Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int J Cancer ; 136(5): E455-69, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25227893

RESUMO

Inherent or acquired drug resistance is a major contributor to epithelial ovarian cancer (EOC) mortality. Novel drugs or drug combinations that produce EOC cell death or resensitize drug resistant cells to standard chemotherapy may improve patient treatment. After conducting drug tolerability studies for the multikinase inhibitors dorsomorphin (DM) and it is structural analogue LDN-193189 (LDN), these drugs were tested in a mouse intraperitoneal xenograft model of EOC. DM significantly increased survival, whereas LDN showed a trend toward increased survival. In vitro experiments using cisplatin (CP)-resistant EOC cell lines, A2780-cp or SKOV3, we determined that pretreatment or cotreatment with DM or LDN resensitized cells to the killing effect of CP or carboplatin (CB). DM was capable of blocking EOC cell cycle and migration, whereas LDN produced a less pronounced effect on cell cycle and no effect on migration. Subsequent analyses using primary human EOC cell samples or additional established EOC cells lines showed that DM or LDN induced a dose-dependent autophagic or cell death response, respectively. DM induced a characteristic morphological change with the appearance of numerous LC3B-containing acidic vacuoles and an increase in LC3BII levels. This was coincident with a decrease in cell growth and the altered cell cycle consistent with DM-induced cytostasis. By contrast, LDN produced a caspase 3-independent, reactive oxygen species-dependent cell death. Overall, DM and LDN possess drug characteristics suitable for adjuvant agents used to treat chemotherapy-sensitive and -resistant EOC.


Assuntos
Adenocarcinoma/tratamento farmacológico , Cistadenocarcinoma Seroso/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Neoplasias Epiteliais e Glandulares/tratamento farmacológico , Neoplasias Ovarianas/tratamento farmacológico , Pirazóis/farmacologia , Pirimidinas/farmacologia , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Adenocarcinoma/patologia , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Carcinoma Epitelial do Ovário , Ciclo Celular/efeitos dos fármacos , Proliferação de Células , Cistadenocarcinoma Seroso/patologia , Feminino , Humanos , Técnicas Imunoenzimáticas , Camundongos , Gradação de Tumores , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Ovarianas/patologia , Células Tumorais Cultivadas , Cicatrização/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Blood ; 122(7): 1162-73, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23733337

RESUMO

Notch signaling regulates several cellular processes including cell fate decisions and proliferation in both invertebrates and mice. However, comparatively less is known about the role of Notch during early human development. Here, we examined the function of Notch signaling during hematopoietic lineage specification from human pluripotent stem cells of both embryonic and adult fibroblast origin. Using immobilized Notch ligands and small interfering RNA to Notch receptors we have demonstrated that Notch1, but not Notch2, activation induced hairy and enhancer of split 1 (HES1) expression and generation of committed hematopoietic progenitors. Using gain- and loss-of-function approaches, this was shown to be attributed to Notch-signaling regulation through HES1, which dictated cell fate decisions from bipotent precursors either to the endothelial or hematopoietic lineages at the clonal level. Our study reveals a previously unappreciated role for the Notch pathway during early human hematopoiesis, whereby Notch signaling via HES1 represents a toggle switch of hematopoietic vs endothelial fate specification.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Células-Tronco Embrionárias/citologia , Endotélio Vascular/citologia , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/citologia , Proteínas de Homeodomínio/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Receptor Notch1/metabolismo , Apoptose , Fatores de Transcrição Hélice-Alça-Hélice Básicos/antagonistas & inibidores , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Biomarcadores/metabolismo , Western Blotting , Diferenciação Celular , Movimento Celular , Proliferação de Células , Células Cultivadas , Derme/citologia , Derme/metabolismo , Células-Tronco Embrionárias/metabolismo , Endotélio Vascular/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Citometria de Fluxo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Proteínas de Homeodomínio/antagonistas & inibidores , Proteínas de Homeodomínio/genética , Humanos , Técnicas Imunoenzimáticas , Células-Tronco Pluripotentes Induzidas/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , RNA Interferente Pequeno/genética , Receptor Notch1/antagonistas & inibidores , Receptor Notch1/genética , Receptores Notch/metabolismo , Transdução de Sinais , Fatores de Transcrição HES-1
3.
Stem Cells ; 30(3): 392-404, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22213600

RESUMO

Recent studies have identified gene signatures in malignant tumors that are associated with human embryonic stem cells, suggesting a molecular relationship between aggressive cancers and pluripotency. Here, we characterize neural precursors (NPs) derived from transformed human embryonic stem cells (N-t-hESCs) that exhibit neoplastic features of human brain tumors. NPs derived from t-hESCs have enhanced cell proliferation and an inability to mature toward the astrocytic lineage, compared with progeny derived from normal human embryonic stem cells (N-hESCs) independent of adherent or neurosphere outgrowth. Intracranial transplantation of NPs derived from N-t-hESCs and N-hESCs into NOD SCID mice revealed development of neuroectoderm tumors exclusively from the N-t-hESCs NPs and not from normal N-hESCs. These tumors infiltrated the ventricles and the cerebellum of recipient mice and displayed morphological, phenotypic, and molecular features associated with classic medulloblastoma including retention of a pluripotent signature. Importantly, N-t-hESCs did not exhibit cytogenetic changes associated with medulloblastoma, suggesting that aberrant cellular and molecular properties precede the acquisition of karyotypic changes thus underscoring the value of this model system of human medulloblastoma. Our study demonstrates that NPs from a starting population of neoplastic human pluripotent parent cells possess brain tumor-initiating cell capacity, thereby providing a model system to investigate initiation and progression of primitive human neural cancers that are difficult to assess using somatic sources.


Assuntos
Neoplasias Encefálicas/patologia , Células-Tronco Embrionárias/patologia , Células-Tronco Pluripotentes/patologia , Animais , Astrócitos/patologia , Diferenciação Celular , Proliferação de Células , Transformação Celular Neoplásica , Células Cultivadas , Modelos Animais de Doenças , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/transplante , Perfilação da Expressão Gênica , Humanos , Meduloblastoma/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Transplante de Neoplasias , Tumores Neuroectodérmicos Primitivos/patologia , Fenótipo , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/transplante
4.
Cell Death Discov ; 9(1): 347, 2023 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-37726268

RESUMO

Sonic Hedgehog (SHH) medulloblastomas (MBs) exhibit an intermediate prognosis and extensive intertumoral heterogeneity. While SHH pathway antagonists are effective in post-pubertal patients, younger patients exhibit significant side effects, and tumors that harbor mutations in downstream SHH pathway genes will be drug resistant. Thus, novel targeted therapies are needed. Here, we performed preclinical testing of the potent MEK inhibitor (MEKi) trametinib on tumor properties across 2 human and 3 mouse SHH MB models in vitro and in 3 orthotopic MB xenograft models in vivo. Trametinib significantly reduces tumorsphere size, stem/progenitor cell proliferation, viability, and migration. RNA-sequencing on human and mouse trametinib treated cells corroborated these findings with decreased expression of cell cycle, stem cell pathways and SHH-pathway related genes concomitant with increases in genes associated with cell death and ciliopathies. Importantly, trametinib also decreases tumor growth and increases survival in vivo. Cell cycle related E2F target gene sets are significantly enriched for genes that are commonly downregulated in both trametinib treated tumorspheres and primary xenografts. However, IL6/JAK STAT3 and TNFα/NFκB signaling gene sets are specifically upregulated following trametinib treatment in vivo indicative of compensatory molecular changes following long-term MEK inhibition. Our study reveals a novel role for trametinib in effectively attenuating SHH MB tumor progression and warrants further investigation of this potent MEK1/2 inhibitor either alone or in combination with other targeted therapies for the treatment of SHH MB exhibiting elevated MAPK pathway activity.

5.
Nat Commun ; 14(1): 2502, 2023 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-37130865

RESUMO

Group 3 medulloblastoma (G3 MB) carries the worst prognosis of all MB subgroups. MYC oncoprotein is elevated in G3 MB tumors; however, the mechanisms that support MYC abundance remain unclear. Using metabolic and mechanistic profiling, we pinpoint a role for mitochondrial metabolism in regulating MYC. Complex-I inhibition decreases MYC abundance in G3 MB, attenuates the expression of MYC-downstream targets, induces differentiation, and prolongs male animal survival. Mechanistically, complex-I inhibition increases inactivating acetylation of antioxidant enzyme SOD2 at K68 and K122, triggering the accumulation of mitochondrial reactive oxygen species that promotes MYC oxidation and degradation in a mitochondrial pyruvate carrier (MPC)-dependent manner. MPC inhibition blocks the acetylation of SOD2 and oxidation of MYC, restoring MYC abundance and self-renewal capacity in G3 MB cells following complex-I inhibition. Identification of this MPC-SOD2 signaling axis reveals a role for metabolism in regulating MYC protein abundance that has clinical implications for treating G3 MB.


Assuntos
Neoplasias Cerebelares , Meduloblastoma , Animais , Masculino , Transportadores de Ácidos Monocarboxílicos , Meduloblastoma/patologia , Neoplasias Cerebelares/patologia , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo
6.
FEBS J ; 289(7): 1765-1778, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-33714236

RESUMO

The cancer stem cell (CSC) model posits that tumors contain subpopulations that display defining features of normal stem cells including self-renewal capacity and differentiation. Tumor cells exhibiting these features are now considered to be responsible for tumor propagation and drug resistance in a wide variety of cancers. Therefore, the identification of robust CSC markers and characterization of CSC-specific molecular signatures may lead to the identification of novel therapeutics that selectively abolish this clinically relevant cell population while preserving normal tissue. Brain tumor researchers have been at the forefront of the CSC field. From initial in vitro cell sorting experiments to the sophisticated bioinformatic technologies that now exquisitely resolve primary brain tumors at a single-cell level, recent glioma and medulloblastoma (MB) studies have integrated developmental state with genomic and transcriptome data to identify the spectrum of cell types that may drive tumor progression. This review will examine the last two decades of CSC studies in the field. Seminal discoveries, emerging controversies, and outstanding questions will be covered with a particular focus on MB, the most common malignant primary brain tumor in children.


Assuntos
Neoplasias Cerebelares , Meduloblastoma , Movimento Celular , Neoplasias Cerebelares/genética , Neoplasias Cerebelares/metabolismo , Neoplasias Cerebelares/patologia , Criança , Humanos , Meduloblastoma/genética , Meduloblastoma/metabolismo , Meduloblastoma/patologia , Células-Tronco Neoplásicas/patologia , Transcriptoma
7.
Commun Biol ; 5(1): 697, 2022 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-35835937

RESUMO

Medulloblastoma (MB) is the most common primary malignant pediatric brain cancer. We recently identified novel roles for the MEK/MAPK pathway in regulating human Sonic Hedgehog (SHH) MB tumorigenesis. The MEK inhibitor, selumetinib, decreased SHH MB growth while extending survival in mouse models. However, the treated mice ultimately succumbed to disease progression. Here, we perform RNA sequencing on selumetinib-treated orthotopic xenografts to identify molecular pathways that compensate for MEK inhibition specifically in vivo. Notably, the JAK/STAT3 pathway exhibits increased activation in selumetinib-treated tumors. The combination of selumetinib and the JAK/STAT3 pathway inhibitor, pacritinib, further reduces growth in two xenograft models and also enhances survival. Multiplex spatial profiling of proteins in drug-treated xenografts reveals shifted molecular dependencies and compensatory changes following combination drug treatment. Our study warrants further investigation into MEK and JAK/STAT3 inhibition as a novel combinatory therapeutic strategy for SHH MB.


Assuntos
Neoplasias Cerebelares , Meduloblastoma , Animais , Neoplasias Cerebelares/tratamento farmacológico , Neoplasias Cerebelares/genética , Criança , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Meduloblastoma/tratamento farmacológico , Meduloblastoma/genética , Meduloblastoma/metabolismo , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo
8.
Neuro Oncol ; 23(11): 1845-1858, 2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34302487

RESUMO

BACKGROUND: Neuro-oncology has grown tremendously since 2010, marked by increasing society membership, specialized clinical expertise, and new journals. Yet, modest improvement in racial/ethnic diversity amongst clinical trial participants, researchers, and clinicians led us to conduct a survey to identify opportunities to enhance diversity and inclusiveness amongst neuro-oncology professionals. METHODS: In summer 2020, the Women and Diversity Committee of the Society for Neuro-Oncology (SNO) distributed an anonymous online survey to members and affiliates including the European Association of Neuro-Oncology (EANO), Asian Society for Neuro-Oncology (ASNO), Society for Neuro-Oncology Latin America (SNOLA) and Society for Neuro-Oncology Sub-Saharan Africa (SNOSSA). The survey captured personal and professional characteristics, biases, effective mentorship qualities, career service metrics, and suggested field/society changes. Results were analyzed by geography, profession, age, racial/ethnic, and sexual identity. Standard descriptive statistics characterized the study population. RESULTS: The 386 respondents were predominantly female (58%) with a median age range of 40-49 years (31%), White (65%), and SNO members (97%). Most worked in North America (77%) in a research profession (67%). A majority of White respondents reported never experiencing biases (64%), while the majority of non-White respondents reported unconscious biases/microaggressions, followed by a lack of/limited mentorship. Qualitative assessments showcased that personal/professional success metrics were linked to needed improvements in diversity and inclusion efforts within the neuro-oncology field. CONCLUSIONS: The prevalence of racial/ethnic biases and poor mentorship rates amongst underrepresented groups in neuro-oncology is high and potentially linked to the limited diverse representation amongst members and affiliates. These findings warrant a swift implementation of equity and inclusion practices within the neuro-oncology field.


Assuntos
Benchmarking , Oncologia , Adulto , Etnicidade , Feminino , Humanos , Pessoa de Meia-Idade , Sociedades , Inquéritos e Questionários
9.
Nat Commun ; 11(1): 3627, 2020 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-32686664

RESUMO

OTX2 is a potent oncogene that promotes tumor growth in Group 3 medulloblastoma. However, the mechanisms by which OTX2 represses neural differentiation are not well characterized. Here, we perform extensive multiomic analyses to identify an OTX2 regulatory network that controls Group 3 medulloblastoma cell fate. OTX2 silencing modulates the repressive chromatin landscape, decreases levels of PRC2 complex genes and increases the expression of neurodevelopmental transcription factors including PAX3 and PAX6. Expression of PAX3 and PAX6 is significantly lower in Group 3 medulloblastoma patients and is correlated with reduced survival, yet only PAX3 inhibits self-renewal in vitro and increases survival in vivo. Single cell RNA sequencing of Group 3 medulloblastoma tumorspheres demonstrates expression of an undifferentiated progenitor program observed in primary tumors and characterized by translation/elongation factor genes. Identification of mTORC1 signaling as a downstream effector of OTX2-PAX3 reveals roles for protein synthesis pathways in regulating Group 3 medulloblastoma pathogenesis.


Assuntos
Carcinogênese/genética , Neoplasias Cerebelares , Meduloblastoma , Fatores de Transcrição Otx/metabolismo , Fator de Transcrição PAX3/genética , Animais , Carcinogênese/metabolismo , Diferenciação Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Neoplasias Cerebelares/genética , Neoplasias Cerebelares/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Meduloblastoma/genética , Meduloblastoma/metabolismo , Células-Tronco Neoplásicas/metabolismo , Oncogenes , Fator de Transcrição PAX3/metabolismo , Fator de Transcrição PAX6/genética , Fator de Transcrição PAX6/metabolismo , Transdução de Sinais/genética
10.
Trends Mol Med ; 14(8): 323-32, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18635398

RESUMO

Although the cancer stem cell (CSC) hypothesis has become an attractive model to account for tumor recurrence, failure to define a cell of origin has created the need to explore alternative models for cancer initiation and maintenance. Recent studies have linked an embryonic stem cell (ESC)-like gene signature with poorly defined high-grade tumors. Here, we review advances in the ESC field with an emphasis on how human pluripotent stem cells (hPSCs) can be used to define early tumorigenic events, including potential miRNA and epigenetic targets, as well as proto-oncogene and tumor suppressor networks that might facilitate hierarchal transformation. These studies allow for investigation of cancer initiation in a manner that cannot be achieved using primary tumors, where only retrospective evaluation of CSC development is possible. By comparing transformed hPSCs with their normal counterparts, we hope to develop novel cell-specific therapies that selectively target CSCs.


Assuntos
Transformação Celular Neoplásica , Neoplasias/etiologia , Neoplasias/fisiopatologia , Células-Tronco Pluripotentes/fisiologia , Progressão da Doença , Células-Tronco de Carcinoma Embrionário , Epigênese Genética , Genes Supressores de Tumor , Terapia Genética , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Neoplasias/patologia , Neoplasias/terapia , Neoplasias Embrionárias de Células Germinativas/patologia , Neoplasias Embrionárias de Células Germinativas/fisiopatologia , Células-Tronco Neoplásicas/patologia , Células-Tronco Neoplásicas/fisiologia , Oncogenes , Proto-Oncogene Mas
11.
Methods Mol Biol ; 1869: 127-142, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30324520

RESUMO

Utilization of human embryonic stem cells (hESCs) as a model system to study highly malignant pediatric cancers has led to significant insight into the molecular mechanisms governing tumor progression and has revealed novel therapeutic targets for these devastating diseases. Here, we describe a method for generating heterogeneous populations of neural precursors from both normal and neoplastic hESCs and the subsequent injection of neoplastic human embryonic neural cells (hENs) into intracerebellar or intracranial xenograft models. Histopathologically, neural tumors derived from neoplastic hENs exhibit features similar to more aggressive medulloblastoma, the most common malignant primary pediatric brain tumor. In this chapter, we will outline the detailed methods for culturing normal and neoplastic neural precursor cells in both adherent and tumorsphere format and the full characterization of the brain tumors generated from these cells in non-obese diabetic severe combined immunodeficiency (NOD SCID) mice.


Assuntos
Neoplasias Encefálicas/patologia , Células-Tronco Embrionárias Humanas/patologia , Modelos Biológicos , Animais , Bovinos , Ensaios de Migração Celular , Humanos , Lentivirus/metabolismo , Camundongos , Células-Tronco Neurais/patologia , Células-Tronco Neurais/transplante , Perfusão , Esferoides Celulares/patologia , Fixação de Tecidos , Transdução Genética
12.
Cancer Res ; 66(3): 1464-72, 2006 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-16452202

RESUMO

Malignant central nervous system (CNS) tumors, such as glioblastoma multiforme, invade the brain and disrupt normal tissue architecture, making complete surgical removal virtually impossible. Here, we have developed and optimized a purification strategy to isolate and identify natural inhibitors of glioma cell invasion in a three-dimensional collagen type I matrix. Inter alpha-trypsin inhibitor heavy chain 2 (ITI H2) was identified from the most inhibitory fractions and its presence was confirmed both as a single protein and in a bikunin-bound form. Stable overexpression in U251 glioma cells validated ITI H2's strong inhibition of human glioma cell invasion together with significant inhibition of cell proliferation and promotion of cell-cell adhesion. Analysis of primary human brain tumors showed significantly higher levels of ITI H2 in normal brain and low-grade tumors compared with high-grade gliomas, indicating an inverse correlation with malignancy. The phosphatidylinositol 3-kinase/Akt signaling cascade seemed to be one of the pathways involved in the effect of ITI H2 on U251 cells. These findings suggest that reduction of ITI H2 expression correlates with brain tumor progression and that targeting factors responsible for its loss or restoring the ITI supply exogenously may serve as potential therapeutic strategies for a variety of CNS tumors.


Assuntos
alfa-Globulinas/isolamento & purificação , Neoplasias Encefálicas/química , Glioma/química , alfa-Globulinas/genética , alfa-Globulinas/metabolismo , alfa-Globulinas/fisiologia , Animais , Western Blotting , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Adesão Celular/fisiologia , Movimento Celular/fisiologia , Regulação para Baixo , Glioma/metabolismo , Glioma/patologia , Humanos , Imuno-Histoquímica , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/genética , Camundongos , Invasividade Neoplásica , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Inibidor da Tripsina de Soja de Kunitz/biossíntese , Inibidor da Tripsina de Soja de Kunitz/genética
13.
Mol Oncol ; 12(4): 495-513, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29377567

RESUMO

Medulloblastoma (MB) is the most common malignant primary pediatric brain cancer. Among the most aggressive subtypes, Group 3 and Group 4 originate from stem/progenitor cells, frequently metastasize, and often display the worst prognosis, yet we know the least about the molecular mechanisms driving their progression. Here, we show that the transcription factor orthodenticle homeobox 2 (OTX2) promotes self-renewal while inhibiting differentiation in vitro and increases tumor initiation from MB stem/progenitor cells in vivo. To determine how OTX2 contributes to these processes, we employed complementary bioinformatic approaches to characterize the OTX2 regulatory network and identified novel relationships between OTX2 and genes associated with neuronal differentiation and axon guidance signaling in Group 3 and Group 4 MB stem/progenitor cells. In particular, OTX2 levels were negatively correlated with semaphorin (SEMA) signaling, as expression of 9 SEMA pathway genes is upregulated following OTX2 knockdown with some being potential direct OTX2 targets. Importantly, this negative correlation was also observed in patient samples, with lower expression of SEMA4D associated with poor outcome specifically in Group 4 tumors. Functional proof-of-principle studies demonstrated that increased levels of select SEMA pathway genes are associated with decreased self-renewal and growth in vitro and in vivo and that RHO signaling, known to mediate the effects of SEMA genes, is contributing to the OTX2 KD phenotype. Our study provides mechanistic insight into the networks controlled by OTX2 in MB stem/progenitor cells and reveals novel roles for axon guidance genes and their downstream effectors as putative tumor suppressors in MB.


Assuntos
Neoplasias Encefálicas/metabolismo , Regulação Neoplásica da Expressão Gênica , Meduloblastoma/metabolismo , Proteínas de Neoplasias/metabolismo , Células-Tronco Neoplásicas/metabolismo , Fatores de Transcrição Otx/metabolismo , Transdução de Sinais , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Humanos , Meduloblastoma/genética , Meduloblastoma/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas de Neoplasias/genética , Células-Tronco Neoplásicas/patologia , Fatores de Transcrição Otx/genética
14.
Cancer Res ; 78(16): 4745-4759, 2018 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-29930101

RESUMO

The extensive heterogeneity both between and within the medulloblastoma subgroups underscores a critical need for variant-specific biomarkers and therapeutic strategies. We previously identified a role for the CD271/p75 neurotrophin receptor (p75NTR) in regulating stem/progenitor cells in the SHH medulloblastoma subgroup. Here, we demonstrate the utility of CD271 as a novel diagnostic and prognostic marker for SHH medulloblastoma using IHC analysis and transcriptome data across 763 primary tumors. RNA sequencing of CD271+ and CD271- cells revealed molecularly distinct, coexisting cellular subsets, both in vitro and in vivo MAPK/ERK signaling was upregulated in the CD271+ population, and inhibiting this pathway reduced endogenous CD271 levels, stem/progenitor cell proliferation, and cell survival as well as cell migration in vitro Treatment with the MEK inhibitor selumetinib extended survival and reduced CD271 levels in vivo, whereas, treatment with vismodegib, a well-known smoothened (SMO) inhibitor currently in clinical trials for the treatment of recurrent SHH medulloblastoma, had no significant effect in our models. Our study demonstrates the clinical utility of CD271 as both a diagnostic and prognostic tool for SHH medulloblastoma tumors and reveals a novel role for MEK inhibitors in targeting CD271+ SHH medulloblastoma cells.Significance: This study identifies CD271 as a specific and novel biomarker of SHH-type medulloblastoma and that targeting CD271+ cells through MEK inhibition represents a novel therapeutic strategy for the treatment of SHH medulloblastoma. Cancer Res; 78(16); 4745-59. ©2018 AACR.


Assuntos
Biomarcadores Tumorais/genética , Meduloblastoma/genética , Proteínas do Tecido Nervoso/genética , Prognóstico , Receptores de Fator de Crescimento Neural/genética , Anilidas/administração & dosagem , Anexina A5/genética , Movimento Celular/genética , Proliferação de Células/genética , Sobrevivência Celular/efeitos dos fármacos , Criança , Inibidores Enzimáticos/administração & dosagem , Feminino , Regulação Neoplásica da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Estimativa de Kaplan-Meier , MAP Quinase Quinase Quinase 1/antagonistas & inibidores , Angiografia por Ressonância Magnética , Masculino , Meduloblastoma/tratamento farmacológico , Meduloblastoma/patologia , Piridinas/administração & dosagem
15.
J Exp Clin Cancer Res ; 36(1): 67, 2017 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-28499442

RESUMO

BACKGROUND: Chemotherapy resistance is one of the major factors contributing to mortality from human epithelial ovarian cancer (EOC). Identifying drugs that can effectively kill chemotherapy-resistant EOC cells would be a major advance in reducing mortality. Glycosylated antitumour ether lipids (GAELs) are synthetic glycolipids that are cytotoxic to a wide range of cancer cells. They appear to induce cancer cell death in an apoptosis-independent manner. METHODS: Herein, the effectiveness of two GAELs, GLN and MO-101, in killing chemotherapy-sensitive and -resistant EOC cells lines and primary cell samples was tested using monolayer, non-adherent aggregate, and non-adherent spheroid cultures. RESULTS: Our results show that EOC cells exhibit a differential sensitivity to the GAELs. Strikingly, both GAELs are capable of inducing EOC cell death in chemotherapy-sensitive and -resistant cells grown as monolayer or non-adherent cultures. Mechanistic studies provide evidence that apoptotic-cell death (caspase activation) contributes to, but is not completely responsible for, GAEL-induced cell killing in the A2780-cp EOC cell line, but not primary EOC cell samples. CONCLUSIONS: Studies using primary EOC cell samples supports previously published work showing a GAEL-induced caspase-independent mechanism of death. GAELs hold promise for development as novel compounds to combat EOC mortality due to chemotherapy resistance.


Assuntos
Antineoplásicos/farmacologia , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Glicolipídeos/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma Epitelial do Ovário , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Humanos , Neoplasias Epiteliais e Glandulares/metabolismo , Neoplasias Ovarianas/metabolismo , Pepstatinas/farmacologia
16.
Oncotarget ; 6(36): 38881-900, 2015 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-26497209

RESUMO

Major research efforts have focused on defining cell surface marker profiles for characterization and selection of brain tumor stem/progenitor cells. Medulloblastoma is the most common primary malignant pediatric brain cancer and consists of 4 molecular subgroups: WNT, SHH, Group 3 and Group 4. Given the heterogeneity within and between medulloblastoma variants, surface marker profiles may be subtype-specific. Here, we employed a high throughput flow cytometry screen to identify differentially expressed cell surface markers in self-renewing vs. non-self-renewing SHH medulloblastoma cells. The top 25 markers were reduced to 4, CD271/p75NTR/NGFR, CD106/VCAM1, EGFR and CD171/NCAM-L1, by evaluating transcript levels in SHH tumors relative to samples representing the other variants. However, only CD271/p75NTR/NGFR and CD171/NCAM-L1 maintain differential expression between variants at the protein level. Functional characterization of CD271, a low affinity neurotrophin receptor, in cell lines and primary cultures suggested that CD271 selects for lower self-renewing progenitors or stem cells. Moreover, CD271 levels were negatively correlated with expression of SHH pathway genes. Our study reveals a novel role for CD271 in SHH medulloblastoma and suggests that targeting CD271 pathways could lead to the design of more selective therapies that lessen the broad impact of current treatments on developing nervous systems.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias Encefálicas/classificação , Meduloblastoma/classificação , Biomarcadores Tumorais/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Citometria de Fluxo , Humanos , Meduloblastoma/genética , Meduloblastoma/metabolismo , Meduloblastoma/patologia , Fenótipo
17.
Neoplasia ; 15(4): 384-98, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23555184

RESUMO

Medulloblastoma (MB) is the most common malignant primary pediatric brain tumor. Major research efforts have focused on characterizing and targeting putative brain tumor stem or propagating cell populations from the tumor mass. However, less is known about the relationship between these cells and highly invasive MB cells that evade current therapies. Here, we dissected MB cellular heterogeneity and directly compared invasion and self-renewal. Analysis of higher versus lower self-renewing tumor spheres and stationary versus migrating adherent MB cells revealed differential expression of the cell surface markers CD271 [p75 neurotrophin receptor (p75NTR)] and CD133. Cell sorting demonstrated that CD271 selects for subpopulations with a higher capacity for self-renewal, whereas CD133 selects for cells exhibiting increased invasion in vitro. CD271 expression is higher in human fetal cerebellum and primary samples of the Shh MB molecular variant and lower in the more aggressive, invasive group 3 and 4 subgroups. Global gene expression analysis of higher versus lower self-renewing MB tumor spheres revealed down-regulation of a cell movement transcription program in the higher self-renewing state and a novel potential role for axon guidance signaling in MB-propagating cells. We have identified a cell surface signature based on CD133/CD271 expression that selects for MB cells with a higher self-renewal potential or invasive capacity in vitro. Our study underscores a previously unappreciated role for CD271 in selecting for MB cell phenotypes and suggests that successful treatment of pediatric brain tumors requires concomitant targeting of a spectrum of transitioning self-renewing and highly infiltrative cell subpopulations.


Assuntos
Proliferação de Células , Neoplasias Cerebelares/patologia , Meduloblastoma/patologia , Células-Tronco Neoplásicas/metabolismo , Adulto , Animais , Antimetabólitos Antineoplásicos/farmacologia , Antígeno CD24/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Separação Celular , Forma Celular , Neoplasias Cerebelares/metabolismo , Resistencia a Medicamentos Antineoplásicos , Exoma , Citometria de Fluxo , Fluoruracila/farmacologia , Humanos , Subunidade alfa de Receptor de Interleucina-3/metabolismo , Meduloblastoma/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Invasividade Neoplásica , Transplante de Neoplasias , Células-Tronco Neoplásicas/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Receptores de Fator de Crescimento Neural/metabolismo , Esferoides Celulares/metabolismo , Esferoides Celulares/fisiologia
18.
PLoS One ; 4(11): e8065, 2009 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-19956629

RESUMO

BACKGROUND: Cancer and normal stem cells (SCs) share proliferative properties of self-renewal and expression of key transcription factors (TFs). Despite similar TF identities, the functional role of specific TFs responsible for retaining SC state has yet to be examined in cancer. METHODOLOGY/PRINCIPAL FINDINGS: Here, we compare the role of Oct4 and Nanog, two-core pluripotent TFs, in transformed (t-hPSCs), and normal human pluripotent stem cells (hPSCs). Unlike normal SCs, self-renewal and survival of t-hPSCs were found to be independent of Oct4. In contrast, t-hPSCs exhibit hypersensitivity to reduction in Nanog and demonstrate complete loss of self-renewal coupled with apoptosis. Dual and sequential knockdown of Oct4 and Nanog revealed that sensitivity of t-hPSCs to Nanog was Oct4 dependent. CONCLUSIONS/SIGNIFICANCE: Our study indicates a bifurcation for the role of two-core SC and cancer related TFs in self-renewal and survival processes. We suggest that the divergent roles of these TFs establish a paradigm to develop novel therapeutics towards selective destruction of aggressive tumors harboring cancer stem cells (CSCs) with similar molecular signatures.


Assuntos
Linhagem Celular Transformada , Células-Tronco Neoplásicas/citologia , Células-Tronco Pluripotentes/citologia , Fatores de Transcrição/metabolismo , Apoptose , Diferenciação Celular , Linhagem Celular , Citometria de Fluxo/métodos , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Lentivirus/metabolismo , Modelos Biológicos , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/metabolismo , Teratoma/metabolismo
19.
Nat Biotechnol ; 27(1): 91-7, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19122652

RESUMO

Cultured human embryonic stem (hES) cells can acquire genetic and epigenetic changes that make them vulnerable to transformation. As hES cells with cancer-cell characteristics share properties with normal hES cells, such as self-renewal, teratoma formation and the expression of pluripotency markers, they may be misconstrued as superior hES cells with enhanced 'stemness'. We characterize two variant hES cell lines (v-hESC-1 and v-hESC-2) that express pluripotency markers at high levels and do not harbor chromosomal abnormalities by standard cytogenetic measures. We show that the two lines possess some features of neoplastic progression, including a high proliferative capacity, growth-factor independence, a 9- to 20-fold increase in frequency of tumor-initiating cells, niche independence and aberrant lineage specification, although they are not malignant. Array comparative genomic hybridization reveals an amplification at 20q11.1-11.2 in v-hESC-1 and a deletion at 5q34a-5q34b;5q3 and a mosaic gain of chromosome 12 in v-hESC-2. These results emphasize the need for functional characterization to distinguish partially transformed and normal hES cells.


Assuntos
Células-Tronco Embrionárias/citologia , Neoplasias/patologia , Diferenciação Celular , Linhagem Celular , Linhagem Celular Tumoral , Aberrações Cromossômicas , Hibridização Genômica Comparativa , Citogenética , Progressão da Doença , Fator 2 de Crescimento de Fibroblastos/metabolismo , Técnicas Genéticas , Humanos , Hibridização de Ácido Nucleico , Fenótipo , Células-Tronco/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA