Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Mol Carcinog ; 62(9): 1417-1427, 2023 09.
Article in English | MEDLINE | ID: mdl-37265438

ABSTRACT

Incessant ovulation is believed to be a potential cause of epithelial ovarian cancer (EOC). Our previous investigations have shown that insulin-like growth factor (IGF2) and hepatocyte growth factor (HGF) in the ovulatory follicular fluid (FF) contributed to the malignant transformation initiated by p53 mutations. Here we examined the individual and synergistic impacts of IGF2 and HGF on enhancing the malignant properties of high-grade serous carcinoma (HGSC), the most aggressive type of EOC, and its precursor lesion, serous tubal intraepithelial carcinoma (STIC). In a mouse xenograft co-injection model, we observed that FF co-injection induced tumorigenesis of STIC-mimicking cells, FE25. Co-injection with IGF2 or HGF partially recapitulated the tumorigenic effects of FF, but co-injection with both resulted in a higher tumorigenic rate than FF. We analyzed the different transformation phenotypes influenced by these FF growth signals through receptor inhibition. The IGF signal was necessary for clonogenicity, while the HGF signal played a crucial role in the migration and invasion of STIC and HGSC cells. Both signals were necessary for the malignant phenotype of anchoring-independent growth but had little impact on cell proliferation. The downstream signals responsible for these HGF activities were identified as the tyrosine-protein kinase Met (cMET)/mitogen-activated protein kinase and cMET/AKT pathways. Together with the previous finding that the FF-IGF2 could mediate clonogenicity and stemness activities via the IGF-1R/AKT/mammalian target of rapamycin and IGF-1R/AKT/NANOG pathways, respectively, this study demonstrated the cooperation of the FF-sourced IGF and HGF growth signals in the malignant transformation and progression of HGSC through both common and distinct signaling pathways. These findings help develop targeted prevention of HGSC.


Subject(s)
Cystadenocarcinoma, Serous , Fallopian Tube Neoplasms , Ovarian Neoplasms , Female , Humans , Mice , Animals , Fallopian Tubes/metabolism , Fallopian Tubes/pathology , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Follicular Fluid/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ovarian Neoplasms/pathology , Tumor Suppressor Protein p53/genetics , Epithelial Cells/metabolism , Carcinogenesis/pathology , Carcinoma, Ovarian Epithelial/pathology , Cystadenocarcinoma, Serous/metabolism , Fallopian Tube Neoplasms/genetics , Fallopian Tube Neoplasms/metabolism , Fallopian Tube Neoplasms/pathology , Cell Transformation, Neoplastic/pathology , Mammals/metabolism
2.
Front Bioeng Biotechnol ; 11: 1110765, 2023.
Article in English | MEDLINE | ID: mdl-36911202

ABSTRACT

Janus kinase/signal transduction and transcription activation (JAK/STAT) pathways were originally thought to be intracellular signaling pathways that mediate cytokine signals in mammals. Existing studies show that the JAK/STAT pathway regulates the downstream signaling of numerous membrane proteins such as such as G-protein-associated receptors, integrins and so on. Mounting evidence shows that the JAK/STAT pathways play an important role in human disease pathology and pharmacological mechanism. The JAK/STAT pathways are related to aspects of all aspects of the immune system function, such as fighting infection, maintaining immune tolerance, strengthening barrier function, and cancer prevention, which are all important factors involved in immune response. In addition, the JAK/STAT pathways play an important role in extracellular mechanistic signaling and might be an important mediator of mechanistic signals that influence disease progression, immune environment. Therefore, it is important to understand the mechanism of the JAK/STAT pathways, which provides ideas for us to design more drugs targeting diseases based on the JAK/STAT pathway. In this review, we discuss the role of the JAK/STAT pathway in mechanistic signaling, disease progression, immune environment, and therapeutic targets.

3.
Cell Cycle ; 22(1): 85-99, 2023 01.
Article in English | MEDLINE | ID: mdl-36004387

ABSTRACT

High-grade serous ovarian cancer (HGSOC) is the most common and malignant type of ovarian cancer, accounting for 70%-80% of mortality. However, the treatment of HGSOC has improved little in the past few decades. Metformin is the first-line medication for the treatment of type 2 diabetes and has now gained more attention in cancer treatment. In this study, we sought to identify potential hub genes that metformin could target in the treatment of HGSOC. We downloaded GSE69428 and GSE69429 in the Gene Expression Omnibus database and performed the bioinformatics analysis. Subsequently, we analyzed the effect of Metformin in HGSOC through biological experiments. Molecular simulation docking was used to predict the interaction of Metformin and CCNE1. We chose CCNE1 for the study based on bioinformatics analysis, literature studies, and preliminary data. We evaluated that CCNE1 is overexpressed in HGSOC tissues and found that HGSOC cells with high CCNE1 expression increase sensitivity to Metformin treatment in the analysis of cell proliferation and anchorage-independent growth. Metformin could inhibit the expression of CCNE1, which is associated with the anti-proliferative effect of tumor cells. Moreover, Metformin could ameliorate the tumor growth in syngeneic orthotopic transplantation mouse models and xenograft tumorigenesis models. Furthermore, molecular simulation docking showed that Metformin may bind to CCNE1 protein, suggesting that CCNE1 could be a potential target for Metformin. Our data revealed that Metformin has antitumor effects on ovarian cancer and CCNE1 could be a potential target for Metformin.


Subject(s)
Carcinoma , Diabetes Mellitus, Type 2 , Metformin , Ovarian Neoplasms , Female , Animals , Mice , Humans , Metformin/pharmacology , Ovarian Neoplasms/pathology , Cell Proliferation , Cell Line, Tumor , Oncogene Proteins , Cyclin E
4.
Exp Cell Res ; 419(2): 113323, 2022 10 15.
Article in English | MEDLINE | ID: mdl-36030968

ABSTRACT

The incidence and mortality of epithelial ovarian cancer (EOC) are increasing in Taiwan and worldwide. The prognosis of this disease has improved little in the last few decades due to insufficient knowledge of the etiology. Previous studies on the role of ovulation in the development of EOC have unveiled IGF2, HGF, and other carcinogens in ovulatory follicular fluid (FF) that exert transformation activities on the exposed fallopian tube fimbria epithelium. However, an orthotopic proof in an animal model is lacking. By using the murine ID8 EOC cells and the syngenic transplantation model, this study explored the effect of FF on the oncogenesis of mouse ovarian cancer. We found FF promoted clonogenicity and anchorage-independent growth of ID8 cells, largely through the IGF-1R and cMET signaling. In contrast, FF modestly promoted cell proliferation independent of the two signals and did not affect cell migration and invasion. Transplantation of ID8 cells into the ovarian bursa of C57BL6/J mice orthotopically grew ovarian tumors and metastasized to the peritoneum with ascites formation. The tumorigenic rate and severity of the disease were positively correlated with the level of IGF-1R and cMET receptors on the cell surface. Our data demonstrated that ovulation, through the signaling of IGF/IGF-1R and HGF/cMET, promotes oncogenic phenotypes in a murine EOC model. The results provide further proof of the carcinogenic effect of ovulation in the development of EOC.


Subject(s)
Ovarian Neoplasms , Animals , Carcinogenesis , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Female , Hepatocyte Growth Factor/metabolism , Humans , Insulin-Like Growth Factor I/metabolism , Mice , Ovarian Neoplasms/pathology , Ovulation , Signal Transduction
5.
Biomedicines ; 10(6)2022 Jun 14.
Article in English | MEDLINE | ID: mdl-35740424

ABSTRACT

Endometriosis is a chronic disease characterized by the ectopic localization of the endometrial tissue in the peritoneal cavity. Consequently, it causes local pathological changes and systemic symptoms, affecting at least one in every ten women. This disease is difficult to diagnose early, it is prone to dissemination, is difficult to eradicate, tends to recur, and is regarded as "a cancer of no kill". Indeed, the development of endometriosis closely resembles that of cancer in the way of mutagenesis, pelvic spreading, and immunological adaptation. While retrograde menstruation has been regarded as the primary cause of endometriosis, the role of ovulation and menstrual stimuli in the development of endometriosis has long been overlooked. The development of ovarian and peritoneal endometrioses, similar to the development of high-grade serous carcinoma in the fallopian tube fimbriae with intraperitoneal metastasis, depends highly on the carcinogens released during ovulation. Moreover, endometriosis carries an extremely hypermutated genome, which is non-inferior to the ultra-mutated endometrial cancer. The hypermutation would lead to an overproduction of new proteins or neoantigens. Because of this, the developing endometriosis may have to turn on the PD-1/PDL-1 "self-tolerance" checkpoint to evade immune surveillance, leaving an Achilles tendon for an immune checkpoint blockade. In this review, we present the double engines and single checkpoint theory of the genesis of endometriosis, provide the current pieces of evidence supporting the hypothesis, and discuss the new directions of prevention and treatment.

6.
Front Immunol ; 13: 806686, 2022.
Article in English | MEDLINE | ID: mdl-35444640

ABSTRACT

Nanodiamond (ND) has been developed as a carrier to conduct various in vivo diagnostic and therapeutic uses. Safety is one of the major considerations, while the hemocompatibility of ND is not clearly addressed. Here we found that, compared to the other sizes of ND with relatively inert properties, treatments of 50 nm ND induced stronger platelet aggregation, platelet pyroptosis, apoptosis and thrombocytopenia in mice. Blockage treatments of soluble P-selectin, reactive oxygen species (ROS), and Nlrp3 inflammasome inhibitors markedly suppressed such adverse effects, suggesting ND-induced platelet activation and pyroptosis involves surface P-selectin-mediated enhancement of mitochondrial superoxide levels and Nlrp3 inflammasome activation. In addition, challenges of NDs induced less platelet pyroptosis and displayed less thrombocytopenia in P-selectin (Selp-/- ), Nlrp3 (Nlrp3-/- ) and caspase-1 (Casp1-/- ) mutants, as compared to the wild type mice. Blockers of P-selectin, ROS, and Nlrp3 inflammasome pathways could be considered as antidotes for ND induced platelet activation and thrombocytopenia.


Subject(s)
Nanodiamonds , Thrombocytopenia , Animals , Apoptosis/physiology , Caspase 1/metabolism , Inflammasomes/metabolism , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , P-Selectin , Platelet Aggregation , Pyroptosis , Reactive Oxygen Species/metabolism
8.
Neoplasia ; 23(11): 1123-1136, 2021 11.
Article in English | MEDLINE | ID: mdl-34688971

ABSTRACT

The fallopian tube fimbrial epithelium, which is exposed to the follicular fluid (FF) contents of ovulation, is regarded as the main origin of ovarian high-grade serous carcinoma. Previously, we found that growth factors in FF, such as IGF2, are responsible for the malignant transformation of fallopian tube epithelium. However, ovulation is a monthly transient event, whereas carcinogenesis requires continuous, long-term exposure. Here, we found the transformation activity of FF sustained for more than 30 days after drainage into the peritoneal fluid (PF). Hepatocyte growth factor (HGF), activated through the ovulation injury-tissue factor-thrombin-HGF activator (HGFA)-HGF cleavage cascade confers a sustained transformation activity to fallopian tube epithelium, high-grade serous carcinoma. Physiologically, the high reserve of the coagulation-HGF cascade sources a sustained level of HGF in PF, then to the blood circulation. This HGF axis promotes the growth of the corpus luteum and repair of tissue injury after ovulation.


Subject(s)
Cell Transformation, Neoplastic/pathology , Corpus Luteum/physiology , Cystadenocarcinoma, Serous/pathology , Fallopian Tube Neoplasms/pathology , Ovarian Neoplasms/pathology , Ovulation , Peptide Hydrolases/metabolism , Adult , Animals , Apoptosis , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Corpus Luteum/injuries , Cystadenocarcinoma, Serous/metabolism , Fallopian Tube Neoplasms/metabolism , Female , Follicular Fluid/metabolism , Hepatocyte Growth Factor/metabolism , Humans , Insulin-Like Growth Factor II/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Ovarian Neoplasms/metabolism , Prognosis , Serine Endopeptidases/metabolism , Thrombin/metabolism , Thromboplastin/metabolism , Xenograft Model Antitumor Assays
9.
J Cancer ; 12(20): 6021-6030, 2021.
Article in English | MEDLINE | ID: mdl-34539876

ABSTRACT

Background: Trp53-/- mice are prone to develop lymphomas at old ages. Factors promoting this tumorigenesis are unknown. Here, we showed human ovulatory follicular fluid (FF) largely promotes lymphomagenesis in Trp53-/- mice at earlier ages. Meanwhile, we clarified that IGF2 and HGF are important cell transforming factors within FF. Methods: To induce tumor formation, 5% FFs, 100 ng/ml IGF2, 20 ng/ml HGF, or both IGF2 and HGF in a volume of 200 µl PBS, was injected into 8-wk-old female Trp53 -/- mice at the mammary fat pad. The injection was repeated weekly for up to 7 weeks or extending to 13 weeks to observe the accumulative incidence of lymphomagenesis. Immunohistochemistry staining and gene rearrangement analysis were used to identify the tumor type. Results: By injecting FF into the mammary fat pad weekly, lymphomas developed in 8/16 (50%) of mice by seven weeks. We identified IGF2 and HGF in FF is largely responsible for this activity. The same weekly injection of IGF2, HGF, and their combination induced lymphomas in 4/11 (36%), 3/8 (38%), and 6/9 (67%) mice, respectively. Interestingly, tumorigenesis was induced only when those were injected into the adipose tissues in the mammary gland, but not when injected into non-adipose sites. We also found this tumor-promoting activity is estradiol (E2)-dependent and relies on estrogen receptor (ER) α expression in the adipose stroma. No tumor or only tiny tumor was yielded when the ovaries were resected or when ER is antagonized. Finally, an extension of the weekly FF-injection to 13 weeks did not further increase the lymphomagenesis rate, suggesting an effect on pre-initiated cancer cells. Conclusions: Taken together, the study disclosed a robust tumor-promoting effect of IGF2 and HGF in the p53 loss-initiated lymphomagenesis depending on an adipose microenvironment in the presence of E2. In light of the clarity of this spontaneous tumor promotion model, we provide a new tool for studying p53-mediated lymphomagenesis and suggest that, as a chemoprevention test, this is a practical model to perform.

10.
Cell Prolif ; 54(5): e13029, 2021 May.
Article in English | MEDLINE | ID: mdl-33768671

ABSTRACT

High-grade serous carcinoma (HGSC) is the most common and malignant histological type of epithelial ovarian cancer, the origin of which remains controversial. Currently, the secretory epithelial cells of the fallopian tube are regarded as the main origin and the ovarian surface epithelial cells as a minor origin. In tubal epithelium, these cells acquire TP53 mutations and expand to a morphologically normal 'p53 signature' lesion, transform to serous tubal intraepithelial carcinoma and metastasize to the ovaries and peritoneum where they develop into HGSC. This shifting paradigm of the main cell of origin has revolutionarily changed the focus of HGSC research. Various cell lines have been derived from the two cellular origins by acquiring immortalization via overexpression of hTERT plus disruption of TP53 and the CDK4/RB pathway. Malignant transformation was achieved by adding canonical driver mutations (such as gain of CCNE1) revealed by The Cancer Genome Atlas or by noncanonical gain of YAP and miR181a. Alternatively, because of the extreme chromosomal instability, spontaneous transformation can be achieved by long passage of murine immortalized cells, whereas in humans, it requires ovulatory follicular fluid, containing regenerating growth factors to facilitate spontaneous transformation. These artificially and spontaneously transformed cell systems in both humans and mice have been widely used to discover carcinogens, oncogenic pathways and malignant behaviours in the development of HGSC. Here, we review the origin, aetiology and carcinogenic mechanism of HGSC and comprehensively summarize the cell models used to study this fatal cancer having multiple cells of origin and overt genomic instability.


Subject(s)
Carcinoma/pathology , Models, Biological , Ovarian Neoplasms/pathology , Animals , Carcinoma/metabolism , Cell Transformation, Neoplastic , Fallopian Tubes/cytology , Fallopian Tubes/metabolism , Fallopian Tubes/pathology , Female , Humans , Ovarian Neoplasms/metabolism , Telomerase/genetics , Telomerase/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
11.
Mod Pathol ; 33(1): 29-37, 2020 01.
Article in English | MEDLINE | ID: mdl-31558785

ABSTRACT

High-grade serous carcinoma is the most common and devastating type of ovarian cancer; its etiology, mechanism of malignant transformation, and origin remain controversial. Recent studies have identified secretory cells at the fimbria of the fallopian tube as the cell-of-origin of high-grade serous carcinoma, acquiring TP53 mutation, evolving to tubal precursor lesions, including "p53 signature" and serous tubal intraepithelial carcinoma, and metastasizing to the ovary as clinically evident ovarian cancer. The etiological mechanisms associated with known epidemiological risk factors, i.e., ovulation and retrograde menstruation, have also been suggested. Mutagens and transforming growth factors, such as reactive oxygen species and insulin-like growth factor axis proteins, as well as the apoptosis-rescuing protein hemoglobin are abundantly present in the ovulatory follicular fluid and peritoneum fluid, which bathes the fimbrial epithelium, and induces malignant transformation after repeated exposure. In accordance with the proposed cleansing effect of progesterone from studies on oral contraceptive use or term pregnancy, a recent study indicated that the p53-null tubal epithelial cells are selectively cleared by progesterone depending on its progesterone receptor. In this report, by analyzing different time effects of oral contraceptive use or pregnancy in the prevention of ovarian cancer and by aligning them with the carcinogenic and cleansing clearance concepts of ovulation and progesterone, as well as the fact of progressive loss of progesterone receptor during tubal transformation, we deduced the natural history of ovarian high-grade serous carcinoma. The natural history begins at the first ovulation and spans for more than 30 years, taking 10 years from the normal tubal epithelium to the "p53 signature" status, another 15 years to progesterone receptor negative serous tubal intraepithelial carcinoma, and a final 5+ years to high-grade serous carcinoma. The estimated natural history may help understand the pathogenesis of high-grade serous carcinoma and defines the window for early detection and chemoprevention.


Subject(s)
Carcinogenesis , Carcinoma, Ovarian Epithelial/pathology , Cystadenocarcinoma, Serous/pathology , Ovulation/physiology , Tumor Suppressor Protein p53/metabolism , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Contraceptives, Oral, Hormonal/pharmacology , Fallopian Tubes/metabolism , Fallopian Tubes/pathology , Female , Humans , Pregnancy , Progesterone/pharmacology
12.
EBioMedicine ; 41: 597-609, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30852161

ABSTRACT

BACKGROUND: The fallopian tube fimbria is regarded as the main tissue of origin and incessant ovulation as the main risk factor of ovarian high-grade serous carcinoma. Previously, we discovered the tumorigenesis activity of human ovulatory follicular fluid (FF) upon injection to the mammary fat pad of Trp53-null mice. We also found a mutagenesis activity of FF-ROS and a apoptosis-rescuing activity of Hb from retrograde menstruation. However, neither of them can explain the tumorigenesis activities of FF. METHODS: From two cohorts of ovulatory FF retrieved from IVF patients, the main growth factor responsible for the transformation of human fimbrial epithelial cells was identified. Mechanism of activation, ways of signal transduction of the growth factor, as well as the cellular and genetic phenotypes of the malignant transformation was characterized. FINDINGS: In this study, we showed that insulin-like growth factor (IGF)-axis proteins, including IGFBP-bound IGF2 as well as the IGFBP-lytic enzyme PAPP-A, are abundantly present in FF. Upon engaging with glycosaminoglycans on the membrane of fimbrial epithelial cells, PAPP-A cleaves IGFBPs and releases IGF2 to bind with IGF-1R. Through the IGF-1R/AKT/mTOR and IGF-1R/AKT/NANOG pathways, FF-IGF leads to stemness and survival, and in the case of TP53/Rb or TP53/CCNE1 loss, to clonal expansion and malignant transformation of fimbrial epithelial cells. By depleting each IGF axis component from FF, we proved that IGF2, IGFBP2/6, and PAPP-A are all essential and confer the majority of the transformation and regeneration activities. INTERPRETATION: This study revealed that the FF-IGF axis functions to regenerate tissue damage after ovulation and promote the transformation of fimbrial epithelial cells that have been initiated by p53- and Rb-pathway disruptions. FUND: The study was supported by grants of the Ministry of Science and Technology, Taiwan (MOST 106-2314-B-303-001-MY2; MOST 105-2314-B-303-017-MY2; MOST 107-2314-B-303-013-MY3), and Buddhist Tzu Chi General Hospital, Taiwan (TCMMP104-04-01).


Subject(s)
Follicular Fluid/metabolism , Insulin-Like Growth Factor II/metabolism , Animals , Carcinogenesis , Chromosome Aberrations , DNA Copy Number Variations , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/transplantation , Fallopian Tubes/cytology , Female , Follicular Fluid/chemistry , Humans , Insulin-Like Growth Factor Binding Proteins/metabolism , Mice , Mice, Inbred NOD , Mice, Knockout , Pregnancy-Associated Plasma Protein-A/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Receptor, IGF Type 1 , Receptors, Somatomedin/antagonists & inhibitors , Receptors, Somatomedin/genetics , Receptors, Somatomedin/metabolism , Signal Transduction , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
13.
Oncotarget ; 9(16): 12781-12795, 2018 Feb 27.
Article in English | MEDLINE | ID: mdl-29560109

ABSTRACT

Mechanisms underlying cold-induced immunosuppression remain unclear. Here we found that cold exposure leads to transient receptor potential melastatin 8 (TRPM8)-dependent, renin-angiotensin-aldosterone system (RAAS)-mediated hypertension, which subsequently induces small molecule and fluid extravasation, increases plasma Ig levels, and elicits immunosuppression. An effect is similar to the clinically-used immunosuppressive treatments of intravenous immunoglobulin (IVIg) against various inflammatory diseases, such as immune thrombocytopenia (ITP). Essential roles of TRPM8 and Ig in cold-induced immunosuppression are supported by the cold-mediated amelioration of ITP and the cold-mediated suppression of bacterial clearance, which were observed in wild-type mice but not in Ig- and TRPM8-deficient mutants. Treatment with antihypertensive drugs aliskiren and losartan drastically reversed high plasma Ig levels and ameliorated cold-induced immunosuppression, indicating the involvement of the RAAS and hypertension. These results indicated that the natively increased plasma Ig level is associated with immunosuppression during periods of cold exposure, and antihypertensive drugs can be useful to manage cold-induced immunosuppression.

14.
Cell Rep ; 18(11): 2557-2565, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28297660

ABSTRACT

High-grade serous ovarian carcinoma (HGSOC) originates mainly from the fallopian tube (FT) epithelium and always carries early TP53 mutations. We previously reported that tumors initiate in the FT fimbria epithelium because of apoptotic failure and the expansion of cells with DNA double-strand breaks (DSB) caused by bathing of the FT epithelial cells in reactive oxygen species (ROSs) and hemoglobin-rich follicular fluid (FF) after ovulation. Because ovulation is frequent and HGSOC is rare, we hypothesized that luteal-phase progesterone (P4) could eliminate p53-defective FT cells. Here we show that P4, via P4 receptors (PRs), induces necroptosis in Trp53-/- mouse oviduct epithelium and in immortalized human p53-defective fimbrial epithelium through the TNF-α/RIPK1/RIPK3/MLKL pathway. Necroptosis occurs specifically at diestrus, recovers at the proestrus phase of the estrus cycle, and can be augmented with P4 supplementation. These results reveal the mechanism of the well-known ability of progesterone to prevent ovarian cancer.


Subject(s)
Apoptosis/drug effects , Epithelial Cells/pathology , Fallopian Tubes/pathology , Neoplasms, Cystic, Mucinous, and Serous/pathology , Ovarian Neoplasms/pathology , Ovarian Neoplasms/prevention & control , Progesterone/pharmacology , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line, Tumor , Cell Survival/drug effects , DNA Breaks, Double-Stranded , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelium/drug effects , Epithelium/pathology , Estrous Cycle/drug effects , Female , Humans , Mice , Necrosis , Neoplasm Grading , Neoplasms, Cystic, Mucinous, and Serous/metabolism , Oviducts/drug effects , Oviducts/pathology , Oviducts/ultrastructure , Receptors, Progesterone/metabolism , Signal Transduction/drug effects
15.
Sci Rep ; 7: 40384, 2017 01 11.
Article in English | MEDLINE | ID: mdl-28074905

ABSTRACT

Our previous studied indicated that eukaryotic translation initiation factor 3a (eIF3a) increases the sensitive of platinum-based chemotherapy in lung cancer. MiRNAs play an important role in lung carcinogenesis and drug response. In this study, we aimed to identify potential endogenous miRNAs that inhibit eIF3a expression and determine their influence of this inhibition on cisplatin resistance. Using bioinformatics analysis prediction and confirmation with dual-luciferase reporter assays, we found that miRNA-488 inhibited eIF3a expression by directly binding to the 3'UTR of eIF3a. In addition, the overexpression of miRNA-488 inhibited cell migration and invasion in A549 cells, and also inhibited cell proliferation, cell cycle progression by elevated P27 expression. Compared to the parental cell line, A549/cisplatin (DDP) resistant cells exhibited a higher level of miRNA-488. Moreover, we found that miRNA-488 was associated with cisplatin resistance in three NSCLC cells (A549, H1299 and SK-MES-1). The mechanism of miRNA-488 induced cisplatin resistance was that miRNA-488 activated nucleotide excision repair (NER) by increasing the expression of Replication Protein A (RPA) 14 and Xeroderma pigmentosum group C (XPC). In conclusion, our results demonstrated that miRNA-488 is a tumor suppressor miRNA that acts by targeting eIF3a. Moreover, miRNA-488 also participates in eIF3a mediated cisplatin resistance in NSCLC cells.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cisplatin/therapeutic use , DNA Repair/genetics , Eukaryotic Initiation Factor-3/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , MicroRNAs/metabolism , Base Sequence , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cisplatin/pharmacology , DNA Repair/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/pathology , MicroRNAs/genetics , Models, Biological , Neoplasm Invasiveness , Signal Transduction/drug effects
16.
J Pathol ; 240(4): 484-494, 2016 12.
Article in English | MEDLINE | ID: mdl-27625309

ABSTRACT

Fallopian tube fimbrial epithelium is considered to be the major site of origin of ovarian high-grade serous carcinoma, with p53 loss being the earliest and universal change. We previously reported that reactive oxygen species (ROS) in the ovulatory follicular fluids (FFs) are mutagenic and cytotoxic to fimbrial epithelial cells, which are bathed in the peritoneal fluid mixed with FFs. Here, we observed that ferryl haemoglobin (Hb), which was abundantly present in ovulatory FFs and pelvic peritoneal fluids, could rescue p53-deficient immortalized fimbrial epithelial (FE25) cells and oviduct epithelial cells from Trp53-null mice from lethal ovulatory ROS stress. Ferryl Hb and FF containing high Hb levels protected FE25 cells from apoptosis, mainly by consuming extracellular ROS and reducing NADPH oxidase-mediated cell death. The remaining extracellular ROS could still induce DNA double-strand breaks in the fimbrial epithelial cells. Our study revealed that ferryl Hb in peritoneal fluid rescued ROS-stressed, DNA-damaged fimbrial epithelial cells from death, and suggested that peritoneal blood from various sources may contribute to the ovulation-induced transformation of Fallopian tube epithelium. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Ascitic Fluid/metabolism , Fallopian Tubes/cytology , Hemoglobins/physiology , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Animals , Apoptosis/physiology , Cell Death/physiology , Cells, Cultured , DNA Breaks, Double-Stranded , Epithelial Cells/cytology , Female , Hemoglobins/analysis , Humans , Menstruation/physiology , Mice, Knockout , NADPH Oxidases/physiology , Ovulation/physiology , Reactive Oxygen Species/analysis
17.
Am J Cancer Res ; 6(5): 957-72, 2016.
Article in English | MEDLINE | ID: mdl-27293991

ABSTRACT

Amounting evidence has demonstrated that phenethyl isothiocyanate (PEITC) is a strong inducer of reactive oxygen species (ROS) and functions as a selective killer to various human cancer cells. However, it remains obscure whether PEITC has potential selective lethality to malignant glioma cells. Thus in this study, we performed multiple analysis such as MTT assay, Hoechst 33258 staining, flow cytometry, foci formation, RT-PCR, Western blot, and transfection to explore the selective lethality of PEITC to malignant glioma cells and the underlying mechanisms. We found that PEITC induced a selective apoptosis and suppressed tumorigenicity and migration of malignant glioma cells. Furthermore, we found PEITC significantly induced GSH depletion, ROS production, caspase-9 and caspase-3 activation, and miR-135a upregulation in malignant glioma cells but not in normal cells. Moreover, PEITC activated the miR-135a-mitochondria dependent apoptosis pathway as demonstrated by downregulation of STAT6, SMAD5 and Bcl-xl while upregulation of Bax expression and Cytochrome-C release in malignant glioma cell lines but not in the immortalized human normal glial HEB cells. Correspondingly, the above PEITC-induced activation of the ROS-MiR-135a-Mitochondria dependent apoptosis pathways in malignant glioma was attenuated by pre-transfection with miR-135a inhibitor, pre-treatment with multidrug resistance-associated protein 1 (MRP1) inhibitor Sch B, or combination with glutathione (GSH). These results revealed that PEITC selectively induced apoptosis of malignant glioma cells through MRP1-mediated export of GSH to activate ROS-MiR-135a-Mitochondria dependent apoptosis pathway, suggesting a potential application of PEITC for treating glioma.

18.
Environ Toxicol ; 31(11): 1663-1673, 2016 Nov.
Article in English | MEDLINE | ID: mdl-26179408

ABSTRACT

Uncontrolled cell proliferation is a common feature of human cancer. Some of herbal extract or plant-derived medicine had been shown as an important source of effective anticancer agents. We previously reported that an n-BuOH-soluble fraction of Kalanchoe tubiflora has antiproliferative activity by inducing mitotic catastrophe. In this study, we showed that the H2 O-soluble fraction of Kalanchoe tubiflora (KT-W) caused cell cycle arrest, and senescence-inducing activities in A549 cells. We used 2 dimensional PAGE to analyze the protein expression levels after KT-W treatment, and identified that the energy metabolism-related proteins and senescence-related proteins were disturbed. In vivo experiments showed that the tumor growths in A549-xenografted nude mice were effectively inhibited by KT-W. Our findings implied that KT-W is a putative antitumor agent by inducing cell cycle arrest and senescence. © 2015 Wiley Periodicals, Inc. Environ Toxicol 31: 1663-1673, 2016.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Kalanchoe , Lung Neoplasms/drug therapy , Plant Extracts/pharmacology , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Lung Neoplasms/pathology , Male , Mice , Mice, Nude , Phytotherapy , Xenograft Model Antitumor Assays
19.
Carcinogenesis ; 36(11): 1419-28, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26363031

ABSTRACT

Ovulation is the strongest risk factor for ovarian high-grade serous carcinoma (HGSC) that largely originates from the fallopian tube fimbriae and always carries loss-of-function mutations of TP53 in both early and late lesions. Mature ovarian follicle contains high level of reactive oxygen species (ROS). When released from ovulation, follicular fluid (FF) bathes the fimbriae and may lead to DNA double-strand break (DSB) and neoplastic transformation. In this study, we examined the mutagenic and tumorigenic activities of human pre-ovulatory FFs. A subset (6/11) of FFs was found with high levels of ROS whereas the antioxidant capacities were indifferent. These ROS(high) FFs induced intracellular ROS and DSBs in the secretory cell population of fimbriae epithelium. When p53 and Rb were turned down, the FF-exposed secretory cells overcame apoptosis and expanded the population carrying ROS and DSB. The cancer initiation and promotion effects of FF were further recapitulated in Trp53 (-/-) mice. When introduced into the mammary fat pad, ROS(high) but not ROS(low) FFs induced early-onset B-cell lymphoma. Cotreatment with physiological concentration of melatonin, a potent antioxidant, ameliorated the mutagenic and tumorigenic effect of ROS(high) FF in vitro and in vivo. The study revealed ROS and mitogens in mature ovarian follicles could initiate the transformation of fimbria epithelium in the context of p53 loss and melatonin is a potent preventive agent.


Subject(s)
Follicular Fluid/physiology , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , Tumor Suppressor Protein p53/genetics , Adult , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Survival , DNA Breaks, Double-Stranded , Epithelium/pathology , Fallopian Tubes/pathology , Female , Humans , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Mutagenesis , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Reactive Oxygen Species/metabolism , Tumor Cells, Cultured
20.
J Immunol ; 195(6): 2743-53, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26259584

ABSTRACT

The mechanisms leading to the life-threatening dengue hemorrhagic fever (DHF) remain elusive. DHF preferentially occurs during secondary dengue infections, suggesting that aberrant immune responses are involved in its development. We previously demonstrated that the autoantibodies elicited by dengue virus (DENV) nonstructural protein 1 (NS1; anti-NS1 Igs) induce plasma leakage and mortality in mice with warfarinized anticoagulant suppression. However, the involved pathogenic Ig fractions of anti-NS1 Igs remain unclear. In this study, the autoreactive Igs in patients with DHF and in NS1-immunized rabbits crossreacted with TNF-related apoptosis-inducing ligand receptor 1 (death receptor [DR]4). Challenges with the DENV in a subcytotoxic dose sensitized endothelial cells to apoptosis. Treatments with the autoantibodies induced proapoptotic activities and suppressed the surface expression of endothelial anticoagulant thrombomodulin. Combined treatments comprising the DENV and DR4 affinity-purified fractions of anti-NS1 IgGs (anti-NS1-DR4 Ig), but not preimmune control IgGs, in subcytotoxic doses led to apoptosis in endothelial cells. Treatments with the anti-NS1-DR4 Ig led to plasma leakage, coagulopathy, and morality in mice with warfarinized anticoagulant suppression. These results suggest that DR4-induced endothelial cell sensitization through NS1-elicited autoantibodies exacerbates anticoagulant suppression, vascular injury, and plasma leakage. Detecting and blocking anti-DR Igs in patients may be novel strategies for managing severe DENV infection.


Subject(s)
Autoantibodies/immunology , Dengue Virus/immunology , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Severe Dengue/pathology , Viral Nonstructural Proteins/immunology , Animals , Antibodies/immunology , Antibodies/pharmacology , Anticoagulants , Apoptosis/immunology , Blood Coagulation , Cell Line , Cell Survival , Chick Embryo , Culicidae , Endothelial Cells/immunology , Endothelial Cells/pathology , Humans , Immunoglobulin G/immunology , Mice , RNA Interference , RNA, Small Interfering , Rabbits , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Severe Dengue/immunology , Thrombomodulin/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL