Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Cell ; 173(2): 321-337.e10, 2018 04 05.
Article in English | MEDLINE | ID: mdl-29625050

ABSTRACT

Genetic alterations in signaling pathways that control cell-cycle progression, apoptosis, and cell growth are common hallmarks of cancer, but the extent, mechanisms, and co-occurrence of alterations in these pathways differ between individual tumors and tumor types. Using mutations, copy-number changes, mRNA expression, gene fusions and DNA methylation in 9,125 tumors profiled by The Cancer Genome Atlas (TCGA), we analyzed the mechanisms and patterns of somatic alterations in ten canonical pathways: cell cycle, Hippo, Myc, Notch, Nrf2, PI-3-Kinase/Akt, RTK-RAS, TGFß signaling, p53 and ß-catenin/Wnt. We charted the detailed landscape of pathway alterations in 33 cancer types, stratified into 64 subtypes, and identified patterns of co-occurrence and mutual exclusivity. Eighty-nine percent of tumors had at least one driver alteration in these pathways, and 57% percent of tumors had at least one alteration potentially targetable by currently available drugs. Thirty percent of tumors had multiple targetable alterations, indicating opportunities for combination therapy.


Subject(s)
Databases, Genetic , Neoplasms/pathology , Signal Transduction/genetics , Genes, Neoplasm , Humans , Neoplasms/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism
2.
Nature ; 573(7775): 526-531, 2019 09.
Article in English | MEDLINE | ID: mdl-31534217

ABSTRACT

Metastasis-the disseminated growth of tumours in distant organs-underlies cancer mortality. Breast-to-brain metastasis (B2BM) is a common and disruptive form of cancer and is prevalent in the aggressive basal-like subtype, but is also found at varying frequencies in all cancer subtypes. Previous studies revealed parameters of breast cancer metastasis to the brain, but its preference for this site remains an enigma. Here we show that B2BM cells co-opt a neuronal signalling pathway that was recently implicated in invasive tumour growth, involving activation by glutamate ligands of N-methyl-D-aspartate receptors (NMDARs), which is key in model systems for metastatic colonization of the brain and is associated with poor prognosis. Whereas NMDAR activation is autocrine in some primary tumour types, human and mouse B2BM cells express receptors but secrete insufficient glutamate to induce signalling, which is instead achieved by the formation of pseudo-tripartite synapses between cancer cells and glutamatergic neurons, presenting a rationale for brain metastasis.


Subject(s)
Brain Neoplasms/physiopathology , Brain Neoplasms/secondary , Receptors, N-Methyl-D-Aspartate/physiology , Signal Transduction/physiology , Synapses/physiology , Animals , Brain Neoplasms/ultrastructure , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Mice , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Neoplasm Metastasis , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/ultrastructure , Synaptic Transmission
3.
Proc Natl Acad Sci U S A ; 116(48): 24184-24195, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31704767

ABSTRACT

MicroRNA-mediated gene regulation has been implicated in various diseases, including cancer. This study examined the role of microRNAs (miRNAs) during tumorigenesis and malignant progression of pancreatic neuroendocrine tumors (PanNETs) in a genetically engineered mouse model. Previously, a set of miRNAs was observed to be specifically up-regulated in a highly invasive and metastatic subtype of mouse and human PanNET. Using functional assays, we now implicate different miRNAs in distinct phenotypes: miR-137 stimulates tumor growth and local invasion, whereas the miR-23b cluster enables metastasis. An algorithm, Bio-miRTa, has been developed to facilitate the identification of biologically relevant miRNA target genes and applied to these miRNAs. We show that a top-ranked miR-137 candidate gene, Sorl1, has a tumor suppressor function in primary PanNETs. Among the top targets for the miR-23b cluster, Acvr1c/ALK7 has recently been described to be a metastasis suppressor, and we establish herein that it is down-regulated by the miR-23b cluster, which is crucial for its prometastatic activity. Two other miR-23b targets, Robo2 and P2ry1, also have demonstrable antimetastatic effects. Finally, we have used the Bio-miRTa algorithm in reverse to identify candidate miRNAs that might regulate activin B, the principal ligand for ALK7, identifying thereby a third family of miRNAs-miRNA-130/301-that is congruently up-regulated concomitant with down-regulation of activin B during tumorigenesis, suggestive of functional involvement in evasion of the proapoptotic barrier. Thus, dynamic up-regulation of miRNAs during multistep tumorigenesis and malignant progression serves to down-regulate distinctive suppressor mechanisms of tumor growth, invasion, and metastasis.


Subject(s)
Cell Transformation, Neoplastic , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/pathology , Activin Receptors, Type I/genetics , Activins/genetics , Algorithms , Animals , Cell Line, Tumor , Computational Biology/methods , Doxycycline/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , LDL-Receptor Related Proteins/genetics , Liver Neoplasms/genetics , Liver Neoplasms/secondary , Membrane Transport Proteins/genetics , Mice , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/mortality , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/mortality , Prognosis , Receptors, LDL/genetics , Xenograft Model Antitumor Assays
4.
Nat Med ; 29(4): 833-845, 2023 04.
Article in English | MEDLINE | ID: mdl-37045996

ABSTRACT

Lung adenocarcinomas (LUADs) display a broad histological spectrum from low-grade lepidic tumors through to mid-grade acinar and papillary and high-grade solid, cribriform and micropapillary tumors. How morphology reflects tumor evolution and disease progression is poorly understood. Whole-exome sequencing data generated from 805 primary tumor regions and 121 paired metastatic samples across 248 LUADs from the TRACERx 421 cohort, together with RNA-sequencing data from 463 primary tumor regions, were integrated with detailed whole-tumor and regional histopathological analysis. Tumors with predominantly high-grade patterns showed increased chromosomal complexity, with higher burden of loss of heterozygosity and subclonal somatic copy number alterations. Individual regions in predominantly high-grade pattern tumors exhibited higher proliferation and lower clonal diversity, potentially reflecting large recent subclonal expansions. Co-occurrence of truncal loss of chromosomes 3p and 3q was enriched in predominantly low-/mid-grade tumors, while purely undifferentiated solid-pattern tumors had a higher frequency of truncal arm or focal 3q gains and SMARCA4 gene alterations compared with mixed-pattern tumors with a solid component, suggesting distinct evolutionary trajectories. Clonal evolution analysis revealed that tumors tend to evolve toward higher-grade patterns. The presence of micropapillary pattern and 'tumor spread through air spaces' were associated with intrathoracic recurrence, in contrast to the presence of solid/cribriform patterns, necrosis and preoperative circulating tumor DNA detection, which were associated with extra-thoracic recurrence. These data provide insights into the relationship between LUAD morphology, the underlying evolutionary genomic landscape, and clinical and anatomical relapse risk.


Subject(s)
Adenocarcinoma of Lung , Adenocarcinoma , Lung Neoplasms , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Neoplasm Recurrence, Local/pathology , Adenocarcinoma of Lung/genetics , Disease Progression , DNA Helicases , Nuclear Proteins , Transcription Factors
5.
Science ; 378(6621): eabl7207, 2022 11 18.
Article in English | MEDLINE | ID: mdl-36395212

ABSTRACT

Many human cancers manifest the capability to circumvent attack by the adaptive immune system. In this work, we identified a component of immune evasion that involves frequent up-regulation of fragile X mental retardation protein (FMRP) in solid tumors. FMRP represses immune attack, as revealed by cancer cells engineered to lack its expression. FMRP-deficient tumors were infiltrated by activated T cells that impaired tumor growth and enhanced survival in mice. Mechanistically, FMRP's immunosuppression was multifactorial, involving repression of the chemoattractant C-C motif chemokine ligand 7 (CCL7) concomitant with up-regulation of three immunomodulators-interleukin-33 (IL-33), tumor-secreted protein S (PROS1), and extracellular vesicles. Gene signatures associate FMRP's cancer network with poor prognosis and response to therapy in cancer patients. Collectively, FMRP is implicated as a regulator that orchestrates a multifaceted barrier to antitumor immune responses.


Subject(s)
Fragile X Mental Retardation Protein , Immune Evasion , Immune Tolerance , Neoplasms , Animals , Humans , Mice , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Neoplasms/immunology , Chemokine CCL7/metabolism , Interleukin-33 , Protein S/metabolism
6.
Cancer Discov ; 11(10): 2638-2657, 2021 10.
Article in English | MEDLINE | ID: mdl-33910926

ABSTRACT

Pancreatic neuroendocrine tumors (PanNET) comprise two molecular subtypes, relatively benign islet tumors (IT) and invasive, metastasis-like primary (MLP) tumors. Until now, the origin of aggressive MLP tumors has been obscure. Herein, using multi-omics approaches, we revealed that MLP tumors arise from IT via dedifferentiation following a reverse trajectory along the developmental pathway of islet ß cells, which results in the acquisition of a progenitor-like molecular phenotype. Functionally, the miR-181cd cluster induces the IT-to-MLP transition by suppressing expression of the Meis2 transcription factor, leading to upregulation of a developmental transcription factor, Hmgb3. Notably, the IT-to-MLP transition constitutes a distinct step of tumorigenesis and is separable from the classic proliferation-associated hallmark, temporally preceding accelerated proliferation of cancer cells. Furthermore, patients with PanNET with elevated HMGB3 expression and an MLP transcriptional signature are associated with higher-grade tumors and worse survival. Overall, our results unveil a new mechanism that modulates cancer cell plasticity to enable malignant progression. SIGNIFICANCE: Dedifferentiation has long been observed as a histopathologic characteristic of many cancers, albeit inseparable from concurrent increases in cell proliferation. Herein, we demonstrate that dedifferentiation is a mechanistically and temporally separable step in the multistage tumorigenesis of pancreatic islet cells, retracing the developmental lineage of islet ß cells.This article is highlighted in the In This Issue feature, p. 2355.


Subject(s)
Cell Transformation, Neoplastic , Gene Expression Regulation , Neuroendocrine Tumors/genetics , Pancreatic Neoplasms/genetics , Animals , Disease Models, Animal , Mice , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/pathology
7.
Sci Rep ; 11(1): 10455, 2021 05 17.
Article in English | MEDLINE | ID: mdl-34001972

ABSTRACT

Lung carcinoids are variably aggressive and mechanistically understudied neuroendocrine neoplasms (NENs). Here, we identified and elucidated the function of a miR-375/yes-associated protein (YAP) axis in lung carcinoid (H727) cells. miR-375 and YAP are respectively high and low expressed in wild-type H727 cells. Following lentiviral CRISPR/Cas9-mediated miR-375 depletion, we identified distinct transcriptomic changes including dramatic YAP upregulation. We also observed a significant decrease in neuroendocrine differentiation and substantial reductions in cell proliferation, transformation, and tumor growth in cell culture and xenograft mouse disease models. Similarly, YAP overexpression resulted in distinct and partially overlapping transcriptomic changes, phenocopying the effects of miR-375 depletion in the same models as above. Transient YAP knockdown in miR-375-depleted cells reversed the effects of miR-375 on neuroendocrine differentiation and cell proliferation. Pathways analysis and confirmatory real-time PCR studies of shared dysregulated target genes indicate that this axis controls neuroendocrine related functions such as neural differentiation, exocytosis, and secretion. Taken together, we provide compelling evidence that a miR-375/YAP axis is a critical mediator of neuroendocrine differentiation and tumorigenesis in lung carcinoid cells.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Carcinoid Tumor/genetics , Lung Neoplasms/genetics , MicroRNAs/metabolism , Neuroendocrine Cells/pathology , Transcription Factors/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Carcinogenesis/genetics , Carcinoid Tumor/pathology , Cell Differentiation/genetics , Cell Proliferation/genetics , Exocytosis/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HEK293 Cells , Humans , Lung Neoplasms/pathology , Mice , Mice, Knockout , MicroRNAs/genetics , Transcription Factors/metabolism , Xenograft Model Antitumor Assays , YAP-Signaling Proteins
8.
Dev Cell ; 49(3): 409-424.e6, 2019 05 06.
Article in English | MEDLINE | ID: mdl-31063757

ABSTRACT

Herein, we report that the TGFß superfamily receptor ALK7 is a suppressor of tumorigenesis and metastasis, as revealed by functional studies in mouse models of pancreatic neuroendocrine and luminal breast cancer, complemented by experimental metastasis assays. Activation in neoplastic cells of the ALK7 signaling pathway by its principal ligand activin B induces apoptosis. During tumorigenesis, cancer cells use two different approaches to evade this barrier, either downregulating activin B and/or downregulating ALK7. Suppressing ALK7 expression additionally contributes to the capability for metastatic seeding. ALK7 is associated with shorter relapse-free survival of various human cancers and distant-metastasis-free survival of breast cancer patients. This study introduces mechanistic insights into primary and metastatic tumor development, in the form of a protective barrier that triggers apoptosis in cells that are not "authorized" to proliferate within a particular tissue, by virtue of those cells expressing ALK7 in a tissue microenvironment bathed in its ligand.


Subject(s)
Activin Receptors, Type I/metabolism , Activins/metabolism , Neoplasms/metabolism , Animals , Apoptosis/physiology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinogenesis , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Female , Heterografts , Homeostasis , Humans , Male , Mice , Mice, Inbred A , Mice, Inbred C57BL , Mice, SCID , Neoplasm Metastasis , Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Signal Transduction , Smad2 Protein/metabolism , Transforming Growth Factor beta/metabolism , Tumor Microenvironment
9.
Cell Rep ; 25(4): 1066-1080.e8, 2018 10 23.
Article in English | MEDLINE | ID: mdl-30355485

ABSTRACT

The discovery of cancer-associated alterations has primarily focused on genetic variants. Nonetheless, altered epigenomes contribute to deregulate transcription and promote oncogenic pathways. Here, we designed an algorithmic approach (RESET) to identify aberrant DNA methylation and associated cis-transcriptional changes across >6,000 human tumors. Tumors exhibiting mutations of chromatin remodeling factors and Wnt signaling displayed DNA methylation instability, characterized by numerous hyper- and hypo-methylated loci. Most silenced and enhanced genes coalesced in specific pathways including apoptosis, DNA repair, and cell metabolism. Cancer-germline antigens (CG) were frequently epigenomically enhanced and their expression correlated with response to anti-PD-1, but not anti-CTLA4, in skin melanoma. Finally, we demonstrated the potential of our approach to explore DNA methylation changes in pediatric tumors, which frequently lack genetic drivers and exhibit epigenomic modifications. Our results provide a pan-cancer map of aberrant DNA methylation to inform functional and therapeutic studies.


Subject(s)
DNA Methylation/genetics , Neoplasms/genetics , Cell Line, Tumor , Child , Epigenesis, Genetic , Gene Silencing , Humans
10.
Cancer Cell ; 32(2): 155-168.e6, 2017 08 14.
Article in English | MEDLINE | ID: mdl-28756993

ABSTRACT

Cancer evolves through the emergence and selection of molecular alterations. Cancer genome profiling has revealed that specific events are more or less likely to be co-selected, suggesting that the selection of one event depends on the others. However, the nature of these evolutionary dependencies and their impact remain unclear. Here, we designed SELECT, an algorithmic approach to systematically identify evolutionary dependencies from alteration patterns. By analyzing 6,456 genomes from multiple tumor types, we constructed a map of oncogenic dependencies associated with cellular pathways, transcriptional readouts, and therapeutic response. Finally, modeling of cancer evolution shows that alteration dependencies emerge only under conditional selection. These results provide a framework for the design of strategies to predict cancer progression and therapeutic response.


Subject(s)
Algorithms , Carcinogenesis , Evolution, Molecular , Neoplasms/genetics , Selection, Genetic , Gene Expression Profiling , Genomics , Humans , Models, Genetic
11.
Cell Rep ; 15(6): 1144-60, 2016 05 10.
Article in English | MEDLINE | ID: mdl-27134166

ABSTRACT

Therapeutic targeting of tumor angiogenesis with VEGF inhibitors results in demonstrable, but transitory efficacy in certain human tumors and mouse models of cancer, limited by unconventional forms of adaptive/evasive resistance. In one such mouse model, potent angiogenesis inhibitors elicit compartmental reorganization of cancer cells around remaining blood vessels. The glucose and lactate transporters GLUT1 and MCT4 are induced in distal hypoxic cells in a HIF1α-dependent fashion, indicative of glycolysis. Tumor cells proximal to blood vessels instead express the lactate transporter MCT1, and p-S6, the latter reflecting mTOR signaling. Normoxic cancer cells import and metabolize lactate, resulting in upregulation of mTOR signaling via glutamine metabolism enhanced by lactate catabolism. Thus, metabolic symbiosis is established in the face of angiogenesis inhibition, whereby hypoxic cancer cells import glucose and export lactate, while normoxic cells import and catabolize lactate. mTOR signaling inhibition disrupts this metabolic symbiosis, associated with upregulation of the glucose transporter GLUT2.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Drug Resistance, Neoplasm , Intestinal Neoplasms/blood supply , Intestinal Neoplasms/metabolism , Neuroendocrine Tumors/blood supply , Neuroendocrine Tumors/metabolism , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/metabolism , Signal Transduction , Stomach Neoplasms/blood supply , Stomach Neoplasms/metabolism , TOR Serine-Threonine Kinases/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Axitinib , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glucose/metabolism , Glutamine/metabolism , Glycolysis/drug effects , Humans , Imidazoles/pharmacology , Imidazoles/therapeutic use , Indazoles/pharmacology , Indazoles/therapeutic use , Indoles/pharmacology , Indoles/therapeutic use , Intestinal Neoplasms/drug therapy , Lactic Acid/metabolism , Membrane Transport Proteins/metabolism , Mice , Models, Biological , Neuroendocrine Tumors/drug therapy , Pancreatic Neoplasms/drug therapy , Pyrroles/pharmacology , Pyrroles/therapeutic use , Signal Transduction/drug effects , Sirolimus/pharmacology , Stomach Neoplasms/drug therapy , Sunitinib , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL