Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
iScience ; 25(11): 105317, 2022 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-36310582

RESUMO

Immunotherapy for cancer that aims to promote T cell anti-tumor activity has changed current clinical practice, where some previously lethal cancers have now become treatable. However, clinical trials with low response rates have been disappointing for pancreatic ductal adenocarcinoma (PDAC). One suggested explanation is the accumulation of dominantly immunosuppressive tumor-associated macrophages and myeloid-derived suppressor cells in the tumor microenvironment (TME). Using retrospectively collected tumor specimens and transcriptomic data from PDAC, we demonstrate that expression of the scavenger receptor MARCO correlates with poor prognosis and a lymphocyte-excluding tumor phenotype. PDAC cell lines produce IL-10 and induce high expression of MARCO in myeloid cells, and this was further enhanced during hypoxic conditions. These myeloid cells suppressed effector T and natural killer (NK) cells and blocked NK cell tumor infiltration and tumor killing in a PDAC 3D-spheroid model. Anti-human MARCO (anti-hMARCO) antibody targeting triggered the repolarization of tumor-associated macrophages and activated the inflammasome machinery, resulting in IL-18 production. This in turn enhanced T cell and NK cell functions. The targeting of MARCO thus remodels the TME and represents a rational approach to make immunotherapy more efficient in PDAC patients.

2.
Nat Commun ; 11(1): 2898, 2020 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-32518258

RESUMO

The sequential generation of layer-specific cortical neurons requires radial glia cells (RGCs) to precisely balance self-renewal and lineage commitment. While specific cell-cycle phases have been associated with these decisions, the mechanisms linking the cell-cycle machinery to cell-fate commitment remain obscure. Using single-cell RNA-sequencing, we find that the strongest transcriptional signature defining multipotent RGCs is that of G2/M-phase, and particularly CYCLIN-B1/2, while lineage-committed progenitors are enriched in G1/S-phase genes, including CYCLIN-D1. These data also reveal cell-surface markers that allow us to isolate RGCs and lineage-committed progenitors, and functionally confirm the relationship between cell-cycle phase enrichment and cell fate competence. Finally, we use cortical electroporation to demonstrate that CYCLIN-B1/2 cooperate with CDK1 to maintain uncommitted RGCs by activating the NOTCH pathway, and that CYCLIN-D1 promotes differentiation. Thus, this work establishes that cell-cycle phase-specific regulators act in opposition to coordinate the self-renewal and lineage commitment of RGCs via core stem cell regulatory pathways.


Assuntos
Ciclina B1/fisiologia , Ciclina B2/fisiologia , Ciclina D1/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Animais , Proteína Quinase CDC2/fisiologia , Ciclo Celular , Diferenciação Celular , Linhagem da Célula , Separação Celular , Córtex Cerebral/embriologia , Feminino , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos C57BL , Neuroglia/metabolismo , Análise de Sequência de RNA , Transdução de Sinais , Células-Tronco/citologia
3.
Proc Natl Acad Sci U S A ; 116(34): 16997-17006, 2019 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-31375625

RESUMO

Despite the discovery of the oxygen-sensitive regulation of HIFα by the von Hippel-Lindau (VHL) protein, the mechanisms underlying the complex genotype/phenotype correlations in VHL disease remain unknown. Some germline VHL mutations cause familial pheochromocytoma and encode proteins that preserve their ability to down-regulate HIFα. While type 1, 2A, and 2B VHL mutants are defective in regulating HIFα, type 2C mutants encode proteins that preserve their ability to down-regulate HIFα. Here, we identified an oxygen-sensitive function of VHL that is abolished by VHL type 2C mutations. We found that BIM-EL, a proapoptotic BH3-only protein, is hydroxylated by EglN3 and subsequently bound by VHL. VHL mutants fail to bind hydroxylated BIM-EL, regardless of whether they have the ability to bind hydroxylated HIFα or not. VHL binding inhibits BIM-EL phosphorylation by extracellular signal-related kinase (ERK) on serine 69. This causes BIM-EL to escape from proteasomal degradation, allowing it to enhance EglN3-induced apoptosis. BIM-EL was rapidly degraded in cells lacking wild-type VHL or in which EglN3 was inactivated genetically or by lack of oxygen, leading to enhanced cell survival and chemotherapy resistance. Combination therapy using ERK inhibitors, however, resensitizes VHL- and EglN3-deficient cells that are otherwise cisplatin-resistant.


Assuntos
Neoplasias das Glândulas Suprarrenais , Proteína 11 Semelhante a Bcl-2/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética , Prolina Dioxigenases do Fator Induzível por Hipóxia/metabolismo , Mutação , Feocromocitoma , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo , Neoplasias das Glândulas Suprarrenais/tratamento farmacológico , Neoplasias das Glândulas Suprarrenais/genética , Neoplasias das Glândulas Suprarrenais/metabolismo , Neoplasias das Glândulas Suprarrenais/patologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Proteína 11 Semelhante a Bcl-2/genética , Hipóxia Celular/efeitos dos fármacos , Hipóxia Celular/genética , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Humanos , Hidroxilação/efeitos dos fármacos , Hidroxilação/genética , Prolina Dioxigenases do Fator Induzível por Hipóxia/genética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Camundongos Knockout , Células PC12 , Feocromocitoma/tratamento farmacológico , Feocromocitoma/metabolismo , Feocromocitoma/patologia , Proteólise/efeitos dos fármacos , Ratos , Proteína Supressora de Tumor Von Hippel-Lindau/genética
4.
Cancer Res ; 77(18): 4985-4997, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28687615

RESUMO

Molecular mechanisms preventing self-renewing brain stem cells from oncogenic transformation are poorly defined. We show that the expression levels of SOX5, SOX6, and SOX21 (SOX5/6/21) transcription factors increase in stem cells of the subventricular zone (SVZ) upon oncogenic stress, whereas their expression in human glioma decreases during malignant progression. Elevated levels of SOX5/6/21 promoted SVZ cells to exit the cell cycle, whereas genetic ablation of SOX5/6/21 dramatically increased the capacity of these cells to form glioma-like tumors in an oncogene-driven mouse brain tumor model. Loss-of-function experiments revealed that SOX5/6/21 prevent detrimental hyperproliferation of oncogene expressing SVZ cells by facilitating an antiproliferative expression profile. Consistently, restoring high levels of SOX5/6/21 in human primary glioblastoma cells enabled expression of CDK inhibitors and decreased p53 protein turnover, which blocked their tumorigenic capacity through cellular senescence and apoptosis. Altogether, these results provide evidence that SOX5/6/21 play a central role in driving a tumor suppressor response in brain stem cells upon oncogenic insult. Cancer Res; 77(18); 4985-97. ©2017 AACR.


Assuntos
Neoplasias Encefálicas/patologia , Transformação Celular Neoplásica/patologia , Glioma/patologia , Células-Tronco Neoplásicas/patologia , Fatores de Transcrição SOXB2/metabolismo , Fatores de Transcrição SOXD/fisiologia , Animais , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Senescência Celular , Feminino , Glioma/genética , Glioma/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Células-Tronco Neoplásicas/metabolismo , Oncogenes , Fatores de Transcrição SOXB2/genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Cell Rep ; 9(5): 1908-1920, 2014 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-25482558

RESUMO

Organ formation and maintenance depends on slowly self-renewing stem cells that supply an intermediate population of rapidly dividing progenitors, but how this proliferative hierarchy is regulated is unknown. By performing genome-wide single-cell and functional analyses in the cortex, we demonstrate that reduced Sox2 expression is a key regulatory signature of the transition between stem cells and rapidly dividing progenitors. In stem cells, Sox2 is expressed at high levels, which enables its repression of proproliferative genes, of which Cyclin D1 is the most potent target. Sox2 confers this function through binding to low-affinity motifs, which facilitate the recruitment of Gro/Tle corepressors in synergy with Tcf/Lef proteins. Upon differentiation, proneural factors reduce Sox2 expression, which derepresses Cyclin D1 and promotes proliferation. Our results show how concentration-dependent Sox2 occupancy of DNA motifs of varying affinities translates into recruitment of repressive complexes, which regulate the proliferative dynamics of neural stem and progenitor cells.


Assuntos
Proliferação de Células , Córtex Cerebral/citologia , Células-Tronco Neurais/fisiologia , Fatores de Transcrição SOXB1/fisiologia , Animais , Ciclo Celular , Diferenciação Celular , Ciclina D1/metabolismo , Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Regiões Promotoras Genéticas , Ligação Proteica , Fatores de Transcrição SOXB1/metabolismo
6.
Dev Cell ; 23(5): 1006-19, 2012 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-23153497

RESUMO

Morphogens orchestrate tissue patterning in a concentration-dependent fashion during vertebrate embryogenesis, yet little is known of how positional information provided by such signals is translated into discrete transcriptional outputs. Here we have identified and characterized cis-regulatory modules (CRMs) of genes operating downstream of graded Shh signaling and bifunctional Gli proteins in neural patterning. Unexpectedly, we find that Gli activators have a noninstructive role in long-range patterning and cooperate with SoxB1 proteins to facilitate a largely concentration-independent mode of gene activation. Instead, the opposing Gli-repressor gradient is interpreted at transcriptional levels, and, together with CRM-specific repressive input of homeodomain proteins, comprises a repressive network that translates graded Shh signaling into regional gene expression patterns. Moreover, local and long-range interpretation of Shh signaling differs with respect to CRM context sensitivity and Gli-activator dependence, and we propose that these differences provide insight into how morphogen function may have mechanistically evolved from an initially binary inductive event.


Assuntos
Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Animais , Animais Geneticamente Modificados , Padronização Corporal , Sistema Nervoso Central/embriologia , Sistema Nervoso Central/metabolismo , Embrião de Galinha , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Neurogênese , Fatores de Transcrição SOXB1/metabolismo , Transdução de Sinais , Proteína GLI1 em Dedos de Zinco
7.
PLoS One ; 6(3): e18454, 2011 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-21483788

RESUMO

The presence of stem cell characteristics in glioma cells raises the possibility that mechanisms promoting the maintenance and self-renewal of tissue specific stem cells have a similar function in tumor cells. Here we characterized human gliomas of various malignancy grades for the expression of stem cell regulatory proteins. We show that cells in high grade glioma co-express an array of markers defining neural stem cells (NSCs) and that these proteins can fulfill similar functions in tumor cells as in NSCs. However, in contrast to NSCs glioma cells co-express neural proteins together with pluripotent stem cell markers, including the transcription factors Oct4, Sox2, Nanog and Klf4. In line with this finding, in high grade gliomas mesodermal- and endodermal-specific transcription factors were detected together with neural proteins, a combination of lineage markers not normally present in the central nervous system. Persistent presence of pluripotent stem cell traits could only be detected in solid tumors, and observations based on in vitro studies and xenograft transplantations in mice imply that this presence is dependent on the combined activity of intrinsic and extrinsic regulatory cues. Together these results demonstrate a general deregulated expression of neural and pluripotent stem cell traits in malignant human gliomas, and indicate that stem cell regulatory factors may provide significant targets for therapeutic strategies.


Assuntos
Glioma/metabolismo , Glioma/patologia , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/patologia , Células-Tronco Pluripotentes/metabolismo , Animais , Western Blotting , Proteínas de Homeodomínio/metabolismo , Humanos , Imuno-Histoquímica , Hibridização In Situ , Técnicas In Vitro , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Camundongos SCID , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição SOXB1/metabolismo , Células Tumorais Cultivadas
8.
Nat Neurosci ; 6(11): 1162-8, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14517545

RESUMO

The generation of neurons from stem cells involves the activity of proneural basic helix-loop-helix (bHLH) proteins, but the mechanism by which these proteins irreversibly commit stem cells to neuronal differentiation is not known. Here we report that expression of the transcription factors Sox1, Sox2 and Sox3 (Sox1-3) is a critical determinant of neurogenesis. Using chick in ovo electroporation, we found that Sox1-3 transcription factors keep neural cells undifferentiated by counteracting the activity of proneural proteins. Conversely, the capacity of proneural bHLH proteins to direct neuronal differentiation critically depends on their ability to suppress Sox1-3 expression in CNS progenitors. These data suggest that the generation of neurons from stem cells depends on the inhibition of Sox1-3 expression by proneural proteins.


Assuntos
Proteínas Aviárias , Diferenciação Celular/fisiologia , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Grupo de Alta Mobilidade/metabolismo , Neurônios/metabolismo , Proteínas Nucleares/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Bromodesoxiuridina/metabolismo , Embrião de Galinha , Proteínas de Ligação a DNA/fisiologia , Drosophila , Proteínas de Drosophila , Eletroporação/métodos , Indução Embrionária , Proteínas do Olho/metabolismo , Proteínas HMGB , Sequências Hélice-Alça-Hélice/fisiologia , Proteínas de Grupo de Alta Mobilidade/fisiologia , Proteínas de Homeodomínio/metabolismo , Proteínas com Homeodomínio LIM , Glicoproteínas de Membrana , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Proteínas de Neurofilamentos/metabolismo , Neuropeptídeos/metabolismo , Proteínas Nucleares/fisiologia , Fator de Transcrição PAX7 , Fosfopiruvato Hidratase/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fatores de Transcrição SOXB1 , Medula Espinal/citologia , Medula Espinal/embriologia , Medula Espinal/fisiologia , Células-Tronco , Fatores de Tempo , Fatores de Transcrição/metabolismo , Tubulina (Proteína)/metabolismo , Vertebrados
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA