Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 157(2): 313-328, 2014 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-24656405

RESUMO

Glioblastoma multiforme (GBM) is the most aggressive form of brain cancer with marginal life expectancy. Based on the assumption that GBM cells gain functions not necessarily involved in the cancerous process, patient-derived glioblastoma cells (GCs) were screened to identify cellular processes amenable for development of targeted treatments. The quinine-derivative NSC13316 reliably and selectively compromised viability. Synthetic chemical expansion reveals delicate structure-activity relationship and analogs with increased potency, termed Vacquinols. Vacquinols stimulate death by membrane ruffling, cell rounding, massive macropinocytic vacuole accumulation, ATP depletion, and cytoplasmic membrane rupture of GCs. The MAP kinase MKK4, identified by a shRNA screen, represents a critical signaling node. Vacquinol-1 displays excellent in vivo pharmacokinetics and brain exposure, attenuates disease progression, and prolongs survival in a GBM animal model. These results identify a vulnerability to massive vacuolization that can be targeted by small molecules and point to the possible exploitation of this process in the design of anticancer therapies.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Piperidinas/farmacologia , Quinolinas/farmacologia , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Xenoenxertos , Humanos , Hidroxiquinolinas/farmacologia , MAP Quinase Quinase 4/metabolismo , Camundongos , Transplante de Neoplasias , Pinocitose/efeitos dos fármacos , Vacúolos/metabolismo , Peixe-Zebra
3.
Proc Natl Acad Sci U S A ; 110(16): E1524-32, 2013 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-23576737

RESUMO

Coherent network activity among assemblies of interconnected cells is essential for diverse functions in the adult brain. However, cellular networks before formations of chemical synapses are poorly understood. Here, embryonic stem cell-derived neural progenitors were found to form networks exhibiting synchronous calcium ion (Ca(2+)) activity that stimulated cell proliferation. Immature neural cells established circuits that propagated electrical signals between neighboring cells, thereby activating voltage-gated Ca(2+) channels that triggered Ca(2+) oscillations. These network circuits were dependent on gap junctions, because blocking prevented electrotonic transmission both in vitro and in vivo. Inhibiting connexin 43 gap junctions abolished network activity, suppressed proliferation, and affected embryonic cortical layer formation. Cross-correlation analysis revealed highly correlated Ca(2+) activities in small-world networks that followed a scale-free topology. Graph theory predicts that such network designs are effective for biological systems. Taken together, these results demonstrate that immature cells in the developing brain organize in small-world networks that critically regulate neural progenitor proliferation.


Assuntos
Encéfalo/embriologia , Proliferação de Células , Rede Nervosa , Células-Tronco Neurais/fisiologia , Animais , Cálcio/metabolismo , Canais de Cálcio Tipo L/metabolismo , Conexina 43/metabolismo , Sinapses Elétricas/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Interferência , Modelos Neurológicos , Células-Tronco Neurais/citologia , Plasmídeos/genética , RNA Interferente Pequeno/genética
4.
Nature ; 451(7177): 460-4, 2008 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-18185516

RESUMO

Stem cell self-renewal implies proliferation under continued maintenance of multipotency. Small changes in numbers of stem cells may lead to large differences in differentiated cell numbers, resulting in significant physiological consequences. Proliferation is typically regulated in the G1 phase, which is associated with differentiation and cell cycle arrest. However, embryonic stem (ES) cells may lack a G1 checkpoint. Regulation of proliferation in the 'DNA damage' S/G2 cell cycle checkpoint pathway is known for its role in the maintenance of chromatin structural integrity. Here we show that autocrine/paracrine gamma-aminobutyric acid (GABA) signalling by means of GABA(A) receptors negatively controls ES cell and peripheral neural crest stem (NCS) cell proliferation, preimplantation embryonic growth and proliferation in the boundary-cap stem cell niche, resulting in an attenuation of neuronal progenies from this stem cell niche. Activation of GABA(A) receptors leads to hyperpolarization, increased cell volume and accumulation of stem cells in S phase, thereby causing a rapid decrease in cell proliferation. GABA(A) receptors signal through S-phase checkpoint kinases of the phosphatidylinositol-3-OH kinase-related kinase family and the histone variant H2AX. This signalling pathway critically regulates proliferation independently of differentiation, apoptosis and overt damage to DNA. These results indicate the presence of a fundamentally different mechanism of proliferation control in these stem cells, in comparison with most somatic cells, involving proteins in the DNA damage checkpoint pathway.


Assuntos
Histonas/metabolismo , Receptores de GABA-A/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Comunicação Autócrina , Blastocisto/citologia , Blastocisto/enzimologia , Blastocisto/metabolismo , Contagem de Células , Ciclo Celular , Linhagem Celular , Proliferação de Células , Tamanho Celular , Dano ao DNA , Agonistas de Receptores de GABA-A , Antagonistas de Receptores de GABA-A , Histonas/deficiência , Histonas/genética , Camundongos , Crista Neural/citologia , Crista Neural/metabolismo , Comunicação Parácrina , Técnicas de Patch-Clamp , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Receptores de GABA-A/genética , Células-Tronco/enzimologia , Ácido gama-Aminobutírico/metabolismo
5.
Proc Natl Acad Sci U S A ; 108(14): 5837-42, 2011 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-21436033

RESUMO

Adult neural stem cell proliferation is dynamic and has the potential for massive self-renewal yet undergoes limited cell division in vivo. Here, we report an epigenetic mechanism regulating proliferation and self-renewal. The recruitment of the PI3K-related kinase signaling pathway and histone H2AX phosphorylation following GABA(A) receptor activation limits subventricular zone proliferation. As a result, NSC self-renewal and niche size is dynamic and can be directly modulated in both directions pharmacologically or by genetically targeting H2AX activation. Surprisingly, changes in proliferation have long-lasting consequences on stem cell numbers, niche size, and neuronal output. These results establish a mechanism that continuously limits proliferation and demonstrates its impact on adult neurogenesis. Such homeostatic suppression of NSC proliferation may contribute to the limited self-repair capacity of the damaged brain.


Assuntos
Células-Tronco Adultas/fisiologia , Ciclo Celular/fisiologia , Proliferação de Células , Reparo do DNA/fisiologia , Epigênese Genética/fisiologia , Histonas/metabolismo , Células-Tronco Neurais/fisiologia , Transdução de Sinais/fisiologia , Animais , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Estatísticas não Paramétricas
6.
Semin Cancer Biol ; 22(5-6): 369-73, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22659342

RESUMO

Epithelial to mesenchymal transition (EMT) during metastasis is initially a two-step process beginning with delamination of cells from the solid tumor followed by acquisition of a migratory phenotype. Several reports indicate that plasma membrane blebbing, associated with cell division, coincides with cell delamination during developmental EMT. This raises a speculative question if blebbing drives EMT in cancer cells in a similar way. Here, we review available data on factors and processes that may support such a connection.


Assuntos
Transição Epitelial-Mesenquimal , Mitose , Neoplasias/genética , Neoplasias/patologia , Movimento Celular , Humanos , Íons/metabolismo , Metástase Neoplásica , Neoplasias/metabolismo
7.
BMC Physiol ; 12: 3, 2012 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-22449203

RESUMO

BACKGROUND: Cardiovascular toxicity is a major limiting factor in drug development and requires multiple cost-effective models to perform toxicological evaluation. Zebrafish is an excellent model for many developmental, toxicological and regenerative studies. Using approaches like morpholino knockdown and electrocardiogram, researchers have demonstrated physiological and functional similarities between zebrafish heart and human heart. The close resemblance of the genetic cascade governing heart development in zebrafish to that of humans has propelled the zebrafish system as a cost-effective model to conduct various genetic and pharmacological screens on developing embryos and larvae. The current report describes a methodology for rapid isolation of adult zebrafish heart, maintenance ex vivo, and a setup to perform quick small molecule throughput screening, including an in-house implemented analysis script. RESULTS: Adult zebrafish were anesthetized and after rapid decapitation the hearts were isolated. The short time required for isolation of hearts allows dissection of multiple fishes, thereby obtaining a large sample size. The simple protocol for ex vivo culture allowed maintaining the beating heart for several days. The in-house developed script and spectral analyses allowed the readouts to be presented either in time domain or in frequency domain. Taken together, the current report offers an efficient platform for performing cardiac drug testing and pharmacological screens. CONCLUSION: The new methodology presents a fast, cost-effective, sensitive and reliable method for performing small molecule screening. The variety of readouts that can be obtained along with the in-house developed analyses script offers a powerful setup for performing cardiac toxicity evaluation by researchers from both academics and industry.


Assuntos
Bioensaio/instrumentação , Avaliação Pré-Clínica de Medicamentos/instrumentação , Citometria de Fluxo/instrumentação , Coração/efeitos dos fármacos , Coração/fisiologia , Técnicas de Cultura de Órgãos/instrumentação , Peixe-Zebra/fisiologia , Animais , Desenho de Equipamento , Análise de Falha de Equipamento
8.
Curr Opin Neurobiol ; 18(3): 232-6, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18638551

RESUMO

The brain develops and functions in a complex ionic milieu, which is a prerequisite for neurotransmitter function and neuronal signaling. Neurotransmitters and ion fluxes are, however, important not only in neuronal signaling, but also in the control of neural differentiation, and in this review, we highlight the recent advances in our understanding of how the gamma-amino butyric acid (GABA) neurotransmitter and ion fluxes are relevant for cell cycle control and neural differentiation. Conversely, proteins previously associated with ion transport across membranes have been endowed with novel ion-independent functions, and we discuss this in the context of gap junctions in cell adhesion and of the neuron-specific K(+)-Cl(-) cotransporter KCC2 in dendritic spine development. Collectively, these findings provide a richer and more complex picture of when ion fluxes are needed in neural development and when they are not.


Assuntos
Transporte de Íons/fisiologia , Neurônios/fisiologia , Neurotransmissores/metabolismo , Transdução de Sinais/fisiologia , Animais , Ciclo Celular/fisiologia , Diferenciação Celular/fisiologia , Ácido gama-Aminobutírico/metabolismo
9.
Mol Cancer Res ; 18(10): 1522-1533, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32665429

RESUMO

Children suffering from neurologic cancers undergoing chemotherapy and radiotherapy are at high risk of reduced neurocognitive abilities likely via damage to proliferating neural stem cells (NSC). Therefore, strategies to protect NSCs are needed. We argue that induced cell-cycle arrest/quiescence in NSCs during cancer treatment can represent such a strategy. Here, we show that hyperpolarization-activated cyclic nucleotide-gated (HCN) ion channels are dynamically expressed over the cell cycle in NSCs, depolarize the membrane potential, underlie spontaneous calcium oscillations and are required to maintain NSCs in the actively proliferating pool. Hyperpolarizing pharmacologic inhibition of HCN channels during exposure to ionizing radiation protects NSCs cells in neurogenic brain regions of young mice. In contrast, brain tumor-initiating cells, which also express HCN channels, remain proliferative during HCN inhibition. IMPLICATIONS: Our finding that NSCs can be selectively rescued while cancer cells remain sensitive to the treatment, provide a foundation for reduction of cognitive impairment in children with neurologic cancers.


Assuntos
Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Neoplasias/tratamento farmacológico , Células-Tronco Neurais/metabolismo , Animais , Proliferação de Células , Humanos , Camundongos
10.
Nat Commun ; 10(1): 2110, 2019 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-31068593

RESUMO

Ribosome biogenesis is a canonical hallmark of cell growth and proliferation. Here we show that execution of Epithelial-to-Mesenchymal Transition (EMT), a migratory cellular program associated with development and tumor metastasis, is fueled by upregulation of ribosome biogenesis during G1/S arrest. This unexpected EMT feature is independent of species and initiating signal, and is accompanied by release of the repressive nucleolar chromatin remodeling complex (NoRC) from rDNA, together with recruitment of the EMT-driving transcription factor Snai1 (Snail1), RNA Polymerase I (Pol I) and the Upstream Binding Factor (UBF). EMT-associated ribosome biogenesis is also coincident with increased nucleolar recruitment of Rictor, an essential component of the EMT-promoting mammalian target of rapamycin complex 2 (mTORC2). Inhibition of rRNA synthesis in vivo differentiates primary tumors to a benign, Estrogen Receptor-alpha (ERα) positive, Rictor-negative phenotype and reduces metastasis. These findings implicate the EMT-associated ribosome biogenesis program with cellular plasticity, de-differentiation, cancer progression and metastatic disease.


Assuntos
Transição Epitelial-Mesenquimal/fisiologia , Pontos de Checagem da Fase G1 do Ciclo Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Regulação Neoplásica da Expressão Gênica , Ribossomos/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral/transplante , Movimento Celular/fisiologia , Nucléolo Celular/metabolismo , Embrião de Galinha , Proteínas Cromossômicas não Histona/metabolismo , DNA Ribossômico/metabolismo , Modelos Animais de Doenças , Feminino , Perfilação da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , RNA Ribossômico/metabolismo , Ribossomos/genética
11.
Stem Cells Dev ; 17(2): 233-43, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18447639

RESUMO

Mouse embryonic stem (ES) cells grown in feeder-free suspension cultures in the presence of leukemia inhibitory factor (LIF) and basic fibroblast growth factor (bFGF) form spheres that retain pluripotency after multiple passages. ES cell-derived spheres of any passage acquired increased competence to differentiate into neurons over time in culture. Eight-day-old spheres produced many neurons upon plating in differentiation conditions whereas 3-day-old spheres produce none, even after monolayer expansion or treatment with blockers of inhibitory signals, indicating the acquisition of a reversible, proto-neurogenic state during sphere development. Gene expression profiling with oligonucleotide microarrays was used to identify the transcriptional changes accompanying this process. Sphere growth was characterized by down-regulation of a subset of ES cell-expressed genes during the first few days of sphere formation, and progressive up-regulation of novel genes over the course of 1 week in culture. Differential gene expression between 3-day-old and 8 day-old spheres was verified by quantitative real-time PCR experiments. Gene Set Enrichment Analysis (GSEA) of microarray data indicated that neurogenic potential in the late stages of sphere development correlated predominantly with up-regulation of pathways related to mitochondrial function, cell metabolism, oxidative stress, hypoxia, and down-regulation of RNA transcription and proteasome machineries, as well as pathways induced by myc and repressed by retinoic acid. We propose that differences in cellular metabolic state brought about by cell-cell contact and paracrine interactions in the sphere niche may play crucial roles in biasing the early stages of ES cell differentiation toward a neuronal phenotype.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/fisiologia , Neurônios/fisiologia , Esferoides Celulares/fisiologia , Algoritmos , Animais , Diferenciação Celular/genética , Células Cultivadas , Análise por Conglomerados , Interpretação Estatística de Dados , Células-Tronco Embrionárias/metabolismo , Perfilação da Expressão Gênica , Camundongos , Neurônios/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Projetos de Pesquisa , Esferoides Celulares/metabolismo
12.
Biochim Biophys Acta Gene Regul Mech ; 1860(2): 175-183, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27863263

RESUMO

The Oct4 gene codes for a transcription factor that plays a critical role in the maintenance of pluripotency in embryonic and cancer stem cells. Its expression thus has to be tightly regulated. We performed biophysical characterization of the promoter region using a combination of UV absorption, CD, and NMR spectroscopies, native PAGE and chemical probing, which was followed by functional studies involving luciferase reporter assays performed in osteosarcoma and human embryonic stem cell lines. We have shown that the evolutionarily conserved G-rich region close to the Oct4 transcription start site in the non-template strand forms a parallel G-quadruplex structure. We characterized its structure and stability upon point mutations in its primary structure. Functional studies then revealed that whereas the wild type quadruplex sequence ensures high reporter gene expression, the expression of mutated variants is significantly decreased proportionally to the destabilizing effect of the mutations on the quadruplex. A ligand, N-methyl mesoporphyrin IX that increases the stability of formed quadruplex rescued the reporter expression of single-mutated variants to the level of wild-type, but it has no effect on a mutated variant that cannot form quadruplex. These data indicate that the quadruplex acts as a strong, positive regulator of Oct4 expression and as such it might serve as a potential target for therapeutic intervention.


Assuntos
Fator 3 de Transcrição de Octâmero/genética , Regiões Promotoras Genéticas/genética , Linhagem Celular Tumoral , Dicroísmo Circular/métodos , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Quadruplex G/efeitos dos fármacos , Genes Reporter/genética , Humanos , Imageamento por Ressonância Magnética/métodos , Mesoporfirinas/farmacologia , Mutação/genética , Osteossarcoma/genética , Regiões Promotoras Genéticas/efeitos dos fármacos , Sítio de Iniciação de Transcrição/efeitos dos fármacos , Sítio de Iniciação de Transcrição/fisiologia
13.
Front Pharmacol ; 8: 726, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29114221

RESUMO

Stem cells display a fundamentally different mechanism of proliferation control when compared to somatic cells. Uncovering these mechanisms would maximize the impact in drug discovery with a higher translational applicability. The unbiased approach used in phenotype-based drug discovery (PDD) programs can offer a unique opportunity to identify such novel biological phenomenon. Here, we describe an integrated phenotypic screening approach, employing a combination of in vitro and in vivo PDD models to identify a small molecule increasing stem cell proliferation. We demonstrate that a combination of both in vitro and in vivo screening models improves hit identification and reproducibility of effects across various PDD models. Using cell viability and colony size phenotype measurement we characterize the structure activity relationship of the lead molecule, and identify that the small molecule inhibits phosphorylation of ERK2 and promotes stem cell proliferation. This study demonstrates a PDD approach that employs combinatorial models to identify compounds promoting stem cell proliferation.

14.
PLoS One ; 12(12): e0188772, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29228002

RESUMO

The cell cycle coordinates core functions such as replication and cell division. However, cell-cycle-regulated transcription in the control of non-core functions, such as cell identity maintenance through specific transcription factors (TFs) and signalling pathways remains unclear. Here, we provide a resource consisting of mapped transcriptomes in unsynchronized HeLa and U2OS cancer cells sorted for cell cycle phase by Fucci reporter expression. We developed a novel algorithm for data analysis that enables efficient visualization and data comparisons and identified cell cycle synchronization of Notch signalling and TFs associated with development. Furthermore, the cell cycle synchronizes with the circadian clock, providing a possible link between developmental transcriptional networks and the cell cycle. In conclusion we find that cell cycle synchronized transcriptional patterns are temporally compartmentalized and more complex than previously anticipated, involving genes, which control cell identity and development.


Assuntos
Ciclo Celular/genética , Neoplasias/metabolismo , Fatores de Transcrição/metabolismo , Transcriptoma , Algoritmos , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Humanos , Neoplasias/genética , Neoplasias/patologia
15.
Cancer Res ; 77(7): 1741-1752, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28087597

RESUMO

Glioma-initiating cells (GIC) are considered the underlying cause of recurrences of aggressive glioblastomas, replenishing the tumor population and undermining the efficacy of conventional chemotherapy. Here we report the discovery that inhibiting T-type voltage-gated Ca2+ and KCa channels can effectively induce selective cell death of GIC and increase host survival in an orthotopic mouse model of human glioma. At present, the precise cellular pathways affected by the drugs affecting these channels are unknown. However, using cell-based assays and integrated proteomics, phosphoproteomics, and transcriptomics analyses, we identified the downstream signaling events these drugs affect. Changes in plasma membrane depolarization and elevated intracellular Na+, which compromised Na+-dependent nutrient transport, were documented. Deficits in nutrient deficit acted in turn to trigger the unfolded protein response and the amino acid response, leading ultimately to nutrient starvation and GIC cell death. Our results suggest new therapeutic targets to attack aggressive gliomas. Cancer Res; 77(7); 1741-52. ©2017 AACR.


Assuntos
Aminoácidos/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo T/fisiologia , Glioma/tratamento farmacológico , Canais de Potássio Cálcio-Ativados/antagonistas & inibidores , Resposta a Proteínas não Dobradas/efeitos dos fármacos , Animais , Transporte Biológico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Morte Celular , Linhagem Celular Tumoral , Di-Hidropiridinas/farmacologia , Glioma/metabolismo , Glioma/patologia , Humanos , Camundongos , Micotoxinas/farmacologia , Células-Tronco Neoplásicas/patologia , Proteômica , Sódio/metabolismo
16.
Springerplus ; 5: 41, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26835223

RESUMO

Pluripotent stem cells are the starting cell type of choice for the development of many cell-based regenerative therapies due to their rapid and unlimited proliferation and broad differentiation potential. The unique pluripotent cell cycle underlies both these properties. Hyperpolarization-activated cyclic nucleotide-gated cation (HCN) family channels have previously been reported to modulate mouse embryonic stem cell (ESC) proliferation and here we characterize the effects of HCN inhibitor ZD7288 on ESC proliferation and stem cell identity. The doubling time of cells treated with the HCN blocker increased by ~30 % due to longer G1 and S phases, resulting in a nearly twofold reduction in ESC numbers after 4 day serum-free culture. Slower progression through S phase was not accompanied by H2AX phosphorylation or cell stalling at transition points, although EdU incorporation in treated cells was reduced. Despite the drastic cell cycle perturbations, the pluripotent status of the cells was not compromised by treatment. Cultures treated with the HCN blocker in maintenance conditions maintained pluripotency marker expression on both RNA and protein level, although we observed a reversible effect on morphology and colony formation frequency. Addition of ZD7288 in differentiating media improved FBS-driven differentiation, but not directed differentiation to neuroectoderm, further indicating that altered cell cycle structure does not necessarily compromise pluripotency and drive ESCs to differentiation. The categorically different outcomes of ZD7288 use during differentiation indicate that cell culture context can be determinative for effects of ion-modulatory molecules and underscores the need for exploring their action in serum-free conditions demanded by potential clinical use.

17.
Oncotarget ; 7(45): 73200-73215, 2016 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-27689322

RESUMO

Glioblastoma multiforme (GBM, astrocytoma grade IV) is the most common malignant primary brain tumor in adults. Addressing the shortage of effective treatment options for this cancer, we explored repurposing of existing drugs into combinations with potent activity against GBM cells. We report that the phytoalexin pterostilbene is a potentiator of two drugs with previously reported anti-GBM activity, the EGFR inhibitor gefitinib and the antidepressant sertraline. Combinations of either of these two compounds with pterostilbene suppress cell growth, viability, sphere formation and inhibit migration in tumor GBM cell (GC) cultures. The potentiating effect of pterostilbene was observed to a varying degree across a panel of 41 patient-derived GCs, and correlated in a case specific manner with the presence of missense mutation of EGFR and PIK3CA and a focal deletion of the chromosomal region 1p32. We identify pterostilbene-induced cell cycle arrest, synergistic inhibition of MAPK activity and induction of Thioredoxin interacting protein (TXNIP) as possible mechanisms behind pterostilbene's effect. Our results highlight a nontoxic stilbenoid compound as a modulator of anticancer drug response, and indicate that pterostilbene might be used to modulate two anticancer compounds in well-defined sets of GBM patients.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Estilbenos/farmacologia , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos Fitogênicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Variações do Número de Cópias de DNA , Sinergismo Farmacológico , Feminino , Gefitinibe , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Mutação , Fenótipo , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/farmacologia , Estilbenos/uso terapêutico , Transcriptoma
18.
Brain Res ; 1051(1-2): 137-44, 2005 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-15996640

RESUMO

Mouse embryonic stem (ES) cells were transplanted into the cochlea of adult guinea pigs in order to explore their survival, differentiation, and possible integration with the host tissue. With the purpose of investigating the possible effect of manipulating the local embryonic microenvironment, ES cells were transplanted into the cochlea with or without an embryonic neuronal cograft consisting of dorsal root ganglion (DRG) tissue. To detect the survival and differentiation of ES cells, cells expressing green fluorescent protein (GFP) were used in combination with immunohistochemical detection of a neuronal marker, neural class III beta-tubulin (TUJ1 antibody). At 4 weeks following transplantation implanted ES cells were found close both to the sensory epithelium, and the spiral ganglion neurons (SGNs) with their peripheral dendritic processes projecting to the organ of Corti. There was a significant difference in the number of surviving TUJ1 (+) ES cells between the DRG cograft group and the non-cograft group (P < 0.01, ANOVA). Neurite-like projections were also identified between TUJ1-positive ES cells and the peripheral dendritic processes from SGNs. The results suggest that an embryonic neuronal microenvironment may be one of the key factors in the survival and differentiation of ES cells in the adult auditory system.


Assuntos
Diferenciação Celular/fisiologia , Cóclea/citologia , Gânglios Espinais/citologia , Sobrevivência de Enxerto , Neurônios/transplante , Transplante de Células-Tronco/métodos , Análise de Variância , Animais , Vias Auditivas/citologia , Vias Auditivas/fisiologia , Sobrevivência Celular/fisiologia , Cóclea/cirurgia , Técnicas de Cocultura , Transplante de Tecido Fetal/métodos , Gânglios Espinais/embriologia , Cobaias , Camundongos , Neurônios/citologia , Células-Tronco/citologia , Células-Tronco/fisiologia , Transplante Heterólogo
19.
Oncotarget ; 6(35): 37083-97, 2015 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-26397227

RESUMO

Radiotherapy in children causes debilitating cognitive decline, partly linked to impaired neurogenesis. Irradiation targets primarily cancer cells but also endogenous neural stem/progenitor cells (NSPCs) leading to cell death or cell cycle arrest. Here we evaluated the effects of lithium on proliferation, cell cycle and DNA damage after irradiation of young NSPCs in vitro.NSPCs were treated with 1 or 3 mM LiCl and we investigated proliferation capacity (neurosphere volume and bromodeoxyuridine (BrdU) incorporation). Using flow cytometry, we analysed apoptosis (annexin V), cell cycle (propidium iodide) and DNA damage (γH2AX) after irradiation (3.5 Gy) of lithium-treated NSPCs.Lithium increased BrdU incorporation and, dose-dependently, the number of cells in replicative phase as well as neurosphere growth. Irradiation induced cell cycle arrest in G1 and G2/M phases. Treatment with 3 mM LiCl was sufficient to increase NSPCs in S phase, boost neurosphere growth and reduce DNA damage. Lithium did not affect the levels of apoptosis, suggesting that it does not rescue NSPCs committed to apoptosis due to accumulated DNA damage.Lithium is a very promising candidate for protection of the juvenile brain from radiotherapy and for its potential to thereby improve the quality of life for those children who survive their cancer.


Assuntos
Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Hipocampo/citologia , Cloreto de Lítio/farmacologia , Células-Tronco Neurais/citologia , Neurogênese/fisiologia , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Pontos de Checagem do Ciclo Celular/efeitos da radiação , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Células Cultivadas , Radioisótopos de Cobalto , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/efeitos da radiação , Feminino , Citometria de Fluxo , Raios gama , Hipocampo/efeitos dos fármacos , Hipocampo/efeitos da radiação , Técnicas In Vitro , Cloreto de Lítio/administração & dosagem , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/efeitos da radiação , Neurogênese/efeitos dos fármacos , Neurogênese/efeitos da radiação
20.
Stem Cells Dev ; 13(4): 421-35, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15345136

RESUMO

Information concerning the development and differentiation of human embryonic stem (hES) cells in vivo is limited. The present study has focused on the in vivo outcome and differentiation of the hESC line HS181, after injection into SCID/beige mice. hES cell-derived teratomas were explored using histological evaluation and by the identification of markers for differentiated cells and tissues. The analyses identified predominant differentiation along a neuronal lineage, the formation of bone/cartilage and epithelia. Fluorescent in situ hybridization (FISH) analysis with a human-specific probe showed the teratomas to be mainly of human origin, with the most organized areas being exclusively human. Importantly, the study revealed interactions between mouse and human tissues, most notably in the formation of vessels. Both mouse and human cells contributed to specific microstructures in which mouse cells could be observed to take on the appropriate histiotypic appearance. Hence, HS181 cells were able to develop into defined mature tissues, supporting the relevant use of this hES cells model for studies of early human development, given the use of appropriate controls for host contribution. Although extensive mitotic activity implicated progenitor cell activity, no detectable multipotent or malignant areas were observed during the observation period. Persisting undifferentiated hESC were not detected.


Assuntos
Desenvolvimento Embrionário/fisiologia , Transplante de Células-Tronco , Células-Tronco/citologia , Transplante Heterólogo/fisiologia , Animais , Osso e Ossos/embriologia , Cartilagem/embriologia , Linhagem Celular , Embrião de Mamíferos , Humanos , Hibridização in Situ Fluorescente , Camundongos , Camundongos SCID
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA