Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Nutr Cancer ; 76(3): 305-315, 2024.
Article in English | MEDLINE | ID: mdl-38185896

ABSTRACT

Dictyopteris divaricata (DD) has been reported to exert diverse pharmacological activities, including anti-inflammatory, antioxidant, and anticancer effects. In this study, we aimed to investigate the anticancer potential of the ethanolic extract of DD (EDD) in non-small cell lung cancer (NSCLC) cells and to explore the underlying mechanism. EDD significantly suppressed cell proliferation in H1299, PC9, and H1975 NSCLC cells. EDD treatment increased the proportion of Annexin V-positive cells and cells in sub-G1 phase, indicating the induction of apoptosis. This observation was further supported by the presence of fragmented nuclei and increased expression of cleaved PARP and cleaved caspase-3 in NSCLC cells following EDD treatment. Mechanistically, EDD decreased the phosphorylation levels of signal transducer and activator of transcription 3 (STAT3) and Src. Transfection of constitutively activated STAT3 into H1975 cells partially attenuated EDD-induced apoptosis, highlighting the contribution of STAT3 inhibition to the anticancer activity of EDD. In addition, we identified fucosterol as a major constituent of EDD that exhibited similar anticancer potential in NSCLC cells. Taken together, our results demonstrate that EDD induces apoptosis in NSCLC cells by inhibiting STAT3 activity. We propose EDD as a potential candidate for the development of therapies targeting NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , STAT3 Transcription Factor/metabolism , Cell Line, Tumor , Apoptosis , Cell Proliferation
2.
Molecules ; 29(3)2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38338342

ABSTRACT

The aim of this study was to investigate the anti-angiogenic effects of the hexane fraction of Adenophora triphylla var. japonica root extract (HAT) and its influence on the development of erlotinib resistance in human lung cancer cells. HAT significantly reduced the migration, invasion, and tube formation of human umbilical vein endothelial cells (HUVECs). The phosphorylation levels of vascular endothelial growth factor receptor 2 (VEGFR2) and its downstream molecules were decreased via HAT, indicating its anti-angiogenic potential in endothelial cells (ECs). A docking analysis demonstrated that ß-sitosterol and lupeol, representative components of HAT, exhibit a high affinity for binding to VEGFR2. In addition, conditioned media from HAT-pretreated H1299 human lung cancer cells attenuated cancer-cell-induced chemotaxis of HUVECs, which was attributed to the decreased expression of angiogenic and chemotactic factors in H1299 cells. Interestingly, co-culture of erlotinib-sensitive PC9 human lung cancer cells with HUVECs induced erlotinib resistance in PC9 cells. However, co-culture with HAT-pretreated HUVECs partially restored the sensitivity of PC9 cells to erlotinib. HAT inhibited the development of erlotinib resistance by attenuating hepatocyte growth factor (HGF) production by ECs. Taken together, our results demonstrate that HAT exerts its anticancer effects by regulating the crosstalk between ECs and lung cancer cells.


Subject(s)
Campanulaceae , Lung Neoplasms , Humans , Erlotinib Hydrochloride/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Vascular Endothelial Growth Factor A/metabolism , Hexanes/pharmacology , Angiogenesis , Angiogenesis Inhibitors/pharmacology , Human Umbilical Vein Endothelial Cells , Neovascularization, Pathologic/drug therapy , Vascular Endothelial Growth Factor Receptor-2 , Cell Movement , Cell Proliferation
3.
Nutr Cancer ; 75(1): 376-387, 2023.
Article in English | MEDLINE | ID: mdl-36004720

ABSTRACT

The aim of this study was to investigate whether the ethanol extract of the Trichosanthes kirilowii root (ETK), traditionally used to treat lung diseases, exhibits anticancer activity in epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI)-resistant non-small cell lung cancer (NSCLC) cells. ETK treatment suppressed the growth of EGFR TKI-resistant NSCLC cells, including H1299, H1975, PC9/ER (erlotinib-resistant PC9) and PC9/GR (gefitinib-resistant PC9) cells, in a concentration- and time-dependent manner. Dose-dependent decline in anchorage-dependent and -independent colony formation was also detected following ETK treatment. We demonstrate that the growth-inhibitory effect of ETK was related to apoptosis induction, based on flow cytometry results showing ETK-induced increase in the percentage of cells with sub-G1 DNA and the population of annexin V-positive cells. Consistently, ETK induced chromatin condensation and cleavage of poly(ADP-ribose) polymerase (PARP). As a molecular mechanism, the phosphorylation level of signal transducer and activator of transcription 3 (STAT3) and Src was decreased by ETK. ETK-induced apoptosis was partially reversed by transfection of constitutively activated STAT3, indicating that STAT3 inactivation mediated ETK-induced apoptosis in EGFR TKI-resistant NSCLC cells. Our results provide basic evidence supporting the role of ETK as a novel therapeutic in EGFR TKI-resistant NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Plant Extracts , Humans , Apoptosis , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm , ErbB Receptors/genetics , ErbB Receptors/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Plant Extracts/pharmacology , Trichosanthes/chemistry
4.
Nutr Cancer ; 75(10): 1945-1957, 2023.
Article in English | MEDLINE | ID: mdl-37870977

