Pleiotrophin Regulates the Ductular Reaction by Controlling the Migration

Total Page:16

File Type:pdf, Size:1020Kb

Pleiotrophin Regulates the Ductular Reaction by Controlling the Migration Gut Online First, published on March 10, 2015 as 10.1136/gutjnl-2014-308176 Hepatology ORIGINAL ARTICLE Gut: first published as 10.1136/gutjnl-2014-308176 on 16 January 2015. Downloaded from Pleiotrophin regulates the ductular reaction by controlling the migration of cells in liver progenitor niches Gregory A Michelotti,1 Anikia Tucker,1 Marzena Swiderska-Syn,1 Mariana Verdelho Machado,1 Steve S Choi,1,2 Leandi Kruger,1 Erik Soderblom,3 J Will Thompson,3 Meredith Mayer-Salman,3 Heather A Himburg,4 Cynthia A Moylan,1,2 Cynthia D Guy,5 Katherine S Garman,1,2 Richard T Premont,1 John P Chute,4 Anna Mae Diehl1 ▸ Additional material is ABSTRACT published online only. To view Objective The ductular reaction (DR) involves Significance of this study please visit the journal online (http://dx.doi.org/10.1136/ mobilisation of reactive-appearing duct-like cells (RDC) gutjnl-2014-308176). along canals of Hering, and myofibroblastic (MF) differentiation of hepatic stellate cells (HSC) in the space 1Division of Gastroenterology, What is already known about this subject? Duke University, Durham, of Disse. Perivascular cells in stem cell niches produce ▸ Various types of liver injury promote a ductular North Carolina, USA pleiotrophin (PTN) to inactivate the PTN receptor, protein reaction (DR) characterised by the periportal 2Section of Gastroenterology, tyrosine phosphatase receptor zeta-1 (PTPRZ1), thereby accumulation of small ductules, myofibroblasts Durham Veterans Affairs augmenting phosphoprotein-dependent signalling. We and collagen matrix. Medical Center, Durham, ▸ Pleiotrophin (PTN) is a heparin-binding growth North Carolina, USA hypothesised that the DR is regulated by PTN/PTPRZ1 3Proteomics Center, signalling. factor that inhibits constitutive tyrosine Duke University, Durham, Design PTN-GFP, PTN-knockout (KO), PTPRZ1-KO, and phosphatase activity of its receptor, protein North Carolina, USA tyrosine phosphatase receptor Z1 (PTPRZ1) to 4 wild type (WT) mice were examined before and after bile Division of Hematology and duct ligation (BDL) for PTN, PTPRZ1 and the DR. RDC regulate fate decisions in various stem/progenitor Oncology, UCLA, Los Angeles, and HSC from WT, PTN-KO, and PTPRZ1-KO mice were cells. California, USA ▸ In the liver, PTN expression is induced in several 5 also treated with PTN to determine effects on Department of Pathology, conditions associated with liver progenitor Duke University, Durham, downstream signaling phosphoproteins, gene expression, accumulation, including bile duct ligation, partial North Carolina, USA http://gut.bmj.com/ growth, and migration. Liver biopsies from patients with hepatectomy and hepatocellular carcinoma. Correspondence to DRs were also interrogated. ▸ Liver regeneration is inhibited in PTN-deficient Professor Anna Mae Diehl, Results Although quiescent HSC and RDC lines mice after partial hepatectomy, suggesting that Division of Gastroenterology, expressed PTN and PTPRZ1 mRNAs, neither PTN nor PTN promotes liver repair. Duke University, Durham, PTPRZ1 protein was demonstrated in healthy liver. BDL NC 27710, USA; induced PTN in MF-HSC and increased PTPRZ1 in MF- What are the new findings? [email protected] HSC and RDC. In WT mice, BDL triggered a DR ▸ Cells in putative liver stem/progenitor niches, that is, GAM and AT contributed characterised by periportal accumulation of collagen, the space of Disse and canals of Hering, express on September 26, 2021 by guest. Protected copyright. equally. RDC and MF-HSC. All aspects of this DR were increased PTN and its receptor, PTPRZ1 and modulate in PTN-KO mice and suppressed in PTPRZ1-KO mice. In PTN/PTPRZ1 expression during liver injury. Received 1 August 2014 ▸ vitro studies revealed PTN-dependent accumulation of PTN-PTPRZ1 signalling controls the mobilisation Revised 19 December 2014 of cells that normally reside in stem/progenitor phosphoproteins that control cell-cell adhesion and Accepted 22 December 2014 niches of adult livers, thereby regulating the migration, with resultant inhibition of cell migration. intensity of the DR. PTPRZ1-positive cells were prominent in the DRs of ▸ PTN-PTPRZ1 interaction in liver cells modulates patients with ductal plate defects and adult cholestatic the tyrosine phosphorylation of factors that diseases. control cell-cell adhesion, cell-matrix interactions Conclusions PTN, and its receptor, PTPRZ1, regulate and cell migration. the DR to liver injury by controlling the migration of ▸ PTPRZ1 is prominent in the DRs of von resident cells in adult liver progenitor niches. Meyenburg complexes, adult polycystic liver disease, primary biliary cirrhosis and primary sclerosing cholangitis. How might it impact on clinical practice in INTRODUCTION the foreseeable future? Various types of liver injury promote a ductular ▸ PTN/PTPRZ1 signalling might be manipulated reaction (DR) characterised by the periportal accu- To cite: Michelotti GA, therapeutically to optimise recovery from chronic mulation of small ductules comprised of cholestatic liver injury. Tucker A, Swiderska-Syn M, fi et al. Gut Published Online reactive-appearing duct-like cells (RDC), myo bro- ▸ Inter-individual differences in PTN/PTPRZ1 First: [please include Day blasts (MF) and collagen matrix. Neither the pathway activity might be exploited as Month Year] doi:10.1136/ mechanisms driving this DR, nor its biological sig- biomarkers for progressive liver fibrosis. gutjnl-2014-308176 nificance, are well understood.1 Bipotent liver Michelotti GA, et al. Gut 2015;0:1–10. doi:10.1136/gutjnl-2014-308176 1 Copyright Article author (or their employer) 2015. Produced by BMJ Publishing Group Ltd (& BSG) under licence. Hepatology progenitors that can differentiate into ductular cells reside along MATERIALS AND METHODS canals of Hering; vestiges of fetal ductal plates that persist Human liver biopsies Gut: first published as 10.1136/gutjnl-2014-308176 on 16 January 2015. Downloaded from around portal tracts in adult livers.2 Multipotent liver progeni- Anonomised unstained liver sections from formalin-fixed paraf- tors have also been identified in submucosal glands within the fin embedded tissue blocks of individuals with normal liver (3), wall of the adult biliary tree.3 Mature hepatocyte and cholangio- adult polycystic disease (n=1), von Meyenburg complexes cytes can also cause RDC with properties of liver progenitors.45 (n=1), primary biliary cirrhosis (n=2) and primary sclerosing It has been suggested that the DR reflects injury-related mobil- cholangitis (n=4) were obtained from the Duke Department of isation of liver progenitors from varying sources.6 We evaluated Pathology in accordance with our Duke IRB-approved protocol. the hypothesis that the DR is regulated by pleiotrophin (PTN) Liver sections were processed for immunohistochemistry as and its receptor, protein tyrosine phosphatase receptor zeta-1 detailed below. (PTPRZ1). PTN regulates fate decisions in various stem/progenitor Mice 7–12 cells. PTN expression has been demonstrated in two condi- PTN-GFP, PTN knockout (KO), and PTPRZ1 KO mice have tions associated with accumulation of liver progenitors; partial been previously described.11 C57BL/6 and SV129 brown WT 13–16 hepatectomy (PH) and hepatocellular carcinoma (HCC). mice were used as strain-specific controls for PTN KO and Bile duct ligation (BDL), a well-established stimulus for the DR, PTPRZ1-KO mice, respectively, and were obtained from The 17 also induces PTN. PTN appears to promote liver growth Jackson Laboratory and Taconic, respectively. because regeneration after PH is inhibited in PTN-deficient To induce cholestatic injury, adult (10–12 weeks) mice (C57BL/ 14 mice. PTN is known to interact with various molecules, 6 WT (n=7), PTN-KO (n=9) mice, SV129 brown WT (n=6), and including syndecans, integrins, anaplastic lymphoma kinase PTPRZ1-KO mice (n=8)) underwent BDL or sham surgery (n=3/ (ALK) and PTPRZ1. Among these, PTPRZ1 is best charac- group). After 14 days, animals were euthanised; blood and tissues terised as a true receptor for PTN because PTN-PTPRZ1 inter- were obtained as described.28 All studies were performed in action inhibits the constitutive tyrosine phosphatase activity of accordance with a Duke University IACUC-approved protocol. PTPRZ1, resulting in accumulation of phosphorylated tyrosine residues in ALK and other proteins that are ordinarily depho- 18 19 Cell culture sphorylated by PTPRZ1. Prolonged PTN-dependent Primary cell isolation from mice phospho-tyrosine-mediated tyrosine kinase activity modulates 920 Primary HSC, LSEC, and hepatocytes were isolated from a signalling in an array of kinase cascades, such as PI3-kinase, 28 30 821 21 12-week-old mice. Akt, extracellular-signal regulated kinase (ERK)1/2, and HSC and LSEC viability and purity were greater than 95% as α β 22 PKC / , providing a mechanism by which PTN might globally previously described,28 30 HSCs were cultured for up to 9 days impact cell fate. Cytoskeletal proteins that regulate migration, in standard Dulbecco’s Modified Eagle Medium supplemented proliferation, and differentiation, are downstream effectors 23–25 with 10% fetal bovine serum, and 1% penicillin-streptomycin. regulated by PTPRZ1. Hepatocytes were used immediately for RNA isolation. Cell In certain stem cell niches, PTN is expressed by endothelial 11 viability exceeded 90% in all experiments as determined by cells. Although resident liver pericytes (a.k.a., hepatic stellate Trypan-blue exclusion. http://gut.bmj.com/ cells (HSC)) produce PTN, it is not known if liver sinusoidal The murine ductular
Recommended publications
  • RNA Isolation and Real-Time PCR Analysis
    Pleiotrophin deletion alters glucose homeostasis, energy metabolism and brown fat thermogenic function. Sevillano, Julio1#; Sánchez-Alonso, María Gracia1#; Zapatería, Begoña1; Calderón, María1; Alcalá, Martín1, Limones, María1; Pita, Jimena1; Gramage, Esther2; Vicente- Rodríguez, Marta2; Horrillo, Daniel4; Medina-Gómez, Gema4; Obregón, María Jesús5; Viana, Marta1; Valladolid-Acebes, Ismael3, Herradón, Gonzalo2, Ramos, María del Pilar1*. Affiliations 1Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad CEU San Pablo, Madrid, Spain. 2Department of Pharmaceutical and Health Sciences, Facultad de Farmacia, Universidad CEU San Pablo, Madrid, Spain. 3The Rolf Luft Research Center for Diabetes and Endocrinology, Department of Molecular Medicine and Surgery, Karolinska Institutet, 171 76, Stockholm, Sweden 4Department of Basic Sciences of Health. Universidad Rey Juan Carlos. Alcorcón. Madrid. Spain. 5Department of Endocrine and Nervous System Pathophysiology, Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain. Keywords: Glucose homeostasis; Metabolism; Adipose tissue; Insulin resistance, Thermogenesis. *To whom correspondence should be addressed: Mª del Pilar Ramos Álvarez, PhD. Department of Chemistry and Biochemistry Facultad de Farmacia, Universidad CEU San Pablo Ctra. Boadilla del Monte km 5,3 28668, Madrid 1 +34-91-3724760 [email protected] Additional Title Page Footnotes #Co-first authors This study was supported by Spanish Ministry of Economy and Competitiveness (SAF2010-19603 and SAF2014-56671-R, SAF2012-32491, BFU2013-47384-R and BFU2016-78951-R) and Community of Madrid (S2010/BMD-2423, S2017/BMD-3864). Running Title: PLEITROPHIN AND ENERGY METABOLISM Aims/hypothesis: Pleiotrophin, a developmentally regulated and highly conserved cytokine, exerts different functions including regulation of cell growth, migration and survival.
    [Show full text]
  • Pleiotrophin Is a Neurotrophic Factor for Spinal Motor Neurons
    Pleiotrophin is a neurotrophic factor for spinal motor neurons Ruifa Mi, Weiran Chen, and Ahmet Ho¨ ke* Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287 Edited by Thomas M. Jessell, Columbia University Medical Center, New York, NY, and approved January 18, 2007 (received for review April 21, 2006) Regeneration in the peripheral nervous system is poor after chronic facial motor neurons against cell death induced by deprivation from denervation. Denervated Schwann cells act as a ‘‘transient target’’ target-derived neurotrophic support. by secreting growth factors to promote regeneration of axons but lose this ability with chronic denervation. We discovered that the Results mRNA for pleiotrophin (PTN) was highly up-regulated in acutely PTN Is Up-Regulated in Denervated Schwann Cells and Muscle After denervated distal sciatic nerves, but high levels of PTN mRNA were Axotomy. To identify candidate neurotrophic factors underlying not maintained in chronically denervated nerves. PTN protected adaptive responses to chronic nerve degeneration, we used focused spinal motor neurons against chronic excitotoxic injury and caused cDNA microarrays to investigate the gene expression of neurotro- increased outgrowth of motor axons out of the spinal cord ex- phic factors in denervated Schwann cells. In microarray experi- plants and formation of ‘‘miniventral rootlets.’’ In neonatal mice, ments, 2 and 7 days after the sciatic nerve transection, PTN mRNA PTN protected the facial motor neurons against cell death induced was up-regulated in the distal denervated segments compared with by deprivation from target-derived growth factors. Similarly, PTN the contralateral side (data not shown). To confirm the up- significantly enhanced regeneration of myelinated axons across a regulation of the PTN mRNA observed in the microarray analysis graft in the transected sciatic nerve of adult rats.
    [Show full text]
  • Related Malignant Phenotypes in the Nf1-Deficient MPNST
    Published OnlineFirst February 19, 2013; DOI: 10.1158/1541-7786.MCR-12-0593 Molecular Cancer Genomics Research RAS/MEK–Independent Gene Expression Reveals BMP2- Related Malignant Phenotypes in the Nf1-Deficient MPNST Daochun Sun1, Ramsi Haddad2,3, Janice M. Kraniak2, Steven D. Horne1, and Michael A. Tainsky1,2 Abstract Malignant peripheral nerve sheath tumor (MPNST) is a type of soft tissue sarcoma that occurs in carriers of germline mutations in Nf1 gene as well as sporadically. Neurofibromin, encoded by the Nf1 gene, functions as a GTPase-activating protein (GAP) whose mutation leads to activation of wt-RAS and mitogen-activated protein kinase (MAPK) signaling in neurofibromatosis type I (NF1) patients' tumors. However, therapeutic targeting of RAS and MAPK have had limited success in this disease. In this study, we modulated NRAS, mitogen-activated protein/extracellular signal–regulated kinase (MEK)1/2, and neurofibromin levels in MPNST cells and determined gene expression changes to evaluate the regulation of signaling pathways in MPNST cells. Gene expression changes due to neurofibromin modulation but independent of NRAS and MEK1/2 regulation in MPNST cells indicated bone morphogenetic protein 2 (Bmp2) signaling as a key pathway. The BMP2-SMAD1/5/8 pathway was activated in NF1-associated MPNST cells and inhibition of BMP2 signaling by LDN-193189 or short hairpin RNA (shRNA) to BMP2 decreased the motility and invasion of NF1-associated MPNST cells. The pathway-specific gene changes provide a greater understanding of the complex role of neurofibromin in MPNST pathology and novel targets for drug discovery. Mol Cancer Res; 11(6); 616–27.
    [Show full text]
  • Investigation of Candidate Genes and Mechanisms Underlying Obesity
    Prashanth et al. BMC Endocrine Disorders (2021) 21:80 https://doi.org/10.1186/s12902-021-00718-5 RESEARCH ARTICLE Open Access Investigation of candidate genes and mechanisms underlying obesity associated type 2 diabetes mellitus using bioinformatics analysis and screening of small drug molecules G. Prashanth1 , Basavaraj Vastrad2 , Anandkumar Tengli3 , Chanabasayya Vastrad4* and Iranna Kotturshetti5 Abstract Background: Obesity associated type 2 diabetes mellitus is a metabolic disorder ; however, the etiology of obesity associated type 2 diabetes mellitus remains largely unknown. There is an urgent need to further broaden the understanding of the molecular mechanism associated in obesity associated type 2 diabetes mellitus. Methods: To screen the differentially expressed genes (DEGs) that might play essential roles in obesity associated type 2 diabetes mellitus, the publicly available expression profiling by high throughput sequencing data (GSE143319) was downloaded and screened for DEGs. Then, Gene Ontology (GO) and REACTOME pathway enrichment analysis were performed. The protein - protein interaction network, miRNA - target genes regulatory network and TF-target gene regulatory network were constructed and analyzed for identification of hub and target genes. The hub genes were validated by receiver operating characteristic (ROC) curve analysis and RT- PCR analysis. Finally, a molecular docking study was performed on over expressed proteins to predict the target small drug molecules. Results: A total of 820 DEGs were identified between
    [Show full text]
  • Genetic Drivers of Pancreatic Islet Function
    | INVESTIGATION Genetic Drivers of Pancreatic Islet Function Mark P. Keller,*,1 Daniel M. Gatti,†,1 Kathryn L. Schueler,* Mary E. Rabaglia,* Donnie S. Stapleton,* Petr Simecek,† Matthew Vincent,† Sadie Allen,‡ Aimee Teo Broman,§ Rhonda Bacher,§ Christina Kendziorski,§ Karl W. Broman,§ Brian S. Yandell,** Gary A. Churchill,†,2 and Alan D. Attie*,2 *Department of Biochemistry, §Department of Biostatistics and Medical Informatics, and **Department of Horticulture, University of Wisconsin–Madison, Wisconsin 53706-1544, †The Jackson Laboratory, Bar Harbor, Maine 06409, and ‡Maine School of Science and Mathematics, Limestone, Maine 06409, ORCID IDs: 0000-0002-7405-5552 (M.P.K.); 0000-0002-4914-6671 (K.W.B.); 0000-0001-9190-9284 (G.A.C.); 0000-0002-0568-2261 (A.D.A.) ABSTRACT The majority of gene loci that have been associated with type 2 diabetes play a role in pancreatic islet function. To evaluate the role of islet gene expression in the etiology of diabetes, we sensitized a genetically diverse mouse population with a Western diet high in fat (45% kcal) and sucrose (34%) and carried out genome-wide association mapping of diabetes-related phenotypes. We quantified mRNA abundance in the islets and identified 18,820 expression QTL. We applied mediation analysis to identify candidate causal driver genes at loci that affect the abundance of numerous transcripts. These include two genes previously associated with monogenic diabetes (PDX1 and HNF4A), as well as three genes with nominal association with diabetes-related traits in humans (FAM83E, IL6ST, and SAT2). We grouped transcripts into gene modules and mapped regulatory loci for modules enriched with transcripts specific for a-cells, and another specific for d-cells.
    [Show full text]
  • 140503 IPF Signatures Supplement Withfigs Thorax
    Supplementary material for Heterogeneous gene expression signatures correspond to distinct lung pathologies and biomarkers of disease severity in idiopathic pulmonary fibrosis Daryle J. DePianto1*, Sanjay Chandriani1⌘*, Alexander R. Abbas1, Guiquan Jia1, Elsa N. N’Diaye1, Patrick Caplazi1, Steven E. Kauder1, Sabyasachi Biswas1, Satyajit K. Karnik1#, Connie Ha1, Zora Modrusan1, Michael A. Matthay2, Jasleen Kukreja3, Harold R. Collard2, Jackson G. Egen1, Paul J. Wolters2§, and Joseph R. Arron1§ 1Genentech Research and Early Development, South San Francisco, CA 2Department of Medicine, University of California, San Francisco, CA 3Department of Surgery, University of California, San Francisco, CA ⌘Current address: Novartis Institutes for Biomedical Research, Emeryville, CA. #Current address: Gilead Sciences, Foster City, CA. *DJD and SC contributed equally to this manuscript §PJW and JRA co-directed this project Address correspondence to Paul J. Wolters, MD University of California, San Francisco Department of Medicine Box 0111 San Francisco, CA 94143-0111 [email protected] or Joseph R. Arron, MD, PhD Genentech, Inc. MS 231C 1 DNA Way South San Francisco, CA 94080 [email protected] 1 METHODS Human lung tissue samples Tissues were obtained at UCSF from clinical samples from IPF patients at the time of biopsy or lung transplantation. All patients were seen at UCSF and the diagnosis of IPF was established through multidisciplinary review of clinical, radiological, and pathological data according to criteria established by the consensus classification of the American Thoracic Society (ATS) and European Respiratory Society (ERS), Japanese Respiratory Society (JRS), and the Latin American Thoracic Association (ALAT) (ref. 5 in main text). Non-diseased normal lung tissues were procured from lungs not used by the Northern California Transplant Donor Network.
    [Show full text]
  • Hydrogel Mediated Delivery of Trophic Factors for Neural Repair Joshua S
    Advanced Review Hydrogel mediated delivery of trophic factors for neural repair Joshua S. Katz1 and Jason A. Burdick∗ Neurotrophins have been implicated in a variety of diseases and their delivery to sites of disease and injury has therapeutic potential in applications including spinal cord injury, Alzheimer’s disease, and Parkinson’s disease. Biodegradable polymers, and specifically, biodegradable water-swollen hydrogels, may be advantageous as delivery vehicles for neurotrophins because of tissue-like properties, tailorability with respect to degradation and release behavior, and a history of biocompatibility. These materials may be designed to degrade via hydrolytic or enzymatic mechanisms and can be used for the sustained delivery of trophic factors in vivo. Hydrogels investigated to date include purely synthetic to purely natural, depending on the application and intended release profiles. Also, flexibility in material processing has allowed for the investigation of injectable materials, the development of scaffolding and porous conduits, and the use of composites for tailored molecule delivery profiles. It is the objective of this review to describe what has been accomplished in this area thus far and to remark on potential future directions in this field. Ultimately, the goal is to engineer optimal biomaterials to deliver molecules in a controlled and dictated manner that can promote regeneration and healing for numerous neural applications. 2008 John Wiley & Sons, Inc. Wiley Interdiscipl. Rev. Nanomed. Nanobiotechnol. 2009 1 128–139
    [Show full text]
  • Macrophages/Microglia in the Glioblastoma Tumor Microenvironment
    International Journal of Molecular Sciences Review Macrophages/Microglia in the Glioblastoma Tumor Microenvironment Jun Ma, Clark C. Chen * and Ming Li * Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA; [email protected] * Correspondence: [email protected] (C.C.C.); [email protected] (M.L.) Abstract: The complex interaction between glioblastoma and its microenvironment has been rec- ognized for decades. Among various immune profiles, the major population is tumor-associated macrophage, with microglia as its localized homolog. The present definition of such myeloid cells is based on a series of cell markers. These good sentinel cells experience significant changes, facilitat- ing glioblastoma development and protecting it from therapeutic treatments. Huge, complicated mechanisms are involved during the overall processes. A lot of effort has been dedicated to crack the mysterious codes in macrophage/microglia recruiting, activating, reprogramming, and functioning. We have made our path. With more and more key factors identified, a lot of new therapeutic methods could be explored to break the ominous loop, to enhance tumor sensitivity to treatments, and to improve the prognosis of glioblastoma patients. However, it might be a synergistic system rather than a series of clear, stepwise events. There are still significant challenges before the light of truth can shine onto the field. Here, we summarize recent advances in this field, reviewing the path we have been on and where we are now. Keywords: glioblastoma; tumor microenvironment; glioblastoma-associated macrophages/microglia; macrophage; microglia Citation: Ma, J.; Chen, C.C.; Li, M. Macrophages/Microglia in the Glioblastoma Tumor Int. J. Mol. Sci. Microenvironment. 1. Glioblastoma Associated Macrophages/Microglia 2021, 22, 5775.
    [Show full text]
  • 1 No. Affymetrix ID Gene Symbol Genedescription Gotermsbp Q Value 1. 209351 at KRT14 Keratin 14 Structural Constituent of Cyto
    1 Affymetrix Gene Q No. GeneDescription GOTermsBP ID Symbol value structural constituent of cytoskeleton, intermediate 1. 209351_at KRT14 keratin 14 filament, epidermis development <0.01 biological process unknown, S100 calcium binding calcium ion binding, cellular 2. 204268_at S100A2 protein A2 component unknown <0.01 regulation of progression through cell cycle, extracellular space, cytoplasm, cell proliferation, protein kinase C inhibitor activity, protein domain specific 3. 33323_r_at SFN stratifin/14-3-3σ binding <0.01 regulation of progression through cell cycle, extracellular space, cytoplasm, cell proliferation, protein kinase C inhibitor activity, protein domain specific 4. 33322_i_at SFN stratifin/14-3-3σ binding <0.01 structural constituent of cytoskeleton, intermediate 5. 201820_at KRT5 keratin 5 filament, epidermis development <0.01 structural constituent of cytoskeleton, intermediate 6. 209125_at KRT6A keratin 6A filament, ectoderm development <0.01 regulation of progression through cell cycle, extracellular space, cytoplasm, cell proliferation, protein kinase C inhibitor activity, protein domain specific 7. 209260_at SFN stratifin/14-3-3σ binding <0.01 structural constituent of cytoskeleton, intermediate 8. 213680_at KRT6B keratin 6B filament, ectoderm development <0.01 receptor activity, cytosol, integral to plasma membrane, cell surface receptor linked signal transduction, sensory perception, tumor-associated calcium visual perception, cell 9. 202286_s_at TACSTD2 signal transducer 2 proliferation, membrane <0.01 structural constituent of cytoskeleton, cytoskeleton, intermediate filament, cell-cell adherens junction, epidermis 10. 200606_at DSP desmoplakin development <0.01 lectin, galactoside- sugar binding, extracellular binding, soluble, 7 space, nucleus, apoptosis, 11. 206400_at LGALS7 (galectin 7) heterophilic cell adhesion <0.01 2 S100 calcium binding calcium ion binding, epidermis 12. 205916_at S100A7 protein A7 (psoriasin 1) development <0.01 S100 calcium binding protein A8 (calgranulin calcium ion binding, extracellular 13.
    [Show full text]
  • Genetic Alterations of Protein Tyrosine Phosphatases in Human Cancers
    Oncogene (2015) 34, 3885–3894 © 2015 Macmillan Publishers Limited All rights reserved 0950-9232/15 www.nature.com/onc REVIEW Genetic alterations of protein tyrosine phosphatases in human cancers S Zhao1,2,3, D Sedwick3,4 and Z Wang2,3 Protein tyrosine phosphatases (PTPs) are enzymes that remove phosphate from tyrosine residues in proteins. Recent whole-exome sequencing of human cancer genomes reveals that many PTPs are frequently mutated in a variety of cancers. Among these mutated PTPs, PTP receptor T (PTPRT) appears to be the most frequently mutated PTP in human cancers. Beside PTPN11, which functions as an oncogene in leukemia, genetic and functional studies indicate that most of mutant PTPs are tumor suppressor genes. Identification of the substrates and corresponding kinases of the mutant PTPs may provide novel therapeutic targets for cancers harboring these mutant PTPs. Oncogene (2015) 34, 3885–3894; doi:10.1038/onc.2014.326; published online 29 September 2014 INTRODUCTION tyrosine/threonine-specific phosphatases. (4) Class IV PTPs include Protein tyrosine phosphorylation has a critical role in virtually all four Drosophila Eya homologs (Eya1, Eya2, Eya3 and Eya4), which human cellular processes that are involved in oncogenesis.1 can dephosphorylate both tyrosine and serine residues. Protein tyrosine phosphorylation is coordinately regulated by protein tyrosine kinases (PTKs) and protein tyrosine phosphatases 1 THE THREE-DIMENSIONAL STRUCTURE AND CATALYTIC (PTPs). Although PTKs add phosphate to tyrosine residues in MECHANISM OF PTPS proteins, PTPs remove it. Many PTKs are well-documented oncogenes.1 Recent cancer genomic studies provided compelling The three-dimensional structures of the catalytic domains of evidence that many PTPs function as tumor suppressor genes, classical PTPs (RPTPs and non-RPTPs) are extremely well because a majority of PTP mutations that have been identified in conserved.5 Even the catalytic domain structures of the dual- human cancers are loss-of-function mutations.
    [Show full text]
  • Inhibition of Metastasis by HEXIM1 Through Effects on Cell Invasion and Angiogenesis
    Oncogene (2013) 32, 3829–3839 & 2013 Macmillan Publishers Limited All rights reserved 0950-9232/13 www.nature.com/onc ORIGINAL ARTICLE Inhibition of metastasis by HEXIM1 through effects on cell invasion and angiogenesis W Ketchart1, KM Smith2, T Krupka3, BM Wittmann1,7,YHu1, PA Rayman4, YQ Doughman1, JM Albert5, X Bai6, JH Finke4,YXu2, AA Exner3 and MM Montano1 We report on the role of hexamethylene-bis-acetamide-inducible protein 1 (HEXIM1) as an inhibitor of metastasis. HEXIM1 expression is decreased in human metastatic breast cancers when compared with matched primary breast tumors. Similarly we observed decreased expression of HEXIM1 in lung metastasis when compared with primary mammary tumors in a mouse model of metastatic breast cancer, the polyoma middle T antigen (PyMT) transgenic mouse. Re-expression of HEXIM1 (through transgene expression or localized delivery of a small molecule inducer of HEXIM1 expression, hexamethylene-bis-acetamide) in PyMT mice resulted in inhibition of metastasis to the lung. Our present studies indicate that HEXIM1 downregulation of HIF-1a protein allows not only for inhibition of vascular endothelial growth factor-regulated angiogenesis, but also for inhibition of compensatory pro- angiogenic pathways and recruitment of bone marrow-derived cells (BMDCs). Another novel finding is that HEXIM1 inhibits cell migration and invasion that can be partly attributed to decreased membrane localization of the 67 kDa laminin receptor, 67LR, and inhibition of the functional interaction of 67LR with laminin. Thus, HEXIM1 re-expression in breast cancer has therapeutic advantages by simultaneously targeting more than one pathway involved in angiogenesis and metastasis. Our results also support the potential for HEXIM1 to indirectly act on multiple cell types to suppress metastatic cancer.
    [Show full text]
  • Heparin/Heparan Sulfate Proteoglycans Glycomic Interactome in Angiogenesis: Biological Implications and Therapeutical Use
    Molecules 2015, 20, 6342-6388; doi:10.3390/molecules20046342 OPEN ACCESS molecules ISSN 1420-3049 www.mdpi.com/journal/molecules Review Heparin/Heparan Sulfate Proteoglycans Glycomic Interactome in Angiogenesis: Biological Implications and Therapeutical Use Paola Chiodelli, Antonella Bugatti, Chiara Urbinati and Marco Rusnati * Section of Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy; E-Mails: [email protected] (P.C.); [email protected] (A.B.); [email protected] (C.U.) * Author to whom correspondence should be addressed; E-Mail: [email protected]; Tel.: +39-030-371-7315; Fax: +39-030-371-7747. Academic Editor: Els Van Damme Received: 26 February 2015 / Accepted: 1 April 2015 / Published: 10 April 2015 Abstract: Angiogenesis, the process of formation of new blood vessel from pre-existing ones, is involved in various intertwined pathological processes including virus infection, inflammation and oncogenesis, making it a promising target for the development of novel strategies for various interventions. To induce angiogenesis, angiogenic growth factors (AGFs) must interact with pro-angiogenic receptors to induce proliferation, protease production and migration of endothelial cells (ECs). The action of AGFs is counteracted by antiangiogenic modulators whose main mechanism of action is to bind (thus sequestering or masking) AGFs or their receptors. Many sugars, either free or associated to proteins, are involved in these interactions, thus exerting a tight regulation of the neovascularization process. Heparin and heparan sulfate proteoglycans undoubtedly play a pivotal role in this context since they bind to almost all the known AGFs, to several pro-angiogenic receptors and even to angiogenic inhibitors, originating an intricate network of interaction, the so called “angiogenesis glycomic interactome”.
    [Show full text]