Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Cardiol Young ; 33(10): 1962-1966, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36424716

RESUMO

BACKGROUND: Endothelial dysfunction is a marked feature of Kawasaki disease during convalescence, but its pathogenesis is currently unclear. Circulating microRNAs (miRNAs) are associated with the progression of Kawasaki disease. However, the role and mechanism of circulating miRNAs in endothelial dysfunction are largely unknown. Kawasaki disease patients were found to have a unique circulating miRNA profile, including upregulation of miRNA-210-3p, miR-184 and miR-19a-3p, compared to non-Kawasaki disease febrile controls. This study aimed to investigate the effects of these three miRNAs on endothelial function. METHODS: Overexpression of miRNAs in human umbilical vein endothelial cells was done by transfection of miRNA mimics. The tube formation assay was used to evaluate the function of human umbilical vein endothelial cells. The potential binding sites of miRNAs on 3'untranslated regions were predicted by using TargetScan database and validated by dual luciferase reporter assay. The protein expression of AGO2, PTEN and VEGF in human umbilical vein endothelial cells was detected by Western blot. Overexpression of AGO2 in human umbilical vein endothelial cells was done by transfection of AGO2 expression plasmids. RESULTS: Overexpression of miRNA-184 and miRNA-19a-3p, but not miR-210-3p, impaired the function of human umbilical vein endothelial cells. Mechanistically, miR-184 and miR-19a-3p could target the 3'untranslated regions of AGO2 mRNA to downregulate its expression and subsequently impede the AGO2/PTEN/VEGF axis. To be noted, the rescue of the expression of AGO2 remarkably recovered the function that was impaired by overexpression of miRNA-184 and miRNA-19a-3p. CONCLUSIONS: This study suggested that miR-184 and miR-19a-3p could target AGO2/PTEN/VEGF axis to induce endothelial dysfunction in Kawasaki disease.


Assuntos
MicroRNAs , Síndrome de Linfonodos Mucocutâneos , Humanos , Células Endoteliais/metabolismo , MicroRNAs/genética , Síndrome de Linfonodos Mucocutâneos/genética , Regiões não Traduzidas , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/genética
2.
Clin Neurol Neurosurg ; 219: 107317, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35750021

RESUMO

OBJECTIVE: As a chronic complication of aneurysmal subarachnoid hemorrhage(aSAH), Shunt dependent hydrocephalus (SDHC) often leads to severe neurological deficits. At present, risk factors of SDHC after aSAH are being refined. So this study aims to investigate independent risk factors and develop a novel score to identify early the patients who require a permanent shunt. METHOD: Five hundred twenty-four patients treated in the first affiliated hospital of Harbin medical university from March 2019 to March 2021 were analyzed. We collected clinical and radiographic data of patients within 72 h after the ictus. The relevant factors were firstly analyzed by univariate analysis, and the significant factors (p < 0.05) were included in the multivariate logistic regression analysis to obtain the independent risk factors with statistical differences. The MAI score was established based on the contribution of different independent risk factors to the outcome. the new score was validated in another cohort (97 patients with aSAH from April and June 2021). RESULT: We enrolled 524 aneurysm patients and 41(7.82%) patients who underwent ventriculoperitoneal shunt (VPS) after aneurysm treatment. Based on univariate and multivariate analysis, Acute Hydrocephalus (OR 6.498,:95% confidence interval (CI) 1.98-21.33, p = 0.002), Intraventricular hemorrhage (OR 3.55,:95%CI 1.189-10.599, p = 0.023) and Modified Fisher score ≥ 3 (OR 5.846, 95%CI 2.649-12.900, p = 0.001) were independent risk factors. The novel score was assigned according to the contribution of different independent risk factors to the results. The MAI score: Modified Fisher grade ≥ 3 (1 point), Acute Hydrocephalus (1 point), Intraventricular hemorrhage (1 point). In the receiver operating characteristic curve analysis, the area under the curve (AUC) for the MAI score is 0.773 (p < 0.0001, 95%CI 0.686-0.861). Patients scoring 2-3 MAI points showed a 10-fold higher risk for shunt dependency than patients scoring 0-1 MAI points (p < 0.001). We performed internal validation of the MAI scoring system. The scoring system reliably predicted SDHC after aSAH. The AUC of the internal validation was 0.950 (p = 0.002, 95%CI 0.863-1.000). CONCLUSION: We develop a novel score based on univariate and multivariate analysis. The effectiveness of the MAI score has been confirmed in this study, which can more accurately predict SDHC after aASH and can be widely used in clinical practice. Prospective studies are needed for validation in the future.


Assuntos
Hidrocefalia , Hemorragia Subaracnóidea , Humanos , Hidrocefalia/diagnóstico por imagem , Hidrocefalia/etiologia , Hidrocefalia/cirurgia , Estudos Prospectivos , Estudos Retrospectivos , Fatores de Risco , Hemorragia Subaracnóidea/complicações , Hemorragia Subaracnóidea/diagnóstico por imagem , Hemorragia Subaracnóidea/cirurgia , Derivação Ventriculoperitoneal/efeitos adversos
3.
Mol Ther ; 30(7): 2537-2553, 2022 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-35570396

RESUMO

Bispecific T cell engagers (BiTEs) are bispecific antibodies that redirect T cells to target antigen-expressing tumors. We hypothesized that BiTE-secreting T cells could be a valuable therapy in solid tumors, with distinct properties in mono- or multi-valent strategies incorporating chimeric antigen receptor (CAR) T cells. Glioblastomas represent a good model for solid tumor heterogeneity, representing a significant therapeutic challenge. We detected expression of tumor-associated epidermal growth factor receptor (EGFR), EGFR variant III, and interleukin-13 receptor alpha 2 (IL13Rα2) on glioma tissues and cancer stem cells. These antigens formed the basis of a multivalent approach, using a conformation-specific tumor-related EGFR targeting antibody (806) and Hu08, an IL13Rα2-targeting antibody, as the single chain variable fragments to generate new BiTE molecules. Compared with CAR T cells, BiTE T cells demonstrated prominent activation, cytokine production, and cytotoxicity in response to target-positive gliomas. Superior response activity was also demonstrated in BiTE-secreting bivalent T cells compared with bivalent CAR T cells in a glioma mouse model at early phase, but not in the long term. In summary, BiTEs secreted by mono- or multi-valent T cells have potent anti-tumor activity in vitro and in vivo with significant sensitivity and specificity, demonstrating a promising strategy in solid tumor therapy.


Assuntos
Glioblastoma , Subunidade alfa2 de Receptor de Interleucina-13 , Animais , Linhagem Celular Tumoral , Receptores ErbB/genética , Receptores ErbB/metabolismo , Glioblastoma/patologia , Imunoterapia Adotiva , Camundongos , Linfócitos T , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Cancer Lett ; 454: 146-157, 2019 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-30978442

RESUMO

Metabolic abnormality is one of the hallmarks of cancer cells, and limiting material supply is a potential breakthrough approach for cancer treatment. Increasing researchers have been involved in the study of glioma cell metabolism reprogramming since the significance of IDH1 was confirmed in glioma. However, the molecular mechanisms underlying metabolic reprogramming induced by methionine deprivation regulates glioma cell proliferation remain unclear. Here we demonstrated that methionine deprivation inhibited glioma cell proliferation via downregulating interleukin 1 receptor antagonist (IL1RN) both in vitro and in vivo, methionine deprivation or knocking down IL1RN induced glioma cell cycle arrest. Moreover, we confirmed that IL1RN is a tumor associated gene and its expression is negatively correlated with the survival time of glioma patients. Altogether these results demonstrate a strong rationale insight that targeting amino acid metabolism such as methionine deprivation/IL1RN related gene therapy may offer novel direction for glioma treatment.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Proteína Antagonista do Receptor de Interleucina 1/biossíntese , Metionina/deficiência , Animais , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Regulação para Baixo , Glioma/patologia , Xenoenxertos , Humanos , Proteína Antagonista do Receptor de Interleucina 1/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transfecção
5.
J Cell Biochem ; 120(1): 622-633, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30596403

RESUMO

Glioma, the most predominant primary malignant brain tumor, remains uncured due to the absence of effective treatments. Hence, it is imperative to develop successful therapeutic agents. This study aimed to explore the antitumor effects and mechanisms of ivermectin (IVM) in glioma cells in vitro and in vivo. The effects of IVM on cell viability, cell cycle arrest, apoptosis rate, and morphological characteristics were determined respectively by MTT assay/colony formation assay, flow cytometry, and transmission electron microscope. In addition, the expression levels of cycle-related and apoptosis-associated proteins were individually examined by Western blot analysis. Moreover, cell proliferation and apoptosis analyses were carried out by TUNEL, Ki-67, cleaved caspase-3, and cleaved caspase-9 immunostaining assay. Our results demonstrated that IVM has a potential dosage-dependent inhibition effect on the apoptosis rate of glioma cells. Meanwhile, the results also revealed that IVM induced apoptosis by increasing caspase-3 and caspase-9 activity, upregulating the expressions of p53 and Bax, downregulating Bcl-2, activating cleaved caspase-3 and cleaved caspase-9, and blocking cell cycle in G0/G1 phase by downregulating levels of CDK2, CDK4, CDK6, cyclin D1, and cyclin E. These findings suggest that IVM has an inhibition effect on the proliferation of glioma cells by triggering cell cycle arrest and inducing cell apoptosis in vitro and in vivo, and probably represents promising agent for treating glioma.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/patologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Glioma/patologia , Ivermectina/farmacologia , Animais , Caspase 3/metabolismo , Caspase 9/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Feminino , Xenoenxertos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Ratos , Carga Tumoral/efeitos dos fármacos
6.
Oncol Rep ; 40(3): 1348-1358, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30015956

RESUMO

Moxidectin (MOX), a broad­spectrum antiparasitic agent, belongs to the milbemycin family and is similar to avermectins in terms of its chemical structure. Previous research has revealed that milbemycins, including MOX, may potentially function as effective multidrug resistance agents. In the present study, the impact of MOX on the viability of glioma cells was examined by MTT and colony formation assay, and the molecular mechanisms underlying MOX­mediated glioma cell apoptosis were explored by using flow cytometry and apoptosis rates. The results demonstrated that MOX exerts an inhibitory effect on glioma cell viability and colony formations in vitro and xenograft growth in vivo and is not active against normal cells. Additionally, as shown by western blot assay, it was demonstrated that MOX arrests the cell cycle at the G0/G1 phase by downregulating the expression levels of cyclin­dependent kinase (CDK)2, CDK4, CDK6, cyclin D1 and cyclin E. Furthermore, it was revealed that MOX is able to induce cell apoptosis by increasing the Bcl­2­associated X protein/B­cell lymphoma 2 ratio and activating the caspase­3/­9 cascade. In conclusion, these results suggest that MOX may inhibit the viability of glioma cells by inducing cell apoptosis and cell cycle arrest, and may be able to function as a potent and promising agent in the treatment of glioma.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/patologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Glioma/patologia , Macrolídeos/farmacologia , Fase de Repouso do Ciclo Celular/efeitos dos fármacos , Animais , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Proliferação de Células/efeitos dos fármacos , Feminino , Glioma/tratamento farmacológico , Glioma/metabolismo , Humanos , Inseticidas/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
7.
J Biomater Appl ; 31(3): 400-10, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27388895

RESUMO

PURPOSE: Achieving a radiographic gross total resection in high-grade gliomas improves overall survival. Many technologies such as intraoperative microscope, intraoperative ultrasound, fluorescence imaging, and intraoperative magnetic resonance imaging have been applied to improve tumor resection. However, most commercial available magnetic resonance imaging contrast agents have limited permeability across the blood-brain barrier and are cleared rapidly from circulation. Fluorescence imaging discriminates tumor from normal tissue and provides a promising new strategy to maximize sage surgical resection of tumor. However, the penetration depth of fluorescence imaging is generally low. MATERIALS AND METHODS: In this study, a new type of magnetite NaGdF4:Yb(3+),Er(3+),Li(+)@NaGdF4 (UCNPs) core-shell nanoparticles, coated with SiO2 and further functionalized with glioma and blood-brain barrier targeting motifs, was prepared for dual-modal in vivo upconversion imaging and magnetic resonance imaging. RESULTS: The as-prepared multifunctional upconversion nanoparticles (UCNPs@SiO2-CX-Lf) were biocompatible, showed strong upconversion luminescence under excitation of 980 nm, and provided high signal-to-noise ratio in vivo. Moreover, UCNPs@SiO2-CX-Lf nanoparticles showed a high relaxivity of 1.25 S(-1 )mM(-1) and were successfully applied as contrast agent for magnetic resonance imaging in tumor xenograft rat model with prolonged tumor signal enhancement. In vivo and magnetic resonance imaging Upconversion Luminescence (UCL) imaging results indicated that these particles can across the blood-brain barrier, bind to glioma, gave bright UCL signal and T1 magnetic resonance imaging contrast. CONCLUSIONS: Targeted UCL and magnetic resonance imaging dual-modal in vivo imaging using Yb(3+)/Er(3+)/Li(+) codoped NaGdF4 core-shell nanostructure can serve as a platform technology for the next generation of intraoperative probes for image-guided tumor resection.


Assuntos
Neoplasias Encefálicas/patologia , Óxido Ferroso-Férrico/química , Glioma/patologia , Medições Luminescentes/métodos , Imageamento por Ressonância Magnética/métodos , Imagem Multimodal/métodos , Animais , Neoplasias Encefálicas/diagnóstico por imagem , Linhagem Celular Tumoral , Meios de Contraste/síntese química , Glioma/diagnóstico por imagem , Humanos , Aumento da Imagem/métodos , Ratos , Ratos Wistar , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
8.
Tumour Biol ; 37(9): 12039-12047, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27177902

RESUMO

Glioblastoma multiform is one of the most common and most aggressive brain tumors in humans. The molecular and cellular mechanisms responsible for the onset and progression of GBM are elusive and controversial. The function of tumor suppressor candidate 3 (TUSC3) has not been previously characterized in GBM. TUSC3 was originally identified as part of an enzyme complex involved in N-glycosylation of proteins, but was recently implicated as a potential tumor suppressor gene in a variety of cancer types. In this study, we demonstrated that the expression levels of TUSC3 were downregulated in both GBM tissues and cells, and also found that overexpression of TUSC3 inhibits GBM cell proliferation and invasion. In addition, the effects of increased levels of methylation on the TUSC3 promoter were responsible for decreased expression of TUSC3 in GBM. Finally, we determined that TUSC3 regulates proliferation and invasion of GBM cells by inhibiting the activity of the Akt signaling pathway.


Assuntos
Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Proteínas de Membrana/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Neoplasias Encefálicas/etiologia , Linhagem Celular Tumoral , Proliferação de Células , Metilação de DNA , Glioblastoma/etiologia , Glicosilação , Humanos , Proteínas de Membrana/genética , Invasividade Neoplásica , Regiões Promotoras Genéticas , Proteínas Supressoras de Tumor/genética
9.
Cytotherapy ; 16(4): 523-34, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24424266

RESUMO

BACKGROUND AIMS: The molecular mechanisms by which stem cell transplantation improves functional recovery after intracerebral hemorrhage (ICH) are not well understood. Accumulating evidence suggests that microglia cells are activated shortly after ICH and that this activation contributes to secondary ICH-induced brain injury. We studied the effect of human amniotic epithelial stem cells (HAESCs) on microglia activation. METHODS: To study the effect of HAESCs in vitro, we used thrombin to activate the microglia cells. Twenty-four hours after thrombin treatment, the levels of tumor necrosis factor-α and interleukin-1ß were measured by enzyme-linked immunosorbent assay. In vivo, the HAESCs were transplanted into the rat striatum 1 day after collagenase-induced ICH. The expression levels of matrix metalloproteinase (MMP)-12 and microglia infiltration in the peri-hematoma tissues were determined 7 days after ICH through the use of reverse transcriptase-polymerase chain reaction and immunohistochemical analysis, respectively. RESULTS: Thrombin-activated microglia expression of tumor necrosis factor-α, interleukin-1ß and MMP-12 was significantly reduced through contact-dependent and paracrine mechanisms when the HAESCs were co-cultured with microglia cells. After transplantation of HAESCs in rat brains, the expression levels of MMP-12 and microglia infiltration in the peri-hematoma tissues were significantly reduced. CONCLUSIONS: Our observations suggest that microglia activation could be inhibited by HAESCs both in vitro and in vivo, which may be an important mechanism by which the transplantation of HAESCs reduces brain edema and ameliorates the neurologic deficits after ICH. Therefore, we hypothesize that methods for suppressing the activation of microglia and reducing the inflammatory response can be used for designing effective treatment strategies for ICH.


Assuntos
Hemorragia Cerebral/terapia , Interleucina-1beta/biossíntese , Metaloproteinase 12 da Matriz/biossíntese , Células-Tronco/citologia , Fator de Necrose Tumoral alfa/biossíntese , Líquido Amniótico/citologia , Animais , Hemorragia Cerebral/patologia , Técnicas de Cocultura , Modelos Animais de Doenças , Células Epiteliais/citologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Microglia/metabolismo , Microglia/transplante , Comunicação Parácrina , Ratos , Trombina/metabolismo
10.
Mol Cells ; 36(1): 17-24, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23807046

RESUMO

The reorganization of brain structures after intracerebral hemorrhage (ICH) insult is crucial to functional outcome. Although the pattern of neuronal rewiring is well-documented after ischemic stroke, the study of brain plasticity after ICH has been focusing on the enhancement of dendritic complexity. Here we hypothesized that functional restoration after ICH involves brain reorganization which may be favorably modulated by stem cell transplantation. In this study, bone marrow stromal cells (BMSCs) were transplanted into the perilesional sites of collagenase-induced ICH in adult rats one day after ICH injury. Forelimb functional recovery was monitored with modified limb placing and vibrissae-elicited forelimb placement tests. Anterograde and retrograde tracing were used to assess the reorganization of bilateral forelimb areas of the sensorimotor cortex. We found that in rats transplanted with BMSCs after ICH injury, axonal sprouting occurred in the contralateral caudal forelimb area of the cortex, and was significantly higher than in ICH rat models that received only the vehicle (P < 0.01). The number of positive neurons in the ipsilateral rostral forelimb area of the cortex of the BMSC group was 1.5-to 4.5-fold greater than in the vehicle group (P < 0.05). No difference was found between the BMSC and vehicle groups in hemispheric atrophy or labeled neurons in the ipsilateral caudal forelimb area (P = 0.193). Scores for improved functional behavior in the BMSC group were in accord with the results from histology. Neuronal plasticity of the denervated corticospinal tract at bilateral forelimb areas of the cortex in the collagenase-induced ICH rat models was significantly enhanced by BMSC transplantation. BMSC transplantation may facilitate functional recovery after ICH injury.


Assuntos
Células da Medula Óssea/citologia , Hemorragia Cerebral/fisiopatologia , Hemorragia Cerebral/terapia , Regeneração Nervosa , Tratos Piramidais/fisiopatologia , Recuperação de Função Fisiológica/fisiologia , Animais , Atrofia , Movimento Celular , Sobrevivência Celular , Cérebro/patologia , Cérebro/fisiopatologia , Colagenases , Modelos Animais de Doenças , Feminino , Membro Anterior/fisiopatologia , Masculino , Neurônios/metabolismo , Neurônios/patologia , Ratos , Ratos Wistar , Coloração e Rotulagem , Estilbamidinas/metabolismo , Células Estromais/transplante
11.
J Mol Neurosci ; 51(3): 933-40, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23430406

RESUMO

Emerging evidence has shown that microRNAs (miRNAs) participate in human carcinogenesis as tumor suppressors or oncogenes. It has been suggested that four common single nucleotide polymorphisms (SNPs; miR-146aG > C, 149C > T, 196a2C > T, and 499A > G) are associated with susceptibility to numerous malignancies. However, published results are inconsistent and inclusive. To further investigate the role of these loci, we examined the association of the miRNA polymorphisms with the risk of gliomas in a Han population in northeastern China. Both miR-146aG > C and 196a2C > T showed allelic differences between glioma patients and healthy controls in the studied population, with an OR of 1.30 (P = 0.0006) and an odds ratio (OR) of 1.25 (P = 0.003), respectively. Logistic regression analysis also revealed that the 146aG > C and 196a2C > T wild-type homozygous carriers had an increased glioma risk compared to the variant carriers. Besides, in pairwise comparisons two SNP combinations were associated with the risk of glioma. Among others, carriers of both homozygous risk genotypes, i.e., 146aGG and 196a2CC were associated with a nearly 4-fold increased risk of glioma (OR = 3.77, P = 1.3 × 10(-4)). Overall, glioma risk increased with increasing numbers of risk variant alleles. These results suggest that the miR-146aG > C and 196a2C > T might influence the risk of developing glioma in a northeastern Han Chinese population.


Assuntos
Neoplasias Encefálicas/genética , Predisposição Genética para Doença , Glioma/genética , MicroRNAs/genética , Polimorfismo de Nucleotídeo Único , Adulto , Idoso , Alelos , Estudos de Casos e Controles , China , Feminino , Homozigoto , Humanos , Masculino , Pessoa de Meia-Idade
12.
J Neuroimaging ; 23(2): 259-61, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22250982

RESUMO

Hyperpneumatization of the temporal bone with extension into the occipital bone and even the parietal bones is a rare condition. We report a case in which the patient suffered periodically from a palpable mass in the parietal-occipital region which originated from extensive occipital bone pneumatization. Computed tomography examination revealed extensive temporal and occipital pneumatization and subperiosteal pneumatoceles, which was corrected by surgery.


Assuntos
Doenças Ósseas/diagnóstico por imagem , Enfisema/diagnóstico por imagem , Osso Occipital/diagnóstico por imagem , Pneumocefalia/diagnóstico por imagem , Osso Temporal/diagnóstico por imagem , Adulto , Doenças Ósseas/complicações , Doenças Ósseas/cirurgia , Diagnóstico Diferencial , Enfisema/etiologia , Enfisema/cirurgia , Humanos , Masculino , Osso Occipital/cirurgia , Periósteo/diagnóstico por imagem , Periósteo/cirurgia , Pneumocefalia/complicações , Pneumocefalia/cirurgia , Radiografia , Osso Temporal/cirurgia , Resultado do Tratamento
13.
PLoS One ; 7(8): e42813, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22952613

RESUMO

OBJECTIVE: We aimed to investigate whether an innovative growth factor-laden scaffold composed of acellular sciatic nerve (ASN) and brain-derived neurotrophic factor (BDNF) could promote axonal regeneration and functional recovery after spinal cord injury (SCI). METHODS: Following complete transection at the thoracic level (T9), we immediately transplanted the grafts between the stumps of the severed spinal cords. We evaluated the functional recovery of the hindlimbs of the operated rats using the BBB locomotor rating scale system every week. Eight weeks after surgery, axonal regeneration was examined using the fluorogold (FG) retrograde tracing method. Electrophysiological analysis was carried out to evaluate the improvement in the neuronal circuits. Immunohistochemistry was employed to identify local injuries and recovery. RESULTS: The results of the Basso-Beattie-Bresnahan (BBB) scale indicated that there was no significant difference between the individual groups. The FG retrograde tracing and electrophysiological analyses indicated that the transplantation of ASN-BDNF provided a permissive environment to support neuron regeneration. CONCLUSION: The ASN-BDNF transplantation provided a promising therapeutic approach to promote axonal regeneration and recovery after SCI, and can be used as part of a combinatory treatment strategy for SCI management.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Nervo Isquiático/fisiopatologia , Traumatismos da Medula Espinal/terapia , Animais , Axônios/patologia , Membrana Basal/patologia , Barreira Hematoencefálica , Eletrofisiologia/métodos , Feminino , Microscopia Eletrônica de Varredura/métodos , Microscopia Eletrônica de Transmissão/métodos , Regeneração Nervosa , Neurônios/metabolismo , Ratos , Nervo Isquiático/ultraestrutura , Medula Espinal/patologia , Transplante Homólogo
14.
J Biomed Mater Res B Appl Biomater ; 100(8): 2131-40, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22848005

RESUMO

A dural tear is a common complication after acute laceration spinal cord injury (ALSCI). An unrepaired dural tear is associated with poor locomotor functional recovery. Spinal duraplasty with biomaterials may promote functional recovery in ALSCI. However, an ideal dural substitute has not yet been found. In this work, we investigated the possibility of using a denuded human amniotic membrane (DHAM) or DHAM seeded on bone marrow stromal cells (DHAM-BMSCs) as duraplasty biomaterials. We patched broken dura with the two novel substitutes in an ALSCI rat model. At the end of the eighth week, we observed that the neural motor function was recovered according to the Basso-Beattie-Bresnahan scale, and the neural loop was successfully reestablished between the ends of the lesions by motor-evoked potentials in the duraplasty groups. Moreover, the DHAM-BMSCs repaired the dura and resulted in a significant reduction in the total lesion and cystic volumes by nearly 10-fold versus the control group (p < 0.01). The levels of neurotrophic factors and NF-200-positive fibers were also improved in the duraplasty groups, compared to the control group. Our data suggest that the two novel substitutes may be promising grafts for patching dural defects to improve locomotor function after ALSCI.


Assuntos
Âmnio , Bioprótese , Células da Medula Óssea , Locomoção , Traumatismos da Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/cirurgia , Doença Aguda , Animais , Potencial Evocado Motor , Feminino , Humanos , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica , Células Estromais
15.
Eur J Pharmacol ; 659(2-3): 108-13, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-21458446

RESUMO

Development of agents to overcome multidrug resistance (MDR) is important in cancer chemotherapy, and the overexpression of P-glycoprotein (P-gp) is one of the major mechanisms of MDR. In this paper, we evaluated the effects of two new milbemycin compounds, milbemycin ß(14) and secomilbemycin D, isolated from fermentation broth of S. bingchenggensis on reversing MDR of adriamycin-resistant human breast carcinoma (MCF-7/adr) cells. We observed that the both milbemycins (5 µM) showed strong potency to increase adriamycin cytotoxicity toward MCF-7/adr cells with reversal fold (RF) of 13.5 and 10.59, respectively. In addition, the mechanisms of milbemycins on reversing P-gp-mediated MDR demonstrated that they significantly increased the accumulations of adriamycin and Rh123 via inhibiting P-gp efflux in MCF-7/adr cells. Furthermore, the results also revealed that milbemycin ß(14) and secomilbemycin D could regulate down the expression of P-gp, but not affect the expression of MDR1 gene. In conclusion, our observations suggest that the two new milbemycin compounds probably represent the promising agents for reversing MDR in cancer therapy.


Assuntos
Doxorrubicina/farmacologia , Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Linhagem Celular Tumoral , Resistência a Múltiplos Medicamentos/genética , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Macrolídeos/química , Macrolídeos/farmacologia , Rodamina 123/metabolismo
16.
Zhonghua Yi Xue Za Zhi ; 84(16): 1386-9, 2004 Aug 17.
Artigo em Chinês | MEDLINE | ID: mdl-15387949

RESUMO

OBJECTIVE: To evaluate the feasibility of monitoring the neural stem cells implanted into the brain by the technique of labeling with superparamagnetic iron oxide (SPIO). METHODS: Neural stem cells were isolated from the cerebral cortex of newborn Wister rats and cultured. SPIO particles and poly-L-lysine were added into the medium to be co-cultured foe one hour. After the formation of neurospheres, Prussian blue staining was conducted and transmission electron microscopy was used to identify the iron particles in these neural stem cells. Sixteen adult female Wistar rats underwent transplantation of labeled neural stem cells into the right side of brain and non-labeled cells were transplanted into the contralateral part as controls. 1, 2, 4, 6, and 7 weeks after the transplantation, MRI examination with the scanning sequences of SE T2WI, FSE T2WI, and GRE T2 * respectively was conducted on the brains of the rats. Four rats at each time point were killed and their brains were taken out to undergo HE staining and Prussian blue staining to track the presence of labeled-cells. RESULTS: After the addition of SPIO the neurospheres continued to proliferate and differentiate normally. Electron microscopy showed vacuolar structures of different sizes under the cytoplasma membrane within and outside which there were high-density iron particles. Prussian blue staining showed numerous blue stained particles in the cytoplasm of the labeled cells. Remarkable low signal change was seen in the right brain transplanted with labeled cells, especially in the condition of scanning sequence of GRET2. Such change could be seen up to 7 weeks after the transplantation. No signal change was found in the left brain. CONCLUSION: SPIO labeling technique is useful in monitoring the outcome of transplanted neural stem cells.


Assuntos
Ferro/farmacocinética , Imageamento por Ressonância Magnética , Nanotecnologia , Óxidos/farmacocinética , Células-Tronco/citologia , Animais , Animais Recém-Nascidos , Diferenciação Celular , Células Cultivadas , Feminino , Óxido Ferroso-Férrico , Indicadores e Reagentes/farmacocinética , Lipídeos , Neurônios/citologia , Neurônios/metabolismo , Ratos , Ratos Wistar , Transplante de Células-Tronco , Células-Tronco/metabolismo , Transfecção/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA