Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Anat Cell Biol ; 57(1): 97-104, 2024 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-38192123

RESUMO

Heavy reliance on glucose metabolism and a reduced capacity to use ketone bodies makes glioblastoma (GBM) a promising candidate for ketone-based therapies. Ketogenic diet (KD) is well-known for its promising effects in controlling tumor growth in GBM. Moreover, synthetic ketone ester (KE) has demonstrated to increase blood ketone levels and enhance animal survival in a metastatic VM-M3 murine tumor model. Here, we compared the efficacy of a KE-supplemented Atkins-type diet (ATD-KE) to a classic KD in controlling tumor progression and enhancing survival in a clinically relevant orthotopic patient-derived xenograft GBM model. Our findings demonstrate that ATD-KE preserves body weight (percent change from the baseline; 112±2.99 vs. 116.9±2.52 and 104.8±3.67), decreases blood glucose (80.55±0.86 vs. 118.6±9.51 and 52.35±3.89 mg/dl), and increases ketone bodies in blood (1.15±0.03 mM vs. 0.55±0.04 and 2.66±0.21 mM) and brain tumor tissue (3.35±0.30 mM vs. 2.04±0.3 and 4.25±0.25 mM) comparable to the KD (results presented for ATD-KE vs. standard diet [STD] and KD, respectively). Importantly, the ATD-KE treatment significantly enhanced survival compared to the STD and was indistinguishable from the KD (47 days in STD vs. 56 days in KD and ATD-KE), suggesting that a nutritionally balanced low carbohydrate ATD combined with KE may be as effective as the KD alone in reducing brain tumor progression. Overall, these data support the rationale for clinical testing of KE-supplemented low-carb diet as an adjunct treatment for brain tumor patients.

2.
Methods Mol Biol ; 2389: 87-94, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34558004

RESUMO

Neural stem cells (NSCs) transplantation enhances plasticity and restores functions in neurological diseases. Therapeutic benefits of NSCs are due to their ability to replace the lost neurons and glial cells and also secreting a wide array of free and membrane-bound bioactive molecules that can reduce the hostility of diseased microenvironment, resolve inflammation, and rescue damaged neural cells. Membrane-encircled spherical nanostructures that are collectively known as extracellular vesicles (EVs) contain mRNA, miRNA, lipids, and specific proteins that affect different biological processes in cells located nearby or at far distances. Using EVs as an alternative non-cell-based therapy has gained huge attention, and developing methods for large-scale production of EVs is of great clinical importance. Here, we describe an efficient method to yield significant quantity of EVs from human NSCs that are expanded under free floating neurosphere assay culture system. Using the neurosphere assay in bioreactors under GMP-compliant conditions can result in scalable NSC-EVs required for human trials.


Assuntos
Vesículas Extracelulares , Células-Tronco Neurais , Transporte Biológico , Separação Celular , Células Cultivadas , Vesículas Extracelulares/metabolismo , Humanos , MicroRNAs/metabolismo , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo
3.
Methods Mol Biol ; 2389: 111-123, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34558007

RESUMO

The renewable source of neural stem cells (NSCs) with multi-lineage differentiation capability toward neurons, astrocytes, and oligodendrocytes represents an ideal supply for cell therapy of central nervous system (CNS) diseases. In spite of this, the clinical use of NSCs is hampered by heterogeneity, poor neuronal cell yield, predominant astrocytic differentiation of NSC progeny, and possible uncontrolled proliferation and tumor formation upon transplantation. The ability to generate highly enriched and defined neural cell populations from the renewable source of NSCs might overcome many of these impediments and pave the way toward their successful clinical applications.Here, we describe a simple method for NSC differentiation and subsequent purification of neuronal progenitor cells, taking advantage of size and granularity differences between neuronal cells and other NSC progeny. This highly enriched neuronal cell population provides an invaluable source of cells for both in vitro and in vivo studies.


Assuntos
Células-Tronco Neurais , Diferenciação Celular , Separação Celular , Células Cultivadas , Neurogênese , Neurônios , Oligodendroglia
4.
Eur J Neurosci ; 54(4): 5620-5637, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34251711

RESUMO

Spinal cord injury (SCI) is a devastating clinical problem that can lead to permanent motor dysfunction. Fingolimod (FTY720) is a sphingosine structural analogue, and recently, its therapeutic benefits in SCI have been reported. The present study aimed to evaluate the therapeutic efficacy of fingolimod-incorporated poly lactic-co-glycolic acid (PLGA) nanoparticles (nanofingolimod) delivered locally together with neural stem/progenitor cells (NS/PCs) transplantation in a mouse model of contusive acute SCI. Fingolimod was encapsulated in PLGA nanoparticles by the emulsion-evaporation method. Mouse NS/PCs were harvested and cultured from embryonic Day 14 (E14) ganglionic eminences. Induction of SCI was followed by the intrathecal delivery of nanofingolimod with and without intralesional transplantation of PuraMatrix-encapsulated NS/PCs. Functional recovery, injury size and the fate of the transplanted cells were evaluated after 28 days. The nanofingolimod particles represented spherical morphology. The entrapment efficiency determined by UV-visible spectroscopy was approximately 90%, and the drug content of fingolimod loaded nanoparticles was 13%. About 68% of encapsulated fingolimod was slowly released within 10 days. Local delivery of nanofingolimod in combination with NS/PCs transplantation led to a stronger improvement in neurological functions and minimized tissue damage. Furthermore, co-administration of nanofingolimod and NS/PCs not only increased the survival of transplanted cells but also promoted their fate towards more oligodendrocytic phenotype. Our data suggest that local release of nanofingolimod in combination with three-dimensional (3D) transplantation of NS/PCs in the acute phase of SCI could be a promising approach to restore the damaged tissues and improve neurological functions.


Assuntos
Nanopartículas , Células-Tronco Neurais , Traumatismos da Medula Espinal , Animais , Diferenciação Celular , Cloridrato de Fingolimode , Glicóis , Camundongos , Células-Tronco Neurais/transplante , Peptídeos , Recuperação de Função Fisiológica , Traumatismos da Medula Espinal/tratamento farmacológico
5.
J Immunother Cancer ; 7(1): 321, 2019 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-31753028

RESUMO

BACKGROUND: Dendritic cell (DC) vaccine efficacy is directly related to the efficiency of DC migration to the lymph node after delivery to the patient. We discovered that a naturally occurring metabolite, sarcosine, increases DC migration in human and murine cells resulting in significantly improved anti-tumor efficacy. We hypothesized that sarcosine induced cell migration was due to chemokine signaling. METHODS: DCs were harvested from the bone marrow of wild type C57BL/6 mice and electroporated with tumor messenger RNA (mRNA). Human DCs were isolated from peripheral blood mononuclear cells (PBMCs). DCs were treated with 20 mM of sarcosine. Antigen specific T cells were isolated from transgenic mice and injected intravenously into tumor bearing mice. DC vaccines were delivered via intradermal injection. In vivo migration was evaluated by flow cytometry and immunofluorescence microscopy. Gene expression in RNA was investigated in DCs via RT-PCR and Nanostring. RESULTS: Sarcosine significantly increased human and murine DC migration in vitro. In vivo sarcosine-treated DCs had significantly increased migration to both the lymph nodes and spleens after intradermal delivery in mice. Sarcosine-treated DC vaccines resulted in significantly improved tumor control in a B16F10-OVA tumor flank model and improved survival in an intracranial GL261-gp100 glioma model. Gene expression demonstrated an upregulation of CXCR2, CXCL3 and CXCL1 in sarcosine- treated DCs. Further metabolic analysis demonstrated the up-regulation of cyclooxygenase-1 and Pik3cg. Sarcosine induced migration was abrogated by adding the CXCR2 neutralizing antibody in both human and murine DCs. CXCR2 neutralizing antibody also removed the survival benefit of sarcosine-treated DCs in the tumor models. CONCLUSION: Sarcosine increases the migration of murine and human DCs via the CXC chemokine pathway. This platform can be utilized to improve existing DC vaccine strategies.


Assuntos
Vacinas Anticâncer/imunologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/imunologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/fisiologia , Receptores CXCR/metabolismo , Sarcosina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transferência Adotiva , Animais , Biomarcadores , Modelos Animais de Doenças , Humanos , Imunoterapia , Camundongos , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/patologia , Neoplasias/terapia , Estresse Oxidativo , Receptores CXCR/genética
6.
BMC Complement Altern Med ; 19(1): 113, 2019 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-31159797

RESUMO

BACKGROUND: Embryonic neural stem cells (eNSCs) are immature precursors of the central nervous system (CNS), with self-renewal and multipotential differentiation capacities. These are regulated by endogenous and exogenous factors such as alpha-linolenic acid (ALA), a plant-based essential omega-3 polyunsaturated fatty acid. METHODS: In this study, we investigated the effects of various concentrations of Alyssum homolocarpum seed oil (AHSO), containing natural ALA, stearic acid (SA), myristic acid (MA), and ß-sitosterol, on proliferation and differentiation of eNSCs, in comparison to controls and to synthetic pure ALA. RESULTS: Treatment with natural AHSO (25 to 75 µM), similar to synthetic ALA, caused a significant ~ 2-fold increase in eNCSs viability, in comparison to controls. To confirm this proliferative activity, treatment of NSCs with 50 or 75 µM AHSO resulted in a significant increase in mRNA levels of notch1, hes-1 and Ki-67and NICD protein expression, in comparison to controls. Moreover, AHSO administration significantly increased the differentiation of eNSCs toward astrocytes (GFAP+) and oligodendrocytes (MBP+) in a dose dependent manner and was more potent than ALA, at similar concentrations, in comparison to controls. Indeed, only high concentrations of 100 µM AHSO, but not ALA, caused a significant increase in the frequency of neurons (ß-III Tubulin+). CONCLUSION: Our data demonstrated that AHSO, a rich source of ALA containing also other beneficial fatty acids, increased the proliferation and stimulated the differentiation of eNSCs. We suggest that AHSO's effects are caused by ß-sitosterol, SA and MA, present within this oil. AHSO could be used in diet to prevent neurodevelopmental syndromes, cognitive decline during aging, and various psychiatric disorders.


Assuntos
Brassicaceae/química , Células-Tronco Neurais/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Óleos de Plantas/farmacologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Antígeno Ki-67/metabolismo , Camundongos , Ácido Mirístico/análise , Células-Tronco Neurais/metabolismo , Óleos de Plantas/química , Sementes/química , Sitosteroides/análise , Ácidos Esteáricos/análise , Ácido alfa-Linolênico/análise
7.
Neuro Oncol ; 21(6): 730-741, 2019 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-30668768

RESUMO

BACKGROUND: The changes induced in host immunity and the tumor microenvironment by chemotherapy have been shown to impact immunotherapy response in both a positive and a negative fashion. Temozolomide is the most common chemotherapy used to treat glioblastoma (GBM) and has been shown to have variable effects on immune response to immunotherapy. Therefore, we aimed to determine the immune modulatory effects of temozolomide that would impact response to immune checkpoint inhibition in the treatment of experimental GBM. METHODS: Immune function and antitumor efficacy of immune checkpoint inhibition were tested after treatment with metronomic dose (MD) temozolomide (25 mg/kg × 10 days) or standard dose (SD) temozolomide (50 mg/kg × 5 days) in the GL261 and KR158 murine glioma models. RESULTS: SD temozolomide treatment resulted in an upregulation of markers of T-cell exhaustion such as LAG-3 and TIM-3 in lymphocytes which was not seen with MD temozolomide. When temozolomide treatment was combined with programmed cell death 1 (PD-1) antibody therapy, the MD temozolomide/PD-1 antibody group demonstrated a decrease in exhaustion markers in tumor infiltrating lymphocytes that was not observed in the SD temozolomide/PD-1 antibody group. Also, the survival advantage of PD-1 antibody therapy in a murine syngeneic intracranial glioma model was abrogated by adding SD temozolomide to treatment. However, when MD temozolomide was added to PD-1 inhibition, it preserved the survival benefit that was seen by PD-1 antibody therapy alone. CONCLUSION: The peripheral and intratumoral immune microenvironments are distinctively affected by dose modulation of temozolomide.


Assuntos
Anticorpos Monoclonais/farmacologia , Neoplasias Encefálicas/imunologia , Glioblastoma/imunologia , Linfócitos do Interstício Tumoral/imunologia , Receptor de Morte Celular Programada 1/imunologia , Linfócitos T/imunologia , Temozolomida/farmacologia , Animais , Antineoplásicos Alquilantes/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Relação Dose-Resposta a Droga , Quimioterapia Combinada , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Humanos , Imunoterapia , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Linfócitos T/efeitos dos fármacos , Linfócitos T/patologia , Células Tumorais Cultivadas , Microambiente Tumoral
8.
Methods Mol Biol ; 1686: 59-67, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29030812

RESUMO

Tumor resistance to conventional therapies is a major challenge toward the eradication of cancer, a life-threatening disease. This resistance mainly results from tumor heterogeneity and more specifically from the existence of "stem-like" cells that remain in a quiescent state for long periods of time and thus escape commonly used anti-cancer drugs resulting in treatment failure. Therefore, targeting this subpopulation would present a viable strategy to overcome tumor burden. This daunting task requires a deep and thorough understanding of the biology of the quiescent stem-cell population, their interaction with tumor microenvironments, and mechanisms used to sustain themselves despite aggressive therapies. In this chapter, we describe detailed technical procedures for the isolation of quiescent or infrequently dividing stem-like cells in cultured glioblastoma tumor cells using carboxy fluorescein succinimidyl ester (CFSE) staining and flow cytometric analysis. Quiescent glioblastoma cells with stem-like features are characterized and subsequently isolated based on their ability to retain the CFSE labeling.


Assuntos
Citometria de Fluxo/métodos , Fluoresceínas/química , Corantes Fluorescentes/química , Glioblastoma/diagnóstico , Células-Tronco Neoplásicas/patologia , Fase de Repouso do Ciclo Celular , Coloração e Rotulagem/métodos , Succinimidas/química , Divisão Celular , Humanos , Células Tumorais Cultivadas
9.
Front Neurosci ; 11: 446, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28824367

RESUMO

Embryonic neural stem cells (eNSCs) could differentiate into neurons, astrocytes and oligodendrocytes. This study was aimed to determine the effect of safflower seed oil, which contains linoleic acid (LA), oleic acid (OA), and palmitic acid (PA), on cultured eNSC proliferation and differentiation, in comparison to linoleic acid alone. Results showed that safflower seed oil, but not LA, increased significantly the viability and proliferation of eNSCs. Moreover, treatment of NSCs by safflower seed oil, but not LA, resulted in a significant increase in mRNA levels of notch1, hes1, and Ki-67, and protein levels of notch intracellular domain (NICD), in comparison to controls, indicating an enhancement of stemness. Finally, safflower seed oil, but not LA, caused an increase in the number of oligodendrocytes (MBP+), astrocytes (GFAP+) and neurons (ß-III tubulin+) of which only the increase in ß-III tubulin positive cells was statistically significant. In summary, OA and PA, present in safflower seed oil may prove beneficial for the enhancement of eNSCs and their neuronal differentiation.

10.
Methods Mol Biol ; 1462: 689-710, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27604746

RESUMO

Traumatic brain injury (TBI) is one of the leading causes of death and disabilities worldwide. It affects approximately 1.5 million people each year and is associated with severe post-TBI symptoms such as sensory and motor deficits. Several neuro-therapeutic approaches ranging from cell therapy interventions such as the use of neural stem cells (NSCs) to drug-based therapies have been proposed for TBI management. Successful cell-based therapies are tightly dependent on reproducible preclinical animal models to ensure safety and optimal therapeutic benefits. In this chapter, we describe the isolation of NSCs from neonatal mouse brain using the neurosphere assay in culture. Subsequently, dissociated neurosphere-derived cells are used for transplantation into the ipsilateral cortex of a controlled cortical impact (CCI) TBI model in C57BL/6 mice. Following intra-cardiac perfusion and brain removal, the success of NSC transplantation is then evaluated using immunofluorescence in order to assess neurogenesis along with gliosis in the ipsilateral coronal brain sections. Behavioral tests including rotarod and pole climbing are conducted to evaluate the motor activity post-treatment intervention.


Assuntos
Lesões Encefálicas Traumáticas/etiologia , Lesões Encefálicas Traumáticas/terapia , Células-Tronco Neurais/citologia , Transplante de Células-Tronco , Animais , Comportamento Animal , Biomarcadores , Lesões Encefálicas Traumáticas/diagnóstico , Lesões Encefálicas Traumáticas/fisiopatologia , Técnicas de Cultura de Células , Sobrevivência Celular , Células Cultivadas , Modelos Animais de Doenças , Feminino , Imunofluorescência , Camundongos , Células-Tronco Neurais/metabolismo , Recuperação de Função Fisiológica , Teste de Desempenho do Rota-Rod , Resultado do Tratamento
11.
Methods Mol Biol ; 1462: 711-31, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27604747

RESUMO

Despite all attempts the problem of regeneration in damaged central nervous system (CNS) has remained challenging due to its cellular complexity and highly organized and sophisticated connections. In this regard, stem cell therapy might serve as a viable therapeutic approach aiming either to support the damaged tissue and hence to reduce the subsequent neurological dysfunctions and impairments or to replace the lost cells and re-establish damaged circuitries. Adult neural stem/progenitor cells (NS/PCs) are one of the outstanding cell sources that can be isolated from the subventricular zone (SVZ) of the lateral ventricles. These cells can differentiate into neurons, astrocytes, and oligodendrocytes. Implanting autologous NS/PCs will greatly benefit the patients by avoiding immune rejection after implantation, better survival, and integration with the host tissue. Developing safe and efficient methods in small animal models will provide us with the opportunity to optimize procedures required to achieve successful human autologous NS/PC transplantation in near future. In this chapter, a highly controlled and safe biopsy method for harvesting stem cell containing tissue from the SVZ of adult rat brain is introduced. Then, isolation and expansion of NS/PCs from harvested specimen as well as the techniques to verify proliferation and differentiation capacity of the resulting NS/PCs are discussed. Finally, a method for assessing the biopsy lesion volume in the brain is described. This safe biopsy method in rat provides a unique tool to study autologous NS/PC transplantation in different CNS injury models.


Assuntos
Separação Celular , Ventrículos Laterais/citologia , Células-Tronco Neurais/citologia , Transplante de Células-Tronco , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Biópsia , Lesões Encefálicas/etiologia , Lesões Encefálicas/patologia , Lesões Encefálicas/terapia , Técnicas de Cultura de Células , Diferenciação Celular , Separação Celular/métodos , Células Cultivadas , Modelos Animais de Doenças , Ventrículos Laterais/metabolismo , Masculino , Células-Tronco Neurais/metabolismo , Oligodendroglia/citologia , Oligodendroglia/metabolismo , Ratos , Traumatismos da Medula Espinal/etiologia , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/terapia , Transplante de Células-Tronco/métodos , Transplante Autólogo
12.
Brain Res ; 1642: 197-208, 2016 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-27038753

RESUMO

Cultivation of neural stem/progenitor cells (NS/PCs) in PuraMatrix (PM) hydrogel is an option for stem cell transplantation. The efficacy of a novel method for placing adult rat NS/PCs in PM (injection method) was compared to encapsulation and surface plating approaches. In addition, the efficacy of injection method for transplantation of autologous NS/PCs was studied in a rat model of brain injury. NS/PCs were obtained from the subventricular zone (SVZ) and cultivated without (control) or with scaffold (three-dimensional cultures; 3D). The effect of different approaches on survival, proliferation, and differentiation of NS/PCs were investigated. In in vivo study, brain injury was induced 45 days after NS/PCs were harvested from the SVZ and phosphate buffered saline, PM, NS/PCs, or PM+NS/PCs were injected into the brain lesion. There was an increase in cell viability and proliferation after injection and surface plating of NS/PCs compared to encapsulation and neural differentiation markers were expressed seven days after culturing the cells. Using injection method, transplantation of NS/PCs cultured in PM resulted in significant reduction of lesion volume, improvement of neurological deficits, and enhancement of surviving cells. In addition, the transplanted cells could differentiate in to neurons, astrocytes, or oligodendrocytes. Our results indicate that the injection and surface plating methods enhanced cell survival and proliferation of NS/PCs and suggest the injection method as a promising approach for transplantation of NS/PCs in brain injury.


Assuntos
Lesões Encefálicas/terapia , Células-Tronco Neurais/fisiologia , Células-Tronco Neurais/transplante , Cultura Primária de Células/métodos , Transplante de Células-Tronco/métodos , Animais , Diferenciação Celular , Proliferação de Células , Sobrevivência Celular , Hidrogel de Polietilenoglicol-Dimetacrilato , Ventrículos Laterais/citologia , Masculino , Córtex Motor/lesões , Células-Tronco Neurais/ultraestrutura , Ratos , Ratos Wistar
13.
Sci Rep ; 6: 23579, 2016 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-27030542

RESUMO

Many neurological injuries are likely too extensive for the limited repair capacity of endogenous neural stem cells (NSCs). An alternative is to isolate NSCs from a donor, and expand them in vitro as transplantation material. Numerous groups have already transplanted neural stem and precursor cells. A caveat to this approach is the undefined phenotypic distribution of the donor cells, which has three principle drawbacks: (1) Stem-like cells retain the capacity to proliferate in vivo. (2) There is little control over the cells' terminal differentiation, e.g., a graft intended to replace neurons might choose a predominantly glial fate. (3) There is limited ability of researchers to alter the combination of cell types in pursuit of a precise treatment. We demonstrate a procedure for differentiating human neural precursor cells (hNPCs) in vitro, followed by isolation of the neuronal progeny. We transplanted undifferentiated hNPCs or a defined concentration of hNPC-derived neurons into mice, then compared these two groups with regard to their survival, proliferation and phenotypic fate. We present evidence suggesting that in vitro-differentiated-and-purified neurons survive as well in vivo as their undifferentiated progenitors, and undergo less proliferation and less astrocytic differentiation. We also describe techniques for optimizing low-temperature cell preservation and portability.


Assuntos
Criopreservação/métodos , Células-Tronco Neurais/citologia , Neurogênese/fisiologia , Neurônios/citologia , Neurônios/transplante , Transplante de Células-Tronco , Animais , Diferenciação Celular , Proliferação de Células , Sobrevivência Celular , Rastreamento de Células/métodos , Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Separação Imunomagnética/métodos , Injeções Intraventriculares , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Células-Tronco Neurais/fisiologia , Neuroglia/citologia , Neuroglia/fisiologia , Neurônios/fisiologia , Fenótipo , Técnicas Estereotáxicas , Transplante Heterólogo
14.
J Nat Med ; 69(3): 387-96, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25860174

RESUMO

Pharmacognostic evaluation of medicinal plants may assess their current applications and possibly results in finding new active components. In this study, ash and extractive values and high performance thin layer chromatography fingerprints of Alyssum homolocarpum (Brassicaceae) seed extracts were investigated to elucidate its composition. Differential scanning calorimetry and gas chromatography-mass spectrometry analysis were employed to determine the components of A. homolocarpum seed oil (AHO). Neurosphere assay, in vitro differentiation and immunofluorescence analysis were performed to evaluate the effects of oral administration of AHO (0.5 or 1 g/kg/day for 14 days) on proliferation and differentiation of neural stem cells (NSCs) in adult male BALB/c mice. Total, acid-insoluble and water-soluble ash values were determined as 45.83 ± 5.85, 6.67 ± 2.89 and 28.33 ± 2.89 mg/g, respectively. The extractive values were 4.90, 0.43 and 0.56 % (w/w) for n-hexane, dichloromethane and ethanolic extracts, respectively. Interestingly, AHO was mainly composed of α-linolenic acid (89.71 %), ß-sitosterol (3.3 mg/g) and campesterol (0.86 mg/g). Administration of AHO at 1 g/kg/day significantly increased proliferation of NSCs, as evidenced by an increase in mean neurosphere-forming frequency per brain (872.7 ± 15.17) and neurosphere diameter (101 ± 2.48 µm) compared to the control group (424.3 ± 59.29 and 78.63 ± 1.7 µm, respectively; P < 0.05). AHO treatment did not affect in vitro differentiation of the harvested NSCs. Our data show that A. homolocarpum seed oil is a rich source of α-linolenic acid and ß-sitosterol with potential therapeutic application to enhance NSC proliferation and recruitment in neurological diseases.


Assuntos
Brassicaceae/química , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células-Tronco Neurais/fisiologia , Extratos Vegetais/farmacologia , Óleos de Plantas/farmacologia , Animais , Células Cultivadas , Colesterol/análogos & derivados , Colesterol/química , Colesterol/isolamento & purificação , Colesterol/farmacologia , Avaliação Pré-Clínica de Medicamentos , Cromatografia Gasosa-Espectrometria de Massas , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Células-Tronco Neurais/efeitos dos fármacos , Fitosteróis/química , Fitosteróis/isolamento & purificação , Fitosteróis/farmacologia , Extratos Vegetais/química , Extratos Vegetais/isolamento & purificação , Óleos de Plantas/química , Óleos de Plantas/isolamento & purificação , Sementes/química , Sitosteroides/química , Sitosteroides/isolamento & purificação , Sitosteroides/farmacologia
15.
Anat Cell Biol ; 48(1): 25-35, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25806119

RESUMO

Certain limitations of the neurosphere assay (NSA) have resulted in a search for alternative culture techniques for brain tumor-initiating cells (TICs). Recently, reports have described growing glioblastoma (GBM) TICs as a monolayer using laminin. We performed a side-by-side analysis of the NSA and laminin (adherent) culture conditions to compare the growth and expansion of GBM TICs. GBM cells were grown using the NSA and adherent culture conditions. Comparisons were made using growth in culture, apoptosis assays, protein expression, limiting dilution clonal frequency assay, genetic affymetrix analysis, and tumorigenicity in vivo. In vitro expansion curves for the NSA and adherent culture conditions were virtually identical (P=0.24) and the clonogenic frequencies (5.2% for NSA vs. 5.0% for laminin, P=0.9) were similar as well. Likewise, markers of differentiation (glial fibrillary acidic protein and beta tubulin III) and proliferation (Ki67 and MCM2) revealed no statistical difference between the sphere and attachment methods. Several different methods were used to determine the numbers of dead or dying cells (trypan blue, DiIC, caspase-3, and annexin V) with none of the assays noting a meaningful variance between the two methods. In addition, genetic expression analysis with microarrays revealed no significant differences between the two groups. Finally, glioma cells derived from both methods of expansion formed large invasive tumors exhibiting GBM features when implanted in immune-compromised animals. A detailed functional, protein and genetic characterization of human GBM cells cultured in serum-free defined conditions demonstrated no statistically meaningful differences when grown using sphere (NSA) or adherent conditions. Hence, both methods are functionally equivalent and remain suitable options for expanding primary high-grade gliomas in tissue culture.

16.
Neurosurgery ; 76(3): 239-47; discussion 247-8, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25603104

RESUMO

BACKGROUND: In 2010, the Patient Protection and Affordable Care Act was passed to expand health insurance, narrow health care disparities, and improve health care quality in the United States. As part of this initiative, the Agency for Healthcare Research and Quality and the Centers for Medicare and Medicaid Services are now tracking quality metrics. OBJECTIVE: To analyze the effects of insurance on the incidence of patient safety indicators (PSIs) and hospital-acquired conditions (HACs) using the Nationwide Inpatient Sample for patients who have brain tumors. METHODS: The Nationwide Inpatient Sample was queried for all hospitalizations between 2002 and 2011 involving patients with brain tumors. Because of the confounding age restriction with Medicare, comparisons were made between Medicaid/self-pay and private insurance. To determine which factors contributed to HACs and PSIs, odds ratios were calculated for each risk factor. Logistic regression models were used to assess the effect of payer status on individual PSIs, HACs, and patient outcomes. RESULTS: Medicaid/self-pay patients had a higher PSI and HAC incidence compared with private insurance patients. The greater incidence of PSIs and HACs correlated with increased length of stay, worse discharge outcomes, and increased in-hospital mortality. CONCLUSION: Variability exists in the incidence of PSIs and HACs in patients with brain tumors based on insurance status. Controlling for both patient and hospital factors can explain these differences. The cause of these disparities should be studied prospectively to begin the process of improving quality metrics in vulnerable patient populations.


Assuntos
Neoplasias Encefálicas , Infecção Hospitalar/epidemiologia , Disparidades em Assistência à Saúde/estatística & dados numéricos , Cobertura do Seguro/estatística & dados numéricos , Patient Protection and Affordable Care Act/estatística & dados numéricos , Adulto , Idoso , Neoplasias Encefálicas/complicações , Neoplasias Encefálicas/economia , Feminino , Mortalidade Hospitalar , Humanos , Pacientes Internados , Cobertura do Seguro/economia , Masculino , Pessoa de Meia-Idade , Alta do Paciente/economia , Patient Protection and Affordable Care Act/economia , Segurança do Paciente , Fatores de Risco , Estados Unidos
17.
J Neurosci Methods ; 225: 81-9, 2014 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-24378338

RESUMO

BACKGROUND: Adult neural stem/progenitor cells (NS/PCs) are one of the outstanding cell sources for therapeutic purposes in the central nervous system diseases. Autologous transplantation of NS/PCs still is a matter of controversy due to the safety issue as well as efficiency of harvesting these cells from the live mammalian brain subventricular zone (SVZ). NEW METHOD: In this new and safe method, a 16-guage semi-automatic biopsy needle was used stereotactically to remove a piece of SVZ. Then, the proliferation and differentiation capacity of obtained cells were assessed. In addition, the safety of the biopsy procedure was analyzed employing the Morris water maze, modified neurologic severity score, passive avoidance and open field tests. RESULTS: Despite being very small in size, the SVZ specimen could generate a large number of progeny with the ability to differentiate into neuronal and glial cells. The biopsy procedure introduced in this study did not have any impact on the behavioral and neurological processes. COMPARISON WITH EXISTING METHOD(S): existing SVZ biopsy methods were uncontrollable techniques which harvested brain tissue by aspiration using a syringe not a semi-automatic biopsy needle. Also, previous methods were not evaluated in terms of behavior and cognition. CONCLUSIONS: This study revealed a considerable safety and efficacy for the stereotactical removal of the adult rat SVZ to harvest NS/PCs for autologous transplantation.


Assuntos
Técnicas Citológicas , Ventrículos Laterais/citologia , Ventrículos Laterais/cirurgia , Células-Tronco Neurais , Técnicas Estereotáxicas , Envelhecimento , Animais , Biópsia , Masculino , Ratos , Ratos Wistar
18.
Future Oncol ; 9(9): 1389-96, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23980685

RESUMO

AIM: Bevacizumab has been reported to result in increased tumor invasion when used to treat malignant glioma. We hypothesized that BMP4 would prevent diffuse tumor infiltration induced by bevacizumab for malignant glioma in a xenograft model. METHODS: Human glioblastoma (GBM) tumor cells were implanted in the striatum of immunocompromised mice. The animals were treated with bevacizumab and BMP4. Tumor growth and invasion were measured. RESULTS: The bevacizumab-treated mice had increased survival compared with control animals (p = 0.02). BMP4 alone did not result in improved survival (p = 1.0). The bevacizumab (p = 0.006) and bevacizumab plus BMP4 (p = 0.006) groups demonstrated significantly decreased total tumor size compared with control. Tumor invasion was significantly decreased in the bevacizumab (p = 0.005), BMP4 (p = 0.04) alone and bevacizumab plus BMP4 (p = 0.002) groups compared with control. No synergistic effect between bevacizumab and BMP4 was observed. CONCLUSION: Bevacizumab treatment did not result in diffuse infiltration of human GBM in a mouse xenograft model. BMP4 did have an independent favorable effect on GBM that was not synergistic with bevacizumab treatment.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Anticorpos Monoclonais Humanizados/uso terapêutico , Proteína Morfogenética Óssea 4/metabolismo , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/prevenção & controle , Glioblastoma/patologia , Glioblastoma/prevenção & controle , Adulto , Animais , Bevacizumab , Neoplasias Encefálicas/metabolismo , Feminino , Glioblastoma/metabolismo , Humanos , Técnicas Imunoenzimáticas , Imageamento por Ressonância Magnética , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Invasividade Neoplásica , Taxa de Sobrevida , Carga Tumoral , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Methods Mol Biol ; 1059: 95-106, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23934837

RESUMO

The renewable source of neural stem cells (NSCs) with multi-lineage differentiation capability towards neurons, astrocytes, and oligodendrocytes represent an ideal supply for cell therapy of central nervous system (CNS) diseases. In spite of this, the clinical use of NSCs is hampered by heterogeneity, poor neuronal cell yield, predominant astrocytic differentiation of NSC progeny and possible uncontrolled proliferation, and tumor formation upon transplantation. The ability to generate highly enriched and defined neural cell populations from the renewable source of NSCs might overcome many of these impediments and pave the way towards their successful clinical applications. Here, we describe a simple method for NSC differentiation and subsequent purification of neuronal progenitor cells, taking advantage of size and granularity differences between neuronal cells and other NSC progeny. This highly enriched neuronal cell population provides an invaluable source of cells for both in vitro and in vivo studies.


Assuntos
Separação Celular/métodos , Células-Tronco Neurais/fisiologia , Animais , Diferenciação Celular , Meios de Cultura , Citometria de Fluxo , Humanos , Cultura Primária de Células
20.
J Vis Exp ; (62)2012 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-22565048

RESUMO

Tumor heterogeneity represents a fundamental feature supporting tumor robustness and presents a central obstacle to the development of therapeutic strategies(1). To overcome the issue of tumor heterogeneity, it is essential to develop assays and tools enabling phenotypic, (epi)genetic and functional identification and characterization of tumor subpopulations that drive specific disease pathologies and represent clinically relevant targets. It is now well established that tumors exhibit distinct sub-fractions of cells with different frequencies of cell division, and that the functional criteria of being slow cycling is positively associated with tumor formation ability in several cancers including those of the brain, breast, skin and pancreas as well as leukemia(2-8). The fluorescent dye carboxyfluorescein succinimidyl ester (CFSE) has been used for tracking the division frequency of cells in vitro and in vivo in blood-borne tumors and solid tumors such as glioblastoma(2,7,8). The cell-permeant non-fluorescent pro-drug of CFSE is converted by intracellular esterases into a fluorescent compound, which is retained within cells by covalently binding to proteins through reaction of its succinimidyl moiety with intracellular amine groups to form stable amide bonds(9). The fluorescent dye is equally distributed between daughter cells upon divisions, leading to the halving of the fluorescence intensity with every cell division. This enables tracking of cell cycle frequency up to eight to ten rounds of division(10). CFSE retention capacity was used with brain tumor cells to identify and isolate a slow cycling subpopulation (top 5% dye-retaining cells) demonstrated to be enriched in cancer stem cell activity(2). This protocol describes the technique of staining cells with CFSE and the isolation of individual populations within a culture of human glioblastoma (GBM)-derived cells possessing differing division rates using flow cytometry(2). The technique has served to identify and isolate a brain tumor slow-cycling population of cells by virtue of their ability to retain the CFSE labeling.


Assuntos
Neoplasias Encefálicas/patologia , Citometria de Fluxo/métodos , Fluoresceínas/química , Corantes Fluorescentes/química , Glioblastoma/patologia , Coloração e Rotulagem/métodos , Succinimidas/química , Processos de Crescimento Celular/fisiologia , Humanos , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA