Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Cancers (Basel) ; 13(13)2021 Jul 02.
Article in English | MEDLINE | ID: mdl-34283056

ABSTRACT

The dysregulation of chromatin and epigenetics has been defined as the overarching cancer hallmark. By disrupting transcriptional regulation in normal cells and mediating tumor progression by promoting cancer cell plasticity, this process has the ability to mediate all defined hallmarks of cancer. In this review, we collect and assess evidence on the contribution of chromatin and epigenetic dysregulation in prostate cancer. We highlight important mechanisms leading to prostate carcinogenesis, the emergence of castration-resistance upon treatment with androgen deprivation therapy, and resistance to antiandrogens. We examine in particular the contribution of chromatin structure and epigenetics to cell lineage commitment, which is dysregulated during tumorigenesis, and cell plasticity, which is altered during tumor progression.

2.
JCI Insight ; 6(9)2021 05 10.
Article in English | MEDLINE | ID: mdl-33986188

ABSTRACT

Glioma stem cells (GSCs) drive propagation and therapeutic resistance of glioblastomas, the most aggressive diffuse brain tumors. However, the molecular mechanisms that maintain the stemness and promote therapy resistance remain poorly understood. Here we report CD109/STAT3 axis as crucial for the maintenance of stemness and tumorigenicity of GSCs and as a mediator of chemoresistance. Mechanistically, CD109 physically interacts with glycoprotein 130 to promote activation of the IL-6/STAT3 pathway in GSCs. Genetic depletion of CD109 abolished the stemness and self-renewal of GSCs and impaired tumorigenicity. Loss of stemness was accompanied with a phenotypic shift of GSCs to more differentiated astrocytic-like cells. Importantly, genetic or pharmacologic targeting of CD109/STAT3 axis sensitized the GSCs to chemotherapy, suggesting that targeting CD109/STAT3 axis has potential to overcome therapy resistance in glioblastoma.


Subject(s)
Antigens, CD/genetics , Astrocytoma/genetics , Brain Neoplasms/genetics , Cytokine Receptor gp130/genetics , Drug Resistance, Neoplasm/genetics , Glioblastoma/genetics , Neoplasm Proteins/genetics , Neoplastic Stem Cells/metabolism , STAT3 Transcription Factor/metabolism , Animals , Antigens, CD/metabolism , Antineoplastic Agents, Alkylating/therapeutic use , Astrocytoma/metabolism , Astrocytoma/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Plasticity/genetics , Cytokine Receptor gp130/metabolism , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Interleukin-6/metabolism , Male , Mice, Nude , Middle Aged , Neoplasm Grading , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Real-Time Polymerase Chain Reaction , Signal Transduction , Temozolomide/therapeutic use
3.
Gastroenterology ; 161(2): 592-607, 2021 08.
Article in English | MEDLINE | ID: mdl-33930428

ABSTRACT

BACKGROUND & AIMS: Inflammatory bowel disease (IBD) is a chronic, relapsing inflammatory disorder associated with an elevated risk of colorectal cancer (CRC). IBD-associated CRC (IBD-CRC) may represent a distinct pathway of tumorigenesis compared to sporadic CRC (sCRC). Our aim was to comprehensively characterize IBD-associated tumorigenesis integrating multiple high-throughput approaches, and to compare the results with in-house data sets from sCRCs. METHODS: Whole-genome sequencing, single nucleotide polymorphism arrays, RNA sequencing, genome-wide methylation analysis, and immunohistochemistry were performed using fresh-frozen and formalin-fixed tissue samples of tumor and corresponding normal tissues from 31 patients with IBD-CRC. RESULTS: Transcriptome-based tumor subtyping revealed the complete absence of canonical epithelial tumor subtype associated with WNT signaling in IBD-CRCs, dominated instead by mesenchymal stroma-rich subtype. Negative WNT regulators AXIN2 and RNF43 were strongly down-regulated in IBD-CRCs and chromosomal gains at HNF4A, a negative regulator of WNT-induced epithelial-mesenchymal transition (EMT), were less frequent compared to sCRCs. Enrichment of hypomethylation at HNF4α binding sites was detected solely in sCRC genomes. PIGR and OSMR involved in mucosal immunity were dysregulated via epigenetic modifications in IBD-CRCs. Genome-wide analysis showed significant enrichment of noncoding mutations to 5'untranslated region of TP53 in IBD-CRCs. As reported previously, somatic mutations in APC and KRAS were less frequent in IBD-CRCs compared to sCRCs. CONCLUSIONS: Distinct mechanisms of WNT pathway dysregulation skew IBD-CRCs toward mesenchymal tumor subtype, which may affect prognosis and treatment options. Increased OSMR signaling may favor the establishment of mesenchymal tumors in patients with IBD.


Subject(s)
Biomarkers, Tumor/genetics , Cell Transformation, Neoplastic/genetics , Colitis-Associated Neoplasms/genetics , DNA Methylation , Epigenesis, Genetic , Inflammatory Bowel Diseases/genetics , Transcriptome , Adult , Aged , Aged, 80 and over , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/pathology , Colitis-Associated Neoplasms/immunology , Colitis-Associated Neoplasms/pathology , DNA Mutational Analysis , Epigenomics , Female , Finland , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Male , Middle Aged , Mutation , Neoplasm Grading , Neoplasm Staging , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide , Sequence Analysis, RNA , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Whole Genome Sequencing
5.
Cancer Cell ; 38(3): 380-399.e13, 2020 09 14.
Article in English | MEDLINE | ID: mdl-32649887

ABSTRACT

Understanding factors that shape the immune landscape across hematological malignancies is essential for immunotherapy development. We integrated over 8,000 transcriptomes and 2,000 samples with multilevel genomics of hematological cancers to investigate how immunological features are linked to cancer subtypes, genetic and epigenetic alterations, and patient survival, and validated key findings experimentally. Infiltration of cytotoxic lymphocytes was associated with TP53 and myelodysplasia-related changes in acute myeloid leukemia, and activated B cell-like phenotype and interferon-γ response in lymphoma. CIITA methylation regulating antigen presentation, cancer type-specific immune checkpoints, such as VISTA in myeloid malignancies, and variation in cancer antigen expression further contributed to immune heterogeneity and predicted survival. Our study provides a resource linking immunology with cancer subtypes and genomics in hematological malignancies.


Subject(s)
Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Leukemia, Myeloid/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Acute Disease , Epigenesis, Genetic , Genomics/methods , HLA Antigens/genetics , Humans , Immunotherapy/methods , Leukemia, Myeloid/immunology , Leukemia, Myeloid/therapy , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphoma, Large B-Cell, Diffuse/immunology , Lymphoma, Large B-Cell, Diffuse/therapy , Multiple Myeloma/genetics , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Mutation , Tumor Suppressor Protein p53/genetics
6.
Cells ; 9(3)2020 03 10.
Article in English | MEDLINE | ID: mdl-32164385

ABSTRACT

Cancer stem cells (CSCs), a small subpopulation of cells existing in the tumor microenvironment promoting cell proliferation and growth. Targeting the stemness of the CSC population would offer a vital therapeutic opportunity. 3,4-Dihydroquinolin-1(2H)-yl)(p-tolyl)methyl)phenol (THTMP), a small synthetic phenol compound, is proposed to play a significant role in controlling the CSC proliferation and survival. We assessed the potential therapeutic effects of THTMP on glioblastoma multiforme (GBM) and its underlying mechanism in various signaling pathways. To fully comprehend the effect of THTMP on the CSCs, CD133+ GBM stem cell (GSC) and CD133- GBM Non-stem cancer cells (NSCC) population from LN229 and SNB19 cell lines was used. Cell cycle arrest, apoptosis assay and transcriptome analysis were performed for individual cell population. THTMP strongly inhibited NSCC and in a subtle way for GSC in a time-dependent manner and inhibit the resistance variants better than that of temozolomide (TMZ). THTMP arrest the CSC cell population at both G1/S and G2/M phase and induce ROS-mediated apoptosis. Gene expression profiling characterize THTMP as an inhibitor of the p53 signaling pathway causing DNA damage and cell cycle arrest in CSC population. We show that the THTMP majorly affects the EGFR and CSC signaling pathways. Specifically, modulation of key genes involved in Wnt, Notch and Hedgehog, revealed the significant role of THTMP in disrupting the CSCs' stemness and functions. Moreover, THTMP inhibited cell growth, proliferation and metastasis of multiple mesenchymal patient-tissue derived GBM-cell lines. THTMP arrests GBM stem cell cycle through the modulation of EGFR and CSC signaling pathways.


Subject(s)
Cell Cycle Checkpoints/genetics , Glioblastoma/genetics , Neoplastic Stem Cells/metabolism , ErbB Receptors/metabolism , Humans , Neoplastic Stem Cells/pathology , Signal Transduction
7.
Histol Histopathol ; 35(7): 673-680, 2020 Jul.
Article in English | MEDLINE | ID: mdl-31660579

ABSTRACT

Deregulation of fibroblast growth factor receptor (FGFR) signaling is tightly associated with numerous human malignancies, including cancer. Indeed, FGFR inhibitors are being tested as anti-tumor drugs in clinical trials. Among gliomas, FGFR3 fusions occur in IDH wild-type diffuse gliomas leading to high FGFR3 protein expression and both, FGFR3 and FGFR1, show elevated expression in aggressive ependymomas. The aim of this study was to uncover the expression of FGFR1 and FGFR3 proteins in choroid plexus tumors and to further characterize FGFR-related as well as other genetic alterations in FGFR3 expressing tumors. Expression levels of FGFR1 and FGFR3 were detected in 15 choroid plexus tumor tissues using immunohistochemistry of tissue microarrays and 6 samples were subjected to whole mount FGFR3 staining. Targeted sequencing was used for deeper molecular analysis of two FGFR3 positive cases. Moderate expression of FGFR1 or FGFR3 was evidenced in one third of the studied choroid plexus tumors. Targeted sequencing of a choroid plexus carcinoma and an atypical choroid plexus papilloma, both with moderate-to-strong FGFR3 expression, revealed lack of protein-altering mutations or fusions in FGFR1 or FGFR3, but TP53 was altered in both tumors. FGFR3 and FGFR1 proteins are expressed in a subpopulation of choroid plexus tumors. Further studies using larger cohorts of patients will allow identification of the clinicopathological implications of FGFR1 and FGFR3 expression in choroid plexus tumors.


Subject(s)
Biomarkers, Tumor/metabolism , Choroid Plexus Neoplasms/metabolism , Receptor, Fibroblast Growth Factor, Type 3/biosynthesis , Tumor Suppressor Protein p53/biosynthesis , Choroid Plexus Neoplasms/pathology , Female , Humans , Male
8.
Nucleic Acids Res ; 47(13): e76, 2019 07 26.
Article in English | MEDLINE | ID: mdl-31329928

ABSTRACT

Existing large gene expression data repositories hold enormous potential to elucidate disease mechanisms, characterize changes in cellular pathways, and to stratify patients based on molecular profiles. To achieve this goal, integrative resources and tools are needed that allow comparison of results across datasets and data types. We propose an intuitive approach for data-driven stratifications of molecular profiles and benchmark our methodology using the dimensionality reduction algorithm t-distributed stochastic neighbor embedding (t-SNE) with multi-study and multi-platform data on hematological malignancies. Our approach enables assessing the contribution of biological versus technical variation to sample clustering, direct incorporation of additional datasets to the same low dimensional representation, comparison of molecular disease subtypes identified from separate t-SNE representations, and characterization of the obtained clusters based on pathway databases and additional data. In this manner, we performed an integrative analysis across multi-omics acute myeloid leukemia studies. Our approach indicated new molecular subtypes with differential survival and drug responsiveness among samples lacking fusion genes, including a novel myelodysplastic syndrome-like cluster and a cluster characterized with CEBPA mutations and differential activity of the S-adenosylmethionine-dependent DNA methylation pathway. In summary, integration across multiple studies can help to identify novel molecular disease subtypes and generate insight into disease biology.


Subject(s)
Cluster Analysis , Computational Biology/methods , Data Mining/methods , Datasets as Topic , Gene Expression Profiling/methods , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/genetics , Phenotype , Algorithms , Databases, Genetic , Genes, Neoplasm , Humans , Leukemia, Myeloid, Acute/classification , Mutation , Sample Size
9.
Cancer Res ; 79(10): 2466-2479, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30940663

ABSTRACT

Large collections of genome-wide data can facilitate the characterization of disease states and subtypes, permitting pan-cancer analysis of molecular phenotypes and evaluation of disease context for new therapeutic approaches. We analyzed 9,544 transcriptomes from more than 30 hematologic malignancies, normal blood cell types, and cell lines, and showed that disease types could be stratified in a data-driven manner. We then identified cluster-specific pathway activity, new biomarkers, and in silico drug target prioritization through interrogation of drug target databases. Using known vulnerabilities and available drug screens, we highlighted the importance of integrating molecular phenotype with drug target expression for in silico prediction of drug responsiveness. Our analysis implicated BCL2 expression level as an important indicator of venetoclax responsiveness and provided a rationale for its targeting in specific leukemia subtypes and multiple myeloma, linked several polycomb group proteins that could be targeted by small molecules (SFMBT1, CBX7, and EZH1) with chronic lymphocytic leukemia, and supported CDK6 as a disease-specific target in acute myeloid leukemia. Through integration with proteomics data, we characterized target protein expression for pre-B leukemia immunotherapy candidates, including DPEP1. These molecular data can be explored using our publicly available interactive resource, Hemap, for expediting therapeutic innovations in hematologic malignancies. SIGNIFICANCE: This study describes a data resource for researching derailed cellular pathways and candidate drug targets across hematologic malignancies.


Subject(s)
Hematologic Neoplasms/genetics , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Hematologic Neoplasms/drug therapy , Humans , Immunotherapy/methods , Internet , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , Lymphoma, B-Cell/drug therapy , Phenotype , Proto-Oncogene Proteins c-bcl-2/genetics , Small Molecule Libraries/therapeutic use , Sulfonamides/therapeutic use , Transcriptome/genetics
10.
Oncotarget ; 10(9): 920-921, 2019 Jan 29.
Article in English | MEDLINE | ID: mdl-30847018
11.
Cancer Res ; 78(19): 5574-5585, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29921698

ABSTRACT

The immunosuppressive microenvironment in glioblastoma (GBM) prevents an efficient antitumoral immune response and enables tumor formation and growth. Although an understanding of the nature of immunosuppression is still largely lacking, it is important for successful cancer treatment through immune system modulation. To gain insight into immunosuppression in GBM, we performed a computational analysis to model relative immune cell content and type of immune response in each GBM tumor sample from The Cancer Genome Atlas RNA-seq data set. We uncovered high variability in immune system-related responses and in the composition of the microenvironment across the cohort, suggesting immunologic diversity. Immune cell compositions were associated with typical alterations such as IDH mutation or inactivating NF1 mutation/deletion. Furthermore, our analysis identified three GBM subgroups presenting different adaptive immune responses: negative, humoral, and cellular-like. These subgroups were linked to transcriptional GBM subtypes and typical genetic alterations. All G-CIMP and IDH-mutated samples were in the negative group, which was also enriched by cases with focal amplification of CDK4 and MARCH9. IDH1-mutated samples showed lower expression and higher DNA methylation of MHC-I-type HLA genes. Overall, our analysis reveals heterogeneity in the immune microenvironment of GBM and identifies new markers for immunosuppression. Characterization of diverse immune responses will facilitate patient stratification and improve personalized immunotherapy in the future.Significance: This study utilizes a computational approach to characterize the immune environments in glioblastoma and shows that glioblastoma immune microenvironments can be classified into three major subgroups, which are linked to typical glioblastoma alterations such as IDH mutation, NF1 inactivation, and CDK4-MARCH9 locus amplification.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/19/5574/F1.large.jpg Cancer Res; 78(19); 5574-85. ©2018 AACR.


Subject(s)
Brain Neoplasms/immunology , Brain Neoplasms/therapy , Glioblastoma/immunology , Glioblastoma/therapy , Mutation , Tumor Microenvironment , Cell Line, Tumor , Computational Biology , Cyclin-Dependent Kinase 4/genetics , DNA Methylation , Gene Expression Regulation, Neoplastic , Genome, Human , Humans , Immune System , Immunosuppression Therapy , Immunotherapy , Isocitrate Dehydrogenase/genetics , Membrane Proteins/genetics , Multigene Family , Neurofibromin 1/genetics , Regression Analysis , Sequence Analysis, RNA , Ubiquitin-Protein Ligases/genetics
13.
Article in English | MEDLINE | ID: mdl-29602769

ABSTRACT

Brain tumors typically arise sporadically and do not affect several family members simultaneously. In the present study, we describe clinical and genetic data from two patients, a mother and her daughter, with familial brain tumors. Exome sequencing revealed a germline missense mutation in the TP53 and ATRX genes in both cases, and a somatic copy-neutral loss of heterozygosity (LOH) in TP53 in both atypical teratoid/rhabdoid tumor (AT/RT) and astrocytoma tumors. ATRX mutation was associated with the loss of ATRX protein expression. In the astrocytoma case, R132C missense mutation was found in the known hotspot site in isocitrate dehydrogenase 1 (IDH1) and LOH was detected in TP53 The mother carried few other somatic alterations, suggesting that the IDH1 mutation and LOH in TP53 were sufficient to drive tumor development. The genome in the AT/RT tumor was atypically aneuploid: Most chromosomes had experienced copy-neutral LOH or whole-chromosome gains. Only Chromosome 18 had normal diploid status. INI1/hSNF5/SMARCB1 was homozygously deleted in the AT/RT tumor. This report provides further information about tumor development in a predisposed genetic background and describes two special Li-Fraumeni cases with a familial brain tumor.

14.
Histol Histopathol ; 33(8): 791-801, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29441509

ABSTRACT

Nuclear factor erythroid 2-related factor 2 (NRF2), DJ1 and sulfiredoxin 1 (SRXN1) are transcription factors which protect cells from the oxidative damage caused by reactive oxygen species and, on the other hand, are associated with resistance to cancer treatments. The immunohistochemical expression of NRF2, DJ1 and SRNX1 was assessed in human grade II-IV astrocytic gliomas. Their association to clinicopathologic and essential molecular factors was evaluated. The RNA expression levels and genetic alterations were analyzed from publicly available datasets. All studied molecules were commonly expressed. The cytoplasmic NRF2 expression was higher in tumors with a higher malignancy grade, whereas the nuclear and cytoplasmic DJ1 expression was associated with a lower grade. The presence of the isocitrate dehyrdogenase 1 mutation (IDH1) was associated with an increasing cytoplasmic and nuclear expression of NRF2 and a nuclear DJ1 expression. When primary grade IV astrocytomas were compared to secondary glioblastomas, nuclear DJ1 was associated with secondary tumors. In grade II-IV tumors, the cytoplasmic NRF2 expression was associated with a poor prognosis, whereas nuclear NRF2 and both cytoplasmic and nuclear DJ1 were associated with a better patient prognosis. Recurrent homozygous deletions of DJ1 were observed, especially in the IDH wild-type samples. When only the glioblastomas were evaluated, nuclear NRF2 and SRNX1 predicted better survival. As a conclusion, NRF2, DJ1 and SNXR1 can be used as prognosticators in gliomas.


Subject(s)
Astrocytoma/enzymology , Biomarkers, Tumor/analysis , Brain Neoplasms/enzymology , Glioblastoma/enzymology , NF-E2-Related Factor 2/analysis , Oxidoreductases Acting on Sulfur Group Donors/analysis , Protein Deglycase DJ-1/analysis , Adult , Aged , Aged, 80 and over , Astrocytoma/genetics , Astrocytoma/pathology , Astrocytoma/therapy , Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Databases, Genetic , Female , Glioblastoma/genetics , Glioblastoma/pathology , Glioblastoma/therapy , Humans , Immunohistochemistry , Isocitrate Dehydrogenase/genetics , Kaplan-Meier Estimate , Male , Middle Aged , Mutation , NF-E2-Related Factor 2/genetics , Neoplasm Grading , Oxidoreductases Acting on Sulfur Group Donors/genetics , Proportional Hazards Models , Protein Deglycase DJ-1/genetics , Time Factors , Treatment Outcome
15.
BMC Cancer ; 17(1): 310, 2017 05 03.
Article in English | MEDLINE | ID: mdl-28468611

ABSTRACT

BACKGROUND: Fibroblast growth factor receptors (FGFRs) are well-known proto-oncogenes in several human malignancies and are currently therapeutically targeted in clinical trials. Among glioma subtypes, activating FGFR1 alterations have been observed in a subpopulation of pilocytic astrocytomas while FGFR3 fusions occur in IDH wild-type diffuse gliomas, resulting in high FGFR3 protein expression. The purpose of this study was to associate FGFR1 and FGFR3 protein levels with clinical features and genetic alterations in ependymoma and pilocytic astrocytoma. METHODS: FGFR1 and FGFR3 expression levels were detected in ependymoma and pilocytic astrocytoma tissues using immunohistochemistry. Selected cases were further analyzed using targeted sequencing. RESULTS: Expression of both FGFR1 and FGFR3 varied within all tumor types. In ependymomas, increased FGFR3 or FGFR1 expression was associated with high tumor grade, cerebral location, young patient age, and poor prognosis. Moderate-to-strong expression of FGFR1 and/or FGFR3 was observed in 76% of cerebral ependymomas. Cases with moderate-to-strong expression of both proteins had poor clinical prognosis. In pilocytic astrocytomas, moderate-to-strong FGFR3 expression was detected predominantly in non-pediatric patients. Targeted sequencing of 12 tumors found no protein-altering mutations or fusions in FGFR1 or FGFR3. CONCLUSIONS: Elevated FGFR3 and FGFR1 protein expression is common in aggressive ependymomas but likely not driven by genetic alterations. Further studies are warranted to evaluate whether ependymoma patients with high FGFR3 and/or FGFR1 expression could benefit from treatment with FGFR inhibitor based therapeutic approaches currently under evaluation in clinical trials.


Subject(s)
Astrocytoma/genetics , Ependymoma/genetics , Glioma/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 3/genetics , Adolescent , Age Factors , Aged , Astrocytoma/epidemiology , Astrocytoma/pathology , Child , Child, Preschool , Ependymoma/epidemiology , Ependymoma/pathology , Female , Gene Expression Regulation, Neoplastic , Genetic Association Studies , Genetic Predisposition to Disease , Glioma/pathology , Humans , Infant , Male , Middle Aged , Mutation , Neoplasm Grading , Prognosis , Signal Transduction , Young Adult
16.
Neuro Oncol ; 19(9): 1206-1216, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28379477

ABSTRACT

BACKGROUND: Inhibitors of fibroblast growth factor receptors (FGFRs) have recently arisen as a promising treatment option for patients with FGFR alterations. Gene fusions involving FGFR3 and transforming acidic coiled-coil protein 3 (TACC3) have been detected in diffuse gliomas and other malignancies, and fusion-positive cases have responded well to FGFR inhibition. As high FGFR3 expression has been detected in fusion-positive tumors, we sought to determine the clinical significance of FGFR3 protein expression level as well as its potential for indicating FGFR3 fusions. METHODS: We performed FGFR3 immunohistochemistry on tissue microarrays containing 676 grades II-IV astrocytomas and 116 grades II-III oligodendroglial tumor specimens. Fifty-one cases were further analyzed using targeted sequencing. RESULTS: Moderate to strong FGFR3 staining was detected in gliomas of all grades, was more common in females, and was associated with poor survival in diffuse astrocytomas. Targeted sequencing identified FGFR3-TACC3 fusions and an FGFR3-CAMK2A fusion in 10 of 15 strongly stained cases, whereas no fusions were found in 36 negatively to moderately stained cases. Fusion-positive cases were predominantly female and negative for IDH and EGFR/PDGFRA/MET alterations. These and moderately stained cases show lower MIB-1 proliferation index than negatively to weakly stained cases. Furthermore, stronger FGFR3 expression was commonly observed in malignant tissue regions of lower cellularity in fusion-negative cases. Importantly, subregional negative FGFR3 staining was also observed in a few fusion-positive cases. CONCLUSIONS: Strong FGFR3 protein expression is indicative of FGFR3 fusions and may serve as a clinically applicable predictive marker for treatment regimens based on FGFR inhibitors.


Subject(s)
Biomarkers, Tumor/analysis , Brain Neoplasms/genetics , Glioma/genetics , Receptor, Fibroblast Growth Factor, Type 3/biosynthesis , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Female , Humans , Male , Microtubule-Associated Proteins/genetics , Middle Aged , Oncogene Fusion , Receptor, Fibroblast Growth Factor, Type 3/analysis , Receptor, Fibroblast Growth Factor, Type 3/genetics , Young Adult
17.
J Clin Pathol ; 68(11): 905-12, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26163539

ABSTRACT

AIMS AND METHODS: Epithelial-mesenchymal transition (EMT) has previously been linked to glioma invasion and progression. To determine whether EMT regulators, Twist and Zeb1, had clinical significance in astrocytic gliomas, the association of Twist and Zeb1 with clinicopathological and molecular factors was studied in 269 astrocytoma samples. RESULTS: Twist and Zeb1 were widely expressed in astrocytic gliomas, but the expression of the former did not correlate with that of the latter. Stronger Twist expression levels were associated with higher WHO grades (p=0.001), whereas Zeb1 did not correlate with WHO grades. We found no association between Twist and proliferation activity (Ki67/MIB-1), p53 status, epidermal growth factor receptor (EGFR) amplification or neural cell adhesion molecule (NCAM) expression. There was no significant difference in Twist or Zeb1 expression when primary and secondary gliomas were analysed. Tumours with high Twist expression were IDH1 negative (p=0.009). High hypoxia-inducible factor-1α expression correlated significantly with positive Twist expression (p<0.001), whereas it was not associated with Zeb1 expression. Zeb1 expression did not correlate with proliferation, EGFR or IDH1. Nevertheless, we did find a correlation between high Zeb1 expression and low p53 expression levels (p=0.027). Positive NCAM expression was significantly associated with Zeb1 positivity (p=0.022). Zeb1 had no association with patient survival, whereas positive Twist expression predicted poor survival for patients in both univariate (p<0.001) and multivariable analyses (p=0.027). CONCLUSIONS: EMT regulators, Twist and Zeb1, are common features of infiltrating astrocytomas, and Twist is upregulated in glioblastomas in particular. Twist may be a novel marker for poor prognosis in glioma patients.


Subject(s)
Astrocytoma/pathology , Biomarkers, Tumor/analysis , Brain Neoplasms/pathology , Epithelial-Mesenchymal Transition/physiology , Nuclear Proteins/biosynthesis , Twist-Related Protein 1/biosynthesis , Adolescent , Adult , Aged , Aged, 80 and over , Astrocytoma/metabolism , Astrocytoma/mortality , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Female , Homeodomain Proteins/analysis , Homeodomain Proteins/biosynthesis , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Nuclear Proteins/analysis , Transcription Factors/analysis , Transcription Factors/biosynthesis , Twist-Related Protein 1/analysis , Up-Regulation , Young Adult , Zinc Finger E-box-Binding Homeobox 1
18.
Proc Natl Acad Sci U S A ; 112(11): 3421-6, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25737557

ABSTRACT

Akt is a robust oncogene that plays key roles in the development and progression of many cancers, including glioma. We evaluated the differential propensities of the Akt isoforms toward progression in the well-characterized RCAS/Ntv-a mouse model of PDGFB-driven low grade glioma. A constitutively active myristoylated form of Akt1 did not induce high-grade glioma (HGG). In stark contrast, Akt2 and Akt3 showed strong progression potential with 78% and 97% of tumors diagnosed as HGG, respectively. We further revealed that significant variations in polarity and hydropathy values among the Akt isoforms in both the pleckstrin homology domain (P domain) and regulatory domain (R domain) were critical in mediating glioma progression. Gene expression profiles from representative Akt-derived tumors indicated dominant and distinct roles for Akt3, consisting primarily of DNA repair pathways. TCGA data from human GBM closely reflected the DNA repair function, as Akt3 was significantly correlated with a 76-gene signature DNA repair panel. Consistently, compared with Akt1 and Akt2 overexpression models, Akt3-expressing human GBM cells had enhanced activation of DNA repair proteins, leading to increased DNA repair and subsequent resistance to radiation and temozolomide. Given the wide range of Akt3-amplified cancers, Akt3 may represent a key resistance factor.


Subject(s)
Brain Neoplasms/genetics , DNA Repair/genetics , Disease Progression , Gene Amplification , Genome, Human , Glioma/genetics , Proto-Oncogene Proteins c-akt/genetics , Animals , Brain Neoplasms/enzymology , Brain Neoplasms/pathology , Cell Line, Tumor , DNA Damage/genetics , DNA Repair/drug effects , DNA Repair/radiation effects , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/radiation effects , Gene Amplification/drug effects , Gene Amplification/radiation effects , Gene Expression Regulation, Neoplastic , Glioma/enzymology , Glioma/pathology , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Protein Structure, Tertiary , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Radiation Tolerance/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/radiation effects , Temozolomide , Transcription, Genetic
19.
Oncotarget ; 6(8): 6235-50, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25749039

ABSTRACT

Prostate cancer is the third most common cause of male cancer death in developed countries, and one of the most comprehensively characterized human cancers. Roughly 60% of prostate cancers harbor gene fusions that juxtapose ETS-family transcription factors with androgen regulated promoters. A second subtype, characterized by SPINK1 overexpression, accounts for 15% of prostate cancers. Here we report the discovery of a new prostate cancer subtype characterized by rearrangements juxtaposing the SMAD inhibitor SKIL with androgen regulated promoters, leading to increased SKIL expression. SKIL fusions were found in 6 of 540 (1.1%) prostate cancers and 1 of 27 (3.7%) cell lines and xenografts. 6 of 7 SKIL-positive cancers were negative for ETS overexpression, suggesting mutual exclusivity with ETS fusions. SKIL knockdown led to growth arrest in PC-3 and LNCaP cell line models of prostate cancer, and its overexpression led to increased invasiveness in RWPE-1 cells. The role of SKIL as a prostate cancer oncogene lends support to recent studies on the role of TGF-ß signaling as a rate-limiting step in prostate cancer progression. Our findings highlight SKIL as an oncogene and potential therapeutic target in 1-2% of prostate cancers, amounting to an estimated 10,000 cancer diagnoses per year worldwide.


Subject(s)
Intracellular Signaling Peptides and Proteins/genetics , Prostatic Neoplasms/classification , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins/genetics , Animals , Cell Line, Tumor , Cohort Studies , Gene Knockdown Techniques , Gene Rearrangement , Heterografts , Humans , Intracellular Signaling Peptides and Proteins/biosynthesis , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Middle Aged , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ets/metabolism , Transcriptome , Transfection
20.
Oncogene ; 34(13): 1619-1628, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-24747968

ABSTRACT

MIR-491 is commonly co-deleted with its adjacent CDKN2A on chromosome 9p21.3 in glioblastoma multiforme (GBM). However, it is not known whether deletion of MIR-491 is only a passenger event or has an important role. Small-RNA sequencing of samples from GBM patients demonstrated that both mature products of MIR-491 (miR-491-5p and -3p) are downregulated in tumors compared with the normal brain. The integration of GBM data from The Cancer Genome Atlas (TCGA), miRNA target prediction and reporter assays showed that miR-491-5p directly targets EGFR, CDK6 and Bcl-xL, whereas miR-491-3p targets IGFBP2 and CDK6. Functionally, miR-491-3p inhibited glioma cell invasion; overexpression of both miR-491-5p and -3p inhibited proliferation of glioma cell lines and impaired the propagation of glioma stem cells (GSCs), thereby prolonging survival of xenograft mice. Moreover, knockdown of miR-491-5p in primary Ink4a-Arf-null mouse glial progenitor cells exacerbated cell proliferation and invasion. Therefore, MIR-491 is a tumor suppressor gene that, by utilizing both mature forms, coordinately controls the key cancer hallmarks: proliferation, invasion and stem cell propagation.


Subject(s)
Brain Neoplasms/pathology , Genes, Tumor Suppressor , Glioblastoma/pathology , MicroRNAs/physiology , Animals , Cell Line, Tumor , Cyclin-Dependent Kinase 6/analysis , ErbB Receptors/analysis , Gene Expression Regulation, Neoplastic , Humans , Insulin-Like Growth Factor Binding Protein 2/analysis , Male , Mice , MicroRNAs/analysis , MicroRNAs/genetics , Neoplasm Invasiveness , bcl-X Protein/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...