Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Mol Ther ; 23(4): 746-56, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25619723

ABSTRACT

Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related death in the world. The multikinase inhibitor sorafenib only demonstrated marginal improvement in overall survival for advanced disease prompted the search for alternative treatment options. Human mesenchymal stem cells (MSCs) have the ability to home to tumor cells. However, its functional roles on the tumor microenvironment remain controversial. Herein, we showed that conditioned media derived from human fetal MSC (CM-hfMSCs) expressed high level of the insulin growth factor binding proteins IGFBPs and can sequester free insulin-like growth factors (IGFs) to inhibit HCC cell proliferation. The inhibitory effect of IGFBPs on IGF signaling was further evident from the reduction of activated IGF-1R and PI3K/Akt, leading eventually to the induction of cell cycle arrest. We also demonstrated that CM-hfMSCs could enhance the therapeutic efficacy of sorafenib and sunitinib. To the best of our knowledge, this is the first report to show that CM-hfMSCs has a tumor-specific, antiproliferative effect that is not observed with normal human hepatocyte cells and patient-derived matched normal tissues. Our results thus suggest that CM-hfMSCs can provide a useful tool to design alternative/adjuvant treatment strategies for HCC, especially in related function to potentiate the effects of chemotherapeutic drugs.


Subject(s)
Carcinoma, Hepatocellular/pathology , Fetus/cytology , Liver Neoplasms/pathology , Mesenchymal Stem Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, IGF Type 1/metabolism , Signal Transduction , Carcinoma, Hepatocellular/drug therapy , Cell Proliferation , Culture Media, Conditioned , Gene Knockdown Techniques , Humans , Indoles/therapeutic use , Liver Neoplasms/drug therapy , Niacinamide/analogs & derivatives , Niacinamide/therapeutic use , Phenylurea Compounds/therapeutic use , Pyrroles/therapeutic use , Receptor, IGF Type 1/genetics , Sorafenib , Sunitinib
2.
BMC Cancer ; 15: 255, 2015 Apr 11.
Article in English | MEDLINE | ID: mdl-25886314

ABSTRACT

BACKGROUND: The treatment of glioblastoma multiforme (GBM) is an unmet clinical need. The 5-year survival rate of patients with GBM is less than 3%. Temozolomide (TMZ) remains the standard first-line treatment regimen for gliomas despite the fact that more than 90% of recurrent gliomas do not respond to TMZ after repeated exposure. We have also independently shown that many of the Asian-derived glioma cell lines and primary cells derived from Singaporean high-grade glioma patients are indeed resistant to TMZ. This issue highlights the need to develop new effective anti-cancer treatment strategies. In a recent study, wild-type epidermal growth factor receptor (wtEGFR) has been shown to phosphorylate a truncated EGFR (known as EGFRvIII), leading to the phosphorylation of STAT proteins and progression in gliomagenesis. Despite the fact that combination of EGFR targeting drugs and rapamycin has been used before, the effect of mono-treatment of Nimotuzumab, rapamycin and combination therapy in human glioma expressing different types of EGFR is not well-studied. Herein, we evaluated the efficacy of dual blockage using monoclonal antibody against EGFR (Nimotuzumab) and an mTOR inhibitor (rapamycin) in Caucasian patient-derived human glioma cell lines, Asian patient-derived human glioma cell lines, primary glioma cells derived from the Mayo GBM xenografts, and primary short-term glioma culture derived from high-grade glioma patients. METHODS: The combination effect of Nimotuzumab and rapamycin was examined in a series of primary human glioma cell lines and glioma cell lines. The cell viability was compared to TMZ treatment alone. Endogenous expressions of EGFR in various GBM cells were determined by western blotting. RESULTS: The results showed that combination of Nimotuzumab with rapamycin significantly enhanced the therapeutic efficacy of human glioma cells compared to single treatment. More importantly, many of the Asian patient-derived glioma cell lines and primary cells derived from Singaporean high-grade gliomas, which showed resistance to TMZ, were susceptible to the combined treatments. CONCLUSIONS: In conclusion, our results strongly suggest that combination usage of Nimotuzumab and rapamycin exert higher cytotoxic activities than TMZ. Our data suggest that this combination may provide an alternative treatment for TMZ-resistant gliomas regardless of the EGFR status.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Sirolimus/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Apoptosis/drug effects , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Drug Resistance, Neoplasm/genetics , ErbB Receptors/genetics , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Mutation , Temozolomide
3.
FASEB J ; 28(10): 4359-68, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25271298

ABSTRACT

Human bone marrow-derived mesenchymal stem cells (MSCs) have the unique ability to home toward injuries or tumor sites. We have previously shown that the tumor-tropic property is dependent on the intrinsic expression and activity of the matrix remodeling gene, matrix metalloproteinase 1 (MMP-1). Herein, crosstalk between MMP-1/protease activated receptor 1 (PAR-1) and the G-protein coupled receptor stromal-derived growth factor 1 (SDF-1)/C-X-C chemokine receptor 4 (CXCR-4) in facilitating cell migration was investigated. Gain-of-function and RNA interference (RNAi) technology were used to evaluate the interplay between the key players. The downstream effect on the tumor-tropic migration of MSCs was investigated using modified Boyden chamber assay. Neutralizing PAR-1 activation using monoclonal antibody and targeted knockdown of MMP-1 using RNAi resulted in decreased expression of SDF-1, which was not observed in control-RNAi-transfected cells. Overexpression of CXCR-4 failed to promote MSC migration; the percentage of migrated cells toward tumor cell conditioned medium was similar to the vector-transduced and the CXCR-4-transduced MSCs. Furthermore, inhibition of SDF-1/CXCR-4 signaling using AMD3100 reduced MSC migration through the deregulation of MMP-1 promoter activities, protein expression, and metalloproteinase activity. Collectively, our results showed that MMP-1-mediated MSC tumor tropism is dependent on crosstalk with the SDF-1/CXCR-4 axis.


Subject(s)
Cell Movement , Chemokine CXCL12/metabolism , Matrix Metalloproteinase 1/metabolism , Mesenchymal Stem Cells/metabolism , Receptors, CXCR4/metabolism , Tumor Microenvironment , Cells, Cultured , Chemokine CXCL12/genetics , Female , Humans , Male , Matrix Metalloproteinase 1/genetics , Mesenchymal Stem Cells/physiology , Middle Aged , Receptor, PAR-1/genetics , Receptor, PAR-1/metabolism , Receptors, CXCR4/genetics
4.
Stem Cells ; 31(1): 146-55, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23034897

ABSTRACT

Tumor tropism of human bone marrow-derived mesenchymal stem cells (MSC) has been exploited for the delivery of therapeutic genes for anticancer therapy. However, the exact contribution of these cells in the tumor microenvironment remains unknown. In this study, we examined the biological effect of MSC on tumor cells. The results showed that MSC inhibited the growth of human glioma cell lines and patient-derived primary glioma cells in vitro. Coadministration of MSC and glioma cells resulted in significant reduction in tumor volume and vascular density, which was not observed when glioma was injected with immortalized normal human astrocytes. Using endothelial progenitor cells (EPC) from healthy donors and HUVEC endothelial cells, the extent of EPC recruitment and capacity to form endothelial tubes was significantly impaired in conditioned media derived from MSC/glioma coculture, suggesting that MSC suppressed tumor angiogenesis through the release of antiangiogenic factors. Further studies using antibody array showed reduced expression of platelet-derived growth factor (PDGF)-BB and interleukin (IL)-1ß in MSC/glioma coculture when compared with controls. In MSC/glioma coculture, PDGF-BB mRNA and the corresponding proteins (soluble and membrane bound forms) as well as the receptors were found to be significantly downregulated when compared with that of glioma cocultured with normal human astrocytes or glioma monoculture. Furthermore, IL-1ß, phosphorylated Akt, and cathepsin B proteins were also reduced in MSC/glioma. Taken together, these data indicated that the antitumor effect of MSC may be mediated through downregulation of PDGF/PDGFR axis, which is known to play a key role in glioma angiogenesis. STEM Cells2013;31:146-155.


Subject(s)
Glioma/pathology , Mesenchymal Stem Cells/physiology , Neovascularization, Pathologic , Proto-Oncogene Proteins c-sis/genetics , Receptors, Platelet-Derived Growth Factor/genetics , Animals , Astrocytes , Becaplermin , Bone Marrow Cells/physiology , Brain Neoplasms/metabolism , Brain Neoplasms/therapy , Cathepsin B/biosynthesis , Cell Line, Tumor , Coculture Techniques , Down-Regulation , Glioma/therapy , Human Umbilical Vein Endothelial Cells , Humans , Interleukin-1beta/biosynthesis , Mesenchymal Stem Cell Transplantation , Mice , Mice, Inbred BALB C , Mice, Nude , Proto-Oncogene Proteins c-akt/biosynthesis , Proto-Oncogene Proteins c-sis/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Platelet-Derived Growth Factor/biosynthesis , Tumor Microenvironment
5.
Cancers (Basel) ; 13(15)2021 Jul 22.
Article in English | MEDLINE | ID: mdl-34359588

ABSTRACT

Glioblastoma (GBM) accounts for more than 50% of all primary malignancies of the brain. Current standard treatment regimen for GBM includes maximal surgical resection followed by radiation and adjuvant chemotherapy. However, due to the heterogeneity of the tumor cells, tumor recurrence is often inevitable. The prognosis of patients with glioma is, thus, dismal. Glioma is a highly angiogenic tumor yet immunologically cold. As such, evolving studies have focused on designing strategies that specifically target the tyrosine kinase receptors of angiokines and encourage immune infiltration. Recent promising results from immunotherapies on other cancer types have prompted further investigations of this therapy in GBM. In this article, we reviewed the pathological angiogenesis and immune reactivity in glioma, as well as its target for drug development, and we discussed future directions in glioma therapy.

6.
J Gene Med ; 12(10): 848-58, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20963807

ABSTRACT

BACKGROUND: Herpes simplex virus type-1 (HSV-1) amplicon vectors are attractive tools for gene transfer because of their large DNA insert capacity, their broad host range of vector transduction and a minimal immune response as a result of the absence of helper viruses during viral packaging. However, the transient gene expression remains a challenge for the translation of HSV-1 amplicon based therapeutic strategies to a clinical setting. Although oriP/EBV nuclear antigen (EBNA)-1 elements of Epstein-Barr virus (EBV) have been successfully employed to achieve prolonged transgene expression, little is known about the stability of the EBNA-1 elements in the context of HSV-1 amplicon viral vectors. METHODS: We have generated HSV/EBV hybrid vectors expressing the mutant EBNA-1 gene with the luciferase reporter gene bicistronically to enable monitoring of EBNA-1 expression in real-time, both in vitro and in vivo. RESULTS: The results obtained showed that the HSV/EBV hybrid vectors could mediate high levels of transgene expression (ranging from approximately two-fold to nine-fold) in primary human tumor cells and human bone marrow-derived mesenchymal stem cells compared to the control vector. Prolonged transgene expression could also be observed in primary patient-derived human hepatocellular carcinoma xenografts and in the mouse brain parenchyma up to a period of 17 and 365 days, respectively. CONCLUSIONS: Taken together, we have demonstrated that these hybrid vectors could be promising tools as carriers of therapeutic genes in mesenchymal stem cells or even provide an alternative non-integrating platform for the generation of induced pluripotent stem cells.


Subject(s)
Genetic Vectors , Herpesvirus 4, Human/genetics , Mesenchymal Stem Cells/virology , Simplexvirus/genetics , Transgenes , Animals , Brain Neoplasms/pathology , Carcinoma, Hepatocellular/pathology , Cell Line , Cell Line, Tumor , Chimera , Epstein-Barr Virus Nuclear Antigens/genetics , Epstein-Barr Virus Nuclear Antigens/metabolism , Feasibility Studies , Female , Gene Expression , Genes, Reporter , Glioma/pathology , HeLa Cells , Helper Viruses , Humans , Liver Neoplasms/pathology , Male , Mice , Mice, Nude , Mice, SCID , Transduction, Genetic , Xenograft Model Antitumor Assays/methods
7.
Stem Cells ; 27(6): 1366-75, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19489099

ABSTRACT

Human mesenchymal stem cells (MSCs) have increasingly been used as cellular vectors for the delivery of therapeutic genes to tumors. However, the precise mechanism of mobilization remains poorly defined. In this study, MSCs that expressed similar cell surface markers and exhibited multilineage differentiation potentials were isolated from various donors. Interestingly, different MSC isolates displayed differential migration ability toward human glioma cells. We hypothesized that distinct molecular signals may be involved in the varied tumor tropisms exhibited by different MSC isolates. To test this hypothesis, gene expression profiles of tumor-trophic MSCs were compared with those of non-tumor-trophic MSCs. Among the various differentially regulated genes, matrix metalloproteinase one (MMP1) gene expression and its protein activities were enhanced by 27-fold and 21-fold, respectively, in highly migrating MSCs compared with poorly migrating MSCs. By contrast, there was no change in the transcriptional levels of other MMPs. Functional inactivation of MMP1 abrogated the migratory potential of MSCs toward glioma-conditioned medium. Conversely, the nonmigratory phenotype of poorly migrating MSC could be rescued in the presence of either recombinant MMP1 or conditioned medium from the highly migrating MSCs. Ectopic expression of MMP1 in these poorly migrating cells also rendered the cells responsive to the signaling cues from the glioma cells in vivo. However, blocking the interaction of MMP1 and its cognate receptor PAR1 effectively diminished the migratory ability of MSCs. Taken together, this study provides, for the first time, supporting evidence that MMP1 is critically involved in the migration capacity of MSCs, acting through the MMP1/PAR1 axis.


Subject(s)
Cell Movement/physiology , Glioma/metabolism , Matrix Metalloproteinase 1/metabolism , Mesenchymal Stem Cells/metabolism , Bone Marrow Cells/cytology , Enzyme-Linked Immunosorbent Assay , Gene Expression Profiling , Gene Transfer Techniques , Humans , Mesenchymal Stem Cell Transplantation , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Transfection
8.
Oncogene ; 38(27): 5367-5380, 2019 07.
Article in English | MEDLINE | ID: mdl-30967630

ABSTRACT

Glioblastoma (GBM) is the most aggressive tumor of the brain. NF1, a tumor suppressor gene and RAS-GTPase, is one of the highly mutated genes in GBM. Dysregulated NF1 expression promotes cell invasion, proliferation, and tumorigenesis. Loss of NF1 expression in glioblastoma is associated with increased aggressiveness of the tumor. Here, we show that NF1-loss in patient-derived glioma cells using shRNA increases self-renewal, heightens cell invasion, and promotes mesenchymal subtype and epithelial mesenchymal transition-specific gene expression that enhances tumorigenesis. The neurofibromin protein contains at least four major domains, with the GAP-related domain being the most well-studied. In this study, we report that the leucine-rich domain (LRD) of neurofibromin inhibits invasion of human glioblastoma cells without affecting their proliferation. Moreover, under conditions tested, the NF1-LRD fails to hydrolyze Ras-GTP to Ras-GDP, suggesting that its suppressive function is independent of Ras signaling. We further demonstrate that rare variants within the NF1-LRD domain found in a subset of the patients are pathogenic and reduce NF1-LRD's invasion suppressive function. Taken together, our results show, for the first time, that NF1-LRD inhibits glioma invasion, and provides evidence of a previously unrecognized function of NF1-LRD in glioma biology.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Leucine/metabolism , Mutation , Neoplasm Invasiveness/genetics , Neurofibromin 1/genetics , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Glioblastoma/pathology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neurofibromin 1/metabolism
9.
Mol Cancer Res ; 5(7): 655-65, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17634421

ABSTRACT

Angiopoietins (ANG-1 and ANG-2) and their TIE-2 receptor tyrosine kinase have wide-ranging effects on tumor malignancy that includes angiogenesis, inflammation, and vascular extravasation. These multifaceted pathways present a valuable opportunity in developing novel inhibition strategies for cancer treatment. However, the regulatory role of ANG-1 and ANG-2 in tumor angiogenesis remains controversial. There is a complex interplay between complementary yet conflicting roles of both the ANGs in shaping the outcome of angiogenesis. Embryonic vascular development suggests that ANG-1 is crucial in engaging interaction between endothelial and perivascular cells. However, recruitment of perivascular cells by ANG-1 has recently been implicated in its antiangiogenic effect on tumor growth. It is becoming clear that TIE-2 signaling may function in a paracrine and autocrine manner directly on tumor cells because the receptor has been increasingly found in tumor cells. In addition, alpha(5)beta(1) and alpha(v)beta(5) integrins were recently recognized as functional receptors for ANG-1 and ANG-2. Therefore, both the ligands may have wide-ranging functions in cellular activities that affect overall tumor development. Collectively, these TIE-2-dependent and TIE-2-independent activities may account for the conflicting findings of ANG-1 and ANG-2 in tumor angiogenesis. These uncertainties have impeded development of a clear strategy to target this important angiogenic pathway. A better understanding of the molecular basis of ANG-1 and ANG-2 activity in the pathophysiologic regulation of angiogenesis may set the stage for novel therapy targeting this pathway.


Subject(s)
Angiopoietins/metabolism , Neoplasms/blood supply , Neovascularization, Pathologic/pathology , Receptors, TIE/metabolism , Angiopoietins/chemistry , Animals , Humans , Receptors, TIE/chemistry
10.
Hum Gene Ther ; 18(3): 222-31, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17355186

ABSTRACT

We previously generated a herpes simplex virus type 1 (HSV-1)-based amplicon vector (denoted pC8-36) in which gene expression from the minimal cyclin A promoter is repressed by preventing the binding of a trans-activating protein, Gal4-NF-YA, to it through selective interaction with the transcriptional repressor protein CDF-1. Because CDF-1 is absent in actively dividing cells, transgene expression conferred by the pC8-36 vector is therefore cell cycle dependent. As gene therapy evolves to become a promising therapeutic modality for many human diseases, there is an increasing need to further improve the kinetics of gene regulation. In the present study, we examined whether the availability of more binding sites for CDF-1 repressor proteins could enhance transgene expression. Using an overlap extension polymerase chain reaction (PCR) method, the CDE and CHR elements within the minimum cyclin A promoter were multimerized to contain two, three, and six copies of the designated CDE/CHR sequence. Interestingly, our results demonstrated that six-copy CDE/CHR sequence motifs (pC8-6CC-Luc) conferred an approximately 20-fold increase in the ratio of cell cycle regulation compared with the previous reported construct. Further, the overall transcriptional activities mediated by pC8-6CC-Luc were stronger compared with the native human survivin promoter, which consists of three copies of the CDE element and one copy of the CHR element. pC8-6CC-Luc contained, in essence, only the synthetic six-copy CDE/CHR sequence motif (about 262 bp). In comparison with other native endogenous promoters, which usually contain many other transcription binding sites, pC8-6CC-Luc amplicon vectors should confer better regulated and consistent transgene expression and may be considered a gene delivery vector of choice to target actively proliferating tumor cells.


Subject(s)
Cell Cycle/genetics , Genetic Therapy , Genetic Vectors/genetics , Herpesvirus 1, Human/genetics , Neoplasms/therapy , Animals , Base Sequence , Binding Sites , Cell Line, Tumor , Cell Proliferation , Cyclin A/genetics , Female , Genetic Engineering , Humans , Inhibitor of Apoptosis Proteins , Luciferases/analysis , Luciferases/genetics , Mice , Mice, Inbred BALB C , Microtubule-Associated Proteins/genetics , Molecular Sequence Data , Neoplasm Proteins/genetics , Repressor Proteins/metabolism , Survivin , Tandem Repeat Sequences , Transgenes/genetics , Virion
11.
J Virol Methods ; 139(2): 166-74, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17074404

ABSTRACT

Herpes simplex virus (HSV)-1 amplicon vectors could be packaged in the presence of replication-competent helper virus or in a helper virus-free system. In the latter system, cytotoxicity due to the expression of de novo viral gene expression is greatly reduced due to the absence of helper virus. However, the titers produced are relatively low in the range of 10(7) and 10(8)TU/ml after sucrose gradient concentration. This may become a limitation to certain gene transfer applications, such as brain disorder studies since the volume of vectors that could be administered is restricted. In contrast, amplicon viral vectors of high titers can be easily generated in the presence of helper viruses. Despite the potential cytotoxicity caused by the presence of helper virus in the latter method of viral packaging, studies involving vector targeting would still require the complementing function of helper virus for the generation of recombinant HSV-1 amplicon vectors with modified viral envelopes. In view of this, the optimal method of purifying Herpes-based viral vectors that confers minimal cytotoxicity for systemic route of viral vector administration is examined. Parameters such as the ratio of amplicon versus helper viruses, the percentage of viral lost, and the extent of liver cytotoxicity induced by these viral vectors purified using different methods were investigated. In addition, the maximum recombinant HSV-1 viral dosage was also determined in vivo. Taken together, these findings may be of importance to the efficient production of contaminant-free HSV-1 amplicon viral vectors required for animal and human studies.


Subject(s)
Gene Transfer Techniques , Genetic Vectors , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/isolation & purification , Animals , Chlorocebus aethiops , Genes, Viral/genetics , Humans , Mice , Mice, Inbred BALB C , Vero Cells
12.
Biomed Res Int ; 2017: 9634172, 2017.
Article in English | MEDLINE | ID: mdl-28630875

ABSTRACT

Glioblastoma is the most aggressive cancer of the brain. The dismal prognosis is largely attributed to the heterogeneous nature of the tumor, which in addition to intrinsic molecular and genetic changes is also influenced by the microenvironmental niche in which the glioma cells reside. The cancer stem cells (CSCs) hypothesis suggests that all cancers arise from CSCs that possess the ability to self-renew and initiate tumor formation. CSCs reside in specialized niches where interaction with the microenvironment regulates their stem cell behavior. The reciprocal interaction between glioma stem cells (GSCs) and cells from the microenvironment, such as endothelial cells, immune cells, and other parenchymal cells, may also promote angiogenesis, invasion, proliferation, and stemness of the GSCs and be likely to have an underappreciated role in their responsiveness to therapy. This crosstalk may also promote molecular transition of GSCs. Hence the inherent plasticity of GSCs can be seen as an adaptive response, changing according to the signaling cue from the niche. Given the association of GSCs with tumor recurrence and treatment sensitivity, understanding this bidirectional crosstalk between GSCs and its niche may provide a framework to identify more effective therapeutic targets and improve treatment outcome.


Subject(s)
Glioblastoma/immunology , Neoplastic Stem Cells/immunology , Neovascularization, Pathologic/immunology , Tumor Microenvironment/immunology , Animals , Glioblastoma/pathology , Glioblastoma/therapy , Humans , Neoplastic Stem Cells/pathology , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/therapy
13.
Nat Commun ; 8(1): 1913, 2017 12 04.
Article in English | MEDLINE | ID: mdl-29203859

ABSTRACT

The interleukin-13 receptor alpha2 (IL-13Rα2) is a cancer-associated receptor overexpressed in human glioblastoma multiforme (GBM). This receptor is undetectable in normal brain which makes it a highly suitable target for diagnostic and therapeutic purposes. However, the pathological role of this receptor in GBM remains to be established. Here we report that IL-13Rα2 alone induces invasiveness of human GBM cells without affecting their proliferation. In contrast, in the presence of the mutant EGFR (EGFRvIII), IL-13Rα2 promotes GBM cell proliferation in vitro and in vivo. Mechanistically, the cytoplasmic domain of IL-13Rα2 specifically binds to EGFRvIII, and this binding upregulates the tyrosine kinase activity of EGFRvIII and activates the RAS/RAF/MEK/ERK and STAT3 pathways. Our findings support the "To Go or To Grow" hypothesis whereby IL-13Rα2 serves as a molecular switch from invasion to proliferation, and suggest that targeting both receptors with STAT3 signaling inhibitor might be a therapeutic approach for the treatment of GBM.


Subject(s)
Brain Neoplasms/genetics , Cell Proliferation/genetics , ErbB Receptors/genetics , Glioblastoma/genetics , Interleukin-13 Receptor alpha2 Subunit/genetics , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Knockdown Techniques , Glioblastoma/metabolism , Glioblastoma/mortality , Glioblastoma/pathology , Humans , In Vitro Techniques , Interleukin-13 Receptor alpha2 Subunit/metabolism , MAP Kinase Kinase Kinases/metabolism , Mice , Mutation , Neoplasm Invasiveness/genetics , Neoplasm Transplantation , RNA, Messenger/metabolism , Survival Rate , raf Kinases/metabolism , ras Proteins/metabolism
14.
Stem Cell Rev Rep ; 12(1): 140-55, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26454429

ABSTRACT

Glioblastoma multiforme is the most malignant tumor of the brain and is challenging to treat due to its highly invasive nature and heterogeneity. Malignant brain tumor displays high metabolic activity which perturbs its redox environment and in turn translates to high oxidative stress. Thus, pushing the oxidative stress level to achieve the maximum tolerable threshold that induces cell death is a potential strategy for cancer therapy. Previously, we have shown that gap junction inhibitor, carbenoxolone (CBX), is capable of enhancing tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) -induced apoptosis in glioma cells. Since CBX is known to induce oxidative stress, we hypothesized that the addition of another potent mediator of oxidative stress, powerful SOD mimic MnTnBuOE-2-PyP(5+) (MnBuOE), could further enhance TRAIL-driven therapeutic efficacy in glioma cells. Our results showed that combining TRAIL + CBX with MnBuOE significantly enhances cell death of glioma cell lines and this enhancement could be further potentiated by CBX pretreatment. MnBuOE-driven cytotoxicity is due to its ability to take advantage of oxidative stress imposed by CBX + TRAIL system, and enhance it in the presence of endogenous reductants, ascorbate and thiol, thereby producing cytotoxic H2O2, and in turn inducing death of glioma cells but not normal astrocytes. Most importantly, combination treatment significantly reduces viability of TRAIL-resistant Asian patient-derived glioma cells, thus demonstrating the potential clinical use of our therapeutic system. It was reported that H2O2 is involved in membrane depolarization-based sensitization of cancer cells toward TRAIL. MnBuOE is entering Clinical Trials as a normal brain radioprotector in glioma patients at Duke University increasing Clinical relevance of our studies.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Biomimetic Materials/pharmacology , Carbenoxolone/pharmacology , Metalloporphyrins/pharmacology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Ascorbic Acid/agonists , Ascorbic Acid/biosynthesis , Astrocytes/cytology , Astrocytes/drug effects , Biomimetic Materials/chemical synthesis , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Drug Combinations , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Gap Junctions/drug effects , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Hydrogen Peroxide/agonists , Hydrogen Peroxide/metabolism , Metalloporphyrins/chemical synthesis , Organ Specificity , Oxidative Stress , Primary Cell Culture , Sulfhydryl Compounds/agonists , Sulfhydryl Compounds/metabolism , Superoxide Dismutase/chemistry
15.
Hum Gene Ther ; 15(5): 495-508, 2004 May.
Article in English | MEDLINE | ID: mdl-15144579

ABSTRACT

We have engineered a novel herpes simplex virus type 1 (HSV-1)-based amplicon viral vector, whereby gene expression is controlled by cell cycle events. In nondividing cells, trans-activation of the cyclin A promoter via interaction of the Gal4/NF-YA fusion protein with the Gal4-binding sites is prevented by the presence of a repressor protein, cell cycle-dependent factor 1 (CDF-1). CDF-1 is specifically expressed during the G(0)/G(1) phase of the cell cycle and its binding site is located within the cyclin A promoter. In actively proliferating cells, trans-activation could take place because of the absence of CDF-1. Our results showed that when all these cell cycle-specific regulatory elements are incorporated in cis into a single HSV-1 amplicon plasmid vector backbone (pC8-36), reporter luciferase activity is greatly enhanced. Transgene expression mediated by this series of HSV-1 amplicon plasmid vectors and amplicon viral vectors could be regulated in a cell cycle-dependent manner in a variety of cell lines. In a further attempt to target transgene expression to a selected group of actively proliferating cells such as glial cells, we have replaced the cytomegalovirus promoter of the pC8-36 amplicon plasmid with the glial cell-specific GFAP enhancer element. With this latter viral construct, cell type-specific and cell cycle-dependent transgene expression could subsequently be demonstrated specifically in glioma-bearing animals. Taken together, our results suggest that this series of cell cycle-regulatable HSV-1 amplicon viral vectors could potentially be adapted as useful tools for the treatment of human cancers.


Subject(s)
Cell Cycle/genetics , Genes, Viral , Genetic Vectors , Glioma/genetics , Herpesvirus 1, Human/genetics , Animals , Cell Line, Tumor , Enhancer Elements, Genetic , Gene Amplification , Gene Expression Regulation, Viral , Gene Transfer Techniques , Genes, Reporter , Genetic Engineering , Glial Fibrillary Acidic Protein/genetics , Glioma/therapy , Glioma/virology , HeLa Cells , Humans , Luciferases/metabolism , Mice , NIH 3T3 Cells , Transgenes
16.
Hum Gene Ther ; 15(8): 719-32, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15319030

ABSTRACT

Glioblastomas account for approximately 20% of all primary brain tumors in adults. Glioblastoma multiforme (GBM) is a highly malignant tumor. In spite of advances in surgery, chemotherapy, and radiotherapy, the life expectancy of the patient with glioblastoma is approximately 11 months. To enhance glioma-specific gene delivery, we employed a 12-mer phage display peptide library to isolate phages that bind specifically to human glioma cell lines. Here, we report the isolation and functional characterization of novel glioma-specific peptides that target transgenes specifically to a wide array of human glioblastomas in vitro and in vivo. One of the isolated peptides, tentatively denoted as MG11, is demonstrated to be glioma specific and gives an in vitro-binding enrichment of more than 5-fold for glioma cells when compared with nonglioma cells. Intravenous injection of phages bearing the MG11 peptide-binding motif enables the phages to home specifically to glioma xenografts. Most significantly, when Lissamine rhodamine-labeled MG11 peptide is injected intratumorally, it targets specifically to glioma xenografts instead of non-glioma-derived xenografts. In summary, our results suggest that the MG11 peptide is able to target specifically to tumors of glial origin, which would allow the design of applications related to the diagnosis and treatment of human gliomas.


Subject(s)
Bacteriophages/genetics , Gene Targeting/methods , Genetic Therapy/methods , Glioblastoma/therapy , Peptides/genetics , Bacteriophages/metabolism , Cell Line, Tumor , Fluorescence , Humans , Luciferases , Nucleic Acid Amplification Techniques , Peptides/metabolism , Rhodamines , Transfection , Transgenes/genetics
17.
Histol Histopathol ; 28(11): 1427-38, 2013 11.
Article in English | MEDLINE | ID: mdl-23832648

ABSTRACT

Human bone marrow is a reservoir containing cells with different self-renewal capabilities, such as mesenchymal stem cells (MSC) and hematopoeitic stem cells (HSC). MSC in particular have been increasingly used in preclinical and clinical treatment of tissue regenerative disorder. Understanding the molecular mechanisms underlying MSC homing and mobilization is critical to the design of rational cell therapy approaches. In this review, we will discuss the key molecular mechanisms that govern the homing of MSC to bone marrow, the mobilization of MSC to tumors and injured sites via circulation, and strategies that enhance MSC migration.


Subject(s)
Cell Movement/physiology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Neoplasms , Signal Transduction/physiology , Animals , Humans , Mesenchymal Stem Cells/cytology , Neoplasms/therapy
18.
Stem Cells Dev ; 22(13): 1870-82, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23428290

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has been used extensively in cancer therapy. However, more than half of glioblastoma multiforme are insensitive to the apoptotic effect of TRAIL. Improvement in therapeutic modalities that enhances the efficacy of TRAIL in glioma is much sought after. In this study, we combined the tumor selectivity of TRAIL and tumor-homing properties of mesenchymal stem cells (MSC) with gap junction (GJ) inhibitory effect of carbenoxolone (CBX) to target orthotopic glioma. MSC were engineered to express TRAIL (MSC-TRAIL) by incorporating the secretable trimeric form of TRAIL into a Herpes Simplex Virus (HSV) type I amplicon vector. Our results showed that combined treatment of MSC-TRAIL and CBX enhanced glioma cell death, especially in three primary human glioma isolates, of which two of those are marginally sensitive to TRAIL. CBX enhanced TRAIL-induced apoptosis through upregulation of death receptor 5, blockade of GJ intercellular communication, and downregulation of connexin 43. Dual arm therapy using TRAIL and CBX prolonged the survival of treated mice by ~27% when compared with the controls in an intracranial glioma model. The enhanced efficacy of TRAIL in combination with CBX coupled with the minimal cytotoxic nature of CBX suggested a favorable clinical usage of this treatment regimen.


Subject(s)
Carbenoxolone/pharmacology , Glioma/therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/biosynthesis , TNF-Related Apoptosis-Inducing Ligand/biosynthesis , Animals , Apoptosis/drug effects , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Cell Line, Tumor , Connexin 43 , Gap Junctions/metabolism , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/pathology , Humans , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Simplexvirus/genetics , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation , Xenograft Model Antitumor Assays
19.
Peptides ; 31(4): 644-50, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20026365

ABSTRACT

Poor prognosis of high grade gliomas coupled with the difficulty of widespread delivery of therapeutic agents prompted the search into new molecular targets. Our aim is to isolate glioma-specific peptide sequences that can be used for targeted delivery of therapeutic drugs and imaging tracer to accurately demarcate tumor volume as a response to therapy. Herein, we describe the isolation and characterization of a glioma-specific peptide sequence, GL1, that interact exclusively with human glioma cells lines and primary glioma cells derived from human biopsy in vitro. Further analysis showed that the receptors for GL1 were located on the external side of the plasma membrane, where the GL1 peptides could bind stably up to a period of 180 min. More importantly, GL1 phages home specifically to human glioma xenograft when administered through tail vein, a phenomenon that was not observed when non-specific phages were used as control. Taken together, our results confirmed that GL1 could represent a novel peptide that target to tumor of glial origins, and could potentially be used as a targeting moiety for the conjugation of therapeutic drugs or diagnostic imaging radiolabels.


Subject(s)
Glioma/metabolism , Ligands , Peptides/isolation & purification , Peptides/metabolism , Amino Acid Sequence , Cell Line, Tumor , Glioma/pathology , Humans , Molecular Sequence Data , Neoplasm Transplantation , Peptides/genetics , Tissue Distribution , Transplantation, Heterologous
20.
Mol Ther ; 15(6): 1129-36, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17426711

ABSTRACT

Our previous studies have shown that transgene expression could be targeted to proliferating cells when cell cycle transcriptional regulatory elements were incorporated into herpes simplex virus type 1 (HSV-1) amplicon backbone vectors. In the study reported here, we further demonstrated the transcriptional activation of transgene expression in association with the onset of cellular proliferation using the mouse partial hepatectomy model. Moreover, transcriptional regulation could be rendered specific to human hepatocellular carcinoma (HCC) cells by inserting the chimeric gene Gal4/NF-YA under the regulation of the HCC-specific hybrid promoter. The hybrid promoter, which consists of four copies of the apolipoprotein E (ApoE) enhancer element inserted upstream of the human alpha1-antitrypsin(hAAT) promoter, induced an higher level of transcription than other liver-specific promoters such as alpha-fetoprotein (AFP) and albumin (Alb) promoter. As a consequence, the enhancement of tissue-specific expression in the context of Gal4/NF-YA fusion proteins enabled the monitoring of transgene expression using a bioluminescence imaging system. Furthermore, these vectors have been shown to be non-toxic and exhibited potent infectivity for proliferating primary HCC cells and HCC cell lines. Together, these results demonstrated that the new hybrid vectors could provide options for the design of safe and efficient systemic gene therapeutic strategies for human HCC.


Subject(s)
Carcinoma, Hepatocellular/therapy , Genetic Therapy/methods , Herpesvirus 1, Human/genetics , Liver Neoplasms/therapy , Transcription, Genetic/genetics , Animals , Apolipoproteins E/genetics , CCAAT-Binding Factor/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line , Cell Line, Tumor , Female , Gene Amplification , Genetic Vectors/genetics , HeLa Cells , Humans , Immunoblotting , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Luciferases/genetics , Luciferases/metabolism , Mice , Mice, Inbred BALB C , Mice, SCID , Microscopy, Fluorescence , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Xenograft Model Antitumor Assays/methods , alpha 1-Antitrypsin/genetics , alpha-Fetoproteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL