Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Cytotherapy ; 20(9): 1191-1201, 2018 09.
Article in English | MEDLINE | ID: mdl-30078654

ABSTRACT

To confirm the anti-tumor effect of engineered neural stem cells (NSCs) expressing cytosine deaminase (CD) and interferon-ß (IFN-ß) with prodrug 5-fluorocytosine (FC), K562 chronic myeloid leukemia (CML) cells were co-cultured with the neural stem cell lines HB1.F3.CD and HB1.F3.CD.IFN-ß in 5-FC containing media. A significant decrease in the viability of K562 cells was observed by the treatment of the NSC lines, HB1.F3.CD and HB1.F3.CD.IFN-ß, compared with the control. A modified trans-well assay showed that engineered human NSCs significantly migrated toward K562 CML cells more than human normal lung cells. In addition, the important chemoattractant factors involved in the specific migration ability of stem cells were found to be expressed in K562 CML cells. In a xenograft mouse model, NSC treatments via subcutaneous and intravenous injections resulted in significant inhibitions of tumor mass growth and extended survival dates of the mice. Taken together, these results suggest that gene therapy using genetically engineered stem cells expressing CD and IFN-ß may be effective for treating CML in these mouse models.


Subject(s)
Neural Stem Cells/transplantation , Animals , Coculture Techniques , Cytosine Deaminase/genetics , Cytosine Deaminase/metabolism , Female , Flucytosine/pharmacology , Genetic Engineering , Genetic Therapy/methods , Humans , Interferon-beta/genetics , Interferon-beta/metabolism , K562 Cells , Leukemia/therapy , Mice, Nude , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Prodrugs , Xenograft Model Antitumor Assays
2.
J Nanosci Nanotechnol ; 15(10): 7929-34, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26726442

ABSTRACT

Human mesenchymal stem cells (MSCs) have the capacity for self-renewal and maintain pluripotency, which is defined by their ability to differentiate into cells such as osteoblasts, neurons, and glial cells. In this study, we report a method for defining the status of human MSCs based on electrochemical detection systems. Gold nano-dot structures were fabricated using a nanoporous alumina mask, and the structural formations were confirmed by scanning electron microscopy (SEM). Human MSCs were allowed to attach to RGD (Arg-Gly-Asp) peptide nanopatterned surfaces, and electrochemical tools were applied to the MSCs attached on the chip surface. The cultured MSCs were shown to differentiate into neural cell types, as indicated by immunocytochemical staining for tyrosine hydroxylase and beta tubulin III. Following treatment with basic fibroblast growth factor (bFGF) for 14 days, most of the B10 cells exhibited bipolar or multipolar morphology with branched processes, and the proportion of B10 cells expressing neuronal cell markers considerably increased. Electrophysiological recordings from MSCs treated with bFGF for 5-14 days were examined with cyclic voltammetry, and the electrochemical signals were shown to increase during differentiation from MSCs to neuronal cells. This human MSC cell line is a useful tool for studying organogenesis, specifically neurogenesis, and in addition, the cell line provides a valuable source of cells for cell therapy. The electrochemical measurement system proposed here could be utilized in electrical cell chips for numerous applications, including cell differentiation, disease diagnosis, drug detection, and on-site monitoring.


Subject(s)
Aluminum Oxide/chemistry , Cell Culture Techniques/methods , Cell Differentiation , Gold/chemistry , Mesenchymal Stem Cells/metabolism , Nanostructures/chemistry , Antigens, Differentiation/biosynthesis , Cell Line , Humans , Mesenchymal Stem Cells/cytology
3.
Oncotarget ; 5(24): 12835-48, 2014 Dec 30.
Article in English | MEDLINE | ID: mdl-25544747

ABSTRACT

In this study, neural stem cells (NSCs)-derived enzyme/prodrug therapy (NDEPT) was used to treat primary lung cancer or metastatic lung cancer in the brain. To confirm the anti-tumor effect of NSCs expressing carboxyl esterase (CE), A549 lung cancer cells were treated with HB1.F3.CE cells and CPT-11. A significant decrease in the viability/proliferation of lung cancer cells was observed compared to negative controls or cells treated with CPT-11 alone. To produce a mouse model of primary lung cancer or lung cancer metastasis to the brain, A549 cells were implanted in the dorsal area of the mouse or right hemisphere. CM-DiI pre-stained stem cells were implanted near the primary lung cancer tumor mass or in the contralateral brain. Two days after stem cells injection, mice were inoculated with CPT-11 (13.5 kg/mouse/day) via intraperitoneal injection. In the primary lung cancer mouse models, tumor mass was 80% lower in response to HB1.F3.CE in conjunction with CPT-11, while it was only reduced by 40% in the group treated with CPT-11 alone. Additionally, therapeutic efficacy of co-treatment with stem cells and CPT-11 was confirmed by detection of apoptosis and necrosis in primary and metastatic lung cancer tissues. By secreting VEGF, tumor cells modulate Erk1/2 and Akt signaling and migration of stem cells. This further increased tumor-selectivity of stem cell/prodrug co-therapy. Overall, these results indicate that NSCs expressing the therapeutic gene may be a powerful tool for treatment of primary lung cancer or metastasis of lung cancer to the brain.


Subject(s)
Camptothecin/analogs & derivatives , Carboxylesterase/biosynthesis , Lung Neoplasms/therapy , Neural Stem Cells/transplantation , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/pharmacology , Carboxylesterase/genetics , Cell Growth Processes/drug effects , Cell Line, Tumor , Disease Models, Animal , Humans , Irinotecan , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Neural Stem Cells/enzymology , Random Allocation , Xenograft Model Antitumor Assays
4.
ACS Nano ; 8(12): 12450-60, 2014 Dec 23.
Article in English | MEDLINE | ID: mdl-25375246

ABSTRACT

Plasmonic photothermal therapy utilizes biologically inert gold nanorods (AuNRs) as tumor-localized antennas that convert light into heat capable of eliminating cancerous tissue. This approach has lower morbidity than surgical resection and can potentially synergize with other treatment modalities including chemotherapy and immunotherapy. Despite these advantages, it is still challenging to obtain heating of the entire tumor mass while avoiding unnecessary collateral damage to surrounding healthy tissue. It is therefore critical to identify innovative methods to distribute an effective concentration of AuNRs throughout tumors without depositing them in surrounding healthy tissue. Here we demonstrate that AuNR-loaded, tumor-tropic neural stem cells (NSCs) can be used to improve the intratumoral distribution of AuNRs. A simple UV-vis technique for measuring AuNR loading within NSCs was established. It was then confirmed that NSC viability is unimpaired following AuNR loading and that NSCs retain AuNRs long enough to migrate throughout tumors. We then demonstrate that intratumoral injections of AuNR-loaded NSCs are more efficacious than free AuNR injections, as evidenced by reduced recurrence rates of triple-negative breast cancer (MDA-MB-231) xenografts following NIR exposure. Finally, we demonstrate that the distribution of AuNRs throughout the tumors is improved when transported by NSCs, likely resulting in the improved efficacy of AuNR-loaded NSCs as compared to free AuNRs. These findings highlight the advantage of combining cellular therapies and nanotechnology to generate more effective cancer treatments.


Subject(s)
Drug Delivery Systems/methods , Gold/chemistry , Gold/therapeutic use , Nanotubes , Neural Stem Cells/metabolism , Phototherapy , Animals , Biological Transport , Cell Line, Tumor , Drug Liberation , Female , Gold/metabolism , Humans , Lasers , Mice
5.
Sci Transl Med ; 5(184): 184ra59, 2013 May 08.
Article in English | MEDLINE | ID: mdl-23658244

ABSTRACT

High-grade gliomas are extremely difficult to treat because they are invasive and therefore not curable by surgical resection; the toxicity of current chemo- and radiation therapies limits the doses that can be used. Neural stem cells (NSCs) have inherent tumor-tropic properties that enable their use as delivery vehicles to target enzyme/prodrug therapy selectively to tumors. We used a cytosine deaminase (CD)-expressing clonal human NSC line, HB1.F3.CD, to home to gliomas in mice and locally convert the prodrug 5-fluorocytosine to the active chemotherapeutic 5-fluorouracil. In vitro studies confirmed that the NSCs have normal karyotype, tumor tropism, and CD expression, and are genetically and functionally stable. In vivo biodistribution studies demonstrated NSC retention of tumor tropism, even in mice pretreated with radiation or dexamethasone to mimic clinically relevant adjuvant therapies. We evaluated safety and toxicity after intracerebral administration of the NSCs in non-tumor-bearing and orthotopic glioma-bearing immunocompetent and immunodeficient mice. We detected no difference in toxicity associated with conversion of 5-fluorocytosine to 5-fluorouracil, no NSCs outside the brain, and no histological evidence of pathology or tumorigenesis attributable to the NSCs. The average tumor volume in mice that received HB1.F3.CD NSCs and 5-fluorocytosine was about one-third that of the average volume in control mice. On the basis of these results, we conclude that combination therapy with HB1.F3.CD NSCs and 5-fluorocytosine is safe, nontoxic, and effective in mice. These data have led to approval of a first-in-human study of an allogeneic NSC-mediated enzyme/prodrug-targeted cancer therapy in patients with recurrent high-grade glioma.


Subject(s)
Glioma/drug therapy , Glioma/therapy , Neural Stem Cells/cytology , Prodrugs/therapeutic use , Animals , Cell Line , Cytosine Deaminase/metabolism , Female , Flow Cytometry , Flucytosine/metabolism , Flucytosine/therapeutic use , Fluorouracil/metabolism , Humans , Male , Mice , Mice, Nude , Neural Stem Cells/metabolism , Prodrugs/metabolism
7.
Environ Toxicol Pharmacol ; 31(3): 397-405, 2011 May.
Article in English | MEDLINE | ID: mdl-21787710

ABSTRACT

Antitumor effects of a ginsenoside Rg(3)-fortified red ginseng preparation (Rg(3)-RGP) were investigated in human non-small cell lung carcinoma (H460) cells using in vitro cytotoxicity assay and in vivo nude mouse xenograft model. Immunomodulatory effects of the preparation were also assessed by measuring the facilitating activities on the nitric oxide (NO) release from peritoneal macrophages, in vitro and in vivo lymphocyte proliferation, and the carbon clearance from circulating blood. In a cell level, Rg(3)-RGP exerted H460 cytotoxicity and facilitated splenocyte proliferation at very high concentrations, without affecting NO production. However, oral administration of Rg(3)-RGP (100-300 mg/kg) enhanced carbon particle-phagocytic index of blood macrophages up to 360-397% of control value. In addition, Rg(3)-RGP significantly increased the splenocyte proliferation (23% at 100mg/kg). In tumor-bearing mice, 28-day oral treatment with Rg(3)-RGP (100mg/kg) remarkably suppressed the tumor growth, leading to the decrease of the tumor volume and weight by 30-31%, which was comparable to the effect (27-29% reduction) of doxorubicin (2mg/kg at 3-day intervals). While Rg(3)-RGP did not cause adverse effects, intravenous injection of doxorubicin markedly decreased body and testes weights, and exhibited severe depletion of spermatogenic cells in the atrophic seminiferous tubules. These results indicate that Rg(3)-RGP exerts antitumor activities via indirect immunomodulatory actions, without causing adverse effects as seen in doxorubicin.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Ginsenosides/pharmacology , Panax/chemistry , Animals , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents, Phytogenic/adverse effects , Body Weight/drug effects , Carbon/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Doxorubicin/pharmacology , Ginsenosides/adverse effects , Heart/drug effects , Humans , Lung Neoplasms/drug therapy , Lymphocytes/drug effects , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , Neoplasm Transplantation , Nitric Oxide/biosynthesis , Organ Size/drug effects , Plant Preparations , Spleen/cytology , Spleen/drug effects , Testis/drug effects
8.
Cancer Res ; 67(17): 8274-84, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17804742

ABSTRACT

Silibinin, a flavonoid isolated from Silybum marianum, has been reported to have cancer chemopreventive and therapeutic effects. Here, we show that treatment with subtoxic doses of silibinin in combination with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) induces rapid apoptosis in TRAIL-resistant glioma cells, but not in human astrocytes, suggesting that this combined treatment may offer an attractive strategy for safely treating gliomas. Although the proteolytic processing of procaspase-3 by TRAIL was partially blocked in glioma cells, cotreatment with silibinin efficiently recovered TRAIL-induced caspase activation in these cells. Silibinin treatment up-regulated DR5, a death receptor of TRAIL, in a transcription factor CHOP-dependent manner. Furthermore, treatment with silibinin down-regulated the protein levels of the antiapoptotic proteins FLIP(L), FLIP(S), and survivin through proteasome-mediated degradation. Taken together, our results show that the activity of silibinin to modulate multiple components in the death receptor-mediated apoptotic pathway is responsible for its ability to recover TRAIL sensitivity in TRAIL-resistant glioma cells.


Subject(s)
Apoptosis/drug effects , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Glioma/pathology , Microtubule-Associated Proteins/genetics , Neoplasm Proteins/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Antioxidants/pharmacology , Astrocytes/drug effects , Caspases/metabolism , Drug Combinations , Drug Evaluation, Preclinical , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Glioma/genetics , Humans , Inhibitor of Apoptosis Proteins , Silybin , Silymarin/pharmacology , Survivin , Transcription Factor CHOP/metabolism , Tumor Cells, Cultured
9.
J Neuroimmunol ; 159(1-2): 66-74, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15652404

ABSTRACT

The effects of the chemokine IL-8 on amyloid beta peptide (Abeta(1-42))-induced responses in cultured human microglia were investigated using RT-PCR, ELISA and immunocytochemistry. Abeta(1-42) (5 microM) applied for 8 h induced the expression and increased the production of the pro-inflammatory cytokines IL-6, IL-1beta, TNF-alpha, the inducible enzyme COX-2 and chemokine IL-8. Microglial treatment with IL-8 added (at 100 ng/mL) with Abeta(1-42) led to enhancement in both expression and production of all of these pro-inflammatory factors compared with peptide alone. Stimulation with IL-8 itself was effective in increasing microglial expression of pro-inflammatory cytokines, IL-8 and COX-2, however, had no effect on protein levels of all these factors. The expression of the anti-inflammatory cytokines IL-10 and TGFbeta(1) remained unchanged from basal levels with stimulation using either Abeta(1-42), IL-8 or the peptide together with IL-8. The actions of IL-8 to potentiate Abeta(1-42)-induced inflammatory mediators may have particular relevance to Alzheimer disease brain which exhibits elevated levels of the chemokine.


Subject(s)
Adjuvants, Immunologic/physiology , Amyloid beta-Peptides/pharmacology , Cytokines/biosynthesis , Inflammation Mediators/metabolism , Interleukin-8/physiology , Microglia/enzymology , Microglia/immunology , Peptide Fragments/pharmacology , Prostaglandin-Endoperoxide Synthases/biosynthesis , Adjuvants, Immunologic/genetics , Adjuvants, Immunologic/pharmacology , Cells, Cultured , Cyclooxygenase 2 , Drug Combinations , Fetus , Humans , Interleukin-1/biosynthesis , Interleukin-1/genetics , Interleukin-1/metabolism , Interleukin-10/biosynthesis , Interleukin-10/genetics , Interleukin-6/biosynthesis , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-8/genetics , Interleukin-8/pharmacology , Membrane Proteins , Microglia/cytology , Microglia/metabolism , Prostaglandin-Endoperoxide Synthases/genetics , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta1 , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL