Rnascope® Manual Reagents Gene Expression Analysis by RNA in Situ Hybridization

Total Page:16

File Type:pdf, Size:1020Kb

Rnascope® Manual Reagents Gene Expression Analysis by RNA in Situ Hybridization RNAscope® Manual Reagents Gene Expression Analysis by RNA In Situ Hybridization Get quantitative molecular detection with morphological context in a single assay The RNAscope® assay is the most advanced RNA in situ hybridization (ISH) methodology based on ACD’s patented technology, with signal EQTPMƤGEXMSRERHWMQYPXERISYWFEGOKVSYRHRSMWIWYTTVIWWMSRXLEX advances RNA analysis in tissues and cells. Unique to this technology, the RNAscope®EWWE]HIPMZIVWUYERXMXEXMZIWIRWMXMZIERHWTIGMƤGQSPIGYPEV detection of RNA species on a cell-by-cell basis with morphological context 2018 >650 publications in a single assay. This enables researchers to visualize which genes are expressed, localize where they are expressed, and quantify the level of expression. 2017 Research Areas >450 publications With more than 1,800 publications since 2011, researchers have embraced the RNAscope® technology in a multitude of research areas, showing the universality of the RNAscope® technology. With its unique features and unequal performances, the RNAscope® technology is an essential tool in any research 2016 and pre-clinical research laboratory. >300 publications • Cancer research 2015 • Stem cell >200 publications • Neuroscience • Immunotherapy 2014 • Xenograft models 95 publications • Infectious disease • Preclinical safety assessment 2013 • Gene and cell therapy 47 publications • And more... 2012 11 publications 2011 Advanced Cell Diagnostics 4 publications Publication Count 1,800+* *In November 2018 2 Advanced Cell Diagnostics Explore the RNAscope® World Principle and features of the RNAscope® Technology ...................................................4 A solution for common research challenges ...................................................................6 >650 publications RNAscope® in situ1ERYEP%WWE];SVOƥS[ .....................................................................8 Step 01. Permeabilize Step 04. Visualize 8MWWYIWIGXMSRWSVGIPPWEVIƤ\IHSRXSWPMHIWERH Each punctate dot signal represents a single pretreated with RNAscope® Pretreatment Kit to target RNA molecule and can be visualized YRQEWOXEVKIX62%ERHTIVQIEFMPM^IGIPPW with a standard microscopes. Pretreatment Reagents / pg. 8 pg. 14 Step 02. Hybridize Step 05. Quantify Double Z probe pools are hybridized 7MRKPIQSPIGYPIWMKREPWEVIUYERXMƤIHSRE to target RNA molecules. cell-by-cell basis by manual counting or automated image analysis with RNAscope® and HALOTM and other RNAscope® Target Probes / pg. 9 software. RNAScope® Control Probes / pg. 9 RNAscope® & HALOTM software/ pg. 14 Step 03. Amplify 7IUYIRXMEPL]FVMHM^EXMSRSJEQTPMƤIVW and labeled probe(s). RNAscope® Detection Reagents / pg. 10 Accessories / pg. 13 RNAscope® Manual Solutions 3 Principle and features of RNAscope® Technology Innovative solution for single RNA molecule detection ERHUYERXMƤGEXMSRMRWMRKPIGIPPW RNAscope® probe design RNAscope® probe hybridization and E Q T P M Ƥ G E X M S R SGGYVWEWEGEWGEHISJIZIRXW A standard target probe consists of a pool of 20 double Z Step 1: Hybridization of 20 ZZ probe pairs to the RNA target probes targeting a region of 1,000 bases. Each Z target probe Step 2:,]FVMHM^EXMSRSJXLITVIEQTPMƤIVXSXLIYTTIV contains three elements: The lower region is complementary regions of the Z probe pairs XSXLIXEVKIX62%ERHMWWIPIGXIHJSVXEVKIXWTIGMƤGL]FVMHM^E- tion and uniform hybridization properties. A spacer sequence Step 3: ,]FVMHM^EXMSRSJQYPXMTPIEQTPMƤIVWTIVTVIEQTPMƤIV PMROWXLIPS[IVVIKMSRXSERYTTIVVIKMSR8LIX[SEHNEGIRX Step 4: ,]FVMHM^EXMSRSJQYPXMTPIPEFIPIHTVSFIWTIVEQTPMƤIV upper regions from a double Z target probe forms a 28 base Serial hybridization events - 20 ZZ probe pairs, multiple FMRHMRKWMXIJSVXLITVIEQTPMƤIV EQTPMƤIVWQYPXMTPIPEFIPIHTVSFIWVIWYPXMRL]FVMHM^EXMSRSJ Two independent Z probes, designed as probe pairs, need to thousands of labeled probes per RNA target. hybridize to the target sequence in tandem in order to enable FMRHMRKSJXLITVIEQTPMƤIV %WMRKPI>TVSFIL]FVMHM^EXMSRSRXSERSRWTIGMƤG62%XEVKIX can happen, but the resulting hybridization of the pre-ampli- ƤIVSRXSXLIYTTIVVIKMSRSJEWMRKPI>[MPPFIYRWXEFPIERH therefore will be removed during the wash steps. This design IRWYVIWEPS[FEGOKVSYRHRSMWIPIZIP #ORNKƒGT Probe Design 2TGCORNKƒGTDKPFKPI site,14 bases Linker Target binding site 18-25 bases Z target probe design Complementary bases Hybridization requires 2TGCORNKƒGT double Z binding Complementary bases #ORNKƒECVKQP Hybridization RNA target 4 Advanced Cell Diagnostics RNAscope® signal detection &IRIƤXWSJXLI62%WGSTI® technology ŵ,MKLWIRWMXMZMX]8LIWIVMEPWMKREPEQTPMƤGEXMSR design increases sensitivity such that a single Labeled probes contain either a chromogenic enzyme or RNA molecule can be detected. EƥYSVSTLSVIWMKREPKIRIVEXMRKSRITYRGXEXIHSXTIV62% ŵ,MKLWTIGMƤGMX]Proprietary probe design target. (See page 14 - Visualize Step). IRWYVIWXEVKIXWTIGMƤGFMRHMRK[LMPIXLIHSYFPI ,]FVMHM^EXMSRSJSRP]XLVII>TVSFITEMVWMWWYJƤGMIRXXSSFXEMR >TVSFIHIWMKRTVIZIRXWWMKREPEQTPMƤGEXMSRSJ a detectable signal by a standard microscope. RSRWTIGMƤGL]FVMHM^EXMSR • Morphological context: Spatial resolution of gene expression in the complex tissue environmet creates a spatial map • Per-cell quantitation: High sensitivity combined with morphological context results in single-molecule detection at single-cell resolution. ŵ9RMZIVWEP;SVOWJSVZMVXYEPP]%2=KIRIJVSQ %2=WTIGMIWMR%2=XMWWYI #ORNKƒGT Labeled probe binding site Labeled probe 2TGCORNKƒGTDKPFKPI 2TGCORNKƒGT #ORNKƒECVKQP RNAscope® Manual Solutions 5 A solution for common research challenges 6ETMHZEPMHEXMSRSJFMSQEVOIVHMWGSZIV] RNA expression analysis to complement ;LIXLIV]SYEVIGLEVEGXIVM^MRKFMSQEVOIVWHMWGSZIVIH or combine with IHC-based protein by single-cell RNA sequencing, NGS, microarray or high analysis throughput qPCR, the RNAscope® XIGLRSPSK]MWEUYMGOERH )\EQMREXMSRSJTVSXIMREWEFMSQEVOIV[MXL IEW]XSSPXSYWIEGVSWWXLIHMJJIVIRXWXEKIWSJXLIFMSQEVOIV immunohistochemistry (IHC) technique is a widely used ® validation. With the RNAscope technology you have access and accepted approach for diagnosis, prognosis, and to unique gene expression information with morphological therapy development for clinical diseases. However, the context: digital RNA expression at single cell level in complex number of high quality and reliable antibodies is limited tissue structure Theses. and IHC is not without issues, and the use of sometimes TSSVP]GLEVEGXIVM^IHERXMFSHMIWERHMRWYJƤGMIRXSZIVEPP standardization often leads to questionable results. At the contrary, RNAscope® technology is based on probe HIWMKRIHXSFILMKLP]WTIGMƤGXSXLIXEVKIXERHVITVSHYGMFP] manufactured. With a unique and reproducible protocol, RNAscope® assay is an ideal solution to validate, supplement or combine with IHC. FIGURE 1. RNAscope®EWWE]EWMQTPIƤVWXƤPXIVMRXEVKIXZEPMHEXMSR 6 Advanced Cell Diagnostics Non-coding RNA expression -RXLIQSWXVIGIRXWXEXMWXMGWJVSQXLI+)2'3()TVSNIGX 8LIHMWGSZIV]SJETVIZMSYWP]YRORS[RYRMZIVWISJPRG62%W (v29, October 2018), the human genome contains 23,643 has created an unprecedented demand for effective RNA in non-coding RNA (ncRNA) genes, surpassing the number situL]FVMHM^EXMSRXSSPW9RPMOITVSXIMRGSHMRKKIRIWJSV[LMGL of protein-coding genes (19,940). Of the non-coding RNA immunohistochemistry (IHC) and RNA in situ hybridization are species, some 30% (7,577) are less than 200 bases long, GSQTPIQIRXEV]JSVQETTMRKKIRII\TVIWWMSRXSWTIGMƤGGIPPW termed as small non-coding RNA. They comprise of transfer in situ, lncRNA expression can only be investigated by RNA RNA (tRNA) and ribosomal RNA (rRNA), as well as RNAs in situ hybridization. The generally lower expression levels of such as snoRNAs, microRNAs, siRNAs, snRNAs, exRNAs lncRNAs than their protein coding counterparts demand the and piRNA. About 60% (16,066) of the non-coding RNAs are highest sensitivity from RNA in situ hybridization methods. longer than 200 bases and are operationally designated as The single-molecule sensitivity and rapid assay development long non-coding RNAs (lncRNAs).The functions of lncRNAs XMQI [IIOW SJ%'(ŭW62%WGSTI®XIGLRSPSK]QEOI are still characterized; their abundance and diversity add to RNAscope® ideally suited for localizing lncRNAs expression to the challenge. Some lncRNAs have been shown to regulate WTIGMƤGGIPPX]TIWERHWYFGIPPYPEVWXVYGXYVIW62%WGSTI® in gene expression through a diversity of mechanisms and situ assays will undoubtedly accelerate lncRNA research and TPE]MQTSVXERXVSPIWMRGLVSQEXMRQSHMƤGEXMSR ,38%-6 become an indispensable tool for lncRNA-based molecular transcriptional and post-transcriptional regulation (ZEB2). diagnostics. Dysregulation of lncRNA is being found to have relevance not only in tumorigenesis, but also to neurological, cardiovascular, developmental and other diseases. “ This technology allows us to directly visualize gene expression in the target 1A3 tissue of interest – for example, within the same sample we can tell whether gene overexpression occurs in benign prostate glands, high grade prostatic intraepithelial neoplasia (HGPIN – a pre-cancerous state) or prostate cancer.” Dr. Mehra, Clinical Assistant Professor of Pathology at Michigan Center for Translational Pathology Hs-PD1 Hs -TIM3 Hs-PDL1 CD45 (Ab) *-+96)7MRKPIGIPPGSI\TVIWWMSRTVSƤPIWSJGLIGOTSMRXQSPIGYPIWMR FIGURE 3. Non-coding PCA3 transcript detection in whole prostate tumor selected non-small cell lung cancer (NSCLC) cores. whole tissue section using RNAscope® technology. RNAscope® Manual Solutions 7 RNAscope® in situ Manual Assay ;SVOƥS[ERHEWWSGMEXIHTVSHYGXW Step 01. RNAscope® Pretreatment Reagents Permeabilize Optimized permeabilization for optimal target accessibility.
Recommended publications
  • Genetics of Familial Non-Medullary Thyroid Carcinoma (FNMTC)
    cancers Review Genetics of Familial Non-Medullary Thyroid Carcinoma (FNMTC) Chiara Diquigiovanni * and Elena Bonora Unit of Medical Genetics, Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; [email protected] * Correspondence: [email protected]; Tel.: +39-051-208-8418 Simple Summary: Non-medullary thyroid carcinoma (NMTC) originates from thyroid follicular epithelial cells and is considered familial when occurs in two or more first-degree relatives of the patient, in the absence of predisposing environmental factors. Familial NMTC (FNMTC) cases show a high genetic heterogeneity, thus impairing the identification of pivotal molecular changes. In the past years, linkage-based approaches identified several susceptibility loci and variants associated with NMTC risk, however only few genes have been identified. The advent of next-generation sequencing technologies has improved the discovery of new predisposing genes. In this review we report the most significant genes where variants predispose to FNMTC, with the perspective that the integration of these new molecular findings in the clinical data of patients might allow an early detection and tailored therapy of the disease, optimizing patient management. Abstract: Non-medullary thyroid carcinoma (NMTC) is the most frequent endocrine tumor and originates from the follicular epithelial cells of the thyroid. Familial NMTC (FNMTC) has been defined in pedigrees where two or more first-degree relatives of the patient present the disease in absence of other predisposing environmental factors. Compared to sporadic cases, FNMTCs are often multifocal, recurring more frequently and showing an early age at onset with a worse outcome. FNMTC cases Citation: Diquigiovanni, C.; Bonora, E.
    [Show full text]
  • Structural Genomics Approach to Investigate Deleterious Impact Of
    www.nature.com/scientificreports OPEN Structural genomics approach to investigate deleterious impact of nsSNPs in conserved telomere maintenance component 1 Arunabh Choudhury1,5, Taj Mohammad2,5, Nikhil Samarth3, Afzal Hussain4, Md. Tabish Rehman4, Asimul Islam2, Mohamed F. Alajmi4, Shailza Singh3 & Md. Imtaiyaz Hassan2* Conserved telomere maintenance component 1 (CTC1) is an important component of the CST (CTC1-STN1-TEN1) complex, involved in maintaining the stability of telomeric DNA. Several non- synonymous single-nucleotide polymorphisms (nsSNPs) in CTC1 have been reported to cause Coats plus syndrome and Dyskeratosis congenital diseases. Here, we have performed sequence and structure analyses of nsSNPs of CTC1 using state-of-the-art computational methods. The structure- based study focuses on the C-terminal OB-fold region of CTC1. There are 11 pathogenic mutations identifed, and detailed structural analyses were performed. These mutations cause a signifcant disruption of noncovalent interactions, which may be a possible reason for CTC1 instability and consequent diseases. To see the impact of such mutations on the protein conformation, all-atom molecular dynamics (MD) simulations of CTC1-wild-type (WT) and two of the selected mutations, R806C and R806L for 200 ns, were carried out. A signifcant conformational change in the structure of the R806C mutant was observed. This study provides a valuable direction to understand the molecular basis of CTC1 dysfunction in disease progression, including Coats plus syndrome. Unlike prokaryotic chromosomes, eukaryotic chromosomes are linear and are much larger in size. Te ends of the eukaryotic chromosome are composed of a specialized protein-DNA complex called telomeres which maintains the stability of the chromosome ends1.
    [Show full text]
  • Structural and Functional Consequences of a Disease Mutation in the Telomere Protein TPP1
    Structural and functional consequences of a disease mutation in the telomere protein TPP1 Kamlesh Bishta,1, Eric M. Smitha,b,1, Valerie M. Tesmera, and Jayakrishnan Nandakumara,b,2 aDepartment of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109; and bProgram in Chemical Biology, University of Michigan, Ann Arbor, MI 48109 Edited by Joachim Lingner, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland, and accepted by Editorial Board Member Dinshaw J. Patel September 29, 2016 (received for review April 8, 2016) Telomerase replicates chromosome ends to facilitate continued cell The discovery of the TEL patch of TPP1 prompted the pre- division. Mutations that compromise telomerase function result in diction that this region could be a hotspot for mutations that stem cell failure diseases, such as dyskeratosis congenita (DC). One cause telomerase-deficiency diseases, such as DC. We recently such mutation (K170Δ), residing in the telomerase-recruitment factor reported a case of a severe variant of DC, Hoyeraal–Hreidarsson TPP1, provides an excellent opportunity to structurally, biochemi- syndrome (23), in which the proband was heterozygous for a cally, and genetically dissect the mechanism of such diseases. We deletion of a single amino acid of the TPP1 protein, namely lysine ACD show through site-directed mutagenesis and X-ray crystallography 170 (K170) (24). Our study placed the gene coding for TPP1 DKC1 TERC TERT that this TPP1 disease mutation deforms the conformation of two protein on a list with 10 other genes ( , , , RTEL1 TINF2 CTC1 NOP10 NHP2 WRAP53 PARN critical amino acids of the TEL [TPP1’s glutamate (E) and leucine-rich , , , , , ,and ) that are found mutated in DC and other telomere-related dis- (L)] patch, the surface of TPP1 that binds telomerase.
    [Show full text]
  • REVIEW Telomeres and Telomerase in Adrenocortical Tissue Maintenance
    131 REVIEW Telomeres and telomerase in adrenocortical tissue maintenance, carcinogenesis, and aging Tobias Else Division of Metabolism, Endocrinology and Diabetes, MEND/Department of Internal Medicine, University of Michigan Health System, 1860 BSRB, 109 Zina Pitcher Pl, Ann Arbor, Michigan 48109-2200, USA (Correspondence should be addressed to T Else; Email: [email protected]) Abstract Telomere dysfunction and telomere maintenance mechanisms contribute to major steps of carcinogenesis. Dysfunctional telomeres lead to the generation of genomic aberrations, such as amplifications and deletions. Telomere maintenance mechanisms, such as telomerase activity and alternative telomere lengthening, provide the basis of malignant cell expansion independent of telomere shortening-induced apoptosis or senescence, ensuring tumor survival. Recent advances highlight the importance of these mechanisms in adrenocortical carcinogenesis. In this review, we will summarize the main models of telomere physiology and their impact on adrenocortical tissue maintenance, aging, and carcinogenesis. Journal of Molecular Endocrinology (2009) 43, 131–141 Overview cycles (BFBs). An indefinite potential of expansion of malignant cells can be secured by the acquisition Mammalian chromosomes end in a stretch of TTAGGG of telomere length maintenance mechanisms (TMMs), nucleotide repeats known as telomeres. There are two which prevent telomere shortening, either telomerase key challenges inherent to these structures. First, activity (TA)-dependent or TA-independent alternative telomeres need to be protected from being recognized mechanisms of telomere lengthening (ALT). as a form of damaged DNA by the DNA surveillance and Here, we will give insight into the current under- from being processed by the DNA repair machinery. standing of how these problems are solved physio- Second, the semi-conservative DNA replication logically and how they are exploited by malignant cells, machinery does not entirely copy linear chromosomes, specifically those of adrenocortical origin.
    [Show full text]
  • Downregulation of Telomerase Maintenance-Related ACD Expression in Patients Undergoing Immunosuppresive Therapy Following Kidney Transplantation
    2224 EXPERIMENTAL AND THERAPEUTIC MEDICINE 10: 2224-2230, 2015 Downregulation of telomerase maintenance-related ACD expression in patients undergoing immunosuppresive therapy following kidney transplantation AGNIESZKA WITKOWSKA1, BARBARA STRZALKA-MROZIK2, ALEKSANDER OWCZAREK3, JOANNA GOLA2, URSZULA MAZUREK2, WLADYSLAW GRZESZCZAK1 and JANUSZ GUMPRECHT1 1Department of Internal Medicine, Diabetology and Nephrology, Medical University of Silesia, 41-800 Zabrze, Silesia; 2Department of Molecular Biology; 3Division of Statistics, Medical University of Silesia, 41-200 Sosnowiec, Silesia, Poland Received August 30, 2014; Accepted July 23, 2015 DOI: 10.3892/etm.2015.2785 Abstract. Chronic administration of immunosuppressants gene transcription, and thus TPP1 protein expression, may has been associated with long-term consequences, including enhance the capacity for cell immortalization, despite normal a higher risk of neoplasm development. The processes regu- levels of other key telomere maintenance factors, in patients lating telomere function exert a major influence on human undergoing immunosuppressive therapy. Furthermore, the cancer biology. The present study aimed to assess the effect of results indicate that TPP1 has potential for use as an early immunosuppressive therapy on the expression of genes asso- clinical marker and/or therapeutic target for cancer in patients ciated with telomere maintenance and protection in patients following organ transplantation. following renal transplantation. A total of 51 patients that had undergone kidney
    [Show full text]
  • Structural and Functional Consequences of a Disease Mutation in the Telomere Protein TPP1
    Structural and functional consequences of a disease mutation in the telomere protein TPP1 Kamlesh Bishta,1, Eric M. Smitha,b,1, Valerie M. Tesmera, and Jayakrishnan Nandakumara,b,2 aDepartment of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109; and bProgram in Chemical Biology, University of Michigan, Ann Arbor, MI 48109 Edited by Joachim Lingner, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland, and accepted by Editorial Board Member Dinshaw J. Patel September 29, 2016 (received for review April 8, 2016) Telomerase replicates chromosome ends to facilitate continued cell The discovery of the TEL patch of TPP1 prompted the pre- division. Mutations that compromise telomerase function result in diction that this region could be a hotspot for mutations that stem cell failure diseases, such as dyskeratosis congenita (DC). One cause telomerase-deficiency diseases, such as DC. We recently such mutation (K170Δ), residing in the telomerase-recruitment factor reported a case of a severe variant of DC, Hoyeraal–Hreidarsson TPP1, provides an excellent opportunity to structurally, biochemi- syndrome (23), in which the proband was heterozygous for a cally, and genetically dissect the mechanism of such diseases. We deletion of a single amino acid of the TPP1 protein, namely lysine ACD show through site-directed mutagenesis and X-ray crystallography 170 (K170) (24). Our study placed the gene coding for TPP1 DKC1 TERC TERT that this TPP1 disease mutation deforms the conformation of two protein on a list with 10 other genes ( , , , RTEL1 TINF2 CTC1 NOP10 NHP2 WRAP53 PARN critical amino acids of the TEL [TPP1’s glutamate (E) and leucine-rich , , , , , ,and ) that are found mutated in DC and other telomere-related dis- (L)] patch, the surface of TPP1 that binds telomerase.
    [Show full text]
  • POT1 Polyclonal Antibody Catalog # AP72003
    10320 Camino Santa Fe, Suite G San Diego, CA 92121 Tel: 858.875.1900 Fax: 858.622.0609 POT1 Polyclonal Antibody Catalog # AP72003 Specification POT1 Polyclonal Antibody - Product Information Application WB Primary Accession Q9NUX5 Reactivity Human, Monkey Host Rabbit Clonality Polyclonal POT1 Polyclonal Antibody - Additional Information Gene ID 25913 Other Names POT1; Protection of telomeres protein 1; hPot1; POT1-like telomere end-binding protein Dilution WB~~Western Blot: 1/500 - 1/2000. ELISA: 1/10000. Not yet tested in other applications. Format Liquid in PBS containing 50% glycerol, 0.5% BSA and 0.02% sodium azide. Storage Conditions -20℃ POT1 Polyclonal Antibody - Protein Information Name POT1 Function Component of the telomerase ribonucleoprotein (RNP) complex that is essential for the replication of chromosome termini. Is a component of the POT1 Polyclonal Antibody - Background double-stranded telomeric DNA-binding TRF1 complex which is involved in the Component of the telomerase regulation of telomere length by cis- ribonucleoprotein (RNP) complex that is inhibition of telomerase. Also acts as a essential for the replication of chromosome single-stranded telomeric DNA- binding termini. Is a component of the double-stranded protein and thus may act as a downstream telomeric DNA- binding TRF1 complex which is effector of the TRF1 complex and may involved in the regulation of telomere length transduce information about telomere Page 1/2 10320 Camino Santa Fe, Suite G San Diego, CA 92121 Tel: 858.875.1900 Fax: 858.622.0609 maintenance and/or length to the telomere by cis-inhibition of telomerase. Also acts as a terminus. Component of the shelterin single-stranded telomeric DNA-binding protein complex (telosome) that is involved in the and thus may act as a downstream effector of regulation of telomere length and the TRF1 complex and may transduce protection.
    [Show full text]
  • ACD (Human) IP-WB Antibody Pair Gene on Chromosome 11, Which Encodes Tripeptidyl-Peptidase I
    ACD (Human) IP-WB Antibody Pair gene on chromosome 11, which encodes tripeptidyl-peptidase I. [provided by RefSeq] Catalog Number: H00065057-PW2 Regulation Status: For research use only (RUO) Product Description: This IP-WB antibody pair set comes with one antibody for immunoprecipitation and another to detect the precipitated protein in western blot. Reactivity: Human Applications: IP-WB (See our web site product page for detailed applications information) Protocols: See our web site at http://www.abnova.com/support/protocols.asp or product page for detailed protocols Supplied Product: Antibody pair set content: 1. Antibody pair for IP: rabbit polyclonal anti-ACD (300 ul) 2. Antibody pair for WB: mouse purified polyclonal anti-ACD (50 ug) Storage Instruction: Store reagents of the antibody pair set at -20°C or lower. Please aliquot to avoid repeated freeze thaw cycle. Reagents should be returned to -20°C storage immediately after use. Entrez GeneID: 65057 Gene Symbol: ACD Gene Alias: PIP1, PTOP, TINT1, TPP1 Gene Summary: This gene encodes a protein that is involved in telomere function. This protein is one of six core proteins in the telosome/shelterin telomeric complex, which functions to maintain telomere length and to protect telomere ends. Through its interaction with other components, this protein plays a key role in the assembly and stabilization of this complex, and it mediates the access of telomerase to the telomere. Multiple transcript variants encoding different isoforms have been found for this gene. This gene, which is also referred to as TPP1, is distinct from the unrelated TPP1 Page 1/1 Powered by TCPDF (www.tcpdf.org).
    [Show full text]
  • Shelterin Genes, Germ Line Mutations and Chronic Lymphocytic Leukemia
    71 Commentary Shelterin genes, germ line mutations and chronic lymphocytic leukemia Irma Slavutsky Laboratorio de Genética de las Neoplasias Linfoides, Instituto de Medicina Experimental (IMEX), CONICET-Academia Nacional de Medicina, Buenos Aires, Argentina Correspondence to: Irma Slavutsky, MD, PhD. Laboratorio de Genética de Neoplasias Linfoides, Instituto de Medicina Experimental, CONICET- Academia Nacional de Medicina, J.A. Pacheco de Melo 3081, C1425AUM-Buenos Aires, Argentina. Email: [email protected]. Comment on: Speedy HE, Kinnersley B, Chubb D, et al. Germline mutations in shelterin complex genes are associated with familial chronic lymphocytic leukemia. Blood 2016. [Epub ahead of print]. Submitted Jan 06, 2017. Accepted for publication Jan 17, 2017. doi: 10.21037/tcr.2017.02.36 View this article at: http://dx.doi.org/10.21037/tcr.2017.02.36 Telomeres are distinctive DNA-protein structures that studies support a dual role for ACD acting in telomere cap the ends of linear chromosomes; they are essential protection preventing DNA damage response and telomere to maintain chromosomal integrity and genome stability. maintenance, preserving telomere function (4). This Telomeres are composed by tandem repeats of the non- member of the shelterin complex also interacts with ataxia codificante DNA sequence TTAGGG bound by the telangiectasia mutated pathway, thus alterations in ACD may shelterin complex. It contains six core proteins: telomeric stimulate chromosomal instability and large accumulation of repeat binding factor 1 (TERF1), TERF2, protection of mutations leading to cancer development (5). In contrast to the telomeres 1 (POT1), adrenocortical dysplasia homolog other shelterin components, TERF2IP is not a protective (ACD), telomeric repeat-binding factor 2-interacting protein, but prevents telomere recombination and fragility.
    [Show full text]
  • Occurrence, Functionality, and Abundance of the TERT Promoter Mutations
    bioRxiv preprint doi: https://doi.org/10.1101/2021.05.03.442397; this version posted June 14, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Occurrence, functionality, and abundance of the TERT promoter mutations Sivaramakrishna Rachakonda1, Jörg D. Hoheisel1, and Rajiv Kumar1,2* 1Division of Functional Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany 2Department of Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska, Prague, Czech Republic Keywords: Telomeres, telomerase, TERT promoter mutations, cancers Abbreviations: ACD: Adrenocortical Dysplasia Protein Homolog (unofficial name TPP1); ARF: A-Raf Proto-Oncogene Serine/Threonine-Protein Kinase; ARID2: AT-Rich Interaction Domain 2; ATAC: Assay for Transposase-Accessible Chromatin; BRAF: B-Raf Proto-Oncogene, Serine/Threonine Kinase; CDKN2A: Cyclin-Dependent Kinase Inhibitor 2A; CRAF: C-Raf Proto- Oncogene, Serine/Threonine Kinase; CST: CTC1-STN1-TEN1 complex; CTNNB1: Catenin Beta 1; ENCODE: Encyclopedia of DNA Elements; ELF: E74 Like ETS Transcription Factor; ERK: Extracellular Signal-Regulated Kinase; ETS: E26 Transformation-specific; ETS1: ETS Proto-Oncogene 1, Transcription Factor; ETS2: ETS Proto-Oncogene 2, Transcription Factor; ETV5: ETS Variant Transcription Factor 5; GABPA: GA Binding Protein Transcription Factor Subunit Alpha; GABPB1: GA Binding Protein Transcription Factor Subunit
    [Show full text]
  • A Cell-Specific Regulatory Region of the Human ABO Blood Group Gene
    www.nature.com/scientificreports OPEN A cell‑specifc regulatory region of the human ABO blood group gene regulates the neighborhood gene encoding odorant binding protein 2B Rie Sano1*, Yoichiro Takahashi1, Haruki Fukuda1, Megumi Harada1, Akira Hayakawa1, Takafumi Okawa1, Rieko Kubo1, Haruo Takeshita2, Junichi Tsukada3 & Yoshihiko Kominato1 The human ABO blood group system is of great importance in blood transfusion and organ transplantation. ABO transcription is known to be regulated by a constitutive promoter in a CpG island and regions for regulation of cell‑specifc expression such as the downstream + 22.6‑kb site for epithelial cells and a site in intron 1 for erythroid cells. Here we investigated whether the + 22.6‑kb site might play a role in transcriptional regulation of the gene encoding odorant binding protein 2B (OBP2B), which is located on the centromere side 43.4 kb from the + 22.6‑kb site. In the gastric cancer cell line KATOIII, quantitative PCR analysis demonstrated signifcantly reduced amounts of OBP2B and ABO transcripts in mutant cells with biallelic deletions of the site created using the CRISPR/Cas9 system, relative to those in the wild‑type cells, and Western blotting demonstrated a corresponding reduction of OBP2B protein in the mutant cells. Moreover, single‑molecule fuorescence in situ hybridization assays indicated that the amounts of both transcripts were correlated in individual cells. These fndings suggest that OBP2B could be co‑regulated by the + 22.6‑kb site of ABO. Te human ABO blood group system is of great importance in blood transfusion and organ transplantation. Te carbohydrate structures of ABO blood group antigens are produced by the A- and B-transferases encoded by the A and B alleles, respectively1.
    [Show full text]
  • Functional Analysis of ACD Mutations in Dyskeratosis Congenita By
    Functional Analysis of ACD Mutations in Dyskeratosis Congenita by Hande Koçak A dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy (Human Genetics) in The University of Michigan 2014 Doctoral Committee: Associate Professor Catherine E. H. Keegan, Chair Professor Sally A. Camper Professor Thomas W. Glover Assistant Professor Jayakrishnan Nandakumar Associate Professor JoAnn M. Sekiguchi © Hande Koçak 2014 DEDIIN To My wonderful parents, Hatice and Basri Yücel Koçak and My wonderful sister, Emine Koçak We made it. ii ACKNOWLEDGEMENTS Foremost, I would like to express my appreciation and thanks to my PhD advisor, Catherine Keegan, for her encouragement, support, and patience. I am sincerely thankful for her research insight and guidance throughout my graduate studies. I am very grateful that Jayakrishnan Nandakumar agreed to become involved in my project and let me spend time in his lab. It is difficult for me to list all that I have learned from him and to express how thankful I am for his help. I cannot find words to express my gratitude to Jayakrishnan Nandakumar, JoAnn Sekiguchi, Thomas Glover and Sally Camper for serving on my thesis committee. They have always been very helpful and supportive. Their advices have kept me focused and goal-oriented during my studies. I have been very lucky to have them in my thesis committee. My special thanks go to JoAnn Sekiguchi for sparing time for me week after week to discuss my data and even just to chat about my concerns. I would like to thank all the former and current members of Keegan Lab.
    [Show full text]