Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Int J Med Sci ; 18(8): 1798-1809, 2021.
Article in English | MEDLINE | ID: mdl-33746597

ABSTRACT

Cell migration and invasion are modulated by epithelial-to-mesenchymal transition (EMT) and the reverse MET process. Despite the detection of microRNA-362 (miR-362, both the miR-362-5p and -3p species) in cancers, none of the identified miR-362 targets is a mesenchymal or epithelial factor to link miR-362 with EMT/MET and metastasis. Focusing on the TGF-ß/SMAD signaling pathway in this work, luciferase assays and western blot data showed that miR-362 targeted and negatively regulated expression of SMAD4 and E-cadherin, but not SNAI1, which is regulated by SMAD4. However, miR-362 knockdown also down-regulated SMAD4 and SNAI1, but up-regulated E-cadherin expression. Wound-healing and transwell assays further showed that miR-362 knockdown suppressed cell migration and invasion, effects which were reversed by over-expressing SMAD4 or SNAI1, or by knocking down E-cadherin in the miR-362 knockdown cells. In orthotopic mice, miR-362 knockdown inhibited metastasis, and displayed the same SMAD4 and E-cadherin expression profiles in the tumors as in the in vitro studies. A scheme is proposed to integrate miR-362 negative regulation via SMAD4, and to explain miR-362 positive regulation of SMAD4 via miR-362 targeting of known SMAD4 suppressors, BRK and DACH1, which would have resulted in SMAD4 depletion and annulment of subsequent involvement in TGF-ß signaling actions. Hence, miR-362 both negatively and positively regulates SMAD4 expression in TGF-ß/SMAD signaling pathway to suppress cell motility and invasiveness and metastasis, and may explain the reported clinical association of anti-miR-362 with suppressed metastasis in various cancers. MiR-362 knockdown in miR-362-positive cancer cells may be used as a therapeutic strategy to suppress metastasis.


Subject(s)
MicroRNAs/metabolism , Neoplasms/genetics , Smad4 Protein/genetics , Animals , Cell Line, Tumor , Cell Movement/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Male , Mice , Neoplasm Invasiveness/genetics , Neoplasms/pathology , Signal Transduction/genetics , Smad4 Protein/metabolism , Transforming Growth Factor beta/metabolism , Xenograft Model Antitumor Assays
2.
J Chin Med Assoc ; 87(1): 12-16, 2024 01 01.
Article in English | MEDLINE | ID: mdl-38016117

ABSTRACT

A maternal inheritance disorder called Leber's hereditary optic neuropathy (LHON) is the most common primary mitochondrial deoxyribonucleic acid (DNA) disorder. In most studies, there are more male patients than female patients, which contradicts the usual pattern in mitochondrial hereditary diseases. This suggests that nuclear DNA (nDNA) may influence the degeneration of retinal ganglion cells (RGCs) in LHON. The primary cause of this is dysfunction in complex I of the electron transport chain, leading to ineffective adenosine triphosphate (ATP) production. In addition to MT-ND4 or MT-ND1 mutations, genes such as PRICKLE3 , YARS2 , and DNAJC30 , which come from nDNA, also play a role in LHON. These three genes affect the electron chain transport differently. PRICKLE3 interacts with ATP synthase (complex V) at Xp11.23, while YARS2 is a tyrosyl-tRNA synthetase 2 involved in mitochondria . DNAJC30 mutations result in autosomal recessive LHON (arLHON). Understanding how genes impact the disease is crucial for developing new treatments. Idebenone has been approved for treating LHON and has shown safety and efficacy in clinical trials. Mesenchymal stem cell-based therapy has also emerged as a potential treatment for LHON by transferring mitochondria into target cells. Gene therapy research focuses on specific gene mutations, and the wild-type ND4 gene target in the adeno-associated viruses (AAV) vector has shown promise in clinical trials as a potential treatment for LHON.


