Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 14(7): 423, 2023 07 13.
Article in English | MEDLINE | ID: mdl-37443071

ABSTRACT

Glioblastoma (GBM) is the most malignant tumor in brain and is highly resistant to therapy. Clinical evidence suggests increased number of cancer stem cells (CSCs) may contribute to the failure of conventional therapies, but the mechanisms associated with acquisition of CSC properties in GBM are not fully understood. We found that DAB2IP suppresses CSC properties by targeting the synaptic proteins neuroligin 3 (NLGN3) in GBM. Furthermore, we showed that GBM-derived NLGN3 has an oncogenic function by inducing CSC properties within GBM. Moreover, elevated NLGN3 transcription mediated by Wnt/ß-catenin signaling pathway resulted in increased secretion of NLGN3 into the surrounding tumor microenvironment. Both condition media containing NLGN3 and recombinant NLGN3 transformed neighboring cells to CSCs, suggesting NLGN3 as a critical component inducing CSC properties. Furthermore, targeting NLGN3-bearing CSCs using upstream Wnt/ß-catenin inhibitors synergistically enhances the efficacy of conventional treatment. Hence, we unveiled the series of regulatory mechanisms for acquisition of CSC properties in GBM progression by Wnt/ß-catenin-mediated NLGN3. These results may provide a new targeting strategy to improve the therapeutic efficacy of GBM treatments.


Subject(s)
Glioblastoma , Humans , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Up-Regulation , beta Catenin/metabolism , Wnt Signaling Pathway , Neoplastic Stem Cells/metabolism , Cell Line, Tumor , Cell Proliferation , Tumor Microenvironment , ras GTPase-Activating Proteins/metabolism
2.
Cell Death Discov ; 8(1): 308, 2022 Jul 05.
Article in English | MEDLINE | ID: mdl-35790735

ABSTRACT

Glioblastoma multiforme (GBM) is the most malignant brain tumor and is refractory to conventional therapies. Although previous studies have proposed that the interaction between gene mutations and the external environment leads to the occurrence of GBM, the pathogenesis of GBM is still unclear and much remains to be studied. Herein, we show an association between human glycoprotein stanniocalcin-2 (STC2) and aggressive GBM progression, and demonstrate the underlying mechanism. Elevated STC2 expression and secretion greatly increase GBM cell growth and invasive phenotypes. Mechanistically, both, conditioned media (CM) containing STC2 and recombinant STC2, can induce the transformation of GBM cells into more malignant phenotypes by upregulating the expression of the epithelial-mesenchymal transition transcription factor, snail family transcription repressor 2 (SNAI2) as well as matrix metalloproteinases (MMPs). Moreover, we further demonstrate that the oncogenic function of STC2 in GBM is mediated through the MAPK signaling pathway. Collectively, these results identify the mechanism of STC2 targeting SNAI2 and MMPs through the MAPK pathway in GBM, and provide insights into a potential therapeutic strategy for GBM.

3.
Cell Death Dis ; 11(9): 771, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32943609

ABSTRACT

Temozolomide (TMZ) is widely used for treating glioblastoma multiforme (GBM), however, the treatment of such brain tumors remains a challenge due to the development of resistance. Increasing studies have found that TMZ treatment could induce autophagy that may link to therapeutic resistance in GBM, but, the precise mechanisms are not fully understood. Understanding the molecular mechanisms underlying the response of GBM to chemotherapy is paramount for developing improved cancer therapeutics. In this study, we demonstrated that the loss of DOC-2/DAB2 interacting protein (DAB2IP) is responsible for TMZ-resistance in GBM through ATG9B. DAB2IP sensitized GBM to TMZ and suppressed TMZ-induced autophagy by negatively regulating ATG9B expression. A higher level of ATG9B expression was associated with GBM compared to low-grade glioma. The knockdown of ATG9B expression in GBM cells suppressed TMZ-induced autophagy as well as TMZ-resistance. Furthermore, we showed that DAB2IP negatively regulated ATG9B expression by blocking the Wnt/ß-catenin pathway. To enhance the benefit of TMZ and avoid therapeutic resistance, effective combination strategies were tested using a small molecule inhibitor blocking the Wnt/ß-catenin pathway in addition to TMZ. The combination treatment synergistically enhanced the efficacy of TMZ in GBM cells. In conclusion, the present study identified the mechanisms of TMZ-resistance of GBM mediated by DAB2IP and ATG9B which provides insight into a potential strategy to overcome TMZ chemo-resistance.


Subject(s)
Autophagy , Central Nervous System Neoplasms/drug therapy , Drug Resistance, Neoplasm , Glioblastoma/drug therapy , Temozolomide/pharmacology , Wnt Proteins/metabolism , beta Catenin/metabolism , Antineoplastic Agents, Alkylating/pharmacology , Autophagy-Related Proteins/metabolism , Cell Line, Tumor , Down-Regulation , Gene Expression Regulation, Neoplastic/drug effects , Humans , Inhibitory Concentration 50 , Membrane Proteins/metabolism , MicroRNAs/metabolism , Signal Transduction , Wnt Signaling Pathway/drug effects , ras GTPase-Activating Proteins/metabolism
4.
Cell Death Dis ; 10(11): 834, 2019 11 04.
Article in English | MEDLINE | ID: mdl-31685812

ABSTRACT

A subpopulation of cancer stem cells (CSCs) plays a critical role of cancer progression, recurrence, and therapeutic resistance. Many studies have indicated that castration-resistant prostate cancer (CRPC) is associated with stem cell phenotypes, which could further promote neuroendocrine transdifferentiation. Although only a small subset of genetically pre-programmed cells in each organ has stem cell capability, CSCs appear to be inducible among a heterogeneous cancer cell population. However, the inductive mechanism(s) leading to the emergence of these CSCs are not fully understood in CRPC. Tumor cells actively produce, release, and utilize exosomes to promote cancer development and metastasis, cancer immune evasion as well as chemotherapeutic resistance; the impact of tumor-derived exosomes (TDE) and its cargo on prostate cancer (PCa) development is still unclear. In this study, we demonstrate that the presence of Cav-1 in TDE acts as a potent driver to induce CSC phenotypes and epithelial-mesenchymal transition in PCa undergoing neuroendocrine differentiation through NFκB signaling pathway. Furthermore, Cav-1 in mCRPC-derived exosomes is capable of inducing radio- and chemo-resistance in recipient cells. Collectively, these data support Cav-1 as a critical driver for mCRPC progression.


Subject(s)
Caveolin 1/metabolism , Exosomes/metabolism , Neoplasm Proteins/metabolism , Paracrine Communication , Prostatic Neoplasms, Castration-Resistant/metabolism , Signal Transduction , Animals , Cell Line, Tumor , Exosomes/pathology , Humans , Male , Mice , Mice, SCID , Prostatic Neoplasms, Castration-Resistant/pathology
5.
Clin Cancer Res ; 25(14): 4542-4551, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31000589

ABSTRACT

PURPOSE: Renal cell carcinoma (RCC) is known to be highly radioresistant but the mechanisms associated with radioresistance have remained elusive. We found DOC-2/DAB2 interactive protein (DAB2IP) frequently downregulated in RCC, is associated with radioresistance. In this study, we investigated the underlying mechanism regulating radioresistance by DAB2IP and developed appropriate treatment. EXPERIMENTAL DESIGN: Several RCC lines with or without DAB2IP expression were irradiated with ionizing radiation (IR) for determining their radiosensitivities based on colony formation assay. To investigate the underlying regulatory mechanism of DAB2IP, immunoprecipitation-mass spectrometry was performed to identify DAB2IP-interactive proteins. PARP-1 expression and enzymatic activity were determined using qRT-PCR, Western blot analysis, and ELISA. In vivo ubiquitination assay was used to test PARP-1 degradation. Furthermore, in vivo mice xenograft model and patient-derived xenograft (PDX) model were used to determine the effect of combination therapy to sensitizing tumors to IR. RESULTS: We notice that DAB2IP-deficient RCC cells acquire IR-resistance. Mechanistically, DAB2IP can form a complex with PARP-1 and E3 ligases that is responsible for degrading PARP-1. Indeed, elevated PARP-1 levels are associated with the IR resistance in RCC cells. Furthermore, PARP-1 inhibitor can enhance the IR response of either RCC xenograft model or PDX model. CONCLUSIONS: In this study, we unveil that loss of DAB2IP resulted in elevated PARP-1 protein is associated with IR-resistance in RCC. These results provide a new targeting strategy to improve the efficacy of radiotherapy of RCC.


Subject(s)
Carcinoma, Renal Cell/pathology , Gene Expression Regulation, Neoplastic/radiation effects , Kidney Neoplasms/pathology , Radiation Tolerance/genetics , ras GTPase-Activating Proteins/antagonists & inhibitors , Animals , Apoptosis , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Cell Proliferation , Down-Regulation , Female , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Radiation, Ionizing , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , ras GTPase-Activating Proteins/genetics
6.
Oncotarget ; 8(40): 66975-66986, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28978010

ABSTRACT

Targeted therapy is a standard of care for metastatic renal cell carcinoma (RCC) but the response rate is not overwhelmed, which only prolongs a short survival of patients due to the onset of therapeutic resistance. Although the mechanisms are not fully understood, the presence of cancer initiating cells (CIC) may underlie the drug resistance. Nevertheless, identifying CIC phenotypes with its biomarkers in RCC appear to be diverse and controversial from many reports. In this study, we took a different approach to focus on the regulatory mechanism in RCC-CIC and unveil DAB2IP-mediated miR-138 expression that plays a critical role in modulating stem-like phenotypes in RCC via targeting the ABC transporter (ABCA13) as well as oncogenic histone methyltransferase EZH2 while down regulation of miR-138 gene expression in RCC is due to epigenetic gene silencing by DNA methyltransferase 1 (DNMT1). We also characterize the individual mechanism by which ABCA13 in RCC-CIC contributes to its drug resistance and. EZH2 maintain stem-like phenotypes. Noticeably, elevated expression of ABCA13 and EZH2 is correlated with overall survival of RCC patients, which can be used as potential prognostic markers. Taken together, this study demonstrates a potent and unique pathway of DAB2IP-mediated miR-138 in modulating CIC phenotypes during RCC progression and also offers a new therapeutic strategy of targeting drug resistant RCC.

7.
Cell Death Dis ; 8(3): e2701, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28333136

ABSTRACT

The current agents used for renal cell carcinoma (RCC) only exhibit the moderate response rate among patients. Development of drug resistance eventually fuels the need of either more potent drugs or new drugs to target the resistant pathways. Oridonin is a diterpenoid isolated from the Chinese medicinal herb Rabdosia rubescens and has been shown to have antitumor activities in many cancers. We previously developed new synthetic methodologies to modify structurally diversified diterpenoids and designed a series of nitrogen-enriched oridonin analogs. In this study, we screened a variety of oridonin analogs based on their cytotoxicity using MTT assay and identify the most potent candidate, namely, CYD-6-17. CYD-6-17 exhibited a high potency to inhibit the in vitro growth of several drug-resistant RCC cells as well as endothelial cells stimulated by tumor cells at nanomolar range. Delivery of CYD-6-17 significantly inhibited RCC tumor growth using xenograft model. Mechanistically, it targeted the 3-phosphoinositide-dependent protein kinase 1 gene that appeared to be a potent regulator of AKT and was associated with patient survival after targeted therapies. This offers a new rational therapeutic regimen of CYD-6-17 to drug-resistant RCC based on its novel mechanism of action.


Subject(s)
3-Phosphoinositide-Dependent Protein Kinases/metabolism , Carcinoma, Renal Cell/drug therapy , Diterpenes, Kaurane/pharmacology , Drug Resistance, Neoplasm/drug effects , Animals , Carcinoma, Renal Cell/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Mice , Proto-Oncogene Proteins c-akt/metabolism
8.
ACS Appl Mater Interfaces ; 8(45): 30722-30734, 2016 Nov 16.
Article in English | MEDLINE | ID: mdl-27786455

ABSTRACT

Prostate cancer is one of the leading causes of cancer death in adult men and is a multistage disease with therapeutic challenges of local recurrent advanced tumors and distant metastatic disease. CD44 is a multifunctional and multistructural cell surface glycoprotein that is involved in cell-cell interactions, cell proliferation, and cell migration. In the study, we produced negatively charged and biocompatible hyaluronic acid-based nanoparticles as a therapeutic system for targeting CD44-positive cancer cells. Subsequently, we confirmed the delivery of bioactive epigallocatechin-3-gallate and site-specific inhibition of prostate tumor growth. In this study, hyaluronic acid-based nanoparticles successfully encapsulated epigallocatechin-3-gallate and were efficiently internalized into cancer cells via CD44 ligand receptor recognition, induced cell cycle arrest at G2/M phase, and inhibited prostate cancer cell growth. Furthermore, in vivo assays indicated that these nanoparticles specifically bind CD44 receptors and increase apoptosis of cancer cells, leading to significant decreases in prostate tumor activity and tumor tissue inflammation.


