Noncoding RNA Therapeutics — Challenges and Potential Solutions

Total Page:16

File Type:pdf, Size:1020Kb

Noncoding RNA Therapeutics — Challenges and Potential Solutions PERSPECTIVES siRNAs or ASOs that influence only a single target gene. Noncoding RNA therapeutics — The diverse functional repertoire of lncRNAs reveals various opportunities for challenges and potential solutions their therapeutic targeting, the means of which need to be adjusted to the mode of Melanie Winkle, Sherien M. El-Daly, Muller Fabbri and George A. Calin action of the lncRNA. LncRNA targeting may include transcriptional inhibition, Abstract | Therapeutic targeting of noncoding RNAs (ncRNAs), such as microRNAs post-transcriptional inhibition, steric (miRNAs) and long noncoding RNAs (lncRNAs), represents an attractive approach hindrance of secondary structure formation for the treatment of cancers, as well as many other diseases. Over the past decade, or protein interactions, introduction of substantial effort has been made towards the clinical application of RNA-based synthetic (for example, circular) lncRNAs, and modulation of lncRNA genomic loci or therapeutics, employing mostly antisense oligonucleotides and small interfering expression patterns via CRISPR–Cas9 and RNAs, with several gaining FDA approval. However, trial results have so far been CRISPR–Cas13, respectively (reviewed in ambivalent, with some studies reporting potent effects whereas others REF.17). An interesting development is the demonstrated limited efficacy or toxicity. Alternative entities such as antimiRNAs exploration of natural antisense transcripts are undergoing clinical testing, and lncRNA-based therapeutics are gaining (NATs): lncRNAs that are transcribed in interest. In this Perspective, we discuss key challenges facing ncRNA therapeutics the antisense direction to coding genes, and negatively regulate them in cis. ASOs — including issues associated with specificity, delivery and tolerability — and focus that target NATs, termed ‘antagoNATs’ on promising emerging approaches that aim to boost their success. have shown very promising preclinical results for gene reactivation in the central Noncoding RNAs (ncRNAs) are generated (TABle 1), aiming at gene modifications nervous system. AntagoNATs successfully from the larger part of the genome that in liver, muscle or the central nervous upregulated brain-derived neurotrophic does not encode proteins but produces system. All these therapeutics are either factor (BDNF), a protein highly involved in noncoding transcripts that regulate gene siRNAs or ASOs that cause specific gene memory formation18, as well as the healthy expression and protein function. The two downregulation, or ASOs that target allele of SCN1A, the haploinsufficiency major classes of ncRNA are the well-studied pre-mRNA splicing (that is, inducing of which causes the brain disorder Dravet short microRNAs (miRNAs) and the exon skipping or inclusion). In addition, syndrome19. Notably, BDNF-AS-targeting more recently identified long ncRNAs a plethora of RNA therapeutics are in antagoNATs were successfully delivered (lncRNAs) (BOx 1). Deregulation of both phase II or III clinical development, including across the blood–brain barrier using a types of transcript has been linked to every newer entities such as miRNA mimics and minimally invasive nasaldepot (MIND) in a cancer investigated to date and affects all antimiRs (TABle 2), but no lncRNA-based mouse model. MIND is aimed at directing major cancer hallmarks1–5. In addition, they therapeutics have entered the clinic. drug delivery to the olfactory submucosal have been linked to complex biological The use of miRNA-based therapeutics space and achieved approximately 40% processes such as immune cell development has dual advantages10,13,14. First, miRNAs are efficacy compared with riskier invasive and function, immune disorders6, neural naturally occurring molecules in human delivery techniques20. Such promising development and neurological diseases7–9. cells, unlike man-made chemotherapy developments suggest that the entrance of Therapeutic targeting of such naturally compounds or ASOs, and therefore have lncRNA-based therapeutics into clinical occurring ncRNAs thus represents a very all the mechanisms in place for their testing is imminent. promising approach for the treatment of processing and downstream target selection The translation of all RNA-based various diseases. (Fig. 1). Second, miRNAs act by targeting therapeutics into the clinic has been Various RNA-based therapies, including multiple genes within one pathway, thus hampered by issues associated with specificity, antisense oligonucleotides (ASOs), small causing a broader yet specific response. delivery and tolerability. Specificity issues interfering RNAs (siRNAs), short hairpin The miR-15–miR-16 cluster, downregulating include undesired on-target effects due RNAs (shRNAs), ASO anti-microRNAs multiple anti-apoptotic factors including to uptake in cells other than the cells of (antimiRs), miRNA mimics, miRNA BCL-2 and MCL1, represents an excellent interest, or off-target effects caused by sponges, therapeutic circular RNAs example of a miRNA acting at multiple either sequence similarities or overdosing (circRNAs) and CRISPR–Cas9-based levels to affect the same cancer hallmark15,16. to levels much higher than expected gene editing have been developed and The use or targeting of naturally occurring endogenously. Delivery issues are related to several excellent reviews describe these miRNAs could therefore represent a three major points: the instability of ‘naked’, agents10–12 (BOx 2). Currently, 11 RNA-based promising alternative to existing RNA-based chemically unmodified RNA structures; therapeutics are approved by the FDA and/or therapies and may potentially boost their inefficient intracellular delivery, which the European Medicines Agency (EMA) therapeutic effects compared with synthetic requires the exploitation of endosomal NATURE REVIEWS | DRUG DISCOVERY VOLUME 20 | AUGUST 2021 | 629 0123456789();: PERSPECTIVES Box 1 | Main characteristics of microRNAs and long noncoding RNAs siRNA was first shown not to trigger an interferon response in animals30, intraocular MicroRNAs (miRNAs) are highly conserved, small, 17- to 25-nucleotide (nt), single-stranded injections of anti-vascular endothelial ncRNAs that act as gene regulators. Their biogenesis is a multistep process (see Fig. 1) involving, growth factor receptor 1 (VEGFR1) siRNAs first, the production of long primary miRNA transcripts (pri-miRNA) by RNA polymerases II and III; bevasiranib (NCT00499590) and AGN second, the processing of pri-miRNAs by the nuclear ribonuclease Drosha and DGCR8 into a 70 nt long precursor miRNA (pre-miRNA) with a stem–loop structure; third, the nuclear export of 211745 (NCT00395057) were later shown pre-miRNAs by exportin 5 and Ran-GTPase; and fourth, the cleavage of pre-miRNAs by RNase III to owe their anti-angiogenic effects to direct enzyme Dicer to yield a mature, double-stranded miRNA. Gene regulation is mediated through the TLR3 stimulation instead of to the desired unwinding of the miRNA by RNA helicase and the incorporation of the miRNA guide strand into silencing effect31, resulting in termination the RNA-induced silencing complex (RISC)274–277. Post-transcriptional gene silencing is mediated of clinical development (TABle 3). The via nucleotide complementarity to mainly the 3′ UTR, or less prevalently the 5′ UTR or coding predominant pathway recognizing RNA region, of target mRNAs278,279. The miRNA ‘seed sequence’ is situated from nucleotides 2 to 7 at therapeutics is TLR signalling, which is the 5′-end of the miRNA sequence. Seed sequence binding with perfect complementarity results mediated through myeloid differentiation in the deadenylation and degradation of the targeted mRNA, whereas binding with imperfect factor 88 (MyD88) and activates various complementarity, which is more common, results in translational inhibition, both of which are pathways, resulting in nuclear factor-κB facilitated by RISC. (NF-κB) activation and the production of Long noncoding RNAs (lncRNAs) are larger transcripts (>200 nt in size) that are synthesized similarly to mRNAs, but not translated into protein280. LncRNAs contain two types of functional pro-inflammatory cytokines (IL-6, IL-8, element, the interactor elements involved in direct physical interaction with other nucleic acids, IL-12 and TNF), or a type I interferon with proteins or lipids, and the structural elements, leading to the occurrence of secondary response, which ultimately leads to the and/or tertiary 3D RNA structures, which direct their functional interactions281. It is this capacity activation of diverse downstream immune to interact with DNA and RNA as well as proteins via base pairing in linear form or chemical responses32. TLR7 and TLR8 stimulation by interactions in secondary structures, that allows lncRNAs to function in more variable ways than miRNA was shown to be dependent on the miRNAs. Gene-regulatory roles have been identified for many lncRNAs, for example, by influencing presence of GU-rich sequences (for example, transcription factor binding or epigenetic marks. Interactions with mRNAs may influence their 5′-UGUGU-3′ or 5′-GUCCUUCAA-3′) stability or rate of translation. Similarly, lncRNA–protein interactions may influence the stability, and caused a IFNα-mediated inflammatory activity or localization of the protein282,283. Furthermore, circular RNAs, which have a similar length range to lncRNAs, have become known for their potent roles as miRNA sponges179,284. response, activation of NF-κB and production of inflammatory cytokines (for example, IL-6 and TNF)
Recommended publications
  • A Phase II Study of the Novel Proteasome Inhibitor Bortezomib In
    Memorial Sloan-Kettering Cance r Center IRB Protocol IRB#: 05-103 A(14) A Phase II Study of the Novel Proteas ome Inhibitor Bortezomib in Combination with Rituximab, Cyclophosphamide and Prednisone in Patients with Relapsed/Refractory I Indolent B-cell Lymphoproliferative Disorders and Mantle Cell Lymphoma (MCL) MSKCC THERAPEUTIC/DIAGNOSTIC PROTOCOL Principal Investigator: John Gerecitano, M.D., Ph.D. Co-Principal Carol Portlock, M.D. Investigator(s): IFormerly: A Phase I/II Study of the Nove l Proteasome Inhibitor Bortezomib in Combinati on with Rituximab, Cyclophosphamide and Prednisone in Patients with Relapsed/Refractory Indolent B-cell Lymphoproliferative Disorders and Mantle Cell Lymphoma (MCL) Amended: 07/25/12 Memorial Sloan-Kettering Cance r Center IRB Protocol IRB#: 05-103 A(14) Investigator(s): Paul Hamlin, M.D. Commack, NY Steven B. Horwitz, M.D. Philip Schulman, M.D. Alison Moskowitz, M.D. Stuart Lichtman, M.D Craig H. Moskowitz, M.D. Stefan Berger, M.D. Ariela Noy, M.D. Julie Fasano, M.D. M. Lia Palomba, M.D., Ph.D. John Fiore, M.D. Jonathan Schatz, M.D. Steven Sugarman, M.D David Straus, M.D. Frank Y. Tsai, M.D. Andrew D. Zelenetz, M.D., Ph.D. Matthew Matasar, M.D Rockville Center, NY Mark L. Heaney, M.D., Ph.D. Pamela Drullinksy, M.D Nicole Lamanna, M.D. Arlyn Apollo, M.D. Zoe Goldberg, M.D. Radiology Kenneth Ng, M.D. Otilia Dumitrescu, M.D. Tiffany Troso-Sandoval, M.D. Andrei Holodny, M.D. Sleepy Hollow, NY Nuclear Medicine Philip Caron, M.D. Heiko Schoder, M.D. Michelle Boyar, M.D.
    [Show full text]
  • (12) United States Patent (10) Patent No.: US 8,580,814 B2 Adelman Et Al
    USOO858O814B2 (12) United States Patent (10) Patent No.: US 8,580,814 B2 Adelman et al. (45) Date of Patent: *Nov. 12, 2013 (54) METHODS OF USING 5,528,823. A 6/1996 Rudy, Jr. et al. (+)-1,4-DIHYDRO-7-(3S4S)-3-METHOXY-4- 3.43wk A E. E. St.Ola tala (METHYLAMINO)-1-PYRROLIDINYL-4- 6,171,857 B1 1/2001 Hendrickson OXO-1-(2-THIAZOLYL)-1.8- 6,291643 B1 9/2001 Zou et al. NAPHTHYRIDNE-3-CARBOXYLIC ACID 6,570,002 B1 5/2003 Hardwicket al. FORTREATMENT OF CANCER 6,641,810 B2 11/2003 Gold 6,641,833 B2 * 1 1/2003 Dang ............................ 424/426 (75) Inventors: Daniel CAdelman, Redwood City, CA 6,696,4836,670,144 B2B1 12/20032/2004 CraigSingh et al. (US); Jeffrey A. Silverman, 6,723,734 B2 4/2004 Kim et al. Burlingame, CA (US) 7,211,562 B2 5/2007 Rosen et al. 7.989,468 B2 * 8/2011 Adelman et al. .............. 514,300 (73) Assignee: Sunesis Pharmaceuticals, Inc., South 3.9. A. S. idaran-Ghera Gh et al.1 San Francisco, CA (US) 2003/0232334 A1* 12/2003 Morris et al. ..................... 435/6 c - r 2004/0106605 A1 6/2004 Carboni et al. .... 514/226.8 (*) Notice: Subject to any disclaimer, the term of this 2004/0132825 A1* 7/2004 Bacopoulos et al. ......... 514/575 patent is extended or adjusted under 35 2005/0203120 A1 9, 2005 Adelman et al. U.S.C. 154(b) by 201 days. 2005/0215583 A1 9, 2005 Arkin et al. 2006, OO25437 A1 2/2006 Adelman et al.
    [Show full text]
  • Universidade Estadual De Campinas Instituto De Biologia
    UNIVERSIDADE ESTADUAL DE CAMPINAS INSTITUTO DE BIOLOGIA VERÔNICA APARECIDA MONTEIRO SAIA CEREDA O PROTEOMA DO CORPO CALOSO DA ESQUIZOFRENIA THE PROTEOME OF THE CORPUS CALLOSUM IN SCHIZOPHRENIA CAMPINAS 2016 1 VERÔNICA APARECIDA MONTEIRO SAIA CEREDA O PROTEOMA DO CORPO CALOSO DA ESQUIZOFRENIA THE PROTEOME OF THE CORPUS CALLOSUM IN SCHIZOPHRENIA Dissertação apresentada ao Instituto de Biologia da Universidade Estadual de Campinas como parte dos requisitos exigidos para a obtenção do Título de Mestra em Biologia Funcional e Molecular na área de concentração de Bioquímica. Dissertation presented to the Institute of Biology of the University of Campinas in partial fulfillment of the requirements for the degree of Master in Functional and Molecular Biology, in the area of Biochemistry. ESTE ARQUIVO DIGITAL CORRESPONDE À VERSÃO FINAL DA DISSERTAÇÃO DEFENDIDA PELA ALUNA VERÔNICA APARECIDA MONTEIRO SAIA CEREDA E ORIENTADA PELO DANIEL MARTINS-DE-SOUZA. Orientador: Daniel Martins-de-Souza CAMPINAS 2016 2 Agência(s) de fomento e nº(s) de processo(s): CNPq, 151787/2F2014-0 Ficha catalográfica Universidade Estadual de Campinas Biblioteca do Instituto de Biologia Mara Janaina de Oliveira - CRB 8/6972 Saia-Cereda, Verônica Aparecida Monteiro, 1988- Sa21p O proteoma do corpo caloso da esquizofrenia / Verônica Aparecida Monteiro Saia Cereda. – Campinas, SP : [s.n.], 2016. Orientador: Daniel Martins de Souza. Dissertação (mestrado) – Universidade Estadual de Campinas, Instituto de Biologia. 1. Esquizofrenia. 2. Espectrometria de massas. 3. Corpo caloso.
    [Show full text]
  • UCSD MOLECULE PAGES Doi:10.6072/H0.MP.A002549.01 Volume 1, Issue 2, 2012 Copyright UC Press, All Rights Reserved
    UCSD MOLECULE PAGES doi:10.6072/H0.MP.A002549.01 Volume 1, Issue 2, 2012 Copyright UC Press, All rights reserved. Review Article Open Access WAVE2 Tadaomi Takenawa1, Shiro Suetsugu2, Daisuke Yamazaki3, Shusaku Kurisu1 WASP family verprolin-homologous protein 2 (WAVE2, also called WASF2) was originally identified by its sequence similarity at the carboxy-terminal VCA (verprolin, cofilin/central, acidic) domain with Wiskott-Aldrich syndrome protein (WASP) and N-WASP (neural WASP). In mammals, WAVE2 is ubiquitously expressed, and its two paralogs, WAVE1 (also called suppressor of cAMP receptor 1, SCAR1) and WAVE3, are predominantly expressed in the brain. The VCA domain of WASP and WAVE family proteins can activate the actin-related protein 2/3 (Arp2/3) complex, a major actin nucleator in cells. Proteins that can activate the Arp2/3 complex are now collectively known as nucleation-promoting factors (NPFs), and the WASP and WAVE families are a founding class of NPFs. The WAVE family has an amino-terminal WAVE homology domain (WHD domain, also called the SCAR homology domain, SHD) followed by the proline-rich region that interacts with various Src-homology 3 (SH3) domain proteins. The VCA domain located at the C-terminus. WAVE2, like WAVE1 and WAVE3, constitutively forms a huge heteropentameric protein complex (the WANP complex), binding through its WHD domain with Abi-1 (or its paralogs, Abi-2 and Abi-3), HSPC300 (also called Brick1), Nap1 (also called Hem-2 and NCKAP1), Sra1 (also called p140Sra1 and CYFIP1; its paralog is PIR121 or CYFIP2). The WANP complex is recruited to the plasma membrane by cooperative action of activated Rac GTPases and acidic phosphoinositides.
    [Show full text]
  • Sirna Interference Or Mirna Mimicry Peipei Wang1,2; Yue Zhou, Ph.D.1,2; Arthur M
    Theranostics 2021, Vol. 11, Issue 18 8771 Ivyspring International Publisher Theranostics 2021; 11(18): 8771-8796. doi: 10.7150/thno.62642 Review Effective tools for RNA-derived therapeutics: siRNA interference or miRNA mimicry Peipei Wang1,2; Yue Zhou, Ph.D.1,2; Arthur M. Richards, M.D., Ph.D.1,2,3 1. Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore. 2. Department of Medicine, National University Health System, 119228 Singapore. 3. Christchurch Heart Institute, Department of Medicine, University of Otago Christchurch, New Zealand. Corresponding author: Peipei Wang, MD, PhD, Cardiovascular Research Institute, Department of Medicine, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Centre for Translational Medicine, MD6, #08-01, 14 Medical Drive, Singapore 117599. Phone: (65) 81613586; Fax: (65) 6775-9715; E-mail: [email protected]. © The author(s). This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions. Received: 2021.05.12; Accepted: 2021.07.30; Published: 2021.08.11 Abstract The approval of the first small interfering RNA (siRNA) drug Patisiran by FDA in 2018 marks a new era of RNA interference (RNAi) therapeutics. MicroRNAs (miRNA), an important post-transcriptional gene regulator, are also the subject of both basic research and clinical trials. Both siRNA and miRNA mimics are ~21 nucleotides RNA duplexes inducing mRNA silencing. Given the well performance of siRNA, researchers ask whether miRNA mimics are unnecessary or developed siRNA technology can pave the way for the emergence of miRNA mimic drugs.
    [Show full text]
  • Preclinical Pharmacologic Evaluation of Pralatrexate and Romidepsin
    Published OnlineFirst February 12, 2015; DOI: 10.1158/1078-0432.CCR-14-2249 Cancer Therapy: Preclinical Clinical Cancer Research Preclinical Pharmacologic Evaluation of Pralatrexate and Romidepsin Confirms Potent Synergy of the Combination in a Murine Model of Human T-cell Lymphoma Salvia Jain1, Xavier Jirau-Serrano2, Kelly M. Zullo2, Luigi Scotto2, Carmine F. Palermo3,4, Stephen A. Sastra3,4,5, Kenneth P. Olive3,4,5, Serge Cremers3, Tiffany Thomas3,YingWei6, Yuan Zhang6, Govind Bhagat3, Jennifer E. Amengual2, Changchun Deng2, Charles Karan8, Ronald Realubit8, Susan E. Bates9, and Owen A. O'Connor2 Abstract Purpose: T-cell lymphomas (TCL) are aggressive diseases, NOG mouse model of TCL were used to explore the in vitro and in which carry a poor prognosis. The emergence of new drugs for vivo activity of pralatrexate and romidepsin in combination. TCL has created a need to survey these agents in a rapid and Corresponding mass spectrometry–based pharmacokinetic and reproducible fashion, to prioritize combinations which should be immunohistochemistry-based pharmacodynamic analyses of prioritized for clinical study. Mouse models of TCL that can be xenograft tumors were performed to better understand a mech- used for screening novel agents and their combinations are anistic basis for the drug:drug interaction. lacking. Developments in noninvasive imaging modalities, such Results: In vitro, pralatrexate and romidepsin exhibited con- as surface bioluminescence (SBL) and three-dimensional ultra- centration-dependent synergism in combination against a panel sound (3D-US), are challenging conventional approaches in of TCL cell lines. In a NOG murine model of TCL, the combination xenograft modeling relying on caliper measurements. The recent of pralatrexate and romidepsin exhibited enhanced efficacy com- approval of pralatrexate and romidepsin creates an obvious pared with either drug alone across a spectrum of tumors using combination that could produce meaningful activity in TCL, complementary imaging modalities, such as SBL and 3D-US.
    [Show full text]
  • Mrna Vaccine Era—Mechanisms, Drug Platform and Clinical Prospection
    International Journal of Molecular Sciences Review mRNA Vaccine Era—Mechanisms, Drug Platform and Clinical Prospection 1, 1, 2 1,3, Shuqin Xu y, Kunpeng Yang y, Rose Li and Lu Zhang * 1 State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Science, Fudan University, Shanghai 200438, China; [email protected] (S.X.); [email protected] (K.Y.) 2 M.B.B.S., School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; [email protected] 3 Shanghai Engineering Research Center of Industrial Microorganisms, Shanghai 200438, China * Correspondence: [email protected]; Tel.: +86-13524278762 These authors contributed equally to this work. y Received: 30 July 2020; Accepted: 30 August 2020; Published: 9 September 2020 Abstract: Messenger ribonucleic acid (mRNA)-based drugs, notably mRNA vaccines, have been widely proven as a promising treatment strategy in immune therapeutics. The extraordinary advantages associated with mRNA vaccines, including their high efficacy, a relatively low severity of side effects, and low attainment costs, have enabled them to become prevalent in pre-clinical and clinical trials against various infectious diseases and cancers. Recent technological advancements have alleviated some issues that hinder mRNA vaccine development, such as low efficiency that exist in both gene translation and in vivo deliveries. mRNA immunogenicity can also be greatly adjusted as a result of upgraded technologies. In this review, we have summarized details regarding the optimization of mRNA vaccines, and the underlying biological mechanisms of this form of vaccines. Applications of mRNA vaccines in some infectious diseases and cancers are introduced. It also includes our prospections for mRNA vaccine applications in diseases caused by bacterial pathogens, such as tuberculosis.
    [Show full text]
  • Stathmin Is Overexpressed in Malignant Mesothelioma
    ANTICANCER RESEARCH 27: 39-44 (2007) Stathmin is Overexpressed in Malignant Mesothelioma JAE Y. KIM1, CHANSONETTE HARVARD1, LIANG YOU1, ZHIDONG XU1, KRISTOPHER KUCHENBECKER1, RICK BAEHNER2 and DAVID JABLONS1 1Thoracic Oncology Laboratory, UCSF Comprehensive Cancer Center, San Francisco, CA 94115; 2Department of Pathology, UCSF, San Francisco, CA 94115, U.S.A. Abstract. Background: Malignant pleural mesothelioma is a unclear (7, 8). Unlike many other epithelial cancers, the highly aggressive cancer, with low overall survival. The activation of ras genes and inactivation of Rb and p53 genes pathogenesis of mesothelioma is poorly understood. The aim of do not seem to be necessary for the development of this study was to identify potential genes overexpressed in mesothelioma (9, 10). Alterations of several molecular mesothelioma. Materials and Methods: A cDNA microarray was pathways, including epidermal growth factor receptor, cell used to identify potential genes that are activated in mesothelioma cycle regulatory genes, and developmental pathways have cell lines. Overexpression of stathmin, a cytosolic protein that been linked to mesothelioma (11, 12). There is also regulates microtubule dynamics, was found. RT-PCR, Western evidence that Simian virus 40 (SV40) may contribute to the blot, and immunohistochemistry were used to confirm develop of mesothelioma, but the exact genetic alterations overexpression in both cell lines and tumor samples. Results: leading to mesothelioma remain unknown (11, 13). Using RT-PCR and Western blot, stathmin overexpression was The aim of this study was to identify potential genes confirmed in seven mesothelioma cell lines. Increased stathmin involved in the pathogenesis of mesothelioma. protein expression was also found in seven out of eight mesothelioma tumor samples.
    [Show full text]
  • Activation of Nuclear Factor-Kappa B Signalling Promotes Cellular Senescence
    Oncogene (2011) 30, 2356–2366 & 2011 Macmillan Publishers Limited All rights reserved 0950-9232/11 www.nature.com/onc ORIGINAL ARTICLE Activation of nuclear factor-kappa B signalling promotes cellular senescence E Rovillain1, L Mansfield1, C Caetano1, M Alvarez-Fernandez2, OL Caballero3, RH Medema2, H Hummerich4 and PS Jat1 1Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK; 2Department of Medical Oncology, University Medical Center, Utrecht, The Netherlands; 3Ludwig Institute for Cancer Research Ltd, New York, NY, USA and 4MRC Prion Unit, UCL Institute of Neurology, London, UK Cellular senescence is a programme of irreversible cell of intrinsic and extrinsic stimuli including progressive cycle arrest that normal cells undergo in response to shortening of telomeres or changes in telomeric struc- progressive shortening of telomeres, changes in telomeric ture or other forms of genotoxic stress such as oncogene structure, oncogene activation or oxidative stress. The activation, DNA damage or oxidative stress resulting in underlying signalling pathways, of major clinicopatholo- a DNA damage response and growth arrest via gical relevance, are unknown. We combined genome-wide activation of the p53 tumour suppressor (Ben-Porath expression profiling with genetic complementation to and Weinberg, 2005; Campisi and d’Adda di Fagagna, identify genes that are differentially expressed when 2007; Adams, 2009). Activation of p53 induces p21CIP1, conditionally immortalised human fibroblasts undergo which prevents cyclin/cdk complexes from phosphor- senescence upon activation of the p16-pRB and p53-p21 ylating the RB family of proteins, thereby activating the tumour suppressor pathways. This identified 816 up and pRB tumour suppressor pathway (Ben-Porath and 961 downregulated genes whose expression was reversed Weinberg, 2005; Campisi and d’Adda di Fagagna, when senescence was bypassed.
    [Show full text]
  • Aptamers and Antisense Oligonucleotides for Diagnosis and Treatment of Hematological Diseases
    International Journal of Molecular Sciences Review Aptamers and Antisense Oligonucleotides for Diagnosis and Treatment of Hematological Diseases Valentina Giudice 1,2,* , Francesca Mensitieri 1, Viviana Izzo 1,2 , Amelia Filippelli 1,2 and Carmine Selleri 1 1 Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; [email protected] (F.M.); [email protected] (V.I.); afi[email protected] (A.F.); [email protected] (C.S.) 2 Unit of Clinical Pharmacology, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, 84131 Salerno, Italy * Correspondence: [email protected]; Tel.: +39-(0)-89965116 Received: 30 March 2020; Accepted: 2 May 2020; Published: 4 May 2020 Abstract: Aptamers or chemical antibodies are single-stranded DNA or RNA oligonucleotides that bind proteins and small molecules with high affinity and specificity by recognizing tertiary or quaternary structures as antibodies. Aptamers can be easily produced in vitro through a process known as systemic evolution of ligands by exponential enrichment (SELEX) or a cell-based SELEX procedure. Aptamers and modified aptamers, such as slow, off-rate, modified aptamers (SOMAmers), can bind to target molecules with less polar and more hydrophobic interactions showing slower dissociation rates, higher stability, and resistance to nuclease degradation. Aptamers and SOMAmers are largely employed for multiplex high-throughput proteomics analysis with high reproducibility and reliability, for tumor cell detection by flow cytometry or microscopy for research and clinical purposes. In addition, aptamers are increasingly used for novel drug delivery systems specifically targeting tumor cells, and as new anticancer molecules. In this review, we summarize current preclinical and clinical applications of aptamers in malignant and non-malignant hematological diseases.
    [Show full text]
  • Differential Gene Expression in Oligodendrocyte Progenitor Cells, Oligodendrocytes and Type II Astrocytes
    Tohoku J. Exp. Med., 2011,Differential 223, 161-176 Gene Expression in OPCs, Oligodendrocytes and Type II Astrocytes 161 Differential Gene Expression in Oligodendrocyte Progenitor Cells, Oligodendrocytes and Type II Astrocytes Jian-Guo Hu,1,2,* Yan-Xia Wang,3,* Jian-Sheng Zhou,2 Chang-Jie Chen,4 Feng-Chao Wang,1 Xing-Wu Li1 and He-Zuo Lü1,2 1Department of Clinical Laboratory Science, The First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China 2Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, P.R. China 3Department of Neurobiology, Shanghai Jiaotong University School of Medicine, Shanghai, P.R. China 4Department of Laboratory Medicine, Bengbu Medical College, Bengbu, P.R. China Oligodendrocyte precursor cells (OPCs) are bipotential progenitor cells that can differentiate into myelin-forming oligodendrocytes or functionally undetermined type II astrocytes. Transplantation of OPCs is an attractive therapy for demyelinating diseases. However, due to their bipotential differentiation potential, the majority of OPCs differentiate into astrocytes at transplanted sites. It is therefore important to understand the molecular mechanisms that regulate the transition from OPCs to oligodendrocytes or astrocytes. In this study, we isolated OPCs from the spinal cords of rat embryos (16 days old) and induced them to differentiate into oligodendrocytes or type II astrocytes in the absence or presence of 10% fetal bovine serum, respectively. RNAs were extracted from each cell population and hybridized to GeneChip with 28,700 rat genes. Using the criterion of fold change > 4 in the expression level, we identified 83 genes that were up-regulated and 89 genes that were down-regulated in oligodendrocytes, and 92 genes that were up-regulated and 86 that were down-regulated in type II astrocytes compared with OPCs.
    [Show full text]
  • Download Article (PDF)
    DNA and RNA Nanotechnology 2015; 2: 42–52 Mini review Open Access Martin Panigaj*, Jakob Reiser Aptamer guided delivery of nucleic acid-based nanoparticles DOI 10.1515/rnan-2015-0005 Evolution of Ligands by Exponential enrichment) [4,5]. Received July 15, 2015; accepted October 3, 2015 Nucleic acid-based aptamers are especially well suited Abstract: Targeted delivery of bioactive compounds is a for the delivery of nucleic acid-based therapeutics. Any key part of successful therapies. In this context, nucleic nucleic acid with therapeutic potential can be linked acid and protein-based aptamers have been shown to to an aptamer sequence [6], resulting in a bivalent bind therapeutically relevant targets including receptors. molecule endowed with a targeting aptamer moiety and In the last decade, nucleic acid-based therapeutics a functional RNA/DNA moiety like a small interfering coupled to aptamers have emerged as a viable strategy for RNA (siRNA), a micro RNA (miRNA), a miRNA antagonist cell specific delivery. Additionally, recent developments (antimiR), deoxyribozymes (DNAzymes), etc. In addition in nucleic acid nanotechnology offer an abundance of to the specific binding, many aptamers upon receptor possibilities to rationally design aptamer targeted RNA recognition elicit antagonistic or agonistic responses that, or DNA nanoparticles involving combinatorial use of in combination with conjugated functional nucleic acids various intrinsic functionalities. Although a host of issues have the potential of synergism. Since the first report including stability, safety and intracellular trafficking describing an aptamer-siRNA delivery approach in 2006 remain to be addressed, aptamers as simple functional many functional RNAs and DNAs conjugated to aptamer chimeras or as parts of multifunctional self-assembled sequences have been tested in vitro and in vivo [7-9].
    [Show full text]