Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Nanomedicine ; 11: 13-23, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26719691

RESUMO

Mesenchymal stem cell (MSC)-based gene therapy is a promising tool for the treatment of various neurological diseases, including brain tumors. However, the tracking of in vivo stem cell migration, distribution, and survival need to be defined for their clinical application. The systemic routes of stem cell delivery must be determined because direct intracerebral injection as a cure for brain tumors is an invasive method. In this study, we show for the first time that near-infrared (NIR) imaging can reveal the distribution and tumor tropism of intravenously injected MSCs in an intracranial xenograft glioma model. MSCs were labeled with NIR fluorescent nanoparticles, and the effects of the NIR dye on cell proliferation and migratory capacity were evaluated in vitro. We investigated the tumor-targeting properties and tissue distribution of labeled MSCs introduced by intravenous injection and followed by in vivo imaging analysis, histological analysis, and real-time quantitative polymerase chain reaction. We observed no cytotoxicity or change in the overall growth rate and characteristics of labeled MSCs compared with control MSCs. NIR fluorescent imaging showed the organ distribution and targeted tumor tropism of systemically injected human MSCs. A significant number of MSCs accumulated specifically at the tumor site in the mouse brain. These results suggest that NIR-based cell tracking is a potentially useful imaging technique to visualize cell survival, migration, and distribution for the application of MSC-mediated therapies in the treatment of malignant gliomas.


Assuntos
Neoplasias Encefálicas/patologia , Glioma/patologia , Medições Luminescentes/métodos , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais , Adulto , Animais , Neoplasias Encefálicas/terapia , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Corantes Fluorescentes/química , Corantes Fluorescentes/farmacocinética , Glioma/terapia , Humanos , Masculino , Células-Tronco Mesenquimais/química , Camundongos Nus , Pessoa de Meia-Idade , Nanopartículas/análise , Distribuição Tecidual , Tropismo , Ensaios Antitumorais Modelo de Xenoenxerto
2.
J Neuroimmunol ; 274(1-2): 20-7, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25005115

RESUMO

We previously demonstrated that interferon ß (IFN-ß)-secreting mesenchymal stem cells (MSCs-IFN-ß) strongly reduced the clinical severity of experimental autoimmune encephalomyelitis (EAE), compared with MSCs alone. Recently, minocycline ameliorates the clinical severity of multiple sclerosis (MS). Herein, we evaluated the effects of a combined treatment of MSCs-IFN-ß and minocycline on EAE mice. The combined treatment significantly alleviated the clinical severity mainly by maintaining the integrity of blood-spinal cord barrier, in a manner likely involving inhibition of microvascular disruption, matrix metalloproteinases, neuroinflammation, and enhancement of immunomodulatory effects. Therefore, this combined treatment has the potential to improve the functional recovery of patients with MS.


Assuntos
Encefalomielite Autoimune Experimental/tratamento farmacológico , Interferon beta/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo , Minociclina/farmacologia , Animais , Antibacterianos/farmacologia , Células Cultivadas , Terapia Combinada , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Feminino , Humanos , Interferon beta/imunologia , Metaloproteinase 2 da Matriz/imunologia , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/imunologia , Metaloproteinase 9 da Matriz/metabolismo , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Medula Espinal/imunologia , Medula Espinal/metabolismo , Células Th1/imunologia , Células Th2/imunologia
3.
Cell Biol Int ; 38(9): 1050-9, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24797366

RESUMO

The beneficial effects of mesenchymal stem cells (MSCs) are mediated partly by the paracrine production of cytoprotective and trophic factors. Vascular endothelial growth factor (VEGF) is released from MSCs as a paracrine trophic factor and contributes to the therapeutic effects of the stem cell by regulating angiogenesis and promoting revascularization in injured tissues. Interleukin-8 (IL-8), an inflammatory chemokine with potent proangiogenic properties, is upregulated in the ischemic brain and has been shown to promote homing of bone marrow-derived cells to injured sites. However, the effect of IL-8 on MSCs paracrine function remains unknown. We found that IL-8 induced VEGF production and phosphorylation of Akt and ERK. Both effects could be blocked by inhibitors (LY294002, PD098059) or siRNA-mediated silencing of Akt and ERK in human bone marrow MSCs (hBM-MSCs). IL-8-induced VEGF production in hBM-MSCs significantly increased tube formation on Matrigel compared with basal secreted VEGF. In a rat stroke model, administration of IL-8-treated hBM-MSCs decreased the infarction volume and increased angiogenesis in the ischemic boundary zone compared with hBM-MSC treatment alone. In conclusion, IL-8 stimulates VEGF production in hBM-MSCs in part via the PI3K/Akt and MAPK/ERK signal transduction pathways and that administration of IL-8-treated hBM-MSCs increases angiogenesis after stroke. This approach may be used to optimize MSC-based therapies for numerous diseases including stroke, myocardial ischemia, and spinal cord injury.


Assuntos
Células da Medula Óssea/citologia , Interleucina-8/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Encéfalo/metabolismo , Células Cultivadas , Cromonas/farmacologia , Modelos Animais de Doenças , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Flavonoides/farmacologia , Humanos , Isquemia/metabolismo , Isquemia/patologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Morfolinas/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Transdução de Sinais/efeitos dos fármacos
4.
Biomed Res Int ; 2014: 129145, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24672780

RESUMO

Numerous studies have reported that mesenchymal stem cells (MSCs) can ameliorate neurological deficits in ischemic stroke models. Among the various hypotheses that have been suggested to explain the therapeutic mechanism underlying these observations, neurogenesis is thought to be critical. To enhance the therapeutic benefits of human bone marrow-derived MSCs (hBM-MSCs), we efficiently modified hBM-MSCs by introduction of the brain-derived neurotrophic factor (BDNF) gene via adenoviral transduction mediated by cell-permeable peptides and investigated whether BDNF-modified hBM-MSCs (MSCs-BDNF) contributed to functional recovery and endogenous neurogenesis in a rat model of middle cerebral artery occlusion (MCAO). Transplantation of MSCs induced the proliferation of 5-bromo-2'-deoxyuridine (BrdU-) positive cells in the subventricular zone. Transplantation of MSCs-BDNF enhanced the proliferation of endogenous neural stem cells more significantly, while suppressing cell death. Newborn cells differentiated into doublecortin (DCX-) positive neuroblasts and Neuronal Nuclei (NeuN-) positive mature neurons in the subventricular zone and ischemic boundary at higher rates in animals with MSCs-BDNF compared with treatment using solely phosphate buffered saline (PBS) or MSCs. Triphenyltetrazolium chloride staining and behavioral analysis revealed greater functional recovery in animals with MSCs-BDNF compared with the other groups. MSCs-BDNF exhibited effective therapeutic potential by protecting cell from apoptotic death and enhancing endogenous neurogenesis.


Assuntos
Isquemia Encefálica/terapia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Neurogênese , Acidente Vascular Cerebral/terapia , Adulto , Animais , Apoptose , Isquemia Encefálica/complicações , Isquemia Encefálica/fisiopatologia , Células Cultivadas , Modelos Animais de Doenças , Proteína Duplacortina , Ensaio de Imunoadsorção Enzimática , Humanos , Marcação In Situ das Extremidades Cortadas , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/fisiopatologia , Infarto da Artéria Cerebral Média/terapia , Masculino , Células-Tronco Mesenquimais/citologia , Pessoa de Meia-Idade , Ratos Sprague-Dawley , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/fisiopatologia , Adulto Jovem
5.
Stem Cells Transl Med ; 3(2): 172-82, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24436439

RESUMO

Because the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) selectively kills tumor cells, it is one of the most promising candidates for cancer treatment. TRAIL-secreting human mesenchymal stem cells (MSC-TRAIL) provide targeted and prolonged delivery of TRAIL in glioma therapy. However, acquired resistance to TRAIL of glioma cells is a major problem to be overcome. We showed a potential therapy that used MSC-TRAIL combined with the chemotherapeutic agent temozolomide (TMZ). The antitumor effects of the combination with MSC-TRAIL and TMZ on human glioma cells were determined by using an in vitro coculture system and an in vivo experimental xenografted mouse model. Intracellular signaling events that are responsible for the TMZ-mediated sensitization to TRAIL-induced apoptosis were also evaluated. Treatment of either TRAIL-sensitive or -resistant human glioma cells with TMZ and MSC-TRAIL resulted in a significant enhancement of apoptosis compared with the administration of each agent alone. We demonstrated that TMZ effectively increased the sensitivity to TRAIL-induced apoptosis via extracellular signal-regulated kinase-mediated upregulation of the death receptor 5 and downregulation of antiapoptotic proteins, such as X-linked inhibitor of apoptosis protein and cellular FLICE-inhibitory protein. Subsequently, this combined treatment resulted in a substantial increase in caspase activation. Furthermore, in vivo survival experiments and bioluminescence imaging analyses showed that treatment using MSC-TRAIL combined with TMZ had greater therapeutic efficacy than did single-agent treatments. These results suggest that the combination of clinically relevant TMZ and MSC-TRAIL is a potential therapeutic strategy for improving the treatment of malignant gliomas.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Dacarbazina/análogos & derivados , Glioma/tratamento farmacológico , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Ligante Indutor de Apoptose Relacionado a TNF/genética , Adulto , Animais , Antineoplásicos Alquilantes/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Dacarbazina/farmacologia , Terapia Genética/métodos , Glioma/patologia , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Masculino , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Temozolomida , Ensaios Antitumorais Modelo de Xenoenxerto , Adulto Jovem
6.
Stem Cell Res Ther ; 4(4): 77, 2013 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-23826999

RESUMO

INTRODUCTION: Multiple sclerosis (MS) is the most common inflammatory demyelinating disorder of the central nervous system (CNS). Minocycline ameliorates the clinical severity of MS and exhibits antiinflammatory, neuroprotective activities, and good tolerance for long-term use, whereas it is toxic to the CNS. Recently, the immunomodulation and neuroprotection capabilities of human bone marrow mesenchymal stem cells (hBM-MSCs) were shown in experimental autoimmune encephalomyelitis (EAE). In this study, we evaluated whether the combination of hBM-MSCs and a low-dose minocycline could produce beneficial effects in EAE mice. METHODS: The sensitivity of hBM-MSCs to minocycline was determined by an established cell-viability assay. Minocycline-treated hBM-MSCs were also characterized with flow cytometry by using MSC surface markers and analyzed for their multiple differentiation capacities. EAE was induced in C57BL/6 mice by using immunization with MOG35-55. Immunopathology assays were used to detect the inflammatory cells, demyelination, and neuroprotection. Interferon gamma (IFN-γ)/tumor necrosis factor alpha (TNF-α) and interleukin-4 (IL-4)/interleukin-10 (IL-10), the hallmark cytokines that direct Th1 and Th2 development, were detected with enzyme-linked immunosorbent assay (ELISA). terminal dUTP nick-end labeling (TUNEL) staining was performed to elucidate the cell apoptosis in the spinal cords of EAE mice. RESULTS: Minocycline did not affect the viability, surface phenotypes, or differentiation capacity of hBM-MSCs, while minocycline affected the viability of astrocytes at a high dose. In vivo efficacy experiments showed that combined treatment, compared to the use of minocycline or hBM-MSCs alone, resulted in a significant reduction in clinical scores, along with attenuation of inflammation, demyelination, and neurodegeneration. Moreover, the combined treatment with hBM-MSCs and minocycline enhanced the immunomodulatory effects, which suppressed proinflammatory cytokines (IFN-γ, TNF-α) and conversely increased anti-inflammatory cytokines (IL-4, IL-10). In addition, TUNEL staining also demonstrated a significant decrease of the number of apoptotic cells in the combined treatment compared with either treatment alone. CONCLUSIONS: The combination of hBM-MSCs and minocycline provides a novel experimental protocol to enhance the therapeutic effects in MS.


Assuntos
Antibacterianos/uso terapêutico , Células da Medula Óssea/metabolismo , Encefalomielite Autoimune Experimental/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo , Minociclina/uso terapêutico , Animais , Apoptose , Diferenciação Celular , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL
7.
Biomed Res Int ; 2013: 696738, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23710456

RESUMO

Interferon-beta (IFN- ß ), a well-established standard treatment for multiple sclerosis (MS), has proved to exhibit clinical efficacy. In this study, we first evaluated the therapeutic effects for MS using human bone marrow-derived mesenchymal stem cells (hBM-MSCs) as delivery vehicles with lesion-targeting capability and IFN- ß as therapeutic gene. We also engineered hBM-MSCs to secret IFN- ß (MSCs-IFN ß ) via adenoviral transduction and confirmed the secretory capacity of MSCs-IFN ß by an ELISA assay. MSCs-IFN ß -treated mice showed inhibition of experimental autoimmune encephalomyelitis (EAE) onset, and the maximum and average score for all animals in each group was significantly lower in the MSCs-IFN ß -treated EAE mice when compared with the MSCs-GFP-treated EAE mice. Inflammatory infiltration and demyelination in the lumbar spinal cord also significantly decreased in the MSCs-IFN ß -treated EAE mice compared to PBS- or MSCs-GFP-treated EAE mice. Moreover, MSCs-IFN ß treatment enhanced the immunomodulatory effects, which suppressed proinflammatory cytokines (IFN-γ and TNF-α) and conversely increased anti-inflammatory cytokines (IL-4 and IL-10). Importantly, injected MSCs-IFN ß migrated into inflamed CNS and significantly reduced further injury of blood-brain barrier (BBB) permeability in EAE mice. Thus, our results provide the rationale for designing novel experimental protocols to enhance the therapeutic effects for MS using hBM-MSCs as an effective gene vehicle to deliver the therapeutic cytokines.


Assuntos
Terapia Genética , Interferon beta/metabolismo , Células-Tronco Mesenquimais/metabolismo , Esclerose Múltipla/tratamento farmacológico , Animais , Células da Medula Óssea/metabolismo , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Interferon beta/administração & dosagem , Interferon beta/genética , Interferon gama/metabolismo , Interleucina-10/metabolismo , Interleucina-4/metabolismo , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Camundongos , Esclerose Múltipla/genética , Esclerose Múltipla/patologia , Fator de Necrose Tumoral alfa/metabolismo
8.
Biochem Biophys Res Commun ; 431(2): 354-9, 2013 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-23261452

RESUMO

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) triggers specific apoptosis in tumor cells and is one of the most promising candidates for cancer gene therapy. However, resistance to TRAIL is one of the main impediments to use of TRAIL in cancer treatment. We showed previously that the lipoxygenase inhibitor MK886 in combination with TRAIL exhibits enhanced antitumor activities compared with each agent alone in human glioma cells. In this study, we elucidated the molecular mechanisms responsible for MK886-mediated sensitization to TRAIL-induced apoptosis. We found that MK886 sensitized glioma cells to TRAIL-induced apoptosis by upregulating the death receptor 5 (DR5) and that specific knockdown of DR5 attenuated cell death. The mechanisms underlying this sensitization involved activation of the MK886-induced p38 mitogen-activated protein kinase (MAPK) pathway and subsequent DR5 overexpression. However, treatment with a specific inhibitor or gene silencing of p38 MAPK abolished both the DR5 induction and the increase in apoptosis caused by TRAIL. Taken together, our findings indicate that the increased expression of DR5 in a p38 MAPK-dependent manner plays an important role in the sensitization of MK886 to TRAIL-induced apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Glioma/metabolismo , Indóis/farmacologia , Inibidores de Lipoxigenase/farmacologia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/biossíntese , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Sinergismo Farmacológico , Técnicas de Silenciamento de Genes , Humanos , Fator de Transcrição CHOP/genética , Fator de Transcrição CHOP/metabolismo , Regulação para Cima , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
9.
Cancer Res ; 72(18): 4807-17, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22962275

RESUMO

The apoptotic ligand TRAIL is believed to have promise as a cancer gene therapy, yet many types of cancer, including gliomas, have exhibited resistance to TRAIL-induced apoptosis. Here, we show that therapeutic combination of the lipoxygenase inhibitor MK886 and TRAIL-secreting human mesenchymal stem cells (MSC-TRAIL) provide targeted and prolonged delivery of TRAIL both in vitro and in orthotopic mouse models of glioma. Treatment of either TRAIL-sensitive or TRAIL-resistant human glioma cells with MK886 and MSC-TRAIL resulted in significantly enhanced apoptosis compared with each agent alone. MK886 effectively increased the sensitivity to TRAIL-induced apoptosis via upregulation of the death receptor 5 and downregulation of the antiapoptotic protein survivin in human glioma cell lines and in primary glioma cells. This regulation was accompanied by a substantial increase in caspase activation after combined treatment. Furthermore, in vivo survival experiments and imaging analysis in orthotopic xenografted mice showed that MSC-based TRAIL gene delivery combined with MK886 into the tumors had greater therapeutic efficacy than single-agent treatment. Together, our findings indicate that MK886 combined with MSC-based TRAIL gene delivery may represent a novel strategy for improving the treatment of malignant gliomas.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias Encefálicas/tratamento farmacológico , Terapia Genética/métodos , Glioma/tratamento farmacológico , Indóis/administração & dosagem , Transplante de Células-Tronco Mesenquimais/métodos , Animais , Western Blotting , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Terapia Combinada , Citometria de Fluxo , Glioma/metabolismo , Humanos , Imuno-Histoquímica , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Nus , RNA Interferente Pequeno , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Eur J Pharmacol ; 689(1-3): 89-95, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22659584

RESUMO

Chlorogenic acid (CGA) has been reported to have various beneficial effects on the cardiovascular and central nervous systems. The purpose of the current study was to investigate whether CGA has protective effects against cerebral ischemia and whether these effects are due to modification of brain edema-related vascular factors. In a rat model of transient middle cerebral artery occlusion (MCAo, 2h of occlusion followed by 22 h of reperfusion), we measured infarct volume and performed behavioral test to evaluate the effects of CGA on brain damage and sensory-motor functional deficits. Brain water content and Evans blue extravasation were measured to evaluate brain edema and blood brain barrier (BBB) damage. Lipid peroxidation (LPO) and the expressions and activities of matrix metalloproteinase (MMP)-2 and MMP-9 were measured to investigate the mechanisms of action. Intraperitoneal injection of CGA (3, 10, and 30 mg/kg) at 0 h and 2h after MCAo dose-dependently reduced infarct volume and sensory-motor functional deficits. It also reduced brain water content and Evans blue extravasation. Mechanistically, CGA reduced LPO and MMPs expressions and activities. These results suggest that CGA reduces brain damage, BBB damage and brain edema by radical scavenging activity and the inhibitory effects on MMP-2 and MMP-9.


Assuntos
Edema Encefálico/tratamento farmacológico , Isquemia Encefálica/tratamento farmacológico , Ácido Clorogênico/uso terapêutico , Modelos Animais de Doenças , Metaloproteinase 2 da Matriz , Metaloproteinase 9 da Matriz , Inibidores de Metaloproteinases de Matriz/uso terapêutico , Animais , Edema Encefálico/enzimologia , Edema Encefálico/patologia , Isquemia Encefálica/enzimologia , Isquemia Encefálica/patologia , Ácido Clorogênico/farmacologia , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Inibidores de Metaloproteinases de Matriz/farmacologia , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Ratos , Ratos Sprague-Dawley
11.
Food Chem Toxicol ; 50(9): 2987-95, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22705330

RESUMO

Angelicae Gigantis (AG) Radix, commonly used medicinal food, has been reported as a promising candidate for inflammatory diseases. However, the anti-allergic effects of AG and its molecular mechanisms have yet to be clarified. The present study investigated the anti-allergy effects of ethanol extracts of AG on mast cell-mediated allergic inflammation in vivo and in vitro. The finding of this study demonstrated that AG reduced anti-dinitrophenyl IgE antibody-induced passive cutaneous anaphylaxis, compound 48/80-induced histamine release, 2,4-dinitrofluoro benzene-induced contact hypersensitivity. In addition, AG inhibited the production of interleukin (IL)-6, IL-8, and TNF-α, as well as the activation of Jun N-terminal kinase and nuclear factor-κB in phorbol 12-myristate 13-acetate plus calcium ionophore A23187-stimulated human mast cells. In conclusion, our results provide a novel insight into the pharmacological actions of AG as a potential candidate for use in allergic inflammatory diseases.


Assuntos
Angelica/química , Hipersensibilidade/prevenção & controle , Inflamação/prevenção & controle , Mastócitos/efeitos dos fármacos , Extratos Vegetais/farmacologia , Animais , Cromatografia Líquida de Alta Pressão , Feminino , Liberação de Histamina/efeitos dos fármacos , Hipersensibilidade/imunologia , Técnicas In Vitro , Inflamação/imunologia , Mastócitos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Ratos , Ratos Sprague-Dawley
12.
J Biomed Biotechnol ; 2012: 987495, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22701311

RESUMO

Temozolomide (TMZ) has become a key therapeutic agent in patients with malignant gliomas; however, its survival benefit remains unsatisfactory. Valproic acid (VPA) has emerged as an anticancer drug via inhibition of histone deacetylases (HDACs), but the therapeutic advantages of a combination with VPA and TMZ remain poorly understood. The main aim of the present study was to determine whether an antitumor effect could be potentiated by a combination of VPA and TMZ, especially in TMZ-resistant cell lines. A combination of VPA and TMZ had a significantly enhanced antitumor effect in TMZ-resistant malignant glioma cells (T98 and U138). This enhanced antitumor effect correlated with VPA-mediated reduced O6-methylguanine-DNA methyltransferase (MGMT) expression, which plays an important role in cellular resistance to alkylating agents. In vitro, the combination of these drugs enhanced the apoptotic and autophagic cell death, as well as suppressed the migratory activities in TMZ-resistant cell lines. Furthermore, in vivo efficacy experiment showed that treatment of combination of VPA and TMZ significantly inhibited tumor growth compared with the monotherapy groups of mice. These results suggest that the clinical efficacy of TMZ chemotherapy in TMZ-resistant malignant glioma may be improved by combination with VPA.


Assuntos
Metilases de Modificação do DNA/genética , Enzimas Reparadoras do DNA/genética , Dacarbazina/análogos & derivados , Glioma/tratamento farmacológico , Proteínas Supressoras de Tumor/genética , Ácido Valproico/administração & dosagem , Animais , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Linhagem Celular Tumoral , Metilases de Modificação do DNA/biossíntese , Enzimas Reparadoras do DNA/biossíntese , Dacarbazina/administração & dosagem , Regulação para Baixo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Sinergismo Farmacológico , Regulação Neoplásica da Expressão Gênica , Glioma/genética , Humanos , Camundongos , Temozolomida , Proteínas Supressoras de Tumor/biossíntese , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Biochem Biophys Res Commun ; 421(3): 585-90, 2012 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-22525671

RESUMO

Suicide gene therapy of glioma based on herpes simplex virus type I thymidine kinase (HSV-TK) and prodrug ganciclovir (GCV) suffers from the lack of efficacy in clinical trials, which is mostly due to low transduction efficacy and absence of bystander effect in tumor cells. Recently, stem cells as cellular delivery vehicles of prodrug converting gene has emerged as a new treatment strategy for malignant glioma. In this study, we evaluated the anti-glioma effect of suicide gene therapy using human bone marrow mesenchymal stem cells expressing HSV-TK (MSCs-TK) combined with valproic acid (VPA), which can upregulate the gap junction proteins and may enhance the bystander effect of suicide gene therapy. Expression of HSV-TK in MSCs was confirmed by RT-PCR analysis and the sensitivity of MSCs-TK to GCV was assessed. A bystander effect was observed in co-cultures of MSCs-TK and U87 glioma cells by GCV in a dose-dependent manner. VPA induced the expression of the gap junction proteins connexin (Cx) 43 and 26 in glioma cell and thereby enhanced the bystander effect in co-culture experiment. The enhanced bystander effect was inhibited by the gap junction inhibitor 18-ß-glycyrrhetinic acid (18-GA). Moreover, the combined treatment with VPA and MSCs-TK synergistically enhanced apoptosis in glioma cells by caspase activation. In vivo efficacy experiments showed that combination treatment of MSCs-TK and VPA significantly inhibited tumor growth and prolonged the survival of glioma-bearing mice compared with single-treatment groups. In addition, TUNEL staining also demonstrated a significant increase in the number of apoptotic cells in the combination treated group compared with single-treatment groups. Taken together, these results provide the rational for designing novel experimental protocols to increase bystander killing effect against intracranial gliomas using MSCs-TK and VPA.


Assuntos
Neoplasias Encefálicas/terapia , Efeito Espectador , Terapia Genética/métodos , Glioma/terapia , Células-Tronco Mesenquimais/enzimologia , Timidina Quinase/genética , Ácido Valproico/administração & dosagem , Animais , Apoptose , Linhagem Celular Tumoral , Conexinas/agonistas , Conexinas/biossíntese , Herpesvirus Humano 1/enzimologia , Humanos , Transplante de Células-Tronco Mesenquimais , Camundongos , Camundongos Nus , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto
14.
J Biomed Biotechnol ; 2012: 362473, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22500090

RESUMO

Numerous studies have shown the benefits of mesenchymal stem cells (MSCs) on the repair of spinal cord injury (SCI) model and on behavioral improvement, but the underlying mechanisms remain unclear. In this study, to investigate possible mechanisms by which MSCs contribute to the alleviation of neurologic deficits, we examined the potential effect of human umbilical cord blood-derived MSCs (hUCB-MSCs) on the endogenous cell proliferation and oligogenesis after SCI. SCI was injured by contusion using a weight-drop impactor and hUCB-MSCs were transplanted into the boundary zone of the injured site. Animals received a daily injection of bromodeoxyuridine (BrdU) for 7 days after treatment to identity newly synthesized cells of ependymal and periependymal cells that immunohistochemically resembled stem/progenitor cells was evident. Behavior analysis revealed that locomotor functions of hUCB-MSCs group were restored significantly and the cavity volume was smaller in the MSCs-transplanted rats compared to the control group. In MSCs-transplanted group, TUNEL-positive cells were decreased and BrdU-positive cells were significantly increased rats compared with control group. In addition, more of BrdU-positive cells expressed neural stem/progenitor cell nestin and oligo-lineage cell such as NG2, CNPase, MBP and glial fibrillary acidic protein typical of astrocytes in the MSC-transplanted rats. Thus, endogenous cell proliferation and oligogenesis contribute to MSC-promoted functional recovery following SCI.


Assuntos
Sangue Fetal/citologia , Transplante de Células-Tronco Mesenquimais , Neurogênese/fisiologia , Traumatismos da Medula Espinal/cirurgia , Cicatrização/fisiologia , Análise de Variância , Animais , Apoptose/fisiologia , Comportamento Animal/fisiologia , Processos de Crescimento Celular/fisiologia , Histocitoquímica , Humanos , Masculino , Células-Tronco Mesenquimais/fisiologia , Ratos , Ratos Sprague-Dawley , Traumatismos da Medula Espinal/metabolismo
15.
Reprod Biol Endocrinol ; 10: 31, 2012 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-22531046

RESUMO

BACKGROUND: Interleukin (IL)-18 is an important regulator of innate and acquired immune responses and has multiple roles in chronic inflammation and autoimmune disorders. Obesity is characterized by low- grade chronic inflammation. IL-18 has been suggested as an adipogenic cytokine that is associated with excess adiposity. The purpose of this study is to evaluate the relationship between IL-18 gene polymorphisms (-137 G/C and -607 C/A) and obesity. METHODS: All 680 subjects were genotyped for the polymorphisms of IL-18 gene promoters (at positions -137 G/C and -607 C/A) using a polymerase chain reaction (271 cases with BMI ≥25 kg/m² and 409 controls with BMI <25 kg/m²). A chi-square test was used to compare the genotype and allele frequencies between the cases and control populations. RESULTS: Analyses of the genotype distributions revealed that IL-18 -607 C/A polymorphism was associated with an increase in body mass index in obese women in the Korean population (chi(2) = 12.301, df = 2, p = 0.015). CONCLUSION: Carriage of the A allele at position -607 in the promoter of the IL-18 gene may have a role in the development of obesity.


Assuntos
Índice de Massa Corporal , Interleucina-18/genética , Obesidade/genética , Adulto , Povo Asiático/genética , Feminino , Frequência do Gene , Haplótipos , Humanos , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único
16.
Stem Cell Res Ther ; 2(5): 38, 2011 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-21939558

RESUMO

INTRODUCTION: Stem cell transplantation is a promising therapeutic strategy for the treatment of stroke. Mesenchymal stem cells (MSCs) are a potential cell source for clinical application because they can be easily obtained and cultivated with a high proliferative capacity. The safety and efficacy of cell therapy depends on the mode of cell administration. To determine the therapeutic potential of intrathecal administration of MSCs by lumbar puncture (LP), we administrated human umbilical cord blood-derived MSCs (hUCB-MSCs) intrathecally into the lumbar spinal cord or intravenously into the tail vein in a rat model of stroke, and then investigated whether hUCB-MSCs could enter the brain, survive, and improve post-stroke neurological functional recovery. METHODS: hUCB-MSCs (1.0 × 10(6)) were administrated three days after stroke induced by occlusion of the middle cerebral artery. The presence of hUCB-MSCs and their survival and differentiation in the brain tissue of the rats was examined by immunohistochemistry. Recovery of coordination of movement after administration of hUCB-MSCs was examined using a Rotarod test and adhesive-removal test on the 7th, 14th, 21st, and 28th days after ischemia. The volume of ischemic lesions seven days after the experimental procedure was evaluated using 2-3-5-triphenyltetrazolium (TTC) staining. RESULTS: Rats receiving hUCB-MSCs intrathecally by LP had a significantly higher number of migrated cells within the ischemic area when compared with animals receiving cells intravenously. In addition, many of the cells administered intrathecally survived and a subset of them expressed mature neural-lineage markers, including the mature neuron marker NeuN and glial fibrillary acidic protein, typical of astrocytes. Animals that received hUCB-MSCs had significantly improved motor function and reduced ischemic damage when compared with untreated control animals. Regardless of the administration route, the group treated with 1 × 10(6) hUCB-MSCs showed better neurological recovery, without significant differences between the two treatment groups. Importantly, intrathecal administration of 5 × 10(5) hUCB-MSCs significantly reduced ischemic damage, but not in the intravenously treated group. Furthermore, the cells administered intrathecally survived and migrated into the ischemic area more extensively, and differentiated significantly into neurons and astrocytes. CONCLUSIONS: Together, these results indicate that intrathecal administration of MSCs by LP may be useful and feasible for MSCs treatment of brain injuries, such as stroke, or neurodegenerative disorders.


Assuntos
Sangue Fetal/citologia , Isquemia/terapia , Células-Tronco Mesenquimais/citologia , Animais , Modelos Animais de Doenças , Humanos , Imuno-Histoquímica , Injeções Intravenosas , Transplante de Células-Tronco Mesenquimais , Ratos , Punção Espinal , Acidente Vascular Cerebral/terapia
17.
Cell Transplant ; 20(11-12): 1855-66, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21375801

RESUMO

The ability of mesenchymal stem cells (MSCs) to differentiate into neural cells makes them potential replacement therapeutic candidates in neurological diseases. Presently, overexpression of brain-derived neurotrophic factor (BDNF), which is crucial in the regulation of neural progenitor cell differentiation and maturation during development, was sufficient to convert the mesodermal cell fate of human umbilical cord blood-derived MSCs (hUCB-MSCs) into a neuronal fate in culture, in the absence of specialized induction chemicals. BDNF overexpressing hUCB-MSCs (MSCs-BDNF) yielded an increased number of neuron-like cells and, surprisingly, increased the expression of neuronal phenotype markers in a time-dependent manner compared with control hUCB-MSCs. In addition, MSCs-BDNF exhibited a decreased labeling for MSCs-related antigens such as CD44, CD73, and CD90, and decreased potential to differentiate into mesodermal lineages. Phosphorylation of the receptor tyrosine kinase B (TrkB), which is a receptor of BDNF, was increased significantly in MSC-BDNF. BDNF overexpression also increased the phosphorylation of ß-catenin and extracellular signal-regulated kinases (ERKs). Inhibition of TrkB availability by treatment with the TrkB-specific inhibitor K252a blocked the BDNF-stimulated phosphorylation of ß-catenin and ERKs, indicating the involvement of both the ß-catenin and ERKs signals in the BDNF-stimulated and TrkB-mediated neural differentiation of hUCB-MSCs. Reduction of ß-catenin availability using small interfering RNA-mediated gene silencing inhibited ERKs phosphorylation. However, ß-catenin activation was maintained. In addition, inhibition of ß-catenin and ERKs expression levels abrogated the BDNF-stimulated upregulation of neuronal phenotype markers. Furthermore, MSC-BDNF survived and migrated more extensively when grafted into the lateral ventricles of neonatal mouse brain, and differentiated significantly into neurons in the olfactory bulb and periventricular astrocytes. These results indicate that BDNF induces the neural differentiation of hUCB-MSCs in culture via the TrkB-mediated phosphorylation of ERKs and ß-catenin and following transplantation into the developing brain.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Encéfalo/crescimento & desenvolvimento , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Neurogênese , Neurônios/citologia , Receptor trkB/metabolismo , Animais , Animais Recém-Nascidos , Encéfalo/patologia , Fator Neurotrófico Derivado do Encéfalo/genética , Carbazóis/farmacologia , Células Cultivadas , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Sangue Fetal/citologia , Humanos , Alcaloides Indólicos/farmacologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Neurônios/metabolismo , Fosforilação/efeitos dos fármacos , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Receptor trkB/antagonistas & inibidores , Regulação para Cima , beta Catenina/antagonistas & inibidores , beta Catenina/genética , beta Catenina/metabolismo
18.
Biochem Biophys Res Commun ; 407(4): 741-6, 2011 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-21439934

RESUMO

In this study, we showed that knocking-down interleukin-8 (IL-8) in glioma cells, or its receptor, CXC chemokine receptor 1 (CXCR1) in hUCB-MSCs reduced hUCB-MSC migration toward glioma cells in a Transwell chamber. In contrast, CXCR1-transfected hUCB-MSCs (CXCR1-MSCs) showed a superior capacity to migrate toward glioma cells in a Transwell chamber compared to primary hUCB-MSCs. Furthermore, these transfected cells also demonstrated the same ability to migrate toward tumors in mice bearing intracranial human gliomas as shown by histological and in vivo imaging analysis. Our findings indicate that overexpression of CXCR1 could be a useful tool for MSC-based gene therapy to achieve a sufficient quantity of therapeutic MSCs that are localized within tumors.


Assuntos
Neoplasias Encefálicas/terapia , Movimento Celular , Terapia Genética/métodos , Glioma/terapia , Células-Tronco Mesenquimais/fisiologia , Receptores de Interleucina-8A/genética , Animais , Linhagem Celular Tumoral , Sangue Fetal/citologia , Técnicas de Silenciamento de Genes , Humanos , Interleucina-8/genética , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Nus , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Hum Gene Ther ; 22(6): 733-43, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21261460

RESUMO

Clinical trials of gene therapy using a viral delivery system for glioma have been limited. Recently, gene therapy using stem cells as the vehicles for delivery of therapeutic agents has emerged as a new treatment strategy for malignant brain tumors. In this study, we used human umbilical cord blood-derived mesenchymal stem cells (UCB-MSCs) as delivery vehicles with glioma-targeting capabilities, and modified interleukin-12 (IL-12p40N220Q; IL-12M) as a novel therapeutic gene. We also engineered UCB-MSCs to secret IL-12M (UCB-MSC-IL12M) via tetrameric cell-permeable peptide (4HP4)-mediated adenoviral transduction. We confirmed the migratory capacity of UCB-MSC-IL12M toward GL26 mouse glioma cells by an in vitro migration assay and in vivo injection of UCB-MSC-IL12M into the ipsilateral hemisphere of implanted gliomas in C57BL/6 mice. In vivo efficacy experiments showed that intratumoral injection of UCB-MSC-IL12M significantly inhibited tumor growth and prolonged the survival of glioma-bearing mice compared with control mice. Antitumor effects were associated with increased local IL-12M levels, followed by interferon-γ secretion and T-cell infiltration in intracranial gliomas, as well as antiangiogenesis. Interestingly, tumor-free mice after UCB-MSC-IL12M treatment were resistant to ipsilateral and contralateral tumor rechallenge, which was closely associated with tumor-specific long-term T-cell immunity. Thus, our results provide the rationale for designing novel experimental protocols to induce long-term antitumor immunity against intracranial gliomas using UCB-MSCs as an effective delivery vehicle for therapeutic cytokines including IL-12M.


Assuntos
Transplante de Células-Tronco de Sangue do Cordão Umbilical , Ganglioglioma/terapia , Terapia Genética/métodos , Interleucina-12/genética , Transplante de Células-Tronco Mesenquimais , Animais , Humanos , Interferon gama/metabolismo , Interleucina-12/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T/imunologia
20.
Int J Oncol ; 38(1): 97-103, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21109930

RESUMO

Mesenchymal stem cells (MSCs) can be used as a delivery vehicle for gene therapy against brain tumors, because these cells have a migratory capacity toward glioma cells. Soluble factors including chemokines or growth factors expressed and released by glioma cells mediate the tropism of MSCs for gliomas. Among them, stromal cell-derived factor-1α (SDF-1α) has been identified as a key molecule related to the tropism of MSC in many cancers containing gliomas. In this study, we found that overexpression of the SDF-1α receptor, CXCR4, on human umbilical cord blood-derived MSCs (hUCB-MSCs) enhanced the migratory capacity of MSCs toward gliomas. We showed that hUCB-MSCs have the migration ability toward the glioma cell lines and primary glioma cells. SDF-1α treatment increased the migration capacity of hUCB-MSCs in a dose-dependent manner and inhibition of SDF-1α or CXCR4 by treatment with the anti-SDF-1α or the CXCR4 antagonist AMD3100 blocked the migration capacity of hUCB-MSCs toward glioma cells. Furthermore, CXCR4-overexpressed hUCB-MSCs (hMSCs-CXCR4) showed a stronger migration capacity toward glioma cells in vitro compared with control MSCs, and also exhibited enhanced migration to glioma cells in an intracranial human malignant glioma xenograft model. These results indicate that SDF-1α/CXCR4 could be involved in recruitment of hUCB-MSCs to glioma cells and that overexpression of CXCR4 may be a useful tool for stem cell-based glioma therapy.


Assuntos
Neoplasias Encefálicas/genética , Sangue Fetal/fisiologia , Glioma/genética , Células-Tronco Mesenquimais/fisiologia , Receptores CXCR4/genética , Animais , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Movimento Celular/fisiologia , Células Cultivadas , Sangue Fetal/citologia , Sangue Fetal/metabolismo , Glioma/metabolismo , Glioma/patologia , Humanos , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Camundongos , Camundongos Nus , Receptores CXCR4/metabolismo , Transdução de Sinais , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...