Subject(s)
Optic Atrophy, Hereditary, Leber , Humans , Male , Female , Optic Atrophy, Hereditary, Leber/therapy , Optic Atrophy, Hereditary, Leber/drug therapy , DNA, Mitochondrial/genetics , Mitochondria , Mutation , Adenosine Triphosphate/therapeutic use
3.
Gastroenterology ; 141(1): 279-91, 291.e1-5, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21640118

ABSTRACT

BACKGROUND & AIMS: Some cancer cells have activities that are similar to those of stem cells from normal tissues, and cell dedifferentiation correlates with poor prognosis. Little is known about the mechanisms that regulate the stem cell-like features of cancer cells; we investigated genes associated with stem cell-like features of colorectal cancer (CRC) cells. METHODS: We isolated colonospheres from primary CRC tissues and cell lines and characterized their gene expression patterns by microarray analysis. We also investigated the biological features of the colonosphere cells. RESULTS: Expanded CRC colonospheres contained cells that expressed high levels of CD44 and CD166, which are markers of colon cancer stem cells, and had many features of cancer stem cells, including chemoresistance and radioresistance, the ability to initiate tumor formation, and activation of epithelial-mesenchymal transition (EMT). SNAIL, an activator of EMT, was expressed at high levels by CRC colonospheres. Overexpression of Snail in CRC cells induced most properties of colonospheres, including cell dedifferentiation. Two hundred twenty-seven SNAIL-activated genes were up-regulated in colonospheres; gene regulatory networks centered around interleukin (IL)-8 and JUN. Blocking IL-8 expression or activity disrupted SNAIL-induced stem cell-like features of colonospheres. We observed that SNAIL activated the expression of IL8 by direct binding to its E3/E4 E-boxes. In CRC tissues, SNAIL and IL-8 were coexpressed with the stem cell marker CD44 but not with CD133 or CD24. CONCLUSIONS: In human CRC tissues, SNAIL regulates expression of IL-8 and other genes to induce cancer stem cell activities. Strategies that disrupt this pathway might be developed to block tumor formation by cancer stem cells.


Subject(s)
Carcinoma/metabolism , Colorectal Neoplasms/metabolism , Interleukin-8/metabolism , Neoplastic Stem Cells/metabolism , Signal Transduction , Transcription Factors/metabolism , Adult , Aged , Aged, 80 and over , Animals , Antibodies/pharmacology , Antigens, CD/metabolism , Antimetabolites, Antineoplastic/pharmacology , Binding Sites , Carcinoma/drug therapy , Carcinoma/genetics , Carcinoma/immunology , Carcinoma/pathology , Cell Adhesion Molecules, Neuronal/metabolism , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Drug Resistance, Neoplasm , E-Box Elements , Epithelial-Mesenchymal Transition/drug effects , Fetal Proteins/metabolism , Fluorouracil/pharmacology , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , HT29 Cells , Humans , Hyaluronan Receptors/metabolism , Interleukin-8/genetics , Interleukin-8/immunology , Mice , Mice, Nude , Middle Aged , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/radiation effects , Oligonucleotide Array Sequence Analysis , Radiation Tolerance , Signal Transduction/drug effects , Signal Transduction/radiation effects , Snail Family Transcription Factors , Spheroids, Cellular , Time Factors , Transcription Factors/genetics , Transfection , Tumor Burden/drug effects , Tumor Cells, Cultured
4.
Acta Biomater ; 101: 484-494, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31672582

ABSTRACT

Nanodiamonds (NDs) are considered to be relatively safe carbon nanomaterials used for the transmission of DNA, proteins and drugs. The feasibility of utilizing the NDs to deliver CRISPR-Cas9 system for gene editing has not been clearly studied. Therefore, in this study, we aimed to use NDs as the carriers of CRISPR-Cas9 components designed to introduce the mutation in RS1 gene associated with X-linked retinoschisis (XLRS). ND particles with a diameter of 3 nm were functionalized by carboxylation of the surface and covalently conjugated with fluorescent mCherry protein. Two linear DNA constructs were attached to the conjugated mCherry: one encoded Cas9 endonuclease and GFP reporter, another encoded sgRNA and contained insert of HDR template designed to introduce RS1 c.625C>T mutation. Such nanoparticles were successfully delivered and internalized by human iPSCs and mouse retinas, the efficiency of internalization was significantly improved by mixing with BSA. The delivery of ND particles led to introduction of RS1 c.625C>T mutation in both human iPSCs and mouse retinas. Rs1 gene editing in mouse retinas resulted in several pathological features typical for XLRS, such as aberrant photoreceptor structure. To conclude, our ND-based CRISPR-Cas9 delivery system can be utilized as a tool for creating in vitro and in vivo disease models of XLRS. STATEMENT OF SIGNIFICANCE: X-linked retinoschisis (XLRS) is a prevalent hereditary retinal disease, which is caused by mutations in RS1 gene, whose product is important for structural organization of the retina. The recent development of genome editing techniques such as CRISPR-Cas9 significantly improved the prospects for better understanding the pathology and development of treatment for this disease. Firstly, gene editing can allow development of appropriate in vitro and in vivo disease models; secondly, CRISPR-Cas9 can be applied for gene therapy by removing the disease-causative mutation in vivo. The major prerequisite for these approaches is to develop safe and efficient CRISPR-Cas9 delivery system. In this study, we tested specifically modified nanodiamonds for such a delivery system. We were able to introduce Rs1 mutation into the mouse retina and, importantly, observed several XLRS-specific effects.


Subject(s)
CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems/genetics , Gene Transfer Techniques , Induced Pluripotent Stem Cells/metabolism , Mutation/genetics , Nanodiamonds/chemistry , Retina/metabolism , Retinoschisis/genetics , Animals , Base Sequence , Eye Proteins/genetics , Gene Editing , Humans , Male , Mice, Inbred C57BL , Photoreceptor Cells, Vertebrate/pathology
5.
Sci Adv ; 6(43)2020 10.
Article in English | MEDLINE | ID: mdl-33097539

ABSTRACT

Leveraging the endogenous homology-directed repair (HDR) pathway, the CRISPR-Cas9 gene-editing system can be applied to knock in a therapeutic gene at a designated site in the genome, offering a general therapeutic solution for treating genetic diseases such as hemoglobinopathies. Here, a combined supramolecular nanoparticle (SMNP)/supramolecular nanosubstrate-mediated delivery (SNSMD) strategy is used to facilitate CRISPR-Cas9 knockin of the hemoglobin beta (HBB) gene into the adeno-associated virus integration site 1 (AAVS1) safe-harbor site of an engineered K562 3.21 cell line harboring the sickle cell disease mutation. Through stepwise treatments of the two SMNP vectors encapsulating a Cas9•single-guide RNA (sgRNA) complex and an HBB/green fluorescent protein (GFP)-encoding plasmid, CRISPR-Cas9 knockin was successfully achieved via HDR. Last, the HBB/GFP-knockin K562 3.21 cells were introduced into mice via intraperitoneal injection to show their in vivo proliferative potential. This proof-of-concept demonstration paves the way for general gene therapeutic solutions for treating hemoglobinopathies.


Subject(s)
CRISPR-Cas Systems , Hemoglobinopathies , Animals , Gene Editing , Genetic Vectors/genetics , Hemoglobinopathies/genetics , Hemoglobinopathies/therapy , Hemoglobins/genetics , Mice
6.
J Pharmacol Exp Ther ; 330(2): 613-23, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19448140

ABSTRACT

Clodronate, a halogenated bisphosphonate, can inhibit the growth of human thyroid carcinoma (TC) cells. Previously, we found that a clodronate-induced Ca(2+) transient was correlated with clodronate-induced growth inhibition in TC cells. However, the details of the signaling process underlying the antiproliferative effect of clodronate on TC cells are not clear. In this study, we investigated the antiproliferative mechanism of clodronate on papillary TC (PTC) cells and xenotransplanted animals using a combination of pharmacological drugs. Reverse transcription-polymerase chain reaction analysis confirmed the endogenous expression of P2Y receptor isoforms in PTC cells. The P2 antagonist suramin not only inhibited the antiproliferative effect of clodronate and ATP on TC cells but also blocked all the Ca(2+) transients induced by clodronate and ATP. The release of Ca(2+) from the endoplasmic reticulum and membrane depolarization of mitochondria was observed during the clodronate-induced Ca(2+) transients. The results of terminal deoxynucleotidyltransferase dUTP nick-end labeling assays and flow cytometry with annexin V and caspase-3 staining suggest that both ATP and clodronate induce apoptosis. Significant inhibition of tumor invasion and colony formation was also observed in clodronate-treated PTC cells. We further demonstrated that only the cAMP inhibitor 9-(tetrahydro-2-furanyl)-9H-purin-6-amine (SQ22536), and not inhibitors of phospholipase C [1-[6-[[17beta-methoxyestra-1,3,5(10)-trien-17-yl]amino]hexyl]-1H-pyrrole-2,5-dione (U73122)] or store-operated Ca(2+) entry (2-aminoethyl diphenylborinate), can significantly reverse the effect of clodronate. Finally, in vivo animal and green fluorescent protein imaging studies further proved that the tumor inhibitory effect of clodronate on xenotransplanted CG3 cells can be reversed by treatment with suramin. In conclusion, we demonstrated that clodronate-induced PTC cell apoptosis and tumor inhibition are partially mediated by the P2Y receptor-cAMP cascade.


Subject(s)
Apoptosis/physiology , Carcinoma, Papillary/drug therapy , Carcinoma, Papillary/pathology , Clodronic Acid/pharmacology , Receptors, Purinergic P2/physiology , Signal Transduction/physiology , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/pathology , Animals , Apoptosis/drug effects , Bone Density Conservation Agents/pharmacology , Carcinoma, Papillary/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Signal Transduction/drug effects , Thyroid Neoplasms/metabolism
7.
J Biomed Sci ; 16: 100, 2009 Nov 16.
Article in English | MEDLINE | ID: mdl-19917110

ABSTRACT

The progression and metastasis of solid tumors, including head and neck squamous cell carcinoma (HNSCC), have been related to the behavior of a small subpopulation of cancer stem cells. Here, we have established a highly malignant HNSCC cell line, SASVO3, from primary tumors using three sequential rounds of xenotransplantation. SASVO3 possesses enhanced tumorigenic ability both in vitro and in vivo. Moreover, SASVO3 exhibits properties of cancer stem cells, including that increased the abilities of sphere-forming, the number of side population cells, the potential of transplanted tumor growth and elevated expression of the stem cell marker Bmi1. Injection of SASVO3 into the tail vein of nude mice resulted in lung metastases. These results are consistent with the postulate that the malignant and/or metastasis potential of HNSCC cells may reside in a stem-like subpopulation.


Subject(s)
Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Head and Neck Neoplasms/pathology , Tumor Cells, Cultured/cytology , Animals , Cell Movement , Cell Proliferation , Disease Progression , Lentivirus/metabolism , Lung Neoplasms/pathology , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation
8.
Childs Nerv Syst ; 25(5): 543-50, 2009 May.
Article in English | MEDLINE | ID: mdl-19225784

ABSTRACT

OBJECTIVES: Resveratrol (RV), a natural polyphenol derived from red wine, recently showed the potential of anticancer and radiosensitizing effects. A recent study has suggested that the cancer stem cells (CSCs) may reflect the clinical refractory malignancy of brain tumors, including medulloblastoma (MB). The aim of the present study is to investigate the possible role of RV in radiosensitivity of MB cells and MB-associated CSCs. MATERIALS AND METHODS: MB-associated CSCs were isolated and cultured by serum-free medium with basic fibroblast growth factor (bFGF) and epidermal growth factor (EGF). The parental MB cells and MB-CSCs were treated with RV in different concentrations and assessed for cell viability. The treatment includes RV alone, radiation alone, or radiation combined with RV. RESULTS: MB-CSCs selected by serum-free medium with bFGF and EGF can form 3D spheroid formation and display enhanced self-renewal and highly co-expressed "stem cell" genes (Oct-4, Nanog, Nestin, and Musashi-1) as well as antiapoptotic genes (Bcl-2 and Bcl-xL). These MB-CSCs showed significant resistance to radiotherapy as compared to the parental MB cells. Importantly, 100 muM RV could effectively inhibit the proliferation of MB-CSCs and significantly enhance the radiosensitivity in RV-treated MB-CSCs. CONCLUSIONS: Our data suggest that RV can effectively inhibit the proliferation and tumorigenicity of MB-CSCs and significantly synergistically enhance radiosensitivity in RV-treated MB-CSCs.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Cell Proliferation/drug effects , Medulloblastoma/drug therapy , Medulloblastoma/radiotherapy , Neoplastic Stem Cells/drug effects , Stilbenes/therapeutic use , Anticarcinogenic Agents/administration & dosage , Cells, Cultured , Chemotherapy, Adjuvant , Humans , Intermediate Filament Proteins/genetics , Neoplastic Stem Cells/metabolism , Nerve Tissue Proteins/genetics , Nestin , Radiotherapy, Adjuvant , Resveratrol , Reverse Transcriptase Polymerase Chain Reaction , Stilbenes/administration & dosage , bcl-2-Associated X Protein/genetics , bcl-X Protein/genetics
9.
Oncotarget ; 7(27): 42485-42501, 2016 Jul 05.
Article in English | MEDLINE | ID: mdl-27285760

ABSTRACT

Glioblastoma multiform (GBM) is one of the most lethal human malignant brain tumors with high risks of recurrence and poor treatment outcomes. The RNA-binding protein Musashi-1 (MSI1) is a marker of neural stem/progenitor cells. Recent study showed that high expression level of MSI1 positively correlates with advanced grade of GBM, where MSI1 increases the growth of GBM. Herein, we explore the roles of MSI1 as well as the underlying mechanisms in the regulation of drug resistance and tumorigenesis of GBM cells. Our results demonstrated that overexpression of MSI1 effectively protected GBM cells from drug-induced apoptosis through down-regulating pro-apoptotic genes; whereas inhibition of AKT withdrew the MSI1-induced anti-apoptosis and cell survival. We further showed that MSI1 robustly promoted the secretion of the pro-inflammatory cytokine IL-6, which was governed by AKT activity. Autonomously, the secreted IL-6 enhanced AKT activity in an autocrine/paracrine manner, forming a positive feedback regulatory loop with the MSI1-AKT pathway. Our results conclusively demonstrated a novel drug resistance mechanism in GBM cells that MSI1 inhibits drug-induced apoptosis through AKT/IL6 regulatory circuit. MSI1 regulates both cellular signaling and tumor-microenvironmental cytokine secretion to create an intra- and intercellular niche for GBM to survive from chemo-drug attack.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Interleukin-6/metabolism , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA-Binding Proteins/metabolism , Animals , Apoptosis , Cell Line, Tumor , Cell Survival , Computational Biology , Drug Resistance, Neoplasm , Humans , Inflammation , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Recurrence, Local , Neoplasm Transplantation , RNA, Small Interfering/metabolism , Signal Transduction
10.
Biochem Pharmacol ; 81(5): 594-605, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21184746

ABSTRACT

In a previous study, BO-1051, an N-mustard linked with a DNA-affinic molecule, was shown to target various types of cancer cell lines. In the present study, we aimed to investigate the cytotoxicity, as well as the underlying mechanism, of BO-1051. We found that BO-1051 simultaneously induced apoptosis and autophagy in hepatocellular carcinoma cell lines. DNA double strand breaks induced by BO-1051 activated the ATM signaling pathway and subsequently resulted in caspase-dependent apoptosis. When autophagy was inhibited in its early or late stages, apoptosis was significantly enhanced. This result indicated autophagy as a cytoprotective effect against BO-1051-induced cell death. We further inhibited ATM activation using an ATM kinase inhibitor or ATM-specific siRNA and found that while apoptosis was blocked, autophagy also diminished in response to BO-1051. We not only determined a signaling pathway induced by BO-1051 but also clarified the linkage between DNA damage-induced apoptosis and autophagy. We also showed that BO-1051-induced autophagy acts as a cytoprotective reaction and downstream target of the ATM-signaling pathway. This research revealed autophagy as a universal cytoprotective response against DNA damage-inducing chemotherapeutic agents, including BO-1051, cisplatin, and doxorubicin, in hepatocellular carcinoma cell lines. Autophagy contributes to the remarkable drug resistance ability of liver cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Cell Cycle Proteins/physiology , DNA-Binding Proteins/physiology , Nitrogen Mustard Compounds/pharmacology , Protein Serine-Threonine Kinases/physiology , Tumor Suppressor Proteins/physiology , Ataxia Telangiectasia Mutated Proteins , Carcinoma, Hepatocellular , Cell Line, Tumor , DNA Damage , Humans , Liver Neoplasms , Signal Transduction
11.
Dent Mater ; 25(8): 1022-9, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19329175

ABSTRACT

OBJECTIVES: This study was to improve human cell growth on titanium (Ti) used for dental implants through formation of a nano-network surface oxide layer created by an electrochemical anodization treatment. METHODS: An electrochemical anodization treatment was used to produce a network oxide layer on Ti surface. Surface characterization of the network layer was carried out using thin film X-ray diffractometer and field emission scanning electron microscopy. Human bone marrow mesenchymal stem cells (hMSCs) were made to express green fluorescent protein (GFP) by retroviral transduction. The GFP signal was measured in situ to assess in vitro and in vivo cell growth on Ti surfaces. In vivo experiments on Ti-supported cell growth were carried out on the back skin of nude mice. Alizarin red staining and immunofluorescent staining were used to observe cell differentiation. RESULTS: A multilayer TiO(2) nano-network was produced rapidly on Ti surface using a simple electrochemical anodization treatment. The TiO(2) nano-network layer on the anodized Ti surfaces significantly improved in vitro and in vivo hMSC growth, as assessed by measurement of GFP fluorescence, relative to hMSC growth on untreated Ti surface. The TiO(2) nano-network layer on the anodized Ti surfaces can also induce the differentiation of hMSCs after 28-day in vivo test. SIGNIFICANCE: The formation of TiO(2) nano-network on the Ti surfaces can increase the hMSC growth in vitro and in vivo.


Subject(s)
Coated Materials, Biocompatible , Mesenchymal Stem Cells/cytology , Nanostructures , Osteogenesis/physiology , Titanium , Animals , Bone Marrow Cells/chemistry , Cell Adhesion , Cell Differentiation , Cell Proliferation , Cells, Cultured , Electrochemical Techniques , Green Fluorescent Proteins , Humans , Mice , Mice, Nude , Osteoblasts/metabolism , Osteopontin/biosynthesis , Subcutaneous Tissue/surgery , Surface Properties
12.
Int J Radiat Oncol Biol Phys ; 74(1): 219-28, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19362240

ABSTRACT

PURPOSE: CD133 has recently been proposed as a marker for cancer stem-like cells (CSC) in brain tumors. The aim of the present study was to investigate the possible role of resveratrol (RV) in radiosensitivity of CD133-positive/-negative cells derived from atypical teratoid/rhabdoid tumors (AT/RT-CD133(+/-)). MATERIALS AND METHODS: AT/RT-CD133(+/-) were isolated and characterized by flow cytometry and quantitative real-time reverse transcription-polymerase chain reaction, and then treated with RV at different doses. Migratory ability, colony formation, apoptotic activity, and xenotransplantation were assessed for RV alone, ionizing radiation (IR) alone, and IR with RV conditions. RESULTS: AT/RT-CD133(+) displayed enhanced self-renewal and highly coexpressed "stem cell" genes and drug-resistant genes, in addition to showing significant resistance to chemotherapeutic agents and radiotherapy as compared with CD133(-) cells. After treatment with 200 microM RV, the in vitro proliferation rates and in vivo tumor restoration abilities of ATRT-CD133(+) were dramatically inhibited. Importantly, treatment with 150 microM RV can effectively inhibit the expression of drug-resistant genes in AT/RT-CD133(+), and further facilitate to the differentiation of CD133(+) into CD133(-). In addition, treatment with 150 microM RV could significantly enhance the radiosensitivity and IR-mediated apoptosis in RV-treated ATRT-CD133(+/-). Kaplan-Meier survival analysis indicated that the mean survival rate of mice with ATRT-CD133(+) that were treated with IR could be significantly improved when IR was combined with 150 microM RV treatment. CONCLUSIONS: AT/RT-CD133(+) exhibit CSC properties and are refractory to IR treatment. Our results suggest that RV treatment plays crucial roles in antiproliferative, proapoptotic, and radiosensitizing effects on treated-CD133(+/-); RV may therefore improve the clinical treatment of AT/RT.


Subject(s)
Apoptosis/drug effects , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/therapeutic use , Rhabdoid Tumor/radiotherapy , Stilbenes/therapeutic use , Teratoma/radiotherapy , AC133 Antigen , Animals , Antigens, CD/analysis , Antigens, Neoplasm/analysis , Biomarkers, Tumor/analysis , Cell Proliferation , Glycoproteins/analysis , Humans , Mice , Mice, Inbred BALB C , Mice, SCID , Peptides/analysis , Radiation Tolerance/genetics , Resveratrol , Rhabdoid Tumor/chemistry , Rhabdoid Tumor/drug therapy , Rhabdoid Tumor/pathology , Teratoma/chemistry , Teratoma/drug therapy , Teratoma/pathology
13.
PLoS One ; 3(7): e2637, 2008 Jul 09.
Article in English | MEDLINE | ID: mdl-18612434

ABSTRACT

CD133 (prominin-1), a 5-transmembrane glycoprotein, has recently been considered to be an important marker that represents the subset population of cancer stem-like cells. Herein we report the isolation of CD133-positive cells (LC-CD133(+)) and CD133-negative cells (LC-CD133(-)) from tissue samples of ten patients with non-small cell lung cancer (LC) and five LC cell lines. LC-CD133(+) displayed higher Oct-4 expressions with the ability to self-renew and may represent a reservoir with proliferative potential for generating lung cancer cells. Furthermore, LC-CD133(+), unlike LC-CD133(-), highly co-expressed the multiple drug-resistant marker ABCG2 and showed significant resistance to chemotherapy agents (i.e., cisplatin, etoposide, doxorubicin, and paclitaxel) and radiotherapy. The treatment of Oct-4 siRNA with lentiviral vector can specifically block the capability of LC-CD133(+) to form spheres and can further facilitate LC-CD133(+) to differentiate into LC-CD133(-). In addition, knock-down of Oct-4 expression in LC-CD133(+) can significantly inhibit the abilities of tumor invasion and colony formation, and increase apoptotic activities of caspase 3 and poly (ADP-ribose) polymerase (PARP). Finally, in vitro and in vivo studies further confirm that the treatment effect of chemoradiotherapy for LC-CD133(+) can be improved by the treatment of Oct-4 siRNA. In conclusion, we demonstrated that Oct-4 expression plays a crucial role in maintaining the self-renewing, cancer stem-like, and chemoradioresistant properties of LC-CD133(+). Future research is warranted regarding the up-regulated expression of Oct-4 in LC-CD133(+) and malignant lung cancer.


Subject(s)
Antigens, CD/analysis , Glycoproteins/analysis , Lung Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Octamer Transcription Factor-3/metabolism , Peptides/analysis , AC133 Antigen , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Antigens, CD/immunology , Apoptosis , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Survival , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Glycoproteins/immunology , Humans , Neoplasm Proteins/metabolism , Octamer Transcription Factor-3/genetics , Peptides/immunology , RNA, Small Interfering/metabolism , Reverse Transcriptase Polymerase Chain Reaction
14.
J Pharmacol Exp Ther ; 323(1): 365-73, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17646428

ABSTRACT

Combretastatin A4 (CA4) is a drug that targets tumor vasculature to inhibit angiogenesis. Whether CA4 has a direct effect on gastric cancer is not known. We herein investigated the effect of CA4 on growth and metastasis of gastric cancer cells at clinically achievable concentration and explored the associated antitumor mechanisms. Nine human gastric cancer cell lines, including two metastatic gastric cancer cell lines (AGS-GFPM1/2), constitutively expressing green fluorescence protein (GFP) were used. These metastatic AGS-GFPM1/2 cells expressed a higher level of phosphorylated serine 473 on AKT (p-AKT). Our results showed that CA4 (0.02-20 microM) has significant in vitro effects on reducing cell attachment, migration, invasiveness, as well as cell cycle G2/M disturbance on p-AKT-positive gastric cancer cells. In addition, a phosphoinositide 3-kinase inhibitor, LY294002 [2-(4-morpholinyl)-8-phenyl-1(4H)-benzopyran-4-one hydrochloride], a specific AKT inhibitor, and 0.2 to 20 microM CA4 displayed a similar response profile on p-AKT-positive cells, suggesting that CA4-induced effect was mediated by inhibition of the PI3 kinase/AKT pathway. The results from in vivo GFP monitoring system indicated that CA4 phosphate (CA4-P; 200 mg/kg) significantly inhibited the s.c. and intra-abdominal growth of xenotransplanted AGS-GFPM2 cells in nude mice. Furthermore, CA4-P treatment showed a remarkable ability to inhibit gastric tumor metastasis as well as attenuate p-AKT expression. In conclusion, our study is the first to find that CA4 inhibited AKT activity in human gastric cancer cells. The decreased AKT activity correlated well with the CA4 antitumor growth response and decrease of metastasis. Further investigation on drugs targeting the PI3 kinase-AKT pathway may provide a new approach for the treatment of human gastric cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cell Differentiation/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Stilbenes/pharmacology , Stomach Neoplasms , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Stilbenes/therapeutic use , Stomach Neoplasms/drug therapy , Stomach Neoplasms/enzymology , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL