Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 24
1.
Oncogene ; 2024 Apr 02.
Article En | MEDLINE | ID: mdl-38565943

Cancer cells employ adaptive mechanisms to survive various stressors, including genotoxic drugs. Understanding the factors promoting survival is crucial for developing effective treatments. In this study, we unveil a previously unexplored long non-coding RNA, JUNI (JUN-DT, LINC01135), which is upregulated by genotoxic drugs through the activation of stress-activated MAPKs, JNK, and p38 and consequently exerts positive control over the expression of its adjacent gene product c-Jun, a well-known oncoprotein, which transduces signals to multiple transcriptional outputs. JUNI regulates cellular migration and has a crucial role in conferring cellular resistance to chemotherapeutic drugs or UV radiation. Depletion of JUNI markedly increases the sensitivity of cultured cells and spheroids to chemotherapeutic agents. We identified 57 proteins interacting with JUNI. The activity of one of them the MAPK phosphatase and inhibitor, DUSP14, is counteracted by JUNI, thereby, facilitating efficient JNK phosphorylation and c-Jun induction when cells are exposed to UV radiation. The antagonistic interplay with DUSP14 contributes not only to c-Jun induction but also augments the survival of UV-exposed cells. In summary, we introduce JUNI as a novel stress-inducible regulator of c-Jun, positioning it as a potential target for enhancing the sensitivity of cancer cells to chemotherapy.

2.
Elife ; 132024 Apr 10.
Article En | MEDLINE | ID: mdl-38597390

Alternative RNA splicing is an essential and dynamic process in neuronal differentiation and synapse maturation, and dysregulation of this process has been associated with neurodegenerative diseases. Recent studies have revealed the importance of RNA-binding proteins in the regulation of neuronal splicing programs. However, the molecular mechanisms involved in the control of these splicing regulators are still unclear. Here, we show that KIS, a kinase upregulated in the developmental brain, imposes a genome-wide alteration in exon usage during neuronal differentiation in mice. KIS contains a protein-recognition domain common to spliceosomal components and phosphorylates PTBP2, counteracting the role of this splicing factor in exon exclusion. At the molecular level, phosphorylation of unstructured domains within PTBP2 causes its dissociation from two co-regulators, Matrin3 and hnRNPM, and hinders the RNA-binding capability of the complex. Furthermore, KIS and PTBP2 display strong and opposing functional interactions in synaptic spine emergence and maturation. Taken together, our data uncover a post-translational control of splicing regulators that link transcriptional and alternative exon usage programs in neuronal development.


Alternative Splicing , Exons , Neurons , Polypyrimidine Tract-Binding Protein , Protein Serine-Threonine Kinases , Animals , Humans , Mice , Exons/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Neurons/metabolism , Phosphorylation , Polypyrimidine Tract-Binding Protein/metabolism , Polypyrimidine Tract-Binding Protein/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism
3.
Nat Methods ; 21(3): 423-434, 2024 Mar.
Article En | MEDLINE | ID: mdl-38374261

Different regions of RNA molecules can often engage in specific interactions with distinct RNA-binding proteins (RBPs), giving rise to diverse modalities of RNA regulation and function. However, there are currently no methods for unbiased identification of RBPs that interact with specific RNA regions in living cells and under endogenous settings. Here we introduce TREX (targeted RNase H-mediated extraction of crosslinked RBPs)-a highly sensitive approach for identifying proteins that directly bind to specific RNA regions in living cells. We demonstrate that TREX outperforms existing methods in identifying known interactors of U1 snRNA, and reveals endogenous region-specific interactors of NORAD long noncoding RNA. Using TREX, we generated a comprehensive region-by-region interactome for 45S rRNA, uncovering both established and previously unknown interactions that regulate ribosome biogenesis. With its applicability to different cell types, TREX is an RNA-centric tool for unbiased positional mapping of endogenous RNA-protein interactions in living cells.


RNA-Binding Proteins , RNA , RNA/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
4.
EMBO J ; 42(11): e110902, 2023 06 01.
Article En | MEDLINE | ID: mdl-37039106

Oncogenic RAS signaling reprograms gene expression through both transcriptional and post-transcriptional mechanisms. While transcriptional regulation downstream of RAS is relatively well characterized, how RAS post-transcriptionally modulates gene expression to promote malignancy remains largely unclear. Using quantitative RNA interactome capture analysis, we here reveal that oncogenic RAS signaling reshapes the RNA-bound proteomic landscape of pancreatic cancer cells, with a network of nuclear proteins centered around nucleolin displaying enhanced RNA-binding activity. We show that nucleolin is phosphorylated downstream of RAS, which increases its binding to pre-ribosomal RNA (rRNA), boosts rRNA production, and promotes ribosome biogenesis. This nucleolin-dependent enhancement of ribosome biogenesis is crucial for RAS-induced pancreatic cancer cell proliferation and can be targeted therapeutically to inhibit tumor growth. Our results reveal that oncogenic RAS signaling drives ribosome biogenesis by regulating the RNA-binding activity of nucleolin and highlight a crucial role for this mechanism in RAS-mediated tumorigenesis.


Genes, ras , Pancreatic Neoplasms , Humans , MAP Kinase Signaling System , Proteomics , Phosphoproteins/metabolism , RNA, Ribosomal/metabolism , RNA/metabolism , Cell Transformation, Neoplastic/genetics , Ribosomes/genetics , Ribosomes/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Nucleolin
5.
Development ; 149(13)2022 07 01.
Article En | MEDLINE | ID: mdl-35723257

Precise vascular patterning is crucial for normal growth and development. The ERG transcription factor drives Delta-like ligand 4 (DLL4)/Notch signalling and is thought to act as a pivotal regulator of endothelial cell (EC) dynamics and developmental angiogenesis. However, molecular regulation of ERG activity remains obscure. Using a series of EC-specific focal adhesion kinase (FAK)-knockout (KO) and point-mutant FAK-knock-in mice, we show that loss of ECFAK, its kinase activity or phosphorylation at FAK-Y397, but not FAK-Y861, reduces ERG and DLL4 expression levels together with concomitant aberrations in vascular patterning. Rapid immunoprecipitation mass spectrometry of endogenous proteins identified that endothelial nuclear-FAK interacts with the deubiquitinase USP9x and the ubiquitin ligase TRIM25. Further in silico analysis confirms that ERG interacts with USP9x and TRIM25. Moreover, ERG levels are reduced in FAKKO ECs via a ubiquitin-mediated post-translational modification programme involving USP9x and TRIM25. Re-expression of ERG in vivo and in vitro rescues the aberrant vessel-sprouting defects observed in the absence of ECFAK. Our findings identify ECFAK as a regulator of retinal vascular patterning by controlling ERG protein degradation via TRIM25/USP9x.


Endothelial Cells , Transcription Factors , Animals , Endothelial Cells/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Mice , Neovascularization, Physiologic/genetics , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitins/metabolism
6.
Cell Death Dis ; 12(11): 1075, 2021 11 11.
Article En | MEDLINE | ID: mdl-34764236

An early event in lung oncogenesis is loss of the tumour suppressor gene LIMD1 (LIM domains containing 1); this encodes a scaffold protein, which suppresses tumorigenesis via a number of different mechanisms. Approximately 45% of non-small cell lung cancers (NSCLC) are deficient in LIMD1, yet this subtype of NSCLC has been overlooked in preclinical and clinical investigations. Defining therapeutic targets in these LIMD1 loss-of-function patients is difficult due to a lack of 'druggable' targets, thus alternative approaches are required. To this end, we performed the first drug repurposing screen to identify compounds that confer synthetic lethality with LIMD1 loss in NSCLC cells. PF-477736 was shown to selectively target LIMD1-deficient cells in vitro through inhibition of multiple kinases, inducing cell death via apoptosis. Furthermore, PF-477736 was effective in treating LIMD1-/- tumours in subcutaneous xenograft models, with no significant effect in LIMD1+/+ cells. We have identified a novel drug tool with significant preclinical characterisation that serves as an excellent candidate to explore and define LIMD1-deficient cancers as a new therapeutic subgroup of critical unmet need.


Carcinoma, Non-Small-Cell Lung/genetics , Intracellular Signaling Peptides and Proteins/deficiency , LIM Domain Proteins/deficiency , Lung Neoplasms/genetics , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Female , Humans , Lung Neoplasms/pathology , Mice , Mice, Inbred NOD , Proof of Concept Study , Transfection
7.
Sci Signal ; 14(691)2021 07 13.
Article En | MEDLINE | ID: mdl-34257105

Synaptic plasticity involves structural modifications in dendritic spines that are modulated by local protein synthesis and actin remodeling. Here, we investigated the molecular mechanisms that connect synaptic stimulation to these processes. We found that the phosphorylation of isoform-specific sites in eEF1A2-an essential translation elongation factor in neurons-is a key modulator of structural plasticity in dendritic spines. Expression of a nonphosphorylatable eEF1A2 mutant stimulated mRNA translation but reduced actin dynamics and spine density. By contrast, a phosphomimetic eEF1A2 mutant exhibited decreased association with F-actin and was inactive as a translation elongation factor. Activation of metabotropic glutamate receptor signaling triggered transient dissociation of eEF1A2 from its regulatory guanine exchange factor (GEF) protein in dendritic spines in a phosphorylation-dependent manner. We propose that eEF1A2 establishes a cross-talk mechanism that coordinates translation and actin dynamics during spine remodeling.


Actins , Dendritic Spines , Peptide Elongation Factor 1/metabolism , Protein Biosynthesis , Actin Cytoskeleton , Actins/genetics , Neuronal Plasticity , Neurons
8.
Br J Cancer ; 125(5): 699-713, 2021 08.
Article En | MEDLINE | ID: mdl-34172930

BACKGROUND: Metastasis is a hallmark of cancer and responsible for most cancer deaths. Migrastatics were defined as drugs interfering with all modes of cancer cell invasion and thus cancers' ability to metastasise. First anti-metastatic treatments have recently been approved. METHODS: We used bioinformatic analyses of publicly available melanoma databases. Experimentally, we performed in vitro target validation (including 2.5D cell morphology analysis and mass spectrometric analysis of RhoA binding partners), developed a new traceable spontaneously metastasising murine melanoma model for in vivo validation, and employed histology (haematoxylin/eosin and phospho-myosin II staining) to confirm drug action in harvested tumour tissues. RESULTS: Unbiased and targeted bioinformatic analyses identified the Rho kinase (ROCK)-myosin II pathway and its various components as potentially relevant targets in melanoma. In vitro validation demonstrated redundancy of several RhoGEFs upstream of RhoA and confirmed ROCK as a druggable target downstream of RhoA. The anti-metastatic effects of two ROCK inhibitors were demonstrated through in vivo melanoma metastasis tracking and inhibitor effects also confirmed ex vivo by digital pathology. CONCLUSIONS: We proposed a migrastatic drug development pipeline. As part of the pipeline, we provide a new traceable spontaneous melanoma metastasis model for in vivo quantification of metastasis and anti-metastatic effects by non-invasive imaging.


Computational Biology/methods , Melanoma/drug therapy , Myosin Type II/metabolism , Protein Kinase Inhibitors/administration & dosage , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Cell Line, Tumor , Cell Movement/drug effects , Humans , Male , Mass Spectrometry , Melanoma/metabolism , Mice , Neoplasm Metastasis , Protein Interaction Maps , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
9.
Science ; 372(6543)2021 05 14.
Article En | MEDLINE | ID: mdl-33986153

Aberrant alternative splicing is a hallmark of cancer, yet the underlying regulatory programs that control this process remain largely unknown. Here, we report a systematic effort to decipher the RNA structural code that shapes pathological splicing during breast cancer metastasis. We discovered a previously unknown structural splicing enhancer that is enriched near cassette exons with increased inclusion in highly metastatic cells. We show that the spliceosomal protein small nuclear ribonucleoprotein polypeptide A' (SNRPA1) interacts with these enhancers to promote cassette exon inclusion. This interaction enhances metastatic lung colonization and cancer cell invasion, in part through SNRPA1-mediated regulation of PLEC alternative splicing, which can be counteracted by splicing modulating morpholinos. Our findings establish a noncanonical regulatory role for SNRPA1 as a prometastatic splicing enhancer in breast cancer.


Alternative Splicing , Breast Neoplasms/pathology , Neoplasm Metastasis/genetics , RNA/genetics , RNA/metabolism , Ribonucleoprotein, U2 Small Nuclear/metabolism , Adaptor Proteins, Signal Transducing/genetics , Algorithms , Animals , Binding Sites , Breast Neoplasms/genetics , Cell Line, Tumor , Disease Progression , Exons , Gene Knockdown Techniques , Humans , Lung Neoplasms/secondary , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness , Neoplasm Transplantation , Nucleic Acid Conformation , Plectin/genetics , Protein Binding , RNA Interference , RNA, Small Nuclear/chemistry , RNA, Small Nuclear/metabolism , RNA-Seq , Ribonucleoprotein, U2 Small Nuclear/genetics , Software , Spliceosomes/metabolism , Tumor Suppressor Proteins/genetics
10.
STAR Protoc ; 2(2): 100462, 2021 06 18.
Article En | MEDLINE | ID: mdl-33912849

Actin-rich protrusions are membrane extensions generated by actin polymerization that drive mesenchymal-like cell migration. Characterization of protrusions proteome is crucial for understanding their function. We present a complete step-by-step protocol based on microporous filter-based fractionation of protrusive cellular domains coupled with sample preparation for quantitative proteomics, mass spectrometric data acquisition, and data analysis. This protocol enables purification, quantification, and analysis of the distribution of proteins present in protrusions and cell bodies. For complete details on the use and execution of this protocol, please refer to Dermit et al. (2020).


Cell Body , Cell Surface Extensions , Proteomics , Cell Body/chemistry , Cell Body/metabolism , Cell Line, Tumor , Cell Surface Extensions/chemistry , Cell Surface Extensions/metabolism , Humans
11.
Dev Cell ; 55(3): 298-313.e10, 2020 11 09.
Article En | MEDLINE | ID: mdl-33171110

Translation of ribosomal protein-coding mRNAs (RP-mRNAs) constitutes a key step in ribosome biogenesis, but the mechanisms that modulate RP-mRNA translation in coordination with other cellular processes are poorly defined. Here, we show that subcellular localization of RP-mRNAs acts as a key regulator of their translation during cell migration. As cells migrate into their surroundings, RP-mRNAs localize to the actin-rich cell protrusions. This localization is mediated by La-related protein 6 (LARP6), an RNA-binding protein that is enriched in protrusions. Protrusions act as hotspots of translation for RP-mRNAs, enhancing RP synthesis, ribosome biogenesis, and the overall protein synthesis in migratory cells. In human breast carcinomas, epithelial-to-mesenchymal transition (EMT) upregulates LARP6 expression to enhance protein synthesis and support invasive growth. Our findings reveal LARP6-mediated mRNA localization as a key regulator of ribosome biogenesis during cell migration and demonstrate a role for this process in cancer progression downstream of EMT.


Cell Movement , Organelle Biogenesis , RNA Transport , Ribosomes/metabolism , Autoantigens/metabolism , Cell Proliferation , Cell Surface Extensions/metabolism , Epithelial-Mesenchymal Transition , Humans , Neoplasms/metabolism , Neoplasms/pathology , Protein Binding , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribonucleoproteins/metabolism , Ribosomal Proteins/metabolism , Subcellular Fractions/metabolism , Transcriptome/genetics , SS-B Antigen
12.
Cancer Discov ; 10(9): 1410-1423, 2020 09.
Article En | MEDLINE | ID: mdl-32513775

Identifying master regulators that drive pathologic gene expression is a key challenge in precision oncology. Here, we have developed an analytic framework, named PRADA, that identifies oncogenic RNA-binding proteins through the systematic detection of coordinated changes in their target regulons. Application of this approach to data collected from clinical samples, patient-derived xenografts, and cell line models of colon cancer metastasis revealed the RNA-binding protein RBMS1 as a suppressor of colon cancer progression. We observed that silencing RBMS1 results in increased metastatic capacity in xenograft mouse models, and that restoring its expression blunts metastatic liver colonization. We have found that RBMS1 functions as a posttranscriptional regulator of RNA stability by directly binding its target mRNAs. Together, our findings establish a role for RBMS1 as a previously unknown regulator of RNA stability and as a suppressor of colon cancer metastasis with clinical utility for risk stratification of patients. SIGNIFICANCE: By applying a new analytic approach to transcriptomic data from clinical samples and models of colon cancer progression, we have identified RBMS1 as a suppressor of metastasis and as a post-transcriptional regulator of RNA stability. Notably, RBMS1 silencing and downregulation of its targets are negatively associated with patient survival.See related commentary by Carter, p. 1261.This article is highlighted in the In This Issue feature, p. 1241.


Colonic Neoplasms/pathology , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/metabolism , Animals , Cell Line, Tumor , Colon/pathology , Colonic Neoplasms/diagnosis , Colonic Neoplasms/genetics , DNA-Binding Proteins/genetics , Gene Knockdown Techniques , Humans , Intestinal Mucosa/pathology , Liver Neoplasms/secondary , Male , Mice , Neoplasm Staging , RNA Stability/genetics , RNA-Binding Proteins/genetics , RNA-Seq , Regulon , Xenograft Model Antitumor Assays
13.
Development ; 147(4)2020 02 26.
Article En | MEDLINE | ID: mdl-32054660

La-related protein 6 (Larp6) is a conserved RNA-binding protein found across eukaryotes that has been suggested to regulate collagen biogenesis, muscle development, ciliogenesis, and various aspects of cell proliferation and migration. Zebrafish have two Larp6 family genes: larp6a and larp6b Viable and fertile single and double homozygous larp6a and larp6b zygotic mutants revealed no defects in muscle structure, and were indistinguishable from heterozygous or wild-type siblings. However, larp6a mutant females produced eggs with chorions that failed to elevate fully and were fragile. Eggs from larp6b single mutant females showed minor chorion defects, but chorions from eggs laid by larp6a;larp6b double mutant females were more defective than those from larp6a single mutants. Electron microscopy revealed defective chorionogenesis during oocyte development. Despite this, maternal zygotic single and double mutants were viable and fertile. Mass spectrometry analysis provided a description of chorion protein composition and revealed significant reductions in a subset of zona pellucida and lectin-type proteins between wild-type and mutant chorions that paralleled the severity of the phenotype. We conclude that Larp6 proteins are required for normal oocyte development, chorion formation and egg activation.


Autoantigens/genetics , Autoantigens/physiology , Chorion/physiology , Oocytes/physiology , Ribonucleoproteins/genetics , Ribonucleoproteins/physiology , Animals , Cell Movement , Cell Proliferation , Collagen/physiology , Egg Proteins/physiology , Female , Gene Editing , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genome , Genotype , Heterozygote , Homozygote , Lectins/physiology , Male , Mutation , Oocytes/cytology , Oogenesis/physiology , Phenotype , Zebrafish , Zona Pellucida/physiology , SS-B Antigen
14.
Mol Biosyst ; 13(12): 2477-2488, 2017 Nov 21.
Article En | MEDLINE | ID: mdl-29051942

Regulation of protein translation constitutes a crucial step in control of gene expression. In comparison to transcriptional regulation, however, translational control has remained a significantly under-studied layer of gene expression. This trend is now beginning to shift thanks to recent advances in next-generation sequencing, proteomics, and microscopy based methodologies which allow accurate monitoring of protein translation rates, from single target messenger RNA molecules to genome-wide scale studies. In this review, we summarize these recent advances, and discuss how they are enabling researchers to study translational regulation in a wide variety of in vitro and in vivo biological systems, with unprecedented depth and spatiotemporal resolution.


Protein Transport/physiology , Animals , Gene Expression Profiling , Gene Expression Regulation , Humans , Protein Biosynthesis , Protein Transport/genetics , RNA, Messenger , Ribosomes/metabolism
15.
Methods Mol Biol ; 1636: 337-352, 2017.
Article En | MEDLINE | ID: mdl-28730490

A major challenge in systems biology is comprehensive mapping of protein interaction networks. Crucially, such interactions are often dynamic in nature, necessitating methods that can rapidly mine the interactome across varied conditions and treatments to reveal change in the interaction networks. Recently, we described a fast mass spectrometry-based method to reveal functional interactions in mammalian cells on a global scale, by revealing spatial colocalizations between proteins (COLA) (Mardakheh et al., Mol Biosyst 13:92-105, 2017). As protein localization and function are inherently linked, significant colocalization between two proteins is a strong indication for their functional interaction. COLA uses rapid complete subcellular fractionation, coupled with quantitative proteomics to generate a subcellular localization profile for each protein quantified by the mass spectrometer. Robust clustering is then applied to reveal significant similarities in protein localization profiles, indicative of colocalization.


Protein Interaction Mapping/methods , Protein Interaction Maps , Proteome , Proteomics/methods , Cell Fractionation/methods , Chromatography, Liquid , Cluster Analysis , Computational Biology/methods , Mass Spectrometry , Protein Binding , Protein Transport , Staining and Labeling , Statistics as Topic , Workflow
16.
Mol Biosyst ; 13(1): 92-105, 2016 Dec 20.
Article En | MEDLINE | ID: mdl-27824369

Localisation and protein function are intimately linked in eukaryotes, as proteins are localised to specific compartments where they come into proximity of other functionally relevant proteins. Significant co-localisation of two proteins can therefore be indicative of their functional association. We here present COLA, a proteomics based strategy coupled with a bioinformatics framework to detect protein-protein co-localisations on a global scale. COLA reveals functional interactions by matching proteins with significant similarity in their subcellular localisation signatures. The rapid nature of COLA allows mapping of interactome dynamics across different conditions or treatments with high precision.


Protein Interaction Mapping/methods , Protein Interaction Maps , Proteome , Proteomics , Cell Fractionation , Cell Line , Chromatography, Liquid , Cluster Analysis , Humans , Intracellular Space/metabolism , Mass Spectrometry , Protein Binding , Protein Transport , Proteomics/methods , Sensitivity and Specificity , Subcellular Fractions
17.
J Cell Sci ; 129(24): 4466-4479, 2016 12 15.
Article En | MEDLINE | ID: mdl-27807006

Directional cell migration involves reorientation of the secretory machinery. However, the molecular mechanisms that control this reorientation are not well characterised. Here, we identify a new Rho effector protein, named FAM65A, which binds to active RHOA, RHOB and RHOC. FAM65A links RHO proteins to Golgi-localising cerebral cavernous malformation-3 protein (CCM3; also known as PDCD10) and its interacting proteins mammalian STE20-like protein kinases 3 and 4 (MST3 and MST4; also known as STK24 and STK26, respectively). Binding of active RHO proteins to FAM65A does not affect the kinase activity of MSTs but results in their relocation from the Golgi in a CCM3-dependent manner. This relocation is crucial for reorientation of the Golgi towards the leading edge and subsequent directional cell migration. Our results reveal a previously unidentified pathway downstream of RHO that regulates the polarity of migrating cells through Golgi reorientation in a FAM65A-, CCM3- and MST3- and MST4-dependent manner.


Cell Movement , Golgi Apparatus/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , rho GTP-Binding Proteins/metabolism , Amino Acid Sequence , Apoptosis Regulatory Proteins/metabolism , Enzyme Activation , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Membrane Proteins/metabolism , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Protein Transport , Proto-Oncogene Proteins/metabolism
18.
PLoS Med ; 13(2): e1001961, 2016 Feb.
Article En | MEDLINE | ID: mdl-26881778

BACKGROUND: The intra-tumor diversity of cancer cells is under intense investigation; however, little is known about the heterogeneity of the tumor microenvironment that is key to cancer progression and evolution. We aimed to assess the degree of microenvironmental heterogeneity in breast cancer and correlate this with genomic and clinical parameters. METHODS AND FINDINGS: We developed a quantitative measure of microenvironmental heterogeneity along three spatial dimensions (3-D) in solid tumors, termed the tumor ecosystem diversity index (EDI), using fully automated histology image analysis coupled with statistical measures commonly used in ecology. This measure was compared with disease-specific survival, key mutations, genome-wide copy number, and expression profiling data in a retrospective study of 510 breast cancer patients as a test set and 516 breast cancer patients as an independent validation set. In high-grade (grade 3) breast cancers, we uncovered a striking link between high microenvironmental heterogeneity measured by EDI and a poor prognosis that cannot be explained by tumor size, genomics, or any other data types. However, this association was not observed in low-grade (grade 1 and 2) breast cancers. The prognostic value of EDI was superior to known prognostic factors and was enhanced with the addition of TP53 mutation status (multivariate analysis test set, p = 9 × 10-4, hazard ratio = 1.47, 95% CI 1.17-1.84; validation set, p = 0.0011, hazard ratio = 1.78, 95% CI 1.26-2.52). Integration with genome-wide profiling data identified losses of specific genes on 4p14 and 5q13 that were enriched in grade 3 tumors with high microenvironmental diversity that also substratified patients into poor prognostic groups. Limitations of this study include the number of cell types included in the model, that EDI has prognostic value only in grade 3 tumors, and that our spatial heterogeneity measure was dependent on spatial scale and tumor size. CONCLUSIONS: To our knowledge, this is the first study to couple unbiased measures of microenvironmental heterogeneity with genomic alterations to predict breast cancer clinical outcome. We propose a clinically relevant role of microenvironmental heterogeneity for advanced breast tumors, and highlight that ecological statistics can be translated into medical advances for identifying a new type of biomarker and, furthermore, for understanding the synergistic interplay of microenvironmental heterogeneity with genomic alterations in cancer cells.


Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , DNA, Neoplasm/genetics , Gene Expression Profiling/methods , Genomics/methods , Neoplasm Staging , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Disease Progression , Female , Humans , Middle Aged , Prognosis , Retrospective Studies , Young Adult
19.
Elife ; 5: e12994, 2016 Jan 14.
Article En | MEDLINE | ID: mdl-26765561

Rho-associated kinases 1 and 2 (ROCK1/2) are Rho-GTPase effectors that control key aspects of the actin cytoskeleton, but their role in proliferation and cancer initiation or progression is not known. Here, we provide evidence that ROCK1 and ROCK2 act redundantly to maintain actomyosin contractility and cell proliferation and that their loss leads to cell-cycle arrest and cellular senescence. This phenotype arises from down-regulation of the essential cell-cycle proteins CyclinA, CKS1 and CDK1. Accordingly, while the loss of either Rock1 or Rock2 had no negative impact on tumorigenesis in mouse models of non-small cell lung cancer and melanoma, loss of both blocked tumor formation, as no tumors arise in which both Rock1 and Rock2 have been genetically deleted. Our results reveal an indispensable role for ROCK, yet redundant role for isoforms 1 and 2, in cell cycle progression and tumorigenesis, possibly through the maintenance of cellular contractility.


Carcinogenesis , Cell Proliferation , rho-Associated Kinases/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Gene Knockout Techniques , Melanoma/pathology , Mice , rho-Associated Kinases/genetics
20.
Dev Cell ; 35(3): 344-57, 2015 Nov 09.
Article En | MEDLINE | ID: mdl-26555054

Polarization of cells into a protrusive front and a retracting cell body is the hallmark of mesenchymal-like cell migration. Many mRNAs are localized to protrusions, but it is unclear to what degree mRNA localization contributes toward protrusion formation. We performed global quantitative analysis of the distributions of mRNAs, proteins, and translation rates between protrusions and the cell body by RNA sequencing (RNA-seq) and quantitative proteomics. Our results reveal local translation as a key determinant of protein localization to protrusions. Accordingly, inhibition of local translation destabilizes protrusions and inhibits mesenchymal-like morphology. Interestingly, many mRNAs localized to protrusions are translationally repressed. Specific cis-regulatory elements within mRNA UTRs define whether mRNAs are locally translated or repressed. Finally, RNAi screening of RNA-binding proteins (RBPs) enriched in protrusions revealed trans-regulators of localized translation that are functionally important for protrusions. We propose that by deciphering the localized mRNA UTR code, these proteins regulate protrusion stability and mesenchymal-like morphology.


Cell Movement/genetics , Cell Surface Extensions/metabolism , Protein Biosynthesis/genetics , RNA, Messenger/genetics , Protein Binding/genetics , Protein Transport , RNA-Binding Proteins/genetics , Sequence Analysis, RNA/methods
...