Subject(s)
Nanoparticles , Cell Line, Tumor , Drug Delivery Systems , Humans , Hyaluronan Receptors , Hyaluronic Acid , Male , Prostatic Neoplasms
9.
Oncotarget ; 7(31): 49961-49971, 2016 Aug 02.
Article in English | MEDLINE | ID: mdl-27363023

ABSTRACT

Breast cancer is one of the most prevalent cancers in women, and nearly half of breast cancer patients develop distant metastatic disease after therapy. Despite the significant advances that have been achieved in understanding breast cancer metastasis in the past decades, metastatic cancer is still hard to cure. Here, we demonstrated an anti-cancer mechanism of docosahexaenoic acid (DHA) that suppressed lung metastasis in breast cancer. DHA could inhibit proliferation and invasion of breast cancer cells in vitro, and this was mainly through blocking Cox-2-PGE2-NF-κB-MMPs cascades. DHA treatment significantly decreased Cox-2 and NF-κB expression as well as nuclear translocation of NF-κB in MDA-MB-231 cells. In addition, DHA also reduced NF-κB binding to DNA which may lead to inactivation of MMPs. Moreover, in vivo studies using Fat-1 transgenic mice showed remarkable decrease of tumor growth and metastasis to EO771 cells to lung in DHA-rich environment. In conclusion, DHA attenuated breast cancer progression and lung metastasis in part through suppressing MMPs, and these findings suggest chemoprevention and potential therapeutic strategy to overcome malignant breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Docosahexaenoic Acids/pharmacology , Matrix Metalloproteinase Inhibitors/pharmacology , Neoplasm Metastasis/prevention & control , Active Transport, Cell Nucleus , Animals , Apoptosis , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Survival , Culture Media, Conditioned , Cyclooxygenase 2/metabolism , Dinoprostone/antagonists & inhibitors , Disease Progression , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/metabolism , Neoplasm Invasiveness
10.
Oncotarget ; 7(35): 56842-56854, 2016 Aug 30.
Article in English | MEDLINE | ID: mdl-27472396

ABSTRACT

Conventional chemotherapy is commonly used for advanced stages of transitional cell carcinoma (TCC) with modest success and high morbidity; however, TCC eventually develops resistance. Muscle invasive bladder cancer (MIBC) is recognized as a lethal disease due to its poor response to traditional chemotherapy. Numerous studies have implicated ß-catenin, a critical effector in Wnt-mediated pathway associated with epithelial-mesenchymal transition and cancer stem cell, is involved in TCC progression, and furthermore closely associated with chemo-resistance. In this study, we discovered a novel natural product analogue CYD 6-17 that has a potent inhibitory effect on TCC cells exhibiting drug resistance to various chemotherapeutics, with an IC50 at nM range. Delivery of CYD 6-17 significantly inhibited the tumor growth using xenograft model but without detectable side effects. Mechanistically, it targeted ß-catenin gene transcription by decreasing the binding of XBP1 to the promoter region, which appeared to be a new regulatory mechanism for ß-catenin gene expression. Clinically, XBP1 expression correlated with the poor overall survival of patients. Overall, this study unveils unique mechanism of ß-catenin gene regulation in advanced TCC and also offers a potential rational therapeutic regimen to MIBC.


Subject(s)
Carcinoma, Transitional Cell/drug therapy , Diterpenes, Kaurane/pharmacology , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/metabolism , X-Box Binding Protein 1/metabolism , beta Catenin/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Computational Biology , Diterpenes, Kaurane/chemistry , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Humans , Inhibitory Concentration 50 , Mice , Neoplasm Transplantation , Promoter Regions, Genetic , Treatment Outcome , Wnt Proteins/metabolism
11.
Clin Cancer Res ; 22(3): 670-9, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26490309

ABSTRACT

PURPOSE: Clinical evidence suggests increased cancer stem cells (CSCs) in a tumor mass may contribute to the failure of conventional therapies because CSCs seem to be more resistant than differentiated tumor cells. Thus, unveiling the mechanism regulating CSCs and candidate target molecules will provide new strategy to cure the patients. EXPERIMENTAL DESIGN: The stem-like cell properties were determined by a prostasphere assay and dye exclusion assay. To find critical stem cell marker and reveal regulation mechanism, basic biochemical and molecular biologic methods, such as quantitative real-time PCR, Western blot, reporter gene assay, and chromatin immunoprecipitation assay, were used. In addition, to determine the effect of combination therapy targeting both CSCs and its progeny, in vitro MTT assay and in vivo xenograft model was used. RESULTS: We demonstrate immortalized normal human prostate epithelial cells, appeared nontumorigenic in vivo, become tumorigenic, and acquire stem cell phenotype after knocking down a tumor suppressor gene. Also, those stem-like cells increase chemoresistance to conventional anticancer reagent. Mechanistically, we unveil that Wnt signaling is a key pathway regulating well-known stem cell marker CD44 by directly interacting to the promoter. Thus, by targeting CSCs using Wnt inhibitors synergistically enhances the efficacy of conventional drugs. Furthermore, the in vivo mouse model bearing xenografts showed a robust inhibition of tumor growth after combination therapy. CONCLUSIONS: Overall, this study provides strong evidence of CSC in castration-resistant prostate cancer. This new combination therapy strategy targeting CSC could significantly enhance therapeutic efficacy of current chemotherapy regimen only targeting non-CSC cells.


Subject(s)
Neoplastic Stem Cells/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Animals , Antineoplastic Agents/pharmacology , Biomarkers , CD24 Antigen/genetics , CD24 Antigen/metabolism , Cell Line, Transformed , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Disease Models, Animal , Docetaxel , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Heterografts , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Male , Mice , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Protein Binding , Taxoids/pharmacology , Tumor Stem Cell Assay , Wnt Signaling Pathway , ras GTPase-Activating Proteins/deficiency , ras GTPase-Activating Proteins/genetics
12.
Asian J Urol ; 3(4): 203-210, 2016 Oct.
Article in English | MEDLINE | ID: mdl-29264188

ABSTRACT

Prostate cancer (PCa) is the most common cause of malignancy in males and the second leading cause of cancer mortality in United States. Current treatments for PCa include surgery, radiotherapy, and androgen-deprivation therapy. Eventually, PCa relapses to an advanced castration-resistant PCa (CRPC) that becomes a systematic disease and incurable. Therefore, identifying cellular components and molecular mechanisms that drive aggressive PCa at early stage is critical for disease prognosis and therapeutic intervention. One potential strategy for aggressive PCa is to target cancer stem cells (CSCs) that are identified by several unique characteristics such as immortal, self-renewal, and pluripotency. Also, CSC is believed to be a major factor contributing to resistance to radiotherapy and conventional chemotherapies. Moreover, CSCs are thought to be the critical cause of metastasis, tumor recurrence and cancer-related death of multiple cancer types, including PCa. In this review, we discuss recent progress made in understanding prostate cancer stem cells (PCSCs). We focus on the therapeutic strategies aimed at targeting specific surface markers of CSCs, the key signaling pathways in the maintenance of self-renewal capacity of CSCs, ATP-binding cassette (ABC) transporters that mediate the drug-resistance of CSCs, dysregulated microRNAs expression profiles in CSCs, and immunotherapeutic strategies developed against PCSCs surface markers.

13.
Nat Med ; 21(12): 1445-54, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26523971

ABSTRACT

Focal malformations of cortical development (FMCDs) account for the majority of drug-resistant pediatric epilepsy. Postzygotic somatic mutations activating the phosphatidylinositol-4,5-bisphosphate-3-kinase (PI3K)-protein kinase B (AKT)-mammalian target of rapamycin (mTOR) pathway are found in a wide range of brain diseases, including FMCDs. It remains unclear how a mutation in a small fraction of cells disrupts the architecture of the entire hemisphere. Within human FMCD-affected brain, we found that cells showing activation of the PI3K-AKT-mTOR pathway were enriched for the AKT3(E17K) mutation. Introducing the FMCD-causing mutation into mouse brain resulted in electrographic seizures and impaired hemispheric architecture. Mutation-expressing neural progenitors showed misexpression of reelin, which led to a non-cell autonomous migration defect in neighboring cells, due at least in part to derepression of reelin transcription in a manner dependent on the forkhead box (FOX) transcription factor FOXG1. Treatments aimed at either blocking downstream AKT signaling or inactivating reelin restored migration. These findings suggest a central AKT-FOXG1-reelin signaling pathway in FMCD and support pathway inhibitors as potential treatments or therapies for some forms of focal epilepsy.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Cell Movement , Extracellular Matrix Proteins/metabolism , Forkhead Transcription Factors/metabolism , Malformations of Cortical Development/metabolism , Malformations of Cortical Development/pathology , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Serine Endopeptidases/metabolism , Animals , Base Sequence , Cell Differentiation , Cell Movement/genetics , Disease Models, Animal , Enzyme Activation , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Green Fluorescent Proteins/metabolism , Humans , Magnetic Resonance Imaging , Malformations of Cortical Development/enzymology , Malformations of Cortical Development/surgery , Mice , Molecular Sequence Data , Mosaicism , Mutation/genetics , Neural Stem Cells/metabolism , Neurons/metabolism , Neurons/pathology , Phenotype , RNA, Small Interfering/metabolism , Real-Time Polymerase Chain Reaction , Recombination, Genetic/genetics , Reelin Protein , Signal Transduction/genetics , TOR Serine-Threonine Kinases/metabolism
14.
BMC Cancer ; 14: 36, 2014 Jan 22.
Article in English | MEDLINE | ID: mdl-24447339

ABSTRACT

BACKGROUND: Combination therapy is key to improving cancer treatment efficacy. Phorbol 12-myristate 13-acetate (PMA), a well-known PKC activator, increases the cytotoxicity of several anticancer drugs. Apicularen A induces cytotoxicity in tumor cells through disrupting microtubule networks by tubulin down-regulation. In this study, we examined whether PMA increases apicularen A-induced cytotoxicity in HeLa cells. METHODS: Cell viability was examined by thiazolyl blue tetrazolium (MTT) assays. To investigate apoptotic potential of apicularen A, DNA fragmentation assays were performed followed by extracting genomic DNA, and caspase-3 activity assays were performed by fluorescence assays using fluorogenic substrate. The cell cycle distribution induced by combination with PMA and apicularen A was examined by flow cytometry after staining with propidium iodide (PI). The expression levels of target proteins were measured by Western blotting analysis using specific antibodies, and α-tubulin mRNA levels were assessed by reverse transcription polymerase chain reaction (RT-PCR). To examine the effect of combination of PMA and apicularen A on the microtubule architecture, α-tubulin protein and nuclei were visualized by immunofluorescence staining using an anti-α-tubulin antibody and PI, respectively. RESULTS: We found that apicularen A induced caspase-dependent apoptosis in HeLa cells. PMA synergistically increased cytotoxicity and apoptotic sub-G1 population induced by apicularen A. These effects were completely blocked by the PKC inhibitors Ro31-8220 and Go6983, while caspase inhibition by Z-VAD-fmk did not prevent cytotoxicity. RNA interference using siRNA against PKCα, but not PKCß and PKCγ, inhibited cytotoxicity induced by combination PMA and apicularen A. PMA increased the apicularen A-induced disruption of microtubule networks by further decreasing α- and ß-tubulin protein levels in a PKC-dependent manner. CONCLUSIONS: These results suggest that the synergy between PMA and apicularen A is involved by PKCα activation and microtubule disruption, and that may inform the development of novel approaches to treat cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Microtubules/drug effects , Uterine Cervical Neoplasms/metabolism , Apoptosis/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Caspase 3/metabolism , Cell Cycle Checkpoints , Cell Survival/drug effects , Drug Synergism , Female , HeLa Cells , Humans , Microtubules/genetics , Microtubules/metabolism , Microtubules/pathology , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase C-alpha/genetics , Protein Kinase C-alpha/metabolism , Protein Kinase Inhibitors/pharmacology , RNA Interference , Tetradecanoylphorbol Acetate/pharmacology , Time Factors , Transfection , Tubulin/genetics , Tubulin/metabolism , Tubulin Modulators/pharmacology , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology
15.
Biochem Biophys Res Commun ; 434(3): 634-40, 2013 May 10.
Article in English | MEDLINE | ID: mdl-23583412

ABSTRACT

Apicularen A is a novel antitumor agent and strongly induces death in tumor cells. In this study, we synthesized apicularen A acetate, an acetyl derivative of apicularen A, and investigated its antitumor effect and mechanism in HM7 colon cancer cells. Apicularen A acetate induced apoptotic cell death and caspase-3 activation; however, the pan-caspase inhibitor Z-VAD-fmk could not prevent this cell death. Apicularen A acetate induced the loss of mitochondrial membrane potential and the translocation of apoptosis-inducing factor (AIF) from mitochondria. In addition, apicularen A acetate significantly decreased tubulin mRNA and protein levels and induced disruption of microtubule networks. Taken together, these results indicate that the mechanism of apicularen A acetate involves caspase-independent apoptotic cell death and disruption of microtubule architecture.


Subject(s)
Apoptosis Inducing Factor/metabolism , Apoptosis/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Colonic Neoplasms/pathology , Down-Regulation/drug effects , Microtubules/drug effects , Tubulin/metabolism , Blotting, Western , Cell Line, Tumor , Colonic Neoplasms/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Humans , Membrane Potential, Mitochondrial/drug effects , Microtubules/metabolism , Protein Transport , Reverse Transcriptase Polymerase Chain Reaction
16.
Mol Endocrinol ; 26(9): 1496-507, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22745193

ABSTRACT

Prostate cancer (PCa) becomes lethal when cancer cells develop into castration-resistant PCa (CRPC). Androgen receptor (AR) gene mutation, altered AR regulation, or overexpression of AR often found in CRPC is believed to become one of the key factors to the lethal phenotype. Here we identify Slug, a member of the Snail family of zinc-finger transcription factors associated with cancer metastasis, as a unique androgen-responsive gene in PCa cells. In addition, the presence of constitutively active AR can induce Slug expression in a ligand-independent manner. Slug overexpression will increase AR protein expression and form a complex with AR. In addition, Slug appears to be a novel coactivator to enhance AR transcriptional activities and AR-mediated cell growth with or without androgen. In vivo, elevated Slug expression provides a growth advantage for PCa cells in androgen-deprived conditions. Most importantly, these observations were validated by several data sets from tissue microarrays. Overall, there is a reciprocal regulation between Slug and AR not only in transcriptional regulation but also in protein bioactivity, and Slug-AR complex plays an important role in accelerating the androgen-independent outgrowth of CRPC.


Subject(s)
Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Transcription Factors/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Fluorescent Antibody Technique , Humans , Immunoprecipitation , Male , Mice , Mice, SCID , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , Reverse Transcriptase Polymerase Chain Reaction , Snail Family Transcription Factors , Transcription Factors/genetics
17.
Pancreatology ; 11(6): 574-84, 2011.
Article in English | MEDLINE | ID: mdl-22213040

ABSTRACT

BACKGROUND/AIMS: ω3-polyunsaturated fatty acids (ω3- PUFAs) are known to possess anticancer properties. However, the relationship between ω3-PUFAs and ß-catenin, one of the key components of the Wnt signaling pathway, in human pancreatic cancer remains poorly characterized. METHODS: Human pancreatic cancer cells (SW1990 and PANC-1) were exposed to two ω3-PUFAs, docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), to investigate the relationship between ω3-PUFAs and the Wnt/ß-catenin signaling pathway in vitro. Mouse pancreatic cancer (PANC02) cells were implanted into fat-1 transgenic mice, which express ω3 desaturases and result in elevated levels of ω3-PUFAs endogenously. The tumor size, levels of Wnt/ß-catenin signaling molecules and apoptosis levels were analyzed to examine the influence of ω3-PUFAs in vivo. RESULTS: DHA and EPA significantly inhibited cell growth and increased cell death in pancreatic cancer cells. DHA also reduced ß-catenin expression, T cell factor/lymphoid-enhancing factor reporter activity and induced ß-catenin/Axin/GSK-3ß complex formation, a known precursor to ß-catenin degradation. Furthermore, Wnt3a, a natural canonical Wnt pathway ligand, reversed DHA-induced growth inhibition in PANC-1 cells. Immunohistochemical analysis showed aberrant upregulation and increased nuclear staining of ß-catenin in tumor tissues from pancreatic cancer patients. However, ß-catenin levels in tumor tissues from fat-1 transgenic mice were reduced with a significant increase in apoptosis compared with those from control mice. CONCLUSION: ω3-PUFAs may be an effective therapy for the chemoprevention and treatment of human pancreatic cancer. and IAP.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/pharmacology , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Pancreatic Neoplasms/drug therapy , Wnt Signaling Pathway/drug effects , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Down-Regulation/drug effects , Drug Screening Assays, Antitumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, Transgenic , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Wnt Signaling Pathway/genetics
18.
Int J Radiat Oncol Biol Phys ; 73(3): 853-60, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19215821

ABSTRACT

PURPOSE: The mechanisms of thymidine phosphorylase (TP) regulation induced by radiation therapy (XRT) in various tumors are poorly understood. We investigated the effect and mechanisms of preoperative XRT on TP expression in rectal cancer tissues. METHODS AND MATERIALS: TP expression and CD68 and monocyte chemoattractant protein-1 (MCP-1) levels in rectal cancer tissues and cancer cell lines were evaluated before and after XRT in Western blotting, immunohistochemistry, enzyme-linked immunoassay, and reverse transcription-polymerase chain reaction studies. Isolated peripheral blood monocytes were used in the study of chemotaxis under the influence of MCP-1 released by irradiated colon cancer cells. RESULTS: Expression of TP was significantly elevated by 9 Gy of XRT in most rectal cancer tissues but not by higher doses of XRT. In keeping with the close correlation of the increase in both TP expression and the number of tumor-associated macrophages (TAMs), anti-TP immunoreactivity was found in the CD68-positive TAMs and not the neoplastic cells. Expression of MCP-1 was increased in most cases after XRT, and this increase was strongly correlated with TP expression. However, this increase in MCP-1 expression occurred in tumor cells and not stromal cells. The XRT upregulated MCP-1 mRNA and also triggered the release of MCP-1 protein from cultured colon cancer cells. The supernatant of irradiated colon cancer cells showed strong chemotactic activity for monocyte migration, but this activity was completely abolished by neutralizing antibody. CONCLUSIONS: Use of XRT induces MCP-1 expression in cancer cells, which causes circulating monocytes to be recruited into TAMs, which then upregulate TP expression in rectal cancer tissues.


Subject(s)
Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Chemokine CCL2/metabolism , Monocytes/physiology , Rectal Neoplasms/metabolism , Thymidine Phosphorylase/metabolism , Up-Regulation/radiation effects , Cell Line, Tumor/enzymology , Cell Line, Tumor/radiation effects , Cell Migration Assays, Leukocyte/methods , Cell Migration Assays, Macrophage , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , HT29 Cells/enzymology , HT29 Cells/radiation effects , Humans , Macrophages/physiology , RNA, Messenger/metabolism , Radiotherapy Dosage , Rectal Neoplasms/radiotherapy , U937 Cells/enzymology , U937 Cells/radiation effects , Up-Regulation/physiology
19.
Autophagy ; 4(5): 650-8, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18424913

ABSTRACT

Rottlerin is widely used as a protein kinase C-delta inhibitor. Recently, several reports have shown the possible apoptosis-inducing effect of rottlerin in some cancer cell lines. Here we report that rottlerin induces not only apoptosis but also autophagy via a PKC-delta-independent pathway in HT1080 human fibrosarcoma cells. Rottlerin treatment induced a dose- and time-dependent inhibition of cell growth, and cytoplasmic vacuolations were markedly shown. These vacuoles were identified as acidic autolysosomes by electron microscopy, acidic vesicular organelle (AVO) staining and transfection of green fluorescent protein-LC3. The LC3-II protein level also increased after treatment with rottlerin. Prolonged exposure to rottlerin eventually caused apoptosis via loss of mitochondrial membrane potential and translocation of AIF from mitochondria to the nucleus. However, the activities of caspase-3, -8 and -9 were not changed, and PARP did not show signs of cleavage. Interestingly, the pretreatment of cells with a specific inhibitor of autophagy (3-methyladenine) accelerated rottlerin-induced apoptosis as revealed by an analysis of the subdiploid fraction and TUNEL assay. Nevertheless, the knockdown of PKC-delta by RNA interference neither affected cell growth nor acidic vacuole formation. Similarly, rottlerin-induced cell death was not prevented by PKC-delta overexpression. Taken together, these findings suggest that rottlerin induces early autophagy and late apoptosis in a PKC-delta-independent manner, and the rottlerin-induced early autophagy may act as a survival mechanism against late apoptosis in HT1080 human fibrosarcoma cells.


Subject(s)
Acetophenones/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Benzopyrans/pharmacology , Fibrosarcoma/enzymology , Fibrosarcoma/pathology , Protein Kinase C-delta/antagonists & inhibitors , Signal Transduction/drug effects , Apoptosis/physiology , Autophagy/physiology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Fibrosarcoma/drug therapy , Humans , Intracellular Membranes/drug effects , Intracellular Membranes/physiology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Protein Kinase C-delta/physiology , Signal Transduction/physiology
20.
Mol Med Rep ; 1(5): 763-8, 2008.
Article in English | MEDLINE | ID: mdl-21479483

ABSTRACT

It has been reported that the SW480 human colon cancer cell line consists of E-type and R-type cells. The long-term tumorigenic potential, invasive and metastatic properties of these subclones have not been characterized. E-type and R-type cells were subcloned using limiting dilution methods from parental SW480 cells. The cell growth rate was determined by MTT colorimetric assay, and colony forming efficiency was analyzed using Matrigel-coated plates. The activity of matrix metalloproteinase (MMP) and of urokinase plasminogen activator (uPA) was assessed by zymography. Invasive and locomotive ability was analyzed using transwell chambers. In situ apoptosis detection of these subclones was also performed. In vivo long-term tumorigenicity and nodal metastasis were evaluated using nude mice. E-type cells produced spontaneously regressive tumors in spite of invasion and lymph node metastasis. In contrast, R-type cells revealed progressively growing tumors without invasion or metastasis. E-type cells exhibited increased apoptosis and invasive and motile ability, as well as strong MMP-9 and -2 activity. Although phorbol 12-myristate 13-acetate treatment induced MMP-9 activity in E-type cells, it had no effect on R-type cells. These findings suggest that E- and R-type cells may have different biological properties in terms of colon cancer progression, regression, invasion and nodal metastasis, and might serve as a useful model for these studies.

SELECTION OF CITATIONS
SEARCH DETAIL
...