Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
2.
Sci Rep ; 13(1): 8087, 2023 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-37208357

RESUMO

Y-box binding protein 1 (YBX1 or YB1) is a therapeutically relevant oncoprotein capable of RNA and DNA binding and mediating protein-protein interactions that drive proliferation, stemness, and resistance to platinum-based therapies. Given our previously published findings, the potential for YB1-driven cisplatin resistance in medulloblastoma (MB), and the limited studies exploring YB1-DNA repair protein interactions, we chose to investigate the role of YB1 in mediating radiation resistance in MB. MB, the most common pediatric malignant brain tumor, is treated with surgical resection, cranio-spinal radiation, and platinum-based chemotherapy, and could potentially benefit from YB1 inhibition. The role of YB1 in the response of MB to ionizing radiation (IR) has not yet been studied but remains relevant for determining potential anti-tumor synergy of YB1 inhibition with standard radiation therapy. We have previously shown that YB1 drives proliferation of cerebellar granular neural precursor cells (CGNPs) and murine Sonic Hedgehog (SHH) group MB cells. While others have demonstrated a link between YB1 and homologous recombination protein binding, functional and therapeutic implications remain unclear, particularly following IR-induced damage. Here we show that depleting YB1 in both SHH and Group 3 MB results not only in reduced proliferation but also synergizes with radiation due to differential response dynamics. YB1 silencing through shRNA followed by IR drives a predominantly NHEJ-dependent repair mechanism, leading to faster γH2AX resolution, premature cell cycle re-entry, checkpoint bypass, reduced proliferation, and increased senescence. These findings show that depleting YB1 in combination with radiation sensitizes SHH and Group 3 MB cells to radiation.


Assuntos
Neoplasias Encefálicas , Neoplasias Cerebelares , Meduloblastoma , Células-Tronco Neurais , Proteína 1 de Ligação a Y-Box , Animais , Humanos , Camundongos , Neoplasias Encefálicas/metabolismo , Proliferação de Células , Neoplasias Cerebelares/patologia , Dano ao DNA , Proteínas Hedgehog/metabolismo , Meduloblastoma/patologia , Células-Tronco Neurais/metabolismo , Proteína 1 de Ligação a Y-Box/metabolismo
3.
Methods Mol Biol ; 2423: 103-107, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34978692

RESUMO

Metastasis of cancer cells leads to 90% of lethality among cancer patients. A crucial step in the hematogenous spread of metastatic cancer is the detachment of cells from the primary tumor followed by invasion through nearby blood vessels (Wong and Hynes. Cell Cycle 5(8):812-817, 2006). This is common to several solid tumors, including medulloblastoma (Van Ommeren et al. Brain Pathol 30:691-702, 2020). Because invasion is a crucial step in metastasis, the development of assays studying invasion are important for identifying antimetastatic drugs. There is always a need to develop better 3D in vitro models that not only mimic the complexity of in vivo architecture of solid tumors and their microenvironment, but are also simple to execute in medium to high throughput. We developed an in vitro coculture invasion assay that relies on the binary interaction between cancer cells and endothelial cells for research on tumor invasion and antimetastatic drug discovery. The goal of the current protocol is to use the simplicity of a two-dimensional endothelial cell culture to create a gel-free physiological substratum that can facilitate cancer cell invasion from a 3D cancer spheroid. This provides a simple and reproducible biomimetic 3D cell-based system for the analysis of invasion capacity in large populations of tumor spheroids. Using this assay, we can compare the effect of invasion inhibitors/activators on cancer spheroids. The results are analyzed by manual scoring of images for the presence or absence of sprouting from cancer spheroids. This enables simple and fast analysis of metastasis, which facilitates multiparameter examination.


Assuntos
Antineoplásicos , Células Endoteliais , Antineoplásicos/farmacologia , Técnicas de Cultura de Células/métodos , Linhagem Celular Tumoral , Técnicas de Cocultura , Humanos , Esferoides Celulares
4.
Methods Mol Biol ; 2423: 109-113, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34978693

RESUMO

Recently, chimeric antigen receptor (CAR) T-cell therapy has begun to be used for solid tumors such as glioblastoma multiforme. Pediatric malignant brain tumors patients develop extensive long-term morbidity of intensive multimodal curative treatment. CAR T-cells treatments could potentially create favorable outcomes and reduce the toxicity of current treatment. T-cell infiltration of solid tumors has been associated with good prognosis. A largely overlooked area of CAR T-cell therapy targeting solid tumors is enhancing the ability of CAR T-cells to migrate and infiltrate solid tumors. A potential reason could be lack of standard in vitro assays which can screen for genetic modifications that result in enhanced T-cell migration in CAR T-cell therapies. We report a novel coculture assay using 3D tumor spheroids cocultured with T-cells to analyze the effect of activating CAR T-cell interventions on cell migration by a simple imaging based readout. This assay can be applied to several different kinds of cancer cell lines in higher throughput as well as toward measuring the efficiency of currently available CAR T-cell therapies in untested solid tumors.


Assuntos
Glioblastoma , Receptores de Antígenos Quiméricos , Movimento Celular , Criança , Glioblastoma/metabolismo , Glioblastoma/terapia , Humanos , Imunoterapia Adotiva/métodos , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T
5.
Methods Mol Biol ; 2423: 181-185, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34978699

RESUMO

Experimental anticancer agents have a history of failing in the late stages of clinical development, which has led to significantly increased losses to stakeholders during the drug development process. A bioinformatics-based approach to predict and derisk a drug development program can save time, effort, and expenses resulting from failure of experimental anticancer agents in preclinical/early clinical stages. We present a two-step in silico ensemble method, involving the comparison of localized gene expression from surrounding tissue with tumor tissue, and subsequent correlation with patient survival data, which can help predict safety and efficacy for siRNA-based drug delivery to internal cancer tissues. This is achieved by reducing the possible off-target effects due to reduced or minimal expression of the drug target in surrounding tissue, and increasing survival probability for patients whose cancers can be controlled/eliminated by siRNA-mediated inhibition of drug target. This kind of approach can be useful for more efficient drug development efforts in oncology through reduction of investment in expensive experimentation during the discovery and preclinical phases; and ultimately support the intended clinical trial design.


Assuntos
Terapêutica com RNAi , Projetos de Pesquisa , Humanos , Oncologia , Assistência Centrada no Paciente , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/uso terapêutico
6.
Mol Oncol ; 16(4): 1009-1025, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34482626

RESUMO

Sonic hedgehog (Shh)-driven medulloblastoma (Shh MB) cells are dependent on constitutive Shh signaling, but targeted treatment of Shh MB has been ineffective due to drug resistance. The purpose of this study was to address the critical role of signal transducer and activator of transcription 3 (STAT3) in Shh signaling and drug resistance in Shh MB cells. Herein, we show that STAT3 is required for Smoothened (Smo)-dependent Shh signaling and, in turn, is reciprocally regulated by Shh signaling, and demonstrate that STAT3 activity is critical for expression of HCK proto-oncogene, Src family tyrosine kinase (Hck) in Shh MB. We also demonstrate that maintained STAT3 activity suppresses p21 expression and promotes colony formation of Shh MB cells, whereas dual treatment with inhibitors of both Smo and STAT3 results in marked synergistic killing and overcomes drug resistance in vitro of Smo antagonist-resistant Shh MB cells. Finally, STAT3 inhibitor treatment significantly prevents in vivo tumor formation in genetically engineered Shh MB mice. Collectively, we show that STAT3 is necessary to maintain Shh signaling and thus is a potential therapeutic target to treat Shh MB and overcome anti-Smo drug resistance.


Assuntos
Neoplasias Cerebelares , Meduloblastoma , Animais , Carcinogênese/genética , Neoplasias Cerebelares/patologia , Proteínas Hedgehog/metabolismo , Humanos , Meduloblastoma/tratamento farmacológico , Meduloblastoma/genética , Meduloblastoma/metabolismo , Camundongos , Fator de Transcrição STAT3/metabolismo , Receptor Smoothened/genética , Receptor Smoothened/metabolismo
7.
Proc Natl Acad Sci U S A ; 117(39): 24205-24212, 2020 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-32934143

RESUMO

The sonic hedgehog subtype of medulloblastoma (SHH MB) is associated with treatment failure and poor outcome. Current strategies utilizing whole brain radiation therapy result in deleterious off-target effects on the normal developing childhood brain. Most conventional chemotherapies remain limited by ineffective blood-brain barrier (BBB) penetrance. These challenges signify an unmet need for drug carriers that can cross the BBB and deliver drugs to targeted sites with high drug-loading efficiency and long-term stability. We herein leverage the enhanced stability and targeting ability of engineered high-density lipoprotein-mimetic nanoparticles (eHNPs) to cross the BBB and deliver a SHH inhibitor effectively to the cancer stem-like cell population in SHH MB. Our microfluidic technology enabled highly reproducible production of multicomponent eHNPs incorporated with apolipoprotein A1, anti-CD15, and a SHH inhibitor (LDE225). We demonstrate the dual-targeted delivery and enhanced therapeutic effect of eHNP-A1-CD15-LDE225 via scavenger receptor class B type 1 (SR-B1) and CD15 on brain SHH MB cells in vitro, ex vivo, and in vivo. Moreover, we show that eHNP-A1 not only serves as a stable drug carrier, but also has a therapeutic effect itself through SR-B1-mediated intracellular cholesterol depletion in SHH MB cells. Through the facilitated and targeted cellular uptake of drugs and direct therapeutic role of this engineered biomimetic nanocarrier in SHH MB, our multifunctional nanoparticle provides intriguing therapeutic promise as an effective and potent nanomedicine for the treatment of SHH MB.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias Cerebelares/tratamento farmacológico , Portadores de Fármacos , Meduloblastoma/tratamento farmacológico , Nanopartículas/química , Animais , Materiais Biomiméticos , Barreira Hematoencefálica , Linhagem Celular Tumoral , HDL-Colesterol , Feminino , Proteínas Hedgehog/antagonistas & inibidores , Humanos , Camundongos Transgênicos , Terapia de Alvo Molecular , Gravidez , Tamoxifeno
8.
Mol Cancer Res ; 14(1): 114-24, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26446920

RESUMO

UNLABELLED: Sonic hedgehog (Shh) signaling is closely coupled with bioenergetics of medulloblastoma, the most common malignant pediatric brain tumor. Shh-associated medulloblastoma arises from cerebellar granule neuron precursors (CGNP), a neural progenitor whose developmental expansion requires signaling by Shh, a ligand secreted by the neighboring Purkinje neurons. Previous observations show that Shh signaling inhibits fatty acid oxidation although driving increased fatty acid synthesis. Proliferating CGNPs and mouse Shh medulloblastomas feature high levels of glycolytic enzymes in vivo and in vitro. Because both of these metabolic processes are closely linked to mitochondrial bioenergetics, the role of Shh signaling in mitochondrial biogenesis was investigated. This report uncovers a surprising decrease in mitochondrial membrane potential (MMP) and overall ATP production in CGNPs exposed to Shh, consistent with increased glycolysis resulting in high intracellular acidity, leading to mitochondrial fragmentation. Ultrastructural examination of mitochondria revealed a spherical shape in Shh-treated cells, in contrast to the elongated appearance in vehicle-treated postmitotic cells. Expression of mitofusin 1 and 2 was reduced in these cells, although their ectopic expression restored the MMP to the nonproliferating state and the morphology to a fused, interconnected state. Mouse Shh medulloblastoma cells featured drastically impaired mitochondrial morphology, restoration of which by ectopic mitofusin expression was also associated with a decrease in the expression of Cyclin D2 protein, a marker for proliferation. IMPLICATIONS: This report exposes a novel role for Shh in regulating mitochondrial dynamics and rescue of the metabolic profile of tumor cells to that of nontransformed, nonproliferating cells and represents a potential avenue for development of medulloblastoma therapeutics.


Assuntos
GTP Fosfo-Hidrolases/metabolismo , Proteínas Hedgehog/metabolismo , Mitofagia , Células-Tronco Neurais/metabolismo , Animais , Animais Recém-Nascidos , Proliferação de Células , Células Cultivadas , Neoplasias Cerebelares , GTP Fosfo-Hidrolases/genética , Glicólise , Meduloblastoma , Potencial da Membrana Mitocondrial , Camundongos , Mitocôndrias/ultraestrutura , Neoplasias Experimentais , Células-Tronco Neurais/citologia
9.
Virol J ; 8: 531, 2011 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-22152111

RESUMO

BACKGROUND: Xenotropic murine leukemia virus (MLV)-related virus (XMRV) is a gammaretrovirus that was discovered in prostate cancer tissues. Recently, it has been proposed that XMRV is a laboratory contaminant and may have originated via a rare recombination event. Host restriction factor APOBEC3G (A3G) has been reported to severely restrict XMRV replication in human peripheral blood mononuclear cells. Interestingly, XMRV infects and replicates efficiently in prostate cancer cells of epithelial origin. It has been proposed that due to lack off or very low levels of A3G protein XMRV is able to productively replicate in these cells. FINDINGS: This report builds on and challenges the published data on the absence of A3G protein in prostate epithelial cells lines. We demonstrate the presence of A3G in prostate epithelial cell lines (LNCaP and DU145) by western blot and mass spectrometry. We believe the discrepancy in A3G detection is may be due to selection and sensitivity of A3G antibodies employed in the prior studies. Our results also indicate that XMRV produced from A3G expressing LNCaP cells can infect and replicate in target cells. Most importantly our data reveal downregulation of A3G in XMRV infected LNCaP and DU145 cells. CONCLUSIONS: We propose that XMRV replicates efficiently in prostate epithelial cells by downregulating A3G expression. Given that XMRV lacks accessory proteins such as HIV-1 Vif that are known to counteract A3G function in human cells, our data suggest a novel mechanism by which retroviruses can counteract the antiviral effects of A3G proteins.


Assuntos
Citidina Desaminase/genética , Próstata/metabolismo , Vírus Relacionado ao Vírus Xenotrópico da Leucemia Murina/fisiologia , Desaminase APOBEC-3G , Sequência de Aminoácidos , Animais , Western Blotting , Linhagem Celular Tumoral , Citidina Desaminase/metabolismo , Regulação para Baixo , Eletroforese em Gel de Poliacrilamida , Células Epiteliais , Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Masculino , Espectrometria de Massas , Camundongos , Dados de Sequência Molecular , Próstata/citologia , Próstata/virologia , Neoplasias da Próstata , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA