Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Cell Biosci ; 13(1): 118, 2023 Jun 28.
Article in English | MEDLINE | ID: mdl-37381062

ABSTRACT

BACKGROUND: Artemisia argyi (A. argyi), also called Chinese mugwort, has been widely used to control pandemic diseases for thousands of years since ancient China due to its anti-microbial infection, anti-allergy, and anti-inflammation activities. Therefore, the potential of A. argyi and its constituents in reducing the infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was investigated in this study. RESULTS: Among the phytochemicals in A. argyi, eriodictyol and umbelliferone were identified to target transmembrane serine protease 2 (TMPRSS2) and angiotensin-converting enzyme 2 (ACE2) proteins, the essential factors for the cellular entry of SARS-CoV-2, in both FRET-based enzymatic assays and molecular docking analyses. These two ingredients of A. argyi suppressed the infection of ACE2-expressed HEK-293 T cells with lentiviral-based pseudo-particles (Vpp) expressing wild-type and variants of SARS-CoV-2 spike (S) protein (SARS-CoV-2 S-Vpp) via interrupting the interaction between S protein and cellular receptor ACE2 and reducing the expressions of ACE2 and TMPRSS2. Oral administration with umbelliferone efficiently prevented the SARS-CoV-2 S-Vpp-induced inflammation in the lung tissues of BALB/c mice. CONCLUSIONS: Eriodictyol and umbelliferone, the phytochemicals of Artemisia argyi, potentially suppress the cellular entry of SARS-CoV-2 by preventing the protein binding activity of the S protein to ACE2.

2.
Mol Ther Nucleic Acids ; 25: 536-553, 2021 Sep 03.
Article in English | MEDLINE | ID: mdl-34589276

ABSTRACT

Mutant p53 (mutp53) commonly loses its DNA binding affinity to p53 response elements (p53REs) and fails to induce apoptosis fully. However, the p53 mutation does not predict chemoresistance in all subtypes of breast cancers, and the critical determinants remain to be identified. In this study, mutp53 was found to mediate chemotherapy-induced long intergenic noncoding RNA-p21 (lincRNA-p21) expression by targeting the G-quadruplex structure rather than the p53RE on its promoter to promote chemosensitivity. However, estrogen receptor alpha (ERα) suppressed mutp53-mediated lincRNA-p21 expression by hijacking mutp53 to upregulate damaged DNA binding protein 2 (DDB2) transcription for subsequent DNA repair and chemoresistance. Levels of lincRNA-p21 positively correlated with the clinical responses of breast cancer patients to neoadjuvant chemotherapy and had an inverse correlation with the ER status and DDB2 level. In contrast, the carboplatin-induced DDB2 expression was higher in ER-positive breast tumor tissues. These results demonstrated that ER status determines the oncogenic function of mutp53 in chemoresistance by switching its target gene preference from lincRNA-p21 to DDB2 and suggest that induction of lincRNA-p21 and targeting DDB2 would be effective strategies to increase the chemosensitivity of mutp53 breast cancer patients.

3.
Cancer Sci ; 112(10): 4234-4245, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34382727

ABSTRACT

Development of acquired resistance to lapatinib, a dual epidermal growth factor receptor (EGFR)/human epidermal growth factor receptor 2 (HER2) tyrosine kinase inhibitor, severely limits the duration of clinical response in advanced HER2-driven breast cancer patients. Although the compensatory activation of the PI3K/Akt survival signal has been proposed to cause acquired lapatinib resistance, comprehensive molecular mechanisms remain required to develop more efficient strategies to circumvent this therapeutic difficulty. In this study, we found that suppression of HER2 by lapatinib still led to Akt inactivation and elevation of FOX3a protein levels, but failed to induce the expression of their downstream pro-apoptotic effector p27kip1 , a cyclin-dependent kinase inhibitor. Elevation of miR-221 was found to contribute to the development of acquired lapatinib resistance by targeting p27kip1 expression. Furthermore, upregulation of miR-221 was mediated by the lapatinib-induced Src family tyrosine kinase and subsequent NF-κB activation. The reversal of miR-221 upregulation and p27kip1 downregulation by a Src inhibitor, dasatinib, can overcome lapatinib resistance. Our study not only identified miRNA-221 as a pivotal factor conferring the acquired resistance of HER2-positive breast cancer cells to lapatinib through negatively regulating p27kip1 expression, but also suggested Src inhibition as a potential strategy to overcome lapatinib resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Drug Resistance, Neoplasm/physiology , Lapatinib/pharmacology , MicroRNAs/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Animals , Breast Neoplasms/chemistry , Breast Neoplasms/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p27/drug effects , Dasatinib/pharmacology , Down-Regulation/drug effects , Drug Resistance, Neoplasm/drug effects , Female , Forkhead Box Protein O3/metabolism , Hepatocyte Nuclear Factor 3-gamma/metabolism , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/drug effects , Microarray Analysis , NF-kappa B p50 Subunit/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Up-Regulation/drug effects , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
4.
Front Pharmacol ; 12: 614673, 2021.
Article in English | MEDLINE | ID: mdl-34267653

ABSTRACT

The proviral integration site for moloney murine leukemia virus 1 (Pim1) is a serine/threonine kinase and able to promote cell proliferation, survival and drug resistance. Overexpression of Pim1 has been observed in many cancer types and is associated with the poor prognosis of breast cancer. However, it remains unclear whether Pim1 kinase is a potential therapeutic target for breast cancer patients. In this study, we found that Pim1 expression was strongly associated with HER2 expression and that HER2-overexpressing breast cancer cells were more sensitive to Pim1 inhibitor-induced inhibitions of cell viability and metastatic ability. Mechanistically, Pim1 inhibitor suppressed the expression of HER2 at least in part through transcriptional level. More importantly, Pim1 inhibitor overcame the resistance of breast cancer cells to HER2 tyrosine kinase inhibitor lapatinib. In summary, downregulation of HER2 by targeting Pim1 may be a promising and effective therapeutic approach not only for anti-cancer growth but also for circumventing lapatinib resistance in HER2-positive breast cancer patients.

5.
Oncogene ; 40(29): 4796-4808, 2021 07.
Article in English | MEDLINE | ID: mdl-34155348

ABSTRACT

The tyrosine kinase inhibitors (TKIs) targeting epidermal growth factor receptor (EGFR) have been widely used for non-small cell lung cancer (NSCLC) patients, but the development of acquired resistance remains a therapeutic hurdle. The reduction of glucose uptake has been implicated in the anti-tumor activity of EGFR TKIs. In this study, the upregulation of the active sodium/glucose co-transporter 1 (SGLT1) was found to confer the development of acquired EGFR TKI resistance and was correlated with the poorer clinical outcome of the NSCLC patients who received EGFR TKI treatment. Blockade of SGLT1 overcame this resistance in vitro and in vivo by reducing glucose uptake in NSCLC cells. Mechanistically, SGLT1 protein was stabilized through the interaction with PKCδ-phosphorylated (Thr678) EGFR in the TKI-resistant cells. Our findings revealed that PKCδ/EGFR axis-dependent SGLT1 upregulation was a critical mechanism underlying the acquired resistance to EGFR TKIs. We suggest co-targeting PKCδ/SGLT1 as a potential strategy to improve the therapeutic efficacy of EGFR TKIs in NSCLC patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Sodium-Glucose Transporter 1 , Up-Regulation
6.
Aging (Albany NY) ; 13(10): 14065-14077, 2021 05 25.
Article in English | MEDLINE | ID: mdl-34037532

ABSTRACT

Long noncoding RNAs play key roles in several cancers, but their potential functions in gastroenteropancreatic neuroendocrine neoplasms remain to be investigated. We performed GeneChip assay to explore differentiated lncRNAs in gastric NENs and peri-cancerous tissues. The regulation of HNF1A-AS1 on biological behavior of GEP-NENs cells and in vivo xenograft model was confirmed by CCK8, colony formation assay, transwell, western blot and qRT-PCR. We next detected the potential transcription factors and the binding sites between them with bioinformatic analysis. qRT-PCR was performed to analyze the exact relationship between them. HNF1A-AS1 expression was decreased in gastric NENs tissues (p < 0.01). Over-expression of HNF1A-AS1 suppressed cellular proliferation, migration and invasion. Knockdown of transcription factor 3 inhibited the expression of HNF1A-AS1 and promoted cellular migration and invasion. Oncostatin M was identified as the downstream target of HNF1A-AS1. Inhibition of transforming growth factor-ß activity inhibited HNF1A-AS1/Oncostatin M-mediated epithelial-mesenchymal transition. Our data suggest that transcription factor 3/HNF1A-AS1/Oncostatin M axis inhibits the tumorigenesis and metastasis of gastroenteropancreatic neuroendocrine neoplasms via transforming growth factor-ß signaling.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Epithelial-Mesenchymal Transition/genetics , Intestinal Neoplasms/genetics , Neuroendocrine Tumors/genetics , Oncostatin M/genetics , Pancreatic Neoplasms/genetics , RNA, Long Noncoding/genetics , Signal Transduction , Stomach Neoplasms/genetics , Transcription, Genetic , Transforming Growth Factor beta/metabolism , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cell Survival/genetics , Down-Regulation/genetics , Gene Expression Regulation, Neoplastic , Humans , Intestinal Neoplasms/pathology , Neoplasm Invasiveness , Neuroendocrine Tumors/pathology , Oncostatin M/metabolism , Pancreatic Neoplasms/pathology , RNA, Long Noncoding/metabolism , Signal Transduction/genetics , Stomach Neoplasms/pathology
7.
Oncogene ; 40(6): 1162-1175, 2021 02.
Article in English | MEDLINE | ID: mdl-33335306

ABSTRACT

Smoker patients with non-small cell lung cancer (NSCLC) have poorer prognosis and survival than those without smoking history. However, the mechanisms underlying the low response rate of those patients to EGFR tyrosine kinase inhibitors (TKIs) are not well understood. Here we report that exposure to cigarette smoke extract enhances glycolysis and attenuates AMP-activated protein kinase (AMPK)-dependent inhibition of mTOR; this in turn reduces the sensitivity of NSCLC cells with wild-type EGFR (EGFRWT) to EGFR TKI by repressing expression of liver kinase B1 (LKB1), a master kinase of the AMPK subfamily, via CpG island methylation. In addition, LKB1 expression is correlated positively with sensitivity to TKI in patients with NSCLC. Moreover, combined treatment of EGFR TKI with AMPK activators synergistically increases EGFR TKI sensitivity. Collectively, the current study suggests that LKB1 may serve as a marker to predict EGFR TKI sensitivity in smokers with NSCLC carrying EGFRWT and that the combination of EGFR TKI and AMPK activator may be a potentially effective therapeutic strategy against NSCLC with EGFRWT.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Protein Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/genetics , AMP-Activated Protein Kinase Kinases , Animals , Carcinoma, Non-Small-Cell Lung/chemically induced , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cigarette Smoking/adverse effects , CpG Islands/drug effects , DNA Methylation/drug effects , Drug Resistance, Neoplasm/genetics , ErbB Receptors/genetics , Heterografts , Humans , Mice , Mutation/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinases/genetics , Signal Transduction/drug effects , Smoking/adverse effects
8.
Artif Organs ; 42(11): E349-E356, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30474888

ABSTRACT

Artificial hearts are effective devices to treat heart failure in clinical practice and can be divided into two categories: artificial hearts and ventricular assist devices. The goal of this work was to investigate the fluidity and biological changes of in vitro sheep blood using a novel alternating current (AC) magnetohydrodynamic blood pump (central magnetic intensity: 0.9 T, alternating current frequency of the electric motor: 0-80 Hz). Blood samples were collected from five sheep and were divided into two groups: the control group (no exposure to an external magnetic field) and the exposed group (3 h of exposure to an alternating magnetic field). The blood cell counts, changes in blood viscosity, and ultrastructural changes of the blood cells under transmission electron microscopy were investigated. This study demonstrated several findings: (i) Continuous sheep blood flow can be achieved; (ii) The blood cell counts remained unchanged after 3 h of exposure to an alternating magnetic field; (iii) Compared with the control group, the high- and low-shear viscosities of the whole blood from the sheep significantly decreased after 3 h of exposure to an alternating magnetic field (P < 0.05 and P < 0.01, respectively). Plasma viscosity was significantly reduced after exposure to high-intensity alternating magnetic fields (P < 0.001); (iv) The cytoplasm of blood cells (especially erythrocytes) became lighter in color in the exposure group compared to the control group, and "beads-on-string" aggregations of black particles appeared. This work provides detailed and reliable scientific research data for the development of this type of blood pump, which may serve as a transition to the clinical artificial heart.


Subject(s)
Heart, Artificial , Hemodynamics , Animals , Blood Cell Count , Blood Flow Velocity , Blood Viscosity , Equipment Design , Erythrocytes/pathology , Female , Heart, Artificial/adverse effects , Magnetic Fields , Sheep
9.
Cancer Sci ; 109(10): 3139-3148, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30033597

ABSTRACT

It has been shown that long noncoding RNAs (lncRNAs) are involved in the carcinogenesis of multiple cancers. However, the roles of lncRNAs in gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) remain elusive. In the present study, we found that lncNEN885 was markedly decreased in human gastric NEN samples compared to adjacent normal tissues by transcriptome sequencing. Functionally, silencing or overexpression of lncNEN885 could not obviously affect cell proliferation or apoptosis in BON-1 or LCC-18 cells but could affect cell migration and invasion as well as wound-healing rates. Furthermore, dysregulation of lncNEN885 affected these biological functions by activating epithelial-mesenchymal transition through increased expression of Snail, vimentin, and N-cadherin as well as decreased E-cadherin levels in BON-1 and LCC-18 cells. Silencing of lncNEN885 could dramatically increase the phosphorylation of glycogen synthase kinase-3ß and decrease the expression of adenomatous polyposis coli and Axin, with the subsequent accumulation of ß-catenin. Taken together, dysregulation of lncNEN885 can regulate cell migration and invasion by activating epithelial-mesenchymal transition process partially through canonical Wnt/ß-catenin signaling in GEP-NEN cells, which may be a novel biomarker for the metastasis of GEP-NENs.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Gastrointestinal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Neuroendocrine Tumors/genetics , Pancreatic Neoplasms/genetics , RNA, Long Noncoding/metabolism , Apoptosis/genetics , Cell Movement/genetics , Cell Proliferation/genetics , Gastrointestinal Neoplasms/pathology , Gastrointestinal Neoplasms/surgery , Gene Expression Profiling , Humans , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neuroendocrine Tumors/pathology , Neuroendocrine Tumors/surgery , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/surgery , Phosphorylation , RNA, Long Noncoding/genetics , RNA, Small Interfering , Stomach/pathology , Wnt Signaling Pathway/genetics , beta Catenin/metabolism
10.
Am J Cancer Res ; 8(12): 2575-2589, 2018.
Article in English | MEDLINE | ID: mdl-30662813

ABSTRACT

Incense burning is common in Asian countries due to the religious beliefs. Environmental exposure to incense burning smoke is a potential risk factor for tumor development and progression of non-small cell lung cancer (NSCLC). Eastern Asia ethnic origin is strongly associated the clinical benefits of epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) in NSCLC patients. However, the impact of the oriental custom of incense burning on the cancer progression and the EGFR TKI-sensitivity of NSCLC remains unclear. Our results showed that long-term exposure to incense burning extract (IBE) increases the cellular proliferation with S phase accumulation and the motility activity of NSCLCs. Interestingly, IBE enhances EGFR signaling activity without affecting its genetic status, and increases the cellular sensitivity of NSCLC cell lines to EGFR TKIs. Auramine, a yellow dye for making incense sticks, was identified as a residual composition in the burning incense smoke, and showed similar EGFR TKI-sensitizing effects. Furthermore, IBE or auramine transcriptionally induce EGFR ligand amphiregulin (AREG) expression for the enhancement of EGFR activity. Neutralization of AREG reduced the viability of IBE-treated cells. These results indicated that exposure to incent smoke may enhance NSCLC progression and their sensitivity to EGFR TKIs through increasing their oncogenic addiction to AREG-induced EGFR signaling.

11.
Mol Oncol ; 11(9): 1273-1287, 2017 09.
Article in English | MEDLINE | ID: mdl-28632938

ABSTRACT

Targeting the MEK/ERK pathway has been viewed as a promising strategy for cancer therapy. However, MEK inhibition leads to the compensatory PI3K/AKT activation and thus contributes to the desensitization of cancer cells to MEK inhibitors. The underlying molecular mechanism of this event is not yet understood. In this study, our data showed that the induction of Akt activity by MEK inhibitors was specifically observed in HER2-positive breast cancer cells. Silence of HER2, or overexpression of HER2 kinase-dead mutant, prevents the induction of Akt activation in response to MEK inhibition, indicating HER2 as a critical regulator for this event. Furthermore, HER2 Thr701 was demonstrated as a direct phosphorylation target of ERK1/2. Inhibition of this specific phosphorylation prolonged the dimerization of HER2 with EGFR in a clathrin-dependent manner, leading to the enhanced activation of HER2 and EGFR tyrosine kinase and their downstream Akt pathway. These results suggest that suppression of ERK-mediated HER2 Thr701 phosphorylation contributes to MEK inhibitor-induced Akt activation.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Feedback, Physiological , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Phosphothreonine/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/metabolism , Amino Acid Sequence , Cell Line, Tumor , Clathrin/metabolism , Down-Regulation , Endocytosis/drug effects , Enzyme Activation/drug effects , Epidermal Growth Factor/pharmacology , ErbB Receptors/chemistry , ErbB Receptors/metabolism , Humans , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase Kinases/metabolism , Models, Biological , Phosphorylation/drug effects , Protein Binding , Protein Multimerization/drug effects , Receptor, ErbB-2/chemistry
12.
Oncotarget ; 8(4): 6929-6939, 2017 Jan 24.
Article in English | MEDLINE | ID: mdl-28036293

ABSTRACT

Neuroendocrine neoplasms (NENs) represent relatively rare tumors. The lack of diagnostic, therapeutic method and prognostic factors makes them a challenge to us. We retrospectively reviewed the data of 205 NENs patients among which 157 cases were followed-up. Proprotein convertase subtilisin/kexin 9 (PCSK9), a regulator of low density lipoprotein cholesterol (LDL-C), was confirmed as a target gene of microRNA-224. We found an increased incidence of NENs from 2012 to 2015. Women were usually diagnosed at earlier stages than men (P < 0.05). Tumor grading was associated with primary tumor site, especially esophagus and cardia NENs all at G3 (P <0.001). Age, tumor grading and LDL-C levels were independent risk factors of digestive NENs. Low LDL-C level was significantly correlated with survival rate and median overall survival (OS, P < 0.05). MicroRNA-224 agomir and PCSK9 siRNA could promote apoptosis and suppress proliferation, invasion of BON-1 cells (P < 0.05), but increase the level of glucocorticoid (GC, P < 0.05). Taken together, age, tumor grading and LDL-C level are independent risk factors of NENs. The miR-224/PCSK9/GC axis binds to tumorigenesis and prognosis of pancreatic NENs (p-NENs).


Subject(s)
MicroRNAs/genetics , Neuroendocrine Tumors/epidemiology , Neuroendocrine Tumors/genetics , Proprotein Convertase 9/genetics , Adolescent , Adult , Aged , Animals , Cardia/pathology , Cell Line, Tumor , Cholesterol, LDL/blood , Esophageal Neoplasms/epidemiology , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Incidence , Male , Mice , Middle Aged , Neoplasm Staging , Neuroendocrine Tumors/pathology , Prognosis , Retrospective Studies , Sex Characteristics , Stomach Neoplasms/epidemiology , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Survival Analysis , Young Adult
13.
Biomed Res Int ; 2014: 168949, 2014.
Article in English | MEDLINE | ID: mdl-24707474

ABSTRACT

Lapatinib, a dual EGFR/HER2 tyrosine kinase inhibitor, has been shown to improve the survival rate of patients with advanced HER2-positive breast cancers. However, the off-target activity of lapatinib in inducing EGFR expression without tyrosine kinase activity was demonstrated to render HER2-negative breast cancer cells more metastatic, suggesting a limitation to the therapeutic effectiveness of this dual inhibitor in HER2-heterogeneous tumors. Therefore, targeting EGFR expression may be a feasible approach to improve the anticancer efficiency of lapatinib-based therapy. Inhibition of HDAC has been previously reported to epigenetically suppress EGFR protein expression. In this study, however, our data indicated that treatment with HDAC inhibitors trichostatin A (TSA), but not suberoylanilide hydroxamic acid (SAHA) or HDAC siRNA, can attenuate both protein and mRNA expressions of EGFR in lapatinib-treated triple-negative breast cancer cells, suggesting that TSA may suppress EGFR expression independently of HDAC inhibition. Nevertheless, TSA reduced EGFR 3'UTR activity and induced the gene expression of microRNA-7, a known EGFR-targeting microRNA. Furthermore, treatment with microRNA-7 inhibitor attenuated TSA-mediated EGFR suppression. These results suggest that TSA induced microRNA-7 expression to downregulate EGFR expression in an HDAC-independent manner.


Subject(s)
ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylases/genetics , Hydroxamic Acids/pharmacology , MicroRNAs/genetics , Quinazolines/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/genetics , Humans , Lapatinib , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/genetics , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics
14.
Breast Cancer Res ; 15(6): R108, 2013 Nov 12.
Article in English | MEDLINE | ID: mdl-24216290

ABSTRACT

INTRODUCTION: Triple-negative breast cancer (TNBC), a subtype of breast cancer with negative expressions of estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 (HER2), is frequently diagnosed in younger women and has poor prognosis for disease-free and overall survival. Due to the lack of known oncogenic drivers for TNBC proliferation, clinical benefit from currently available targeted therapies is limited, and new therapeutic strategies are urgently needed. METHODS: Triple-negative breast cancer cell lines were treated with proteasome inhibitors in combination with lapatinib (a dual epidermal growth factor receptor (EGFR)/HER2 tyrosine kinase inhibitor). Their in vitro and in vivo viability was examined by MTT assay, clonogenic analysis, and orthotopic xenograft mice model. Luciferase reporter gene, immunoblot, and RT-qPCR, immunoprecipitation assays were used to investigate the molecular mechanisms of action. RESULTS: Our data showed that nuclear factor (NF)-κB activation was elicited by lapatinib, independent of EGFR/HER2 inhibition, in TNBCs. Lapatinib-induced constitutive activation of NF-κB involved Src family kinase (SFK)-dependent p65 and IκBα phosphorylations, and rendered these cells more vulnerable to NF-κB inhibition by p65 small hairpin RNA. Lapatinib but not other EGFR inhibitors synergized the anti-tumor activity of proteasome inhibitors both in vitro and in vivo. Our results suggest that treatment of TNBCs with lapatinib may enhance their oncogene addiction to NF-κB, and thus augment the anti-tumor activity of proteasome inhibitors. CONCLUSIONS: These findings suggest that combination therapy of a proteasome inhibitor with lapatinib may benefit TNBC patients.


Subject(s)
NF-kappa B/metabolism , Proteasome Inhibitors/pharmacology , Quinazolines/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Boronic Acids/administration & dosage , Boronic Acids/pharmacology , Bortezomib , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride , Female , Gefitinib , Humans , I-kappa B Kinase/metabolism , Lapatinib , Mice, SCID , Phosphorylation/drug effects , Pyrazines/administration & dosage , Pyrazines/pharmacology , Quinazolines/administration & dosage , Receptor, ErbB-2/antagonists & inhibitors , Triple Negative Breast Neoplasms/metabolism , Xenograft Model Antitumor Assays
15.
Mol Pharmacol ; 83(4): 857-69, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23355539

ABSTRACT

Lapatinib, a dual epidermal growth factor receptor (EGFR)/human epidermal growth factor receptor 2 (HER2) kinase inhibitor, showed clinical benefits in advanced HER2-positive breast cancer patients. Because some triple-negative breast cancers (TNBCs) frequently overexpress EGFR, the antitumor activity of lapatinib in such diseases was also tested. However, the results showed a worse event-free survival rate. It remains unknown whether and how lapatinib elicits the aggressiveness of such cancer cells. In this study, our results demonstrated that lapatinib facilitated axillary and lung metastases of triple-negative MDA-MB-231 breast cancer cells without affecting their viability, leading to worse survival in orthotopic xenograft mice. The lapatinib-increased motility was attributed by the elevation of EGFR through the downregulation of microRNA-7 and by the subsequent overexpression of cyclooxygenase-2 (COX-2). Strikingly, independent of its kinase activity, the elevated EGFR at least partly stabilized COX-2 expression by enhancing the binding of HuR to COX-2 mRNA. Our results suggest that lapatinib may increase the migration and invasion of MDA-MB-231 cells by upregulating EGFR and COX-2 through the downregulation of microRNA-7, providing a potential explanation for the worse clinical outcome of TNBC patients who receive lapatinib-based treatment. These findings also shed new light on the molecular mechanism of COX-2 mRNA stabilization by EGFR in a kinase-independent manner.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cyclooxygenase 2/biosynthesis , Gene Expression Regulation, Enzymologic , Neoplasm Invasiveness , Quinazolines/therapeutic use , Receptor, ErbB-2/metabolism , Up-Regulation/genetics , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/genetics , Cell Line, Tumor , ErbB Receptors/metabolism , Female , Humans , Lapatinib , Mice , Quinazolines/pharmacology , Severe Combined Immunodeficiency/genetics , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
16.
PLoS One ; 6(6): e21428, 2011.
Article in English | MEDLINE | ID: mdl-21731744

ABSTRACT

BACKGROUND: The sensitivity of non-small cell lung cancer (NSCLC) patients to EGFR tyrosine kinase inhibitors (TKIs) is strongly associated with activating EGFR mutations. Although not as sensitive as patients harboring these mutations, some patients with wild-type EGFR (wtEGFR) remain responsive to EGFR TKIs, suggesting that the existence of unexplored mechanisms renders most of wtEGFR-expressing cancer cells insensitive. METHODOLOGY/PRINCIPAL FINDINGS: Here, we show that acquired resistance of wtEGFR-expressing cancer cells to an EGFR TKI, gefitinib, is associated with elevated expression of breast cancer resistance protein (BCRP/ABCG2), which in turn leads to gefitinib efflux from cells. In addition, BCRP/ABCG2 expression correlates with poor response to gefitinib in both cancer cell lines and lung cancer patients with wtEGFR. Co-treatment with BCRP/ABCG2 inhibitors enhanced the anti-tumor activity of gefitinib. CONCLUSIONS/SIGNIFICANCE: Thus, BCRP/ABCG2 expression may be a predictor for poor efficacy of gefitinib treatment, and targeting BCRP/ABCG2 may broaden the use of gefitinib in patients with wtEGFR.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/metabolism , Neoplasm Proteins/metabolism , Quinazolines/pharmacology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/genetics , Animals , Cell Line, Tumor , Drug Screening Assays, Antitumor , Gefitinib , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Quinazolines/metabolism , RNA, Small Interfering/metabolism
17.
J Biol Chem ; 286(23): 20558-68, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21487020

ABSTRACT

Epidermal growth factor receptor (EGFR), an aberrantly overexpressed or activated receptor-tyrosine kinase in many cancers, plays a pivotal role in cancer progression and has been an attractive target for cancer therapy. Gefitinib and erlotinib, two EGFR-tyrosine kinase inhibitors, have been approved for non-small cell lung cancer. However, durable clinical efficacy of these EGFR inhibitors is severely limited by the emergence of acquired resistance. For example, the expression of breast cancer-resistant protein (BCRP/ABCG2) has been shown to confer acquired resistance of wild-type EGFR (wtEGFR)-expressing cancer cells to gefitinib. However, the underlying molecular mechanisms still remain unclear. Here, we show that wtEGFR expression is elevated in the nucleus of acquired gefitinib-resistant cancer cells. Moreover, nuclear translocation of EGFR requires phosphorylation at Ser-229 by Akt. In the nucleus, EGFR then targets the proximal promoter of BCRP/ABCG2 and thereby enhances its gene transcription. The nuclear EGFR-mediated BCRP/ABCG2 expression may contribute at least in part to the acquired resistance of wtEGFR-expressing cancer cells to gefitinib. Our findings shed light on the role of nuclear EGFR in the sensitivity of wtEGFR-expressing cancer cells to EGFR tyrosine kinase inhibitors and also deciphered a putative molecular mechanism contributing to gefitinib resistance through BCRP/ABCG2 expression.


Subject(s)
ATP-Binding Cassette Transporters/biosynthesis , Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Cell Nucleus/metabolism , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Neoplasm Proteins/biosynthesis , Proto-Oncogene Proteins c-akt/metabolism , Quinazolines/pharmacology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/genetics , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Nucleus/genetics , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , Female , Gefitinib , Gene Expression Regulation, Neoplastic/genetics , Humans , Neoplasm Proteins/genetics , Phosphorylation/drug effects , Phosphorylation/genetics , Proto-Oncogene Proteins c-akt/genetics
18.
J Med Virol ; 80(6): 989-96, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18428145

ABSTRACT

Multiple interferon-stimulated genes (ISGs) involving T-cell activation are upregulated during initial interferon-alpha-based therapy for chronic hepatitis C virus (HCV) infection. However, the long-term impact on therapeutic outcome in patients remains unknown. In this study, the effects of anti-HCV therapy on the surface expression of HLA-ABC, CD86, and CD28 were longitudinally assessed. These proteins are integral membrane receptors of antigen presentation and triggering of costimulatory signals for activating CD8+ T cells. Peripheral blood mononuclear cells were collected at baseline and post-treatment for 1 day, and 2, 4, 12, and 24 weeks, respectively. This treatment led to a time-related elevation of membrane levels of HLA-ABC and CD86 on B-cells and monocytes in patients with a sustained response (n = 23), but not in those without (n = 8). Meanwhile, upregulation of CD28 on CD4+ and CD8+ T cells was comparable in both groups of sustained responders and non-responders. Steady increases in the B cells' surface and intracellular HLA-ABC were observed, thus, the surface-to-intracellular ratios did not alter over the period of treatment. Furthermore, multivariate analysis shows that increased HLA-ABC on monocytes by week 12 correlates significantly with sustained response (P = 0.033). In conclusion, differential modulation of T-cell activation ISGs, such as HLA-ABC and CD86 might correlate with the outcome of interferon-alpha-based therapy in chronic hepatitis C patients.


Subject(s)
Antigens, Surface/metabolism , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Hepatitis C, Chronic/drug therapy , Interferon-alpha/pharmacology , Leukocytes, Mononuclear/drug effects , Ribavirin/pharmacology , Adult , Aged , Antigens, Surface/genetics , Antiviral Agents/administration & dosage , Biomarkers/analysis , Female , Hepatitis C, Chronic/metabolism , Humans , Interferon alpha-2 , Interferon-alpha/administration & dosage , Interferon-alpha/therapeutic use , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Recombinant Proteins , Ribavirin/administration & dosage , Ribavirin/therapeutic use , Time Factors , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...