Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
J Oncol ; 2020: 2319194, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33178271

RESUMO

Glioma is the most common and deadly tumor in central nervous system. According to previous studies, long noncoding RNAs (lncRNA) and transcription factors were significant factors of gliomas progression by regulating gliomas immune microenvironment. In our study, we built two independent cohorts from CGGA and TCGA. And we extracted 253 immune-related lncRNA correlated with prognosis. After LASSO analysis and multivariate Cox regression analysis, 8 immune-related lncRNA were used to construct classifier. The effectiveness of classifier was confirmed in both CGGA (AUC = 0.869) and TCGA (AUC = 0.902) cohorts. The correlation between transcription factors and immune-related lncRNA was calculated by WCGNA. Eventually, we built a network between 8 lncRNA and transcription factors. The function of core immune-related lncRNA in gliomas immune microenvironment was also investigated by CIBERTSORT. Our research provided a strong classifier of immune-related lncRNA to predict gliomas patient outcome. We also found the correlation between core immune-related lncRNA and transcription factors. These results may stimulate new strategy of immunotherapy in gliomas patients.

2.
Cancer Sci ; 106(7): 857-66, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25940092

RESUMO

Vasculogenic mimicry (VM) refers to the process by which highly aggressive tumor cells mimic endothelial cells to form vessel-like structures that aid in supplying enough nutrients to rapidly growing tumors. Histone deacetylases (HDACs) regulate the expression and activity of numerous molecules involved in cancer initiation and progression. Notably, HDAC3 is overexpressed in the majority of carcinomas. However, thus far, no data are available to support the role of HDAC3 in VM. In this study, we subjected glioma specimens to immunohistochemical and histochemical double-staining methods and found that VM and HDAC3 expression were related to the pathological grade of gliomas. The presence of VM correlated with HDAC3 expression in glioma tissues. The formation of tubular structures, as determined by the tube formation assay to evaluate VM, was impaired in U87MG cells when transfected by siRNA or treated with an HDAC3 inhibitor. Importantly, the expression of VM-related molecules such as MMP-2/14 and laminin5γ2 was also affected when HDAC3 expression was altered. Furthermore, U87MG cells were treated with a phosphoinositide 3-kinase (PI3K) inhibitor or/and ERK inhibitor and found that the PI3K and ERK signaling pathways play key roles in VM; whereas, in VM, the two signaling pathways did not act upstream or downstream from each other. Taken together, our findings showed that HDAC3 contributed to VM in gliomas, possibly through the PI3K/ERK-MMPs-laminin5γ2 signaling pathway, which could potentially be a novel therapeutic target for gliomas.


Assuntos
Glioma/enzimologia , Histona Desacetilases/metabolismo , Neovascularização Patológica/enzimologia , Adulto , Linhagem Celular Tumoral , Feminino , Expressão Gênica , Glioma/irrigação sanguínea , Histona Desacetilases/genética , Humanos , Laminina/metabolismo , Sistema de Sinalização das MAP Quinases , Masculino , Metaloproteinases da Matriz/metabolismo , Pessoa de Meia-Idade , Fosfatidilinositol 3-Quinases/metabolismo
3.
Mol Med Rep ; 12(1): 165-72, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25760394

RESUMO

The poor therapeutic effect of traditional antiangiogenic therapy on glioblastoma multiforme (GBM) may be attributed to vasculogenic mimicry (VM), which was previously reported to be promoted by cancer stem-like cells (SLCs). All-trans retinoic acid (ATRA), a potent reagent which drives differentiation, was reported to be able to eradicate cancer SLCs in certain malignancies. The aim of the present study was to investigate the effects of ATRA on the VM formation ability of U87 glioblastoma SLCs. The expression of cancer SLC markers CD133 and nestin was detected using immunocytochemistry in order to identify U87 SLCs. In addition, the differentiation of these SLCs was observed through detecting the expression of glial fibrillary acidic protein (GFAP), ß-tubulin III and galactosylceramidase (Galc) using immunofluorescent staining. The results showed that the expression levels of GFAP, ß-tubulin III and Galc were upregulated following treatment with ATRA in a dose-dependent manner. Furthermore, ATRA significantly reduced the proliferation, invasiveness, tube formation and vascular endothelial growth factor (VEGF) secretion of U87 SLCs. In conclusion, the VM formation ability of SLCs was found to be negatively correlated with differentiation. These results therefore suggested that ATRA may serve as a promising novel agent for the treatment of GBM due to its role in reducing VM formation.


Assuntos
Inibidores da Angiogênese/farmacologia , Biomarcadores Tumorais/genética , Células-Tronco Neoplásicas/efeitos dos fármacos , Neovascularização Patológica/prevenção & controle , Neuroglia/efeitos dos fármacos , Tretinoína/farmacologia , Antígeno AC133 , Antígenos CD/genética , Antígenos CD/metabolismo , Biomarcadores Tumorais/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Galactosilceramidase/genética , Galactosilceramidase/metabolismo , Expressão Gênica , Proteína Glial Fibrilar Ácida/agonistas , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Nestina/genética , Nestina/metabolismo , Neuroglia/metabolismo , Neuroglia/patologia , Peptídeos/genética , Peptídeos/metabolismo , Tubulina (Proteína)/agonistas , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
4.
J Neurotrauma ; 32(7): 506-15, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25225747

RESUMO

Adipocyte-derived stem cells have emerged as a novel source of stem cell therapy for their autologous and readily accessible and pluripotent potential to differentiate into different lineages such as neural stem cells (NSCs) and endothelial progenitor cells (EPCs). Transplantation of NSCs and EPCs has been promising for the repair of brain injury. We explored using co-transplanted hydrogel scaffold to improve the survival of the transplanted cells and recovery of neurological function. Adult Wistar rats were transplanted with EPC-hydrogel, NSC-hydrogel, NSC-EPC-hydrogel, EPC only, or NSC only 7 days after cortical contusion injury. Behavioral tests were performed to evaluate neurological function before, and 1, 2, 3, and 4 weeks after transplantation. Size of injury, extent of vascularization, as well as the survival and differentiation of the transplanted EPCs and NSCs, were evaluated at week 5. All transplantation groups displayed significantly better neurological function compared with the control groups. Improved neurological function correlated with significantly smaller injury volumes than that of the saline group. Using immunostaining, we have shown that while transplanted NSCs differentiated into both neurons and astrocytes, the EPCs were incorporated into vessel epithelia. The extent of reactive gliosis (based on glial fibrillary acidic protein immunostaining) was significantly reduced in all treatment groups (NSC-EPC-hydrogel, NSC-hydrogel, and EPC-hydrogel) when compared with the saline group, with the highest reduction in the NSC-EPC-hydrogel transplantation group. Thus, co-transplantation of hydrogel scaffold provides a more conducive environment for the survival and differentiation of NSCs and EPCs at the site of brain injury, leading to improved vascularization and better recovery of neurological function.


Assuntos
Adipócitos/transplante , Lesões Encefálicas/terapia , Recuperação de Função Fisiológica/fisiologia , Transplante de Células-Tronco/métodos , Animais , Comportamento Animal/fisiologia , Lesões Encefálicas/fisiopatologia , Modelos Animais de Doenças , Hidrogel de Polietilenoglicol-Dimetacrilato , Masculino , Atividade Motora/fisiologia , Ratos , Ratos Wistar , Alicerces Teciduais , Resultado do Tratamento
5.
J Neurooncol ; 112(3): 339-45, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23417321

RESUMO

A previous report has confirmed the existence and clinical significance of vasculogenic mimicry (VM) in glioma. However, its conclusions about the negative clinical significance of VM in glioblastoma are based on a small group of patients and, thus, might be unconvincing. The aim of the present study was to reevaluate the clinical significance of VM in glioblastoma. Patients were classified as VM-positive or VM-negative according to CD34 and periodic acid-Schiff staining. The association between VM and the clinical characteristics of the patients was analyzed. Univariate and multivariate analyses were carried out to identify the independent prognostic factors for overall survival using the Cox regression hazard model. Survival times were estimated using the Kaplan-Meier method and compared using the log-rank test. Of all 86 glioblastomas, 23 were found to have VM. The presence of VM in glioblastoma was not associated with gender, age, Karnofsky performance status, hydrocephalus, tumor burden, microvessel density, tumor relapse, or the extent of tumor resection. The univariate and multivariate analyses revealed that VM is an independent prognostic factor for overall survival. The median survival time for patients with VM was 11.17 months compared with 16.10 months for those without VM (P = 0.017). In addition to VM, an age of 65 years or older, a KPS of 60 or less, a large tumor burden are significant prognostic factors for patient survival. Our data suggest that VM might be an independent adverse prognostic factor in newly diagnosed GBM, further prospective studies are needed to answer this question.


Assuntos
Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/patologia , Glioblastoma/irrigação sanguínea , Glioblastoma/patologia , Adolescente , Adulto , Idoso , Antígenos CD34/análise , Antígenos CD34/biossíntese , Neoplasias Encefálicas/mortalidade , Feminino , Glioblastoma/mortalidade , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Prognóstico , Modelos de Riscos Proporcionais , Adulto Jovem
6.
Br J Neurosurg ; 27(2): 187-93, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22984981

RESUMO

OBJECTIVE: The purpose of this study was to evaluate the safety and efficacy of transorbital puncture for the retreatment of previously embolized cavernous sinus dural arteriovenous fistulas (DAVFs) via a superior ophthalmic vein (SOV) approach. MATERIALS AND METHODS: During a 12-year period, 9 consecutive patients with previously embolized cavernous sinus DAVFs underwent retreatment via the transorbital SOV approach. RESULTS: All of the nine cases of previously embolized cavernous sinus DAVFs were successfully embolized. Clinical follow-ups were conducted in all nine cases at the duration of 17-141 months (61.22 ± 39.13 months). No recanalization occurred during the follow-up period. A subtle ptosis appeared in two patients and disappeared in one of the two cases after a 4-year follow-up. One patient suffered from paroxysmal positional vertigo and bruit for nearly 2 years after the treatment, but the follow-up angiography demonstrated no recurrence. One patient had persistent visual impairment caused by the initial venous stasis retinopathy. One patient with a history of a procedure-related transient decrease in visual acuity had it return to the normal level. The remaining four cases had clear improvement in the ocular symptoms and became completely asymptomatic during the follow-up period. No patient worsened or developed new symptoms. CONCLUSION: The approach of surgical cannulation of the SOV for the retreatment of previously embolized cavernous sinus DAVFs was proved feasible and efficient, especially when the transarterial and transfemoral venous approaches were inaccessible. However, if the SOV is not dilated enough or is located deeply in the orbit, transorbital venous puncture access may not be possible.


Assuntos
Cateterismo/métodos , Seio Cavernoso/anormalidades , Malformações Vasculares do Sistema Nervoso Central/terapia , Olho/irrigação sanguínea , Veias/cirurgia , Adulto , Idoso , Seio Cavernoso/diagnóstico por imagem , Malformações Vasculares do Sistema Nervoso Central/diagnóstico por imagem , Embolização Terapêutica , Procedimentos Endovasculares/métodos , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Complicações Pós-Operatórias/etiologia , Radiografia , Retratamento , Estudos Retrospectivos , Trombose Venosa/cirurgia , Adulto Jovem
7.
Neurosci Lett ; 534: 327-32, 2013 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-23220650

RESUMO

MicroRNAs (miRNAs) are small noncoding RNAs that function as tumor suppressors or oncogenes. MicroRNA-107 (miR-107), a transcriptional target of p53, is deregulated in many cancer cell lines. Here, we showed that miR-107 is down-regulated in glioma tissues and cell lines, in particular, p53-mutated U251 and A172. Transfection of wild-type p53 into these cells stimulated miR-107 expression. To investigate the role of miR-107 in tumorigenesis, we constructed a lentiviral vector overexpressing miR-107. Notably, miR-107 inhibited proliferation and arrested the cell cycle at the G0-G1 phase in glioma cells. Transduction of Lenti-GFP-miR-107 into glioma cells inhibited CDK6 and Notch-2 protein expression. Our findings collectively demonstrate that p53-induced miR-107 suppresses brain tumor cell growth and down-regulates CDK6 and Notch-2 expression, supporting its tumor suppressor role and utility as a target for glioma therapy.


Assuntos
Quinase 6 Dependente de Ciclina/biossíntese , MicroRNAs/metabolismo , Receptor Notch2/biossíntese , Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Regulação para Baixo , Glioma , Humanos , Lentivirus/genética , MicroRNAs/genética , Transdução Genética , Transfecção , Proteína Supressora de Tumor p53/genética
8.
Neuroreport ; 23(8): 513-8, 2012 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-22525837

RESUMO

Human placental decidua basalis-derived mesenchymal stem cells (DBMSCs) have been identified as valuable sources for cell transplantation. In this study, we found that DBMSCs could be induced to form neural stem cells in the form of neurospheres. These neurospheres were further differentiated into dopamine neuron-like cells with a cocktail of cytokines. The differentiated DBMSCs were verified through the presence of a neuron-like morphology, the expression of specific dopamine neuron makers, and the production of dopamine. In addition, this differentiation capacity of DBMSCs was not affected by long-term culture, and the cells maintained a normal karyotype in vitro. The dopamine neuronal differentiation and the relative safety transplantation potential of DBMSCs may facilitate stem cell therapeutic approaches to Parkinson's disease.


Assuntos
Diferenciação Celular/fisiologia , Decídua/citologia , Neurônios Dopaminérgicos/fisiologia , Células-Tronco Mesenquimais/citologia , Antígenos CD/metabolismo , Células Cultivadas , Cromatografia Líquida de Alta Pressão , Dopamina/metabolismo , Feminino , Humanos , Recém-Nascido , Cariotipagem , Masculino , Proteínas do Tecido Nervoso/metabolismo , Gravidez
9.
Cancer Biol Ther ; 13(5): 341-8, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22258034

RESUMO

Vasculogenic mimicry (VM), a process involving the formation of a tubular structure by highly invasive and genetically dysregulated tumor cells, can supplement the function of blood vessels to transport nutrients and oxygen to maintain the growth of tumor cells in many malignant tumors. We aimed to explore the existence of VM and its clinical significance in medulloblastoma in this study. VM was identified in 9 out of 41 (22%) medulloblastoma tissues. Immunohistochemical studies revealed that the presence of VM was associated with the expression of MMP-2, MMP-14, EphA2 and laminin 5γ2. Tumor tissues with VM were associated with lower microvessel density (MVD), which was indirect evidence of the blood supply function of VM. Survival analysis and log-rank tests showed that patients with VM had shorter overall survival time than those without VM. Multivariate analysis and the Cox proportional hazards model identified VM as independent prognostic factor for overall survival. Our results confirmed the existence of VM for the first time and revealed that VM is a strong independent prognostic factor for survival in patients with medulloblastoma.


Assuntos
Neoplasias Cerebelares/irrigação sanguínea , Meduloblastoma/irrigação sanguínea , Adolescente , Adulto , Neoplasias Cerebelares/metabolismo , Neoplasias Cerebelares/patologia , Criança , Pré-Escolar , Feminino , Humanos , Imuno-Histoquímica , Lactente , Masculino , Meduloblastoma/metabolismo , Meduloblastoma/patologia , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Adulto Jovem
10.
J Neuroimmunol ; 242(1-2): 9-15, 2012 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-22123613

RESUMO

Glioma, the most common tumor of the central nervous system (CNS), currently results in a high rate of morbidity and mortality. The expression of CD133, a stem-like cell marker expressed in the glioma cells, is believed to lead to tumorigenesis in the human brain. Thus, it is necessary to find a proper method to specifically kill the CD133(+) glioma cells. Dendritic cell (DC)/tumor hybrids are proven to be able to induce an effective immune response, leading to killing of glioma cells in vitro. We isolated CD133(+) cells from a population of primary glioma cells, and cultured autologous DCs and T cells at the same time. Next, we electrofused the DCs with the CD133(+) glioma cells and with CD133- ones, in order to explore a new strategy for glioma therapy. We then exposed the T cells to five separate groups of cells: DC/CD133(+) hybrids, DC/CD133(-) hybrids, DCs alone, unsorted glioma cells alone and mixed DCs-glioma cells. A cytotoxicity assay showed that T cells stimulated by either type of hybrid were able to kill cultured autologous glioma cells significantly more effectively than those stimulated by the other three cell types (P<0.05). The amounts of IFN-γ secreted by T cells stimulated by the two types of fused cells were obviously increased compared to those stimulated by the other three cell types (P<0.05). However, no significant differences were noted between the effects of the two hybrids, neither in the cytotoxicity assay nor in the IFN-γ release assay (P>0.05). Therefore, both DC/CD133(+) and DC/CD133(-) hybrids can cause significant T cell immune responses in vitro. There were no significant differences between the immune responses caused by the two types of hybrids.


Assuntos
Antígenos CD/biossíntese , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/prevenção & controle , Células Dendríticas/imunologia , Glioma/imunologia , Glioma/prevenção & controle , Glicoproteínas/biossíntese , Ativação Linfocitária/imunologia , Subpopulações de Linfócitos T/imunologia , Antígeno AC133 , Neoplasias Encefálicas/patologia , Fusão Celular , Células Cultivadas , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Técnicas Eletroquímicas/métodos , Glioma/patologia , Humanos , Células Híbridas , Peptídeos , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/patologia , Células Tumorais Cultivadas
11.
Cancer Lett ; 312(2): 168-77, 2011 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-21924825

RESUMO

Immunotoxins have shown great promise as an alternative treatment for brain malignancies such as gliomas, but their failure to penetrate into the tumor mass remains a major problem. Mesenchymal stem cells exhibit tropism to tumor tissue and may serve as a cellular vehicle for the delivery and local production of antitumor agents. In this study, we used human bone marrow-derived mesenchymal stem cells (hMSCs) as a vehicle for the targeted delivery of EphrinA1-PE38, a very specific immunotoxin against the EphA2 receptor that is overexpressed in gliomas. hMSCs were transduced with adenovirus to express secretable EphrinA1-PE38. Our invitro assays confirmed the expression, release and selective killing effect of the immunotoxin produced by hMSCs. Furthermore, the intratumoral injection of engineered hMSCs was effective at inhibiting tumor growth in a malignant glioma tumor model. These results indicate that gene therapy utilizing EphrinA1-PE38-secreting hMSCs may provide a novel approach for the local treatment of malignant gliomas.


Assuntos
Células da Medula Óssea/patologia , Neoplasias Encefálicas/tratamento farmacológico , Glioma/tratamento farmacológico , Imunotoxinas/uso terapêutico , Células-Tronco Mesenquimais/patologia , Receptor EphA2/imunologia , Animais , Sequência de Bases , Western Blotting , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Primers do DNA , Feminino , Glioma/patologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Reação em Cadeia da Polimerase
12.
Cytotherapy ; 13(1): 46-53, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20735164

RESUMO

BACKGROUND AIMS: This study aimed to observe nine factors expressed in rat ischemic brain after transplantation of bone marrow stromal cells (BMSC) and/or endothelial progenitor cells (EPC). These factors were vascular endothelial growth factor (VEGF), stromal cell-derived factor-1 (SDF-1), basic fibroblast growth factor (bFGF), insulin-like growth factor (IGF-l), transforming growth factor-ß (TGF-ß), platelet-derived growth factor-BB (PDGF-BB), brain-derived neurotrophic factor (BDNF), glial cell line-derived neurotrophic factor (GDNF) and nerve growth factor (NGF). METHODS: Adult Wistar rats were divided randomly into four groups: a vehicle group, BMSC group, EPC group and BMSC combined with EPC group. The rats were subjected to middle cerebral artery occlusion (MCAO) then implanted intravenously with 3 × 10(6) BMSC, EPC, BMSC/EPC or phosphate-buffered saline (PBS) 24 h after MCAO. Neurologic functional deficits were measured on days 1, 7, 14, 28 after transplantation. On day 7 after transplantation, quantitative reverse transcription (qRT)-polymerase chain reaction (PCR) and Western blot were employed to detect the expression of VEGF, SDF-1, bFGF, IGF-l, TGF-ß, PDGF-BB, BDNF, GDNF and NGF. RESULTS: The neurologic evaluation found that the neurologic severity scores were no different between the four groups on day 1, and the scores of rats in the BMSC/EPC group were significantly lower than those of rats in the other groups on days 7, 14 and 28 after transplantation. The expressions of bFGF, VEGF and BNDF were significantly higher in the BMSC/EPC group compared with the other groups. CONCLUSIONS: The intravenous transplantation of BMSC combined with EPC could promote the functional rehabilitation of rats with focal cerebral ischemia, and the mechanism may be related to the enhanced expression of factors.


Assuntos
Células da Medula Óssea/citologia , Isquemia Encefálica/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Citocinas/metabolismo , Células Endoteliais/transplante , Transplante de Células-Tronco , Animais , Comportamento Animal , Células da Medula Óssea/metabolismo , Isquemia Encefálica/patologia , Isquemia Encefálica/fisiopatologia , Isquemia Encefálica/terapia , Células Endoteliais/citologia , Microvasos/patologia , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Ratos , Ratos Wistar , Células-Tronco/citologia , Células-Tronco/metabolismo , Células Estromais/citologia , Células Estromais/transplante
13.
Neurochem Res ; 35(10): 1522-9, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20658188

RESUMO

Mesenchymal stem cells are capable of differentiating into dopaminergic-like cells, but currently no report has been available to describe the induction of human umbilical vein mesenchymal stem cells (HUVMSCs) into dopaminergic-like cells. In this study, we induced HUVMSCs in vitro into neurospheres constituted by neural stem-like cells, and further into cells bearing strong morphological, phenotypic and functional resemblances with dopaminergic-like cells. These HUVMSC-derived dopaminergic-like cells, after grafting into the brain of a rat model of Parkinson's disease (PD), showed a partial therapeutic effect in terms of the behavioral improvement. Nerve growth factor was reported to improve the local microenvironment of the grafted cells, and we therefore further tested the effect of dopaminergic-like cell grafting combined with nerve growth factor (NGF) administration at the site of cell transplantation. The results showed that NGF administration significantly promoted the survival of the grafted cells in the host brain and enhanced the content of dopaminergic in the local brain tissue. Behavioral test demonstrated a significant improvement of the motor function of the PD rats after dopaminergic-like cell grafting with NGF administration as compared with that of rats receiving the cell grafting only. These results suggest that transplantation of the dopaminergic-like cells combined with NGF administration may represent a new strategy of stem cell therapy for PD.


Assuntos
Dopamina/metabolismo , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Atividade Motora , Fator de Crescimento Neural/uso terapêutico , Doença de Parkinson/terapia , Veias Umbilicais/citologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Diferenciação Celular , Humanos , Células-Tronco Mesenquimais/citologia , Doença de Parkinson/metabolismo , Doença de Parkinson/psicologia , Ratos , Ratos Sprague-Dawley
14.
Neuroimmunomodulation ; 17(4): 270-8, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20203533

RESUMO

LINGO-1 (leucine-rich repeat and Ig domain-containing, Nogo receptor-interacting protein) is an important component of the NgR receptor complex involved in RhoA activation and axon regeneration. The authors report on passive immunization with LINGO-1 polyclonal antiserum, a therapeutic approach to overcome NgR-mediated growth inhibition after spinal cord injury (SCI). The intrathecally administered high-titer rabbit-derived antiserum can be detected around the injury site within a wide time window; it blocks LINGO-1 in vivo with high molecular specificity. In this animal model, passive immunization with LINGO-1 antiserum significantly decreased RhoA activation and increased neuronal survival. Adult rats immunized in this manner show recovery of certain hindlimb motor functions after dorsal hemisection of the spinal cord. Thus, passive immunotherapy with LINGO-1 polyclonal antiserum may represent a promising repair strategy following acute SCI.


Assuntos
Citoproteção/efeitos dos fármacos , Imunização Passiva/métodos , Proteínas de Membrana/antagonistas & inibidores , Proteínas do Tecido Nervoso/antagonistas & inibidores , Recuperação de Função Fisiológica/efeitos dos fármacos , Traumatismos da Medula Espinal/tratamento farmacológico , Animais , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/imunologia , Citoproteção/imunologia , Modelos Animais de Doenças , Feminino , Soros Imunes/imunologia , Soros Imunes/farmacologia , Injeções Espinhais , Proteínas de Membrana/imunologia , Degeneração Neural/tratamento farmacológico , Degeneração Neural/imunologia , Degeneração Neural/fisiopatologia , Proteínas do Tecido Nervoso/imunologia , Paralisia/tratamento farmacológico , Paralisia/imunologia , Paralisia/fisiopatologia , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/imunologia , Traumatismos da Medula Espinal/imunologia , Traumatismos da Medula Espinal/fisiopatologia , Resultado do Tratamento , Proteína rhoA de Ligação ao GTP/efeitos dos fármacos , Proteína rhoA de Ligação ao GTP/metabolismo
15.
Int J Cancer ; 127(9): 2222-9, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-20127864

RESUMO

Inhibition of tumor neovascularization has profound effects on the growth of solid tumors. Our previous studies have shown the effect of VEGF165-PE38 recombinant immunotoxin on proliferation and apoptosis in human umbilical vein endothelial cells in vitro. In this study, we explored the direct inhibition of angiogenesis in chick chorioallantoic membrane and antiangiogenic therapy in a malignant glioma model. HEK293 cells were transfected with the pVEGF165PE38-IRES2-EGFP plasmid. ELISA was used to confirm the expression of VEGF165-PE38 in the transfected cells. These cells released 1396 + or - 131.9 pg VEGF165-PE38/1x10(4) cells/48 h into the culture medium and the supernatant was capable of inhibiting the growth of capillary-like structures in chick chorioallantoic membrane assay. In a murine malignant glioma model, plasmid was directly administered via multiple local intratumoral delivery. After day 16 the tumor volume in mice treated with pVEGF165PE38-IRES2-EGFP was significantly lower than that in mice in the control groups. Immunohistochemistry studies showed that the treated group had decreased expression of CD31. Quantitative analysis of microvessel density in the treated group was 1.99 + or - 0.69/0.74 mm(2), and was significantly lower than that in the control groups (9.33 + or - 1.99/0.74 mm(2), 8.09 + or - 1.39/0.74 mm(2) and 8.49 + or - 1.69/0.74 mm(2)). Immunohistochemistry analysis indicated that immunotoxin VEGF165-PE38 was distributed in the treated group in malignant glioma tissue. Our findings provide evidence that the in vivo production of VEGF165-PE38 through gene therapy using a eukaryotic expression plasmid had potential antiangiogenic activity in malignant glioma in vivo.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Terapia Genética , Glioma/terapia , Imunotoxinas/uso terapêutico , Fator A de Crescimento do Endotélio Vascular/genética , ADP Ribose Transferases/uso terapêutico , Animais , Toxinas Bacterianas/uso terapêutico , Linhagem Celular Tumoral , Exotoxinas/uso terapêutico , Estudos de Viabilidade , Glioma/irrigação sanguínea , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Plasmídeos , Pseudomonas/metabolismo , Transfecção , Fatores de Virulência/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto , Exotoxina A de Pseudomonas aeruginosa
16.
Nan Fang Yi Ke Da Xue Xue Bao ; 29(9): 1787-9, 2009 Sep.
Artigo em Chinês | MEDLINE | ID: mdl-19778790

RESUMO

OBJECTIVE: To investigate the biocompatibility of a novel cavernous nickel-titanium alloy with rat bone marrow stromal cells (BMSCs) in vitro. METHODS: Rat BMSCs were cultured on the surface of compact, microporous and macroporous nickel-titanium alloys, and the cell proliferation on day 3 during the culture was assessed using MTT assay. On day 7 of the cell culture, the cells were labeled with Hoechst33342 for cell counting under a fluorescence microscope. Scanning electron microscopy (SEM) was performed on day 7 of cell culture to observe the morphological changes of the cells. RESULTS: The cell proliferation rate and cell numbers differed significantly between the cavernous alloy groups and the compact alloy group (P<0.05), but similar between the former two groups (P>0.05). SEM showed that compared with the compact alloy, microporous and macroporous nickel-titanium alloys had better biocompatibility with the BMSCs, and the cells on the surface of the cavernous alloys had normal cell morphology. CONCLUSION: Cavernous nickel-titanium alloy has good biocompatibility and can promote the adhesion, aggregation and proliferation of rat BMSCs in vitro.


Assuntos
Materiais Biocompatíveis/farmacologia , Células da Medula Óssea/citologia , Teste de Materiais/métodos , Níquel/farmacologia , Células Estromais/citologia , Titânio/farmacologia , Animais , Adesão Celular , Células Cultivadas , Feminino , Masculino , Ratos , Ratos Sprague-Dawley
17.
Acta Neurochir (Wien) ; 151(11): 1483-92, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19499175

RESUMO

PURPOSE: Spinal cord injury (SCI) results in severe neurological deficit. However, the functional recovery following SCI is very poor due to the neural lost and limited axonal regeneration. To date, there was no effective treatment. Recent studies have shown that bone marrow stromal cells (BMSCs) transplantated into the central nervous system (CNS) can survive and differentiate into neuronal-like cells. Additionally, granulocyte colony-stimulating factor (G-CSF) can mobilize hematopoietic stem cells and inhibit neural cell apoptosis. Thus, we aimed to evaluate the combined effect of BMSC transplantation and G-CSF administration on rats with traverse spinal cord injury. METHODS: BMSCs were cultured in vitro, labeled with Hoechst33342, and then transplanted into the lesion site with or without G-CSF administration (50 microg/kg/day) for 5 subsequent days. The groups included an untreated control, along with treatment by G-CSF alone, BMSCs alone, and G-CSF + BMSCs. RESULTS: In this study, by the end of eighth week after SCI injury, the animals in group treated with G-CSF + BMSCs showed higher BBB scores than the other two groups. Morphometric assessment showed that the lesion areas in the rats of the G-CSF + BMSCs group were much smaller. Compared with the control, BMSC, and G-CSF groups, less expression of apoptosis cells and more neural-cell markers around the spinal cord injury were found in rats treated with G-CSF + BMSCs. CONCLUSIONS: The animals with the combination treatment achieved a better functional and morphologic recovery, although partial. This synergistic effect between BMSCs and G-CSF may be attributed to extrinsic and endogenous neurogenesis in the traverse spinal cord injury.


Assuntos
Transplante de Medula Óssea/métodos , Movimento Celular/efeitos dos fármacos , Sobrevivência de Enxerto/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/cirurgia , Células Estromais/efeitos dos fármacos , Células Estromais/transplante , Animais , Animais Recém-Nascidos , Benzimidazóis , Biomarcadores/análise , Biomarcadores/metabolismo , Técnicas de Cultura de Células , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Células Cultivadas , Modelos Animais de Doenças , Feminino , Corantes Fluorescentes , Sobrevivência de Enxerto/fisiologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Regeneração Nervosa/efeitos dos fármacos , Regeneração Nervosa/fisiologia , Proteínas do Tecido Nervoso/análise , Proteínas do Tecido Nervoso/metabolismo , Neurogênese/efeitos dos fármacos , Neurogênese/fisiologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/efeitos dos fármacos , Recuperação de Função Fisiológica/fisiologia , Traumatismos da Medula Espinal/fisiopatologia , Células Estromais/fisiologia , Resultado do Tratamento
18.
J Neurosci Methods ; 179(1): 45-50, 2009 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-19428510

RESUMO

Bone marrow stroma cells-derived neural stem cells (BMSCs-D-NSCs) transplantation is a promising strategy for the treatment of nervous system disorders. The development of a non-invasive method to follow the fate of BMSCs-D-NSCs in vivo is very important for the future application of this treatment. In this paper, we show for the first time, that BMSCs-D-NSCs from rhesus monkeys can be labeled in vitro with the superparamagnetic iron oxide (SPIO) contrast agent Feridex and Poly-L-lysine (PLL) without affecting morphology, cell cycle, telomerase activity, proliferation and differentiation ability of the labeled cells. Furthermore, when autografted into the striatum, these cells survived, differentiated and were incorporated into the brain, and could be reliably tracked using MRI, as confirmed by histological examination of the grafting sites with PKH(67) fluorescence. These results suggest that Feridex labeling of BMSCs-D-NSCs is feasible, efficient and safe for MRI tracing following autografting into the rhesus monkey nervous system.


Assuntos
Células da Medula Óssea/citologia , Encéfalo/citologia , Imageamento por Ressonância Magnética/métodos , Neurônios/fisiologia , Transplante de Células-Tronco , Células-Tronco/fisiologia , Animais , Apoptose , Células da Medula Óssea/ultraestrutura , Encéfalo/ultraestrutura , Ciclo Celular , Sobrevivência Celular , Dextranos , Óxido Ferroso-Férrico , Imuno-Histoquímica , Ferro , Macaca mulatta , Nanopartículas de Magnetita , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Neurônios/citologia , Neurônios/ultraestrutura , Óxidos , Células-Tronco/citologia , Células-Tronco/ultraestrutura , Células Estromais/fisiologia , Células Estromais/ultraestrutura , Telomerase/metabolismo , Transplante Autólogo
19.
Biotechnol Lett ; 31(2): 181-9, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18923908

RESUMO

Human mesenchymal stem cells-like cells (hMSCs-like cells) were used as a tumor treatment platform for the systemic delivery of immunotoxin genes. VEGF165-PE38 recombinant immunotoxin served as the model system. hMSCs-like cells were isolated, expanded, and electroporated with the pIRES2-VEGF165PE38-EGFP plasmid. RT-PCR and ELISA were used to confirm the expression of VEGF165-PE38 in the transfected hMSCs-like cells. These cells released 1390 +/- 137 pg VEGF165-PE38/10(4)cells over 48 h into the culture medium and the supernatant was capable of selectively killing human umbilical vein endothelial cells (HUVECs) and increasing apoptosis in these cells. In contrast, RPMI8226 was not inhibited by identical supernatants. Thus, these results lay the foundation for further studies on the potential role of hMSCs-like cells as a targeted therapeutic delivery vehicle for immunotoxins.


Assuntos
Células Endoteliais/citologia , Células Endoteliais/imunologia , Imunotoxinas/imunologia , Células-Tronco Mesenquimais/imunologia , Fator A de Crescimento do Endotélio Vascular/imunologia , Comunicação Celular/imunologia , Sobrevivência Celular , Células Cultivadas , Humanos , Imunotoxinas/administração & dosagem , Transfecção , Fator A de Crescimento do Endotélio Vascular/administração & dosagem , Fator A de Crescimento do Endotélio Vascular/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...