Funktionelle in Vitro Und in Vivo Charakterisierung Des Putativen Tumorsuppressorgens SFRP1 Im Humanen Mammakarzinom

Total Page:16

File Type:pdf, Size:1020Kb

Funktionelle in Vitro Und in Vivo Charakterisierung Des Putativen Tumorsuppressorgens SFRP1 Im Humanen Mammakarzinom Funktionelle in vitro und in vivo Charakterisierung des putativen Tumorsuppressorgens SFRP1 im humanen Mammakarzinom Von der Fakult¨at fur¨ Mathematik, Informatik und Naturwissenschaften der RWTH Aachen University zur Erlangung des akademischen Grades einer Doktorin der Naturwissenschaften genehmigte Dissertation vorgelegt von Diplom-Biologin Laura Huth (geb. Franken) aus Julich¨ Berichter: Universit¨atsprofessor Dr. rer. nat. Edgar Dahl Universit¨atsprofessor Dr. rer. nat. Ralph Panstruga Tag der mundlichen¨ Prufung:¨ 6. August 2014 Diese Dissertation ist auf den Internetseiten der Hochschulbibliothek online verfugbar.¨ Zusammenfassung Krebserkrankungen stellen weltweit eine der h¨aufigsten Todesursachen dar. Aus diesem Grund ist die Aufkl¨arung der zugrunde liegenden Mechanismen und Ur- sachen ein essentielles Ziel der molekularen Onkologie. Die Tumorforschung der letzten Jahre hat gezeigt, dass die Entstehung solider Karzinome ein Mehrstufen- Prozess ist, bei dem neben Onkogenen auch Tumorsuppresorgene eine entschei- dende Rolle spielen. Viele der heute bekannten Gene des WNT-Signalweges wur- den bereits als Onkogene oder Tumorsuppressorgene charakterisiert. Eine Dere- gulation des WNT-Signalweges wird daher mit der Entstehung und Progression vieler humaner Tumorentit¨aten wie beispielsweise auch dem Mammakarzinom, der weltweit h¨aufigsten Krebserkrankung der Frau, assoziiert. SFRP1, ein nega- tiver Regulator der WNT-Signalkaskade, wird in Brusttumoren haupts¨achlich durch den epigenetischen Mechanismus der Promotorhypermethylierung inakti- viert. Im Rahmen dieser Arbeit wurde die Beziehung eines SFRP1-Verlustes zur molekularen Entstehung humaner Brusttumoren entschlusselt.¨ Zun¨achst kl¨arte eine detaillierte, funktionelle Charakterisierung den Einfluss von SFRP1 auf tumorassoziierte Eigenschaften von Mammakarzinomzellen auf. Voraussetzung hierfur¨ war die Etablierung von gain-of-function Modellen in jeweils zwei basalen sowie luminalen Mammakarzinomzelllinien. Fur¨ die basalen BT-20 und MDA- MB-231 Zelllinien wurde nach der SFRP1-Reexpression eine signifikant vermin- derte Proliferationsf¨ahigkeit und Kolonienbildung sowie eine erh¨ohte Adh¨asions- und Apoptoserate festgestellt. Die luminalen Mammakarzinomzelllinien MCF7 und SK-BR-3 wiesen im Gegensatz dazu nach konstitutiver SFRP1-Expression keine Unterschiede in den untersuchten tumorassoziierten Eigenschaften vergli- chen mit SFRP1 defizienten Zellen auf. Interessanterweise konnten durch ei- ne zus¨atzliche Transfektion mit WNT1 und daraus resultierender Aktivierung des kanonischen WNT-Signalweges auch in den beiden luminalen in vitro Tu- 3 Zusammenfassung mormodellen eine signifikant verminderte Kolonienbildung sowie eine erh¨ohte Adh¨asions- und Apoptoserate beobachtet werden. Ein Einfluss von SFRP1 auf die Migration konnte in keiner der vier untersuchten Mammakarzinomzelllini- en aufgezeigt werden. Weiterhin konnte durch Hinzugabe von rekombinantem SFRP1-Protein in das Kulturmedium das Kolonienwachstum in basalen Mam- makarzinomzelllinien signifikant reduziert werden. Ein in vivo Tumorgenesemo- dell belegte schließlich, dass SFRP1 exprimierende MDA-MB-231 Mammakar- zinomzellen nach Implantation in das Brustgewebe weiblicher, immunsuppri- mierter Nacktm¨ause ein signifikant reduziertes Tumorwachstum im Vergleich zur Kontrollgruppe aufwiesen. Insgesamt wurde somit eine eindeutig tumor- suppressive Funktion von SFRP1 sowohl in vitro als auch in vivo festgestellt. Anschließende Microarray Genexpressionsanalysen des basalen BT-20 sowie des luminalen SK-BR-3 Tumormodells trugen zu einem genaueren Verst¨andnis bei, welche Gene und Signalkaskaden der epithelialen Brustzelle durch einen Verlust von SFRP1 maßgeblich beeinflusst werden. Anhand von Gen-Ontologie Analysen wurde dabei neben der bereits bekannten Funktion von SFRP1 als ne- gativer Regulator des WNT-Signalweges eine Assoziation zur BMP- sowie Smoo- thened-Signalkaskade hergestellt. Zudem konnte eine Vielzahl von Genen iden- tifiziert werden, die nach SFRP1-Reexpression differenzielle Expressionsmuster aufwiesen wie beispielsweise BDNF, LY96, MMP1, ST18, BIN1 und RERGL. Das Gen BDNF ruckte¨ dabei in den Fokus der Untersuchungen als potenzi- elles Target der SFRP1-vermittelten Signaltransduktion. Eine Ko-Regulation von SFRP1 und BNDF konnte sowohl in den luminalen SK-BR-3 als auch den basalen BT-20-Zellen gezeigt werden. Eine Untersuchung der mRNA- sowie der Proteinexpression in prim¨aren Mammakarzinomen best¨atigte eine positive Korrelation der SFRP1- und BDNF-Expression. Weiterhin zeigten Patientinnen mit einer hohen SFRP1- und BDNF-Expression ein signifikant verl¨angertes Ge- samtuberleben.¨ Eine BDNF-Uberexpression¨ in humanen Mammakarzinomzellen resultierte in einer verringerten Proliferationsrate. Weitere Ver¨anderungen der tumorassoziierten Eigenschaften nach einer BDNF-Reexpression wurden nicht beobachtet. In dieser Arbeit ist es gelungen, die Funktion von SFRP1 als Tumorsup- pressor mittels unterschiedlichster funktioneller Untersuchungen herauszustellen sowie zellul¨are Signalwege und Gene zu identifizieren, die durch einen SFRP1- Verlust beeinflusst werden. 4 Summary Cancer represents a leading cause of death worldwide. The elucidation of the underlying mechanisms and causes therefore is an essential goal of molecular oncology research. Tumor research during the past few years has shown that the formation of solid cancers is a multistep process in which not only oncogenes but also tumor suppressor genes play a crucial role. Many of the currently known genes of the WNT signaling pathway have been characterized as oncogenes or tumor suppressor genes. Thus, a deregulation of the WNT signaling pathway leads to the development and progression of cancer in many human tumor types such as breast cancer, the world's most common cancer in women. SFRP1, a negative regulator of the WNT signaling pathway, is inactivated in breast tumors mainly due to the epigenetic mechanism of promoter hypermethylation. In this study the relationship between a loss of SFRP1 and the molecular pathogenesis of human breast cancer was deciphered. First, a detailed functional characterization clarified the influence of SFRP1 on tumor-associated proper- ties of breast cancer cells. This required the establishment of gain-of-function models each in two basal and luminal breast cancer cell lines. In the basal- like cell lines BT-20 and MDA-MB-231 a significantly reduced proliferation rate and colony formation capability were detectable. Furthermore an increased ad- hesion and apoptosis rate were determined after SFRP1-reexpression. By con- trast, the luminal-like breast cancer cell lines MCF7 and SK-BR-3 exhibited no differences in the examined tumor-associated properties after constitutive SFRP1-expression compared with SFRP1-deficient cells. Interestingly, an ad- ditional transfection with WNT1 and the resulting activation of the canonical WNT signaling pathway led to a significantly decreased colony formation capa- bility and increased adhesion and apoptosis rates in both luminal-like in vitro tumor models. An effect of SFRP1 on the migration behavior could not be de- tected in any of the four breast cancer cell lines. Furthermore, the colony growth 5 Summary of basal-like breast cancer cell lines was significantly reduced by the addition of recombinant SFRP1 protein into the culture medium. Finally, an in vivo tumorigenesis model documented that SFRP1-expressing MDA-MB-231 breast cancer cells showed a significantly reduced tumor growth after implantation in the breast tissue of female, immunocompromised nude mice in comparison to the control group. Overall, a tight tumor suppressive function of SFRP1 was observed for both in vitro and in vivo models. Subsequently, a microarray based gene expression analysis of the basal-like BT-20 and the luminal-like SK-BR-3 tumor models contributed to a more pre- cise understanding which genes and signaling pathways of the mammary epi- thelial cells are significantly affected by the loss of SFRP1-expression. Using gene ontology analysis an association to BMP and Smoothened signaling cas- cades was identified in addition to the already known function of SFRP1 as a negative regulator of the WNT signaling pathway. Additionally, a multiplicity of genes has been identified, which showed differential expression patterns after SFRP1-reexpression such as BDNF, LY96, MMP1, ST18, BIN1 and RERGL. The BDNF gene was brought into the focus of this investigation as a potential target of the SFRP1-mediated signal transduction. A co-regulation of SFRP1 and BDNF could be identified as well within the the luminal-like SK-BR-3 as the basal-like BT-20 cells. Studying mRNA as well as protein expression in primary breast carcinomas a positive correlation of SFRP1 and BDNF-expression could be confirmed. Furthermore, patients with high SFRP1- and BDNF-expression showed a significantly extended overall survival. BDNF-overexpression in hu- man breast cancer cells resulted in a decreased proliferation rate. However, a change of other tumor-associated characteristics could not be observed after BDNF-reexpression. In this work the function of SFRP1 as a tumor suppressor was successfully highlighted on basis of different functional tests. Furthermore, cellular pathways and genes were identified that are affected by a loss of SFRP1-expression. 6 Abkurzungsverzeichnis¨ A Adenin ADH Atypische duktale Hyperplasie AMV Avian Myoblastosis Virus ATP Adenosintriphosphat BDNF brain-derived neurotrophic factor BIN1 bridging integrator 1 bp Basenpaare bzw. beziehungsweise
Recommended publications
  • Changes of Liver Transcriptome Profiles Following Oxidative Stress in Streptozotocin-Induced Diabetes in Mice
    Changes of liver transcriptome profiles following oxidative stress in streptozotocin-induced diabetes in mice Shuren Guo1, Xiaohuan Mao2, Yunmeng Yan1, Yan Zhang1 and Liang Ming1 1 Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China 2 Department of Clinical Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou Uni- versity, Zhengzhou, Henan, People's Republic of China ABSTRACT Background. Oxidative-stress (OS) was causal in the development of cell dysfunction and insulin resistance. Streptozotocin (STZ) was an alkylation agent that increased reactive oxygen species (ROS) levels. Here we aimed to explore the oxidative-stress and related RNAs in the liver of STZ-induced diabetic mice. Methods. RNA-sequencing was performed using liver tissues from STZ induced diabetic mice and controls. Pathway and Gene Ontology (GO) analyses were utilized to annotate the target genes. The differentially expressed RNAs involved in the peroxisome pathway were validated by qRT-PCR. The glucose metabolite and OS markers were measured in the normal control (NC) and STZ-induced diabetic mellitus (DM) group. Results. The levels of serum Fasting insulin, HbA1c, Malondialdehyde (MDA) and 8-iso-prostaglandin F2α (8-iso-PGF2α) were significant higher in DM groups than NC group, while SOD activity decreased significantly in DM groups. We found 416 lncRNAs and 910 mRNAs were differentially expressed in the STZ-induced diabetic mice compared to the control group. OS associated RNAs were differentially expressed in the liver of STZ-induced diabetic mice. Conclusion. This study confirmed that the OS was increased in the STZ-induced DM mice as evidenced by the increase of lipid peroxidation product MDA and 8-iso-PGF2α, Submitted 11 November 2019 identified aberrantly expressed lncRNAs and mRNAs in STZ-induced diabetic mice.
    [Show full text]
  • Proquest Dissertations
    Modeling Uncertainty in Data Integration for Improving Protein Function Assignment Brenton E. Louie A dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy University of Washington 2008 Program Authorized to Offer Degree: Medical Education and Biomedical Informatics UMI Number: 3318214 INFORMATION TO USERS The quality of this reproduction is dependent upon the quality of the copy submitted. Broken or indistinct print, colored or poor quality illustrations and photographs, print bleed-through, substandard margins, and improper alignment can adversely affect reproduction. In the unlikely event that the author did not send a complete manuscript and there are missing pages, these will be noted. Also, if unauthorized copyright material had to be removed, a note will indicate the deletion. ® UMI UMI Microform 3318214 Copyright 2008 by ProQuest LLC. All rights reserved. This microform edition is protected against unauthorized copying under Title 17, United States Code. ProQuest LLC 789 E. Eisenhower Parkway PO Box 1346 Ann Arbor, Ml 48106-1346 University of Washington Graduate School This is to certify that I have examined this copy of a doctoral dissertation by Brenton E. Louie and have found that it is complete and satisfactory in all respects, and that any and all revisions required by the final examining committee have been made. Chair of the Supervisory Committee: Peter Tar^zv-Hornocfey- h Reading Committee: &/&L f OMA^J "("fcf^^cM Peter Tarczy-Hornoch Dan Suciu V In presenting this dissertation in partial fulfillment of the requirements for the doctoral degree at the University of Washington, I agree that the Library shall make its copies freely available for inspection.
    [Show full text]
  • WO 2015/130968 A2 3 September 2015 (03.09.2015) P O P C T
    (12) INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PCT) (19) World Intellectual Property Organization International Bureau (10) International Publication Number (43) International Publication Date WO 2015/130968 A2 3 September 2015 (03.09.2015) P O P C T (51) International Patent Classification: Inc., 75 Francis Street, Boston, MA 021 15 (US). YOSEF, C12Q 1/68 (2006.01) Nir; 1520 Laurel Ave., Richmond, CA 94805 (US). (21) International Application Number: (74) Agents: KOWALSKI, Thomas J. et al; Vedder Price PCT/US20 15/0 17826 P.C., 1633 Broadway, New York, NY 1001 9 (US). (22) International Filing Date: (81) Designated States (unless otherwise indicated, for every 26 February 2015 (26.02.2015) kind of national protection available): AE, AG, AL, AM, AO, AT, AU, AZ, BA, BB, BG, BH, BN, BR, BW, BY, (25) Filing Language: English BZ, CA, CH, CL, CN, CO, CR, CU, CZ, DE, DK, DM, (26) Publication Language: English DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, HN, HR, HU, ID, IL, IN, IR, IS, JP, KE, KG, KN, KP, KR, (30) Priority Data: KZ, LA, LC, LK, LR, LS, LU, LY, MA, MD, ME, MG, 61/945,641 27 February 2014 (27.02.2014) US MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, (71) Applicants: THE BROAD INSTITUTE INC. [US/US]; PA, PE, PG, PH, PL, PT, QA, RO, RS, RU, RW, SA, SC, 415 Main Street, Cambridge, MA 02142 (US). THE SD, SE, SG, SK, SL, SM, ST, SV, SY, TH, TJ, TM, TN, BRIGHAM AND WOMEN'S HOSPITAL, INC.
    [Show full text]
  • Understanding Chronic Kidney Disease: Genetic and Epigenetic Approaches
    University of Pennsylvania ScholarlyCommons Publicly Accessible Penn Dissertations 2017 Understanding Chronic Kidney Disease: Genetic And Epigenetic Approaches Yi-An Ko Ko University of Pennsylvania, [email protected] Follow this and additional works at: https://repository.upenn.edu/edissertations Part of the Bioinformatics Commons, Genetics Commons, and the Systems Biology Commons Recommended Citation Ko, Yi-An Ko, "Understanding Chronic Kidney Disease: Genetic And Epigenetic Approaches" (2017). Publicly Accessible Penn Dissertations. 2404. https://repository.upenn.edu/edissertations/2404 This paper is posted at ScholarlyCommons. https://repository.upenn.edu/edissertations/2404 For more information, please contact [email protected]. Understanding Chronic Kidney Disease: Genetic And Epigenetic Approaches Abstract The work described in this dissertation aimed to better understand the genetic and epigenetic factors influencing chronic kidney disease (CKD) development. Genome-wide association studies (GWAS) have identified single nucleotide polymorphisms (SNPs) significantly associated with chronic kidney disease. However, these studies have not effectively identified target genes for the CKD variants. Most of the identified variants are localized to non-coding genomic regions, and how they associate with CKD development is not well-understood. As GWAS studies only explain a small fraction of heritability, we hypothesized that epigenetic changes could explain part of this missing heritability. To identify potential gene targets of the genetic variants, we performed expression quantitative loci (eQTL) analysis, using genotyping arrays and RNA sequencing from human kidney samples. To identify the target genes of CKD-associated SNPs, we integrated the GWAS-identified SNPs with the eQTL results using a Bayesian colocalization method, coloc. This resulted in a short list of target genes, including PGAP3 and CASP9, two genes that have been shown to present with kidney phenotypes in knockout mice.
    [Show full text]
  • Myopia in African Americans Is Significantly Linked to Chromosome 7P15.2-14.2
    Genetics Myopia in African Americans Is Significantly Linked to Chromosome 7p15.2-14.2 Claire L. Simpson,1,2,* Anthony M. Musolf,2,* Roberto Y. Cordero,1 Jennifer B. Cordero,1 Laura Portas,2 Federico Murgia,2 Deyana D. Lewis,2 Candace D. Middlebrooks,2 Elise B. Ciner,3 Joan E. Bailey-Wilson,1,† and Dwight Stambolian4,† 1Department of Genetics, Genomics and Informatics and Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, Tennessee, United States 2Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore, Maryland, United States 3The Pennsylvania College of Optometry at Salus University, Elkins Park, Pennsylvania, United States 4Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, United States Correspondence: Joan E. PURPOSE. The purpose of this study was to perform genetic linkage analysis and associ- Bailey-Wilson, NIH/NHGRI, 333 ation analysis on exome genotyping from highly aggregated African American families Cassell Drive, Suite 1200, Baltimore, with nonpathogenic myopia. African Americans are a particularly understudied popula- MD 21131, USA; tion with respect to myopia. [email protected]. METHODS. One hundred six African American families from the Philadelphia area with a CLS and AMM contributed equally to family history of myopia were genotyped using an Illumina ExomePlus array and merged this work and should be considered co-first authors. with previous microsatellite data. Myopia was initially measured in mean spherical equiv- JEB-W and DS contributed equally alent (MSE) and converted to a binary phenotype where individuals were identified as to this work and should be affected, unaffected, or unknown.
    [Show full text]
  • Supplementary Table S4. FGA Co-Expressed Gene List in LUAD
    Supplementary Table S4. FGA co-expressed gene list in LUAD tumors Symbol R Locus Description FGG 0.919 4q28 fibrinogen gamma chain FGL1 0.635 8p22 fibrinogen-like 1 SLC7A2 0.536 8p22 solute carrier family 7 (cationic amino acid transporter, y+ system), member 2 DUSP4 0.521 8p12-p11 dual specificity phosphatase 4 HAL 0.51 12q22-q24.1histidine ammonia-lyase PDE4D 0.499 5q12 phosphodiesterase 4D, cAMP-specific FURIN 0.497 15q26.1 furin (paired basic amino acid cleaving enzyme) CPS1 0.49 2q35 carbamoyl-phosphate synthase 1, mitochondrial TESC 0.478 12q24.22 tescalcin INHA 0.465 2q35 inhibin, alpha S100P 0.461 4p16 S100 calcium binding protein P VPS37A 0.447 8p22 vacuolar protein sorting 37 homolog A (S. cerevisiae) SLC16A14 0.447 2q36.3 solute carrier family 16, member 14 PPARGC1A 0.443 4p15.1 peroxisome proliferator-activated receptor gamma, coactivator 1 alpha SIK1 0.435 21q22.3 salt-inducible kinase 1 IRS2 0.434 13q34 insulin receptor substrate 2 RND1 0.433 12q12 Rho family GTPase 1 HGD 0.433 3q13.33 homogentisate 1,2-dioxygenase PTP4A1 0.432 6q12 protein tyrosine phosphatase type IVA, member 1 C8orf4 0.428 8p11.2 chromosome 8 open reading frame 4 DDC 0.427 7p12.2 dopa decarboxylase (aromatic L-amino acid decarboxylase) TACC2 0.427 10q26 transforming, acidic coiled-coil containing protein 2 MUC13 0.422 3q21.2 mucin 13, cell surface associated C5 0.412 9q33-q34 complement component 5 NR4A2 0.412 2q22-q23 nuclear receptor subfamily 4, group A, member 2 EYS 0.411 6q12 eyes shut homolog (Drosophila) GPX2 0.406 14q24.1 glutathione peroxidase
    [Show full text]
  • ITRAQ-Based Quantitative Proteomic Analysis of Processed Euphorbia Lathyris L
    Zhang et al. Proteome Science (2018) 16:8 https://doi.org/10.1186/s12953-018-0136-6 RESEARCH Open Access ITRAQ-based quantitative proteomic analysis of processed Euphorbia lathyris L. for reducing the intestinal toxicity Yu Zhang1, Yingzi Wang1*, Shaojing Li2*, Xiuting Zhang1, Wenhua Li1, Shengxiu Luo1, Zhenyang Sun1 and Ruijie Nie1 Abstract Background: Euphorbia lathyris L., a Traditional Chinese medicine (TCM), is commonly used for the treatment of hydropsy, ascites, constipation, amenorrhea, and scabies. Semen Euphorbiae Pulveratum, which is another type of Euphorbia lathyris that is commonly used in TCM practice and is obtained by removing the oil from the seed that is called paozhi, has been known to ease diarrhea. Whereas, the mechanisms of reducing intestinal toxicity have not been clearly investigated yet. Methods: In this study, the isobaric tags for relative and absolute quantitation (iTRAQ) in combination with the liquid chromatography-tandem mass spectrometry (LC-MS/MS) proteomic method was applied to investigate the effects of Euphorbia lathyris L. on the protein expression involved in intestinal metabolism, in order to illustrate the potential attenuated mechanism of Euphorbia lathyris L. processing. Differentially expressed proteins (DEPs) in the intestine after treated with Semen Euphorbiae (SE), Semen Euphorbiae Pulveratum (SEP) and Euphorbiae Factor 1 (EFL1) were identified. The bioinformatics analysis including GO analysis, pathway analysis, and network analysis were done to analyze the key metabolic pathways underlying the attenuation mechanism through protein network in diarrhea. Western blot were performed to validate selected protein and the related pathways. Results: A number of differentially expressed proteins that may be associated with intestinal inflammation were identified.
    [Show full text]
  • Non-Synonymous Single Nucleotide Polymorphisms in the P2X
    Int. J. Mol. Sci. 2014, 15, 13344-13371; doi:10.3390/ijms150813344 OPEN ACCESS International Journal of Molecular Sciences ISSN 1422-0067 www.mdpi.com/journal/ijms Review Non-Synonymous Single Nucleotide Polymorphisms in the P2X Receptor Genes: Association with Diseases, Impact on Receptor Functions and Potential Use as Diagnosis Biomarkers Emily A. Caseley 1,†, Stephen P. Muench 1, Sebastien Roger 2, Hong-Ju Mao 3, Stephen A. Baldwin 1 and Lin-Hua Jiang 1,4,†,* 1 School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK; E-Mails: [email protected] (E.A.C.); [email protected] (S.P.M.); [email protected] (S.A.B.) 2 Inserm U1069, University of Tours, Tours 37032, France; E-Mail: [email protected] 3 State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Science, Shanghai 200050, China; E-Mail: [email protected] 4 Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang 453003, China † These authors contributed equally to this work. * Author to whom correspondence should be addressed; E-Mail: [email protected]; Tel.: +44-0-113-343-4231. Received: 6 June 2014; in revised form: 10 July 2014 / Accepted: 14 July 2014 / Published: 30 July 2014 Abstract: P2X receptors are Ca2+-permeable cationic channels in the cell membranes, where they play an important role in mediating a diversity of physiological and pathophysiological functions of extracellular ATP. Mammalian cells express seven P2X receptor genes. Single nucleotide polymorphisms (SNPs) are widespread in the P2RX genes encoding the human P2X receptors, particularly the human P2X7 receptor.
    [Show full text]
  • Co-Stimulation of Purinergic P2X4 and Prostanoid EP3 Receptors Triggers Synergistic Degranulation in Murine Mast Cells
    International Journal of Molecular Sciences Article Co-Stimulation of Purinergic P2X4 and Prostanoid EP3 Receptors Triggers Synergistic Degranulation in Murine Mast Cells Kazuki Yoshida 1, Makoto Tajima 1, Tomoki Nagano 1, Kosuke Obayashi 1, Masaaki Ito 1, Kimiko Yamamoto 2 and Isao Matsuoka 1,* 1 Laboratory of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Gunma 370-0033, Japan; [email protected] (K.Y.); [email protected] (M.T.); [email protected] (T.N.); [email protected] (K.O.); [email protected] (M.I.) 2 Department of Biomedical Engineering, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan; [email protected] * Correspondence: [email protected]; Tel.: +81-27-352-1180 Received: 4 October 2019; Accepted: 16 October 2019; Published: 17 October 2019 Abstract: Mast cells (MCs) recognize antigens (Ag) via IgE-bound high affinity IgE receptors (Fc"RI) and trigger type I allergic reactions. Fc"RI-mediated MC activation is regulated by various G protein-coupled receptor (GPCR) agonists. We recently reported that ionotropic P2X4 receptor (P2X4R) stimulation enhanced Fc"RI-mediated degranulation. Since MCs are involved in Ag-independent hypersensitivity, we investigated whether co-stimulation with ATP and GPCR agonists in the absence of Ag affects MC degranulation. Prostaglandin E2 (PGE2) induced synergistic degranulation when bone marrow-derived MCs (BMMCs) were co-stimulated with ATP, while pharmacological analyses revealed that the effects of PGE2 and ATP were mediated by EP3 and P2X4R, respectively. Consistently, this response was absent in BMMCs prepared from P2X4R-deficient mice.
    [Show full text]
  • Produktinformation
    Produktinformation Diagnostik & molekulare Diagnostik Laborgeräte & Service Zellkultur & Verbrauchsmaterial Forschungsprodukte & Biochemikalien Weitere Information auf den folgenden Seiten! See the following pages for more information! Lieferung & Zahlungsart Lieferung: frei Haus Bestellung auf Rechnung SZABO-SCANDIC Lieferung: € 10,- HandelsgmbH & Co KG Erstbestellung Vorauskassa Quellenstraße 110, A-1100 Wien T. +43(0)1 489 3961-0 Zuschläge F. +43(0)1 489 3961-7 [email protected] • Mindermengenzuschlag www.szabo-scandic.com • Trockeneiszuschlag • Gefahrgutzuschlag linkedin.com/company/szaboscandic • Expressversand facebook.com/szaboscandic CSTL1 Antibody Product Code CSB-PA006101GA01HU Abbreviation CSTL1-Specific Storage Upon receipt, store at -20°C or -80°C. Avoid repeated freeze. Uniprot No. Q9H114 Immunogen Human CSTL1-Specific Raised In Rabbit Species Reactivity Human,Mouse Tested Applications ELISA,IHC Storage Buffer PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. -20°C, Avoid freeze / thaw cycles. Purification Method Antigen Affinity purified Isotype IgG Alias cystatin-like 1;CSTL1;RCET11;dJ322G13.4 ; Product Type Purified Rabbit Anti Human PolyClonal Antibody Species Homo sapiens (Human) Target Names CSTL1 Target Details The cystatin superfamily encompasses proteins that contain multiple cystatin- like sequences. Some of the members are active cysteine protease inhibitors, while others have lost or perhaps never acquired this inhibitory activity. There are three inhibitory families in the superfamily, including the type 1 cystatins (stefins), type 2 cystatins and the kininogens. The type 2 cystatin proteins are a class of cysteine proteinase inhibitors found in a variety of human fluids and secretions. The cystatin locus on chromosome 20 contains the majority of the type 2 cystatin genes and pseudogenes. This gene is located at the telomeric end of the cystatin locus and encodes a type 2 cystatin-like protein.
    [Show full text]
  • High-Fat Diet Alters the Retinal Transcriptome in the Absence of Gut Microbiota
    cells Article High-Fat Diet Alters the Retinal Transcriptome in the Absence of Gut Microbiota David Dao 1, Bingqing Xie 2,3 , Urooba Nadeem 4, Jason Xiao 1, Asad Movahedan 5, Mark D’Souza 2, Vanessa Leone 6,7 , Seenu M. Hariprasad 1, Eugene B. Chang 7, Dinanath Sulakhe 3 and Dimitra Skondra 1,* 1 Department of Ophthalmology and Visual Science, University of Chicago, Chicago, IL 60637, USA; [email protected] (D.D.); [email protected] (J.X.); [email protected] (S.M.H.) 2 Center for Research Informatics, University of Chicago, Chicago, IL 60637, USA; [email protected] (B.X.); [email protected] (M.D.) 3 Department of Medicine, University of Chicago, Chicago, IL 60637, USA; [email protected] 4 Department of Pathology, University of Chicago, Chicago, IL 60637, USA; [email protected] 5 Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT 06437, USA; [email protected] 6 Department of Animal Biologics and Metabolism, University of Wisconsin, Madison, WI 53706, USA; [email protected] 7 Knapp Center for Biomedical Discovery, Department of Medicine, Microbiome Medicine Program, University of Chicago, Chicago, IL 60637, USA; [email protected] * Correspondence: [email protected] Abstract: The relationship between retinal disease, diet, and the gut microbiome has shown increasing importance over recent years. In particular, high-fat diets (HFDs) are associated with development and progression of several retinal diseases, including age-related macular degeneration (AMD) and Citation: Dao, D.; Xie, B.; Nadeem, diabetic retinopathy. However, the complex, overlapping interactions between diet, gut microbiome, U.; Xiao, J.; Movahedan, A.; D’Souza, and retinal homeostasis are poorly understood.
    [Show full text]
  • Histone H3.3 and Its Proteolytically Processed Form Drive a Cellular Senescence Programme
    ARTICLE Received 21 Mar 2014 | Accepted 9 Sep 2014 | Published 14 Nov 2014 DOI: 10.1038/ncomms6210 Histone H3.3 and its proteolytically processed form drive a cellular senescence programme Luis F. Duarte1,2,3, Andrew R.J. Young4, Zichen Wang3,5, Hsan-Au Wu1,3, Taniya Panda1,2, Yan Kou3,5, Avnish Kapoor1,2,w, Dan Hasson1,2, Nicholas R. Mills1,2, Avi Ma’ayan5, Masashi Narita4 & Emily Bernstein1,2 The process of cellular senescence generates a repressive chromatin environment, however, the role of histone variants and histone proteolytic cleavage in senescence remains unclear. Here, using models of oncogene-induced and replicative senescence, we report novel histone H3 tail cleavage events mediated by the protease Cathepsin L. We find that cleaved forms of H3 are nucleosomal and the histone variant H3.3 is the preferred cleaved form of H3. Ectopic expression of H3.3 and its cleavage product (H3.3cs1), which lacks the first 21 amino acids of the H3 tail, is sufficient to induce senescence. Further, H3.3cs1 chromatin incorporation is mediated by the HUCA histone chaperone complex. Genome-wide transcriptional profiling revealed that H3.3cs1 facilitates transcriptional silencing of cell cycle regulators including RB/E2F target genes, likely via the permanent removal of H3K4me3. Collectively, our study identifies histone H3.3 and its proteolytically processed forms as key regulators of cellular senescence. 1 Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, New York 10029, USA. 2 Department of Dermatology, Icahn School of Medicine at Mount Sinai, One Gustave L.
    [Show full text]