Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Small ; 19(52): e2302280, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37649234

RESUMO

Glioblastoma (GBM) is the most common primary malignant brain cancer in adults with a dismal prognosis. Temozolomide (TMZ) is the first-in-line chemotherapeutic; however, resistance is frequent and multifactorial. While many molecular and genetic factors have been linked to TMZ resistance, the role of the solid tumor morphology and the tumor microenvironment, particularly the blood-brain barrier (BBB), is unknown. Here, the authors investigate these using a complex in vitro model for GBM and its surrounding BBB. The model recapitulates important clinical features such as a dense tumor core with tumor cells that invade along the perivascular space; and a perfusable BBB with a physiological permeability and morphology that is altered in the presence of a tumor spheroid. It is demonstrated that TMZ sensitivity decreases with increasing cancer cell spatial organization, and that the BBB can contribute to TMZ resistance. Proteomic analysis with next-generation low volume sample workflows of these cultured microtissues revealed potential clinically relevant proteins involved in tumor aggressiveness and TMZ resistance, demonstrating the utility of complex in vitro models for interrogating the tumor microenvironment and therapy validation.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Humanos , Temozolomida/farmacologia , Temozolomida/uso terapêutico , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Barreira Hematoencefálica/metabolismo , Microambiente Tumoral , Proteômica , Resistencia a Medicamentos Antineoplásicos/genética , Linhagem Celular Tumoral , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Pharmacol Res ; 182: 106308, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35714825

RESUMO

This review describes recent technological advances applied to glioblastoma (GBM), a brain tumor with dismal prognosis. International consortial efforts suggest the presence of molecular subtypes within histologically identical GBM tumors. This emphasizes that future treatment decisions should no longer be made based solely on morphological analyses, but must now take into consideration such molecular and cellular heterogeneity. The use of single-cell technologies has advanced our understanding and assignation of functional subtypes revealing therapeutic vulnerabilities. Our team has developed stratification approaches in the past few years, and we have been able to identify patient cohorts enriched for various signaling pathways. Importantly, our Glioportal brain tumor resource has been established under the National Neuroscience Institute Tissue Bank in 2021. This resource offers preclinical capability to validate working hypotheses established from patient clinical datasets. This review highlights recent developments with the ultimate goal of assigning functional meaning to molecular subtypes, revealing therapeutic vulnerabilities.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Humanos , Terapia de Alvo Molecular , Medicina de Precisão , Prognóstico
3.
Anal Chem ; 93(4): 2377-2384, 2021 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-33443405

RESUMO

Recent genomic studies on the glioblastoma (GBM) subtypes (e.g., mesenchymal, proneural, and classical) pave a way for effective clinical treatments of the recurrent brain tumor. However, identification of the GBM subtype is complicated by the intratumoral heterogeneity that results in coexistence of multiple subtypes within the tissue specimen. Here, we present the use of hyperspectral stimulated Raman scattering (SRS) microscopy for rapid, label-free molecular assessment of GBM intratumoral heterogeneity with submicron resolution. We develop a unique label-free Raman imaging diagnostic platform consisting of the spectral focusing hyperspectral SRS imaging of the large-area GBM tissue specimens, SRS images, and spectrum retrieval using the multivariate curve resolution algorithm and subtype classification based on the quadratic support vector machine model for rapid molecular subtyping of GBMs. Both the stain-free SRS histological images and 2D subtype maps can be obtained within 20-30 min which is superior to the days of the conventional single-cell RNA sequencing. While the SRS histology assesses the demyelination status as a new diagnostic feature, the SRS mapping provides a new insight into intratumoral heterogeneity across GBM tissue specimens. We find that the major proportions of the GBM tissues agree with the diagnostic results of the genomic analysis, but nontrivial portions of the remaining SRS image tiles in the specimens are found to belong to other molecular subtypes, implying the substantial degree of GBM heterogeneity. The rapid SRS imaging diagnostic platform developed has shown the ability of unveiling tumor heterogeneity in GBM tissues accurately, which would promote the improvement of the GBM-targeted therapy in near future.


Assuntos
Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Técnicas Histológicas , Microscopia Óptica não Linear/métodos , Neoplasias Encefálicas/diagnóstico por imagem , Glioblastoma/diagnóstico por imagem , Humanos , Sensibilidade e Especificidade
4.
Proc Natl Acad Sci U S A ; 115(22): E5086-E5095, 2018 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-29764999

RESUMO

Competitive BET bromodomain inhibitors (BBIs) targeting BET proteins (BRD2, BRD3, BRD4, and BRDT) show promising preclinical activities against brain cancers. However, the BET protein-dependent glioblastoma (GBM)-promoting transcriptional network remains elusive. Here, with mechanistic exploration of a next-generation chemical degrader of BET proteins (dBET6), we reveal a profound and consistent impact of BET proteins on E2F1- dependent transcriptional program in both differentiated GBM cells and brain tumor-initiating cells. dBET6 treatment drastically reduces BET protein genomic occupancy, RNA-Pol2 activity, and permissive chromatin marks. Subsequently, dBET6 represses the proliferation, self-renewal, and tumorigenic ability of GBM cells. Moreover, dBET6-induced degradation of BET proteins exerts superior antiproliferation effects compared to conventional BBIs and overcomes both intrinsic and acquired resistance to BBIs in GBM cells. Our study reveals crucial functions of BET proteins and provides the rationale and therapeutic merits of targeted degradation of BET proteins in GBM.


Assuntos
Antineoplásicos/farmacologia , Fator de Transcrição E2F1 , Glioblastoma , Proteínas Serina-Treonina Quinases , Proteínas de Ligação a RNA , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos , Fator de Transcrição E2F1/antagonistas & inibidores , Fator de Transcrição E2F1/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/metabolismo , Domínios Proteicos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Ligação a RNA/antagonistas & inibidores , Proteínas de Ligação a RNA/metabolismo , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/metabolismo
5.
Anal Chem ; 90(17): 10249-10255, 2018 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-30070837

RESUMO

We report the development and implementation of an epi-detected spectral-focusing hyperspectral stimulated Raman scattering (SRS) imaging technique for label-free biomolecular subtyping of glioblastomas (GBMs). The hyperspectral SRS imaging technique developed generates SRS image stacks (from 2800 to 3020 cm-1 at 7 cm-1 intervals) within 30 s through controlling the time delay between the chirped pump and Stokes beams. SRS images at representative Raman shifts (e.g., 2845, 2885, and 2935 cm-1) delineate the biochemical variations and morphological differences between proneural and mesenchymal subtypes of GBMs. Multivariate curve resolution (MCR) analysis on hyperspectral SRS images enables the quantification of major biomolecule distributions in mesenchymal and proneural GBMs. Further principal component analysis (PCA) and linear discriminant analysis (LDA) together with leave-one SRS spectrum-out, cross-validation (LOOCV) yields a diagnostic sensitivity of 96.7% (29/30) and specificity of 88.9% (28/36) for differentiation between mesenchymal and proneural subtypes of GBMs. This study shows great potential of applying hyperspectral SRS imaging technique developed for rapid, label-free molecular subtyping of GBMs in neurosurgery.


Assuntos
Neoplasias Encefálicas/classificação , Glioblastoma/classificação , Microscopia Óptica não Linear/métodos , Análise Espectral Raman/métodos , Humanos , Análise Multivariada , Análise de Componente Principal
6.
EMBO Rep ; 15(2): 165-74, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24413555

RESUMO

Drosophila larval brain neuroblasts divide asymmetrically to balance between self-renewal and differentiation. Here, we demonstrate that the SCF(Slimb) E3 ubiquitin ligase complex, which is composed of Cul1, SkpA, Roc1a and the F-box protein Supernumerary limbs (Slimb), inhibits ectopic neuroblast formation and regulates asymmetric division of neuroblasts. Hyperactivation of Akt leads to similar neuroblast overgrowth and defects in asymmetric division. Slimb associates with Akt in a protein complex, and SCF(S)(limb) acts through SAK and Akt to inhibit neuroblast overgrowth. Moreover, Beta-transducin repeat containing, the human ortholog of Slimb, is frequently deleted in highly aggressive gliomas, suggesting a conserved tumor suppressor-like function.


Assuntos
Divisão Celular Assimétrica , Proteínas de Ciclo Celular/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Células-Tronco Neurais/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Drosophila/crescimento & desenvolvimento , Proteínas de Drosophila/genética , Células-Tronco Neurais/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transducina/metabolismo , Ubiquitina-Proteína Ligases/genética
7.
Adv Exp Med Biol ; 951: 47-56, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27837553

RESUMO

The Cancer Genome Atlas effort has generated significant interest in a new paradigm shift in tumor tissue analysis, patient diagnosis and subsequent treatment decision. Findings have highlighted the limitation of sole reliance on histology, which can be confounded by inter-observer variability. Such studies demonstrate that histologically similar grade IV brain tumors can be divided into four molecular subtypes based on gene expression, with each subtype demonstrating unique genomic aberrations and clinical outcome. These advances indicate that curative therapeutic strategies must now take into account the molecular information in tumor tissue, with the goal of identifying molecularly stratified patients that will most likely to receive treatment benefit from targeted therapy. This in turn spares non-responders from chemotherapeutic side effects and financial costs. In advancing clinical stage drug candidates, the banking of brain tumor tissue necessitates the acquisition of not just tumor tissue with clinical history and robust follow-up, but also high quality molecular information such as somatic mutation, transcriptomic and DNA methylation profiles which have been shown to predict patient survival independent of current clinical indicators. Additionally, the derivation of cell lines from such tumor tissue facilitates the development of clinically relevant patient-derived xenograft mouse models that can prospectively reform the tumor for further studies, yet have retrospective clinical history to associate bench and in vivo findings with clinical data. This represents a core capability of Precision Medicine where the focus is on understanding inter- and intra-tumor heterogeneity so as to best tailor therapies that will result in improved treatment outcomes.


Assuntos
Bancos de Espécimes Biológicos/estatística & dados numéricos , Neoplasias Encefálicas/terapia , Regulação Neoplásica da Expressão Gênica , Glioblastoma/terapia , Proteínas de Neoplasias/genética , Transcriptoma , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Biologia Computacional/métodos , Metilação de DNA , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Camundongos , Mutação , Gradação de Tumores , Proteínas de Neoplasias/metabolismo , Medicina de Precisão , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Neuro Oncol ; 26(7): 1247-1261, 2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38366847

RESUMO

BACKGROUND: Precision treatment of glioblastoma is increasingly focused on molecular subtyping, with the mesenchymal subtype particularly resistant to temozolomide. Here, we aim to develop a targeted therapy for temozolomide resensitization in the mesenchymal subtype. METHODS: We integrated kinomic profiles and kinase inhibitor screens from patient-derived proneural and mesenchymal glioma-propagating cells and public clinical datasets to identify key protein kinases implicated in temozolomide resistance. RNAseq, apoptosis assays, and comet assays were used to examine the role of p38MAPK signaling and adaptive chemoresistance in mesenchymal cells. The efficacy of dual p38MAPK and MEK/ERK inhibition using ralimetinib (selective orally active p38MAPK inhibitor; phase I/II for glioblastoma) and binimetinib (approved MEK1/2 inhibitor for melanoma; phase II for high-grade glioma) in primary and recurrent mesenchymal tumors was evaluated using an intracranial patient-derived tumor xenograft model, focusing on survival analysis. RESULTS: Our transcriptomic-kinomic integrative analysis revealed p38MAPK as the prime target whose gene signature enables patient stratification based on their molecular subtypes and provides prognostic value. Repurposed p38MAPK inhibitors synergize favorably with temozolomide to promote intracellular retention of temozolomide and exacerbate DNA damage. Mesenchymal cells exhibit adaptive chemoresistance to p38MAPK inhibition through a pH-/calcium-mediated MEK/ERK pathway. Dual p38MAPK and MEK inhibition effectively maintain temozolomide sensitivity in primary and recurrent intracranial mesenchymal glioblastoma xenografts. CONCLUSIONS: Temozolomide resistance in mesenchymal glioblastoma is associated with p38MAPK activation. Adaptive chemoresistance in p38MAPK-resistant cells is mediated by MEK/ERK signaling. Adjuvant therapy with dual p38MAPK and MEK inhibition prolongs temozolomide sensitivity, which can be developed into a precision therapy for the mesenchymal subtype.


Assuntos
Neoplasias Encefálicas , Resistencia a Medicamentos Antineoplásicos , Glioblastoma , Temozolomida , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas Quinases p38 Ativadas por Mitógeno , Temozolomida/farmacologia , Humanos , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Glioblastoma/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Animais , Camundongos , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Antineoplásicos Alquilantes/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Células Tumorais Cultivadas , Proliferação de Células/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Prognóstico
9.
Nat Commun ; 14(1): 4278, 2023 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-37460561

RESUMO

Current technologies to subtype glioblastoma (GBM), the most lethal brain tumor, require highly invasive brain biopsies. Here, we develop a dedicated analytical platform to achieve direct and multiplexed profiling of circulating RNAs in extracellular vesicles for blood-based GBM characterization. The technology, termed 'enzyme ZIF-8 complexes for regenerative and catalytic digital detection of RNA' (EZ-READ), leverages an RNA-responsive transducer to regeneratively convert and catalytically enhance signals from rare RNA targets. Each transducer comprises hybrid complexes - protein enzymes encapsulated within metal organic frameworks - to configure strong catalytic activity and robust protection. Upon target RNA hybridization, the transducer activates directly to liberate catalytic complexes, in a target-recyclable manner; when partitioned within a microfluidic device, these complexes can individually catalyze strong chemifluorescence reactions for digital RNA quantification. The EZ-READ platform thus enables programmable and reliable RNA detection, across different-sized RNA subtypes (miRNA and mRNA), directly in sample lysates. When clinically evaluated, the EZ-READ platform established composite signatures for accurate blood-based GBM diagnosis and subtyping.


Assuntos
Neoplasias Encefálicas , Glioblastoma , MicroRNAs , Humanos , MicroRNAs/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , RNA Mensageiro , Hibridização de Ácido Nucleico , Glioblastoma/genética , Glioblastoma/patologia
10.
J Biol Chem ; 285(38): 29231-8, 2010 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-20630868

RESUMO

Although mutations in the parkin gene are frequently associated with familial Parkinsonism, emerging evidence suggests that parkin also plays a role in cancers as a putative tumor suppressor. Supporting this, we show here that parkin expression is dramatically reduced in several breast cancer-derived cell lines as well as in primary breast cancer tissues. Importantly, we found that ectopic parkin expression in parkin-deficient breast cancer cells mitigates their proliferation rate both in vitro and in vivo, as well as reduces the capacity of these cells to migrate. Cell cycle analysis revealed the arrestment of a significant percentage of parkin-expressing breast cancer cells at the G1-phase. However, we did not observe significant changes in the levels of the G1-associated cyclin D1 and E. On the other hand, the level of cyclin-dependent kinase 6 (CDK6) is dramatically and selectively elevated in parkin-expressing breast cancer cells, the extent of which correlates well with the expression of parkin. Interestingly, a recent study demonstrated that CDK6 restrains the proliferation of breast cancer cells. Taken together, our results support a negative role for parkin in tumorigenesis and provide a potential mechanism by which parkin exerts its suppressing effects on breast cancer cell proliferation.


Assuntos
Neoplasias da Mama/metabolismo , Quinase 6 Dependente de Ciclina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Neoplasias da Mama/genética , Ciclo Celular/genética , Ciclo Celular/fisiologia , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Quinase 6 Dependente de Ciclina/genética , Feminino , Fase G1/genética , Fase G1/fisiologia , Humanos , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Ubiquitina-Proteína Ligases/genética
11.
Cells ; 10(1)2021 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-33466414

RESUMO

Parkinson's disease (PD) is an age-dependent neurodegenerative condition. Leucine-rich repeat kinase 2 (LRRK2) mutations are the most frequent cause of sporadic and autosomal dominant PD. The exact role of LRRK2 protective variants (R1398H, N551K) together with a pathogenic mutant (G2019S) in aging and neurodegeneration is unknown. We generated the following myc-tagged UAS-LRRK2 transgenic Drosophila: LRRK2 (WT), N551K, R1398H, G2019S single allele, and double-mutants (N551K/G2019S or R1398H/G2019S). The protective variants alone were able to suppress the phenotypic effects caused by the pathogenic LRRK2 mutation. Next, we conducted RNA-sequencing using mRNA isolated from dopaminergic neurons of these different groups of transgenic Drosophila. Using pathway enrichment analysis, we identified the top 10 modules (p < 0.05), with "LRRK2 in neurons in Parkinson's disease" among the candidates. Further dissection of this pathway identified the most significantly modulated gene nodes such as eEF1A2, ACTB, eEF1A, and actin cytoskeleton reorganization. The induction of the pathway was successfully restored by the R1398H protective variant and R1398H-G2019S or N551K-G2019S rescue experiments. The oxidoreductase family of genes was also active in the pathogenic mutant and restored in protective and rescue variants. In summary, we provide in vivo evidence supporting the neuroprotective effects of LRRK2 variants. RNA sequencing of dopaminergic neurons identified upregulation of specific gene pathways in the Drosophila carrying the pathogenic variant, and this was restored in the rescue phenotypes. Using protective gene variants, our study identifies potential new targets and provides proof of principle of a new therapeutic approach that will further our understanding of aging and neurodegeneration in PD.


Assuntos
Proteínas de Drosophila/metabolismo , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/metabolismo , Mutação , Doença de Parkinson/metabolismo , Animais , Proteínas de Drosophila/genética , Drosophila melanogaster , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Organismos Geneticamente Modificados , Doença de Parkinson/genética
12.
Theranostics ; 11(11): 5127-5142, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33859738

RESUMO

Hypoxic microenvironment is a hallmark of solid tumors, especially glioblastoma. The strong reliance of glioma-propagating cells (GPCs) on hypoxia-induced survival advantages is potentially exploitable for drug development. Methods: To identify key signaling pathways for hypoxia adaptation by patient-derived GPCs, we performed a kinase inhibitor profiling by screening 188 small molecule inhibitors against 130 different kinases in normoxia and hypoxia. Potential kinase candidates were prioritized for in vitro and in vivo investigations using a ranking algorithm that integrated information from the kinome connectivity network and estimated patients' survival based on expression status. Results: Hypoxic drug screen highlighted extensive modifications of kinomic landscape and a crucial functionality of c-MET-PI3K. c-MET inhibitors diminished phosphorylation of c-MET and PI3K in GPCs subjected to hypoxia, suggesting its role in the hypoxic adaptation of GPCs. Mechanistically, the inhibition of c-MET and PI3K impaired antioxidant defense, leading to oxidative catastrophe and apoptosis. Repurposed c-MET inhibitors PF04217903 and tivantinib exhibited hypoxic-dependent drug synergism with temozolomide, resulting in reduced tumor load and growth of GPC xenografts. Detailed analysis of bulk and single-cell glioblastoma transcriptomes associates the cellular subpopulation over-expressing c-MET with inflamed, hypoxic, metastatic, and stem-like phenotypes. Conclusions: Thus, our "bench to bedside (the use of patient-derived GPCs and xenografts for basic research) and back (validation with independent glioblastoma transcriptome databases)" analysis unravels the novel therapeutic indications of c-MET and PI3K/Akt inhibitors for the treatment of glioblastoma, and potentially other cancers, in the hypoxic tumor microenvironment.


Assuntos
Glioma/genética , Hipóxia/genética , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas c-met/genética , Animais , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Linhagem Celular Tumoral , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Glioma/tratamento farmacológico , Humanos , Hipóxia/tratamento farmacológico , Masculino , Camundongos , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Pirazinas/farmacologia , Pirrolidinonas/farmacologia , Quinolinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Temozolomida/farmacologia , Transcriptoma/efeitos dos fármacos , Transcriptoma/genética , Triazóis/farmacologia
13.
Dev Cell ; 56(20): 2841-2855.e8, 2021 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-34559979

RESUMO

Glioblastoma are heterogeneous tumors composed of highly invasive and highly proliferative clones. Heterogeneity in invasiveness could emerge from discrete biophysical properties linked to specific molecular expression. We identified clones of patient-derived glioma propagating cells that were either highly proliferative or highly invasive and compared their cellular architecture, migratory, and biophysical properties. We discovered that invasiveness was linked to cellular fitness. The most invasive cells were stiffer, developed higher mechanical forces on the substrate, and moved stochastically. The mechano-chemical-induced expression of the formin FMN1 conferred invasive strength that was confirmed in patient samples. Moreover, FMN1 expression was also linked to motility in other cancer and normal cell lines, and its ectopic expression increased fitness parameters. Mechanistically, FMN1 acts from the microtubule lattice and promotes a robust mechanical cohesion, leading to highly invasive motility.


Assuntos
Movimento Celular/fisiologia , Forminas/metabolismo , Glioblastoma/metabolismo , Invasividade Neoplásica/patologia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Proteínas Fetais/metabolismo , Glioblastoma/patologia , Humanos , Proteínas dos Microfilamentos/metabolismo
14.
Sci Adv ; 7(18)2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33931443

RESUMO

Molecular profiling of the most aggressive brain tumor glioblastoma (GBM) on the basis of gene expression, DNA methylation, and genomic variations advances both cancer research and clinical diagnosis. The enhancer architectures and regulatory circuitries governing tumor-intrinsic transcriptional diversity and subtype identity are still elusive. Here, by mapping H3K27ac deposition, we analyze the active regulatory landscapes across 95 GBM biopsies, 12 normal brain tissues, and 38 cell line counterparts. Analyses of differentially regulated enhancers and super-enhancers uncovered previously unrecognized layers of intertumor heterogeneity. Integrative analysis of variant enhancer loci and transcriptome identified topographies of transcriptional enhancers and core regulatory circuitries in four molecular subtypes of primary tumors: AC1-mesenchymal, AC1-classical, AC2-proneural, and AC3-proneural. Moreover, this study reveals core oncogenic dependency on super-enhancer-driven transcriptional factors, long noncoding RNAs, and druggable targets in GBM. Through profiling of transcriptional enhancers, we provide clinically relevant insights into molecular classification, pathogenesis, and therapeutic intervention of GBM.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Cromatina/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Glioblastoma/patologia , Humanos
15.
Stem Cells ; 27(1): 29-39, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18845764

RESUMO

Cancer stem cells have been shown to initiate and sustain tumor growth. In many instances, clinical material is limited, compounded by a lack of methods to preserve such cells at convenient time points. Although brain tumor-initiating cells grown in a spheroid manner have been shown to maintain their integrity through serial transplantation in immune-compromised animals, practically, it is not always possible to have access to animals of suitable ages to continuously maintain these cells. We therefore explored vitrification as a cryopreservation technique for brain tumor-initiating cells. Tumor neurospheres were derived from five patients with glioblastoma multiforme (GBM). Cryopreservation in 90% serum and 10% dimethyl sulfoxide yielded greatest viability and could be explored in future studies. Vitrification yielded cells that maintained self-renewal and multipotentiality properties. Karyotypic analyses confirmed the presence of GBM hallmarks. Upon implantation into NOD/SCID mice, our vitrified cells reformed glioma masses that could be serially transplanted. Transcriptome analysis showed that the vitrified and nonvitrified samples in either the stem-like or differentiated states clustered together, providing evidence that vitrification does not change the genotype of frozen cells. Upon induction of differentiation, the transcriptomes of vitrified cells associated with the original primary tumors, indicating that tumor stem-like cells are a genetically distinct population from the differentiated mass, underscoring the importance of working with the relevant tumor-initiating population. Our results demonstrate that vitrification of brain tumor-initiating cells preserves the biological phenotype and genetic profiles of the cells. This should facilitate the establishment of a repository of tumor-initiating cells for subsequent experimental designs.


Assuntos
Criopreservação , Glioblastoma/patologia , Neurônios/patologia , Esferoides Celulares/patologia , Antígeno AC133 , Animais , Antígenos CD/metabolismo , Agregação Celular , Diferenciação Celular , Proliferação de Células , Forma Celular , Sobrevivência Celular , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Glicoproteínas/metabolismo , Humanos , Cariotipagem , Camundongos , Camundongos SCID , Células-Tronco Multipotentes/patologia , Células-Tronco Neoplásicas/patologia , Peptídeos/metabolismo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
17.
J Neurosurg ; 111(2): 238-46, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19199503

RESUMO

OBJECT: The study of tumor cell growth and invasion in cancer biology is often limited by the inability to visualize tumor cell behavior in real time in animal models. The authors provide evidence that glioma cells are heterogeneous,with a subset responsible for increased invasiveness. The use of bioluminescence (BL) imaging to investigate dynamic aspects of glioma progression are discussed. METHODS: Glioblastoma multiforme-initiating cells were generated under conditions typically used to sustain neural stem cells. The invasiveness potential was determined using a Matrigel chamber. The presence of an "invasiveness gene signature" that correlated with patient survival outcome was ascertained through microarray gene expression analysis. To measure invasiveness, the authors devised a method focussed on BL imaging and tested it in vitro and in vivo using a zebrafish xenograft model. Bioluminescence imaging signals were verified using known inhibitors of glioma growth: AEE788, N-[(3,5-Difluorophenyl)acetyl]-L-alanyl-2-phenylglycine-1,1-dimethylethyl ester, and compound E. RESULTS: The authors' data support the idea that glioblastoma multiforme-initiating cells are heterogeneous and possess an invasive subset; BL imaging was used as a readout method to assess this invasive subset. The in vitro data obtained using a known glioma growth inhibitor, AEE788, showed that BL imaging could detect cellular movement and invasion even before overall cell death was detectable on conventional viability assays. Further work using a zebrafish tumor xenograft model supported the efficacy of BL imaging in monitoring changes in tumor load. CONCLUSIONS: The authors used optically transparent zebrafish and high-resolution confocal imaging to track tumor growth in vivo and demonstrate the efficacy of this model for screening antitumor and antiangiogenic compounds.The integration of zebrafish transgenic technology into human cancer biological studies may aid in the development of cancer models targeting specific organs, tissues, or cell types within tumors. Zebrafish could also provide a cost-effective means for the rapid development of therapeutic agents directed at blocking tumor growth and invasion.


Assuntos
Glioblastoma/patologia , Medições Luminescentes/métodos , Invasividade Neoplásica/patologia , Animais , Movimento Celular , Progressão da Doença , Humanos , Invasividade Neoplásica/genética , Análise de Sequência com Séries de Oligonucleotídeos , Purinas/farmacologia , Transplante Heterólogo , Células Tumorais Cultivadas , Peixe-Zebra
18.
Nat Commun ; 10(1): 3601, 2019 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-31399589

RESUMO

Intratumoral heterogeneity is a hallmark of glioblastoma (GBM) tumors, thought to negatively influence therapeutic outcome. Previous studies showed that mesenchymal tumors have a worse outcome than the proneural subtype. Here we focus on STAT3 as its activation precedes the proneural-mesenchymal transition. We first establish a STAT3 gene signature that stratifies GBM patients into STAT3-high and -low cohorts. STAT3 inhibitor treatment selectively mitigates STAT3-high cell viability and tumorigenicity in orthotopic mouse xenograft models. We show the mechanism underlying resistance in STAT3-low cells by combining STAT3 signature analysis with kinome screen data on STAT3 inhibitor-treated cells. This allows us to draw connections between kinases affected by STAT3 inhibitors, their associated transcription factors and target genes. We demonstrate that dual inhibition of IGF-1R and STAT3 sensitizes STAT3-low cells and improves survival in mice. Our study underscores the importance of serially profiling tumors so as to accurately target individuals who may demonstrate molecular subtype switching.


Assuntos
Predisposição Genética para Doença/genética , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Animais , Sobrevivência Celular , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Humanos , Imidazóis/farmacologia , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Camundongos , Pirazinas/farmacologia , Pirazóis/farmacologia , Pirimidinas/farmacologia , Receptor IGF Tipo 1/antagonistas & inibidores , Receptor IGF Tipo 1/genética , Fator de Transcrição STAT3/antagonistas & inibidores , Temozolomida/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Transl Stroke Res ; 10(1): 91-103, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-29569041

RESUMO

The transient receptor potential melastatin 4 (TRPM4) channel has been suggested to play a key role in the treatment of ischemic stroke. However, in vivo evaluation of TRPM4 channel, in particular by direct channel suppression, is lacking. In this study, we used multimodal imaging to assess edema formation and quantify the amount of metabolically functional brain salvaged after a rat model of stroke reperfusion. TRPM4 upregulation in endothelium emerges as early as 2 h post-stroke induction. Expression of TRPM4 channel was suppressed directly in vivo by treatment with siRNA; scrambled siRNA was used as a control. T2-weighted MRI suggests that TRPM4 inhibition successfully reduces edema by 30% and concomitantly salvages functionally active brain, measured by 18F-FDG-PET. These in vivo imaging results correlate well with post-mortem 2,3,5-triphenyltetrazolium chloride (TTC) staining which exhibits a 34.9% reduction in infarct volume after siRNA treatment. Furthermore, in a permanent stroke model, large areas of brain tissue displayed both edema and significant reductions in metabolic activity which was not shown in transient models with or without TRPM4 inhibition, indicating that tissue salvaged by TRPM4 inhibition during stroke reperfusion may survive. Evans Blue extravasation and hemoglobin quantification in the ipsilateral hemisphere were greatly reduced, suggesting that TRPM4 inhibition can improve BBB integrity after ischemic stroke reperfusion. Our results support the use of TRPM4 blocker for early stroke reperfusion.


Assuntos
Regulação da Expressão Gênica/fisiologia , Infarto da Artéria Cerebral Média/tratamento farmacológico , Imagem Multimodal/métodos , Traumatismo por Reperfusão/tratamento farmacológico , Canais de Cátion TRPM/metabolismo , Animais , Barreira Hematoencefálica/patologia , Barreira Hematoencefálica/fisiopatologia , Edema Encefálico , Modelos Animais de Doenças , Fluordesoxiglucose F18/farmacocinética , Lateralidade Funcional , Processamento de Imagem Assistida por Computador , Infarto da Artéria Cerebral Média/complicações , Masculino , Análise em Microsséries , Fosfopiruvato Hidratase/metabolismo , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/uso terapêutico , Ratos , Ratos Wistar , Traumatismo por Reperfusão/complicações , Canais de Cátion TRPM/antagonistas & inibidores , Canais de Cátion TRPM/genética , Fator de von Willebrand/metabolismo
20.
Oncogene ; 38(27): 5367-5380, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30967630

RESUMO

Glioblastoma (GBM) is the most aggressive tumor of the brain. NF1, a tumor suppressor gene and RAS-GTPase, is one of the highly mutated genes in GBM. Dysregulated NF1 expression promotes cell invasion, proliferation, and tumorigenesis. Loss of NF1 expression in glioblastoma is associated with increased aggressiveness of the tumor. Here, we show that NF1-loss in patient-derived glioma cells using shRNA increases self-renewal, heightens cell invasion, and promotes mesenchymal subtype and epithelial mesenchymal transition-specific gene expression that enhances tumorigenesis. The neurofibromin protein contains at least four major domains, with the GAP-related domain being the most well-studied. In this study, we report that the leucine-rich domain (LRD) of neurofibromin inhibits invasion of human glioblastoma cells without affecting their proliferation. Moreover, under conditions tested, the NF1-LRD fails to hydrolyze Ras-GTP to Ras-GDP, suggesting that its suppressive function is independent of Ras signaling. We further demonstrate that rare variants within the NF1-LRD domain found in a subset of the patients are pathogenic and reduce NF1-LRD's invasion suppressive function. Taken together, our results show, for the first time, that NF1-LRD inhibits glioma invasion, and provides evidence of a previously unrecognized function of NF1-LRD in glioma biology.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Leucina/metabolismo , Mutação , Invasividade Neoplásica/genética , Neurofibromina 1/genética , Animais , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Glioblastoma/patologia , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Neurofibromina 1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA