Androgen-Induced Programs for Prostate Epithelial Growth and Invasion Arise in Embryogenesis and Are Reactivated in Cancer

Total Page:16

File Type:pdf, Size:1020Kb

Androgen-Induced Programs for Prostate Epithelial Growth and Invasion Arise in Embryogenesis and Are Reactivated in Cancer Oncogene (2008) 27, 7180–7191 & 2008 Macmillan Publishers Limited All rights reserved 0950-9232/08 $32.00 www.nature.com/onc ORIGINAL ARTICLE Androgen-induced programs for prostate epithelial growth and invasion arise in embryogenesis and are reactivated in cancer EM Schaeffer1,2,3,5, L Marchionni3,5, Z Huang1,2, B Simons1, A Blackman3,WYu3, G Parmigiani1,3,4 and DM Berman1,2,3 1Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; 2Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; 3Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA and 4Department of Biostatistics, Johns Hopkins University School of Medicine, Baltimore, MD, USA Cancer cells differentiate along specific lineages that most significant links to the development and cancer, we largelydetermine their clinical and biologic behavior. highlight coordinate induction of the transcription factor Distinct cancer phenotypes from different cells and organs Sox9 and suppression of the proapoptotic phospholipid- likelyresult from unique gene expression repertoires binding protein Annexin A1 that link earlyprostate established in the embryo and maintained after malignant development to earlyprostate carcinogenesis. These transformation. We used comprehensive gene expression results credential earlyprostate development as a reliable analysis to examine this concept in the prostate, an organ and valid model system for the investigation of genes and with a tractable developmental program and a high pathways that drive prostate cancer. propensityfor cancer. We focused on gene expression in Oncogene (2008) 27, 7180–7191; doi:10.1038/onc.2008.327; the murine prostate rudiment at three time points during published online 15 September 2008 the first 48 h of exposure to androgen, which initiates proliferation and invasion of prostate epithelial buds into Keywords: prostate organogenesis; androgen signaling; surrounding urogenital sinus mesenchyme. Here, we show prostate cancer; microarray that androgen exposure regulates genes previouslyim- plicated in prostate carcinogenesis comprising pathways for the phosphatase and tensin homolog (PTEN), fibroblast growth factor (FGF)/mitogen-activated protein kinase (MAPK), and Wnt signaling along with cellular Introduction programs regulating such ‘hallmarks’ of cancer as angiogenesis, apoptosis, migration and proliferation. We The discovery of proto-oncogenes in the 1970’s (Stehelin found statisticallysignificant evidence for novel androgen- et al., 1976) and tumor suppressor genes a decade later induced gene regulation events that establish and/or (Friend et al., 1986) launched an effort to identify a maintain prostate cell fate. These include modulation of common genetic basis for all cancers. This approach has gene expression through microRNAs, expression of yielded significant insights into common molecular specific transcription factors, and regulation of their machinery regulating tumor initiation and growth predicted targets. Byquerying public gene expression (Hanahan and Weinberg, 2000), but fails to account databases from other tissues, we found that rather than for dramatic differences in the behaviors of tumors generallycharacterizing androgen exposure or epithelial arising in different sites. As revealed by recent genomic budding, the earlyprostate development program more approaches, the site of origin is the dominant influence closelyresembles the program for human prostate cancer. on gene expression in cancers (Ramaswamy et al., 2001) Most importantly, early androgen-regulated genes and and specifies particular oncogenic mutations (Garraway functional themes associated with prostate development and Sellers, 2006). Tissue-specific behaviors of cancers were highlyenriched in contrasts between increasingly almost certainly reflect lineage-specific epigenetic pro- lethal forms of prostate cancer, confirming a ‘reactivation’ grams that operate during embryogenesis in cells and of embryonic pathways for proliferation and invasion in tissues from which cancers arise. Indeed, reawakening of prostate cancer progression. Among the genes with the embryonic programs has long been posited for cancer (Bailey and Cushing, 1925), and could underlie tissue Correspondence: Associate Professor DM Berman, Department of specific modes of regulating critical aspects of the Pathology, Johns Hopkins University School of Medicine and Sidney malignant phenotype such as survival, angiogenesis, Kimmel Cancer Center, CRB2 Room 545, 1550 Orleans street, invasion and migration. The advent of comprehensive Baltimore, MD, 21231 USA. genomic profiling techniques now permits this hypo- E-mail: [email protected] thesis to be tested, and to link cancer to embryogenesis 5These authors contributed equally to this work. Received 31 March 2008; revised 18 July 2008; accepted 1 August 2008; through cellular pathways that define specific lineages published online 15 September 2008 and organs (Garraway and Sellers, 2006). Identification Embryonic gene expression in prostate cancer EM Schaeffer et al 7181 of these pathways and their functions should identify urogenital sinus (UGS), an embryonic rudiment present targets for more specific and less toxic cancer therapies. in both sexes. These interactions lead to outgrowth of The prostate gland represents a prime target for such buds from the UGS epithelium (UGE) that proliferate an analysis as the genetic causes of prostate cancer are and invade surrounding urogenital sinus mesenchyme. poorly understood and because there are excellent AR expression has been described in both UGE and models of prostate development and homeostasis. While urogenital sinus mesenchyme (Drews et al., 2001; B other cancers have canonical cancer initiating mutations Simons, EM Schaeffer and DM Berman, unpublished (for example, K-ras for pancreatic carcinoma and observations), although epithelial AR is dispensable for Adenomatosis Polyposis Coli for colon carcinoma), induction of prostate development (Cunha et al., 2004). the driving force for prostate cancer initiation in humans remains uncertain. Answers may lie in cellular programs activated by androgen receptor (AR) signal- Results ing, which strictly controls prostate epithelial cell fate in embryogenesis and in cancer. Hormonal manipulation of mouse prostate development Upon binding androgen, AR signals through genomic In mouse, epithelial expression of the androgen-sensitive and nongenomic modes (Manolagas et al., 2002). homeobox family member Nkx3. 1 begins by embryonic Genomic responses entail nuclear translocation of day 16 (e16), and is the earliest reported marker of liganded AR and activation of transcription at regula- prostate development (Bhatia-Gaur et al., 1999). We tory regions containing AR-binding sites. Nongenomic comprehensively profiled gene expression in the UGS signaling, in contrast, comprises protein–protein inter- between e16 and e17.5, when prostate buds first emerge. actions in the cytoplasm and is exceedingly rapid, with The developmental fate of the UGS is bipotential in measurable responses within minutes of androgen both sexes and depends soley on androgens to drive exposure (Kousteni et al., 2001). AR signaling, through prostate formation in males (Jost, 1968). The absence of genomic and/or nongenomic routes, is necessary and androgens leads to vaginal/urethral formation in sufficient for prostate organogenesis. In response to females (Wilson et al., 1980). Taking advantage of this circulating testosterone and its local conversion to the dynamic, we induced prostate development in the more potent androgen, dihydrotestosterone, AR signal- androgen-naı¨ ve yet androgen-sensitive female UGS with ing induces epithelial-mesenchymal interactions in the precisely timed 6 and 12 h intrauterine exposure 15.5–16 d.p.c. 17.5 d.p.c. Mouse public domain Molecular Histologic Prostate degeneration/regeneration (GSE5901; Wang et al., 2007) Mesenchyme Epithelium Lung development (GSE1423; Lu et al., 2004) UGS Androgen-dependent gene expression in salivary gland (GSE3995: Treister et al., 2006) Androgen 48 h Endogenous (testis) Human public domain data Macrodissected prostate cancer profiling (Lapointe et al., 2004) Exogenous (injection) 12 h Androgen 6 h LCM prostate cancer profiling (GSE6099: Tomlins et al., 2007) Androgen start Differential gene expression analysis * Gene expression measurements: preprocessing, quality control evaluation * Statistical analysis of gene expression (Linear model analysis and empirical Bayes approach) * Gene cross-referencing with Entrez gene and homologene identifiers for cross platform and organismal analysis Analysis of functional annotation (AFA) * Enrichment evaluation using: Gene Ontology (GO), KEGG pathways and TFBS targets in prostate development and human cancer progression by Wilcoxon rank-sum test * Enrichment evaluation for mouse expression signatures in human prostate tumors by Wilcoxon rank-sum test Figure 1 Flowchart of data acquisition and analysis. (a) Schematic of early prostate development. The embryonic prostate rudiment, the urogenital sinus (UGS). Mesenchyme (light blue) surrounds epithelium (darker green). In the mouse, prostate-specific gene expression begins by embryonic day 16 (e16) followed by prostate epithelial budding at e17.5. Prostate development proceeds spontaneously in males in response to endogenous androgens or can be engineered in females in response to exogenous androgens. We comprehensively profiled androgen-induced gene
Recommended publications
  • Structural and Functional Properties of Platelet-Derived Growth Factor and Stem Cell Factor Receptors
    Downloaded from http://cshperspectives.cshlp.org/ on September 28, 2021 - Published by Cold Spring Harbor Laboratory Press Structural and Functional Properties of Platelet-Derived Growth Factor and Stem Cell Factor Receptors Carl-Henrik Heldin and Johan Lennartsson Ludwig Institute for Cancer Research, Uppsala University, SE-751 24 Uppsala, Sweden Correspondence: [email protected] The receptors for platelet-derived growth factor (PDGF) and stem cell factor (SCF) are members of the type III class of PTK receptors, which are characterized by five Ig-like domains extracellularly and a split kinase domain intracellularly. The receptors are activated by ligand-induced dimerization, leading to autophosphorylation on specific tyrosine resi- dues. Thereby the kinase activities of the receptors are activated and docking sites for down- stream SH2 domain signal transduction molecules are created; activation of these pathways promotes cell growth, survival, and migration. These receptors mediate important signals during the embryonal development, and control tissue homeostasis in the adult. Their over- activity is seen in malignancies and other diseases involving excessive cell proliferation, such as atherosclerosis and fibrotic diseases. In cancer, mutations of PDGF and SCF receptors— including gene fusions, point mutations, and amplifications—drive subpopulations of cer- tain malignancies, such as gastrointestinal stromal tumors, chronic myelomonocytic leuke- mia, hypereosinophilic syndrome, glioblastoma, acute myeloid leukemia, mastocytosis, and melanoma. he type III tyrosine kinase receptor family with kinases, and a less well-conserved carboxy- Tconsists of platelet-derived growth factor terminal tail. The ligands for these receptors are (PDGF) receptor a and b, stem cell factor all dimeric molecules, and on binding they in- (SCF) receptor (Kit), colony-stimulating fac- duce receptor dimerization.
    [Show full text]
  • TRANSCRIPTIONAL REGULATION of Hur in RENAL STRESS
    TRANSCRIPTIONAL REGULATION OF HuR IN RENAL STRESS DISSERTATION Presented in Partial Fulfillment of the Requirements for the Degree Doctor of Philosophy in the Graduate School of The Ohio State University By Sudha Suman Govindaraju Graduate Program in Biochemistry The Ohio State University 2014 Dissertation Committee: Dr. Beth S. Lee, Ph.D., Advisor Dr. Kathleen Boris-Lawrie, Ph.D. Dr. Sissy M. Jhiang, Ph.D. Dr. Arthur R. Strauch, Ph.D Abstract HuR is a ubiquitously expressed RNA-binding protein that affects the post- transcriptional life of thousands of cellular mRNAs by regulating transcript stability and translation. HuR can post-transcriptionally regulate gene expression and modulate cellular responses to stress, differentiation, proliferation, apoptosis, senescence, inflammation, and the immune response. It is an important mediator of survival during cellular stress, but when inappropriately expressed, can promote oncogenic transformation. Not surprisingly, the expression of HuR itself is tightly regulated at multiple transcriptional and post-transcriptional levels. Previous studies demonstrated the existence of two alternate HuR transcripts that differ in their 5’ untranslated regions and have markedly different translatabilities. These forms were also found to be reciprocally expressed following cellular stress in kidney proximal tubule cell lines, and the shorter, more readily translatable variant was shown to be regulated by Smad 1/5/8 pathway and bone morphogenetic protein-7 (BMP-7) signaling. In this study, the factors that promote transcription of the longer alternate form were identified. NF-κB was shown to be important for expression of the long HuR mRNA, as was a newly identified region with potential for binding the Sp/KLF families of transcription factors.
    [Show full text]
  • In Vivo Studies Using the Classical Mouse Diversity Panel
    The Mouse Diversity Panel Predicts Clinical Drug Toxicity Risk Where Classical Models Fail Alison Harrill, Ph.D The Hamner-UNC Institute for Drug Safety Sciences 0 The Importance of Predicting Clinical Adverse Drug Reactions (ADR) Figure: Cath O’Driscoll Nature Publishing 2004 Risk ID PGx Testing 1 People Respond Differently to Drugs Pharmacogenetic Markers Identified by Genome-Wide Association Drug Adverse Drug Risk Allele Reaction (ADR) Abacavir Hypersensitivity HLA-B*5701 Flucloxacillin Hepatotoxicity Allopurinol Cutaneous ADR HLA-B*5801 Carbamazepine Stevens-Johnson HLA-B*1502 Syndrome Augmentin Hepatotoxicity DRB1*1501 Ximelagatran Hepatotoxicity DRB1*0701 Ticlopidine Hepatotoxicity HLA-A*3303 Average preclinical populations and human hepatocytes lack the diversity to detect incidence of adverse events that occur only in 1/10,000 people. Current Rodent Models of Risk Assessment The Challenge “At a time of extraordinary scientific progress, methods have hardly changed in several decades ([FDA] 2004)… Toxicologists face a major challenge in the twenty-first century. They need to embrace the new “omics” techniques and ensure that they are using the most appropriate animals if their discipline is to become a more effective tool in drug development.” -Dr. Michael Festing Quantitative geneticist Toxicol Pathol. 2010;38(5):681-90 Rodent Models as a Strategy for Hazard Characterization and Pharmacogenetics Genetically defined rodent models may provide ability to: 1. Improve preclinical prediction of drugs that carry a human safety risk 2.
    [Show full text]
  • Podocyte Specific Knockdown of Klf15 in Podocin-Cre Klf15flox/Flox Mice Was Confirmed
    SUPPLEMENTARY FIGURE LEGENDS Supplementary Figure 1: Podocyte specific knockdown of Klf15 in Podocin-Cre Klf15flox/flox mice was confirmed. (A) Primary glomerular epithelial cells (PGECs) were isolated from 12-week old Podocin-Cre Klf15flox/flox and Podocin-Cre Klf15+/+ mice and cultured at 37°C for 1 week. Real-time PCR was performed for Nephrin, Podocin, Synaptopodin, and Wt1 mRNA expression (n=6, ***p<0.001, Mann-Whitney test). (B) Real- time PCR was performed for Klf15 mRNA expression (n=6, *p<0.05, Mann-Whitney test). (C) Protein was also extracted and western blot analysis for Klf15 was performed. The representative blot of three independent experiments is shown in the top panel. The bottom panel shows the quantification of Klf15 by densitometry (n=3, *p<0.05, Mann-Whitney test). (D) Immunofluorescence staining for Klf15 and Wt1 was performed in 12-week old Podocin-Cre Klf15flox/flox and Podocin-Cre Klf15+/+ mice. Representative images from four mice in each group are shown in the left panel (X 20). Arrows show colocalization of Klf15 and Wt1. Arrowheads show a lack of colocalization. Asterisk demonstrates nonspecific Wt1 staining. “R” represents autofluorescence from RBCs. In the right panel, a total of 30 glomeruli were selected in each mouse and quantification of Klf15 staining in the podocytes was determined by the ratio of Klf15+ and Wt1+ cells to Wt1+ cells (n=6 mice, **p<0.01, unpaired t test). Supplementary Figure 2: LPS treated Podocin-Cre Klf15flox/flox mice exhibit a lack of recovery in proteinaceous casts and tubular dilatation after DEX administration.
    [Show full text]
  • TFEB Regulates Murine Liver Cell Fate During Development and Regeneration ✉ Nunzia Pastore 1,2 , Tuong Huynh1,2, Niculin J
    ARTICLE https://doi.org/10.1038/s41467-020-16300-x OPEN TFEB regulates murine liver cell fate during development and regeneration ✉ Nunzia Pastore 1,2 , Tuong Huynh1,2, Niculin J. Herz1,2, Alessia Calcagni’ 1,2, Tiemo J. Klisch1,2, Lorenzo Brunetti 3,4, Kangho Ho Kim5, Marco De Giorgi6, Ayrea Hurley6, Annamaria Carissimo7, Margherita Mutarelli7, Niya Aleksieva 8, Luca D’Orsi7, William R. Lagor6, David D. Moore 5, ✉ Carmine Settembre7,9, Milton J. Finegold10, Stuart J. Forbes8 & Andrea Ballabio 1,2,7,9 1234567890():,; It is well established that pluripotent stem cells in fetal and postnatal liver (LPCs) can differentiate into both hepatocytes and cholangiocytes. However, the signaling pathways implicated in the differentiation of LPCs are still incompletely understood. Transcription Factor EB (TFEB), a master regulator of lysosomal biogenesis and autophagy, is known to be involved in osteoblast and myeloid differentiation, but its role in lineage commitment in the liver has not been investigated. Here we show that during development and upon regen- eration TFEB drives the differentiation status of murine LPCs into the progenitor/cho- langiocyte lineage while inhibiting hepatocyte differentiation. Genetic interaction studies show that Sox9, a marker of precursor and biliary cells, is a direct transcriptional target of TFEB and a primary mediator of its effects on liver cell fate. In summary, our findings identify an unexplored pathway that controls liver cell lineage commitment and whose dysregulation may play a role in biliary cancer. 1 Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, TX 77030, USA. 2 Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA.
    [Show full text]
  • A Role for Histone Modification in the Mechanism of Action of Antidepressant and Stimulant Drugs: a Dissertation
    University of Massachusetts Medical School eScholarship@UMMS GSBS Dissertations and Theses Graduate School of Biomedical Sciences 2007-12-28 A Role for Histone Modification in the Mechanism of Action of Antidepressant and Stimulant Drugs: a Dissertation Frederick Albert Schroeder University of Massachusetts Medical School Let us know how access to this document benefits ou.y Follow this and additional works at: https://escholarship.umassmed.edu/gsbs_diss Part of the Amino Acids, Peptides, and Proteins Commons, Cells Commons, Enzymes and Coenzymes Commons, Genetic Phenomena Commons, Mental Disorders Commons, Nervous System Commons, and the Therapeutics Commons Repository Citation Schroeder FA. (2007). A Role for Histone Modification in the Mechanism of Action of Antidepressant and Stimulant Drugs: a Dissertation. GSBS Dissertations and Theses. https://doi.org/10.13028/7bk0-a687. Retrieved from https://escholarship.umassmed.edu/gsbs_diss/370 This material is brought to you by eScholarship@UMMS. It has been accepted for inclusion in GSBS Dissertations and Theses by an authorized administrator of eScholarship@UMMS. For more information, please contact [email protected]. A Dissertation Presented by Frederick Albert Schroeder Submitted to the Faculty of the University of Massachusetts Graduate School of Biomedical Sciences Worcester, Massachusetts, USA in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY December 28, 2007 Program in Neuroscience A Role for Histone Modification in the Mechanism of Action of Antidepressant and Stimulant Drugs A Dissertation Presented By Frederick Albert Schroeder Approved as to style and content by: _____________________________________ Alonzo Ross, Ph.D., Chair of Committee _____________________________________ Pradeep Bhide, Ph.D., Member of Committee _____________________________________ Craig L.
    [Show full text]
  • Plant Mitogen-Activated Protein Kinase Signaling Cascades Guillaume Tena*, Tsuneaki Asai†, Wan-Ling Chiu‡ and Jen Sheen§
    392 Plant mitogen-activated protein kinase signaling cascades Guillaume Tena*, Tsuneaki Asai†, Wan-Ling Chiu‡ and Jen Sheen§ Mitogen-activated protein kinase (MAPK) cascades have components that link sensors/receptors to target genes emerged as a universal signal transduction mechanism that and other cellular responses. connects diverse receptors/sensors to cellular and nuclear responses in eukaryotes. Recent studies in plants indicate that In the past few years, it has become apparent that mitogen- MAPK cascades are vital to fundamental physiological functions activated protein kinase (MAPK) cascades play some of the involved in hormonal responses, cell cycle regulation, abiotic most essential roles in plant signal transduction pathways stress signaling, and defense mechanisms. New findings have from cell division to cell death (Figure 1). MAPK cascades revealed the complexity and redundancy of the signaling are evolutionarily conserved signaling modules with essen- components, the antagonistic nature of distinct pathways, and tial regulatory functions in eukaryotes, including yeasts, the use of both positive and negative regulatory mechanisms. worms, flies, frogs, mammals and plants. The recent enthu- siasm for plant MAPK cascades is backed by numerous Addresses studies showing that plant MAPKs are activated by hor- Department of Molecular Biology, Massachusetts General Hospital, mones, abiotic stresses, pathogens and pathogen-derived Department of Genetics, Harvard Medical School, Wellman 11, elicitors, and are also activated at specific stages during the 50 Blossom Street, Boston, Massachusetts 02114, USA cell cycle [2]. Until recently, studies of MAPK cascades in *e-mail: [email protected] †e-mail: [email protected] plants were focused on cDNA cloning [3,4] and used a ‡e-mail: [email protected] MAPK in-gel assay, MAPK and tyrosine-phosphate anti- §e-mail: [email protected] bodies, and kinase inhibitors to connect signals to MAPKs Current Opinion in Plant Biology 2001, 4:392–400 [2].
    [Show full text]
  • Whole Exome Sequencing in ADHD Trios from Single and Multi-Incident Families Implicates New Candidate Genes and Highlights Polygenic Transmission
    European Journal of Human Genetics (2020) 28:1098–1110 https://doi.org/10.1038/s41431-020-0619-7 ARTICLE Whole exome sequencing in ADHD trios from single and multi-incident families implicates new candidate genes and highlights polygenic transmission 1,2 1 1,2 1,3 1 Bashayer R. Al-Mubarak ● Aisha Omar ● Batoul Baz ● Basma Al-Abdulaziz ● Amna I. Magrashi ● 1,2 2 2,4 2,4,5 6 Eman Al-Yemni ● Amjad Jabaan ● Dorota Monies ● Mohamed Abouelhoda ● Dejene Abebe ● 7 1,2 Mohammad Ghaziuddin ● Nada A. Al-Tassan Received: 26 September 2019 / Revised: 26 February 2020 / Accepted: 10 March 2020 / Published online: 1 April 2020 © The Author(s) 2020. This article is published with open access Abstract Several types of genetic alterations occurring at numerous loci have been described in attention deficit hyperactivity disorder (ADHD). However, the role of rare single nucleotide variants (SNVs) remains under investigated. Here, we sought to identify rare SNVs with predicted deleterious effect that may contribute to ADHD risk. We chose to study ADHD families (including multi-incident) from a population with a high rate of consanguinity in which genetic risk factors tend to 1234567890();,: 1234567890();,: accumulate and therefore increasing the chance of detecting risk alleles. We employed whole exome sequencing (WES) to interrogate the entire coding region of 16 trios with ADHD. We also performed enrichment analysis on our final list of genes to identify the overrepresented biological processes. A total of 32 rare variants with predicted damaging effect were identified in 31 genes. At least two variants were detected per proband, most of which were not exclusive to the affected individuals.
    [Show full text]
  • Figure S1. Representative Report Generated by the Ion Torrent System Server for Each of the KCC71 Panel Analysis and Pcafusion Analysis
    Figure S1. Representative report generated by the Ion Torrent system server for each of the KCC71 panel analysis and PCaFusion analysis. (A) Details of the run summary report followed by the alignment summary report for the KCC71 panel analysis sequencing. (B) Details of the run summary report for the PCaFusion panel analysis. A Figure S1. Continued. Representative report generated by the Ion Torrent system server for each of the KCC71 panel analysis and PCaFusion analysis. (A) Details of the run summary report followed by the alignment summary report for the KCC71 panel analysis sequencing. (B) Details of the run summary report for the PCaFusion panel analysis. B Figure S2. Comparative analysis of the variant frequency found by the KCC71 panel and calculated from publicly available cBioPortal datasets. For each of the 71 genes in the KCC71 panel, the frequency of variants was calculated as the variant number found in the examined cases. Datasets marked with different colors and sample numbers of prostate cancer are presented in the upper right. *Significantly high in the present study. Figure S3. Seven subnetworks extracted from each of seven public prostate cancer gene networks in TCNG (Table SVI). Blue dots represent genes that include initial seed genes (parent nodes), and parent‑child and child‑grandchild genes in the network. Graphical representation of node‑to‑node associations and subnetwork structures that differed among and were unique to each of the seven subnetworks. TCNG, The Cancer Network Galaxy. Figure S4. REVIGO tree map showing the predicted biological processes of prostate cancer in the Japanese. Each rectangle represents a biological function in terms of a Gene Ontology (GO) term, with the size adjusted to represent the P‑value of the GO term in the underlying GO term database.
    [Show full text]
  • Supplemental Information IRF4 Transcription Factor-Dependent Cd11b+ Dendritic Cells in Human and Mouse Control Mucosal IL-17 C
    Immunity, Volume 38 Supplemental Information IRF4 Transcription Factor-Dependent CD11b+ Dendritic Cells in Human and Mouse Control Mucosal IL-17 Cytokine Responses Andreas Schlitzer, Naomi McGovern, Pearline Teo, Teresa Zelante, Koji Atarashi, Donovan Low, Adrian W.S. Ho, Peter See, Amanda Shin, Pavandip Singh Wasan, Guillaume Hoeffel, Benoit Malleret, Alexander Heiseke, Samantha Chew, Laura Jardine, Harriet A. Purvis, Catharien M.U. Hilkens, John Tam, Michael Poidinger, E. Richard Stanley, Anne B. K rug, Laurent Renia, Baalasubramanian Sivasankar, Lai Guan Ng, Matthew Collin, Paola Ricciardi-Castagnoli, Kenya Honda, Muzlifah Haniffa, and Florent Ginhoux Supplemental Inventory 1. Supplemental Figures and Tables Figure S1, Related to Figure 1 Figure S2, Related to Figure 2 and 3 Figure S3, Related to Figure 4 Figure S4, Related to Figure 5 Figure S5, Related to Figure 5 Figure S6, Related to Figure 7 Table S1, Related to Figure 3 Table S2, Related to Figure 6 Table S3, Related to Figure 6 2. Supplemental Experimental Procedures 3. Supplemental References Supplementary figure 1 Sorting strategy for mouse lung and small intestinal DC A Sorting strategy, Lung Singlets Dapi-CD45+ 250K 250K 250K 105 200K 200K 200K 104 A I - 150K 150K 150K C P C C 3 10 S A S S 100K 100K S 100K D S S 102 50K 50K 50K 0 0 0 0 0 50K 100K 150K 200K 250K 0 50K 100K 150K 200K 250K 0 103 104 105 0 103 104 105 FSC FSC-W CD45 GR1 Auto Fluor.- MHCII+ GR1- SSClow CD11c+ CD11b+ 250K 105 105 105 200K 4 104 10 104 150K I 3 I 3 4 3 0 10 3 C 2 H 10 10 100K 1 S D C D S C M 2 C 10 50K 0 0 0 0 0 102 103 104 105 0 103 104 105 0 103 104 105 0 103 104 105 Auto fluor.
    [Show full text]
  • A Human Population-Based Organotypic in Vitro Model for Cardiotoxicity Screening1
    ALTEX preprint published July 8, 2018 doi:10.14573/altex.1805301 Research Article A human population-based organotypic in vitro model for cardiotoxicity screening1 Fabian A. Grimm1, Alexander Blanchette1, John S. House2, Kyle Ferguson1, Nan-Hung Hsieh1, Chimeddulam Dalaijamts1, Alec A. Wright1, Blake Anson5, Fred A. Wright3,4, Weihsueh A. Chiu1, Ivan Rusyn1 1Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA; 2Bioinformatics Research Center, 3Department of Biological Sciences, and 4Department of Statistics, North Carolina State University, Raleigh, NC, USA; 5Cellular Dynamics International, Madison, WI, USA Abstract Assessing inter-individual variability in responses to xenobiotics remains a substantial challenge, both in drug development with respect to pharmaceuticals and in public health with respect to environmental chemicals. Although approaches exist to characterize pharmacokinetic variability, there are no methods to routinely address pharmacodynamic variability. In this study, we aimed to demonstrate the feasibility of characterizing inter-individual variability in a human in vitro model. Specifically, we hypothesized that genetic variability across a population of iPSC- derived cardiomyocytes translates into reproducible variability in both baseline phenotypes and drug responses. We measured baseline and drug-related effects in iPSC-derived cardiomyocytes from 27 healthy donors on kinetic Ca2+ flux and high-content live cell imaging. Cells were treated in concentration-response with cardiotoxic drugs: isoproterenol (β- adrenergic receptor agonist/positive inotrope), propranolol (β-adrenergic receptor antagonist/negative inotrope), and cisapride (hERG channel inhibitor/QT prolongation). Cells from four of the 27 donors were further evaluated in terms of baseline and treatment-related gene expression. Reproducibility of phenotypic responses was evaluated across batches and time.
    [Show full text]
  • A Flexible Microfluidic System for Single-Cell Transcriptome Profiling
    www.nature.com/scientificreports OPEN A fexible microfuidic system for single‑cell transcriptome profling elucidates phased transcriptional regulators of cell cycle Karen Davey1,7, Daniel Wong2,7, Filip Konopacki2, Eugene Kwa1, Tony Ly3, Heike Fiegler2 & Christopher R. Sibley 1,4,5,6* Single cell transcriptome profling has emerged as a breakthrough technology for the high‑resolution understanding of complex cellular systems. Here we report a fexible, cost‑efective and user‑ friendly droplet‑based microfuidics system, called the Nadia Instrument, that can allow 3′ mRNA capture of ~ 50,000 single cells or individual nuclei in a single run. The precise pressure‑based system demonstrates highly reproducible droplet size, low doublet rates and high mRNA capture efciencies that compare favorably in the feld. Moreover, when combined with the Nadia Innovate, the system can be transformed into an adaptable setup that enables use of diferent bufers and barcoded bead confgurations to facilitate diverse applications. Finally, by 3′ mRNA profling asynchronous human and mouse cells at diferent phases of the cell cycle, we demonstrate the system’s ability to readily distinguish distinct cell populations and infer underlying transcriptional regulatory networks. Notably this provided supportive evidence for multiple transcription factors that had little or no known link to the cell cycle (e.g. DRAP1, ZKSCAN1 and CEBPZ). In summary, the Nadia platform represents a promising and fexible technology for future transcriptomic studies, and other related applications, at cell resolution. Single cell transcriptome profling has recently emerged as a breakthrough technology for understanding how cellular heterogeneity contributes to complex biological systems. Indeed, cultured cells, microorganisms, biopsies, blood and other tissues can be rapidly profled for quantifcation of gene expression at cell resolution.
    [Show full text]