Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 16 de 16
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Mol Cancer Ther ; : OF1-OF13, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38904222

RÉSUMÉ

KRAS is the most frequently mutated oncogene in human cancer and facilitates uncontrolled growth through hyperactivation of the receptor tyrosine kinase (RTK)/mitogen-activated protein kinase (MAPK) pathway. The Son of Sevenless homolog 1 (SOS1) protein functions as a guanine nucleotide exchange factor (GEF) for the RAS subfamily of small GTPases and represents a druggable target in the pathway. Using a structure-based drug discovery approach, MRTX0902 was identified as a selective and potent SOS1 inhibitor that disrupts the KRAS:SOS1 protein-protein interaction to prevent SOS1-mediated nucleotide exchange on KRAS and translates into an anti-proliferative effect in cancer cell lines with genetic alterations of the KRAS-MAPK pathway. MRTX0902 augmented the antitumor activity of the KRAS G12C inhibitor adagrasib when dosed in combination in eight out of 12 KRAS G12C-mutant human non-small cell lung cancer and colorectal cancer xenograft models. Pharmacogenomic profiling in preclinical models identified cell cycle genes and the SOS2 homolog as genetic co-dependencies and implicated tumor suppressor genes (NF1 and PTEN) in resistance following combination treatment. Lastly, combined vertical inhibition of RTK/MAPK pathway signaling by MRTX0902 with inhibitors of EGFR or RAF/MEK led to greater downregulation of pathway signaling and improved antitumor responses in KRAS-MAPK pathway-mutant models. These studies demonstrate the potential clinical application of dual inhibition of SOS1 and KRAS G12C and additional SOS1 combination strategies that will aide in the understanding of SOS1 and RTK/MAPK biology in targeted cancer therapy.

2.
Mol Cancer ; 23(1): 123, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38849845

RÉSUMÉ

BACKGROUND: Pediatric-type diffuse high-grade glioma (pHGG) is the most frequent malignant brain tumor in children and can be subclassified into multiple entities. Fusion genes activating the MET receptor tyrosine kinase often occur in infant-type hemispheric glioma (IHG) but also in other pHGG and are associated with devastating morbidity and mortality. METHODS: To identify new treatment options, we established and characterized two novel orthotopic mouse models harboring distinct MET fusions. These included an immunocompetent, murine allograft model and patient-derived orthotopic xenografts (PDOX) from a MET-fusion IHG patient who failed conventional therapy and targeted therapy with cabozantinib. With these models, we analyzed the efficacy and pharmacokinetic properties of three MET inhibitors, capmatinib, crizotinib and cabozantinib, alone or combined with radiotherapy. RESULTS: Capmatinib showed superior brain pharmacokinetic properties and greater in vitro and in vivo efficacy than cabozantinib or crizotinib in both models. The PDOX models recapitulated the poor efficacy of cabozantinib experienced by the patient. In contrast, capmatinib extended survival and induced long-term progression-free survival when combined with radiotherapy in two complementary mouse models. Capmatinib treatment increased radiation-induced DNA double-strand breaks and delayed their repair. CONCLUSIONS: We comprehensively investigated the combination of MET inhibition and radiotherapy as a novel treatment option for MET-driven pHGG. Our seminal preclinical data package includes pharmacokinetic characterization, recapitulation of clinical outcomes, coinciding results from multiple complementing in vivo studies, and insights into molecular mechanism underlying increased efficacy. Taken together, we demonstrate the groundbreaking efficacy of capmatinib and radiation as a highly promising concept for future clinical trials.


Sujet(s)
Tumeurs du cerveau , Gliome , Protéines proto-oncogènes c-met , Tests d'activité antitumorale sur modèle de xénogreffe , Animaux , Humains , Gliome/anatomopathologie , Gliome/traitement médicamenteux , Gliome/génétique , Gliome/thérapie , Protéines proto-oncogènes c-met/antagonistes et inhibiteurs , Protéines proto-oncogènes c-met/génétique , Protéines proto-oncogènes c-met/métabolisme , Souris , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/génétique , Tumeurs du cerveau/radiothérapie , Benzamides/pharmacologie , Benzamides/usage thérapeutique , Lignée cellulaire tumorale , Protéines de fusion oncogènes/génétique , Protéines de fusion oncogènes/métabolisme , Femelle , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Pyridines/pharmacologie , Pyridines/usage thérapeutique , Crizotinib/pharmacologie , Crizotinib/usage thérapeutique , Modèles animaux de maladie humaine , Enfant , Grading des tumeurs , Anilides/pharmacologie , Imidazoles , Triazines
3.
Science ; 384(6700): eadk0775, 2024 06 07.
Article de Anglais | MEDLINE | ID: mdl-38843331

RÉSUMÉ

How the KRAS oncogene drives cancer growth remains poorly understood. Therefore, we established a systemwide portrait of KRAS- and extracellular signal-regulated kinase (ERK)-dependent gene transcription in KRAS-mutant cancer to delineate the molecular mechanisms of growth and of inhibitor resistance. Unexpectedly, our KRAS-dependent gene signature diverges substantially from the frequently cited Hallmark KRAS signaling gene signature, is driven predominantly through the ERK mitogen-activated protein kinase (MAPK) cascade, and accurately reflects KRAS- and ERK-regulated gene transcription in KRAS-mutant cancer patients. Integration with our ERK-regulated phospho- and total proteome highlights ERK deregulation of the anaphase promoting complex/cyclosome (APC/C) and other components of the cell cycle machinery as key processes that drive pancreatic ductal adenocarcinoma (PDAC) growth. Our findings elucidate mechanistically the critical role of ERK in driving KRAS-mutant tumor growth and in resistance to KRAS-ERK MAPK targeted therapies.


Sujet(s)
Carcinome du canal pancréatique , Extracellular Signal-Regulated MAP Kinases , Régulation de l'expression des gènes tumoraux , Système de signalisation des MAP kinases , Mutation , Tumeurs du pancréas , Protéines proto-oncogènes p21(ras) , Transcriptome , Animaux , Humains , Souris , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/métabolisme , Lignée cellulaire tumorale , Résistance aux médicaments antinéoplasiques/génétique , Extracellular Signal-Regulated MAP Kinases/métabolisme , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Protéines proto-oncogènes p21(ras)/génétique , Protéines proto-oncogènes p21(ras)/métabolisme , Cellules HEK293
4.
Cancer Cell ; 42(3): 413-428.e7, 2024 Mar 11.
Article de Anglais | MEDLINE | ID: mdl-38402609

RÉSUMÉ

KRASG12C inhibitors (adagrasib and sotorasib) have shown clinical promise in targeting KRASG12C-mutated lung cancers; however, most patients eventually develop resistance. In lung patients with adenocarcinoma with KRASG12C and STK11/LKB1 co-mutations, we find an enrichment of the squamous cell carcinoma gene signature in pre-treatment biopsies correlates with a poor response to adagrasib. Studies of Lkb1-deficient KRASG12C and KrasG12D lung cancer mouse models and organoids treated with KRAS inhibitors reveal tumors invoke a lineage plasticity program, adeno-to-squamous transition (AST), that enables resistance to KRAS inhibition. Transcriptomic and epigenomic analyses reveal ΔNp63 drives AST and modulates response to KRAS inhibition. We identify an intermediate high-plastic cell state marked by expression of an AST plasticity signature and Krt6a. Notably, expression of the AST plasticity signature and KRT6A at baseline correlates with poor adagrasib responses. These data indicate the role of AST in KRAS inhibitor resistance and provide predictive biomarkers for KRAS-targeted therapies in lung cancer.


Sujet(s)
Acétonitriles , Carcinome épidermoïde , Tumeurs du poumon , Pipérazines , Pyrimidines , Animaux , Souris , Humains , Protéines proto-oncogènes p21(ras) , Gènes ras , Mutation
5.
Cancer Discov ; 13(11): 2412-2431, 2023 11 01.
Article de Anglais | MEDLINE | ID: mdl-37552839

RÉSUMÉ

Previous studies implicated protein arginine methyltransferase 5 (PRMT5) as a synthetic lethal target for MTAP-deleted (MTAP del) cancers; however, the pharmacologic characterization of small-molecule inhibitors that recapitulate the synthetic lethal phenotype has not been described. MRTX1719 selectively inhibited PRMT5 in the presence of MTA, which is elevated in MTAP del cancers, and inhibited PRMT5-dependent activity and cell viability with >70-fold selecti-vity in HCT116 MTAP del compared with HCT116 MTAP wild-type (WT) cells. MRTX1719 demonstrated dose-dependent antitumor activity and inhibition of PRMT5-dependent SDMA modification in MTAP del tumors. In contrast, MRTX1719 demonstrated minimal effects on SDMA and viability in MTAP WT tumor xenografts or hematopoietic cells. MRTX1719 demonstrated marked antitumor activity across a panel of xenograft models at well-tolerated doses. Early signs of clinical activity were observed including objective responses in patients with MTAP del melanoma, gallbladder adenocarcinoma, mesothelioma, non-small cell lung cancer, and malignant peripheral nerve sheath tumors from the phase I/II study. SIGNIFICANCE: PRMT5 was identified as a synthetic lethal target for MTAP del cancers; however, previous PRMT5 inhibitors do not selectively target this genotype. The differentiated binding mode of MRTX1719 leverages the elevated MTA in MTAP del cancers and represents a promising therapy for the ∼10% of patients with cancer with this biomarker. See related commentary by Mulvaney, p. 2310. This article is featured in Selected Articles from This Issue, p. 2293.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Humains , Lignée cellulaire tumorale , Mutations synthétiques létales , Antienzymes/pharmacologie , Protein-arginine N-methyltransferases
6.
Nat Commun ; 12(1): 4089, 2021 07 02.
Article de Anglais | MEDLINE | ID: mdl-34215733

RÉSUMÉ

Pediatric high-grade glioma (pHGG) is a major contributor to cancer-related death in children. In vitro and in vivo disease models reflecting the intimate connection between developmental context and pathogenesis of pHGG are essential to advance understanding and identify therapeutic vulnerabilities. Here we report establishment of 21 patient-derived pHGG orthotopic xenograft (PDOX) models and eight matched cell lines from diverse groups of pHGG. These models recapitulate histopathology, DNA methylation signatures, mutations and gene expression patterns of the patient tumors from which they were derived, and include rare subgroups not well-represented by existing models. We deploy 16 new and existing cell lines for high-throughput screening (HTS). In vitro HTS results predict variable in vivo response to PI3K/mTOR and MEK pathway inhibitors. These unique new models and an online interactive data portal for exploration of associated detailed molecular characterization and HTS chemical sensitivity data provide a rich resource for pediatric brain tumor research.


Sujet(s)
Hétérogénéité génétique/effets des médicaments et des substances chimiques , Gliome/traitement médicamenteux , Gliome/génétique , Animaux , Tumeurs du cerveau , Lignée cellulaire tumorale , Prolifération cellulaire , Enfant , Modèles animaux de maladie humaine , Régulation de l'expression des gènes tumoraux , Gliome/anatomopathologie , Tests de criblage à haut débit , Humains , Souris , Mutation , Inhibiteurs de protéines kinases/usage thérapeutique , Sérine-thréonine kinases TOR , Tests d'activité antitumorale sur modèle de xénogreffe
7.
Int J Radiat Oncol Biol Phys ; 106(4): 838-847, 2020 03 15.
Article de Anglais | MEDLINE | ID: mdl-31785339

RÉSUMÉ

PURPOSE: Optimal radiation therapy (RT) target margins for diffuse intrinsic pontine glioma (DIPG) are unknown. We sought to define disease progression patterns in a contemporary cohort treated with conformal RT using different clinical target volume (CTV) margins. METHODS AND MATERIALS: We reviewed 105 patients with newly diagnosed DIPG treated with conformal conventionally fractionated RT at our institution from 2006 to 2014. CTV margins were classified as standard (1 cm) for 60 patients and extended (2-3 cm) for 45 patients. Survival and cumulative incidence of progression in treatment groups were compared by log-rank and Gray's tests, respectively. Cox proportional hazard models identified predictors of survival. RESULTS: For 97 patients evaluated with magnetic resonance imaging at progression, the cumulative incidences of isolated local, isolated distant, and synchronous disease progression at 1 year were 62.6%, 12.3%, and 7.2%, respectively, and did not differ significantly according to the CTV margin. Central dosimetric progression (Vprogression95% ≥95%) was observed in 80 of 81 evaluable patients. Median progression-free survival and overall survival (OS) were 7.6 months (95% confidence interval, 6.9-8.2) and 11.3 months (95% confidence interval, 10.0-12.8), respectively, and did not differ significantly according to margin status. DIPG survival prediction risk group (standard vs high, P = .02; intermediate vs high, P = .009) and development of distant metastasis (P = .003) were independent predictors of OS. For the 41 patients (39%) with a pathologic diagnosis, H3.3 K27M mutation was associated with shorter OS (hazard ratio [HR], 0.41; P =.02), whereas H3.1 K27M and ACVR1 mutations were associated with longer OS (HR, 3.56; P =.004 and HR, 2.58; P =.04, respectively). CONCLUSIONS: All patients who experienced local failure showed progression within the high-dose volume, and there was no apparent survival or tumor-control benefit to extending the CTV margins beyond 1 cm. Given the increasing use of reirradiation, standardizing the CTV margin to 1 cm may improve retreatment tolerance.


Sujet(s)
Tumeurs du tronc cérébral/radiothérapie , Gliome infiltrant du tronc cérébral/radiothérapie , Radiothérapie conformationnelle , Tumeurs du tronc cérébral/imagerie diagnostique , Tumeurs du tronc cérébral/anatomopathologie , Enfant , Enfant d'âge préscolaire , Gliome infiltrant du tronc cérébral/imagerie diagnostique , Gliome infiltrant du tronc cérébral/anatomopathologie , Évolution de la maladie , Femelle , Humains , Mâle , Études rétrospectives , Analyse de survie , Tomodensitométrie
8.
Nat Commun ; 10(1): 3718, 2019 08 16.
Article de Anglais | MEDLINE | ID: mdl-31420543

RÉSUMÉ

High throughput omics approaches provide an unprecedented opportunity for dissecting molecular mechanisms in cancer biology. Here we present deep profiling of whole proteome, phosphoproteome and transcriptome in two high-grade glioma (HGG) mouse models driven by mutated RTK oncogenes, PDGFRA and NTRK1, analyzing 13,860 proteins and 30,431 phosphosites by mass spectrometry. Systems biology approaches identify numerous master regulators, including 41 kinases and 23 transcription factors. Pathway activity computation and mouse survival indicate the NTRK1 mutation induces a higher activation of AKT downstream targets including MYC and JUN, drives a positive feedback loop to up-regulate multiple other RTKs, and confers higher oncogenic potency than the PDGFRA mutation. A mini-gRNA library CRISPR-Cas9 validation screening shows 56% of tested master regulators are important for the viability of NTRK-driven HGG cells, including TFs (Myc and Jun) and metabolic kinases (AMPKa1 and AMPKa2), confirming the validity of the multiomics integrative approaches, and providing novel tumor vulnerabilities.


Sujet(s)
Tumeurs du cerveau/génétique , Analyse de profil d'expression de gènes , Gliome/génétique , Protéomique , AMP-Activated Protein Kinases/métabolisme , Animaux , Tumeurs du cerveau/métabolisme , Modèles animaux de maladie humaine , Rétrocontrôle physiologique , Gliome/métabolisme , Souris , Mutation , Protéine oncogène p65(gag-jun)/métabolisme , Phosphopeptides/métabolisme , Phosphoprotéines/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-myc/métabolisme , Récepteur au PDGF alpha/génétique , Récepteur trkA/génétique , Transduction du signal , Biologie des systèmes , Régulation positive
9.
Acta Neuropathol ; 137(1): 123-137, 2019 01.
Article de Anglais | MEDLINE | ID: mdl-30267146

RÉSUMÉ

Double minute chromosomes are extrachromosomal circular DNA fragments frequently found in brain tumors. To understand their evolution, we characterized the double minutes in paired diagnosis and relapse tumors from a pediatric high-grade glioma and four adult glioblastoma patients. We determined the full structures of the major double minutes using a novel approach combining multiple types of supporting genomic evidence. Among the double minutes identified in the pediatric patient, only one carrying EGFR was maintained at high abundance in both samples, whereas two others were present in only trace amounts at diagnosis but abundant at relapse, and the rest were found either in the relapse sample only or in the diagnosis sample only. For the EGFR-carrying double minutes, we found a secondary somatic deletion in all copies at relapse, after erlotinib treatment. However, the somatic mutation was present at very low frequency at diagnosis, suggesting potential resistance to the EGFR inhibitor. This mutation caused an in-frame RNA transcript to skip exon 16, a novel transcript isoform absent in EST database, as well as about 700 RNA-seq of normal brains that we reviewed. We observed similar patterns involving longitudinal copy number shift of double minutes in another four pairs (diagnosis/relapse) of adult glioblastoma. Overall, in three of five paired tumor samples, we found that although the same oncogenes were amplified at diagnosis and relapse, they were amplified on different double minutes. Our results suggest that double minutes readily evolve, increasing tumor heterogeneity rapidly. Understanding patterns of double minute evolution can shed light on future therapeutic solutions to brain tumors carrying such variants.


Sujet(s)
Tumeurs du cerveau/diagnostic , Encéphale/anatomopathologie , Glioblastome/génétique , Récidive tumorale locale/anatomopathologie , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Enfant , Génomique , Glioblastome/diagnostic , Gliome/génétique , Humains , Mâle , Mutation/génétique , Récidive tumorale locale/diagnostic , Récidive tumorale locale/génétique , Récidive
10.
Neuro Oncol ; 18(7): 928-38, 2016 07.
Article de Anglais | MEDLINE | ID: mdl-26683138

RÉSUMÉ

BACKGROUND: Improved therapies for high-grade glioma (HGG) are urgently needed as the median survival for grade IV gliomas is only 15 months. Bone morphogenetic protein (BMP) signaling plays critical and complex roles in many types of cancer, including glioma, with most of the recently published work focusing on BMP-mediated regulation of glioma stem cells (GSCs). We hypothesized that BMP signaling may be an important modulator of tumorigenic properties in glioma cells outside of the GSC compartment. METHODS: We used a human HGG tissue microarray and performed immunohistochemistry for phospho-Smads1,5,8. To examine the role of BMP signaling in tumorigenic astrocytes, transgenic mice were used to delete the BMP type IA receptor (Bmpr1a) and generate astrocytes transformed with oncogenic Ras and homozygous deletion of p53. The cells were transplanted orthotopically into immunocompetent adult host mice. RESULTS: First we established that BMP signaling is active within the vast majority of HGG tumor cells. Mice implanted with BMPR1a-knockout transformed astrocytes showed an increase in median survival compared with mice that received BMPR1a-intact transformed astrocytes (52.5 vs 16 days). In vitro analysis showed that deletion of BMPR1a in oncogenic astrocytes resulted in decreased proliferation, decreased invasion, decreased migration, and increased expression of stemness markers. In addition, inhibition of BMP signaling in murine cells and astrocytoma cells with a small molecule BMP receptor kinase inhibitor resulted in similar tumor suppressive effects in vitro. CONCLUSION: BMP inhibition may represent a viable therapeutic approach in adult HGG.


Sujet(s)
Astrocytes/métabolisme , Récepteurs de la protéine morphogénique osseuse de type I/génétique , Protéines morphogénétiques osseuses/métabolisme , Carcinogenèse/génétique , Régulation de l'expression des gènes/génétique , Gliome/métabolisme , Transduction du signal , Animaux , Prolifération cellulaire , Transformation cellulaire néoplasique , Gliome/génétique , Souris transgéniques
11.
Cancer Lett ; 368(1): 79-87, 2015 Nov 01.
Article de Anglais | MEDLINE | ID: mdl-26235139

RÉSUMÉ

The bone morphogenetic protein (BMP) pathway belonging to the Transforming Growth Factor beta (TGFß) family of secreted cytokines/growth factors is an important regulator of cancer. BMP ligands have been shown to play both tumor suppressive and promoting roles in human cancers. We have found that BMP ligands are amplified in human ovarian cancers and that BMP receptor expression correlates with poor progression-free-survival (PFS). Furthermore, active BMP signaling has been observed in human ovarian cancer tissue. We also determined that ovarian cancer cell lines have active BMP signaling in a cell autonomous fashion. Inhibition of BMP signaling with a small molecule receptor kinase antagonist is effective at reducing ovarian tumor sphere growth. Furthermore, BMP inhibition can enhance sensitivity to Cisplatin treatment and regulates gene expression involved in platinum resistance in ovarian cancer. Overall, these studies suggest targeting the BMP pathway as a novel source to enhance chemo-sensitivity in ovarian cancer.


Sujet(s)
Antinéoplasiques/pharmacologie , Protéines morphogénétiques osseuses/antagonistes et inhibiteurs , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tumeurs de l'ovaire/métabolisme , Pyrazoles/pharmacologie , Quinoléines/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Récepteurs de la protéine morphogénique osseuse/métabolisme , Protéines morphogénétiques osseuses/génétique , Protéines morphogénétiques osseuses/métabolisme , Cisplatine/pharmacologie , Survie sans rechute , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/anatomopathologie , Sphéroïdes de cellules , Cellules cancéreuses en culture
12.
Oncotarget ; 6(26): 22890-904, 2015 Sep 08.
Article de Anglais | MEDLINE | ID: mdl-26274893

RÉSUMÉ

Bone Morphogenetic Proteins (BMPs) are secreted cytokines/growth factors belonging to the Transforming Growth Factor ß (TGFß) family. BMP ligands have been shown to be overexpressed in human breast cancers. Normal and cancerous breast tissue display active BMP signaling as indicated by phosphorylated Smads 1, 5 and 9. We combined mice expressing the MMTV.PyMT oncogene with mice having conditional knockout (cKO) of BMP receptor type 1a (BMPR1a) using whey acidic protein (WAP)-Cre and found this deletion resulted in delayed tumor onset and significantly extended survival. Immunofluorescence staining revealed that cKO tumors co-expressed Keratin 5 and mesenchymal cell markers such as Vimentin. This indicates that epithelial-to-mesenchymal (EMT)-like transitions occurred in cKO tumors. We performed microarray analysis on these tumors and found changes that support EMT-like changes. We established primary tumor cell lines and found that BMPR1a cKO had slower growth in vitro and in vivo upon implantation. cKO tumor cells had reduced migration in vitro. We analyzed human databases from TCGA and survival data from microarrays to confirm BMPR1a tumor promoting functions, and found that high BMPR1a gene expression correlates with decreased survival regardless of molecular breast cancer subtype. In conclusion, the data indicate that BMP signaling through BMPR1a functions as a tumor promoter.


Sujet(s)
Récepteurs de la protéine morphogénique osseuse de type I/déficit , Tumeurs du sein/métabolisme , Animaux , Récepteurs de la protéine morphogénique osseuse de type I/génétique , Récepteurs de la protéine morphogénique osseuse de type I/métabolisme , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Humains , Souris , Souris de lignée C57BL , Analyse sur microréseau , Métastase tumorale , Transduction du signal
13.
Transl Oncogenomics ; 7: 1-9, 2015.
Article de Anglais | MEDLINE | ID: mdl-25987829

RÉSUMÉ

Glioblastoma multiforme (GBM) is a grade IV glioma with a median survival of 15 months. Recently, bone morphogenetic protein (BMP) signaling has been shown to promote survival in xenograft murine models. To gain a better understanding of the role of BMP signaling in human GBMs, we examined the genomic alterations of 90 genes associated with BMP signaling in GBM patient samples. We completed this analysis using publically available datasets compiled through Te Cancer Genome Atlas and the Glioma Molecular Diagnostic Initiative. Here we show how mRNA expression is altered in GBM samples and how that is associated with patient survival, highlighting both known and novel associations between BMP signaling and GBM biology.

14.
Mol Oncol ; 9(1): 179-91, 2015 Jan.
Article de Anglais | MEDLINE | ID: mdl-25205038

RÉSUMÉ

Bone Morphogenetic Protein (BMP) receptors mediate a diverse range of signals to regulate both development and disease. BMP activity has been linked to both tumor promoting and suppressive functions in both tumor cells and their surrounding microenvironment. We sought to investigate the requirement for BMPR2 in stromal fibroblasts during mammary tumor formation and metastasis. We utilized FSP1 (Fibroblast Specific Protein-1) promoter driven Cre to genetically delete BMPR2 in mice expressing the MMTV.PyVmT mammary carcinoma oncogene. We found that abrogation of stromal BMPR2 expression via FSP1 driven Cre resulted in increased tumor metastasis. Additionally, similar to epithelial BMPR2 abrogation, stromal loss of BMPR2 results in increased inflammatory cell infiltration. We proceeded to isolate and establish fibroblast cell lines without BMPR2 and found a cell autonomous increase in inflammatory cytokine secretion. Fibroblasts were co-implanted with syngeneic tumor cells and resulted in accelerated tumor growth and increased metastasis when fibroblasts lacked BMPR2. We observed that the loss of BMPR2 results in increased chemokine expression, which facilitates inflammation by a sustained increase in myeloid cells. The chemokines increased in BMPR2 deleted cells correlated with poor outcome in human breast cancer patients. We conclude that BMPR2 has tumor suppressive functions in the stroma by regulating inflammation.


Sujet(s)
Récepteurs de la protéine morphogénique osseuse de type II/génétique , Fibroblastes/métabolisme , Délétion de gène , Tumeurs expérimentales de la mamelle/métabolisme , Protéines suppresseurs de tumeurs/génétique , Animaux , Récepteurs de la protéine morphogénique osseuse de type II/métabolisme , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Femelle , Fibroblastes/anatomopathologie , Humains , Inflammation/génétique , Inflammation/métabolisme , Inflammation/anatomopathologie , Tumeurs expérimentales de la mamelle/génétique , Tumeurs expérimentales de la mamelle/anatomopathologie , Souris , Métastase tumorale , Protéines suppresseurs de tumeurs/métabolisme
15.
J Neuropathol Exp Neurol ; 71(8): 729-40, 2012 Aug.
Article de Anglais | MEDLINE | ID: mdl-22805776

RÉSUMÉ

Human high-grade gliomas (HGGs) are known for their histologic diversity. To address the role of cell of origin in glioma phenotype, transgenic mice were generated in which oncogenic Ras and p53 deletion were targeted to neural stem/progenitor cells (NSPCs) and mature astrocytes. The hGFAP-Cre/Kras/p53 mice develop multifocal HGGs that vary histopathologically and with respect to the expression of markers associated with NSPCs. One HGG pattern strongly expressed markers of NSPCs and arose near the subventricular zone. Additional nonoverlapping patterns that recapitulate human HGG variants were present simultaneously in the same brain. These neoplastic foci were more often cortical or leptomeningeal based, and the neoplastic cells lacked expression of NSPC markers. To determine whether cell of origin determines tumor phenotype, astrocytes and NSPCs were harvested from neonatal mutant pups. Onorthotopic transplantation, early-passage astrocytes and NSPCs formed tumors that differed in engraftment rates, latency to clinical signs, histopathology, and protein expression. Astrocyte-derivedtumors were more aggressive, had giant-cell histology, and glial fibrillary acidic protein expression. The NSPC-derived tumors retained NSPC markers and showed evidence of differentiation along astrocytic, oligodendroglial, and neuronal lineages. These results indicate that identical tumorigenic stimuli produce markedly different glioma phenotypes, depending on the differentiation status of the transformed cell.


Sujet(s)
Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Gènes p53/génétique , Gliome/génétique , Gliome/anatomopathologie , Protéines G ras/génétique , Animaux , Astrocytes/anatomopathologie , Astrocytes/physiologie , Technique de Western , Lignage cellulaire , Technique d'immunofluorescence , Protéine gliofibrillaire acide/génétique , Protéine gliofibrillaire acide/métabolisme , Immunohistochimie , Souris , Souris knockout , Souris transgéniques , Transplantation tumorale , Réaction de polymérisation en chaine en temps réel , Recombinaison génétique , Analyse de survie
16.
Cancer Res ; 71(15): 5276-86, 2011 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-21653679

RÉSUMÉ

Vitamin D(3) is a promising preventative and therapeutic agent for prostate cancer, but its implementation is hampered by a lack of understanding about its mechanism of action. Upon treatment with 1α,25-dihydroxyvitamin D(3) [1,25(OH)(2)D(3), vitamin D(3)], the metabolically active form of vitamin D(3), adult prostate progenitor/stem cells (PrP/SC) undergo cell-cycle arrest, senescence, and differentiation to an androgen receptor-positive luminal epithelial cell fate. Microarray analyses of control- and vitamin D(3)-treated PrP/SCs revealed global gene expression signatures consistent with induction of differentiation. Interestingly, one of the most highly upregulated genes by vitamin D(3) was the proinflammatory cytokine interleukin-1α (IL-1α). Systems biology analyses supported a central role for IL-1α in the vitamin D(3) response in PrP/SCs. siRNA-mediated knockdown of IL-1α abrogated vitamin D(3)-induced growth suppression, establishing a requirement for IL-1α in the antiproliferative effects of vitamin D(3) in PrP/SCs. These studies establish a system to study the molecular profile of PrP/SC differentiation, proliferation, and senescence, and they point to an important new role for IL-1α in vitamin D(3) signaling in PrP/SCs.


Sujet(s)
Calcitriol/pharmacologie , Interleukine-1 alpha/physiologie , Prostate/cytologie , Cellules souches/effets des médicaments et des substances chimiques , Animaux , Cycle cellulaire/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Division cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées/effets des médicaments et des substances chimiques , Cellules cultivées/métabolisme , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Test clonogénique , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Gènes du rétinoblastome , Gènes p16 , Interleukine-1 alpha/biosynthèse , Interleukine-1 alpha/génétique , Mâle , Souris , Souris knockout , Séquençage par oligonucléotides en batterie , Prostate/effets des médicaments et des substances chimiques , Petit ARN interférent/pharmacologie , Récepteur calcitriol/déficit , Récepteur calcitriol/génétique , Cellules souches/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE