAP treatment activated the caspase cascade. PARP and caspase-3 molecules were turned into cleaved forms in the cells that were exposed to MeAP (50 µg/ml), which indicated that apoptosis occurred. These findings indicated that MeAP induced apoptosis in IMS-M2 and BaF3/E-N cells (Fig. 6).

Discussion

NTRK1, NTRK2, and NTRK3 encode the tyrosine kinases TrkA, TrkB, and TrkC, respectively. In a variety of tumor types, oncogenic gene fusions involving members of the NTRK family have been identified and appear to result in constitutive Trk kinase activity (33). Orally administered larotrectinib is a highly selective inhibitor that inhibits all three Trk protein isoforms at nanomolar concentrations (34). Significant tumor regressions have been reported with larotrectinib in a child with congenital fibrosarcoma harboring an EN fusion and an adult with a soft tissue sarcoma harboring an LMNA-NTRK1 fusion (35,36). Larotrectinib has also demonstrated preliminary activity in a number of other cancer types with NTRK fusion positivity (23). In July 2016, the FDA designated larotrectinib as a breakthrough therapy for the treatment of unresectable or metastatic solid tumors with NTRK fusions.

The EN fusion gene is expressed at low levels in AML, however it exerts a potent transforming effect on numerous cell lines, including hematopoietic cells, and transformed cells can induce tumors in nude mice (13,37). Previous research documented two cases of EN-AML, one with AML M2 and severe myelofibrosis at the time of diagnosis, as well as rapidly spreading leukemia cells to multiple organs, and the other with primary myelofibrosis and progression to AML M7. The EN fusion gene may be involved in the pathogenesis of acute myeloid leukemia (37).

Due to the rarity of EN-positive cases in the world (22), the established cell lines were also rare and not commercially available. Extreme difficulty exists in obtaining cell lines with EN fusion. In the present study, two EN-positive cells were utilised, one from a human (IMS-M2 cell line) and the other from transfected mouse BaF3 cells (BaF3/EN). It was demonstrated that BaF3/EN cell growth was dependent on EN-signalling (13).

Because the number of cases is so low, there is almost no literature on the use of medicinal herbs to treat diseases caused by the fusion gene EN. This may be the first study to mention the use of medicinal herbs to prevent leukemia cell proliferation caused by the EN fusion gene.

Currently, reports on the ability of inhibition of the proliferation of cancer cells in A. paniculata have primarily focused on andrographolide compounds, which are found in high concentrations in leaves (38). Indeed, this compound was shown to induce cell cycle arrest and mitochondrial-mediated apoptosis in the leukemia cell line HL-60 with an IC50 of 14.01 µg/ml after a 24-h experiment (39). Other research has demonstrated that the IC50 for this compound's toxicity to MCF-7 breast cancer cell lines is 500 g/ml. Furthermore, scientists have investigated the effects of this compound on many other cancer cell lines and discovered a positive inhibitory effect on the non-small lung cancer cell line A549, as well as on the nasopharyngeal cancer cell lines (HSC-2, HSC-3, and HSC-4) (40,41).

In addition, MeAP has been demonstrated to inhibit the proliferation of colon cancer (HT-29 cells) (5), human alveolar basal epithelial cell line (A-549), human breast adenocarcinoma cell line (MCF-7), human embryonic kidney (HEK), human cervical cancer cell line (HeLa), and human invasive ductal carcinoma cell line (BT-544), as demonstrated in previous study (42). Furthermore, The GC-MS chromatogram of MeAP extracts displayed 21 peaks, indicating the presence of 21 distinct phytochemical compounds. It was determined that 2(5H)-furanone (14.73%), quinic acid (QA; 17.32%), and phytol (11.43%) were the most abundant phytochemicals (42).

In the present study, the IC50 values of MeAP were determined to be 12.38±0.57 µg/ml (IMS-M2) and 13.06±0.49 µg/ml (BaF3/EN). The SI value helps to evaluate the specificity of an extract for certain cells. A high SI value indicates a more selective extract. An SI value >3 units indicates the general toxicity of a compound (28). The results in the present study revealed that MeAP had an SI value of >3, which indicated that it exerted marked cytotoxic potential and was very effective at killing leukemia cells that had EN.

Moreover, MeAP treatment inhibited EN phosphorylation and induced apoptosis in these cells. Therefore, it was concluded that the inhibitory effect of MeAP on AML cell lines harboring the EN fusion gene was dependent on dose, time, and cell density. MeAP can stop the viability of EN-carrying AML cells by blocking the phosphorylation of EN, which causes the cells to die.

The limitation of the present study is that no knockout or overexpression cell line with its parental cell line was used, such as IMS-M2 vs. IMS-M2 EN knockout or BaF3 vs. BaF3/EN, to provide solid evidence for the selective activity of MeAP. Future research should be conducted to address this issue.

Acknowledgements

We would like to thank Professor Yuko Sato (University of Tokyo, Japan) for providing the IMS-M2 cell line used in the present study.

Funding

Funding: The present study was supported by Thu Dau Mot University under grant no. DT.21.1-058.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

HTC and BTKL conceived and designed the study. VNT, NTQ and HTC performed the experiments; VNT, NTQ, BTKL and HTC acquired and analyzed the data, as well as wrote and revised the manuscript critically for important intellectual content. BTKL and HTC confirm the authenticity of all the raw data. All authors have read and agreed to the published version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Hossain MS, Urbi Z, Sule A and Hafizur Rahman KM: Andrographis paniculata (Burm. f.) Wall. ex Nees: A review of ethnobotany, phytochemistry, and pharmacology. ScientificWorldJournal. 2014(274905)2014.PubMed/NCBI View Article : Google Scholar

2 

Davis C, Chan BY, Zhen Ong AS, Koh Y, Wen Yap AFH, Goh SH and Vidyarthi AR: An evaluation of a medical student international service-learning experience in Southeast Asia. Educ Health (Abingdon). 34:3–10. 2021.PubMed/NCBI View Article : Google Scholar

3 

Jadhav AK and Karuppayil SM: Andrographis paniculata (Burm F) Wall ex Nees: Antiviral properties. Phytother Res. 35:5365–5373. 2021.PubMed/NCBI View Article : Google Scholar

4 

Sa-Ngiamsuntorn K, Suksatu A, Pewkliang Y, Thongsri P, Kanjanasirirat P, Manopwisedjaroen S, Charoensutthivarakul S, Wongtrakoongate P, Pitiporn S, Chaopreecha J, et al: Anti-SARS-CoV-2 activity of Andrographis paniculata extract and its major component andrographolide in human lung epithelial cells and cytotoxicity evaluation in major organ cell representatives. J Nat Prod. 84:1261–1270. 2021.PubMed/NCBI View Article : Google Scholar

5 

Kumar RA, Sridevi K, Kumar NV, Nanduri S and Rajagopal S: Anticancer and immunostimulatory compounds from Andrographis paniculata. J Ethnopharmacol. 92:291–295. 2004.PubMed/NCBI View Article : Google Scholar

6 

Rajagopal S, Kumar RA, Deevi DS, Satyanarayana C and Rajagopalan R: Andrographolide, a potential cancer therapeutic agent isolated from Andrographis paniculata. J Exp Ther Oncol. 3:147–158. 2003.PubMed/NCBI View Article : Google Scholar

7 

Ahmad S, Ahmad S, Arshad M and Afzal M: Andrographia paniculata a miracle herbs for cancer treatment: In vivo and in vitro studies against aflatoxin B1 toxicity. Egypt J Med Hum Genet. 15:163–171. 2014.

8 

Li L, Yue GG, Lee JK, Wong EC, Fung KP, Yu J, Lau CB and Chiu PW: The adjuvant value of Andrographis paniculata in metastatic esophageal cancer treatment-from preclinical perspectives. Sci Rep. 7(854)2017.PubMed/NCBI View Article : Google Scholar

9 

Hodroj MH, Jardaly A, Abi Raad S, Zouein A and Rizk S: Andrographolide potentiates the antitumor effect of topotecan in acute myeloid leukemia cells through an intrinsic apoptotic pathway. Cancer Manag Res. 10:1079–1088. 2018.PubMed/NCBI View Article : Google Scholar

10 

Malik Z, Parveen R, Parveen B, Zahiruddin S, Aasif Khan M, Khan A, Massey S, Ahmad S and Husain SA: Anticancer potential of andrographolide from Andrographis paniculata (Burm.f.) Nees and its mechanisms of action. J Ethnopharmacol. 272(113936)2021.PubMed/NCBI View Article : Google Scholar

11 

Lee YC, Lin HH, Hsu CH, Wang CJ, Chiang TA and Chen JH: Inhibitory effects of andrographolide on migration and invasion in human non-small cell lung cancer A549 cells via down-regulation of PI3K/Akt signaling pathway. Eur J Pharmacol. 632:23–32. 2010.PubMed/NCBI View Article : Google Scholar

12 

Amatu A, Sartore-Bianchi A and Siena S: NTRK gene fusions as novel targets of cancer therapy across multiple tumour types. ESMO Open. 1(e000023)2016.PubMed/NCBI View Article : Google Scholar

13 

Chi HT, Ly BT, Kano Y, Tojo A, Watanabe T and Sato Y: ETV6-NTRK3 as a therapeutic target of small molecule inhibitor PKC412. Biochem Biophys Res Commun. 429:87–92. 2012.PubMed/NCBI View Article : Google Scholar

14 

Knezevich SR, McFadden DE, Tao W, Lim JF and Sorensen PH: A novel ETV6-NTRK3 gene fusion in congenital fibrosarcoma. Nat Genet. 18:184–187. 1998.PubMed/NCBI View Article : Google Scholar

15 

Tognon C, Knezevich SR, Huntsman D, Roskelley CD, Melnyk N, Mathers JA, Becker L, Carneiro F, MacPherson N, Horsman D, et al: Expression of the ETV6-NTRK3 gene fusion as a primary event in human secretory breast carcinoma. Cancer Cell. 2:367–376. 2002.PubMed/NCBI View Article : Google Scholar

16 

Eguchi M, Eguchi-Ishimae M, Tojo A, Morishita K, Suzuki K, Sato Y, Kudoh S, Tanaka K, Setoyama M, Nagamura F, et al: Fusion of ETV6 to neurotrophin-3 receptor TRKC in acute myeloid leukemia with t(12;15)(p13;q25). Blood. 93:1355–1363. 1999.PubMed/NCBI

17 

Setoyama M, Tojo A, Nagamura F, Asano S, Ishimae M, Eguchi M and Kamada N: A unique translocation of the gene in a case of acute myelogenous leukemia with inv(12)(p13q15). Blood. 92:1454–1455. 1998.PubMed/NCBI

18 

Skálová A, Vanecek T, Sima R, Laco J, Weinreb I, Perez-Ordonez B, Starek I, Geierova M, Simpson RH, Passador-Santos F, et al: Mammary analogue secretory carcinoma of salivary glands, containing the ETV6-NTRK3 fusion gene: A hitherto undescribed salivary gland tumor entity. Am J Surg Pathol. 34:599–608. 2010.PubMed/NCBI View Article : Google Scholar

19 

Forghieri F, Morselli M, Potenza L, Maccaferri M, Pedrazzi L, Paolini A, Bonacorsi G, Artusi T, Giacobbi F, Corradini G, et al: Chronic eosinophilic leukaemia with ETV6-NTRK3 fusion transcript in an elderly patient affected with pancreatic carcinoma. Eur J Haematol. 86:352–355. 2011.PubMed/NCBI View Article : Google Scholar

20 

Rubin BP, Chen CJ, Morgan TW, Xiao S, Grier HE, Kozakewich HP, Perez-Atayde AR and Fletcher JA: Congenital mesoblastic nephroma t(12;15) is associated with ETV6-NTRK3 gene fusion: Cytogenetic and molecular relationship to congenital (infantile) fibrosarcoma. Am J Pathol. 153:1451–1458. 1998.PubMed/NCBI View Article : Google Scholar

21 

Leeman-Neill RJ, Kelly LM, Liu P, Brenner AV, Little MP, Bogdanova TI, Evdokimova VN, Hatch M, Zurnadzy LY, Nikiforova MN, et al: ETV6-NTRK3 is a common chromosomal rearrangement in radiation-associated thyroid cancer. Cancer. 120:799–807. 2014.PubMed/NCBI View Article : Google Scholar

22 

Solomon JP, Benayed R, Hechtman JF and Ladanyi M: Identifying patients with NTRK fusion cancer. Ann Oncol. 30 (Suppl 8):viii16–viii22. 2019.PubMed/NCBI View Article : Google Scholar

23 

Lapeña LM, Caldas MCS, Ramírez C, Basilio MS, Junco PT, Rodríguez-Laguna L, Martínez-González V, Marín-Manzano E, Perez-Martinez A and Lopez-Gutierrez JC: Larotrectinib as an effective therapy in congenital infantile fibrosarcoma: Report of two cases. European J Pediatr Surg Rep. 10:e76–e79. 2022.PubMed/NCBI View Article : Google Scholar

24 

Ernst MS, Lysack JT, Hyrcza MD, Chandarana SP and Hao D: TRK inhibition with entrectinib in metastatic salivary secretory carcinoma (SC): A case report. Curr Oncol. 29:3933–3939. 2022.PubMed/NCBI View Article : Google Scholar

25 

Chi HT, Thuong NTL and Ly BTK: Sphagneticola trilobata (L.) Pruski (asteraceae) methanol extract induces apoptosis in leukemia cells through suppression of BCR/ABL. Plants (Basel). 10(980)2021.PubMed/NCBI View Article : Google Scholar

26 

Ly BT, Chi HT, Yamagishi M, Kano Y, Hara Y, Nakano K, Sato Y and Watanabe T: Inhibition of FLT3 expression by green tea catechins in FLT3 mutated-AML cells. PLoS One. 8(e66378)2013.PubMed/NCBI View Article : Google Scholar

27 

Ala AA, Olotu BB and Ohia CMD: Assessment of cytotoxicity of leaf extracts of Andrographis paniculata and Aspilia africana on murine cells in vitro. Arch Basic Appl Med. 6:61–65. 2018.PubMed/NCBI

28 

Mahavorasirikul W, Viyanant V, Chaijaroenkul W, Itharat A and Na-Bangchang K: Cytotoxic activity of Thai medicinal plants against human cholangiocarcinoma, laryngeal and hepatocarcinoma cells in vitro. BMC Complement Altern Med. 10(55)2010.PubMed/NCBI View Article : Google Scholar

29 

Iijima Y, Okuda K, Tojo A, Tri NK, Setoyama M, Sakaki Y, Asano S, Tokunaga K, Kruh GD and Sato Y: Transformation of Ba/F3 cells and Rat-1 cells by ETV6/ARG. Oncogene. 21:4374–4383. 2002.PubMed/NCBI View Article : Google Scholar

30 

Ziegler U and Groscurth P: Morphological features of cell death. News Physiol Sci. 19:124–128. 2004.PubMed/NCBI View Article : Google Scholar

31 

Doonan F and Cotter TG: Morphological assessment of apoptosis. Methods. 44:200–204. 2008.PubMed/NCBI View Article : Google Scholar

32 

Tognon C, Garnett M, Kenward E, Kay R, Morrison K and Sorensen PH: The chimeric protein tyrosine kinase ETV6-NTRK3 requires both Ras-Erk1/2 and PI3-kinase-Akt signaling for fibroblast transformation. Cancer Res. 61:8909–8916. 2001.PubMed/NCBI

33 

Vaishnavi A, Le AT and Doebele RC: TRKing down an old oncogene in a new era of targeted therapy. Cancer Discov. 5:25–34. 2015.PubMed/NCBI View Article : Google Scholar

34 

Vaishnavi A, Capelletti M, Le AT, Kako S, Butaney M, Ercan D, Mahale S, Davies KD, Aisner DL, Pilling AB, et al: Oncogenic and drug-sensitive NTRK1 rearrangements in lung cancer. Nat Med. 19:1469–1472. 2013.PubMed/NCBI View Article : Google Scholar

35 

Nagasubramanian R, Wei J, Gordon P, Rastatter JC, Cox MC and Pappo A: Infantile fibrosarcoma with NTRK3-ETV6 fusion successfully treated with the tropomyosin-related kinase inhibitor LOXO-101. Pediatr Blood Cancer. 63:1468–1470. 2016.PubMed/NCBI View Article : Google Scholar

36 

Doebele RC, Davis LE, Vaishnavi A, Le AT, Estrada-Bernal A, Keysar S, Jimeno A, Varella-Garcia M, Aisner DL, Li Y, et al: An oncogenic NTRK fusion in a patient with soft-tissue sarcoma with response to the tropomyosin-related kinase inhibitor LOXO-101. Cancer Discov. 5:1049–1057. 2015.PubMed/NCBI View Article : Google Scholar

37 

Zhou F and Chen B: Acute myeloid leukemia carrying ETV6 mutations: Biologic and clinical features. Hematology. 23:608–612. 2018.PubMed/NCBI View Article : Google Scholar

38 

Chao WW and Lin BF: Isolation and identification of bioactive compounds in Andrographis paniculata (Chuanxinlian). Chin Med. 5(17)2010.PubMed/NCBI View Article : Google Scholar

39 

Cheung HY, Cheung SH, Li J, Cheung CS, Lai WP, Fong WF and Leung FM: Andrographolide isolated from Andrographis paniculata induces cell cycle arrest and mitochondrial-mediated apoptosis in human leukemic HL-60 cells. Planta Med. 71:1106–1111. 2005.PubMed/NCBI View Article : Google Scholar

40 

Lin HH, Tsai CW, Chou FP, Wang CJ, Hsuan SW, Wang CK and Chen JH: Andrographolide down-regulates hypoxia-inducible factor-1α in human non-small cell lung cancer A549 cells. Toxicol Appl Pharmacol. 250:336–345. 2011.PubMed/NCBI View Article : Google Scholar

41 

Suzuki R, Matsushima Y, Okudaira N, Sakagami H and Shirataki Y: Cytotoxic components against human oral squamous cell carcinoma isolated from Andrographis paniculata. Anticancer Res. 36:5931–5935. 2016.PubMed/NCBI View Article : Google Scholar

42 

Anoor PK, Yadav AN, Rajkumar K, Kande R, Tripura C, Naik KS and Burgula S: Methanol extraction revealed anticancer compounds quinic acid, 2(5H)-furanone and phytol in Andrographis paniculata. Mol Clin Oncol. 17(151)2022.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

July-2023
Volume 19 Issue 1

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chi HT, Tram VN, Quan NT and Ly BT: <em>Andrographis paniculata</em> methanol extract suppresses the phosphorylation of ETV6‑NTRK3. Biomed Rep 19: 47, 2023
APA
Chi, H.T., Tram, V.N., Quan, N.T., & Ly, B.T. (2023). <em>Andrographis paniculata</em> methanol extract suppresses the phosphorylation of ETV6‑NTRK3. Biomedical Reports, 19, 47. https://doi.org/10.3892/br.2023.1630
MLA
Chi, H. T., Tram, V. N., Quan, N. T., Ly, B. T."<em>Andrographis paniculata</em> methanol extract suppresses the phosphorylation of ETV6‑NTRK3". Biomedical Reports 19.1 (2023): 47.
Chicago
Chi, H. T., Tram, V. N., Quan, N. T., Ly, B. T."<em>Andrographis paniculata</em> methanol extract suppresses the phosphorylation of ETV6‑NTRK3". Biomedical Reports 19, no. 1 (2023): 47. https://doi.org/10.3892/br.2023.1630