Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Sci Rep ; 10(1): 6751, 2020 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-32317665

RESUMO

SOX3 is a transcription factor expressed within the developing and adult nervous system where it mostly functions to help maintain neural precursors. Sox3 is also expressed in other locations, notably within the spermatogonial stem/progenitor cell population in postnatal testis. Independent studies have shown that Sox3 null mice exhibit a spermatogenic block as young adults, the mechanism of which remains poorly understood. Using a panel of spermatogonial cell marker genes, we demonstrate that Sox3 is expressed within the committed progenitor fraction of the undifferentiated spermatogonial pool. Additionally, we use a Sox3 null mouse model to define a potential role for this factor in progenitor cell function. We demonstrate that Sox3 expression is required for transition of undifferentiated cells from a GFRα1+ self-renewing state to the NGN3 + transit-amplifying compartment. Critically, using chromatin immunoprecipitation, we demonstrate that SOX3 binds to a highly conserved region in the Ngn3 promoter region in vivo, indicating that Ngn3 is a direct target of SOX3. Together these studies indicate that SOX3 functions as a pro-commitment factor in spermatogonial stem/progenitor cells.


Assuntos
Células-Tronco Germinativas Adultas/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas do Tecido Nervoso/genética , Regiões Promotoras Genéticas , Fatores de Transcrição SOXB1/genética , Espermatogônias/metabolismo , Testículo/metabolismo , Células-Tronco Germinativas Adultas/citologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial , Masculino , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/metabolismo , Proteína com Dedos de Zinco da Leucemia Promielocítica/genética , Proteína com Dedos de Zinco da Leucemia Promielocítica/metabolismo , Ligação Proteica , Fatores de Transcrição SOXB1/deficiência , Transdução de Sinais , Espermatogênese/genética , Espermatogônias/citologia , Espermatogônias/crescimento & desenvolvimento , Testículo/citologia , Testículo/crescimento & desenvolvimento , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
2.
Theranostics ; 9(2): 424-435, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30809284

RESUMO

Tumorigenic cells, when facing a hostile environment, may enter a dormant state, leading to long-term tumor survival, relapse, and metastasis. To date, the molecular mechanism of tumor cell dormancy remains poorly understood. Methods: A soft, 3-dimentional (3D) fibrin gel culture system was used to mechanically select and grow highly malignant and tumorigenic melanoma tumor-repopulating cells (TRCs). We cultured control melanoma TRCs, TRCs with Sox2 knockdown, TRCs with Sox2 knockout, and a 2D control for in vitro and in vivo experiments. Western blotting, immunofluorescence, and flow cytometry analysis were performed to examine TRC dormancy and exit from dormancy. Results: Under a low-expression condition, we show that Sox2, a stemness molecule participates in dormancy regulation of highly tumorigenic cells that can repopulate a tumor (TRCs). Intriguingly, complete depletion of Sox2 via knockout results in dormancy exit and growth resumption of melanoma TRCs in culture and elevation of melanoma TRC apoptosis. Mice that are injected subcutaneously with Sox2-depleted melanoma TRCs do not form tumors and survive much longer than those injected with melanoma TRCs. We found that complete depletion of Sox2 promotes nuclear translocation of phosphorylated STAT3, where it binds to the p53 gene promoter, thus activating the p53-caspase3 cascade. Conclusion: These findings provide a novel insight into the role of the Sox2 gene in tumor cell stemness, tumor dormancy, and apoptosis.


Assuntos
Regulação Neoplásica da Expressão Gênica , Melanoma Experimental/patologia , Células-Tronco Neoplásicas/patologia , Fatores de Transcrição SOXB1/metabolismo , Animais , Apoptose , Proliferação de Células , Camundongos , Recidiva Local de Neoplasia , Fatores de Transcrição SOXB1/deficiência
3.
Nature ; 548(7665): 52-57, 2017 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-28746310

RESUMO

It has been proposed that the hypothalamus helps to control ageing, but the mechanisms responsible remain unclear. Here we develop several mouse models in which hypothalamic stem/progenitor cells that co-express Sox2 and Bmi1 are ablated, as we observed that ageing in mice started with a substantial loss of these hypothalamic cells. Each mouse model consistently displayed acceleration of ageing-like physiological changes or a shortened lifespan. Conversely, ageing retardation and lifespan extension were achieved in mid-aged mice that were locally implanted with healthy hypothalamic stem/progenitor cells that had been genetically engineered to survive in the ageing-related hypothalamic inflammatory microenvironment. Mechanistically, hypothalamic stem/progenitor cells contributed greatly to exosomal microRNAs (miRNAs) in the cerebrospinal fluid, and these exosomal miRNAs declined during ageing, whereas central treatment with healthy hypothalamic stem/progenitor cell-secreted exosomes led to the slowing of ageing. In conclusion, ageing speed is substantially controlled by hypothalamic stem cells, partially through the release of exosomal miRNAs.


Assuntos
Envelhecimento/genética , Envelhecimento/fisiologia , Exossomos/genética , Hipotálamo/citologia , Hipotálamo/fisiologia , Longevidade/fisiologia , MicroRNAs/genética , Células-Tronco Neurais/fisiologia , Envelhecimento/líquido cefalorraquidiano , Envelhecimento/patologia , Animais , Microambiente Celular , Exossomos/metabolismo , Hipotálamo/patologia , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Inflamação , Longevidade/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/líquido cefalorraquidiano , MicroRNAs/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/transplante , Complexo Repressor Polycomb 1/deficiência , Proteínas Proto-Oncogênicas/deficiência , Fatores de Transcrição SOXB1/deficiência , Fatores de Tempo
4.
Oncotarget ; 7(30): 47095-47110, 2016 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-27283990

RESUMO

Type II germ cell cancers (GCC) are divided into seminomas, which are highly similar to primordial germ cells and embryonal carcinomas (EC), often described as malignant counterparts to embryonic stem cells.Previously, we demonstrated that the development of GCCs is a highly plastic process and strongly influenced by the microenvironment. While orthotopic transplantation into the testis promotes seminomatous growth of the seminoma-like cell line TCam-2, ectopic xenotransplantation into the flank initiates reprogramming into an EC-like fate.During this reprogramming, BMP signaling is inhibited, leading to induction of NODAL signaling, upregulation of pluripotency factors and downregulation of seminoma markers, like SOX17. The pluripotency factor and EC-marker SOX2 is strongly induced.Here, we adressed the molecular role of SOX2 in this reprogramming. Using CRISPR/Cas9-mediated genome-editing, we established SOX2-deficient TCam-2 cells. Xenografting of SOX2-deficient cells into the flank of nude mice resulted in maintenance of a seminoma-like fate, indicated by the histology and expression of OCT3/4, SOX17, TFAP2C, PRDM1 and PRAME. In SOX2-deficient cells, BMP signaling is inhibited, but NODAL signaling is not activated. Thus, SOX2 appears to be downstream of BMP signaling but upstream of NODAL activation. So, SOX2 is an essential factor in acquiring an EC-like cell fate from seminomas.A small population of differentiated cells was identified resembling a mixed non-seminoma. Analyses of these cells revealed downregulation of the pluripotency and seminoma markers OCT3/4, SOX17, PRDM1 and TFAP2C. In contrast, the pioneer factor FOXA2 and its target genes were upregulated, suggesting that FOXA2 might play an important role in induction of non-seminomatous differentiation.


Assuntos
Carcinoma Embrionário/patologia , Fatores de Transcrição SOXB1/metabolismo , Seminoma/patologia , Animais , Carcinoma Embrionário/genética , Carcinoma Embrionário/metabolismo , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Reprogramação Celular/fisiologia , Técnicas de Inativação de Genes , Xenoenxertos , Humanos , Masculino , Camundongos , Camundongos Nus , Proteína Nodal/metabolismo , Fatores de Transcrição SOXB1/deficiência , Fatores de Transcrição SOXB1/genética , Seminoma/genética , Seminoma/metabolismo , Transfecção
5.
J Histochem Cytochem ; 63(9): 721-33, 2015 09.
Artigo em Inglês | MEDLINE | ID: mdl-26001828

RESUMO

SRY (sex determining region Y)-box 2 (SOX2) plays an important role in tumor cell metastasis and apoptosis. Laryngeal squamous cell carcinoma (LSCC), responsible for 1.5% of all cancers, is one of the most common head and neck malignancies. Accumulating evidence shows that SOX2 is overexpressed in several human tumors, including lung cancer, esophageal carcinoma, pancreatic carcinoma, breast cancer, ovarian carcinoma and glioma. Our study aimed to investigate the silencing effects of SOX2 expression using RNA interference (RNAi) on various biological processes in laryngeal cancer TU212 cells, including proliferation, apoptosis, invasion and metastasis. We also studied the involvement of the MAPK/JNK signaling pathway in the biological effects of SOX2 siRNA in TU212 cells. We found that silencing SOX2 decreased the proliferation, migration, and invasion of TU212 cells, and induced apoptosis. This effect of silencing SOX2 could be reversed by silencing MAP4K4. Therefore, we consider SOX2 as a key regulator of the upstream MAP4K4/JNK signaling pathways that could be a potential therapeutic target in the treatment of patients with or prevention of laryngeal cancer.


Assuntos
Apoptose/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Neoplasias Laríngeas/patologia , Sistema de Sinalização das MAP Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Interferência de RNA , Fatores de Transcrição SOXB1/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Células Clonais/patologia , Progressão da Doença , Regulação para Baixo/genética , Ativação Enzimática/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Neoplasias Laríngeas/genética , Invasividade Neoplásica , Metástase Neoplásica , Fosforilação/genética , RNA Interferente Pequeno/genética , Fatores de Transcrição SOXB1/deficiência
6.
PLoS One ; 10(2): e0116839, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25642713

RESUMO

Melanoma tissues and cell lines are heterogeneous, and include cells with invasive, proliferative, stem cell-like, and differentiated properties. Such heterogeneity likely contributes to the aggressiveness of the disease and resistance to therapy. One model suggests that heterogeneity arises from rare cancer stem cells (CSCs) that produce distinct cancer cell lineages. Another model suggests that heterogeneity arises through reversible cellular plasticity, or phenotype-switching. Recent work indicates that phenotype-switching may include the ability of cancer cells to dedifferentiate to a stem cell-like state. We set out to investigate the phenotype-switching capabilities of melanoma cells, and used unbiased methods to identify genes that may control such switching. We developed a system to reversibly synchronize melanoma cells between 2D-monolayer and 3D-stem cell-like growth states. Melanoma cells maintained in the stem cell-like state showed a striking upregulation of a gene set related to development and neural stem cell biology, which included SRY-box 2 (SOX2) and Inhibitor of DNA Binding 4 (ID4). A gene set related to cancer cell motility and invasiveness was concomitantly downregulated. Intense and pervasive ID4 protein expression was detected in human melanoma tissue samples, suggesting disease relevance for this protein. SiRNA knockdown of ID4 inhibited switching from monolayer to 3D-stem cell-like growth, and instead promoted switching to a highly differentiated, neuronal-like morphology. We suggest that ID4 is upregulated in melanoma as part of a stem cell-like program that facilitates further adaptive plasticity. ID4 may contribute to disease by preventing stem cell-like melanoma cells from progressing to a normal differentiated state. This interpretation is guided by the known role of ID4 as a differentiation inhibitor during normal development. The melanoma stem cell-like state may be protected by factors such as ID4, thereby potentially identifying a new therapeutic vulnerability to drive differentiation to the normal cell phenotype.


Assuntos
Diferenciação Celular , Regulação Neoplásica da Expressão Gênica , Proteínas Inibidoras de Diferenciação/metabolismo , Melanoma/metabolismo , Melanoma/patologia , Adesão Celular , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Humanos , Proteínas Inibidoras de Diferenciação/deficiência , Proteínas Inibidoras de Diferenciação/genética , Melanoma/genética , Invasividade Neoplásica , Células-Tronco Neoplásicas/patologia , Fenótipo , RNA Interferente Pequeno/genética , Fatores de Transcrição SOXB1/deficiência , Fatores de Transcrição SOXB1/genética , Transcrição Gênica
7.
Oncotarget ; 6(5): 2992-3002, 2015 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-25576924

RESUMO

Cellular reprogramming to iPSCs has uncovered unsuspected links between tumor suppressors and pluripotency factors. Using this system, it was possible to identify tumor suppressor p27 as a repressor of Sox2 during differentiation. This led to the demonstration that defects in the repression of Sox2 can contribute to tumor development. The members of the retinoblastoma family of pocket proteins, pRb, p107 and p130, are negative regulators of the cell cycle with tumor suppressor activity and with roles in differentiation. In this work we studied the relative contribution of the retinoblastoma family members to the regulation of Sox2 expression. We found that deletion of Rb or p130 leads to impaired repression of Sox2, a deffect amplified by inactivation of p53. We also identified binding of pRb and p130 to an enhancer with crucial regulatory activity on Sox2 expression. Using cellular reprogramming we tested the impact of the defective repression of Sox2 and confirmed that Rb deficiency allows the generation of iPSCs in the absence of exogenous Sox2. Finally, partial depletion of Sox2 positive cells reduced the pituitary tumor development initiated by Rb loss in vivo. In summary, our results show that Sox2 repression by pRb is a relevant mechanism of tumor suppression.


Assuntos
Células-Tronco Pluripotentes Induzidas/metabolismo , Proteína do Retinoblastoma/metabolismo , Proteína p130 Retinoblastoma-Like/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Transcrição Gênica , Animais , Reprogramação Celular , Epigênese Genética , Regulação da Expressão Gênica no Desenvolvimento , Regulação Neoplásica da Expressão Gênica , Genótipo , Células HEK293 , Humanos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco Neoplásicas/metabolismo , Fenótipo , Neoplasias Hipofisárias/genética , Neoplasias Hipofisárias/metabolismo , Interferência de RNA , Proteína do Retinoblastoma/deficiência , Proteína do Retinoblastoma/genética , Proteína p107 Retinoblastoma-Like/genética , Proteína p107 Retinoblastoma-Like/metabolismo , Proteína p130 Retinoblastoma-Like/deficiência , Proteína p130 Retinoblastoma-Like/genética , Fatores de Transcrição SOXB1/deficiência , Fatores de Transcrição SOXB1/genética , Transfecção , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
8.
J Clin Invest ; 122(10): 3635-46, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22945632

RESUMO

Sex-determining region Y (SRY) box 2 (SOX2) haploinsufficiency causes a form of hypopituitarism in humans that is characterized by gonadotrophin deficiency known as hypogonadotrophic hypogonadism. Here, we conditionally deleted Sox2 in mice to investigate the pathogenesis of hypogonadotrophic hypogonadism. First, we found that absence of SOX2 in the developing Rathke pouch of conditional embryos led to severe anterior lobe hypoplasia with drastically reduced expression of the pituitary-specific transcription factor POU class 1 homeobox 1 (POU1F1) as well as severe disruption of somatotroph and thyrotroph differentiation. In contrast, corticotrophs, rostral-tip POU1F1-independent thyrotrophs, and, interestingly, lactotrophs and gonadotrophs were less affected. Second, we identified a requirement for SOX2 in normal proliferation of periluminal progenitors; in its absence, insufficient precursors were available to produce all cell lineages of the anterior pituitary. Differentiated cells derived from precursors exiting cell cycle at early stages, including corticotrophs, rostral-tip thyrotrophs, and gonadotrophs, were generated, while hormone-producing cells originating from late-born precursors, such as somatotrophs and POU1F1-dependent thyrotrophs, were severely reduced. Finally, we found that 2 previously characterized patients with SOX2 haploinsufficiency and associated hypogonadotrophic hypogonadism had a measurable response to gonadotropin-releasing hormone (GnRH) stimulation, suggesting that it is not the absence of gonadotroph differentiation, but rather the deficient hypothalamic stimulation of gonadotrophs, that underlies typical hypogonadotrophic hypogonadism.


Assuntos
Hipogonadismo/genética , Sistema Hipotálamo-Hipofisário/fisiologia , Fatores de Transcrição SOXB1/fisiologia , Animais , Diferenciação Celular , Linhagem da Célula , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Hormônio Liberador de Gonadotropina/uso terapêutico , Heterozigoto , Proteínas de Homeodomínio/genética , Humanos , Hipogonadismo/tratamento farmacológico , Hipogonadismo/fisiopatologia , Camundongos , Camundongos Knockout , Organogênese/genética , Organogênese/fisiologia , Adeno-Hipófise/anormalidades , Adeno-Hipófise/embriologia , Adeno-Hipófise/metabolismo , Adeno-Hipófise/patologia , Proteínas Repressoras/genética , Fatores de Transcrição SOXB1/deficiência , Fatores de Transcrição SOXB1/genética , Somatotrofos/patologia , Tireotrofos/patologia , Fator de Transcrição Pit-1/deficiência
9.
PLoS One ; 7(8): e43628, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22928007

RESUMO

The cancer stem cell (CSC) model posits the presence of a small number of CSCs in the heterogeneous cancer cell population that are ultimately responsible for tumor initiation, as well as cancer recurrence and metastasis. CSCs have been isolated from a variety of human cancers and are able to generate a hierarchical and heterogeneous cancer cell population. CSCs are also resistant to conventional chemo- and radio-therapies. Here we report that ionizing radiation can induce stem cell-like properties in heterogeneous cancer cells. Exposure of non-stem cancer cells to ionizing radiation enhanced spherogenesis, and this was accompanied by upregulation of the pluripotency genes Sox2 and Oct3/4. Knockdown of Sox2 or Oct3/4 inhibited radiation-induced spherogenesis and increased cellular sensitivity to radiation. These data demonstrate that ionizing radiation can activate stemness pathways in heterogeneous cancer cells, resulting in the enrichment of a CSC subpopulation with higher resistance to radiotherapy.


Assuntos
Raios gama , Células-Tronco Neoplásicas/efeitos da radiação , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Técnicas de Silenciamento de Genes , Células Hep G2 , Humanos , Células-Tronco Neoplásicas/metabolismo , Fator 3 de Transcrição de Octâmero/deficiência , Fator 3 de Transcrição de Octâmero/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição SOXB1/deficiência , Fatores de Transcrição SOXB1/genética
10.
PLoS One ; 7(8): e41335, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22912670

RESUMO

SOX2 is an important stem cell marker and plays important roles in development and carcinogenesis. However, the role of SOX2 in Epithelial-Mesenchymal Transition has not been investigated. We demonstrated, for the first time, that SOX2 is involved in the Epithelial-Mesenchymal Transition (EMT) process as knock downof SOX2 in colorectal cancer (CRC) SW620 cells induced a Mesenchymal-Epithelial Transition (MET) process with recognized changes in the expression of key genes involved in the EMT process including E-cadherin and vimentin. In addition, we provided a link between SOX2 activity and the WNT pathway by showing that knock down of SOX2 reduced the WNT pathway activity in colorectal cancer (CRC) cells. We further demonstrated that SOX2 is involved in cell migration and invasion in vitro and in metastasis in vivo for CRC cells, and that the process might be mediated through the MMP2 activity. Finally, an IHC analysis of 44 cases of colorectal cancer patients suggested that SOX2 is a prognosis marker for metastasis of colorectal cancers.


Assuntos
Neoplasias Colorretais/patologia , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica/genética , Inativação Gênica , Neoplasias Hepáticas/secundário , Fatores de Transcrição SOXB1/deficiência , Fatores de Transcrição SOXB1/genética , Animais , Linhagem Celular Tumoral , Movimento Celular/genética , Neoplasias Colorretais/genética , Regulação para Baixo/genética , Feminino , Humanos , Metástase Linfática , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Invasividade Neoplásica , Transdução de Sinais/genética , Fatores de Transcrição TCF/metabolismo , beta Catenina/metabolismo
11.
Mech Dev ; 129(1-4): 1-12, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22522080

RESUMO

Haploinsufficiency for the HMG-box transcription factor SOX2 results in abnormalities of the human ventral forebrain and its derivative structures. These defects include anophthalmia (absence of eye), microphthalmia (small eye) and hypothalamic hamartoma (HH), an overgrowth of the ventral hypothalamus. To determine how Sox2 deficiency affects the morphogenesis of the ventral diencephalon and eye, we generated a Sox2 allelic series (Sox2(IR), Sox2(LP), and Sox2(EGFP)), allowing for the generation of mice that express germline hypomorphic levels (<40%) of SOX2 protein and that faithfully recapitulate SOX2 haploinsufficient human phenotypes. We find that Sox2 hypomorphism significantly disrupts the development of the posterior hypothalamus, resulting in an ectopic protuberance of the prechordal floor, an upregulation of Shh signaling, and abnormal hypothalamic patterning. In the anterior diencephalon, both the optic stalks and optic cups (OC) of Sox2 hypomorphic (Sox2(HYP)) embryos are malformed. Furthermore, Sox2(HYP) eyes exhibit a loss of neural potential and coloboma, a common phenotype in SOX2 haploinsufficient humans that has not been described in a mouse model of SOX2 deficiency. These results establish for the first time that germline Sox2 hypomorphism disrupts the morphogenesis and patterning of the hypothalamus, optic stalk, and the early OC, establishing a model of the development of the abnormalities that are observed in SOX2 haploinsufficient humans.


Assuntos
Anormalidades do Olho/genética , Hipotálamo/anormalidades , Fatores de Transcrição SOXB1/genética , Animais , Diencéfalo/anormalidades , Diencéfalo/patologia , Modelos Animais de Doenças , Embrião de Mamíferos/anormalidades , Embrião de Mamíferos/patologia , Haploinsuficiência , Humanos , Hipotálamo/patologia , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica de Varredura , Especificidade de Órgãos , Fatores de Transcrição SOXB1/deficiência
12.
Stem Cells ; 30(6): 1064-75, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22415968

RESUMO

Glioblastoma multiforme (GBM) ranks among the deadliest types of cancer and given these new therapies are urgently needed. To identify molecular targets, we queried a microarray profiling 467 human GBMs and discovered that polo-like kinase 1 (PLK1) was highly expressed in these tumors and that it clustered with the proliferative subtype. Patients with PLK1-high tumors were more likely to die from their disease suggesting that current therapies are inactive against such tumors. This prompted us to examine its expression in brain tumor initiating cells (BTICs) given their association with treatment failure. BTICs isolated from patients expressed 110-470 times more PLK1 than normal human astrocytes. Moreover, BTICs rely on PLK1 for survival because the PLK1 inhibitor BI2536 inhibited their growth in tumorsphere cultures. PLK1 inhibition suppressed growth, caused G(2) /M arrest, induced apoptosis, and reduced the expression of SOX2, a marker of neural stem cells, in SF188 cells. Consistent with SOX2 inhibition, the loss of PLK1 activity caused the cells to differentiate based on elevated levels of glial fibrillary acidic protein and changes in cellular morphology. We then knocked glial fibrillary acidic protein (GFAP) down SOX2 with siRNA and showed that it too inhibited cell growth and induced cell death. Likewise, in U251 cells, PLK1 inhibition suppressed cell growth, downregulated SOX2, and induced cell death. Furthermore, BI2536 delayed tumor growth of U251 cells in an orthotopic brain tumor model, demonstrating that the drug is active against GBM. In conclusion, PLK1 level is elevated in GBM and its inhibition restricts the growth of brain cancer cells.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Proteínas de Ciclo Celular/antagonistas & inibidores , Glioblastoma/tratamento farmacológico , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Pteridinas/farmacologia , Fatores de Transcrição SOXB1/deficiência , Animais , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Progressão da Doença , Glioblastoma/enzimologia , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Camundongos , Terapia de Alvo Molecular , Células-Tronco Neurais , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Análise de Sobrevida , Transfecção , Quinase 1 Polo-Like
13.
J Cell Sci ; 125(Pt 4): 932-42, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22421361

RESUMO

Wnt-ß-catenin signaling regulates cell fate during organ development and postnatal tissue maintenance, but its contribution to specification of distinct lung epithelial lineages is still unclear. To address this question, we used a Cre recombinase (Cre)-LoxP approach to activate canonical Wnt signaling ectopically in developing lung endoderm. We found that persistent activation of canonical Wnt signaling within distal lung endoderm was permissive for normal development of alveolar epithelium, yet led to the loss of developing bronchiolar epithelium and ectasis of distal conducting airways. Activation of canonical Wnt led to ectopic expression of a lymphoid-enhancing factor and a T-cell factor (LEF and TCF, respectively) and absence of SRY (sex-determining region Y)-box 2 (SOX2) and tumor protein p63 (p63) expression in proximal derivatives. Conditional loss of SOX2 in airways phenocopied epithelial differentiation defects observed with ectopic activation of canonical Wnt. Our data suggest that Wnt negatively regulates a SOX2-dependent signaling program required for developmental progression of the bronchiolar lineage.


Assuntos
Células Epiteliais/citologia , Células Epiteliais/metabolismo , Pulmão/citologia , Fatores de Transcrição SOXB1/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo , Animais , Apoptose , Bronquíolos/citologia , Bronquíolos/metabolismo , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Endoderma/metabolismo , Feminino , Regulação da Expressão Gênica , Genes Reporter , Pulmão/metabolismo , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Fosfoproteínas/metabolismo , Estabilidade Proteica , Alvéolos Pulmonares/citologia , Alvéolos Pulmonares/metabolismo , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição SOXB1/deficiência , Fatores de Transcrição TCF/metabolismo , Transativadores/metabolismo , Transcrição Gênica , Proteínas Wnt/metabolismo , beta Catenina/biossíntese , beta Catenina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA