Supplementary Figure S1

Total Page:16

File Type:pdf, Size:1020Kb

Supplementary Figure S1 Supplementary Figure S1 A 40 20 20 0 TCGA 0 −20 PETACC-3 Agendia (GSE42284) −40 Agendia (ICO) −20 Agendia (VHB) Principal component 2 −60 Principal component 4 Marisa et al. (GSE39582) Schlicker et al. (GSE35896) −80 −40 GSE75316 GSE75315 −60 −40 −20 0 20 40 60 80 −40 −20 0 20 40 Principal component 1 Principal component 3 B 4 3 2 1 0 supplementary Figure S1. Clustering of colorectal cancer BRAF V600E mutant patients. A: Principal component analysis of BRAF mutant patients. The four first principal components which explain the most of data variation are shown. Patients are labeled according to the cohort to which they belong. B: Hierarchical clustering of BRAF mutant patients using the Ward method to compute distance between patients. Supplementary Figure S2 A Row Z−Score - 4 - 2 0 2 4 BM1 BM2 200 150 100 50 BRAF V600E mutant patients 1000 2000 3000 4000 5000 6000 7000 8000 9000 .6 .4 .2 0 BM Subtype Genes B BM1 BM2 NMF clustering after 0 MSI status correction BM2 BM1 0.5 Unsupervised BM1 51 9 Cohen’s kappa = 0.82 Proportion NMF clustering [95%CI = 0.73-0.90] 6 125 BM subtypes BM2 MSI corrected 1 supplementary Figure S2. Subtyping of CRC BRAF mutant patients. A: Heatmap displaying genewise and patientwise double clustering of 218 CRC BRAF mutant patients. Clustering was performed using the Ward method to compute distance between genes and between patients. Gene expression is displayed as standardized z-score. B: NMF clustering after adjustment for MSI status, the BM subtype calls were then compared to the original calls computed without any adjustment. High agreement is shown between the two methods by the Cohen’s kappa coefficient of 0.82. Supplementary Figure S3 EMT signature 0.6 BM Subtype 0.5 0.4 423 0.3 Site 0 0 0 0.2 0 0 53 0 EMT 0 0 8 Enrichment score 0.1 0.0 147 0 0 0 0 0 0 0 0 0 0 0 0 0 0 BM1 BM2 1 0 0 0 48 Ranked 476 genes MSI status Gender KRAS signature 0.30 BM Subtype 0.25 0.20 448 0.15 Site 0 0 0.10 0 0 0 28 0 0.05 KRAS 0 0 8 Enrichment score 0.00 170 0 0 -0.05 0 0 0 0 0 0 0 2 0 0 0 0 1 0 0 0 BM1 BM2 46 Ranked 476 genes MSI status Gender E2F signature 0.0 BM Subtype -0.1 -0.2 -0.3 452 -0.4 Site 0 0 -0.5 0 0 0 -0.6 24 0 E2F 0 0 7 Enrichment score -0.7 -0.8 172 0 0 1 0 0 0 0 0 0 3 0 0 0 0 1 0 0 0 BM1 BM2 45 Ranked 476 genes MSI status Gender G2M signature 0.0 -0.1 BM Subtype -0.2 -0.3 454 -0.4 Site 0 0 -0.5 0 0 0 22 0 -0.6 G2M 0 0 7 Enrichment score -0.7 172 0 0 -0.8 1 0 0 0 0 0 0 4 0 1 0 0 0 0 0 0 BM1 BM2 48 Ranked 476 genes MSI status Gender supplementary Figure S3. Gene Set Enrichment Analysis of BRAF mutant patients. The 476 BRAF mutant specific genes were used to perform a standard GSEA analysis between BM1 and BM2. EMT (MSigDB ref: M5930) and KRAS (MSigDB ref: M5953) signatures were found enriched in BM1 while G2M (MSigDB ref: M5901) and E2F (MSigDB ref: M5925) signatures were enriched in BM2. Venn diagrams on the right side highlight the number of genes that are shared or specific between the respective signatures and the indicated clinical factors. Supplementary Figure S4 BM1 BRAF/KRAS WT (WT2) Positive signatures BM2 KRAS mutant BM subtypes Vs WT2 Negative signatures COMPLEMENT KRAS SIGNALING COAGULATION EPITHELIAL MESENCHYMAL TRANSITION w Z−Score INFLAMMATORY RESPONSE o R UV RESPONSE 3 APICAL JUNCTION 2 1 MYOGENESIS 0 TNFA SIGNALING VIA NFKB −1 GLYCOLYSIS −2 E2F TARGETS −3 G2M CHECKPOINT MITOTIC SPINDLE ESTROGEN RESPONSE LATE G2M CHECKPOINT MTORC1 SIGNALING MITOTIC SPINDLE E2F TARGETS ALLOGRAFT REJECTION GLYCOLYSIS IL2 STAT5 SIGNALING MYOGENESIS EPITHELIAL MESENCHYMAL TRANSITION APICAL JUNCTION ESTROGEN RESPONSE EARLY XENOBIOTIC METABOLISM COAGULATION UV RESPONSE APOPTOSIS BM subtypes Vs KRAS mutant COMPLEMENT INFLAMMATORY RESPONSE KRAS SIGNALING EPITHELIAL MESENCHYMAL TRANSITION COAGULATION UV RESPONSE MYOGENESIS GLYCOLYSIS E2F TARGETS ESTROGEN RESPONSE LATE MITOTIC SPINDLE G2M CHECKPOINT G2M CHECKPOINT MITOTIC SPINDLE E2F TARGETS MTORC1 SIGNALING ALLOGRAFT REJECTION GLYCOLYSIS MYOGENESIS ESTROGEN RESPONSE EARLY APICAL JUNCTION EPITHELIAL MESENCHYMAL TRANSITION APOPTOSIS XENOBIOTIC METABOLISM supplementary Figure S4. Pathway analysis comparison between BM subtypes, KRAS/BRAF wild-type and KRAS mutant CRCs. Single sample gene set enrichment analysis (ssGSEA) was performed for BM1 versus KRAS/BRAF wild-type (WT2) patients (top panel, top heatmap, 69 patients per group) or versus KRAS mutant patients (bottom panel, top heatmap, 69 patients per group) and similarly for BM2 versus WT2 patients (top panel, bottom heatmap, 149 patients per group) or versus KRAS mutant patients (bottom panel, bottom heatmap, 149 patients per group). These analyses were performed using the hallmark gene signature collection from MSigDB and using the only the 476 BM-specific genes. Supplementary Figure S5 Signature scores FDR Row scaled average signature score -1 -0.5 0 0.5 1 BM1 WT2 KRAS mut BM2 All genes 476 BM-specific genes MITOTIC SPINDLE MYC TARGETS CHOLESTEROL HOMEOSTASIS Enriched OXIDATIVE PHOSPHORYLATION in UV RESPONSE (DOWN) GLYCOLYSIS BM2 UNFOLDED PROTEIN RESPONSE 40 E2F TARGETS MTORC1 SIGNALING 30 DNA REPAIR G2M CHECKPOINT 20 BILE ACID METABOLISM PEROXISOME 10 MYC TARGETS FATTY ACID METABOLISM 0 PI3K/AKT/MTOR SIGNALING -log10(FDR) 10 SPERMATOGENESIS PROTEIN SECRETION 20 UV RESPONSE (UP) HYPOXIA Enriched APICAL_SURFACE in IL2/STAT5 SIGNALING TNFA SIGNALING VIA NFKB BM1 APOPTOSIS P53 PATHWAY COMPLEMENT INFLAMMATORY RESPONSE KRAS SIGNALING IL6/JAK/STAT3 SIGNALING ALLOGRAFT REJECTION INTERFERON ALPHA RESPONSE INTERFERON GAMMA RESPONSE PANCREAS BETA CELLS REACTIVE OXIGEN SPECIES PATHWAY HEME METABOLISM XENOBIOTIC METABOLISM WNT/BETA-CATENIN SIGNALING ESTROGENE RESPONSE LATE ANDROGEN RESPONSE LATE ANGIOGENESIS COAGULATION EPITHELIAL MESENCHYMAL TRANSITION KRAS SIGNALING (UP) TGF-B SIGNALING MYOGENESIS UV RESPONSE (DOWN) APICAL JUNCTION NOTCH SIGNALING HEDGEHOG SIGNALING ESTROGEN RESPONSE EARLY ADIPOGENESIS KRAS SIGNALING (DOWN) supplementary Figure S5. Molecular characterization of BM subtypes. Heatmap displaying the average ssGSEA hallmark signature scores for BM1, BM2, KRAS/BRAF wild-type (WT2) and KRAS mutant using all gene profiles (compared to Figure 3 where only the 476 BM-specific genes were used). False discovery rates adjusted p-values (FDR) for BM1 versus BM2 statistical analyses appear on the right under the form of a heatmap for both all genes and for the 476 BM-specific genes. Supplementary Figure S6 M2882 M1417 44 182 M5930 M5953 M11513 M2878 26 1 8 1 42 6 0 0 1 1 0 15 163 0 132 130 0 0 0 2 0 0 1 2 19 0 7 4 0 0 0 0 0 0 4 0 0 0 0 3 0 6 1 0 0 0 0 0 0 0 5 0 5 0 1 1 170 25 194 79 EMT signatures KRAS signatures M8191 M5956 M10573 M5915 APICAL JUNCTION M12829 43 M1484 M1396 M2632 M5925 4 20 M8627 151 116 M5901 0 4 1 3 65 8 136 0 2 9 0 60 1 11 0 0 37 1 0 0 12 113 0 0 12 le signatures 12 0 0 0 0 c 1 0 y 11 0 c 10 339 3 9 1 1 4 Cell M543 M14894 MTORC1 HALLMARK M7561 M16909 M5924 46 535 M1919 1 3 15 176 1 1 329 0 11 0 mTOR-related signatures 1 0 10 supplementary Figure S6. MSigDB gene set overlap. Venn diagram showing the number of genes that are overlap between collections of signatures tested in Figure 3. Supplementary Figure S7 100 Neutrophils Eosinophils Mast cells resting Dendritic cells activated Dendritic cells resting 75 Macrophages M2 Macrophages M1 Macrophages M0 Monocytes NK cells activated 50 NK cells resting T cells gamma delta T cells regulatory (Tregs) T cells follicular helper Relative percentage T cells CD4 memory activated 25 T cells CD4 memory resting T cells CD4 naive T cells CD8 Plasma cells B cells memory B cells naive 0 BM1 BM2 Mast cells activated Dendritic cells resting Macrophages M2 Macrophages M1 Monocytes Plasma cells 40 40 20 30 15 30 20 30 15 20 20 10 20 10 10 10 5 10 10 5 Relative percentage 0 0 0 0 0 0 FDR 5.1.10-3 3.5.10-2 2.8.10-6 3.8.10-2 5.5.10-3 5.5.10-3 Macrophage-associated genes 4 CYBB 2 CD163 CD84 0 MS4A4A GM2A row z-score −2 CD68 MARCO −4 APOE CHIT1 CHI3L1 BCAT1 Subtype FN1 RAI14 BM1 COLEC12 BM2 COL8A2 SCARB2 ATG7 MSI status CXCL5 MSS SCG5 GPC4 MSI ME1 Not available FDX1 supplementary Figure S7. Immune profiles of BM subtypes. A CIBERSORT analysis that reveals the immune landscape of BM subtypes in terms of relative percentage for each immune subset (top panel). The bottom panel displays the relative percentage of immune subsets that were found significantly enriched (FDR < 0.01) for each BM subtypes. Bottom heatmap displays the expression of macrophage-associated genes according to BM subtypes. MSI status is displayed as well. Supplementary Figure S8 β-value A TCGA patients 0 0.2 0.4 0.6 0.8 Illumina 450k 1 BRAF WT BRAF Mutant Methylated Not methylated Probes more methylated in BRAF mutant 15’591 / 374’801 (4.2 %) Probes more methylated in BRAF WT 196 / 374’801 (0.05 %) Differentially methylated Differentially probes (15’591) Probes not significantly different 359’014 / 374’801 (95.8 %) B TCGA patients Illumina 27k Illumina 450k BM1 BM2 MSS All DNA methylation probes All DNA in common between the two platforms (22’333) MSI C Illumina 27k Illumina 450k methylation probes MLH1 D Illumina 27k Illumina 450k SLC16A8 CLDN15 RPS6KA2 SPAG6 C16orf95 NGEF KLK8 TIMP2 METRN DMRT2 NGEF TBC1D13 PLCH2 PSD KIF17 AHRR RHOT1 FAM124B NLRP2 CCBP2 COL18A1 C9orf47 NCRNA00176 DNAJB6 MAPK15 MTERF QSOX2 PSTPIP1 MTERF MCAM PCDHB12 SSTR5 TSSC1 SLC27A6 RAMP1 TRIM29 CD4 SECTM1 HIST1H3H PSMG3 GRAMD2 HSD17B7P2 GDPD5 HLA−DPB2 HSD17B7 GRIN2A AGK LINC00982 CD40 NR5A2 FBXO34 Differentially methylated probes Differentially VPS18 GRB14 SOGA1 supplementary Figure S8.
Recommended publications
  • Systematic Screening for Potential Therapeutic Targets in Osteosarcoma Through a Kinome-Wide CRISPR-Cas9 Library
    Cancer Biol Med 2020. doi: 10.20892/j.issn.2095-3941.2020.0162 ORIGINAL ARTICLE Systematic screening for potential therapeutic targets in osteosarcoma through a kinome-wide CRISPR-Cas9 library Yuanzhong Wu*, Liwen Zhou*, Zifeng Wang, Xin Wang, Ruhua Zhang, Lisi Zheng, Tiebang Kang Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China ABSTRACT Objective: Osteosarcoma is the most common primary malignant bone tumor. However, the survival of patients with osteosarcoma has remained unchanged during the past 30 years, owing to a lack of efficient therapeutic targets. Methods: We constructed a kinome-targeting CRISPR-Cas9 library containing 507 kinases and 100 nontargeting controls and screened the potential kinase targets in osteosarcoma. The CRISPR screening sequencing data were analyzed with the Model-based Analysis of Genome-wide CRISPR/Cas9 Knockout (MAGeCK) Python package. The functional data were applied in the 143B cell line through lenti-CRISPR-mediated gene knockout. The clinical significance of kinases in the survival of patients with osteosarcoma was analyzed in the R2: Genomics Analysis and Visualization Platform. Results: We identified 53 potential kinase targets in osteosarcoma. Among these targets, we analyzed 3 kinases, TRRAP, PKMYT1, and TP53RK, to validate their oncogenic functions in osteosarcoma. PKMYT1 and TP53RK showed higher expression in osteosarcoma than in normal bone tissue, whereas TRRAP showed no significant difference. High expression of all 3 kinases was associated with relatively poor prognosis in patients with osteosarcoma. Conclusions: Our results not only offer potential therapeutic kinase targets in osteosarcoma but also provide a paradigm for functional genetic screening by using a CRISPR-Cas9 library, including target design, library construction, screening workflow, data analysis, and functional validation.
    [Show full text]
  • The Role of the Mtor Pathway in Developmental Reprogramming Of
    THE ROLE OF THE MTOR PATHWAY IN DEVELOPMENTAL REPROGRAMMING OF HEPATIC LIPID METABOLISM AND THE HEPATIC TRANSCRIPTOME AFTER EXPOSURE TO 2,2',4,4'- TETRABROMODIPHENYL ETHER (BDE-47) An Honors Thesis Presented By JOSEPH PAUL MCGAUNN Approved as to style and content by: ________________________________________________________** Alexander Suvorov 05/18/20 10:40 ** Chair ________________________________________________________** Laura V Danai 05/18/20 10:51 ** Committee Member ________________________________________________________** Scott C Garman 05/18/20 10:57 ** Honors Program Director ABSTRACT An emerging hypothesis links the epidemic of metabolic diseases, such as non-alcoholic fatty liver disease (NAFLD) and diabetes with chemical exposures during development. Evidence from our lab and others suggests that developmental exposure to environmentally prevalent flame-retardant BDE47 may permanently reprogram hepatic lipid metabolism, resulting in an NAFLD-like phenotype. Additionally, we have demonstrated that BDE-47 alters the activity of both mTOR complexes (mTORC1 and 2) in hepatocytes. The mTOR pathway integrates environmental information from different signaling pathways, and regulates key cellular functions such as lipid metabolism, innate immunity, and ribosome biogenesis. Thus, we hypothesized that the developmental effects of BDE-47 on liver lipid metabolism are mTOR-dependent. To assess this, we generated mice with liver-specific deletions of mTORC1 or mTORC2 and exposed these mice and their respective controls perinatally to
    [Show full text]
  • Autophagy Processes Are Dependent on EGF Receptor Signaling
    www.oncotarget.com Oncotarget, 2018, Vol. 9, (No. 54), pp: 30289-30303 Research Paper Autophagy processes are dependent on EGF receptor signaling Vincenzo De Iuliis1, Antonio Marino1, Marika Caruso1, Sabrina Capodifoglio1, Vincenzo Flati2, Anna Marynuk3, Valeria Marricareda3, Sebastiano Ursi1, Paola Lanuti4, Claudio Talora5, Pio Conti6, Stefano Martinotti1,7 and Elena Toniato7 1Unit of Predictive Medicine, SS Annunziata University Hospital of Chieti, Chieti, Italy 2Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy 3Odessa National Medical University, Odesa, Odessa Oblsat, Ucraina 4Department of Medicine and Aging Sciences, University G. d’Annunzio of Chieti, Chieti, Italy 5Department of Molecular Medicine, University of Rome “La Sapienza”, Rome, Italy 6Postgraduate Medical School, University of Chieti, Chieti, Italy 7Department of Medical, Oral and Biotechnological Sciences, University G. d’Annunzio of Chieti, Chieti, Italy Correspondence to: Elena Toniato, email: [email protected] Keywords: apoptosis; autophagy; Beclin 1; EGF receptor; MAPK pathway Received: May 03, 2018 Accepted: June 13, 2018 Published: July 13, 2018 Copyright: De Iuliis et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License 3.0 (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. ABSTRACT Autophagy is a not well-understood conserved mechanism activated during nutritional deprivation in order to maintain cellular homeostasis. In the present study, we investigated the correlations between autophagy, apoptosis and the MAPK pathways in melanoma cell lines. We demonstrated that during starvation the EGF receptor mediated signaling activates many proteins involved in the MAPK pathway.
    [Show full text]
  • Proteomic Landscape of Tissue-Specific Cyclin E Functions in Vivo
    RESEARCH ARTICLE Proteomic Landscape of Tissue-Specific Cyclin E Functions in Vivo Junko Odajima1,2, Siddharth Saini3, Piotr Jung1,2, Yasmine Ndassa-Colday1,4, Scott Ficaro1,4, Yan Geng1,2, Eugenio Marco5, Wojciech Michowski1,2, Yaoyu E. Wang6, James A. DeCaprio7, Larisa Litovchick3, Jarrod Marto1,4, Piotr Sicinski1,2* 1 Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America, 2 Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America, 3 Department of Internal Medicine and Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, United States of America, 4 Department of Biochemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States of America, 5 Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute and Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America, 6 Center for Cancer Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America, 7 Department of a11111 Medical Oncology, Dana-Farber Cancer Institute, and Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America * [email protected] Abstract OPEN ACCESS E-type cyclins (cyclins E1 and E2) are components of the cell cycle machinery that has Citation: Odajima J, Saini S, Jung P, Ndassa- Colday Y, Ficaro S, Geng Y, et al. (2016) Proteomic been conserved from yeast to humans. The major function of E-type cyclins is to drive cell Landscape of Tissue-Specific Cyclin E Functions in division. It is unknown whether in addition to their `core' cell cycle functions, E-type cyclins Vivo. PLoS Genet 12(11): e1006429.
    [Show full text]
  • Integrin-Linked Kinase Controls Renal Branching Morphogenesis Via Dual Specificity Phosphatase 8
    BASIC RESEARCH www.jasn.org Integrin-linked Kinase Controls Renal Branching Morphogenesis via Dual Specificity Phosphatase 8 †‡ Joanna Smeeton,* Priya Dhir,* Di Hu,* Meghan M. Feeney,* Lin Chen,* and † Norman D. Rosenblum* § *Program in Developmental and Stem Cell Biology, and §Division of Nephrology, The Hospital for Sick Children, Toronto, Ontario, Canada; and †Departments of Paediatrics, and ‡Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada ABSTRACT Integrin-linked kinase (ILK) is an intracellular scaffold protein with critical cell-specific functions in the embryonic and mature mammalian kidney. Previously, we demonstrated a requirement for Ilk during ureteric branching and cell cycle regulation in collecting duct cells in vivo. Although in vitro data indicate that ILK controls p38 mitogen-activated protein kinase (p38MAPK) activity, the contribution of ILK- p38MAPK signaling to branching morphogenesis in vivo is not defined. Here, we identified genes that are regulated by Ilk in ureteric cells using a whole-genome expression analysis of whole-kidney mRNA in mice with Ilk deficiency in the ureteric cell lineage. Six genes with expression in ureteric tip cells, including Wnt11, were downregulated, whereas the expression of dual-specificity phosphatase 8 (DUSP8) was upregulated. Phosphorylation of p38MAPK was decreased in kidney tissue with Ilk deficiency, but no significant decrease in the phosphorylation of other intracellular effectors previously shown to control renal morphogenesis was observed. Pharmacologic inhibition of p38MAPK activity in murine inner med- ullary collecting duct 3 (mIMCD3) cells decreased expression of Wnt11, Krt23,andSlo4c1.DUSP8over- expression in mIMCD3 cells significantly inhibited p38MAPK activation and the expression of Wnt11 and Slo4c1. Adenovirus-mediated overexpression of DUSP8 in cultured embryonic murine kidneys decreased ureteric branching and p38MAPK activation.
    [Show full text]
  • Ncounter® Human Autoimmune Profiling Panel
    nCounter® Human AutoImmune Profiling Panel - Gene and Probe Details Official Symbol Accession Alias / Previous Symbol Official Full Name Other targets or Isoform Information ACE NM_000789.2 DCP1;angiotensin I converting enzyme (peptidyl-dipeptidase A) 1 angiotensin I converting enzyme ACIN1 NM_001164815.1 ACINUS;apoptotic chromatin condensation inducer in the nucleus apoptotic chromatin condensation inducer 1 ACP5 NM_001611.3 acid phosphatase 5, tartrate resistant CTRN2;ARP1 (actin-related protein 1, yeast) homolog B (centractin beta),ARP1 actin-related ACTR1B NM_005735.3 protein 1 homolog B, centractin beta ARP1 actin related protein 1 homolog B ADAM17 NM_003183.4 TACE;tumor necrosis factor, alpha, converting enzyme ADAM metallopeptidase domain 17 ADAR NM_001111.3 IFI4,G1P1;interferon-induced protein 4 adenosine deaminase, RNA specific ADORA2A NM_000675.3 ADORA2 adenosine A2a receptor AGER NM_001136.3 advanced glycosylation end-product specific receptor AGT NM_000029.3 SERPINA8;serpin peptidase inhibitor, clade A, member 8 angiotensinogen AHR NM_001621.3 aryl hydrocarbon receptor AICDA NM_020661.2 activation-induced cytidine deaminase activation induced cytidine deaminase AIM2 NM_004833.1 absent in melanoma 2 APECED;autoimmune regulator (autoimmune polyendocrinopathy candidiasis ectodermal AIRE NM_000383.2 dystrophy) autoimmune regulator AKT1 NM_001014432.1 v-akt murine thymoma viral oncogene homolog 1 AKT serine/threonine kinase 1 AKT2 NM_001626.4 v-akt murine thymoma viral oncogene homolog 2 AKT serine/threonine kinase 2 AKT3 NM_005465.4
    [Show full text]
  • Autophagy Proteins Regulate ERK Phosphorylation
    ARTICLE Received 20 Aug 2013 | Accepted 22 Oct 2013 | Published 18 Nov 2013 DOI: 10.1038/ncomms3799 OPEN Autophagy proteins regulate ERK phosphorylation Nuria Martinez-Lopez1,2, Diana Athonvarangkul1,2, Priti Mishall3, Srabani Sahu1,2 & Rajat Singh1,2,4,5 Autophagy is a conserved pathway that maintains cellular quality control. Extracellular signal- regulated kinase (ERK) controls various aspects of cell physiology including proliferation. Multiple signalling cascades, including ERK, have been shown to regulate autophagy, however whether autophagy proteins (ATG) regulate cell signalling is unknown. Here we show that growth factor exposure increases the interaction of ERK cascade components with ATG proteins in the cytosol and nucleus. ERK and its upstream kinase MEK localize to the extra- luminal face of autophagosomes. ERK2 interacts with ATG proteins via its substrate-binding domains. Deleting Atg7 or Atg5 or blocking LC3 lipidation or ATG5–ATG12 conjugation decreases ERK phosphorylation. Conversely, increasing LC3-II availability by silencing the cysteine protease ATG4B or acute trehalose exposure increases ERK phosphorylation. Decreased ERK phosphorylation in Atg5 À / À cells does not occur from overactive phos- phatases. Our findings thus reveal an unconventional function of ATG proteins as cellular scaffolds in the regulation of ERK phosphorylation. 1 Department of Medicine, Albert Einstein College of Medicine, Bronx, New York 10461, USA. 2 Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York 10461, USA. 3 Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA. 4 Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, New York 10461, USA. 5 Diabetes Research Center, Albert Einstein College of Medicine, Bronx, New York 10461, USA.
    [Show full text]
  • Supplementary Table 5.List of the 220 Most Frequently Amplified Genes In
    Supplementary Table 5. List of the 220 most frequently amplified genes in this study. The table includes their chromosomal location, the amplification frequency in ER-positive female breast cancer with associated p-value for difference in proportions, the preference for surrogate intrinsic molecular subtype, and associations with clinical, pathological and genetic characteristics. Potentially druggable gene categories, clinical actionability and known drug interactions are indicated per gene. Gene Full name chr location % amp in FFPE % amps in FF total % amp % amp ER+ FBC* p-value MBC vs ER+ FBC** % in lumA-like % in lumB-like p-value BRCA2 germline Age Hist type ER status PR status HER2 status Grade MAI Size LN SNV load PIK3CA mut KM (OS)*** KM (5Y OS)*** druggable gene category# clinically actionable?## known drug interactions?### THBS1 thrombospondin 1 15q14 37% 9% 30% 0.1% <0.0001 23% 35% 0.128 ns ns ns ns ns ns ns ns ns ns ns ns 0.642 p=0.832 cell surface, tumor suppressor, drug resistance, external side of plasma membrane no none PRKDC protein kinase, DNA-activated, catalytic polypeptide 8q11.21 35% 7% 27% 10.9% <0.0001 26% 30% 0.595 ns ns ns ns ns ns ns ns ns ns ns ns 0.838 p=0.903 (serine threonine) kinase, druggable genome, PI3 kinase, tumor suppressor, TF complex, TF binding, DNA repair yes DNA-PK INHIBITOR V (DNA-PK inhibitor); WORTMANNIN (PI3K inhibitor); SF1126 (PI3 kinase/mTOR inhibitor) TBX3 T-box 3 12q24.21 34% 7% 27% 0.1% <0.0001 20% 35% 0.053 ns ns ns ns ns ns ns ns ns ns ns ns 0.439 p=0.264 tumor suppressor, TF binding
    [Show full text]
  • Impaired SNF2L Chromatin Remodeling Prolongs Accessibility at Promoters Enriched for Fos/Jun Binding Sites and Delays Granule Neuron Differentiation
    fnmol-14-680280 June 30, 2021 Time: 16:59 # 1 ORIGINAL RESEARCH published: 06 July 2021 doi: 10.3389/fnmol.2021.680280 Impaired SNF2L Chromatin Remodeling Prolongs Accessibility at Promoters Enriched for Fos/Jun Binding Sites and Delays Granule Neuron Differentiation Laura R. Goodwin1,2, Gerardo Zapata1,2, Sara Timpano1, Jacob Marenger1 and David J. Picketts1,2,3* 1 Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada, 2 Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada, 3 Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada Chromatin remodeling proteins utilize the energy from ATP hydrolysis to mobilize Edited by: nucleosomes often creating accessibility for transcription factors within gene regulatory Veronica Martinez Cerdeño, University of California, Davis, elements. Aberrant chromatin remodeling has diverse effects on neuroprogenitor United States homeostasis altering progenitor competence, proliferation, survival, or cell fate. Previous Reviewed by: work has shown that inactivation of the ISWI genes, Smarca5 (encoding Snf2h) and Mitsuhiro Hashimoto, Fukushima Medical University, Japan Smarca1 (encoding Snf2l) have dramatic effects on brain development. Smarca5 Koji Shibasaki, conditional knockout mice have reduced progenitor expansion and severe forebrain Nagasaki University, Japan hypoplasia, with a similar effect on the postnatal growth of the cerebellum. In contrast, *Correspondence: Smarca1 mutants exhibited enlarged forebrains
    [Show full text]
  • Extracellular-Signal Regulated Kinase: a Central Molecule Driving Epithelial-Mesenchymal Transition in Cancer
    University of Massachusetts Medical School eScholarship@UMMS Open Access Articles Open Access Publications by UMMS Authors 2019-06-13 Extracellular-Signal Regulated Kinase: A Central Molecule Driving Epithelial-Mesenchymal Transition in Cancer Monserrat Olea-Flores Autonomous University of Guerrero Et al. Let us know how access to this document benefits ou.y Follow this and additional works at: https://escholarship.umassmed.edu/oapubs Part of the Amino Acids, Peptides, and Proteins Commons, Cancer Biology Commons, Cell Biology Commons, Enzymes and Coenzymes Commons, and the Molecular Biology Commons Repository Citation Olea-Flores M, Zuniga-Eulogio MD, Mendoza-Catalan MA, Rodriguez-Ruiz HA, Castaneda-Saucedo E, Ortuno-Pineda C, Padilla-Benavides T, Navarro-Tito N. (2019). Extracellular-Signal Regulated Kinase: A Central Molecule Driving Epithelial-Mesenchymal Transition in Cancer. Open Access Articles. https://doi.org/10.3390/ijms20122885. Retrieved from https://escholarship.umassmed.edu/oapubs/ 3883 Creative Commons License This work is licensed under a Creative Commons Attribution 4.0 License. This material is brought to you by eScholarship@UMMS. It has been accepted for inclusion in Open Access Articles by an authorized administrator of eScholarship@UMMS. For more information, please contact [email protected]. International Journal of Molecular Sciences Review Extracellular-Signal Regulated Kinase: A Central Molecule Driving Epithelial–Mesenchymal Transition in Cancer Monserrat Olea-Flores 1 , Miriam Daniela Zuñiga-Eulogio 1 , Miguel Angel Mendoza-Catalán 2 , Hugo Alberto Rodríguez-Ruiz 2, Eduardo Castañeda-Saucedo 1, Carlos Ortuño-Pineda 2, Teresita Padilla-Benavides 3,* and Napoleón Navarro-Tito 1,* 1 Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av.
    [Show full text]
  • ERK8 Is a Negative Regulator of O-Galnac Glycosylation and Cell Migration
    RESEARCH ARTICLE elife.elifesciences.org ERK8 is a negative regulator of O-GalNAc glycosylation and cell migration Joanne Chia1, Keit Min Tham1, David James Gill1, Emilie Anne Bard-Chapeau1†, Frederic A Bard1,2* 1Institute of Molecular and Cell Biology, Singapore, Singapore; 2Department of Biochemistry, National University of Singapore, Singapore, Singapore Abstract ER O-glycosylation can be induced through relocalisation GalNAc-Transferases from the Golgi. This process markedly stimulates cell migration and is constitutively activated in more than 60% of breast carcinomas. How this activation is achieved remains unclear. Here, we screened 948 signalling genes using RNAi and imaging. We identified 12 negative regulators of O-glycosylation that all control GalNAc-T sub-cellular localisation. ERK8, an atypical MAPK with high basal kinase activity, is a strong hit and is partially localised at the Golgi. Its inhibition induces the relocation of GalNAc-Ts, but not of KDEL receptors, revealing the existence of two separate COPI-dependent pathways. ERK8 down-regulation, in turn, activates cell motility. In human breast and lung carcinomas, ERK8 expression is reduced while ER O-glycosylation initiation is hyperactivated. In sum, ERK8 appears as a constitutive brake on GalNAc-T relocalisation, and the loss of its expression could drive cancer aggressivity through increased cell motility. DOI: 10.7554/eLife.01828.001 *For correspondence: Introduction [email protected] GalNAc-type O-linked glycans are polysaccharides present on secreted and membrane-inserted pro- †Present address: Novartis teins (Tran and Ten Hagen, 2013). Traditionally associated with mucin-like proteins, recent advances in Institutes for BioMedical mass spectrometric analysis have revealed O-glycosylation on hundreds of different proteins (Steentoft Research, Basel, Switzerland et al., 2013).
    [Show full text]
  • Nanostring®: Product Data Sheet | Ncounter® GX Human Kinase
    PRODUCT DATA SHEET nCounter® GX Human Kinase Kit nCounter® GX Human Kinase Kit Product Highlights Highly Curated • Our expert bio-informaticists use a very rigorous process in selecting the most meaningful set of genes Efficient • Multiplexed assay profiles 522 human kinase genes in a single reaction Cost-effective • Gold-standard data at a fraction of the cost Quick Turnaround Time • Complete kit with all consumables ready to ship next day nCounter® GX Human Kinase Kit The nCounter GX Human Kinase Kit is a comprehensive list of 522 human The nCounter Human Kinase Kit represents 99% of the KinBase content for genes known to be differentially expressed in the kinome. Human. With the nCounter GX Human Kinase Kit, scientists can leverage a pre-designed The final nCounter GX Human Kinase Kit consists of 522 protein kinase- panel to accelerate their research and quickly generate expression data for a related genes and 14 internal reference genes. For the gene list and additional large panel of protein kinase-related genes. information about this gene set, visit the nCounter Pre-built Panels product page at: www.nanostring.com. The gene list was compiled using the KinBase database at www.kinase.com. Home > Products > nCounter Gene Expression CodeSets > Pre-built Panels The database is based on the publication, The Protein Kinase Complement of the Human Genome, by G Manning, DB Whyte, R Martinez, T Hunter, S Sudarsanam (2002). Science 298:1912-1934. Molecules That Count® Translational Research Gene Expression miRNA Expression Copy Number Variation 1 PRODUCT DATA SHEET nCounter® GX Human Kinase Kit nCounter® Analysis System Overview The nCounter Analysis System from NanoString offers a cost-effective way to easily profile hundreds of gene transcripts simultaneously with high sensitivity and precision.
    [Show full text]