ABSTRACT

The roots of Trichosanthes kirilowii (TK) have been used in traditional oriental medicine for the treatment of respiratory diseases. In this study, we investigated whether an ethanolic root extract of TK (ETK) can regulate the metastatic potency of epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI)-resistant human lung cancer cells. The relative migration and invasion abilities of erlotinib-resistant PC9 (PC9/ER) and gefitinib-resistant PC9 (PC9/GR) cells were higher than those of parental PC9 cells. Mesenchymal markers were overexpressed, whereas epithelial markers were downregulated in resistant cells, suggesting that resistant cells acquired the EMT phenotype. ETK reduced migration and invasion of resistant cells. The expression levels of N-cadherin and Twist were downregulated, whereas Claudin-1 was upregulated by ETK, demonstrating that ETK suppresses EMT. As a molecular mechanism, Src was dephosphorylated by ETK. The anti-metastatic effect of ETK was reduced by transfecting PC9/ER cells with a constitutively active form of c-Src. Dasatinib downregulated N-cadherin, Twist, and vimentin, suggesting that Src regulates EMT in resistant cells. Notably, CuB played a key role in mediating the anti-metastatic activity of ETK. Collectively, our results demonstrate that ETK can attenuate the metastatic ability of EGFR-TKI-resistant lung cancer cells by inhibiting Src-mediated EMT.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Trichosanthes , Humans , Lung Neoplasms/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Trichosanthes/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Protein Kinase Inhibitors/pharmacology , Drug Resistance, Neoplasm , Cell Line, Tumor , Cadherins
5.
Molecules ; 28(8)2023 Apr 12.
Article in English | MEDLINE | ID: mdl-37110648

ABSTRACT

Previous studies have indicated that the adrenergic receptor signaling pathway plays a fundamental role in chronic stress-induced cancer metastasis. In this study, we investigated whether an ethanol extract of Perilla frutescens leaves (EPF) traditionally used to treat stress-related symptoms by moving Qi could regulate the adrenergic agonist-induced metastatic ability of cancer cells. Our results show that adrenergic agonists including norepinephrine (NE), epinephrine (E), and isoproterenol (ISO) increased migration and invasion of MDA-MB-231 human breast cancer cells and Hep3B human hepatocellular carcinoma cells. However, such increases were completely abrogated by EPF treatment. E/NE induced downregulation of E-cadherin and upregulation of N-cadherin, Snail, and Slug. Such effects were clearly reversed by pretreatment with EPF, suggesting that the antimetastatic activity of EPF could be related to epithelial-mesenchymal transition (EMT) regulation. EPF suppressed E/NE-stimulated Src phosphorylation. Inhibition of Src kinase activity with dasatinib completely suppressed the E/NE-induced EMT process. Transfecting MDA-MB-231 cells with constitutively activated Src (SrcY527F) diminished the antimigration effect of EPF. Taken together, our results demonstrate that EPF can suppress the adrenergic agonist-promoted metastatic ability of cancer cells by inhibiting Src-mediated EMT. This study provides basic evidence supporting the probable use of EPF to prevent metastasis in cancer patients, especially those under chronic stress.


Subject(s)
Perilla frutescens , Humans , Perilla frutescens/metabolism , Adrenergic Agonists/pharmacology , Epithelial-Mesenchymal Transition , Signal Transduction , src-Family Kinases/metabolism , Cell Line, Tumor , Cell Movement , Neoplasm Invasiveness
6.
Breast Cancer Res Treat ; 192(3): 553-561, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35107713

ABSTRACT

PURPOSE: We previously constructed a nomogram for predicting the risk of arm lymphedema following contemporary breast cancer treatment. This nomogram should be validated in patients with different background characteristics before use. Therefore, we aimed to externally validate the nomogram in a large multi-institutional cohort. METHODS: Overall, 8835 patients who underwent breast cancer surgery during 2007-2017 were identified. Data of variables in the nomogram and arm lymphedema were collected. The nomogram was validated externally using C-index and integrated area under the curve (iAUC) with 1000 bootstrap samples and by calibration plots. RESULTS: Overall, 1377 patients (15.6%) developed lymphedema. The median time from surgery to lymphedema development was 11.4 months. Lymphedema rates at 2, 3, and 5 years were 11.2%, 13.1%, and 15.6%, respectively. Patients with lymphedema had significantly higher body mass index (median, 24.1 kg/m2 vs. 23.4 kg/m2) and a greater number of removed nodes (median, 17 vs. 6) and more frequently underwent taxane-based chemotherapy (85.7% vs. 41.9%), total mastectomy (73.1% vs. 52.1%), conventionally fractionated radiotherapy (71.9% vs. 54.2%), and regional nodal irradiation (70.7% vs 22.4%) than those who did not develop lymphedema (all P < 0.001). The C-index of the nomogram was 0.7887, and iAUC was 0.7628, indicating good predictive accuracy. Calibration plots confirmed that the predicted lymphedema risks were well correlated with the actual lymphedema rates. CONCLUSION: This nomogram, which was developed using factors related to multimodal breast cancer treatment and was validated in a large multi-institutional cohort, can well predict the risk of breast cancer-related lymphedema.


Subject(s)
Breast Neoplasms , Lymphedema , Breast Neoplasms/surgery , Female , Humans , Lymphedema/epidemiology , Lymphedema/etiology , Lymphedema/surgery , Mastectomy , Nomograms , Risk Factors
7.
Nutr Cancer ; 74(5): 1837-1848, 2022.
Article in English | MEDLINE | ID: mdl-34533079

ABSTRACT

The root bark of Morus alba L. (MA) used in traditional oriental medicine exerts various bioactivities including anticancer effects. In this study, we investigated the molecular mechanism underlying the methylene chloride extract of MA (MEMA)-induced apoptosis in colorectal cancer (CRC) cells. We observed that MEMA decreased cell viability and colony formation in both HCT116 p53+/+ cells and HCT116 p53-/- cells. In addition, MEMA increased the sub-G1 phase DNA content, the annexin V-positive cell population, and the expression of apoptosis marker proteins in both cell lines, indicating that MEMA induced apoptosis regardless of the p53 status. Interestingly, the phosphorylation level, transcriptional activity, and target genes expression of signal transducer and activator of transcription 3 (STAT3) were commonly decreased by MEMA. The overexpression of constitutively active STAT3 in HCT116 cells reversed MEMA-induced apoptosis, demonstrating that MEMA-triggered apoptosis was mediated by the inactivation of STAT3. Taken together, we suggest that MEMA can be applied not only to p53 wild-type CRC in the early stages but also to p53-mutant advanced CRC with hyperactivated STAT3. Even though a wide range of studies are required to validate the anticancer effects of MEMA, we propose MEMA as a novel material for the treatment of CRC.


Subject(s)
Colorectal Neoplasms , Morus , Apoptosis , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , HCT116 Cells , Humans , Plant Bark , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
8.
Aesthetic Plast Surg ; 46(2): 937-946, 2022 04.
Article in English | MEDLINE | ID: mdl-34761289

ABSTRACT

BACKGROUND: In advanced breast cancer, radiotherapy is recommended as adjuvant therapy following breast reconstructive surgery. This inevitably led to growing concerns over possible complications of radiotherapy on implants. In this experimental animal study, we investigated the utility of acellular dermal matrix (ADM) wraps around implants as preventive management for radiotherapy complications. METHODS: Black mice (C57NL6; n = 32) were assigned to groups that either received radiation or did not: groups A and B underwent surgery using implants without radiotherapy; while groups C and D underwent surgery using implants with radiotherapy for one and three months, respectively. The hemispheric silicone implants with an 0.8-cm-diameter were inserted on the left back of each mouse, and implants wrapped by ADM were inserted on the right back. The Clinic 23EX LINAC model was used for irradiation at 10 Gy. The samples were evaluated by gross assessment, histological analysis, immunohistochemical analysis, and the Western blotting test. RESULTS: The H&E staining analysis showed that membrane thickness is smallest in group A, followed by groups C, D, and B. In a Masson trichrome histological analysis, collagen fibers became less dense and more widespread over time in the groups that received an ADM. Immunohistochemistry findings were similarly constant. However, the expression of TGF-ß1 was increased in the irradiated groups, whereas it was decreased in the non-irradiated groups as observed over time. CONCLUSIONS: Radiotherapy was shown to increase risk factors for capsular contracture, including inflammatory response, pseudoepithelium, thinning of membrane, and TGF-ß1 expression over time; however, the accompanying framework using an ADM as a barrier between implant and tissue was shown to be effective in alleviating these risks. NO LEVEL ASSIGNED: This journal requires that authors assign a level of evidence to each submission to which Evidence-Based Medicine rankings are applicable. This excludes Review Articles, Book Reviews, and manuscripts that concern Basic Science, Animal Studies, Cadaver Studies, and Experimental Studies. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .


Subject(s)
Acellular Dermis , Breast Implants , Implant Capsular Contracture , Mammaplasty , Radiotherapy , Acellular Dermis/radiation effects , Animals , Capsules , Humans , Implant Capsular Contracture/etiology , Implant Capsular Contracture/prevention & control , Mice , Radiotherapy/adverse effects , Silicones , Transforming Growth Factor beta1
9.
Molecules ; 27(7)2022 Apr 06.
Article in English | MEDLINE | ID: mdl-35408753

ABSTRACT

The aim of this study was to investigate the anticancer effects of the root extract of Peucedanum praeruptorum Dunn (EPP) in human non-small-cell lung cancer (NSCLC) cells and explore the mechanisms of action. We used four types of human lung cancer cell lines, including H1299 (epidermal growth factor receptor (EGFR) wild-type), PC9 (EGFR Glu746-Ala750 deletion mutation in exon 19; EGFR tyrosine kinase inhibitor (TKI)-sensitive), H1975 (EGFR L858R/T790M double-mutant; EGFR TKI-resistant), and PC9/ER (erlotinib-resistant) cells. EPP suppressed cell growth and the colony formation of NSCLC cells in a concentration-dependent manner. EPP stimulated chromatin condensation, increased the percentage of sub-G1 phase cells, and enhanced the proportion of annexin V-positive cells, demonstrating that EPP triggered apoptosis in NSCLC cells regardless of the EGFR mutation and EGFR TKI resistance status. The phosphorylation level of the signal transducer and activator of transcription 3 (STAT3) and AKT was decreased by EPP. The expression of STAT3 target genes was also downregulated by EPP. EPP reversed hepatocyte growth factor (HGF)-induced MET phosphorylation and gefitinib resistance. Taken together, our results demonstrate that EPP exerted anticancer effects not only in EGFR TKI-sensitive NSCLC cells, but also in EGFR TKI-resistant NSCLC cells, by suppressing MET activity.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm/genetics , ErbB Receptors/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mutation , Plant Extracts/pharmacology , Protein Kinase Inhibitors/pharmacology
10.
Molecules ; 27(18)2022 Sep 13.
Article in English | MEDLINE | ID: mdl-36144682

ABSTRACT

Chronic stress has been reported to stimulate the release of catecholamines, including norepinephrine (NE) and epinephrine (E), which promote cancer progression by activating the adrenergic receptor (AR). Although previous studies showed that ß2-AR mediated chronic stress-induced tumor growth and metastasis, the underlying mechanism has not been fully explored. In this study, we aimed to investigate the molecular mechanism by which ß2-AR exerts a pro-metastatic function in hepatocarcinoma (HCC) cells and breast cancer (BC) cells. Our results showed that Hep3B human HCC cells and MDA-MB-231 human BC cells exhibited the highest ADRB2 expression among diverse HCC and BC cell lines. NE, E, and isoprenaline (ISO), adrenergic agonists commonly increased the migration and invasion of Hep3B cells and MDA-MB-231 cells. The phosphorylation level of Src was significantly increased by E/NE. Dasatinib, a Src kinase inhibitor, blocked E/NE-induced migration and invasion, indicating that AR agonists enhanced the mobility of cancer cells by activating Src. ADRB2 knockdown attenuated E/NE-induced Src phosphorylation, as well as the metastatic ability of cancer cells, suggesting the essential role of ß2-AR. Taken together, our results demonstrate that chronic stress-released catecholamines promoted the migration and invasion of HCC cells and BC cells via ß2-AR-mediated Src activation.


Subject(s)
Receptors, Adrenergic, beta-2 , Signal Transduction , Adrenergic Agonists , Catecholamines , Cell Line, Tumor , Dasatinib , Epinephrine/pharmacology , Humans , Isoproterenol/pharmacology , Neoplastic Processes , Norepinephrine/metabolism , Norepinephrine/pharmacology , Receptors, Adrenergic, beta-2/genetics , src-Family Kinases/genetics , src-Family Kinases/metabolism
11.
Int J Colorectal Dis ; 36(6): 1279-1286, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33547945

ABSTRACT

PURPOSE: We analyzed the safety and feasibility of preoperative short-course radiotherapy (SCRT) followed by consolidation chemotherapy for patients with locally advanced rectal cancer (LARC). METHODS: From April 2018 to May 2019, 19 patients with LARC were treated with SCRT followed by three cycles of consolidation chemotherapy with leucovorin, fluorouracil, and oxaliplatin (FOLFOX6) before surgery. Adjuvant chemotherapy relied on oxaliplatin. Tumor response, patient compliance, and toxicities were analyzed. RESULTS: The median age was 60 years (range 44-71), and 16 of the patients were male. The median tumor height was 5 cm (range 0-9) from anal verge. All patients received a total dose of 25 Gy in five fractions. The number of cycles of FOLFOX6 before surgery was three in 17, four in one, five in one. Five patients required dose reductions in consolidation chemotherapy. The median interval between initiation of SCRT and surgery was 10.6 weeks (range 8.6-16.4). A pathologic complete response was seen in two patients (11%). Grade III toxicities to the preoperative treatment were seen in five patients (26%): diarrhea in two, a decreased white blood cell count in one, and anemia in two. Postoperative complications arising within 30 days developed in five patients (26%). During the median follow-up period of 20.4 months, there was no tumor recurrence. CONCLUSION: Preoperative SCRT followed by oxaliplatin-based consolidation chemotherapy showed acceptable toxicity and feasibility in patients with LARC. Prospective randomized trials are warranted to verify the efficacy and safety of this treatment strategy compared with conventional long-course concurrent chemoradiotherapy.


Subject(s)
Neoadjuvant Therapy , Rectal Neoplasms , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Chemoradiotherapy/adverse effects , Consolidation Chemotherapy , Female , Fluorouracil/adverse effects , Humans , Leucovorin/adverse effects , Male , Middle Aged , Neoplasm Recurrence, Local , Neoplasm Staging , Oxaliplatin , Prospective Studies , Rectal Neoplasms/drug therapy , Rectal Neoplasms/radiotherapy
12.
Int J Mol Sci ; 22(10)2021 May 13.
Article in English | MEDLINE | ID: mdl-34068421

ABSTRACT

Resistance to epidermal growth factor receptor tyrosine kinase inhibitors (EGFR TKIs) is a major obstacle in managing lung cancer. The root of Scutellaria baicalensis (SB) traditionally used for fever clearance and detoxification possesses various bioactivities including anticancer effects. The purpose of this study was to investigate whether SB exhibited anticancer activity in EGFR TKI-resistant lung cancer cells and to explore the underlying mechanism. We used four types of human lung cancer cell lines, including H1299 (EGFR wildtype; EGFR TKI-resistant), H1975 (acquired TKI-resistant), PC9/ER (acquired erlotinib-resistant), and PC9/GR (acquired gefitinib-resistant) cells. The ethanol extract of SB (ESB) decreased cell viability and suppressed colony formation in the four cell lines. ESB stimulated nuclear fragmentation and the cleavage of poly(ADP-ribose) polymerase (PARP) and caspase-3. Consistently, the proportion of sub-G1 phase cells and annexin V+ cells were significantly elevated by ESB, indicating that ESB induced apoptotic cell death in EGFR TKI-resistant cells. ESB dephosphorylated signal transducer and activator of transcription 3 (STAT3) and downregulated the target gene expression. The overexpression of constitutively active STAT3 reversed ESB-induced apoptosis, suggesting that ESB triggered apoptosis in EGFR TKI-resistant cells by inactivating STAT3. Taken together, we propose the potential use of SB as a novel therapeutic for lung cancer patients with EGFR TKI resistance.


Subject(s)
Drug Resistance, Neoplasm , Lung Neoplasms/pathology , Plant Extracts/pharmacology , Protein Kinase Inhibitors/pharmacology , STAT3 Transcription Factor/antagonists & inhibitors , Scutellaria baicalensis/chemistry , Apoptosis , Cell Proliferation , ErbB Receptors/antagonists & inhibitors , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Plant Roots/chemistry , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Tumor Cells, Cultured
13.
Biochem Biophys Res Commun ; 527(4): 889-895, 2020 07 05.
Article in English | MEDLINE | ID: mdl-32430175

ABSTRACT

Tumor-associated macrophages (TAMs) are closely related with poor prognosis of cancers. The current study investigated whether lupeol regulates TAMs by focusing on the recruitment and polarization of macrophages. We found that lupeol suppressed the recruitment of THP-1 macrophages (THP-1 cells differentiated into macrophages) towards H1299 lung carcinoma cells by inhibiting plasminogen activator inhibitor-1 (PAI-1) production from H1299 cells. The reduced migration of THP-1 macrophages by lupeol was recovered by adding recombinant human PAI-1 as a chemoattractant. Knockdown of PAI-1 or treatment of tiplaxtinin, a PAI-1 inhibitor, in H1299 cells abrogated the chemotaxis of macrophages. Furthermore, lupeol suppressed the interleukin (IL)-4- and IL-13-induced M2 macrophage polarization. The mRNA expression of M2 macrophage markers and the phosphorylation of signal transducer and activator of transcription 6 (STAT6) were commonly decreased by lupeol in RAW264.7 cells. In addition, lupeol-suppressed M2 macrophage polarization led to the reduced migration of Lewis lung carcinoma (LLC) cells. Taken together, our results suggest that lupeol attenuates PAI-1-mediated macrophage recruitment towards cancer cells and inhibits M2 macrophage polarization.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Macrophage Activation/drug effects , Pentacyclic Triterpenes/pharmacology , Plasminogen Activator Inhibitor 1/immunology , Tumor-Associated Macrophages/drug effects , Animals , Cell Line , Cell Line, Tumor , Cell Movement/drug effects , Humans , Lung Neoplasms/immunology , Mice , RAW 264.7 Cells , Tumor-Associated Macrophages/immunology
14.
Phytother Res ; 34(12): 3333-3344, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32677743

ABSTRACT

Tumor-associated macrophages (TAMs) promote tumor growth and metastasis, and are closely related with poor prognosis of cancers. Therefore, TAMs have been an attractive target in cancer therapy. This study investigated whether the root bark of Morus alba L. (MA) regulates TAMs. Methylene chloride extract of MA (MEMA) decreased the migration of RAW264.7 cells and THP-1 macrophages toward cancer cells via inhibition of focal adhesion kinase and Src activity. In addition, MEMA inhibited the phorbol myristate acetate-stimulated secretion of plasminogen activator inhibitor-1 from cancer cells, leading to the decreased chemotaxis of macrophages. Finally, MEMA-suppressed M2 macrophage polarization induced by interleukin (IL)-4/IL-13 or IL-6. MEMA downregulated the mRNA expression of M2 macrophage markers and decreased the phosphorylation of signal transducer and activator of transcription (STAT) 6 and STAT3 in RAW264.7 cells. Suppression of M2 polarization of macrophages by MEMA resulted in the reduced migration of Lewis lung carcinoma cells when the conditioned media from RAW264.7 cells was used as a chemoattractant. Taken together, our results demonstrate that MEMA regulates TAMs by blocking the recruitment of macrophages into tumor microenvironments and by inhibiting M2 polarization of macrophages.


Subject(s)
Morus/chemistry , Plant Bark/chemistry , Tumor-Associated Macrophages/drug effects , Humans , Transfection
15.
Int J Mol Sci ; 20(9)2019 May 07.
Article in English | MEDLINE | ID: mdl-31067694

ABSTRACT

The root bark of Morus alba L. (MA) has been traditionally used for the treatment of various lung diseases in Korea. Although recent research has demonstrated its anticancer effects in several cancer cells, it is still unclear whether MA inhibits the migratory ability of lung cancer cells. The present study investigated the effects of MA on the migration of lung cancer cells and explored the underlying mechanism. Results from a transwell assay and wound-healing assay demonstrated that methylene chloride extracts of MA (MEMA) suppressed the migration and invasion of H1299, H460, and A549 human non-small-cell lung cancer (NSCLC) cells in a concentration-dependent manner. Results from Western blot analyses showed that MEMA reduced the phosphorylation of STAT3 and Src. In addition, MEMA downregulated the expression of epithelial-mesenchymal transition (EMT) marker proteins including Slug, Snail, Vimentin, and N-cadherin, while upregulating the expression of Occludin-a tight-junction protein. The regulation of EMT markers and the decrease of migration by MEMA treatment were reversed once phospho-mimetic STAT3 (Y705D) or Src (Y527F) was transfected into H1299 cells. In conclusions, MEMA inhibited the migratory activity of human NSCLC cells through blocking Src/STAT3-mediated EMT.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Movement/drug effects , Epithelial-Mesenchymal Transition/drug effects , Lung Neoplasms/metabolism , Morus/chemistry , Plant Extracts/pharmacology , Cell Line, Tumor , Humans , Plant Bark/chemistry , STAT3 Transcription Factor/metabolism , src-Family Kinases/metabolism
16.
Biochem Biophys Res Commun ; 505(1): 194-200, 2018 10 20.
Article in English | MEDLINE | ID: mdl-30243717

ABSTRACT

This study was designed to validate the anticancer effects of morusin in human non-small cell lung cancer (NSCLC) cells. Morusin suppressed the cell growth and colony formation in a concentration-dependent manner in H1299, H460 and H292 cells. These anticancer activities were related with apoptosis induction proved by the accumulation of chromatin condensation, PARP cleavage, increase of sub-G1 phage and annexin V-positive cell population. Interestingly, signal transducer and activator of transcription 3 (STAT3) was dephosphorylated by morusin. Morusin suppressed the transcriptional activity of STAT3 and down-regulated the expression of STAT3 target genes. In addition, morusin inhibited the phosphorylation of epithelial growth factor receptor (EGFR), an upstream regulator of STAT3. The docking study showed that morusin directly binds to the tyrosine kinase domain of EGFR. Furthermore, the anticancer effects of morusin were consistently observed in erlotinib-resistant H1975 cells expressing L858R and T790 M mutant EGFR, suggesting that morusin can be used for the advanced NSCLC with acquired resistance to EGFR TKI. Taken together, our results demonstrate that morusin induced apoptosis in human NSCLC cells regardless of EGFR mutation status through inhibition of EGFR/STAT3 activation.


Subject(s)
Apoptosis/drug effects , Flavonoids/pharmacology , STAT3 Transcription Factor/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , ErbB Receptors/chemistry , ErbB Receptors/genetics , ErbB Receptors/metabolism , Erlotinib Hydrochloride/pharmacology , Flavonoids/chemistry , Flavonoids/metabolism , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Molecular Docking Simulation , Mutation , Phosphorylation/drug effects , Protein Binding , Protein Kinase Inhibitors/pharmacology , STAT3 Transcription Factor/genetics
17.
Gynecol Oncol ; 148(3): 449-455, 2018 03.
Article in English | MEDLINE | ID: mdl-29329882

ABSTRACT

OBJECTIVE: Lymph node involvement is an important prognostic factor in patients with cervical cancer. However, the prognostic significance of lymph node response to chemoradiotherapy remains unclear. We retrospectively analyzed the relationship between residual lymph node status after definitive chemoradiotherapy and survival. METHODS: We enrolled 117 patients with node-positive cervical cancer. All patients were treated with definitive chemoradiotherapy in our institution, from 2006 to 2016. The median follow-up period was 41months (range, 6-128months). The criterion for a positive lymph node was defined as a maximum short axis diameter of ≥8mm on pretreatment magnetic resonance imaging (MRI)/computed tomography (CT) scans. Posttreatment pelvic MRI was obtained 3months after the completion of chemoradiotherapy. Residual primary tumor was defined as any residual lesion identified upon clinical examination and/or MRI. Residual lymph node was defined as any lymph node with a short axis diameter of ≥8mm posttreatment, according to MRI/CT. RESULTS: At follow-up, 3months after chemoradiotherapy, we observed residual primary tumor in 30 patients (25.6%), and residual lymph node in 31 patients (26.5%). The presence of residual lymph node was associated with worse overall survival according to multivariate analysis (hazard ratio, 3.04; 95% confidence interval, 1.43-6.44; p=0.004). In the 5-year time-dependent ROC analysis of survival prediction, the presence of residual lymph node showed an AUC value of 0.72. CONCLUSIONS: The presence of residual lymph node after chemoradiotherapy was associated with worse survival in patients with node-positive cervical cancer.


Subject(s)
Adenocarcinoma/therapy , Carcinoma, Adenosquamous/therapy , Carcinoma, Squamous Cell/therapy , Lymph Nodes/pathology , Uterine Cervical Neoplasms/therapy , Adenocarcinoma/diagnostic imaging , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Carcinoma, Adenosquamous/diagnostic imaging , Carcinoma, Adenosquamous/pathology , Carcinoma, Squamous Cell/diagnostic imaging , Carcinoma, Squamous Cell/pathology , Chemoradiotherapy , Female , Humans , Magnetic Resonance Imaging , Middle Aged , Neoplasm Staging , Neoplasm, Residual , Pelvis , Prognosis , Proportional Hazards Models , Retrospective Studies , Survival Rate , Tomography, X-Ray Computed , Uterine Cervical Neoplasms/diagnostic imaging , Uterine Cervical Neoplasms/pathology , Young Adult
18.
Biol Pharm Bull ; 41(5): 713-721, 2018.
Article in English | MEDLINE | ID: mdl-29709909

ABSTRACT

The fruit of Citrus unshiu MARKOVICH used for various purposes in traditional medicine has various pharmacological properties including antioxidant, anti-inflammatory, and antibacterial effects. Recently, the possibility of anti-cancer activity of the extracts or components of this fruit has been reported; however, the exact mechanism has not yet been fully understood. In this study, we evaluated the anti-proliferative effect of water extract of C. unshiu peel (WECU) on human breast cancer MCF-7 cells and investigated the underlying mechanism. Our results showed that reduction of MCF-7 cell survival by WECU was associated with the induction of apoptosis. WECU-induced apoptotic cell death was related to the activation of caspase-8 and -9, representative initiate caspases of extrinsic and intrinsic apoptosis pathways, respectively, and increase in the Bax : Bcl-2 ratio accompanied by cleavage of poly(ADP-ribose) polymerase (PARP). WECU also increased the mitochondrial dysfunction and cytosolic release of cytochrome c. In addition, AMP-activated protein kinase (AMPK) and its downstream target molecule, acetyl-CoA carboxylase, were activated in a concentration-dependent manner in WECU-treated cells. In contrast, compound C, an AMPK inhibitor, significantly inhibited WECU-induced apoptosis, while inhibiting increased expression of Bax and decreased expression of Bcl-2 by WECU and inhibition of WECU-induced PARP degradation. Furthermore, WECU provoked the production of reactive oxygen species (ROS); however, the activation of AMKP and apoptosis by WECU were prevented, when the ROS production was blocked by antioxidant N-acetyl cysteine. Therefore, our data indicate that WECU suppresses MCF-7 cell proliferation by activating the intrinsic and extrinsic apoptosis pathways through ROS-dependent AMPK pathway activation.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/metabolism , Citrus , Plant Extracts/pharmacology , Reactive Oxygen Species/metabolism , Apoptosis/drug effects , Caspases/metabolism , Cell Survival/drug effects , Fruit , Humans , MCF-7 Cells , Membrane Potential, Mitochondrial/drug effects
19.
Mar Drugs ; 15(6)2017 May 30.
Article in English | MEDLINE | ID: mdl-28555064

ABSTRACT

It is well known that fucoidan, a natural sulfated polysaccharide present in various brown algae, mediates anticancer effects through the induction of cell cycle arrest and apoptosis. Nevertheless, the role of tumor suppressor p53 in the mechanism action of fucoidan remains unclear. Here, we investigated the anticancer effect of fucoidan on two p53 isogenic HCT116 (p53+/+ and p53-/-) cell lines. Our results showed that inhibition of cell viability, induction of apoptosis and DNA damage by treatment with fucoidan were similar in two cell lines. Flow cytometric analysis revealed that fucoidan resulted in G1 arrest in the cell cycle progression, which correlated with the inhibition of phosphorylation of retinoblastoma protein (pRB) and concomitant association of pRB with the transcription factor E2Fs. Furthermore, treatment with fucoidan obviously upregulated the expression of cyclin-dependent kinase (CDK) inhibitors, such as p21WAF1/CIP1 and p27KIP1, which was paralleled by an enhanced binding with CDK2 and CDK4. These events also commonly occurred in both cell lines, suggesting that fucoidan triggered G1 arrest and apoptosis in HCT116 cells by a p53-independent mechanism. Thus, given that most tumors exhibit functional p53 inactivation, fucoidan could be a possible therapeutic option for cancer treatment regardless of the p53 status.


Subject(s)
Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Colorectal Neoplasms/drug therapy , G1 Phase/drug effects , Polysaccharides/pharmacology , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Colorectal Neoplasms/metabolism , Cyclin-Dependent Kinase 2/metabolism , DNA Damage/drug effects , E2F Transcription Factors/metabolism , HCT116 Cells , Humans , Phosphorylation/drug effects , Retinoblastoma Protein/metabolism , Up-Regulation/drug effects
20.
Mol Cancer ; 14: 113, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-26041671

ABSTRACT

BACKGROUND: Therapeutic interventions in the insulin-like growth factor receptor (IGF-1R) pathway were expected to provide clinical benefits; however, IGF-1R tyrosine kinase inhibitors (TKIs) have shown limited antitumor efficacy, and the mechanisms conveying resistance to these agents remain elusive. METHODS: The expression and activation of the IGF-1R and Src were assessed via the analysis of a publicly available dataset, as well as immunohistochemistry, Western blotting, RT-PCR, and in vitro kinase assays. The efficacy of IGF-1R TKIs alone or in combination with Src inhibitors was analyzed using MTT assays, colony formation assays, flow cytometric analysis, and xenograft tumor models. RESULTS: The co-activation of IGF-1R and Src was observed in multiple human NSCLC cell lines as well as in a tissue microarray (n = 353). The IGF-1R and Src proteins mutually phosphorylate on their autophosphorylation sites. In high-pSrc-expressing NSCLC cells, linsitinib treatment initially inactivated the IGF-1R pathway but led a Src-dependent reactivation of downstream effectors. In low-pSrc-expressing NSCLC cells, linsitinib treatment decreased the turnover of the IGF-1R and Src proteins, ultimately amplifying the reciprocal co-activation of IGF-1R and Src. Co-targeting IGF-1R and Src significantly suppressed the proliferation and tumor growth of both high-pSrc-expressing and low-pSrc-expressing NSCLC cells in vitro and in vivo and the growth of patient-derived tissues in vivo. CONCLUSIONS: Reciprocal activation between Src and IGF-1R occurs in NSCLC. Src causes IGF-1R TKI resistance by acting as a key downstream modulator of the cross-talk between multiple membrane receptors. Targeting Src is a clinically applicable strategy to overcome resistance to IGF-1R TKIs.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Molecular Targeted Therapy , Receptor, IGF Type 1/metabolism , Signal Transduction , src-Family Kinases/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Humans , Imidazoles/pharmacology , Lung Neoplasms/pathology , Mice , Models, Biological , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein Stability/drug effects , Pyrazines/pharmacology , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL