17 results on '"Mesirov JP"'
Search Results
2. Circular extrachromosomal DNA promotes tumor heterogeneity in high-risk medulloblastoma.
- Author
-
Chapman OS, Luebeck J, Sridhar S, Wong IT, Dixit D, Wang S, Prasad G, Rajkumar U, Pagadala MS, Larson JD, He BJ, Hung KL, Lange JT, Dehkordi SR, Chandran S, Adam M, Morgan L, Wani S, Tiwari A, Guccione C, Lin Y, Dutta A, Lo YY, Juarez E, Robinson JT, Korshunov A, Michaels JA, Cho YJ, Malicki DM, Coufal NG, Levy ML, Hobbs C, Scheuermann RH, Crawford JR, Pomeroy SL, Rich JN, Zhang X, Chang HY, Dixon JR, Bagchi A, Deshpande AJ, Carter H, Fraenkel E, Mischel PS, Wechsler-Reya RJ, Bafna V, Mesirov JP, and Chavez L
- Subjects
- Humans, DNA, Circular, Retrospective Studies, Oncogenes, Medulloblastoma genetics, Neoplasms genetics, Cerebellar Neoplasms genetics
- Abstract
Circular extrachromosomal DNA (ecDNA) in patient tumors is an important driver of oncogenic gene expression, evolution of drug resistance and poor patient outcomes. Applying computational methods for the detection and reconstruction of ecDNA across a retrospective cohort of 481 medulloblastoma tumors from 465 patients, we identify circular ecDNA in 82 patients (18%). Patients with ecDNA-positive medulloblastoma were more than twice as likely to relapse and three times as likely to die within 5 years of diagnosis. A subset of tumors harbored multiple ecDNA lineages, each containing distinct amplified oncogenes. Multimodal sequencing, imaging and CRISPR inhibition experiments in medulloblastoma models reveal intratumoral heterogeneity of ecDNA copy number per cell and frequent putative 'enhancer rewiring' events on ecDNA. This study reveals the frequency and diversity of ecDNA in medulloblastoma, stratified into molecular subgroups, and suggests copy number heterogeneity and enhancer rewiring as oncogenic features of ecDNA., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
3. The GPCR-Gα s -PKA signaling axis promotes T cell dysfunction and cancer immunotherapy failure.
- Author
-
Wu VH, Yung BS, Faraji F, Saddawi-Konefka R, Wang Z, Wenzel AT, Song MJ, Pagadala MS, Clubb LM, Chiou J, Sinha S, Matic M, Raimondi F, Hoang TS, Berdeaux R, Vignali DAA, Iglesias-Bartolome R, Carter H, Ruppin E, Mesirov JP, and Gutkind JS
- Subjects
- Mice, Animals, Signal Transduction, Mice, Transgenic, Immunotherapy, Tumor Microenvironment, CD8-Positive T-Lymphocytes, Neoplasms
- Abstract
Immune checkpoint blockade (ICB) targeting PD-1 and CTLA-4 has revolutionized cancer treatment. However, many cancers do not respond to ICB, prompting the search for additional strategies to achieve durable responses. G-protein-coupled receptors (GPCRs) are the most intensively studied drug targets but are underexplored in immuno-oncology. Here, we cross-integrated large singe-cell RNA-sequencing datasets from CD8
+ T cells covering 19 distinct cancer types and identified an enrichment of Gαs -coupled GPCRs on exhausted CD8+ T cells. These include EP2 , EP4 , A2A R, β1 AR and β2 AR, all of which promote T cell dysfunction. We also developed transgenic mice expressing a chemogenetic CD8-restricted Gαs -DREADD to activate CD8-restricted Gαs signaling and show that a Gαs -PKA signaling axis promotes CD8+ T cell dysfunction and immunotherapy failure. These data indicate that Gαs -GPCRs are druggable immune checkpoints that might be targeted to enhance the response to ICB immunotherapies., (© 2023. The Author(s), under exclusive licence to Springer Nature America, Inc.)- Published
- 2023
- Full Text
- View/download PDF
4. Germline modifiers of the tumor immune microenvironment implicate drivers of cancer risk and immunotherapy response.
- Author
-
Pagadala M, Sears TJ, Wu VH, Pérez-Guijarro E, Kim H, Castro A, Talwar JV, Gonzalez-Colin C, Cao S, Schmiedel BJ, Goudarzi S, Kirani D, Au J, Zhang T, Landi T, Salem RM, Morris GP, Harismendy O, Patel SP, Alexandrov LB, Mesirov JP, Zanetti M, Day CP, Fan CC, Thompson WK, Merlino G, Gutkind JS, Vijayanand P, and Carter H
- Subjects
- Germ Cells, Germ-Line Mutation, Inhibition, Psychological, Macrophages, Tumor Microenvironment genetics, Immunotherapy, Neoplasms genetics, Neoplasms therapy
- Abstract
With the continued promise of immunotherapy for treating cancer, understanding how host genetics contributes to the tumor immune microenvironment (TIME) is essential to tailoring cancer screening and treatment strategies. Here, we study 1084 eQTLs affecting the TIME found through analysis of The Cancer Genome Atlas and literature curation. These TIME eQTLs are enriched in areas of active transcription, and associate with gene expression in specific immune cell subsets, such as macrophages and dendritic cells. Polygenic score models built with TIME eQTLs reproducibly stratify cancer risk, survival and immune checkpoint blockade (ICB) response across independent cohorts. To assess whether an eQTL-informed approach could reveal potential cancer immunotherapy targets, we inhibit CTSS, a gene implicated by cancer risk and ICB response-associated polygenic models; CTSS inhibition results in slowed tumor growth and extended survival in vivo. These results validate the potential of integrating germline variation and TIME characteristics for uncovering potential targets for immunotherapy., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
5. Variant Review with the Integrative Genomics Viewer.
- Author
-
Robinson JT, Thorvaldsdóttir H, Wenger AM, Zehir A, and Mesirov JP
- Subjects
- High-Throughput Nucleotide Sequencing, Humans, Polymorphism, Single Nucleotide, Sequence Alignment, Sequence Analysis, DNA, Computational Biology methods, Genomics methods, Neoplasms genetics, Software
- Abstract
Manual review of aligned reads for confirmation and interpretation of variant calls is an important step in many variant calling pipelines for next-generation sequencing (NGS) data. Visual inspection can greatly increase the confidence in calls, reduce the risk of false positives, and help characterize complex events. The Integrative Genomics Viewer (IGV) was one of the first tools to provide NGS data visualization, and it currently provides a rich set of tools for inspection, validation, and interpretation of NGS datasets, as well as other types of genomic data. Here, we present a short overview of IGV's variant review features for both single-nucleotide variants and structural variants, with examples from both cancer and germline datasets. IGV is freely available at https://www.igv.org Cancer Res; 77(21); e31-34. ©2017 AACR ., (©2017 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
6. Decomposing Oncogenic Transcriptional Signatures to Generate Maps of Divergent Cellular States.
- Author
-
Kim JW, Abudayyeh OO, Yeerna H, Yeang CH, Stewart M, Jenkins RW, Kitajima S, Konieczkowski DJ, Medetgul-Ernar K, Cavazos T, Mah C, Ting S, Van Allen EM, Cohen O, Mcdermott J, Damato E, Aguirre AJ, Liang J, Liberzon A, Alexe G, Doench J, Ghandi M, Vazquez F, Weir BA, Tsherniak A, Subramanian A, Meneses-Cime K, Park J, Clemons P, Garraway LA, Thomas D, Boehm JS, Barbie DA, Hahn WC, Mesirov JP, and Tamayo P
- Subjects
- Biomarkers, Tumor metabolism, Cell Line, Tumor, Gene Expression Profiling methods, Genes, ras genetics, Genome, Humans, MAP Kinase Signaling System, Neoplasms pathology, Precision Medicine, Gene Expression Regulation, Neoplastic, Neoplasms genetics
- Abstract
The systematic sequencing of the cancer genome has led to the identification of numerous genetic alterations in cancer. However, a deeper understanding of the functional consequences of these alterations is necessary to guide appropriate therapeutic strategies. Here, we describe Onco-GPS (OncoGenic Positioning System), a data-driven analysis framework to organize individual tumor samples with shared oncogenic alterations onto a reference map defined by their underlying cellular states. We applied the methodology to the RAS pathway and identified nine distinct components that reflect transcriptional activities downstream of RAS and defined several functional states associated with patterns of transcriptional component activation that associates with genomic hallmarks and response to genetic and pharmacological perturbations. These results show that the Onco-GPS is an effective approach to explore the complex landscape of oncogenic cellular states across cancers, and an analytic framework to summarize knowledge, establish relationships, and generate more effective disease models for research or as part of individualized precision medicine paradigms., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
7. Dependency of a therapy-resistant state of cancer cells on a lipid peroxidase pathway.
- Author
-
Viswanathan VS, Ryan MJ, Dhruv HD, Gill S, Eichhoff OM, Seashore-Ludlow B, Kaffenberger SD, Eaton JK, Shimada K, Aguirre AJ, Viswanathan SR, Chattopadhyay S, Tamayo P, Yang WS, Rees MG, Chen S, Boskovic ZV, Javaid S, Huang C, Wu X, Tseng YY, Roider EM, Gao D, Cleary JM, Wolpin BM, Mesirov JP, Haber DA, Engelman JA, Boehm JS, Kotz JD, Hon CS, Chen Y, Hahn WC, Levesque MP, Doench JG, Berens ME, Shamji AF, Clemons PA, Stockwell BR, and Schreiber SL
- Subjects
- Cadherins metabolism, Cell Death, Cell Line, Tumor, Cell Lineage, Cell Transdifferentiation, Drug Resistance, Neoplasm genetics, Epithelial-Mesenchymal Transition, Humans, Iron metabolism, Lipid Peroxides metabolism, Male, Melanoma drug therapy, Melanoma enzymology, Melanoma metabolism, Melanoma pathology, Mesoderm drug effects, Mesoderm enzymology, Mesoderm metabolism, Mesoderm pathology, Neoplasms genetics, Neoplasms pathology, Phospholipid Hydroperoxide Glutathione Peroxidase, Prostatic Neoplasms drug therapy, Prostatic Neoplasms enzymology, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology, Proteomics, Proto-Oncogene Proteins B-raf genetics, Reproducibility of Results, Zinc Finger E-box-Binding Homeobox 1 genetics, Glutathione Peroxidase metabolism, Lipid Peroxidation drug effects, Neoplasms drug therapy, Neoplasms enzymology
- Abstract
Plasticity of the cell state has been proposed to drive resistance to multiple classes of cancer therapies, thereby limiting their effectiveness. A high-mesenchymal cell state observed in human tumours and cancer cell lines has been associated with resistance to multiple treatment modalities across diverse cancer lineages, but the mechanistic underpinning for this state has remained incompletely understood. Here we molecularly characterize this therapy-resistant high-mesenchymal cell state in human cancer cell lines and organoids and show that it depends on a druggable lipid-peroxidase pathway that protects against ferroptosis, a non-apoptotic form of cell death induced by the build-up of toxic lipid peroxides. We show that this cell state is characterized by activity of enzymes that promote the synthesis of polyunsaturated lipids. These lipids are the substrates for lipid peroxidation by lipoxygenase enzymes. This lipid metabolism creates a dependency on pathways converging on the phospholipid glutathione peroxidase (GPX4), a selenocysteine-containing enzyme that dissipates lipid peroxides and thereby prevents the iron-mediated reactions of peroxides that induce ferroptotic cell death. Dependency on GPX4 was found to exist across diverse therapy-resistant states characterized by high expression of ZEB1, including epithelial-mesenchymal transition in epithelial-derived carcinomas, TGFβ-mediated therapy-resistance in melanoma, treatment-induced neuroendocrine transdifferentiation in prostate cancer, and sarcomas, which are fixed in a mesenchymal state owing to their cells of origin. We identify vulnerability to ferroptic cell death induced by inhibition of a lipid peroxidase pathway as a feature of therapy-resistant cancer cells across diverse mesenchymal cell-state contexts.
- Published
- 2017
- Full Text
- View/download PDF
8. Leveraging premalignant biology for immune-based cancer prevention.
- Author
-
Spira A, Disis ML, Schiller JT, Vilar E, Rebbeck TR, Bejar R, Ideker T, Arts J, Yurgelun MB, Mesirov JP, Rao A, Garber J, Jaffee EM, and Lippman SM
- Subjects
- Germ Cells metabolism, Humans, Immune System pathology, Models, Biological, Neoplasm Proteins metabolism, Tumor Microenvironment, Neoplasms immunology, Neoplasms prevention & control, Precancerous Conditions pathology
- Abstract
Prevention is an essential component of cancer eradication. Next-generation sequencing of cancer genomes and epigenomes has defined large numbers of driver mutations and molecular subgroups, leading to therapeutic advances. By comparison, there is a relative paucity of such knowledge in premalignant neoplasia, which inherently limits the potential to develop precision prevention strategies. Studies on the interplay between germ-line and somatic events have elucidated genetic processes underlying premalignant progression and preventive targets. Emerging data hint at the immune system's ability to intercept premalignancy and prevent cancer. Genetically engineered mouse models have identified mechanisms by which genetic drivers and other somatic alterations recruit inflammatory cells and induce changes in normal cells to create and interact with the premalignant tumor microenvironment to promote oncogenesis and immune evasion. These studies are currently limited to only a few lesion types and patients. In this Perspective, we advocate a large-scale collaborative effort to systematically map the biology of premalignancy and the surrounding cellular response. By bringing together scientists from diverse disciplines (e.g., biochemistry, omics, and computational biology; microbiology, immunology, and medical genetics; engineering, imaging, and synthetic chemistry; and implementation science), we can drive a concerted effort focused on cancer vaccines to reprogram the immune response to prevent, detect, and reject premalignancy. Lynch syndrome, clonal hematopoiesis, and cervical intraepithelial neoplasia which also serve as models for inherited syndromes, blood, and viral premalignancies, are ideal scenarios in which to launch this initiative., Competing Interests: The authors declare no conflict of interest.
- Published
- 2016
- Full Text
- View/download PDF
9. Characterizing genomic alterations in cancer by complementary functional associations.
- Author
-
Kim JW, Botvinnik OB, Abudayyeh O, Birger C, Rosenbluh J, Shrestha Y, Abazeed ME, Hammerman PS, DiCara D, Konieczkowski DJ, Johannessen CM, Liberzon A, Alizad-Rahvar AR, Alexe G, Aguirre A, Ghandi M, Greulich H, Vazquez F, Weir BA, Van Allen EM, Tsherniak A, Shao DD, Zack TI, Noble M, Getz G, Beroukhim R, Garraway LA, Ardakani M, Romualdi C, Sales G, Barbie DA, Boehm JS, Hahn WC, Mesirov JP, and Tamayo P
- Subjects
- Drug Resistance, Neoplasm genetics, Genes, Neoplasm genetics, Genetic Predisposition to Disease genetics, Genome, Human genetics, Humans, Mutation genetics, Neoplasms diagnosis, Signal Transduction genetics, Biomarkers, Tumor genetics, Chromosome Mapping methods, Genome-Wide Association Study methods, Neoplasm Proteins genetics, Neoplasms genetics, Polymorphism, Single Nucleotide genetics
- Abstract
Systematic efforts to sequence the cancer genome have identified large numbers of mutations and copy number alterations in human cancers. However, elucidating the functional consequences of these variants, and their interactions to drive or maintain oncogenic states, remains a challenge in cancer research. We developed REVEALER, a computational method that identifies combinations of mutually exclusive genomic alterations correlated with functional phenotypes, such as the activation or gene dependency of oncogenic pathways or sensitivity to a drug treatment. We used REVEALER to uncover complementary genomic alterations associated with the transcriptional activation of β-catenin and NRF2, MEK-inhibitor sensitivity, and KRAS dependency. REVEALER successfully identified both known and new associations, demonstrating the power of combining functional profiles with extensive characterization of genomic alterations in cancer genomes.
- Published
- 2016
- Full Text
- View/download PDF
10. Systematic interrogation of 3q26 identifies TLOC1 and SKIL as cancer drivers.
- Author
-
Hagerstrand D, Tong A, Schumacher SE, Ilic N, Shen RR, Cheung HW, Vazquez F, Shrestha Y, Kim SY, Giacomelli AO, Rosenbluh J, Schinzel AC, Spardy NA, Barbie DA, Mermel CH, Weir BA, Garraway LA, Tamayo P, Mesirov JP, Beroukhim R, and Hahn WC
- Subjects
- Breast Neoplasms genetics, Carcinoma, Non-Small-Cell Lung genetics, Cell Line, Tumor, Cell Proliferation, DEAD-box RNA Helicases metabolism, DNA Copy Number Variations genetics, Epithelial-Mesenchymal Transition genetics, Female, Gene Amplification genetics, Gene Expression Regulation, Neoplastic, Humans, Intracellular Signaling Peptides and Proteins metabolism, Lung Neoplasms genetics, Mammary Glands, Human cytology, Membrane Transport Proteins metabolism, Ovarian Neoplasms genetics, Protein Binding, Proto-Oncogene Proteins metabolism, RNA Interference, RNA, Small Interfering, Snail Family Transcription Factors, Transcription Factors biosynthesis, Chromosomes, Human, Pair 3 genetics, Intracellular Signaling Peptides and Proteins genetics, Membrane Transport Proteins genetics, Neoplasm Invasiveness genetics, Neoplasms genetics, Proto-Oncogene Proteins genetics
- Abstract
Unlabelled: 3q26 is frequently amplified in several cancer types with a common amplified region containing 20 genes. To identify cancer driver genes in this region, we interrogated the function of each of these genes by loss- and gain-of-function genetic screens. Specifically, we found that TLOC1 (SEC62) was selectively required for the proliferation of cell lines with 3q26 amplification. Increased TLOC1 expression induced anchorage-independent growth, and a second 3q26 gene, SKIL (SNON), facilitated cell invasion in immortalized human mammary epithelial cells. Expression of both TLOC1 and SKIL induced subcutaneous tumor growth. Proteomic studies showed that TLOC1 binds to DDX3X, which is essential for TLOC1-induced transformation and affected protein translation. SKIL induced invasion through upregulation of SLUG (SNAI2) expression. Together, these studies identify TLOC1 and SKIL as driver genes at 3q26 and more broadly suggest that cooperating genes may be coamplified in other regions with somatic copy number gain., Significance: These studies identify TLOC1 and SKIL as driver genes in 3q26. These observations provide evidence that regions of somatic copy number gain may harbor cooperating genes of different but complementary functions., (©2013 AACR.)
- Published
- 2013
- Full Text
- View/download PDF
11. Cancer vulnerabilities unveiled by genomic loss.
- Author
-
Nijhawan D, Zack TI, Ren Y, Strickland MR, Lamothe R, Schumacher SE, Tsherniak A, Besche HC, Rosenbluh J, Shehata S, Cowley GS, Weir BA, Goldberg AL, Mesirov JP, Root DE, Bhatia SN, Beroukhim R, and Hahn WC
- Subjects
- ATPases Associated with Diverse Cellular Activities, Animals, Cell Line, Tumor, Chromosome Deletion, Gene Dosage, Genes, Tumor Suppressor, Humans, Mice, Mice, Nude, Neoplasm Transplantation, Neoplasms metabolism, Proteasome Endopeptidase Complex genetics, Proteasome Endopeptidase Complex metabolism, Transplantation, Heterologous, Genes, Essential, Genomic Instability, Neoplasms genetics
- Abstract
Due to genome instability, most cancers exhibit loss of regions containing tumor suppressor genes and collateral loss of other genes. To identify cancer-specific vulnerabilities that are the result of copy number losses, we performed integrated analyses of genome-wide copy number and RNAi profiles and identified 56 genes for which gene suppression specifically inhibited the proliferation of cells harboring partial copy number loss of that gene. These CYCLOPS (copy number alterations yielding cancer liabilities owing to partial loss) genes are enriched for spliceosome, proteasome, and ribosome components. One CYCLOPS gene, PSMC2, encodes an essential member of the 19S proteasome. Normal cells express excess PSMC2, which resides in a complex with PSMC1, PSMD2, and PSMD5 and acts as a reservoir protecting cells from PSMC2 suppression. Cells harboring partial PSMC2 copy number loss lack this complex and die after PSMC2 suppression. These observations define a distinct class of cancer-specific liabilities resulting from genome instability., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
12. The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity.
- Author
-
Barretina J, Caponigro G, Stransky N, Venkatesan K, Margolin AA, Kim S, Wilson CJ, Lehár J, Kryukov GV, Sonkin D, Reddy A, Liu M, Murray L, Berger MF, Monahan JE, Morais P, Meltzer J, Korejwa A, Jané-Valbuena J, Mapa FA, Thibault J, Bric-Furlong E, Raman P, Shipway A, Engels IH, Cheng J, Yu GK, Yu J, Aspesi P Jr, de Silva M, Jagtap K, Jones MD, Wang L, Hatton C, Palescandolo E, Gupta S, Mahan S, Sougnez C, Onofrio RC, Liefeld T, MacConaill L, Winckler W, Reich M, Li N, Mesirov JP, Gabriel SB, Getz G, Ardlie K, Chan V, Myer VE, Weber BL, Porter J, Warmuth M, Finan P, Harris JL, Meyerson M, Golub TR, Morrissey MP, Sellers WR, Schlegel R, and Garraway LA
- Subjects
- Antineoplastic Agents pharmacology, Cell Line, Tumor, Cell Lineage, Chromosomes, Human genetics, Clinical Trials as Topic methods, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Genes, ras genetics, Genome, Human genetics, Genomics, Humans, Mitogen-Activated Protein Kinase Kinases antagonists & inhibitors, Mitogen-Activated Protein Kinase Kinases metabolism, Neoplasms genetics, Neoplasms metabolism, Pharmacogenetics, Plasma Cells cytology, Plasma Cells drug effects, Plasma Cells metabolism, Precision Medicine methods, Receptor, IGF Type 1 antagonists & inhibitors, Receptor, IGF Type 1 metabolism, Receptors, Aryl Hydrocarbon genetics, Receptors, Aryl Hydrocarbon metabolism, Sequence Analysis, DNA, Topoisomerase Inhibitors pharmacology, Databases, Factual, Drug Screening Assays, Antitumor methods, Encyclopedias as Topic, Models, Biological, Neoplasms drug therapy, Neoplasms pathology
- Abstract
The systematic translation of cancer genomic data into knowledge of tumour biology and therapeutic possibilities remains challenging. Such efforts should be greatly aided by robust preclinical model systems that reflect the genomic diversity of human cancers and for which detailed genetic and pharmacological annotation is available. Here we describe the Cancer Cell Line Encyclopedia (CCLE): a compilation of gene expression, chromosomal copy number and massively parallel sequencing data from 947 human cancer cell lines. When coupled with pharmacological profiles for 24 anticancer drugs across 479 of the cell lines, this collection allowed identification of genetic, lineage, and gene-expression-based predictors of drug sensitivity. In addition to known predictors, we found that plasma cell lineage correlated with sensitivity to IGF1 receptor inhibitors; AHR expression was associated with MEK inhibitor efficacy in NRAS-mutant lines; and SLFN11 expression predicted sensitivity to topoisomerase inhibitors. Together, our results indicate that large, annotated cell-line collections may help to enable preclinical stratification schemata for anticancer agents. The generation of genetic predictions of drug response in the preclinical setting and their incorporation into cancer clinical trial design could speed the emergence of 'personalized' therapeutic regimens.
- Published
- 2012
- Full Text
- View/download PDF
13. Carcinoma-associated fibroblast-like differentiation of human mesenchymal stem cells.
- Author
-
Mishra PJ, Mishra PJ, Humeniuk R, Medina DJ, Alexe G, Mesirov JP, Ganesan S, Glod JW, and Banerjee D
- Subjects
- Animals, Base Sequence, Cell Division, Cell Line, Tumor, Culture Media, Conditioned, DNA Primers, Fibroblasts cytology, Fluorescent Antibody Technique, Humans, Mice, Mice, Nude, Reverse Transcriptase Polymerase Chain Reaction, Cell Differentiation, Mesenchymal Stem Cells cytology, Neoplasms pathology
- Abstract
Carcinoma-associated fibroblasts (CAF) have recently been implicated in important aspects of epithelial solid tumor biology, such as neoplastic progression, tumor growth, angiogenesis, and metastasis. However, neither the source of CAFs nor the differences between CAFs and fibroblasts from nonneoplastic tissue have been well defined. In this study, we show that human bone marrow-derived mesenchymal stem cells (hMSCs) exposed to tumor-conditioned medium (TCM) over a prolonged period of time assume a CAF-like myofibroblastic phenotype. More importantly, these cells exhibit functional properties of CAFs, including sustained expression of stromal-derived factor-1 (SDF-1) and the ability to promote tumor cell growth both in vitro and in an in vivo coimplantation model, and expression of myofibroblast markers, including alpha-smooth muscle actin and fibroblast surface protein. hMSCs induced to differentiate to a myofibroblast-like phenotype using 5-azacytidine do not promote tumor cell growth as efficiently as hMSCs cultured in TCM nor do they show increased SDF-1 expression. Furthermore, gene expression profiling revealed similarities between TCM-exposed hMSCs and CAFs. Taken together, these data suggest that hMSCs are a source of CAFs and can be used in the modeling of tumor-stroma interactions. To our knowledge, this is the first report showing that hMSCs become activated and resemble carcinoma-associated myofibroblasts on prolonged exposure to conditioned medium from MDAMB231 human breast cancer cells.
- Published
- 2008
- Full Text
- View/download PDF
14. Metagenes and molecular pattern discovery using matrix factorization.
- Author
-
Brunet JP, Tamayo P, Golub TR, and Mesirov JP
- Subjects
- Algorithms, Central Nervous System Neoplasms classification, Central Nervous System Neoplasms genetics, Data Interpretation, Statistical, Leukemia classification, Leukemia genetics, Medulloblastoma genetics, Neoplasms genetics, Computational Biology, Models, Genetic, Neoplasms classification
- Abstract
We describe here the use of nonnegative matrix factorization (NMF), an algorithm based on decomposition by parts that can reduce the dimension of expression data from thousands of genes to a handful of metagenes. Coupled with a model selection mechanism, adapted to work for any stochastic clustering algorithm, NMF is an efficient method for identification of distinct molecular patterns and provides a powerful method for class discovery. We demonstrate the ability of NMF to recover meaningful biological information from cancer-related microarray data. NMF appears to have advantages over other methods such as hierarchical clustering or self-organizing maps. We found it less sensitive to a priori selection of genes or initial conditions and able to detect alternative or context-dependent patterns of gene expression in complex biological systems. This ability, similar to semantic polysemy in text, provides a general method for robust molecular pattern discovery.
- Published
- 2004
- Full Text
- View/download PDF
15. Estimating dataset size requirements for classifying DNA microarray data.
- Author
-
Mukherjee S, Tamayo P, Rogers S, Rifkin R, Engle A, Campbell C, Golub TR, and Mesirov JP
- Subjects
- Algorithms, Computational Biology methods, Computer Simulation, Gene Expression Profiling classification, Humans, Models, Molecular, Neoplasms metabolism, Gene Expression Profiling methods, Neoplasms classification, Neoplasms genetics, Oligonucleotide Array Sequence Analysis
- Abstract
A statistical methodology for estimating dataset size requirements for classifying microarray data using learning curves is introduced. The goal is to use existing classification results to estimate dataset size requirements for future classification experiments and to evaluate the gain in accuracy and significance of classifiers built with additional data. The method is based on fitting inverse power-law models to construct empirical learning curves. It also includes a permutation test procedure to assess the statistical significance of classification performance for a given dataset size. This procedure is applied to several molecular classification problems representing a broad spectrum of levels of complexity.
- Published
- 2003
- Full Text
- View/download PDF
16. Multiclass cancer diagnosis using tumor gene expression signatures.
- Author
-
Ramaswamy S, Tamayo P, Rifkin R, Mukherjee S, Yeang CH, Angelo M, Ladd C, Reich M, Latulippe E, Mesirov JP, Poggio T, Gerald W, Loda M, Lander ES, and Golub TR
- Subjects
- Biomarkers, Tumor, Cluster Analysis, Humans, Multigene Family, Neoplasms genetics, Gene Expression Profiling, Neoplasms classification, Neoplasms diagnosis
- Abstract
The optimal treatment of patients with cancer depends on establishing accurate diagnoses by using a complex combination of clinical and histopathological data. In some instances, this task is difficult or impossible because of atypical clinical presentation or histopathology. To determine whether the diagnosis of multiple common adult malignancies could be achieved purely by molecular classification, we subjected 218 tumor samples, spanning 14 common tumor types, and 90 normal tissue samples to oligonucleotide microarray gene expression analysis. The expression levels of 16,063 genes and expressed sequence tags were used to evaluate the accuracy of a multiclass classifier based on a support vector machine algorithm. Overall classification accuracy was 78%, far exceeding the accuracy of random classification (9%). Poorly differentiated cancers resulted in low-confidence predictions and could not be accurately classified according to their tissue of origin, indicating that they are molecularly distinct entities with dramatically different gene expression patterns compared with their well differentiated counterparts. Taken together, these results demonstrate the feasibility of accurate, multiclass molecular cancer classification and suggest a strategy for future clinical implementation of molecular cancer diagnostics.
- Published
- 2001
- Full Text
- View/download PDF
17. Chemosensitivity prediction by transcriptional profiling.
- Author
-
Staunton JE, Slonim DK, Coller HA, Tamayo P, Angelo MJ, Park J, Scherf U, Lee JK, Reinhold WO, Weinstein JN, Mesirov JP, Lander ES, and Golub TR
- Subjects
- Gene Expression Profiling, Humans, Neoplasms drug therapy, Oligonucleotide Array Sequence Analysis methods, Predictive Value of Tests, Tumor Cells, Cultured, Drug Resistance, Neoplasm genetics, Neoplasms genetics, Transcription, Genetic
- Abstract
In an effort to develop a genomics-based approach to the prediction of drug response, we have developed an algorithm for classification of cell line chemosensitivity based on gene expression profiles alone. Using oligonucleotide microarrays, the expression levels of 6,817 genes were measured in a panel of 60 human cancer cell lines (the NCI-60) for which the chemosensitivity profiles of thousands of chemical compounds have been determined. We sought to determine whether the gene expression signatures of untreated cells were sufficient for the prediction of chemosensitivity. Gene expression-based classifiers of sensitivity or resistance for 232 compounds were generated and then evaluated on independent sets of data. The classifiers were designed to be independent of the cells' tissue of origin. The accuracy of chemosensitivity prediction was considerably better than would be expected by chance. Eighty-eight of 232 expression-based classifiers performed accurately (with P < 0.05) on an independent test set, whereas only 12 of the 232 would be expected to do so by chance. These results suggest that at least for a subset of compounds genomic approaches to chemosensitivity prediction are feasible.
- Published
- 2001
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.