Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 14 de 14
1.
Food Chem X ; 20: 100886, 2023 Dec 30.
Article En | MEDLINE | ID: mdl-38144837

Loquat fruits are among the most popular Chinese fruits because of their unique taste and aroma. The quality profiles of these fruits during 18 days of shelf-life at 20 °C were elucidated by headspace gas chromatography-ion mobility spectrometry (HS-GC-IMS), E-nose, and E-tongue. During shelf-life period, the properties and variations of 43 (20 aldehydes, 7 esters, 6 ketones, 1 alcohol, and 1 furan) volatile flavored compounds were determined by GC-IMS, which showed that the contents of methyl 3-methyl butanoate, ethyl acetate, and dimethyl ketone gradually decrease with prolonged shelf-life time, while (E)-2-heptenal, heptanal, (E)-2-pentenal, 1-penten-3-one 3-pentanone and 2-pentylfuran increase. The PCA based on the signal intensity of GC-IMS and E-nose, revealed that loquat fruits are well distinguished at different shelf-life times. The taste profile alternates as the storage time increases, along with higher pH, and lower amounts of total soluble solids, vitamin C, and total phenolics. The visual plots of GC-IMS, E-nose, and E-tongue had good consistency, and they characterized the aroma characteristics of loquat fruits well during different shelf-life periods. The findings of this research provide a useful understanding of the flavors of loquat fruits during their prolonged shelf-life, and a potential research basis for advancements in the loquat industry.

2.
Int J Med Sci ; 20(7): 901-917, 2023.
Article En | MEDLINE | ID: mdl-37324189

DPY30, a core subunit of the SET1/MLL histone H3K4 methyltransferase complexes, plays an important role in diverse biological functions through the epigenetic regulation of gene transcription, especially in cancer development. However, its involvement in human colorectal carcinoma (CRC) has not been elucidated yet. Here we demonstrated that DPY30 was overexpressed in CRC tissues, and significantly associated with pathological grading, tumor size, TNM stage, and tumor location. Furthermore, DPY30 knockdown remarkably suppressed the CRC cell proliferation through downregulation of PCNA and Ki67 in vitro and in vivo, simultaneously induced cell cycle arrest at S phase by downregulating Cyclin A2. In the mechanistic study, RNA-Seq analysis revealed that enriched gene ontology of cell proliferation and cell growth was significantly affected. And ChIP result indicated that DPY30 knockdown inhibited H3 lysine 4 trimethylation (H3K4me3) and attenuated interactions between H3K4me3 with PCNA, Ki67 and cyclin A2 respectively, which led to the decrease of H3K4me3 establishment on their promoter regions. Taken together, our results demonstrate overexpression of DPY30 promotes CRC cell proliferation and cell cycle progression by facilitating the transcription of PCNA, Ki67 and cyclin A2 via mediating H3K4me3. It suggests that DPY30 may serve as a potential therapeutic molecular target for CRC.


Colorectal Neoplasms , Cyclin A2 , Humans , Cyclin A2/genetics , Transcription Factors , Epigenesis, Genetic , Ki-67 Antigen , Proliferating Cell Nuclear Antigen , Cell Proliferation/genetics , Cell Cycle/genetics , Colorectal Neoplasms/genetics
3.
Phytother Res ; 35(10): 5741-5753, 2021 Oct.
Article En | MEDLINE | ID: mdl-34355433

Currently available therapies for hepatocellular carcinoma (HCC), with a high morbidity and high mortality, are only marginally effective and with sharp adverse side effects, which makes it compulsory to explore novel and more effective anticancer molecules. Chinese medicinal herbs exhibited prominent anticancer effects and were applied to supplement clinical cancer treatment. Here, we reported a compound, trilobolide-6-O-isobutyrate (TBB), isolated from the flowers of Wedelia trilobata with a markedly cytotoxic effect on HCC cells. We found that TBB time- and dose-dependently inhibited HCC cells' growth and colony formation in vitro. Moreover, TBB induced cell cycle arrest at the G2/M phase, mitochondrial caspase-dependent apoptosis, and suppressed migration and invasion, as well as the glycolysis of HCC cells. Mechanistically, our data indicated that TBB inhibited the STAT3 pathway activation by directly interacting with the TYR 640/657 sites of the STAT3 protein and decreasing the level of p-STAT3. TBB also regulated the expression of PCNA, Ki67, Cyclin B1, Cyclin E, Bax, Bcl2, MMP2/9, and PGK1 through the inhibition of the IL-6/STAT3 signaling pathway. Lastly, we confirmed that TBB effectively eliminated tumor growth without causing overt toxicity to healthy tissues in the xenograft tumor model. The exploration of anticancer activity and the underlying mechanism of TBB suggested its usage as a promising chemotherapeutic agent for HCC.


Carcinoma, Hepatocellular , Liver Neoplasms , Apoptosis , Butyrates , Carcinogenesis , Carcinoma, Hepatocellular/drug therapy , Cell Line, Tumor , Cell Proliferation , Furans , Humans , Interleukin-6/metabolism , Isobutyrates , Liver Neoplasms/drug therapy , STAT3 Transcription Factor/metabolism , Signal Transduction
4.
Eur J Pharmacol ; 908: 174370, 2021 Oct 05.
Article En | MEDLINE | ID: mdl-34324855

Colorectal cancer (CRC) is the most fatal gastrointestinal tumor and it is urge to explore powerful drugs for the treatment. Diosgenin (DSG) as a new steroidal had been reported exerts anti-tumor activity in multiple cancers, including CRC. However, the potential mechanism of DSG suppresses CRC remains further to be revealed. Here, we reported that DSG inhibited proliferation of CRC cells in dose- and time-dependent manner, induced apoptosis by modulating p53 and Bcl-2 family proteins expression to mediate mitochondrial apoptosis pathway, suppressed migration and invasion by reducing MMP-9 (matrix metalloproteinase) and decreased aerobic glycolysis by mediating glucose transporter (GLUT) like GLUT3 and GLUT4, and pyruvate carboxylase PC downregulation. Intriguingly, mechanistic study suggests those phenotypes involved DSG inhibited cAMP/PKA/CREB pathway in CRC cells, and result to inhibit the phosphorylation of CREB to regulate the transcription of genes above-mentioned. Finally, nude mice xenograft tumor model further indicated that DSG could be a great agent to suppress the growth of CRC cells in vivo and have no obvious side effects. Taken together, we revealed a unique mechanism that DSG suppresses CRC cells through cAMP/PKA/CREB pathway and DSG is a promising candidate drug for CRC treatment.


Diosgenin , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation , Mice , Signal Transduction
5.
Reprod Biol Endocrinol ; 18(1): 119, 2020 Nov 23.
Article En | MEDLINE | ID: mdl-33225937

BACKGROUND: Endometriosis is a benign gynecological disease with obviously feature of estrogen-dependence and inflammatory response. The applications of primary endometriotic stromal cells in research of endometriosis are restricted for short life span, dedifferentiation of hormone and cytokine responsiveness. The objective of this study was to establish and characterize immortalized human endometriotic stromal cells (ihESCs). METHODS: The endometriotic samples were from a patient with ovarian endometriosis and the primary endometriotic stromal cells were isolated from the endometriotic tissues. The primary cells were infected by lentivirus to establish telomerase reverse transcriptase (hTERT)-induced immortalized cells. Quantification of mRNA and proteins was examined by quantitative real-time polymerase chain reaction (qRT-PCR) and Western Blot. CCK-8 assay and EdU labeling assay were assigned to assess the growth of ihESCs. Karyotype assay was performed to detect the chromosomes of ihESCs. Colony formation assay and nude mouse tumorigenicity assay were used to evaluate colony-formation and tumorigenesis abilities. RESULTS: ihESCs continuously overexpressed hTERT via infection of lentivirus and significant extended the life span reaching 31 passages. The morphology, proliferation and karyotype of ihESCs remained unchanged. The expression of epithelial-mesenchymal transition (EMT) markers, estrogen-metabolizing proteins and estrogen/progesterone receptors (ERs and PRs) were unaltered. Furthermore, the treatment of estrogen increased the proliferation and EMT of ihESCs. Lipopolysaccharides (LPS) and IL-1ß remarkably induced inflammatory response. The clonogenesis ability of ihESCs was consistent with primary cells, which were much lower than Ishikawa cells. In addition, nude mouse tumorigenicity assay demonstrated that ihESCs were unable to trigger tumor formation. CONCLUSION: This study established and characterized an immortalized endometriotic stromal cell line that exhibited longer life span and kept the cellular morphology and physiological function as the primary cells. The immortalized cells remained normal feedback to estrogen and inflammatory response. Moreover, the immortalized cells were not available with tumorigenic ability. Therefore, ihESCs would be serviceable as in vitro cell tool to investigate the pathogenesis of endometriosis.


Endometriosis/genetics , Endometrium/metabolism , Gene Expression , Stromal Cells/metabolism , Animals , Cell Line, Transformed , Cell Line, Tumor , Cell Proliferation/genetics , Cell Survival/genetics , Cells, Cultured , Endometriosis/metabolism , Endometriosis/pathology , Endometrium/pathology , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Mice, Inbred BALB C , Mice, Nude , Stromal Cells/cytology , Transplantation, Heterologous/methods , Tumor Burden/genetics
6.
J Cell Mol Med ; 24(18): 10693-10704, 2020 09.
Article En | MEDLINE | ID: mdl-32725958

Endometriosis is a benign gynaecological disease appearing with pelvic pain, rising dysmenorrhoea and infertility seriously impacting on 10% of reproductive-age females. This research attempts to demonstrate the function and molecular mechanism of RhoA/ROCK pathway on epithelial-mesenchymal transition (EMT) and proliferation in endometriosis. The expression of Rho family was abnormally changed in endometriotic lesions; in particular, RhoA and ROCK1/2 were significantly elevated. Overexpression of RhoA in human eutopic endometrial epithelial cells (eutopic EECs) enhanced the cell mobility, epithelial-mesenchymal transition (EMT) and proliferation, and RhoA knockdown exhibited the opposite function. Oestrogen up-regulated the RhoA activity and expression of RhoA and ROCK1/2. RhoA overexpression reinforced the effect of oestrogen on promoting EMT and proliferation, and RhoA knockdown impaired the effect of oestrogen. oestrogen receptor α (ERα) was involved with the regulation of oestrogen on EMT and proliferation and up-regulated RhoA activity and expression of RhoA and ROCK1/2. The function of ERα was modulated by the change in RhoA expression. Furthermore, phosphorylated ERK that was enhanced by oestrogen and ERα promoted the protein expression of RhoA/ROCK pathway. Endometriosis mouse model revealed that oestrogen enhanced the size and weight of endometriotic lesions. The expression of RhoA and phosphorylated ERK in mouse endometriotic lesions was significantly elevated by oestrogen. We conclude that abnormal activated RhoA/ROCK pathway in endometriosis is responsible for the function of oestrogen/ERα/ERK signalling, which promoted EMT and proliferation and resulted in the development of endometriosis.


Endometriosis/pathology , Endometrium/pathology , Epithelial-Mesenchymal Transition/physiology , Estrogens/physiology , Signal Transduction/physiology , rho-Associated Kinases/physiology , rhoA GTP-Binding Protein/physiology , Adult , Animals , Cells, Cultured , Disease Models, Animal , Endometriosis/surgery , Endometrium/drug effects , Endometrium/transplantation , Epithelial-Mesenchymal Transition/drug effects , Estradiol/pharmacology , Estrogen Receptor alpha/drug effects , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/physiology , Female , Gene Expression Regulation , Humans , Mice , Mice, Inbred BALB C , Ovarian Cysts/etiology , Ovarian Cysts/surgery , RNA Interference , RNA, Messenger/biosynthesis , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Signal Transduction/drug effects , rho-Associated Kinases/biosynthesis , rho-Associated Kinases/genetics , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/biosynthesis , rhoA GTP-Binding Protein/genetics
7.
Am J Reprod Immunol ; 83(3): e13213, 2020 03.
Article En | MEDLINE | ID: mdl-31802565

PROBLEM: The application of primary eutopic endometrial cells from endometriosis patients in research is restricted for short life span, dedifferentiation of hormone responsiveness. METHOD OF STUDY: Human telomerase reverse transcriptase (hTERT)-induced immortalized cells (iheESCs) were infected by lentivirus. mRNA level was examined by qRT-PCR, and protein expression was quantified by Western blot. CCK-8 and EdU assay were assigned to assess the proliferation. The migration and invasion of cells were assessed by transwell assay. Clone formation assay and nude mouse tumorigenicity assay were used to evaluate colony-formation and tumorigenesis abilities. RESULTS: hTERT mRNA and protein were significantly expressed higher in iheESCs compared to primary cells. iheESCs grew without morphological change for 42 passages which is much longer than 18 passages of primary cells. There was no obvious difference between primary cells and iheESCs in growth, mobility, and chromosome karyotype. Furthermore, the expression of epithelial-mesenchymal transition (EMT) markers and estrogen/progesterone receptors remained unchanged. The decidualization of iheESCs could be induced by progesterone and cAMP. Estrogen increased the proliferation and mobility of iheESCs, and lipopolysaccharides (LPS) induced the IL-1ß and IL-6 promoting inflammatory response. The colony-forming ability of iheESCs, like primary cells, was lower than Ishikawa cells. In addition, tumorigenicity assay indicated that iheESCs were unable to trigger tumor formation in BALB/c nude mouse. CONCLUSIONS: This study established and characterized iheESCs that kept the cellular physiology of primary cells and were not available with tumorigenic ability. Thus, iheESCs would be useful as in vitro cell model to investigate pathogenesis of endometriosis.


Endometriosis/pathology , Endometrium/cytology , Stromal Cells/cytology , Animals , Carcinogenesis , Cell Line, Transformed , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , Female , Humans , Mice , Mice, Nude , Telomerase/metabolism , Tumor Stem Cell Assay
8.
Onco Targets Ther ; 12: 9093-9104, 2019.
Article En | MEDLINE | ID: mdl-31806994

PURPOSE: Diosgenin (DSG) is the precursor of steroid hormones and plays a crucial part in the proliferation of various carcinomas including human colorectal cancer and gastric carcinoma. Nevertheless, its specific features and mechanisms in human cholangiocarcinoma (CCA) remain unknown. METHODS: MTS assay, colony-forming assay, and EdU assay were performed to determine the role of DSG on the progression of human CCA cells. The distributions of cell cycle, the ratio of apoptosis, and the mitochondrial membrane potential (ΔΨm) were studied by flow cytometry (FCM). AO/EB and Hoechst 33258 staining were performed to observe the morphological features of cell apoptosis. TEM was performed to observe the ultrastructures of QBC939 and HuCCT1 cells. The mRNA and protein expression of mitochondrial apoptotic pathway and GSK3ß/ß-catenin pathway were further confirmed by qPCR and Western blotting. The xenograft tumor model of HuCCT1 cells was built. Immunohistochemistry of tumor tissues was performed. RESULTS: Our results indicated that DSG inhibited the progression of six CCA cell lines. In vivo tumor studies also indicated that DSG significantly inhibited tumor growth in xenografts in nude mice. The expression of mitosis-promoting factor cyclinB1 was decreased along with the elevating level of cell cycle inhibitor p21, resulting in arresting CCA cell cycles at G2/M phase. Furthermore, DSG induced apoptosis with the increased expressions of cytosol cytochrome C, cleaved-caspase-3, cleaved-PARP1 and the Bax/Bcl-2 ratio. Mechanistically, our study showed that GSK3ß/ß-catenin pathway was involved in the apoptosis of CCA cells. Thus, DSG might provide a new clue for the drug therapy of CCA. CONCLUSION: In our data, DSG was found to have efficient antitumor potential of human CCA cells in vitro and in vivo.

9.
Pathol Oncol Res ; 25(3): 849-858, 2019 Jul.
Article En | MEDLINE | ID: mdl-30361906

Vasculogenic mimicry (VM) is a new pattern of blood supplement independent of endothelial vessels, which is related with tumor invasion, metastasis and prognosis. However, the role of VM in the prognosis of cancer patients is controversial. This study aimed to perform a meta-analysis of the published data to attempt to clarify the prognostic value of VM in the digestive cancer. Relevant studies were retrieved from the PubMed, Web of Science, Cochrane Library, Chinese National Knowledge Infrastructure and VIP databases published before March 29, 2018. Studies were included if they detected VM in the digestive cancer and analyzed the overall survival (OS) or disease-free survival (DFS) according to VM status. Two independent reviewers screened the studies, extracted data, and evaluated the quality of included studies with the Newcastle-Ottawa scale. Meta-analysis was performed using STATA 12.0 software. A total of 22 studies with 2411 patients were included in this meta-analysis. Meta-analysis showed that VM was related with the poor OS (HR = 2.30, 95% CI: 2.06-2.56, P < 0.001) and DFS (HR = 2.60, 95% CI: 2.07-3.27, P < 0.001) of patients with digestive cancer. Subgroup analysis showed VM was related with tumor differentiation, lymph node metastasis and TNM stage. Moreover, the present meta-analysis was reliable, and there was no obvious publication bias. This meta-analysis suggested that VM was a poor prognosis of digestive cancer patients. Further large and well-designed studies are required.


Gastrointestinal Neoplasms/pathology , Neovascularization, Pathologic/pathology , Cell Differentiation/physiology , Disease-Free Survival , Humans , Lymphatic Metastasis/pathology , Prognosis
10.
Dig Dis Sci ; 63(12): 3348-3358, 2018 12.
Article En | MEDLINE | ID: mdl-30155836

BACKGROUND: Aberrant expression of retinoic acid receptor α (RARα) was correlated with diverse carcinomas such as acute promyelocytic leukemia and colorectal carcinoma. Nevertheless, the function and mechanism of RARα in esophageal carcinoma (EC) remain unclear. AIM: To investigate the expression of RARα in EC and its effect in the tumorigenesis of EC. METHODS AND RESULTS: In immunohistochemistry study, RARα was overexpressed in human EC tissues, and its overexpression was closely related to the pathological differentiation, lymph node metastasis, and clinical stages in EC patients. Functionally, RARα knockdown suppressed the proliferation and metastasis of EC cells through downregulating the expression of PCNA, Ki67, MMP7, and MMP9, as well as enhanced drug susceptibility of EC cells to 5-fluorouracil and cisplatin. Mechanistically, RARα knockdown inhibited the activity of Wnt/ß-catenin pathway through reducing the phosphorylation level of GSK3ß at Ser-9 and inducing phosphorylation level at Tyr-216, which resulted in downregulation of its downstream targets such as MMP7, MMP9, and P-gP. CONCLUSIONS: Our results demonstrated that RARα knockdown suppressed the tumorigenicity of EC via Wnt/ß-catenin pathway. RARα might be a potential molecular target for EC clinical therapy.


Esophageal Neoplasms , Gene Expression Regulation, Neoplastic , Retinoic Acid Receptor alpha/metabolism , Wnt Signaling Pathway/physiology , Carcinogenesis/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Survival , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Female , Gene Knockout Techniques/methods , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasm Staging , Tumor Stem Cell Assay/methods
11.
Int J Biol Macromol ; 106: 587-595, 2018 Jan.
Article En | MEDLINE | ID: mdl-28807689

The polysaccharides (AVP) was obtained from abalone (Haliotis discus hannai Ino) viscera, using the alkaline protease to enzymolysis, sevage method and repeated freezing and thawing method to remove protein and hydrogen peroxide method to depigment. The total sugar content was 46.27±1.5% and uronic acid, sulfate radical, hexosamine and protein contents were 17.44±0.22%, 16.98±0.15%, 0.65±0.02% and 1.64±0.13% in AVP respectively. The main monosaccharide compositions of AVP were d-galactose, d-xylose, d-mannose, d-glucose and d-glucuronic acid. MTT assay showed AVP had a significant anti-tumor activity to gastric carcinoma cells, especially to MGC 803, while it had no influence upon proliferation of normal stomach cells GES 1. The results of Morphological changes, cell migration ability and AO/EB staining indicated that MGC803 cells underwent apoptosis in a dose-dependent manner induced by AVP. Moreover, the western blotting results showed that the expressions of survivin, Bcl-2 and VEGF were decreased, while the expression of Bax and p53 were increased in a dose-dependent manner of AVP. The results suggested that AVP might be a potential anti-tumor agent securely and naturally.


Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Epithelial Cells/drug effects , Gastropoda/chemistry , Gene Expression Regulation, Neoplastic , Polysaccharides/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Apoptosis/drug effects , Cell Line , Cell Line, Tumor , Cell Movement/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gastric Mucosa/metabolism , Hexosamines/chemistry , Hexosamines/isolation & purification , Humans , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Organ Specificity , Polysaccharides/chemistry , Polysaccharides/isolation & purification , Proteins/chemistry , Proteins/isolation & purification , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Stomach/pathology , Survivin , Tumor Suppressor Protein p53/agonists , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Uronic Acids/chemistry , Uronic Acids/isolation & purification , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Viscera/chemistry , bcl-2-Associated X Protein/agonists , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
12.
Tumour Biol ; 39(3): 1010428317694320, 2017 Mar.
Article En | MEDLINE | ID: mdl-28347224

Crocodile choline, an active compound isolated from Crocodylus siamensis, was found to exert potent anti-cancer activities against human gastric cancer cells in vitro and in vivo. Our study revealed that crocodile choline led to cell cycle arrest at the G2/M phase through attenuating the expressions of cyclins, Cyclin B1, and CDK-1. Furthermore, crocodile choline accelerated apoptosis through the mitochondrial apoptotic pathway with the decrease in mitochondrial membrane potential, the increase in reactive oxygen species production and Bax/Bcl-2 ratio, and the activation of caspase-3 along with the release of cytochrome c. In addition, this study, for the first time, shows that Notch pathway is remarkably deregulated by crocodile choline. The combination of crocodile choline and Notch1 short interfering RNA led to dramatically increased cytotoxicity than observed with either agent alone. Notch1 short interfering RNA sensitized and potentiated the capability of crocodile choline to suppress the cell progression and invasion of gastric cancer. Taken together, these data suggested that crocodile choline was a potent progression inhibitor of gastric cancer cells, which was correlated with mitochondrial apoptotic pathway and Notch pathway. Combining Notch1 inhibitors with crocodile choline might represent a novel approach for gastric cancer.


Apoptosis/drug effects , Choline/administration & dosage , Receptor, Notch1/biosynthesis , Stomach Neoplasms/drug therapy , Alligators and Crocodiles/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols , CDC2 Protein Kinase , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cyclin B1/biosynthesis , Cyclin-Dependent Kinases/biosynthesis , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mitochondria/drug effects , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Receptor, Notch1/genetics , Signal Transduction/drug effects , bcl-2-Associated X Protein/biosynthesis
13.
Int J Oncol ; 50(1): 241-251, 2017 Jan.
Article En | MEDLINE | ID: mdl-27878242

Apoptosis, programmed cell death under physiological or pathological conditions, plays a critical role in the tissue homeostasis of eukaryotes. It is desirable to prevent the occurrence and metastasis of cancer through inducing apoptosis. Our previous study demonstrated that apoptosis could be induced by extract from crocodile in human cholangiocarcinoma. ESC-3, a novel cytotoxic compound isolated from the extract induced apoptosis in Mz-ChA-1 cells via the mitochondria-dependent pathway in a dose-dependent manner. In this study, ESC-3 significantly inhibited the proliferation of A2780 cells and arrested the cells at G2/M phase. After exposure to ESC-3, A2780 cells displayed typical morphological changes and the ability of colony-forming was remarkably inhibited. ESC-3 could significantly upregulate the expression of Bax proteins while Bcl-2 protein remained unchanged, resulting in the elevation of Bax/Bcl-2 ratio, which usually could induce apoptosis. The critical protein of Wnt signaling (ß-catenin) was significantly downregulated, whereas Hes1, the downstream protein of Notch signaling, was remarkably attenuated through upregulating the expression of P53. In addition, xenograft models demonstrated that ESC-3 effectively suppressed the growth of OvCa tumors (T/C=42%). Western blot analysis of PCNA and VEGF confirmed that ESC-3 could inhibit the growth and metastasis of OvCa tumors. In conclusion, apoptosis could be induced by ESC-3 through Wnt/ß-catenin and Notch signaling in vitro and in vivo, and might have therapeutic potential for the treatment of human OvCa.


Carcinoma/drug therapy , Neoplasm Proteins/biosynthesis , Ovarian Neoplasms/drug therapy , Tissue Extracts/administration & dosage , Alligators and Crocodiles , Animals , Apoptosis/drug effects , Carcinoma/genetics , Carcinoma/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mitochondria/drug effects , Mitochondria/pathology , Neoplasm Proteins/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Receptors, Notch/drug effects , Tissue Extracts/chemistry , Wnt Signaling Pathway/drug effects
...