Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Neuro Oncol ; 26(6): 1052-1066, 2024 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-38271182

RESUMO

BACKGROUND: Compared to minimally invasive brain metastases (MI BrM), highly invasive (HI) lesions form abundant contacts with cells in the peritumoral brain parenchyma and are associated with poor prognosis. Reactive astrocytes (RAs) labeled by phosphorylated STAT3 (pSTAT3) have recently emerged as a promising therapeutic target for BrM. Here, we explore whether the BrM invasion pattern is influenced by pSTAT3+ RAs and may serve as a predictive biomarker for STAT3 inhibition. METHODS: We used immunohistochemistry to identify pSTAT3+ RAs in HI and MI human and patient-derived xenograft (PDX) BrM. Using PDX, syngeneic, and transgenic mouse models of HI and MI BrM, we assessed how pharmacological STAT3 inhibition or RA-specific STAT3 genetic ablation affected BrM growth in vivo. Cancer cell invasion was modeled in vitro using a brain slice-tumor co-culture assay. We performed single-cell RNA sequencing of human BrM and adjacent brain tissue. RESULTS: RAs expressing pSTAT3 are situated at the brain-tumor interface and drive BrM invasive growth. HI BrM invasion pattern was associated with delayed growth in the context of STAT3 inhibition or genetic ablation. We demonstrate that pSTAT3+ RAs secrete Chitinase 3-like-1 (CHI3L1), which is a known STAT3 transcriptional target. Furthermore, single-cell RNA sequencing identified CHI3L1-expressing RAs in human HI BrM. STAT3 activation, or recombinant CHI3L1 alone, induced cancer cell invasion into the brain parenchyma using a brain slice-tumor plug co-culture assay. CONCLUSIONS: Together, these data reveal that pSTAT3+ RA-derived CHI3L1 is associated with BrM invasion, implicating STAT3 and CHI3L1 as clinically relevant therapeutic targets for the treatment of HI BrM.


Assuntos
Astrócitos , Neoplasias Encefálicas , Proteína 1 Semelhante à Quitinase-3 , Invasividade Neoplásica , Fator de Transcrição STAT3 , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT3/genética , Humanos , Proteína 1 Semelhante à Quitinase-3/metabolismo , Proteína 1 Semelhante à Quitinase-3/genética , Animais , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/secundário , Neoplasias Encefálicas/genética , Astrócitos/metabolismo , Astrócitos/patologia , Camundongos , Camundongos Transgênicos , Proliferação de Células , Ensaios Antitumorais Modelo de Xenoenxerto , Células Tumorais Cultivadas
2.
Cancer Cell ; 41(9): 1637-1649.e11, 2023 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-37652007

RESUMO

A high percentage of patients with brain metastases frequently develop neurocognitive symptoms; however, understanding how brain metastasis co-opts the function of neuronal circuits beyond a tumor mass effect remains unknown. We report a comprehensive multidimensional modeling of brain functional analyses in the context of brain metastasis. By testing different preclinical models of brain metastasis from various primary sources and oncogenic profiles, we dissociated the heterogeneous impact on local field potential oscillatory activity from cortical and hippocampal areas that we detected from the homogeneous inter-model tumor size or glial response. In contrast, we report a potential underlying molecular program responsible for impairing neuronal crosstalk by scoring the transcriptomic and mutational profiles in a model-specific manner. Additionally, measurement of various brain activity readouts matched with machine learning strategies confirmed model-specific alterations that could help predict the presence and subtype of metastasis.


Assuntos
Neoplasias Encefálicas , Humanos , Neoplasias Encefálicas/genética , Encéfalo , Perfilação da Expressão Gênica , Aprendizado de Máquina , Mutação
3.
Nat Med ; 28(4): 752-765, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35411077

RESUMO

Whole-brain radiotherapy (WBRT) is the treatment backbone for many patients with brain metastasis; however, its efficacy in preventing disease progression and the associated toxicity have questioned the clinical impact of this approach and emphasized the need for alternative treatments. Given the limited therapeutic options available for these patients and the poor understanding of the molecular mechanisms underlying the resistance of metastatic lesions to WBRT, we sought to uncover actionable targets and biomarkers that could help to refine patient selection. Through an unbiased analysis of experimental in vivo models of brain metastasis resistant to WBRT, we identified activation of the S100A9-RAGE-NF-κB-JunB pathway in brain metastases as a potential mediator of resistance in this organ. Targeting this pathway genetically or pharmacologically was sufficient to revert the WBRT resistance and increase therapeutic benefits in vivo at lower doses of radiation. In patients with primary melanoma, lung or breast adenocarcinoma developing brain metastasis, endogenous S100A9 levels in brain lesions correlated with clinical response to WBRT and underscored the potential of S100A9 levels in the blood as a noninvasive biomarker. Collectively, we provide a molecular framework to personalize WBRT and improve its efficacy through combination with a radiosensitizer that balances therapeutic benefit and toxicity.


Assuntos
Neoplasias Encefálicas , Melanoma , Neoplasias Encefálicas/secundário , Irradiação Craniana , Humanos , Melanoma/radioterapia
4.
EMBO Mol Med ; 14(3): e14552, 2022 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-35174975

RESUMO

We report a medium-throughput drug-screening platform (METPlatform) based on organotypic cultures that allows to evaluate inhibitors against metastases growing in situ. By applying this approach to the unmet clinical need of brain metastasis, we identified several vulnerabilities. Among them, a blood-brain barrier permeable HSP90 inhibitor showed high potency against mouse and human brain metastases at clinically relevant stages of the disease, including a novel model of local relapse after neurosurgery. Furthermore, in situ proteomic analysis applied to metastases treated with the chaperone inhibitor uncovered a novel molecular program in brain metastasis, which includes biomarkers of poor prognosis and actionable mechanisms of resistance. Our work validates METPlatform as a potent resource for metastasis research integrating drug-screening and unbiased omic approaches that is compatible with human samples. Thus, this clinically relevant strategy is aimed to personalize the management of metastatic disease in the brain and elsewhere.


Assuntos
Antineoplásicos , Neoplasias Encefálicas , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Barreira Hematoencefálica , Neoplasias Encefálicas/tratamento farmacológico , Camundongos , Recidiva Local de Neoplasia , Proteômica
5.
Front Oncol ; 11: 714428, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34540682

RESUMO

Uncovering the complexity of the microenvironment that emerges in brain disorders is key to identify potential vulnerabilities that might help challenging diseases affecting this organ. Recently, genomic and proteomic analyses, especially at the single cell level, have reported previously unrecognized diversity within brain cell types. The complexity of the brain microenvironment increases during disease partly due to the immune infiltration from the periphery that contributes to redefine the brain connectome by establishing a new crosstalk with resident brain cell types. Within the rewired brain ecosystem, glial cell subpopulations are emerging hubs modulating the dialogue between the Immune System and the Central Nervous System with important consequences in the progression of brain tumors and other disorders. Single cell technologies are crucial not only to define and track the origin of disease-associated cell types, but also to identify their molecular similarities and differences that might be linked to specific brain injuries. These altered molecular patterns derived from reprogramming the healthy brain into an injured organ, might provide a new generation of therapeutic targets to challenge highly prevalent and lethal brain disorders that remain incurable with unprecedented specificity and limited toxicities. In this perspective, we present the most relevant clinical and pre-clinical work regarding the characterization of the heterogeneity within different components of the microenvironment in the healthy and injured brain with a special interest on single cell analysis. Finally, we discuss how understanding the diversity of the brain microenvironment could be exploited for translational purposes, particularly in primary and secondary tumors affecting the brain.

6.
Front Immunol ; 10: 1314, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31244853

RESUMO

The neuro-immune axis has emerged as a key aspect to understand the normal function of the Central Nervous System (CNS) as well as the pathophysiology of many brain disorders. As such, it may represent a promising source for novel therapeutic targets. Glial cells, and in particular the extensively studied microglia, play important roles in brain disorders. Astrocytes, in their reactive state, have been shown to positively and negatively modulate the progression of multiple CNS disorders. These seemingly opposing effects, might stem from their underlying heterogeneity, an aspect that has recently come to light. In this article we will discuss the link between reactive astrocytes and the neuro-immune axis with a perspective on their potential importance in brain tumors. Based on the gained knowledge from studies in other CNS disorders, reactive astrocytes are undoubtfully emerging as a key component of the neuro-immune axis, with ability to modulate both the innate and adaptive branches of the immune system. Lastly, we will discuss how we can exploit our improved understanding of the basic biology of astrocytes to further enhance the efficacy of emerging immune-based therapies in primary brain tumors and brain metastasis.


Assuntos
Astrócitos/imunologia , Neoplasias Encefálicas/imunologia , Animais , Astrócitos/patologia , Neoplasias Encefálicas/patologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Comunicação Celular/imunologia , Homeostase , Humanos , Imunidade Inata , Imunomodulação , Microglia/imunologia , Microglia/patologia , Modelos Imunológicos , Modelos Neurológicos , Monócitos/imunologia , Monócitos/patologia , Neuroimunomodulação , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia
7.
Nat Med ; 24(7): 1024-1035, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29892069

RESUMO

The brain microenvironment imposes a particularly intense selective pressure on metastasis-initiating cells, but successful metastases bypass this control through mechanisms that are poorly understood. Reactive astrocytes are key components of this microenvironment that confine brain metastasis without infiltrating the lesion. Here, we describe that brain metastatic cells induce and maintain the co-option of a pro-metastatic program driven by signal transducer and activator of transcription 3 (STAT3) in a subpopulation of reactive astrocytes surrounding metastatic lesions. These reactive astrocytes benefit metastatic cells by their modulatory effect on the innate and acquired immune system. In patients, active STAT3 in reactive astrocytes correlates with reduced survival from diagnosis of intracranial metastases. Blocking STAT3 signaling in reactive astrocytes reduces experimental brain metastasis from different primary tumor sources, even at advanced stages of colonization. We also show that a safe and orally bioavailable treatment that inhibits STAT3 exhibits significant antitumor effects in patients with advanced systemic disease that included brain metastasis. Responses to this therapy were notable in the central nervous system, where several complete responses were achieved. Given that brain metastasis causes substantial morbidity and mortality, our results identify a novel treatment for increasing survival in patients with secondary brain tumors.


Assuntos
Astrócitos/patologia , Neoplasias Encefálicas/secundário , Fator de Transcrição STAT3/metabolismo , Animais , Encéfalo/patologia , Neoplasias Encefálicas/patologia , Sobrevivência Celular , Marcação de Genes , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Imunidade Inata , Camundongos , Fosforilação , Microambiente Tumoral
8.
Nat Med ; 24(9): 1481, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29921958

RESUMO

In the version of this article originally published, the names of three authors were incorrect. The authors were listed as "Coral Fustero-Torres", "Elena Pineiro" and "Melchor Sánchez-Martínez". Their respective names are "Coral Fustero-Torre", "Elena Piñeiro-Yáñez" and "Melchor Sanchez-Martinez". The errors have been corrected in the print, HTML and PDF versions of this article.

9.
Acta Neuropathol ; 135(4): 581-599, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29350274

RESUMO

The discovery of genes and molecular pathways involved in the formation of brain metastasis would direct the development of therapeutic strategies to prevent this deadly complication of cancer. By comparing gene expression profiles of Estrogen Receptor negative (ER-) primary breast tumors between patients who developed metastasis to brain and to organs other than brain, we found that T lymphocytes promote the formation of brain metastases. To functionally test the ability of T cells to promote brain metastasis, we used an in vitro blood-brain barrier (BBB) model. By co-culturing T lymphocytes with breast cancer cells, we confirmed that T cells increase the ability of breast cancer cells to cross the BBB. Proteomics analysis of the tumor cells revealed Guanylate-Binding Protein 1 (GBP1) as a key T lymphocyte-induced protein that enables breast cancer cells to cross the BBB. The GBP1 gene appeared to be up-regulated in breast cancer of patients who developed brain metastasis. Silencing of GBP1 reduced the ability of breast cancer cells to cross the in vitro BBB model. In addition, the findings were confirmed in vivo in an immunocompetent syngeneic mouse model. Co-culturing of ErbB2 tumor cells with activated T cells induced a significant increase in Gbp1 expression by the cancer cells. Intracardial inoculation of the co-cultured tumor cells resulted in preferential seeding to brain. Moreover, intracerebral outgrowth of the tumor cells was demonstrated. The findings point to a role of T cells in the formation of brain metastases in ER- breast cancers, and provide potential targets for intervention to prevent the development of cerebral metastases.


Assuntos
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/secundário , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proteínas de Ligação ao GTP/metabolismo , Linfócitos T/metabolismo , Adulto , Idoso , Animais , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Células Cultivadas , Técnicas de Cocultura , Feminino , Proteínas de Ligação ao GTP/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Pessoa de Meia-Idade , Metástase Neoplásica/fisiopatologia , Transplante de Neoplasias , Proteoma , RNA Mensageiro/metabolismo
10.
Front Oncol ; 7: 298, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29312881

RESUMO

Brain metastasis, the secondary growth of malignant cells within the central nervous system (CNS), exceeds the incidence of primary brain tumors (i.e., gliomas) by tenfold and are seemingly on the rise owing to the emergence of novel targeted therapies that are more effective in controlling extracranial disease relatively to intracranial lesions. Despite the fact that metastasis to the brain poses a unmet clinical problem, with afflicted patients carrying significant morbidity and a fatal prognosis, our knowledge as to how metastatic cells manage to adapt to the tissue environment of the CNS remains limited. Answering this question could pave the way for novel and more specific therapeutic modalities in brain metastasis by targeting the specific makeup of the brain metastatic niche. In regard to this, astrocytes have emerged as the major host cell type that cancer cells encounter and interact with during brain metastasis formation. Similarly to other CNS disorders, astrocytes become reactive and respond to the presence of cancer cells by changing their phenotype and significantly influencing the outcome of disseminated cancer cells within the CNS. Here, we summarize the current knowledge on the contribution of reactive astrocytes in brain metastasis by focusing on the signaling pathways and types of interactions that play a crucial part in the communication with cancer cells and how these could be translated into innovative therapies.

11.
Oncotarget ; 7(29): 45060-45078, 2016 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-27286263

RESUMO

C3G, a Guanine nucleotide Exchange Factor (GEF) for Rap1 and R-Ras, has been shown to play important roles in development and cancer. Previous studies determined that C3G regulates cell death through down-regulation of p38α MAPK activity. Here, we found that C3G knock-down in MEFs and HCT116 cells promotes migration and invasion through Rap1-mediated p38α hyper-activation. These effects of C3G were inhibited by Rap1 knock-down or inactivation. The enhanced migration observed in C3G depleted HCT116 cells was associated with reduction in E-cadherin expression, internalization of ZO-1, actin cytoskeleton reorganization and decreased adhesion. We also found that matrix metalloproteases MMP2 and MMP9 are involved in the pro-invasive effect of C3G down-regulation. Additionally, our studies revealed that both C3G and p38α collaborate to promote growth of HCT116 cells in vitro and in vivo, possibly by enhancing cell survival. In fact, knocking-down C3G or p38α individually or together promoted cell death in vitro, although only the double C3G-p38α silencing was able to increase cell death within tumors. Notably, we found that the pro-tumorigenic function of C3G does not depend on p38α or Rap1 activation. Altogether, our studies uncover novel mechanisms by which C3G controls key aspects of tumorigenesis.


Assuntos
Carcinogênese/metabolismo , Neoplasias Colorretais/patologia , Fator 2 de Liberação do Nucleotídeo Guanina/metabolismo , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Proteínas rap1 de Ligação ao GTP/metabolismo , Animais , Movimento Celular/fisiologia , Neoplasias Colorretais/metabolismo , Ativação Enzimática/fisiologia , Fibroblastos/metabolismo , Técnicas de Silenciamento de Genes , Células HCT116 , Xenoenxertos , Humanos , Masculino , Camundongos , Camundongos Nus , Invasividade Neoplásica/patologia
12.
J Biol Chem ; 290(7): 4383-97, 2015 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-25548290

RESUMO

p38 MAPKs regulate migration and invasion. However, the mechanisms involved are only partially known. We had previously identified fibulin 3, which plays a role in migration, invasion, and tumorigenesis, as a gene regulated by p38α. We have characterized in detail how p38 MAPK regulates fibulin 3 expression and its role. We describe here for the first time that p38α, p38γ, and p38δ down-regulate fibulin 3 expression. p38α has a stronger effect, and it does so through hypermethylation of CpG sites in the regulatory sequences of the gene. This would be mediated by the DNA methylase, DNMT3A, which is down-regulated in cells lacking p38α, but once re-introduced represses Fibulin 3 expression. p38α through HuR stabilizes dnmt3a mRNA leading to an increase in DNMT3A protein levels. Moreover, by knocking-down fibulin 3, we have found that Fibulin 3 inhibits migration and invasion in MEFs by mechanisms involving p38α/ß inhibition. Hence, p38α pro-migratory/invasive effect might be, at least in part, mediated by fibulin 3 down-regulation in MEFs. In contrast, in HCT116 cells, Fibulin 3 promotes migration and invasion through a mechanism dependent on p38α and/or p38ß activation. Furthermore, Fibulin 3 promotes in vitro and in vivo tumor growth of HCT116 cells through a mechanism dependent on p38α, which surprisingly acts as a potent inducer of tumor growth. At the same time, p38α limits fibulin 3 expression, which might represent a negative feed-back loop.


Assuntos
Movimento Celular , Neoplasias do Colo/patologia , Metilação de DNA , Embrião de Mamíferos/metabolismo , Proteínas da Matriz Extracelular/genética , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Proteína Quinase 14 Ativada por Mitógeno/fisiologia , Animais , Western Blotting , Adesão Celular , Proliferação de Células , Células Cultivadas , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Regulação para Baixo , Embrião de Mamíferos/citologia , Proteínas da Matriz Extracelular/metabolismo , Fibroblastos/citologia , Humanos , Masculino , Camundongos , Camundongos Knockout , Camundongos Nus , Invasividade Neoplásica , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Elementos de Resposta/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Cicatrização , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA