Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Development ; 147(18)2020 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-32895288

RESUMO

The central nervous system (CNS) contains a complex network of blood vessels that promote normal tissue development and physiology. Abnormal control of blood vessel morphogenesis and maturation is linked to the pathogenesis of various neurodevelopmental diseases. The CNS-specific genes that regulate blood vessel morphogenesis in development and disease remain largely unknown. Here, we have characterized functions for the gene encoding prion protein 2 (Prnd) in CNS blood vessel development and physiology. Prnd encodes the glycosylphosphatidylinositol (GPI)-linked protein doppel, which is expressed on the surface of angiogenic vascular endothelial cells, but is absent in quiescent endothelial cells of the adult CNS. During CNS vascular development, doppel interacts with receptor tyrosine kinases and activates cytoplasmic signaling pathways involved in endothelial cell survival, metabolism and migration. Analysis of mice genetically null for Prnd revealed impaired CNS blood vessel morphogenesis and associated endothelial cell sprouting defects. Prnd-/- mice also displayed defects in endothelial barrier integrity. Collectively, these data reveal novel mechanisms underlying doppel control of angiogenesis in the developing CNS, and may provide new insights about dysfunctional pathways that cause vascular-related CNS disorders.


Assuntos
Barreira Hematoencefálica/metabolismo , Células Endoteliais/metabolismo , Neovascularização Patológica/metabolismo , Neovascularização Fisiológica/fisiologia , Proteínas Priônicas/metabolismo , Animais , Sistema Nervoso Central/metabolismo , Citoplasma/metabolismo , Proteínas Ligadas por GPI/metabolismo , Camundongos , Morfogênese/fisiologia , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais/fisiologia
2.
Int J Mol Sci ; 21(19)2020 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-32998285

RESUMO

Glioblastoma multiforme (GBM) is the most common malignant type of astrocytic tumors. GBM patients have a poor prognosis with a median survival of approximately 15 months despite the "Stupp" Regimen and high tumor recurrence due to the tumor resistance to chemotherapy. In this study, we co-cultured GBM cells with human astrocytes in three-dimensional (3D) poly(ethylene glycol) dimethyl acrylate (PEGDA) microwells to mimic the tumor microenvironment. We treated 3D co- and mono-cultured cells with Temozolomide (TMZ) and the nuclear factor-κB (NF-κB) inhibitor Bay 11-7082 and investigated the combined effect of the drugs. We assessed the expressions of glial fibrillary acidic protein (GFAP) and vimentin that play a role in the tumor malignancy and activation of the astrocytes as well as Notch-1 and survivin that play a role in GBM malignancy after the drug treatment to understand how astrocytes induced GBM drug response. Our results showed that in the co-culture, astrocytes increased GBM survival and resistance after combined drug treatment compared to mono-cultures. These data restated the importance of 3D cell culture to mimic the tumor microenvironment for drug screening.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Neuroglia/efeitos dos fármacos , Nitrilas/farmacologia , Sulfonas/farmacologia , Temozolomida/farmacologia , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Técnicas de Cocultura/métodos , Resistencia a Medicamentos Antineoplásicos/genética , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Modelos Biológicos , Neuroglia/metabolismo , Neuroglia/patologia , Cultura Primária de Células , Receptor Notch1/genética , Receptor Notch1/metabolismo , Transdução de Sinais , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/metabolismo , Esferoides Celulares/patologia , Survivina/genética , Survivina/metabolismo , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/genética , Vimentina/genética , Vimentina/metabolismo
3.
IEEE Open J Eng Med Biol ; 4: 251-258, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38196976

RESUMO

Glioblastoma Multiforme (GBM) is the most malignant type of all brain tumors. Current GBM treatment options include surgery, followed by radiation and chemotherapy. However, GBM can become resistant to therapy, resulting in tumor recurrence. GBM cells develop resistance to treatments by either downregulating cell death pathways (CD95) or upregulating cell survival pathways (NF-κB (p65)). Healthy tissues can be affected by the increased therapeutic dose. Therefore, it is important to develop a method that can only target GBM tumor cells, thereby reducing the non-specific uptake which will reduce the side effects. Here we demonstrate an application of novel priori activation of apoptosis pathways of tumor technology (AAAPT), which has been used to demonstrate the effect of targeted tumor sensitizers to make chemotherapy work at lower doses in breast, lung and prostate cancers. Treatment of GBM spheroids with AAAPT in 3D PEGDA microwells, showed an increase in cell death, an upregulation of cell death pathways, and a downregulation of cell survival pathways, in comparison to Temozolomide (TMZ), an oral alkylating agent, which is a commonly used chemotherapy in the treatment of GBM. The dose of AAAPT sensitizers may provide a promising method to increase treatment efficacy and reduce off-target toxicity, as an alternative to existing methods which cause significant off-target damage.

4.
Sci Rep ; 13(1): 4911, 2023 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-36966190

RESUMO

Brain growth occurs during the first 2 weeks of postnatal development in rats. This developmental period is equivalent to the third trimester of human gestation. Dendritic arborization, axonal growth, and gliogenesis are observed along with a strong maturation of neurotransmission during this critical development period. Furthermore, nicotine exposure during early development causes deficiencies in sensory and cognitive processing in adults. In this study, we further investigated the gene expression of neuron groups and the influence of perinatal nicotine exposure on gene expressions of neurons within the sub-regions of the ventral tegmental area (VTA) in 1 week, 2 week and 3-week-old rat pups. We exposed pregnant rats to nicotine perinatally on gestational day 7 through postnatal day 14. Pups are exposed to nicotine during pregnancy and through breastfeeding to investigate its effect in rat pups during early neuronal development. Real time PCR was used to find the relative expressions of gamma-aminobutyric acid (GABA), dopamine, and glutamate neuron markers within the three sub-regions of the VTA including the parabrachial pigmented nucleus (PBP), parainterfascicular (PIF), and paranigral nucleus (PN). Our results indicated that during early maturation, the dopamine marker tyrosine hydroxylase (TH) showed a consistently increased significance in PN sub-region compared to PIF and PBP. These results suggest that following perinatal nicotine exposure, VTA dopamine neurons, especially within the PN sub-region, are significantly excited starting from birth.


Assuntos
Nicotina , Efeitos Tardios da Exposição Pré-Natal , Gravidez , Humanos , Feminino , Ratos , Animais , Nicotina/efeitos adversos , Nicotina/metabolismo , Área Tegmentar Ventral/metabolismo , Transcriptoma , Dopamina/metabolismo , Ratos Sprague-Dawley , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Neurônios Dopaminérgicos/metabolismo
5.
Sci Rep ; 11(1): 8706, 2021 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-33888815

RESUMO

The ventral tegmental area (VTA) is the origin of dopaminergic neurons and the dopamine (DA) reward pathway. This pathway has been widely studied in addiction and drug reinforcement studies and is believed to be the central processing component of the reward circuit. In this study, we used a well-established rat model to expose mother dams to alcohol, nicotine-alcohol, and saline perinatally. DA and non-DA neurons collected from the VTA of the rat pups were used to study expression profiles of miRNAs and mRNAs. miRNA pathway interactions, putative miRNA-mRNA target pairs, and downstream modulated biological pathways were analyzed. In the DA neurons, 4607 genes were differentially upregulated and 4682 were differentially downregulated following nicotine-alcohol exposure. However, in the non-DA neurons, only 543 genes were differentially upregulated and 506 were differentially downregulated. Cell proliferation, differentiation, and survival pathways were enriched after the treatments. Specifically, in the PI3K/AKT signaling pathway, there were 41 miRNAs and 136 mRNAs differentially expressed in the DA neurons while only 16 miRNAs and 20 mRNAs were differentially expressed in the non-DA neurons after the nicotine-alcohol exposure. These results depicted that chronic nicotine and alcohol exposures during pregnancy differentially affect both miRNA and gene expression profiles more in DA than the non-DA neurons in the VTA. Understanding how the expression signatures representing specific neuronal subpopulations become enriched in the VTA after addictive substance administration helps us to identify how neuronal functions may be altered in the brain.


Assuntos
Neurônios Dopaminérgicos/efeitos dos fármacos , Etanol/administração & dosagem , Exposição Materna , Nicotina/administração & dosagem , Área Tegmentar Ventral/efeitos dos fármacos , Animais , Neurônios Dopaminérgicos/metabolismo , Feminino , Masculino , MicroRNAs/metabolismo , Gravidez , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Área Tegmentar Ventral/citologia , Área Tegmentar Ventral/metabolismo
6.
Sci Rep ; 10(1): 2419, 2020 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-32051445

RESUMO

Chronic nicotine exposure during pregnancy has been shown to induce physiological and anatomical alterations in offspring. Previously, we investigated the complexity of dopamine (DA) neuron firing in the sub-regions of the ventral tegmental area (VTA) following perinatal nicotine exposure. Using approximate entropy, we found that within the middle sub-region, the parainterfascicular nucleus (PIF), there was higher complexity indicating more random neural firing and a less homogeneous neuron population. Therefore, we sought to investigate the neuron populations within the sub-regions of the VTA following perinatal nicotine exposure. We used real time PCR in order to find the relative quantity of glutamate to γ-aminobutyric acid (GABA), DA, and glutamate neurons within three sub-regions: the parabrachial pigmented nucleus (PBP), parainterfascicular nucleus (PIF), and paranigral nucleus (PN). Our results showed that the PIF region of the VTA contained a more diverse population of neurons resulting in a more complex system. In addition, we found that DA neurons are more activated in PN sub-region of the VTA, which mediates the rewarding effects of drugs including nicotine. Lastly, using immunohistochemistry, we observed an overall decrease in DA neurons following perinatal nicotine exposure.


Assuntos
Expressão Gênica/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Nicotina/efeitos adversos , Agonistas Nicotínicos/efeitos adversos , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Área Tegmentar Ventral/efeitos dos fármacos , Animais , Feminino , Perfil Genético , Masculino , Neurônios/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/genética , Ratos Sprague-Dawley , Área Tegmentar Ventral/citologia
7.
Sci Rep ; 10(1): 15016, 2020 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-32929144

RESUMO

Nicotine and alcohol are two of the most commonly used and abused recreational drugs, are often used simultaneously, and have been linked to significant health hazards. Furthermore, patients diagnosed with dependence on one drug are highly likely to be dependent on the other. Several studies have shown the effects of each drug independently on gene expression within many brain regions, including the ventral tegmental area (VTA). Dopaminergic (DA) neurons of the dopamine reward pathway originate from the VTA, which is believed to be central to the mechanism of addiction and drug reinforcement. Using a well-established rat model for both nicotine and alcohol perinatal exposure, we investigated miRNA and mRNA expression of dopaminergic (DA) neurons of the VTA in rat pups following perinatal alcohol and joint nicotine-alcohol exposure. Microarray analysis was then used to profile the differential expression of both miRNAs and mRNAs from DA neurons of each treatment group to further explore the altered genes and related biological pathways modulated. Predicted and validated miRNA-gene target pairs were analyzed to further understand the roles of miRNAs within these networks following each treatment, along with their post transcription regulation points affecting gene expression throughout development. This study suggested that glutamatergic synapse and axon guidance pathways were specifically enriched and many miRNAs and genes were significantly altered following alcohol or nicotine-alcohol perinatal exposure when compared to saline control. These results provide more detailed insight into the cell proliferation, neuronal migration, neuronal axon guidance during the infancy in rats in response to perinatal alcohol/ or nicotine-alcohol exposure.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Etanol/farmacologia , Redes Reguladoras de Genes , Nicotina/farmacologia , Efeitos Tardios da Exposição Pré-Natal/genética , Área Tegmentar Ventral/metabolismo , Animais , Neurônios Dopaminérgicos/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Masculino , MicroRNAs/genética , MicroRNAs/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Transcriptoma , Área Tegmentar Ventral/citologia , Área Tegmentar Ventral/efeitos dos fármacos
8.
Sci Rep ; 10(1): 13352, 2020 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-32770097

RESUMO

Glioblastoma (GBM) is the most malignant brain tumor characterized by intrinsic or acquired resistance to chemotherapy. GBM tumors show nuclear factor-κB (NF-κB) activity that has been associated with tumor formation, growth, and increased resistance to therapy. We investigated the effect of NF-κB inhibitor BAY 11-7082 with Temozolomide (TMZ) on the signaling pathways in GBM pathogenesis. GBM cells and patient-derived GBM cells cultured in 3D microwells were co-treated with BAY 11-7082 and TMZ or BAY 11-7082 and TMZ alone, and combined experiments of cell proliferation, apoptosis, wound healing assay, as well as reverse-phase protein arrays, western blot and immunofluorescence staining were used to evaluate the effects of drugs on GBM cells. The results revealed that the co-treatment significantly altered cell proliferation by decreasing GBM viability, suppressed NF-κB pathway and enhanced apoptosis. Moreover, it was found that the co-treatment of BAY 11-7082 and TMZ significantly contributed to a decrease in the migration pattern of patient-derived GBM cells by modulating actin cytoskeleton pathway. These findings suggest that in addition to TMZ treatment, NF-κB can be used as a potential target to increase the treatment's outcomes. The drug combination strategy, which is significantly improved by NF-κB inhibitor could be used to better understand the underlying mechanism of GBM pathways in vivo and as a potential therapeutic tool for GBM treatment.


Assuntos
Citoesqueleto de Actina/metabolismo , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , NF-kappa B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Temozolomida/uso terapêutico , Fator de Transcrição RelA/metabolismo , Citoesqueleto de Actina/efeitos dos fármacos , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática , Humanos , Immunoblotting , NF-kappa B/antagonistas & inibidores , Nitrilas/farmacologia , Sulfonas/farmacologia
9.
Artigo em Inglês | MEDLINE | ID: mdl-35402955

RESUMO

Glioblastoma multiforme (GBM) is the most common malignant primary brain tumor, accounting for 50% of all cases. GBM patients have a five-year survival rate of merely 5.6% and a median overall survival of 14.6 months with the "Stupp" regimen, 20.9 months with tumor treatment fields (TTF, OptuneR) in patients who participated in clinical trials, and 11 months for all GBM patients prior to TTF use. Objective: Our group recently developed a brain cancer chip which generates tumor spheroids, and provides large-scale assessments on the response of tumor cells to various concentrations and combinations of drugs. This platform could optimize the use of tumor samples derived from GBM patients to provide valuable insight on the tumor growth and responses to drug therapies. To minimize any sample loss in vitro, we improved our brain cancer chip system by adding an additional laminar flow distribution layer, which reduces sample loss during cell seeding and prevents spheroids from escaping from the microwells. Methods: In this study, we cultured 3D spheroids from GBM cell lines and patient-derived GBM cells in vitro, and investigated the effect of the combination of Temozolomide and nuclear factor-κB inhibitor on tumor growth. Results: Our study revealed that these drugs have synergistic effects in inhibiting spheroid formation when used in combination. Conclusions: These results suggest that the brain cancer chip enables large-scale, inexpensive and sample-effective drug screening to 3D cancer tumors in vitro, and could be applied to related tissue engineering drug screening studies.

10.
Cancers (Basel) ; 11(9)2019 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-31540485

RESUMO

Diffuse invasion into adjacent brain matter by glioblastoma (GBM) is largely responsible for their dismal prognosis. Previously, we showed that the TWIST1 (TW) bHLH transcription factor and its regulated gene periostin (POSTN) promote invasive phenotypes of GBM cells. Since TW functional effects are regulated by phosphorylation and dimerization, we investigated how phosphorylation of serine 68 in TW regulates TW dimerization, POSTN expression, and invasion in glioma cells. Compared with wild-type TW, the hypophosphorylation mutant, TW(S68A), impaired TW heterodimerization with the E12 bHLH transcription factor and cell invasion in vitro but had no effect on TW homodimerization. Overexpression of TW:E12 forced dimerization constructs (FDCs) increased glioma cell invasion and upregulated pro-invasive proteins, including POSTN, in concert with cytoskeletal reorganization. By contrast, TW:TW homodimer FDCs inhibited POSTN expression and cell invasion in vitro. Further, phosphorylation of analogous PXSP phosphorylation sites in TW:E12 FDCs (TW S68 and E12 S139) coordinately regulated POSTN and PDGFRa mRNA expression. These results suggested that TW regulates pro-invasive phenotypes in part through coordinated phosphorylation events in TW and E12 that promote heterodimer formation and regulate downstream targets. This new mechanistic understanding provides potential therapeutic strategies to inhibit TW-POSTN signaling in GBM and other cancers.

11.
Cancers (Basel) ; 10(6)2018 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-29857493

RESUMO

With the progress of immunotherapy in cancer, oncolytic viruses (OVs) have attracted more and more attention during the past decade. Due to their cancer-selective and immunogenic properties, OVs are considered ideal candidates to be combined with immunotherapy to increase both specificity and efficacy in cancer treatment. OVs preferentially replicate in and lyse cancer cells, resulting in in situ autovaccination leading to adaptive anti-virus and anti-tumor immunity. The main challenge in OV approaches is how to redirect the host immunity from anti-virus to anti-tumor and optimize the clinical outcome of cancer patients. Here, we summarize the conceptual updates on oncolytic virotherapy and immunotherapy in cancer, and the development of strategies to enhance the virus-mediated anti-tumor immune response, including: (1) arm OVs with cytokines to modulate innate and adaptive immunity; (2) combining OVs with immune checkpoint inhibitors to release T cell inhibition; (3) combining OVs with immune co-stimulators to enhance T cell activation. Future studies need to be enforced on developing strategies to augment the systemic effect on metastasized tumors.

12.
Sci Rep ; 8(1): 15423, 2018 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-30337660

RESUMO

Glioblastoma multiforme (GBM), an extremely invasive and high-grade (grade IV) glioma, is the most common and aggressive form of brain cancer. It has a poor prognosis, with a median overall survival of only 11 months in the general GBM population and 14.6 to 21 months in clinical trial participants with standard GBM therapies, including maximum safe craniotomy, adjuvant radiation, and chemotherapies. Therefore, new approaches for developing effective treatments, such as a tool for assessing tumor cell drug response before drug treatments are administered, are urgently needed to improve patient survival. To address this issue, we developed an improved brain cancer chip with a diffusion prevention mechanism that blocks drugs crossing from one channel to another. In the current study, we demonstrate that the chip has the ability to culture 3D spheroids from patient tumor specimen-derived GBM cells obtained from three GBM patients. Two clinical drugs used to treat GBM, temozolomide (TMZ) and bevacizumab (Avastin, BEV), were applied and a range of relative concentrations was generated by the microfluidic channels in the brain cancer chip. The results showed that TMZ works more effectively when used in combination with BEV compared to TMZ alone. We believe that this low-cost brain cancer chip could be further developed to generate optimal combination of chemotherapy drugs tailored to individual GBM patients.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Encefálicas/patologia , Sobrevivência Celular , Avaliação Pré-Clínica de Medicamentos/métodos , Glioblastoma/patologia , Dispositivos Lab-On-A-Chip , Esferoides Celulares/patologia , Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Humanos , Esferoides Celulares/efeitos dos fármacos , Células Tumorais Cultivadas
13.
Mol Cancer Res ; 14(12): 1277-1287, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27655131

RESUMO

Glioblastoma is a primary brain cancer that is resistant to all treatment modalities. This resistance is due, in large part, to invasive cancer cells that disperse from the main tumor site, escape surgical resection, and contribute to recurrent secondary lesions. The adhesion and signaling mechanisms that drive glioblastoma cell invasion remain enigmatic, and as a result there are no effective anti-invasive clinical therapies. Here we have characterized a novel adhesion and signaling pathway comprised of the integrin αvß8 and its intracellular binding partner, Spinophilin (Spn), which regulates glioblastoma cell invasion in the brain microenvironment. We show for the first time that Spn binds directly to the cytoplasmic domain of ß8 integrin in glioblastoma cells. Genetically targeting Spn leads to enhanced invasive cell growth in preclinical models of glioblastoma. Spn regulates glioblastoma cell invasion by modulating the formation and dissolution of invadopodia. Spn-regulated invadopodia dynamics are dependent, in part, on proper spatiotemporal activation of the Rac1 GTPase. Glioblastoma cells that lack Spn showed diminished Rac1 activities, increased numbers of invadopodia, and enhanced extracellular matrix degradation. Collectively, these data identify Spn as a critical adhesion and signaling protein that is essential for modulating glioblastoma cell invasion in the brain microenvironment. IMPLICATIONS: Tumor cell invasion is a major clinical obstacle in glioblastoma and this study identifies a new signaling pathway regulated by Spinophilin in invasive glioblastoma. Mol Cancer Res; 14(12); 1277-87. ©2016 AACR.


Assuntos
Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Integrinas/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Podossomos/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Sítios de Ligação , Neoplasias Encefálicas/metabolismo , Adesão Celular , Linhagem Celular Tumoral , Movimento Celular , Glioblastoma/metabolismo , Humanos , Integrinas/química , Camundongos , Proteínas dos Microfilamentos/química , Invasividade Neoplásica , Transplante de Neoplasias , Proteínas do Tecido Nervoso/química , Ligação Proteica , Transdução de Sinais
14.
IEEE Trans Nanobioscience ; 14(7): 790-6, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26571536

RESUMO

Glioblastoma (GBM) is the most common form of primary brain tumor with a high infiltrative capacity, increased vascularity, and largely elusive tumor progression mechanism. The current GBM treatment methods do not increase the patient survival rate and studies using two-dimensional (2D) cell cultures and in vivo animal models to investigate GBM behavior and mechanism have limitations. Therefore, there is an increasing need for in vitro three-dimensional (3D) models that closely mimic in vivo microenvironment of the GBM tumors to understand the underlying mechanisms of the tumor progression. In this study we propose to use a 3D in vitro model to overcome these limitations, using poly (ethylene glycol) dimethyl acrylate (PEGDA) hydrogel-based microwells and co-culture GBM (U87) cells and endothelial cells (HUVEC) in the 3D microwells to provide a 3D in vitro simulation of the tumor microenvironment. Furthermore, we investigated the gene expression differences of co-cultures by quantitative real-time PCR. Our results suggested that the relative expression profiles of tumor angiogenesis markers, PECAM1/CD31, and VEGFR2, in co-cultures are consistent with in vivo GBM studies. Furthermore, we suggest that our microwell platform could provide robust and useful 3D co-culture models for high-throughput drug screening and treatment of the GBM.


Assuntos
Comunicação Celular , Técnicas de Cocultura/instrumentação , Células Endoteliais/fisiologia , Glioblastoma/fisiopatologia , Dispositivos Lab-On-A-Chip , Microambiente Tumoral/fisiologia , Células Cultivadas , Células Endoteliais/patologia , Desenho de Equipamento , Análise de Falha de Equipamento , Glioblastoma/patologia , Humanos , Esferoides Celulares/patologia , Esferoides Celulares/fisiologia
15.
IEEE J Transl Eng Health Med ; 3: 4300108, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-27170911

RESUMO

Glioblastoma multiforme (GBM) is the most common and malignant primary brain tumor in adults because of its highly invasive behavior. The existing treatment for GBM, which involves a combination of resection, chemotherapy, and radiotherapy, has a very limited success rate with a median survival rate of <1 year. This is mainly because of the failure of early detection and effective treatment. We designed a novel 3-D GBM cell culture model based on microwells that could mimic in vitro environment and help to bypass the lack of suitable animal models for preclinical toxicity tests. Microwells were fabricated from simple and inexpensive polyethylene glycol material for the control of in vitro 3-D culture. We applied the 3-D micropatterning system to GBM (U-87) cells using the photolithography technique to control the cell spheroids' shape, size, and thickness. Our preliminary results suggested that uniform GBM spheroids can be formed in 3-D, and the size of these GBM spheroids depends on the size of microwells. The viability of the spheroids generated in this manner was quantitatively evaluated using live/dead assay and shown to improve over 21 days. We believe that in vitro 3-D cell culture model could help to reduce the time of the preclinical brain tumor growth studies. The proposed novel platform could be useful and cost-effective for high-throughput screening of cancer drugs and assessment of treatment responses.

16.
IEEE Trans Nanobioscience ; 14(8): 946-51, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26661633

RESUMO

Glioblastoma (GBM) is the most aggressive brain tumor, with 12-15 months median survival time despite current treatment efforts. Among the alternative treatment approaches that have gained acceptance over the last decade is the use of replication-competent oncolytic adenoviruses, which are promising due to their relatively low toxicity and tumor-specific targeting. Three-dimensional (3D) tumor models can mimic the physiological microenvironment of GBM tumors and provide valuable information about the interaction between tumor cells and adenoviruses. Therefore, robust in vitro 3D tumor models are critical to investigate the mechanisms underlying tumor progression and explore the cytotoxicity effect of the adenovirus on tumor cells. In this study, we used a hydrogel microwell platform to generate in vitro 3D GBM spheroids and studied their interactions with the Delta-24-RGD adenovirus. The results showed that the cultured 3D spheroids were successfully infected by the Delta-24-RGD. A significant cell lysis was observed. Cell viability was decreased approximately 37%, 54% and 65% with 10, 50, and 100 MOIs, respectively. The infection of the Delta-24-RGD was found more effective on 3D spheroids when compared to 2D monolayer cell culture. These results implicate that our hydrogel microwell platform could provide a promising 3D model to investigate the oncolytic potential of the viruses in vitro.


Assuntos
Adenoviridae/genética , Glioblastoma , Modelos Biológicos , Esferoides Celulares/fisiologia , Microambiente Tumoral/fisiologia , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Técnicas de Transferência de Genes , Células Endoteliais da Veia Umbilical Humana , Humanos , Hidrogéis/química , Oligopeptídeos/genética , Polietilenoglicóis/química , Esferoides Celulares/virologia , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa