51
|
Moore SM, Skowronska-Krawczyk D, Chao DL. Emerging Concepts for RNA Therapeutics for Inherited Retinal Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1185:85-89. [PMID: 31884593 DOI: 10.1007/978-3-030-27378-1_14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Inherited retinal diseases (IRD) encompass a wide spectrum of hereditary blindness with significant genetic heterogeneity. Therapeutics regulating gene expression on an RNA level have significant promise for treating IRD. In this review, we review the molecular basis of oligonucleotide therapeutics such as ribozymes, RNA interference (RNAi), antisense oligonucleotides (ASO), CRISPRi/a, and their applications to treatments of IRD.
Collapse
Affiliation(s)
- Spencer M Moore
- Medical Scientist Training Program, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Dorota Skowronska-Krawczyk
- Department of Ophthalmology, Shiley Eye Institute, University of California, San Diego, La Jolla, CA, USA
| | - Daniel L Chao
- Department of Ophthalmology, Shiley Eye Institute, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
52
|
Xiong Y, Chang LL, Tran B, Dai T, Zhong R, Mao YC, Zhu YZ. ZYZ-803, a novel hydrogen sulfide-nitric oxide conjugated donor, promotes angiogenesis via cross-talk between STAT3 and CaMKII. Acta Pharmacol Sin 2020; 41:218-228. [PMID: 31316179 PMCID: PMC7468320 DOI: 10.1038/s41401-019-0255-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 05/21/2019] [Indexed: 02/05/2023]
Abstract
Endothelial angiogenesis plays a vital role in recovery from chronic ischemic injuries. ZYZ-803 is a hybrid donor of hydrogen sulfide (H2S) and nitric oxide (NO). Previous studies showed that ZYZ-803 stimulated endothelial cell angiogenesis both in vitro and in vivo. In this study, we investigated whether the signal transducer and activator of transcription 3 (STAT3) and Ca2+/CaM-dependent protein kinase II (CaMKII) signaling was involved in ZYZ-803-induced angiogenesis. Treatment with ZYZ-803 (1 μM) significantly increased the phosphorylation of STAT3 (Tyr705) and CaMKII (Thr286) in human umbilical vein endothelial cells (HUVECs), these two effects had a similar time course. Pretreatment with WP1066 (STAT3 inhibitor) or KN93 (CAMKII inhibitor) blocked ZYZ-803-induced STAT3/CAMKII activation and significantly suppressed the proliferation and migration of HUVECs. In addition, pretreatment with the inhibitors significantly decreased ZYZ-803-induced tube formations along with the outgrowths of branch-like microvessels in aortic rings. In the mice with femoral artery ligation, administration of ZYZ-803 significantly increased the blood perfusion and vascular density in the hind limb, whereas co-administration of WP1066 or KN93 abrogated ZYZ-803-induced angiogenesis. By using STAT3 siRNA, we further explored the cross-talk between STAT3 and CaMKII in ZYZ-803-induced angiogenesis. We found that STAT3 knockdown suppressed ZYZ-803-induced HUVEC angiogenesis and affected CaMKII expression. ZYZ-803 treatment markedly enhanced the interaction between CaMKII and STAT3. ZYZ-803 treatment induced the nuclear translocation of STAT3. We demonstrated that both STAT3 and CaMKII functioned as positive regulators in ZYZ-803-induced endothelial angiogenesis and STAT3 was important in ZYZ-803-induced CaMKII activation, which highlights the beneficial role of ZYZ-803 in STAT3/CaMKII-related cardiovascular diseases.
Collapse
Affiliation(s)
- Ying Xiong
- Institute of Biomedical Science and School of Pharmacy, Fudan University, Shanghai, 200032, China
| | - Ling-Ling Chang
- Institute of Biomedical Science and School of Pharmacy, Fudan University, Shanghai, 200032, China
| | - Bahieu Tran
- Institute of Biomedical Science and School of Pharmacy, Fudan University, Shanghai, 200032, China
| | - Tao Dai
- Institute of Biomedical Science and School of Pharmacy, Fudan University, Shanghai, 200032, China
| | - Rui Zhong
- Institute of Biomedical Science and School of Pharmacy, Fudan University, Shanghai, 200032, China
| | - Yi-Cheng Mao
- Institute of Biomedical Science and School of Pharmacy, Fudan University, Shanghai, 200032, China.
| | - Yi-Zhun Zhu
- Institute of Biomedical Science and School of Pharmacy, Fudan University, Shanghai, 200032, China.
- School of Pharmacy, Macau University of Science and Technology, Macau, China.
| |
Collapse
|
53
|
Bartoszewski R, Sikorski AF. Editorial focus: understanding off-target effects as the key to successful RNAi therapy. Cell Mol Biol Lett 2019; 24:69. [PMID: 31867046 PMCID: PMC6902517 DOI: 10.1186/s11658-019-0196-3] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 12/03/2019] [Indexed: 12/21/2022] Open
Abstract
With the first RNA interference (RNAi) drug (ONPATTRO (patisiran)) on the market, we witness the RNAi therapy field reaching a critical turning point, when further improvements in drug candidate design and delivery pipelines should enable fast delivery of novel life changing treatments to patients. Nevertheless, ignoring parallel development of RNAi dedicated in vitro pharmacological profiling aiming to identify undesirable off-target activity may slow down or halt progress in the RNAi field. Since academic research is currently fueling the RNAi development pipeline with new therapeutic options, the objective of this article is to briefly summarize the basics of RNAi therapy, as well as to discuss how to translate basic research into better understanding of related drug candidate safety profiles early in the process.
Collapse
Affiliation(s)
- Rafal Bartoszewski
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - Aleksander F. Sikorski
- Department of Cytobiochemistry, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| |
Collapse
|
54
|
A. Lea R, K. Niakan K. Human germline genome editing. Nat Cell Biol 2019; 21:1479-1489. [DOI: 10.1038/s41556-019-0424-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 10/25/2019] [Indexed: 12/14/2022]
|
55
|
Seira O, Liu J, Assinck P, Ramer M, Tetzlaff W. KIF2A characterization after spinal cord injury. Cell Mol Life Sci 2019; 76:4355-4368. [PMID: 31041455 PMCID: PMC11105463 DOI: 10.1007/s00018-019-03116-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 04/05/2019] [Accepted: 04/24/2019] [Indexed: 01/23/2023]
Abstract
Axons in the central nervous system (CNS) typically fail to regenerate after injury. This failure is multi-factorial and caused in part by disruption of the axonal cytoskeleton. The cytoskeleton, in particular microtubules (MT), plays a critical role in axonal transport and axon growth during development. In this regard, members of the kinesin superfamily of proteins (KIFs) regulate the extension of primary axons toward their targets and control the growth of collateral branches. KIF2A negatively regulates axon growth through MT depolymerization. Using three different injury models to induce SCI in adult rats, we examined the temporal and cellular expression of KIF2A in the injured spinal cord. We observed a progressive increase of KIF2A expression with maximal levels at 10 days to 8 weeks post-injury as determined by Western blot analysis. KIF2A immunoreactivity was present in axons, spinal neurons and mature oligodendrocytes adjacent to the injury site. Results from the present study suggest that KIF2A at the injured axonal tips may contribute to neurite outgrowth inhibition after injury, and that its increased expression in inhibitory spinal neurons adjacent to the injury site might contribute to an intrinsic wiring-control mechanism associated with neuropathic pain. Further studies will determine whether KIF2A may be a potential target for the development of regeneration-promoting or pain-preventing therapies.
Collapse
Affiliation(s)
- Oscar Seira
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Centre, University of British Columbia (UBC), 818 West 10th Avenue, Vancouver, BC, V5Z 1M9, Canada.
- Department of Zoology, University of British Columbia (UBC), Vancouver, Canada.
| | - Jie Liu
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Centre, University of British Columbia (UBC), 818 West 10th Avenue, Vancouver, BC, V5Z 1M9, Canada
| | - Peggy Assinck
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Centre, University of British Columbia (UBC), 818 West 10th Avenue, Vancouver, BC, V5Z 1M9, Canada
- Graduate Program in Neuroscience, University of British Columbia (UBC), Vancouver, Canada
- MRC Centre for Regenerative Medicine, The University of Edinburgh, Edinburgh, UK
| | - Matt Ramer
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Centre, University of British Columbia (UBC), 818 West 10th Avenue, Vancouver, BC, V5Z 1M9, Canada
- Department of Zoology, University of British Columbia (UBC), Vancouver, Canada
| | - Wolfram Tetzlaff
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Centre, University of British Columbia (UBC), 818 West 10th Avenue, Vancouver, BC, V5Z 1M9, Canada
- Department of Zoology, University of British Columbia (UBC), Vancouver, Canada
- Department of Surgery, University of British Columbia (UBC), Vancouver, Canada
| |
Collapse
|
56
|
Park R, Kim H, Jang M, Jo D, Park YI, Namkoong S, Lee JI, Jang IS, Park J. LNX1 contributes to tumor growth by down-regulating p53 stability. FASEB J 2019; 33:13216-13227. [PMID: 31533005 DOI: 10.1096/fj.201900366r] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The well-known tumor suppressor p53 inhibits the formation of various cancers by inducing cell cycle arrest and apoptosis. Although p53 mutations are commonly found in many cancers, p53 is functionally inactivated in tumor cells that retain wild-type p53. Here, we show that the ligand of numb protein X1 (LNX1) inhibited p53-dependent transcription by decreasing the half-life of p53. We generated LNX1 knockout (KO) cells in p53 wild-type cancer cells (A549, HCT116, and MCF7) using the clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein 9 gene-editing system. LNX1 KO activated p53-dependent transcription by increasing the stability of p53. Moreover, lentivirus-mediated overexpression of LNX1 decreased p53 protein levels and inhibited p53-dependent transcription. LNX1 interacted with p53 and mouse double minute 2 (MDM2) and increased the ubiquitination of p53 in an MDM2-dependent manner. Finally, we demonstrated that LNX1 was required for efficient tumor growth both in cell culture and in a mouse tumor xenograft model. These results collectively indicated that LNX1 contributed to tumor growth by inhibiting p53-dependent signaling in p53 wild-type cancer cells.-Park, R., Kim, H., Jang, M., Jo, D., Park, Y.-I., Namkoong, S., Lee, J. I., Jang, I.-S., Park, J. LNX1 contributes to tumor growth by down-regulating p53 stability.
Collapse
Affiliation(s)
- Rackhyun Park
- Division of Biological Science and Technology, Yonsei University, Wonju, South Korea
| | - Hyunju Kim
- Division of Biological Science and Technology, Yonsei University, Wonju, South Korea
| | - Minsu Jang
- Division of Biological Science and Technology, Yonsei University, Wonju, South Korea
| | - Daum Jo
- Division of Biological Science and Technology, Yonsei University, Wonju, South Korea
| | - Yea-In Park
- Division of Biological Science and Technology, Yonsei University, Wonju, South Korea
| | - Sim Namkoong
- Department of Biochemistry, Kangwon National University, Chuncheon, South Korea
| | - Jin I Lee
- Division of Biological Science and Technology, Yonsei University, Wonju, South Korea
| | - Ik-Soon Jang
- Division of Bioconvergence Analysis, Korea Basic Science Institute, Daejeon, South Korea
| | - Junsoo Park
- Division of Biological Science and Technology, Yonsei University, Wonju, South Korea
| |
Collapse
|
57
|
Ingusci S, Verlengia G, Soukupova M, Zucchini S, Simonato M. Gene Therapy Tools for Brain Diseases. Front Pharmacol 2019; 10:724. [PMID: 31312139 PMCID: PMC6613496 DOI: 10.3389/fphar.2019.00724] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 06/05/2019] [Indexed: 01/20/2023] Open
Abstract
Neurological disorders affecting the central nervous system (CNS) are still incompletely understood. Many of these disorders lack a cure and are seeking more specific and effective treatments. In fact, in spite of advancements in knowledge of the CNS function, the treatment of neurological disorders with modern medical and surgical approaches remains difficult for many reasons, such as the complexity of the CNS, the limited regenerative capacity of the tissue, and the difficulty in conveying conventional drugs to the organ due to the blood-brain barrier. Gene therapy, allowing the delivery of genetic materials that encodes potential therapeutic molecules, represents an attractive option. Gene therapy can result in a stable or inducible expression of transgene(s), and can allow a nearly specific expression in target cells. In this review, we will discuss the most commonly used tools for the delivery of genetic material in the CNS, including viral and non-viral vectors; their main applications; their advantages and disadvantages. We will discuss mechanisms of genetic regulation through cell-specific and inducible promoters, which allow to express gene products only in specific cells and to control their transcriptional activation. In addition, we will describe the applications to CNS diseases of post-transcriptional regulation systems (RNA interference); of systems allowing spatial or temporal control of expression [optogenetics and Designer Receptors Exclusively Activated by Designer Drugs (DREADDs)]; and of gene editing technologies (CRISPR/Cas9, Zinc finger proteins). Particular attention will be reserved to viral vectors derived from herpes simplex type 1, a potential tool for the delivery and expression of multiple transgene cassettes simultaneously.
Collapse
Affiliation(s)
- Selene Ingusci
- Department of Medical Sciences and National Institute of Neuroscience, University of Ferrara, Ferrara, Italy
| | - Gianluca Verlengia
- Department of Medical Sciences and National Institute of Neuroscience, University of Ferrara, Ferrara, Italy.,Division of Neuroscience, University Vita-Salute San Raffaele, Milan, Italy
| | - Marie Soukupova
- Department of Medical Sciences and National Institute of Neuroscience, University of Ferrara, Ferrara, Italy
| | - Silvia Zucchini
- Department of Medical Sciences and National Institute of Neuroscience, University of Ferrara, Ferrara, Italy.,Technopole of Ferrara, LTTA Laboratory for Advanced Therapies, Ferrara, Italy
| | - Michele Simonato
- Department of Medical Sciences and National Institute of Neuroscience, University of Ferrara, Ferrara, Italy.,Division of Neuroscience, University Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
58
|
Zimmer AM, Pan YK, Chandrapalan T, Kwong RWM, Perry SF. Loss-of-function approaches in comparative physiology: is there a future for knockdown experiments in the era of genome editing? ACTA ACUST UNITED AC 2019; 222:222/7/jeb175737. [PMID: 30948498 DOI: 10.1242/jeb.175737] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Loss-of-function technologies, such as morpholino- and RNAi-mediated gene knockdown, and TALEN- and CRISPR/Cas9-mediated gene knockout, are widely used to investigate gene function and its physiological significance. Here, we provide a general overview of the various knockdown and knockout technologies commonly used in comparative physiology and discuss the merits and drawbacks of these technologies with a particular focus on research conducted in zebrafish. Despite their widespread use, there is an ongoing debate surrounding the use of knockdown versus knockout approaches and their potential off-target effects. This debate is primarily fueled by the observations that, in some studies, knockout mutants exhibit phenotypes different from those observed in response to knockdown using morpholinos or RNAi. We discuss the current debate and focus on the discrepancies between knockdown and knockout phenotypes, providing literature and primary data to show that the different phenotypes are not necessarily a direct result of the off-target effects of the knockdown agents used. Nevertheless, given the recent evidence of some knockdown phenotypes being recapitulated in knockout mutants lacking the morpholino or RNAi target, we stress that results of knockdown experiments need to be interpreted with caution. We ultimately argue that knockdown experiments should not be discontinued if proper control experiments are performed, and that with careful interpretation, knockdown approaches remain useful to complement the limitations of knockout studies (e.g. lethality of knockout and compensatory responses).
Collapse
Affiliation(s)
- Alex M Zimmer
- Department of Biology, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Yihang K Pan
- Department of Biology, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | | | | | - Steve F Perry
- Department of Biology, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| |
Collapse
|
59
|
Brachs S, Polack J, Brachs M, Jahn-Hofmann K, Elvert R, Pfenninger A, Bärenz F, Margerie D, Mai K, Spranger J, Kannt A. Genetic Nicotinamide N-Methyltransferase ( Nnmt) Deficiency in Male Mice Improves Insulin Sensitivity in Diet-Induced Obesity but Does Not Affect Glucose Tolerance. Diabetes 2019; 68:527-542. [PMID: 30552109 DOI: 10.2337/db18-0780] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 12/03/2018] [Indexed: 11/13/2022]
Abstract
Antisense oligonucleotide knockdown (ASO-KD) of nicotinamide N-methyltransferase (NNMT) in high-fat diet (HFD)-fed mice has been reported to reduce weight gain and plasma insulin levels and to improve glucose tolerance. Using NNMT-ASO-KD or NNMT knockout mice (NNMT-/-), we tested the hypothesis that Nnmt deletion protects against diet-induced obesity and its metabolic consequences in males and females on obesity-inducing diets. We also examined samples from a human weight reduction (WR) study for adipose NNMT (aNNMT) expression and plasma 1-methylnicotinamide (MNAM) levels. In Western diet (WD)-fed female mice, NNMT-ASO-KD reduced body weight, fat mass, and insulin level and improved glucose tolerance. Although NNMT-/- mice fed a standard diet had no obvious phenotype, NNMT-/- males fed an HFD showed strongly improved insulin sensitivity (IS). Furthermore, NNMT-/- females fed a WD showed reduced weight gain, less fat, and lower insulin levels. However, no improved glucose tolerance was observed in NNMT-/- mice. Although NNMT expression in human fat biopsy samples increased during WR, corresponding plasma MNAM levels significantly declined, suggesting that other mechanisms besides aNNMT expression modulate circulating MNAM levels during WR. In summary, upon NNMT deletion or knockdown in males and females fed different obesity-inducing diets, we observed sex- and diet-specific differences in body composition, weight, and glucose tolerance and estimates of IS.
Collapse
Affiliation(s)
- Sebastian Brachs
- Department of Endocrinology and Metabolism, Charité-Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
- Center for Cardiovascular Research (CCR), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - James Polack
- Department of Endocrinology and Metabolism, Charité-Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
- Center for Cardiovascular Research (CCR), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Maria Brachs
- Department of Endocrinology and Metabolism, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Center for Cardiovascular Research (CCR), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Ralf Elvert
- Sanofi Research and Development, Frankfurt am Main, Germany
| | | | - Felix Bärenz
- Sanofi Research and Development, Frankfurt am Main, Germany
| | | | - Knut Mai
- Department of Endocrinology and Metabolism, Charité-Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
- Center for Cardiovascular Research (CCR), Charité-Universitätsmedizin Berlin, Berlin, Germany
- Clinical Research Unit, Berlin Institute of Health (BIH), Berlin, Germany
| | - Joachim Spranger
- Department of Endocrinology and Metabolism, Charité-Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
- Center for Cardiovascular Research (CCR), Charité-Universitätsmedizin Berlin, Berlin, Germany
- Clinical Research Unit, Berlin Institute of Health (BIH), Berlin, Germany
| | - Aimo Kannt
- Sanofi Research and Development, Frankfurt am Main, Germany
- Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
60
|
Iannitti T, Scarrott JM, Likhite S, Coldicott IRP, Lewis KE, Heath PR, Higginbottom A, Myszczynska MA, Milo M, Hautbergue GM, Meyer K, Kaspar BK, Ferraiuolo L, Shaw PJ, Azzouz M. Translating SOD1 Gene Silencing toward the Clinic: A Highly Efficacious, Off-Target-free, and Biomarker-Supported Strategy for fALS. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 12:75-88. [PMID: 30195799 PMCID: PMC6023790 DOI: 10.1016/j.omtn.2018.04.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/28/2018] [Accepted: 04/28/2018] [Indexed: 12/13/2022]
Abstract
Of familial amyotrophic lateral sclerosis (fALS) cases, 20% are caused by mutations in the gene encoding human cytosolic Cu/Zn superoxide dismutase (hSOD1). Efficient translation of the therapeutic potential of RNAi for the treatment of SOD1-ALS patients requires the development of vectors that are free of significant off-target effects and with reliable biomarkers to discern sufficient target engagement and correct dosing. Using adeno-associated virus serotype 9 to deliver RNAi against hSOD1 in the SOD1G93A mouse model, we found that intrathecal injection of the therapeutic vector via the cisterna magna delayed onset of disease, decreased motor neuron death at end stage by up to 88%, and prolonged the median survival of SOD1G93A mice by up to 42%. To our knowledge, this is the first report to demonstrate no significant off-target effects linked to hSOD1 silencing, providing further confidence in the specificity of this approach. We also report the measurement of cerebrospinal fluid (CSF) hSOD1 protein levels as a biomarker of effective dosing and efficacy of hSOD1 knockdown. Together, these data provide further confidence in the safety of the clinical therapeutic vector. The CSF biomarker will be a useful measure of biological activity for translation into human clinical trials.
Collapse
Affiliation(s)
- Tommaso Iannitti
- University of Sheffield, Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, Sheffield, UK
| | - Joseph M Scarrott
- University of Sheffield, Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, Sheffield, UK
| | - Shibi Likhite
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, College of Medicine and Public Health, The Ohio State University, Columbus, OH, USA
| | - Ian R P Coldicott
- University of Sheffield, Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, Sheffield, UK
| | - Katherine E Lewis
- University of Sheffield, Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, Sheffield, UK
| | - Paul R Heath
- University of Sheffield, Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, Sheffield, UK
| | - Adrian Higginbottom
- University of Sheffield, Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, Sheffield, UK
| | - Monika A Myszczynska
- University of Sheffield, Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, Sheffield, UK
| | - Marta Milo
- University of Sheffield, Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, Sheffield, UK
| | - Guillaume M Hautbergue
- University of Sheffield, Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, Sheffield, UK
| | - Kathrin Meyer
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, College of Medicine and Public Health, The Ohio State University, Columbus, OH, USA
| | - Brian K Kaspar
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, College of Medicine and Public Health, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Laura Ferraiuolo
- University of Sheffield, Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, Sheffield, UK
| | - Pamela J Shaw
- University of Sheffield, Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, Sheffield, UK.
| | - Mimoun Azzouz
- University of Sheffield, Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, Sheffield, UK.
| |
Collapse
|
61
|
Zotti M, Dos Santos EA, Cagliari D, Christiaens O, Taning CNT, Smagghe G. RNA interference technology in crop protection against arthropod pests, pathogens and nematodes. PEST MANAGEMENT SCIENCE 2018; 74:1239-1250. [PMID: 29194942 DOI: 10.1002/ps.4813] [Citation(s) in RCA: 197] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 10/09/2017] [Accepted: 11/22/2017] [Indexed: 05/03/2023]
Abstract
Scientists have made significant progress in understanding and unraveling several aspects of double-stranded RNA (dsRNA)-mediated gene silencing during the last two decades. Now that the RNA interference (RNAi) mechanism is well understood, it is time to consider how to apply the acquired knowledge to agriculture and crop protection. Some RNAi-based products are already available for farmers and more are expected to reach the market soon. Tailor-made dsRNA as an active ingredient for biopesticide formulations is considered a raw material that can be used for diverse purposes, from pest control and bee protection against viruses to pesticide resistance management. The RNAi mechanism works at the messenger RNA (mRNA) level, exploiting a sequence-dependent mode of action, which makes it unique in potency and selectivity compared with conventional agrochemicals. Furthermore, the use of RNAi in crop protection can be achieved by employing plant-incorporated protectants through plant transformation, but also by non-transformative strategies such as the use of formulations of sprayable RNAs as direct control agents, resistance factor repressors or developmental disruptors. In this review, RNAi is presented in an agricultural context (discussing products that have been launched on the market or will soon be available), and we go beyond the classical presentation of successful examples of RNAi in pest-insect control and comprehensively explore its potential for the control of plant pathogens, nematodes and mites, and to fight against diseases and parasites in beneficial insects. Moreover, we also discuss its use as a repressor for the management of pesticide-resistant weeds and insects. Finally, this review reports on the advances in non-transformative dsRNA delivery and the production costs of dsRNA, and discusses environmental considerations. © 2017 Society of Chemical Industry.
Collapse
Affiliation(s)
- Moises Zotti
- Department of Crop Protection, Molecular Entomology, Federal University of Pelotas, Pelotas, Brazil
| | - Ericmar Avila Dos Santos
- Department of Crop Protection, Molecular Entomology, Federal University of Pelotas, Pelotas, Brazil
| | - Deise Cagliari
- Department of Crop Protection, Molecular Entomology, Federal University of Pelotas, Pelotas, Brazil
| | - Olivier Christiaens
- Department of Crop Protection, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Clauvis Nji Tizi Taning
- Department of Crop Protection, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Guy Smagghe
- Department of Crop Protection, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| |
Collapse
|
62
|
Li H, Su B, Qin G, Ye Z, Elaswad A, Alsaqufi A, Perera DA, Qin Z, Odin R, Vo K, Drescher D, Robinson D, Dong S, Zhang D, Shang M, Abass N, Das SK, Bangs M, Dunham RA. Repressible Transgenic Sterilization in Channel Catfish, Ictalurus punctatus, by Knockdown of Primordial Germ Cell Genes with Copper-Sensitive Constructs. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2018; 20:324-342. [PMID: 29679251 DOI: 10.1007/s10126-018-9819-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 11/24/2017] [Indexed: 06/08/2023]
Abstract
Repressible knockdown approaches were investigated to manipulate for transgenic sterilization in channel catfish, Ictalurus punctatus. Two primordial germ cell (PGC) marker genes, nanos and dead end, were targeted for knockdown and an off-target gene, vasa, was monitored. Two potentially copper-sensitive repressible promoters, yeast ctr3 (M) and ctr3-reduced (Mctr), were coupled with four knockdown strategies separately including: ds-sh RNA targeting the 5' end (N1) or 3' end (N2) of channel catfish nanos, full-length cDNA sequence of channel catfish nanos for overexpression (cDNA), and ds-sh RNA-targeting channel catfish dead end (DND). Each construct had an untreated group and treated group with copper sulfate as the repressor compound. Spawning rates of full-sibling P1 fish exposed or not exposed to the constructs as treated and untreated embryos were 85 and 54%, respectively, indicating potential sterilization of fish and repression of the constructs. In F1 fish, mRNA expressions of PGC marker genes for most constructs were downregulated in the untreated group and the knockdown was repressed in the treated group. Gonad development in transgenic, untreated F1 channel catfish was reduced compared to non-transgenic fish for MctrN2, MN1, MN2, and MDND. For 3-year-old adults, gonad size in the transgenic untreated group was 93.4% smaller than the non-transgenic group for females and 92.3% for males. However, mean body weight of transgenic females (781.8 g) and males (883.8 g) was smaller than of non-transgenic counterparts (984.2 and 1254.3 g) at 3 years of age, a 25.8 and 41.9% difference for females and males, respectively. The results indicate that repressible transgenic sterilization is feasible for reproductive control of fish, but negative pleiotropic effects can result.
Collapse
Affiliation(s)
- Hanbo Li
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Baofeng Su
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
- National and Local United Engineering Laboratory for Freshwater Fish Breeding, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fisheries Sciences, Harbin, 150070, China
| | - Guyu Qin
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Zhi Ye
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Ahmed Elaswad
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
- Department of Animal Wealth Development, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Ahmed Alsaqufi
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
- Department of Aquaculture and Animal Production, King Faisal University, Al Ahsa, 31982, Saudi Arabia
| | - Dayan A Perera
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
- Research and Development Corporation, Gus R. Douglass Institute, West Virginia State University, Institute, WV, 25112, USA
| | - Zhenkui Qin
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Ramji Odin
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Khoi Vo
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
| | - David Drescher
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Dalton Robinson
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Sheng Dong
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Dan Zhang
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Mei Shang
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
- National and Local United Engineering Laboratory for Freshwater Fish Breeding, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fisheries Sciences, Harbin, 150070, China
| | - Nermeen Abass
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Sanjay K Das
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
- ICAR Research Complex for NEH Region, Umiam, Meghalaya, 793103, India
| | - Max Bangs
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Rex A Dunham
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL, 36849, USA.
| |
Collapse
|
63
|
Wu SJ, Niknafs YS, Kim SH, Oravecz-Wilson K, Zajac C, Toubai T, Sun Y, Prasad J, Peltier D, Fujiwara H, Hedig I, Mathewson ND, Khoriaty R, Ginsburg D, Reddy P. A Critical Analysis of the Role of SNARE Protein SEC22B in Antigen Cross-Presentation. Cell Rep 2018; 19:2645-2656. [PMID: 28658614 DOI: 10.1016/j.celrep.2017.06.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 04/05/2017] [Accepted: 05/31/2017] [Indexed: 12/22/2022] Open
Abstract
Cross-presentation initiates immune responses against tumors and viral infections by presenting extracellular antigen on MHC I to activate CD8+ T cell-mediated cytotoxicity. In vitro studies in dendritic cells (DCs) established SNARE protein SEC22B as a specific regulator of cross-presentation. However, the in vivo contribution of SEC22B to cross-presentation has not been tested. To address this, we generated DC-specific Sec22b knockout (CD11c-Cre Sec22bfl/fl) mice. Contrary to the paradigm, SEC22B-deficient DCs efficiently cross-present both in vivo and in vitro. Although in vitro small hairpin RNA (shRNA)-mediated Sec22b silencing in bone-marrow-derived dendritic cells (BMDCs) reduced cross-presentation, treatment of SEC22B-deficient BMDCs with the same shRNA produced a similar defect, suggesting the Sec22b shRNA modulates cross-presentation through off-target effects. RNA sequencing of Sec22b shRNA-treated SEC22B-deficient BMDCs demonstrated several changes in the transcriptome. Our data demonstrate that contrary to the accepted model, SEC22B is not necessary for cross-presentation, cautioning against extrapolating phenotypes from knockdown studies alone.
Collapse
Affiliation(s)
- S Julia Wu
- Program in Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Medical Scientist Training Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Yashar S Niknafs
- Medical Scientist Training Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Stephanie H Kim
- Program in Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Medical Scientist Training Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Katherine Oravecz-Wilson
- Division of Hematology/Oncology, Department of Internal Medicine, Michigan Medicine, Ann Arbor, MI 48109, USA
| | - Cynthia Zajac
- Division of Hematology/Oncology, Department of Internal Medicine, Michigan Medicine, Ann Arbor, MI 48109, USA
| | - Tomomi Toubai
- Division of Hematology/Oncology, Department of Internal Medicine, Michigan Medicine, Ann Arbor, MI 48109, USA
| | - Yaping Sun
- Division of Hematology/Oncology, Department of Internal Medicine, Michigan Medicine, Ann Arbor, MI 48109, USA
| | - Jayendra Prasad
- Division of Hematology/Oncology, Department of Internal Medicine, Michigan Medicine, Ann Arbor, MI 48109, USA
| | - Daniel Peltier
- Division of Pediatric Hematology/Oncology, Department of Pediatrics and Communicable Diseases, Michigan Medicine, Ann Arbor, MI 48109, USA
| | - Hideaki Fujiwara
- Division of Hematology/Oncology, Department of Internal Medicine, Michigan Medicine, Ann Arbor, MI 48109, USA
| | - Israel Hedig
- Division of Hematology/Oncology, Department of Internal Medicine, Michigan Medicine, Ann Arbor, MI 48109, USA
| | - Nathan D Mathewson
- Dana Farber Cancer Institute, Harvard University, Cambridge, MA 02215, USA
| | - Rami Khoriaty
- Division of Hematology/Oncology, Department of Internal Medicine, Michigan Medicine, Ann Arbor, MI 48109, USA
| | - David Ginsburg
- Division of Hematology/Oncology, Department of Internal Medicine, Michigan Medicine, Ann Arbor, MI 48109, USA; Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI 48109, USA; Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Pavan Reddy
- Program in Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Division of Hematology/Oncology, Department of Internal Medicine, Michigan Medicine, Ann Arbor, MI 48109, USA.
| |
Collapse
|
64
|
Li H, Su B, Qin G, Ye Z, Alsaqufi A, Perera DA, Shang M, Odin R, Vo K, Drescher D, Robinson D, Zhang D, Abass N, Dunham RA. Salt Sensitive Tet-Off-Like Systems to Knockdown Primordial Germ Cell Genes for Repressible Transgenic Sterilization in Channel Catfish, Ictalurus punctatus. Mar Drugs 2017; 15:md15060155. [PMID: 28561774 PMCID: PMC5484105 DOI: 10.3390/md15060155] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 05/22/2017] [Accepted: 05/25/2017] [Indexed: 01/03/2023] Open
Abstract
Repressible knockdown approaches were investigated for transgenic sterilization in channel catfish, Ictalurus punctatus. Two primordial germ cell (PGC) marker genes, nanos and dead end, were targeted for knockdown, and an off-target gene, vasa, was monitored. Two potentially salt sensitive repressible promoters, zebrafish adenylosuccinate synthase 2 (ADSS) and zebrafish racemase (Rm), were each coupled with four knockdown strategies: ds-sh RNA targeting the 5′ end (N1) or 3′ end (N2) of channel catfish nanos, full-length cDNA sequence of channel catfish nanos for overexpression (cDNA) and ds-sh RNA targeting channel catfish dead end (DND). Each construct had an untreated group and treated group with sodium chloride as the repressor compound. Spawning rates of full-sibling P1 fish exposed or not exposed to the constructs as treated and untreated embryos were 93% and 59%, respectively, indicating potential sterilization of fish and repression of the constructs. Although the mRNA expression data of PGC marker genes were inconsistent in P1 fish, most F1 individuals were able to downregulate the target genes in untreated groups and repress the knockdown process in treated groups. The results indicate that repressible transgenic sterilization is feasible for reproductive control of fish, but more data from F2 or F3 are needed for evaluation.
Collapse
Affiliation(s)
- Hanbo Li
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA.
| | - Baofeng Su
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA.
- National and Local United Engineering Laboratory for Freshwater Fish Breeding, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fisheries Sciences, Harbin 150070, China.
| | - Guyu Qin
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA.
| | - Zhi Ye
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA.
| | - Ahmed Alsaqufi
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA.
- Department of Aquaculture and Animal production, King Faisal University, Al Ahsa 31982, Saudi Arabia.
| | - Dayan A Perera
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA.
- Research and Development Corporation, Gus R. Douglass Institute, West Virginia State University, WV 25112, USA.
| | - Mei Shang
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA.
- National and Local United Engineering Laboratory for Freshwater Fish Breeding, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fisheries Sciences, Harbin 150070, China.
| | - Ramjie Odin
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA.
| | - Khoi Vo
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA.
| | - David Drescher
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA.
| | - Dalton Robinson
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA.
| | - Dan Zhang
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA.
| | - Nermeen Abass
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA.
- Department of Agricultural Botany, Faculty of Agriculture Saba-Basha, Alexandria University, Alexandria City, P.O. Box 2153, Egypt.
| | - Rex A Dunham
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA.
| |
Collapse
|
65
|
Pineda RH, Nedumaran B, Hypolite J, Pan XQ, Wilson S, Meacham RB, Malykhina AP. Altered expression and modulation of the two-pore-domain (K 2P) mechanogated potassium channel TREK-1 in overactive human detrusor. Am J Physiol Renal Physiol 2017; 313:F535-F546. [PMID: 28539337 DOI: 10.1152/ajprenal.00638.2016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 05/10/2017] [Accepted: 05/18/2017] [Indexed: 01/25/2023] Open
Abstract
Detrusor overactivity (DO) is the abnormal response of the urinary bladder to physiological stretch during the filling phase of the micturition cycle. The mechanisms of bladder smooth muscle compliance upon the wall stretch are poorly understood. We previously reported that the function of normal detrusor is regulated by TREK-1, a member of the mechanogated subfamily of two-pore-domain potassium (K2P) channels. In the present study, we aimed to identify the changes in expression and function of TREK-1 channels under pathological conditions associated with DO, evaluate the potential relationship between TREK-1 channels and cytoskeletal proteins in the human bladder, and test the possibility of modulation of TREK-1 channel expression by small RNAs. Expression of TREK-1 channels in DO specimens was 2.7-fold decreased compared with control bladders and was associated with a significant reduction of the recorded TREK-1 currents. Isolated DO muscle strips failed to relax when exposed to a TREK-1 channel opener. Immunocytochemical labeling revealed close association of TREK-1 channels with cell cytoskeletal proteins and caveolins, with caveolae microdomains being severely disrupted in DO specimens. Small activating RNA (saRNA) tested in vitro provided evidence that expression of TREK-1 protein could be partially upregulated. Our data confirmed a significant downregulation of TREK-1 expression in human DO specimens and provided evidence of close association between the channel, cell cytoskeleton, and caveolins. Upregulation of TREK-1 expression by saRNA could be a future step for the development of in vivo pharmacological and genetic approaches to treat DO in humans.
Collapse
Affiliation(s)
- Ricardo H Pineda
- Division of Urology, Department of Surgery, University of Colorado Denver, Aurora, Colorado; and
| | - Balachandar Nedumaran
- Division of Urology, Department of Surgery, University of Colorado Denver, Aurora, Colorado; and
| | - Joseph Hypolite
- Division of Urology, Department of Surgery, University of Colorado Denver, Aurora, Colorado; and
| | - Xiao-Qing Pan
- Division of Urology, Department of Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Shandra Wilson
- Division of Urology, Department of Surgery, University of Colorado Denver, Aurora, Colorado; and
| | - Randall B Meacham
- Division of Urology, Department of Surgery, University of Colorado Denver, Aurora, Colorado; and
| | - Anna P Malykhina
- Division of Urology, Department of Surgery, University of Colorado Denver, Aurora, Colorado; and
| |
Collapse
|
66
|
Herrera-Carrillo E, Berkhout B. Novel AgoshRNA molecules for silencing of the CCR5 co-receptor for HIV-1 infection. PLoS One 2017; 12:e0177935. [PMID: 28542329 PMCID: PMC5443530 DOI: 10.1371/journal.pone.0177935] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 05/05/2017] [Indexed: 12/20/2022] Open
Abstract
Allogeneic transplantation of blood stem cells from a CCR5-Δ32 homozygous donor to an HIV-infected individual, the "Berlin patient", led to a cure. Since then there has been a search for approaches that mimic this intervention in a gene therapy setting. RNA interference (RNAi) has evolved as a powerful tool to regulate gene expression in a sequence-specific manner and can be used to inactivate the CCR5 mRNA. Short hairpin RNA (shRNA) molecules can impair CCR5 expression, but these molecules may cause unintended side effects and they will not be processed in cells that lack Dicer, such as monocytes. Dicer-independent RNAi pathways have opened opportunities for new AgoshRNA designs that rely exclusively on Ago2 for maturation. Furthermore, AgoshRNA processing yields a single active guide RNA, thus reducing off-target effects. In this study, we tested different AgoshRNA designs against CCR5. We selected AgoshRNAs that potently downregulated CCR5 expression on human T cells and peripheral blood mononuclear cells (PBMC) and that had no apparent adverse effect on T cell development as assessed in a competitive cell growth assay. CCR5 knockdown significantly protected T cells from CCR5 tropic HIV-1 infection.
Collapse
Affiliation(s)
- Elena Herrera-Carrillo
- Laboratory of Experimental Virology, Department of Medical Microbiology, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Ben Berkhout
- Laboratory of Experimental Virology, Department of Medical Microbiology, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
67
|
Feldcamp LA, Boutros PC, Raymond R, Fletcher PJ, Nobrega JN, Wong AHC. Pdxdc1 modulates prepulse inhibition of acoustic startle in the mouse. Transl Psychiatry 2017; 7:e1125. [PMID: 28485732 PMCID: PMC5534953 DOI: 10.1038/tp.2017.85] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Revised: 03/11/2017] [Accepted: 03/16/2017] [Indexed: 12/12/2022] Open
Abstract
Current antipsychotic medications used to treat schizophrenia all target the dopamine D2 receptor. Although these drugs have serious side effects and limited efficacy, no novel molecular targets for schizophrenia treatment have been successfully translated into new medications. To identify novel potential treatment targets for schizophrenia, we searched for previously unknown molecular modulators of acoustic prepulse inhibition (PPI), a schizophrenia endophenotype, in the mouse. We examined six inbred mouse strains that have a range of PPI, and used microarrays to determine which mRNA levels correlated with PPI across these mouse strains. We examined several brain regions involved in PPI and schizophrenia: hippocampus, striatum, and brainstem, found a number of transcripts that showed good correlation with PPI level, and confirmed this with real-time quantitative PCR. We then selected one candidate gene for further study, Pdxdc1 (pyridoxal-dependent decarboxylase domain containing 1), because it is a putative enzyme that could metabolize catecholamine neurotransmitters, and thus might be a feasible target for new medications. We determined that Pdxdc1 mRNA and protein are both strongly expressed in the hippocampus and levels of Pdxdc1 are inversely correlated with PPI across the six mouse strains. Using shRNA packaged in a lentiviral vector, we suppressed Pdxdc1 protein levels in the hippocampus and increased PPI by 70%. Our results suggest that Pdxdc1 may regulate PPI and could be a good target for further investigation as a potential treatment for schizophrenia.
Collapse
Affiliation(s)
- L A Feldcamp
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Campbell Family Mental Health Research Institute, The Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - P C Boutros
- Ontario Institute for Cancer Research, Toronto, ON, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - R Raymond
- Neuroimaging Section, The Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - P J Fletcher
- Campbell Family Mental Health Research Institute, The Centre for Addiction and Mental Health, Toronto, ON, Canada.,Department of Psychology, University of Toronto, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - J N Nobrega
- Neuroimaging Section, The Centre for Addiction and Mental Health, Toronto, ON, Canada.,Department of Psychology, University of Toronto, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - A H C Wong
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Campbell Family Mental Health Research Institute, The Centre for Addiction and Mental Health, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
68
|
Kanungo J. Puromycin-resistant lentiviral control shRNA vector, pLKO.1 induces unexpected cellular differentiation of P19 embryonic stem cells. Biochem Biophys Res Commun 2017; 486:481-485. [PMID: 28322785 DOI: 10.1016/j.bbrc.2017.03.066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 03/15/2017] [Indexed: 10/19/2022]
Abstract
RNA silencing is used as a common method for investigating loss-of-function effects of genes of interest. In mammalian cells, RNA interference (RNAi) or RNA silencing can be achieved by transient siRNA (small or short interfering RNA) transfection or by stable shRNA (short hairpin RNA) systems. Various vectors are used for efficient delivery of shRNA. Lentiviral vectors offer an efficient delivery system for stable and long-term expression of the shRNA in mammalian cells. The widely used lentiviral pLKO.1 plasmid vector is very popular in RNAi studies. A large RNAi database, a TRC (the RNAi Consortium) library, was established based on the pLKO.1-TRC plasmid vector. This plasmid (also called pLKO.1-puro) has a puromycin-resistant gene for selection in mammalian cells along with designs for generating lentiviral particles as well for RNA silencing. While using the pLKO.1-puro TRC control shRNA plasmid for transfection in murine P19 embryonic stem (ES) cells, it was unexpectedly discovered that this plasmid vector induced robust endodermal differentiation. Since P19 ES cells are pluripotent and respond to external stimuli that have the potential to alter the phenotype and thus its stemness, other cell types used in RNA silencing studies do not display the obvious effect and therefore, may affect experiments in subtle ways that would go undetected. This study for the first time provides evidence that raises concern and warrants extreme caution while using the pLKO.1-puro control shRNA vector because of its unexpected non-specific effects on cellular integrity.
Collapse
Affiliation(s)
- Jyotshna Kanungo
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR 72079, USA.
| |
Collapse
|
69
|
Adeola OA. Treatment of Influenza: Prospects of Post-Transcriptional Gene Silencing Through Synthetic siRNAs. EXPLORATORY RESEARCH AND HYPOTHESIS IN MEDICINE 2017; 2:1-2. [DOI: 10.14218/erhm.2016.00013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
70
|
Moulton JD. Using Morpholinos to Control Gene Expression. CURRENT PROTOCOLS IN NUCLEIC ACID CHEMISTRY 2017; 68:4.30.1-4.30.29. [PMID: 28252184 PMCID: PMC7162182 DOI: 10.1002/cpnc.21] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Morpholino oligonucleotides are stable, uncharged, water-soluble molecules used to block complementary sequences of RNA, preventing processing, read-through, or protein binding at those sites. Morpholinos are typically used to block translation of mRNA and to block splicing of pre-mRNA, though they can block other interactions between biological macromolecules and RNA. Morpholinos are effective, specific, and lack non-antisense effects. They work in any cell that transcribes and translates RNA, but must be delivered into the nuclear/cytosolic compartment to be effective. Morpholinos form stable base pairs with complementary nucleic acid sequences but apparently do not bind to proteins to a significant extent. They are not recognized by any proteins and do not undergo protein-mediated catalysis-nor do they mediate RNA cleavage by RNase H or the RISC complex. This work focuses on techniques and background for using Morpholinos. © 2017 by John Wiley & Sons, Inc.
Collapse
|
71
|
|
72
|
Bradford BJ, Cooper CA, Tizard ML, Doran TJ, Hinton TM. RNA interference-based technology: what role in animal agriculture? ANIMAL PRODUCTION SCIENCE 2017. [DOI: 10.1071/an15437] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Animal agriculture faces a broad array of challenges, ranging from disease threats to adverse environmental conditions, while attempting to increase productivity using fewer resources. RNA interference (RNAi) is a biological phenomenon with the potential to provide novel solutions to some of these challenges. Discovered just 20 years ago, the mechanisms underlying RNAi are now well described in plants and animals. Intracellular double-stranded RNA triggers a conserved response that leads to cleavage and degradation of complementary mRNA strands, thereby preventing production of the corresponding protein product. RNAi can be naturally induced by expression of endogenous microRNA, which are critical in the regulation of protein synthesis, providing a mechanism for rapid adaptation of physiological function. This endogenous pathway can be co-opted for targeted RNAi either through delivery of exogenous small interfering RNA (siRNA) into target cells or by transgenic expression of short hairpin RNA (shRNA). Potentially valuable RNAi targets for livestock include endogenous genes such as developmental regulators, transcripts involved in adaptations to new physiological states, immune response mediators, and also exogenous genes such as those encoded by viruses. RNAi approaches have shown promise in cell culture and rodent models as well as some livestock studies, but technical and market barriers still need to be addressed before commercial applications of RNAi in animal agriculture can be realised. Key challenges for exogenous delivery of siRNA include appropriate formulation for physical delivery, internal transport and eventual cellular uptake of the siRNA; additionally, rigorous safety and residue studies in target species will be necessary for siRNA delivery nanoparticles currently under evaluation. However, genomic incorporation of shRNA can overcome these issues, but optimal promoters to drive shRNA expression are needed, and genetic engineering may attract more resistance from consumers than the use of exogenous siRNA. Despite these hurdles, the convergence of greater understanding of RNAi mechanisms, detailed descriptions of regulatory processes in animal development and disease, and breakthroughs in synthetic chemistry and genome engineering has created exciting possibilities for using RNAi to enhance the sustainability of animal agriculture.
Collapse
|
73
|
Michler T, Große S, Mockenhaupt S, Röder N, Stückler F, Knapp B, Ko C, Heikenwalder M, Protzer U, Grimm D. Blocking sense-strand activity improves potency, safety and specificity of anti-hepatitis B virus short hairpin RNA. EMBO Mol Med 2016; 8:1082-98. [PMID: 27473329 PMCID: PMC5009812 DOI: 10.15252/emmm.201506172] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Hepatitis B virus (HBV) is a promising target for therapies based on RNA interference (RNAi) since it replicates via RNA transcripts that are vulnerable to RNAi silencing. Clinical translation of RNAi technology, however, requires improvements in potency, specificity and safety. To this end, we systematically compared different strategies to express anti-HBV short hairpin RNA (shRNA) in a pre-clinical immunocompetent hepatitis B mouse model. Using recombinant Adeno-associated virus (AAV) 8 vectors for delivery, we either (i) embedded the shRNA in an artificial mi(cro)RNA under a liver-specific promoter; (ii) co-expressed Argonaute-2, a rate-limiting cellular factor whose saturation with excess RNAi triggers can be toxic; or (iii) co-delivered a decoy ("TuD") directed against the shRNA sense strand to curb off-target gene regulation. Remarkably, all three strategies minimised adverse side effects as compared to a conventional shRNA vector that caused weight loss, liver damage and dysregulation of > 100 hepatic genes. Importantly, the novel AAV8 vector co-expressing anti-HBV shRNA and TuD outperformed all other strategies regarding efficiency and persistence of HBV knock-down, thus showing substantial promise for clinical translation.
Collapse
Affiliation(s)
- Thomas Michler
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, München, Germany German Center for Infection Research (DZIF), partner site München, München, Germany
| | - Stefanie Große
- Department of Infectious Diseases/Virology, Cluster of Excellence CellNetworks, Heidelberg University Hospital, Heidelberg, Germany BioQuant, University of Heidelberg, Heidelberg, Germany
| | - Stefan Mockenhaupt
- Department of Infectious Diseases/Virology, Cluster of Excellence CellNetworks, Heidelberg University Hospital, Heidelberg, Germany BioQuant, University of Heidelberg, Heidelberg, Germany
| | - Natalie Röder
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, München, Germany
| | - Ferdinand Stückler
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Bettina Knapp
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Chunkyu Ko
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, München, Germany
| | - Mathias Heikenwalder
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, München, Germany
| | - Ulrike Protzer
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, München, Germany German Center for Infection Research (DZIF), partner site München, München, Germany
| | - Dirk Grimm
- Department of Infectious Diseases/Virology, Cluster of Excellence CellNetworks, Heidelberg University Hospital, Heidelberg, Germany BioQuant, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
74
|
Swamy MN, Wu H, Shankar P. Recent advances in RNAi-based strategies for therapy and prevention of HIV-1/AIDS. Adv Drug Deliv Rev 2016; 103:174-186. [PMID: 27013255 PMCID: PMC4935623 DOI: 10.1016/j.addr.2016.03.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 03/10/2016] [Accepted: 03/11/2016] [Indexed: 12/15/2022]
Abstract
RNA interference (RNAi) provides a powerful tool to silence specific gene expression and has been widely used to suppress host factors such as CCR5 and/or viral genes involved in HIV-1 replication. Newer nuclease-based gene-editing technologies, such as zinc finger nucleases (ZFN), transcription activator-like effector nucleases (TALEN) and the clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 system, also provide powerful tools to ablate specific genes. Because of differences in co-receptor usage and the high mutability of the HIV-1 genome, a combination of host factors and viral genes needs to be suppressed for effective prevention and treatment of HIV-1 infection. Whereas the continued presence of small interfering/short hairpin RNA (si/shRNA) mediators is needed for RNAi to be effective, the continued expression of nucleases in the gene-editing systems is undesirable. Thus, RNAi provides the only practical way for expression of multiple silencers in infected and uninfected cells, which is needed for effective prevention/treatment of infection. There have been several advances in the RNAi field in terms of si/shRNA design, targeted delivery to HIV-1 susceptible cells, and testing for efficacy in preclinical humanized mouse models. Here, we comprehensively review the latest advances in RNAi technology towards prevention and treatment of HIV-1.
Collapse
Affiliation(s)
- Manjunath N Swamy
- Center of Emphasis in Infectious Disease, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA.
| | - Haoquan Wu
- Center of Emphasis in Infectious Disease, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA
| | - Premlata Shankar
- Center of Emphasis in Infectious Disease, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA.
| |
Collapse
|
75
|
Johnson E, Chase K, McGowan S, Mondo E, Pfister E, Mick E, Friedline RH, Kim JK, Sapp E, DiFiglia M, Aronin N. Safety of Striatal Infusion of siRNA in a Transgenic Huntington's Disease Mouse Model. J Huntingtons Dis 2016; 4:219-229. [PMID: 26444021 DOI: 10.3233/jhd-150163] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The immune system In Huntington's disease (HD) is activated and may overreact to some therapies. RNA interference using siRNA lowers mutant huntingtin (mHTT) protein but could increase immune responses. OBJECTIVE To examine the innate immune response following siRNA infusion into the striatum of wild-type (WT) and HD transgenic (YAC128) mice. METHODS siRNAs (2'-O-methyl phosphorothioated) were infused unilaterally into striatum of four month-old WT and YAC128 mice for 28 days. Microglia number and morphology (resting (normal), activated, dystrophic), cytokine levels, and DARPP32-positive neurons were measured in striatum immediately or 14 days post-infusion. Controls included contralateral untreated striatum, and PBS and sham treated striata. RESULTS The striata of untreated YAC128 mice had significantly fewer resting microglia and more dystrophic microglia than WT mice, but no difference from WT in the proportion of activated microglia or total number of microglia. siRNA infusion increased the total number of microglia in YAC128 mice compared to PBS treated and untreated striata and increased the proportion of activated microglia in WT and YAC128 mice compared to untreated striata and sham treated groups. Cytokine levels were low and siRNA infusion resulted in only modest changes in those levels. siRNA infusion did not change the number of DARPP32-positive neurons. CONCLUSION Findings suggest that siRNA infusion may be a safe method for lowering mHTT levels in the striatum in young animals, since treatment does not produce a robust cytokine response or cause neurotoxicity. The potential long-term effects of a sustained increase in total and activated microglia after siRNA infusion in HD mice need to be explored.
Collapse
Affiliation(s)
- Emily Johnson
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Kathryn Chase
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Sarah McGowan
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Erica Mondo
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Edith Pfister
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Eric Mick
- Department of Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, MA
| | - Randall H Friedline
- Department of Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Jason K Kim
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA.,Department of Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Ellen Sapp
- Department of Massachusetts General Hospital, Charlestown, MA
| | - Marian DiFiglia
- Department of Massachusetts General Hospital, Charlestown, MA
| | - Neil Aronin
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| |
Collapse
|
76
|
Williams A, Hayashi T, Wolozny D, Yin B, Su TC, Betenbaugh MJ, Su TP. The non-apoptotic action of Bcl-xL: regulating Ca(2+) signaling and bioenergetics at the ER-mitochondrion interface. J Bioenerg Biomembr 2016; 48:211-25. [PMID: 27155879 PMCID: PMC6737942 DOI: 10.1007/s10863-016-9664-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 04/26/2016] [Indexed: 12/20/2022]
Abstract
Bcl-2 family proteins are known to competitively regulate Ca(2+); however, the specific inter-organelle signaling pathways and related cellular functions are not fully elucidated. In this study, a portion of Bcl-xL was detected at the ER-mitochondrion interface or MAM (mitochondria-associated ER membrane) in association with type 3 inositol 1,4,5-trisphosphate receptors (IP3R3); an association facilitated by the BH4 and transmembrane domains of Bcl-xL. Moreover, increasing Bcl-xL expression enhanced transient mitochondrial Ca(2+) levels upon ER Ca(2+) depletion induced by short-term, non-apoptotic incubation with thapsigargin (Tg), while concomitantly reducing cytosolic Ca(2+) release. These mitochondrial changes appear to be IP3R3-dependent and resulted in decreased NAD/NADH ratios and higher electron transport chain oxidase activity. Interestingly, extended Tg exposure stimulated ER stress, but not apoptosis, and further enhanced TCA cycling. Indeed, confocal analysis indicated that Bcl-xL translocated to the MAM and increased its interaction with IP3R3 following extended Tg treatment. Thus, the MAM is a critical cell-signaling junction whereby Bcl-xL dynamically interacts with IP3R3 to coordinate mitochondrial Ca(2+) transfer and alters cellular metabolism in order to increase the cells' bioenergetic capacity, particularly during periods of stress.
Collapse
Affiliation(s)
- Abasha Williams
- Cellular Pathobiology Section, IRP, NIDA, NIH, DHHS, 333 Cassell Drive, Baltimore, MD, 21224, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD, 21218, USA
- Division of Biotechnology Review and Research II, FDA/CDER/OPS/OBP, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - Teruo Hayashi
- Cellular Pathobiology Section, IRP, NIDA, NIH, DHHS, 333 Cassell Drive, Baltimore, MD, 21224, USA
- Seiwakai Nishikawa Hospital, 293-2 Minato-Machi, Hamada, Shimane, 697-0052, Japan
| | - Daniel Wolozny
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD, 21218, USA
| | - Bojiao Yin
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD, 21218, USA
| | - Tzu-Chieh Su
- Cellular Pathobiology Section, IRP, NIDA, NIH, DHHS, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Michael J Betenbaugh
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD, 21218, USA.
| | - Tsung-Ping Su
- Cellular Pathobiology Section, IRP, NIDA, NIH, DHHS, 333 Cassell Drive, Baltimore, MD, 21224, USA.
| |
Collapse
|
77
|
Shatizadeh Malekshahi S, Arefian E, Salimi V, Mokhtari Azad T, Yavarian J. Potential siRNA Molecules for Nucleoprotein and M2/L Overlapping Region of Respiratory Syncytial Virus: In Silico Design. Jundishapur J Microbiol 2016; 9:e34304. [PMID: 27303618 PMCID: PMC4902852 DOI: 10.5812/jjm.34304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 12/05/2015] [Accepted: 02/16/2016] [Indexed: 11/28/2022] Open
Abstract
Background Human respiratory syncytial virus (RSV) is a leading cause of severe lower respiratory tract disease in the pediatric population, elderly and in immunosuppressed individuals. Respiratory syncytial virus is also responsible for bronchiolitis, pneumonia, and chronic obstructive pulmonary infections in all age groups. With this high disease burden and the lack of an effective RSV treatment and vaccine, there is a clear need for discovery and development of novel, effective and safe drugs to prevent and treat RSV disease. The most innovative approach is the use of small interfering RNAs (siRNAs) which represent a revolutionary new concept in human therapeutics. The nucleoprotein gene of RSV which is known as the most conserved gene and the M2/L mRNA, which encompass sixty-eight overlapping nucleotides, were selected as suitable targets for siRNA design. Objectives The present study is aimed to design potential siRNAs for silencing nucleoprotein and an overlapping region of M2-L coding mRNAs by computational analysis. Materials and Methods Various computational methods (target alignment, similarity search, secondary structure prediction, and RNA interaction calculation) have been used for siRNA designing against different strains of RSV. Results In this study, seven siRNA molecules were rationally designed against the nucleoprotein gene and validated using various computational methods for silencing different strains of RSV. Additionally, three effective siRNA molecules targeting the overlapping region of M2/L mRNA were designed. Conclusions This approach provides insight and a validated strategy for chemical synthesis of an antiviral RNA molecule which meets many sequence features for efficient silencing and treatment at the genomic level.
Collapse
Affiliation(s)
| | - Ehsan Arefian
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, IR Iran
| | - Vahid Salimi
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, IR Iran
| | - Talat Mokhtari Azad
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, IR Iran
| | - Jila Yavarian
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, IR Iran
- Corresponding author: Jila Yavarian, Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, IR Iran. Tel/Fax: +98-2188962343, E-mail:
| |
Collapse
|
78
|
Ma AC, McNulty MS, Poshusta TL, Campbell JM, Martínez-Gálvez G, Argue DP, Lee HB, Urban MD, Bullard CE, Blackburn PR, Man TK, Clark KJ, Ekker SC. FusX: A Rapid One-Step Transcription Activator-Like Effector Assembly System for Genome Science. Hum Gene Ther 2016; 27:451-63. [PMID: 26854857 PMCID: PMC4931509 DOI: 10.1089/hum.2015.172] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Transcription activator-like effectors (TALEs) are extremely effective, single-molecule DNA-targeting molecular cursors used for locus-specific genome science applications, including high-precision molecular medicine and other genome engineering applications. TALEs are used in genome engineering for locus-specific DNA editing and imaging, as artificial transcriptional activators and repressors, and for targeted epigenetic modification. TALEs as nucleases (TALENs) are effective editing tools and offer high binding specificity and fewer sequence constraints toward the targeted genome than other custom nuclease systems. One bottleneck of broader TALE use is reagent accessibility. For example, one commonly deployed method uses a multitube, 5-day assembly protocol. Here we describe FusX, a streamlined Golden Gate TALE assembly system that (1) is backward compatible with popular TALE backbones, (2) is functionalized as a single-tube 3-day TALE assembly process, (3) requires only commonly used basic molecular biology reagents, and (4) is cost-effective. More than 100 TALEN pairs have been successfully assembled using FusX, and 27 pairs were quantitatively tested in zebrafish, with each showing high somatic and germline activity. Furthermore, this assembly system is flexible and is compatible with standard molecular biology laboratory tools, but can be scaled with automated laboratory support. To demonstrate, we use a highly accessible and commercially available liquid-handling robot to rapidly and accurately assemble TALEs using the FusX TALE toolkit. Together, the FusX system accelerates TALE-based genomic science applications from basic science screening work for functional genomics testing and molecular medicine applications.
Collapse
Affiliation(s)
- Alvin C Ma
- 1 Department of Biochemistry and Molecular Biology, Mayo Clinic , Rochester, Minnesota.,2 Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong , Hong Kong
| | - Melissa S McNulty
- 1 Department of Biochemistry and Molecular Biology, Mayo Clinic , Rochester, Minnesota
| | - Tanya L Poshusta
- 1 Department of Biochemistry and Molecular Biology, Mayo Clinic , Rochester, Minnesota
| | - Jarryd M Campbell
- 1 Department of Biochemistry and Molecular Biology, Mayo Clinic , Rochester, Minnesota
| | | | - David P Argue
- 1 Department of Biochemistry and Molecular Biology, Mayo Clinic , Rochester, Minnesota
| | - Han B Lee
- 1 Department of Biochemistry and Molecular Biology, Mayo Clinic , Rochester, Minnesota
| | - Mark D Urban
- 1 Department of Biochemistry and Molecular Biology, Mayo Clinic , Rochester, Minnesota
| | - Cassandra E Bullard
- 1 Department of Biochemistry and Molecular Biology, Mayo Clinic , Rochester, Minnesota
| | - Patrick R Blackburn
- 1 Department of Biochemistry and Molecular Biology, Mayo Clinic , Rochester, Minnesota
| | - Toni K Man
- 2 Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong , Hong Kong
| | - Karl J Clark
- 1 Department of Biochemistry and Molecular Biology, Mayo Clinic , Rochester, Minnesota
| | - Stephen C Ekker
- 1 Department of Biochemistry and Molecular Biology, Mayo Clinic , Rochester, Minnesota
| |
Collapse
|
79
|
Yu-Wai-Man C, Tagalakis AD, Manunta MD, Hart SL, Khaw PT. Receptor-targeted liposome-peptide-siRNA nanoparticles represent an efficient delivery system for MRTF silencing in conjunctival fibrosis. Sci Rep 2016; 6:21881. [PMID: 26905457 PMCID: PMC4764806 DOI: 10.1038/srep21881] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 02/03/2016] [Indexed: 12/22/2022] Open
Abstract
There is increasing evidence that the Myocardin-related transcription factor/Serum response factor (MRTF/SRF) pathway plays a key role in fibroblast activation and that knocking down MRTF can lead to reduced scarring and fibrosis. Here, we have developed a receptor-targeted liposome-peptide-siRNA nanoparticle as a non-viral delivery system for MRTF-B siRNA in conjunctival fibrosis. Using 50 nM siRNA, the MRTF-B gene was efficiently silenced by 76% and 72% with LYR and LER nanoparticles, respectively. The silencing efficiency was low when non-targeting peptides or siRNA alone or liposome-siRNA alone were used. LYR and LER nanoparticles also showed higher silencing efficiency than PEGylated LYR-P and LER-P nanoparticles. The nanoparticles were not cytotoxic using different liposomes, targeting peptides, and 50 nM siRNA. Three-dimensional fibroblast-populated collagen matrices were also used as a functional assay to measure contraction in vitro, and showed that MRTF-B LYR nanoparticles completely blocked matrix contraction after a single transfection treatment. In conclusion, this is the first study to develop and show that receptor-targeted liposome-peptide-siRNA nanoparticles represent an efficient and safe non-viral siRNA delivery system that could be used to prevent fibrosis after glaucoma filtration surgery and other contractile scarring conditions in the eye.
Collapse
Affiliation(s)
- Cynthia Yu-Wai-Man
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, United Kingdom
| | - Aristides D Tagalakis
- Wolfson Centre for Gene Therapy of Childhood Disease, UCL Institute of Child Health, London, United Kingdom
| | - Maria D Manunta
- Wolfson Centre for Gene Therapy of Childhood Disease, UCL Institute of Child Health, London, United Kingdom
| | - Stephen L Hart
- Wolfson Centre for Gene Therapy of Childhood Disease, UCL Institute of Child Health, London, United Kingdom
| | - Peng T Khaw
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, United Kingdom
| |
Collapse
|
80
|
Barrett GL, Naim T, Trieu J, Huang M. In vivo knockdown of basal forebrain p75 neurotrophin receptor stimulates choline acetyltransferase activity in the mature hippocampus. J Neurosci Res 2016; 94:389-400. [DOI: 10.1002/jnr.23717] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 12/15/2015] [Accepted: 01/11/2016] [Indexed: 11/09/2022]
Affiliation(s)
- Graham L. Barrett
- Department of Physiology; University of Melbourne; Melbourne Victoria Australia
| | - Timur Naim
- Department of Physiology; University of Melbourne; Melbourne Victoria Australia
| | - Jennifer Trieu
- Department of Physiology; University of Melbourne; Melbourne Victoria Australia
| | - Mengjie Huang
- Department of Physiology; University of Melbourne; Melbourne Victoria Australia
| |
Collapse
|
81
|
Di Rocco G, Verdina A, Gatti V, Virdia I, Toietta G, Todaro M, Stassi G, Soddu S. Apoptosis induced by a HIPK2 full-length-specific siRNA is due to off-target effects rather than prevalence of HIPK2-Δe8 isoform. Oncotarget 2016; 7:1675-86. [PMID: 26625198 PMCID: PMC4811489 DOI: 10.18632/oncotarget.6423] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 11/15/2015] [Indexed: 01/05/2023] Open
Abstract
Small interfering RNAs (siRNAs) are widely used to study gene function and extensively exploited for their potential therapeutic applications. HIPK2 is an evolutionary conserved kinase that binds and phosphorylates several proteins directly or indirectly related to apoptosis. Recently, an alternatively spliced isoform skipping 81 nucleotides of exon 8 (Hipk2-Δe8) has been described. Selective depletion of Hipk2 full-length (Hipk2-FL) with a specific siRNA that spares the Hipk2-Δe8 isoform has been shown to strongly induce apoptosis, suggesting an unpredicted dominant-negative effect of Hipk2-FL over the Δe8 isoform. From this observation, we sought to take advantage and assessed the therapeutic potential of generating Hipk2 isoform unbalance in tumor-initiating cells derived from colorectal cancer patients. Strong reduction of cell viability was induced in vitro and in vivo by the originally described exon 8-specific siRNA, supporting a potential therapeutic application. However, validation analyses performed with additional exon8-specific siRNAs with different stabilities showed that all exon8-targeting siRNAs can induce comparable Hipk2 isoform unbalance but only the originally reported e8-siRNA promotes cell death. These data show that loss of viability does not depend on the prevalence of Hipk2-Δe8 isoform but it is rather due to microRNA-like off-target effects.
Collapse
Affiliation(s)
- Giuliana Di Rocco
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - Alessandra Verdina
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - Veronica Gatti
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - Ilaria Virdia
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - Gabriele Toietta
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - Matilde Todaro
- Department of Surgical and Oncological Sciences, Cellular and Molecular Pathophysiology Laboratory, University of Palermo, Palermo, Italy
| | - Giorgio Stassi
- Department of Surgical and Oncological Sciences, Cellular and Molecular Pathophysiology Laboratory, University of Palermo, Palermo, Italy
| | - Silvia Soddu
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
82
|
Roberts TC, Ezzat K, El Andaloussi S, Weinberg MS. Synthetic SiRNA Delivery: Progress and Prospects. Methods Mol Biol 2016; 1364:291-310. [PMID: 26472459 DOI: 10.1007/978-1-4939-3112-5_23] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Small interfering RNA (siRNA) is a powerful tool for modulating gene expression by RNA interference (RNAi). Duplex RNA oligonucleotides induce cleavage of homologous target transcripts, thereby enabling posttranscriptional silencing of potentially any gene. As such, siRNAs may have utility as novel pharmaceuticals for a wide range of diseases. However, a lack of "drug-likeness," physiological barriers, and potential toxicities have meant that systemic delivery of SiRNAs in vivo remains a major challenge. Here we discuss various strategies that have been employed to solve the problem of SiRNA delivery. These include chemical modification of the SiRNA, direct conjugation to bioactive moieties, and nanoparticle formulations.
Collapse
Affiliation(s)
- Thomas C Roberts
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA, 92037, USA
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Kariem Ezzat
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK
| | - Samir El Andaloussi
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK
- Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Marc S Weinberg
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA, 92037, USA.
- Antiviral Gene Therapy Research Unit, Department of Molecular Medicine and Haematology, University of the Witwatersrand Medical School, Johannesburg, WITS 2050, South Africa.
- HIV Pathogenesis Research Unit, Department of Molecular Medicine and Haematology, University of the Witwatersrand Medical School, Johannesburg, WITS 2050, South Africa.
| |
Collapse
|
83
|
Dueck H, Eberwine J, Kim J. Variation is function: Are single cell differences functionally important?: Testing the hypothesis that single cell variation is required for aggregate function. Bioessays 2015; 38:172-80. [PMID: 26625861 PMCID: PMC4738397 DOI: 10.1002/bies.201500124] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
There is a growing appreciation of the extent of transcriptome variation across individual cells of the same cell type. While expression variation may be a byproduct of, for example, dynamic or homeostatic processes, here we consider whether single-cell molecular variation per se might be crucial for population-level function. Under this hypothesis, molecular variation indicates a diversity of hidden functional capacities within an ensemble of identical cells, and this functional diversity facilitates collective behavior that would be inaccessible to a homogenous population. In reviewing this topic, we explore possible functions that might be carried by a heterogeneous ensemble of cells; however, this question has proven difficult to test, both because methods to manipulate molecular variation are limited and because it is complicated to define, and measure, population-level function. We consider several possible methods to further pursue the hypothesis that variation is function through the use of comparative analysis and novel experimental techniques.
Collapse
Affiliation(s)
- Hannah Dueck
- Genomics and Computational Biology Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
| | - James Eberwine
- Genomics and Computational Biology Graduate Group, University of Pennsylvania, Philadelphia, PA, USA.,Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, USA.,Penn Program in Single Cell Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Junhyong Kim
- Genomics and Computational Biology Graduate Group, University of Pennsylvania, Philadelphia, PA, USA.,Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, USA.,Penn Program in Single Cell Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Biology, University of Pennsylvania, Philadelphia, PA, USA.,Department of Computer and Information Science, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
84
|
Kashi K, Henderson L, Bonetti A, Carninci P. Discovery and functional analysis of lncRNAs: Methodologies to investigate an uncharacterized transcriptome. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2015; 1859:3-15. [PMID: 26477492 DOI: 10.1016/j.bbagrm.2015.10.010] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 10/08/2015] [Accepted: 10/13/2015] [Indexed: 01/15/2023]
Abstract
It is known that more than 70% of mammalian genomes are transcribed, yet the vast majority of transcripts do not code for proteins. Are these noncoding transcripts merely transcriptional noise, or do they serve a biological purpose? Recent developments in genomic analysis technologies, especially sequencing methods, have allowed researchers to create a large atlas of transcriptomes, study subcellular localization, and investigate potential interactions with proteins for a growing number of transcripts. Here, we review the current methodologies available for discovering and investigating functions of long noncoding RNAs (lncRNAs), which require a wide variety of applications to study their potential biological roles. This article is part of a Special Issue entitled: Clues to long noncoding RNA taxonomy1, edited by Dr. Tetsuro Hirose and Dr. Shinichi Nakagawa.
Collapse
Affiliation(s)
- Kaori Kashi
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, RIKEN Yokohama Campus, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Lindsey Henderson
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, RIKEN Yokohama Campus, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Alessandro Bonetti
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, RIKEN Yokohama Campus, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Piero Carninci
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, RIKEN Yokohama Campus, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan.
| |
Collapse
|
85
|
Tantikanjana T, Nasrallah JB. Ligand-Mediated cis-Inhibition of Receptor Signaling in the Self-Incompatibility Response of the Brassicaceae. PLANT PHYSIOLOGY 2015; 169:1141-54. [PMID: 26269543 PMCID: PMC4587449 DOI: 10.1104/pp.15.00572] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 08/11/2015] [Indexed: 05/02/2023]
Abstract
The inhibition of self-pollination in self-incompatible Brassicaceae is based on allele-specific trans-activation of the highly polymorphic S-locus receptor kinase (SRK), which is displayed at the surface of stigma epidermal cells, by its even more polymorphic pollen coat-localized ligand, the S-locus cysteine-rich (SCR) protein. In an attempt to achieve constitutive activation of SRK and thus facilitate analysis of self-incompatibility (SI) signaling, we coexpressed an Arabidopsis lyrata SCR variant with its cognate SRK receptor in the stigma epidermal cells of Arabidopsis (Arabidopsis thaliana) plants belonging to the C24 accession, in which expression of SRK and SCR had been shown to exhibit a robust SI response. Contrary to expectation, however, coexpression of SRK and SCR was found to inhibit SRK-mediated signaling and to disrupt the SI response. This phenomenon, called cis-inhibition, is well documented in metazoans but has not as yet been reported for plant receptor kinases. We demonstrate that cis-inhibition of SRK, like its trans-activation, is based on allele-specific interaction between receptor and ligand. We also show that stigma-expressed SCR causes entrapment of its SRK receptor in the endoplasmic reticulum, thus disrupting the proper targeting of SRK to the plasma membrane, where the receptor would be available for productive interaction with its pollen coat-derived SCR ligand. Although based on an artificial cis-inhibition system, the results suggest novel strategies of pollination control for the generation of hybrid cultivars and large-scale seed production from hybrid plants in Brassicaceae seed crops and, more generally, for inhibiting cell surface receptor function and manipulating signaling pathways in plants.
Collapse
Affiliation(s)
- Titima Tantikanjana
- Section of Plant Biology, School of Integrative Plant Sciences, Cornell University, Ithaca, New York 14953
| | - June B Nasrallah
- Section of Plant Biology, School of Integrative Plant Sciences, Cornell University, Ithaca, New York 14953
| |
Collapse
|
86
|
siRNA Versus miRNA as Therapeutics for Gene Silencing. MOLECULAR THERAPY. NUCLEIC ACIDS 2015; 4:e252. [PMID: 26372022 PMCID: PMC4877448 DOI: 10.1038/mtna.2015.23] [Citation(s) in RCA: 703] [Impact Index Per Article: 70.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 07/18/2015] [Indexed: 02/06/2023]
Abstract
Discovered a little over two decades ago, small interfering RNAs (siRNAs) and microRNAs (miRNAs) are noncoding RNAs with important roles in gene regulation. They have recently been investigated as novel classes of therapeutic agents for the treatment of a wide range of disorders including cancers and infections. Clinical trials of siRNA- and miRNA-based drugs have already been initiated. siRNAs and miRNAs share many similarities, both are short duplex RNA molecules that exert gene silencing effects at the post-transcriptional level by targeting messenger RNA (mRNA), yet their mechanisms of action and clinical applications are distinct. The major difference between siRNAs and miRNAs is that the former are highly specific with only one mRNA target, whereas the latter have multiple targets. The therapeutic approaches of siRNAs and miRNAs are therefore very different. Hence, this review provides a comparison between therapeutic siRNAs and miRNAs in terms of their mechanisms of action, physicochemical properties, delivery, and clinical applications. Moreover, the challenges in developing both classes of RNA as therapeutics are also discussed.
Collapse
|
87
|
Deciphering Seed Sequence Based Off-Target Effects in a Large-Scale RNAi Reporter Screen for E-Cadherin Expression. PLoS One 2015; 10:e0137640. [PMID: 26361354 PMCID: PMC4567318 DOI: 10.1371/journal.pone.0137640] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 08/19/2015] [Indexed: 12/28/2022] Open
Abstract
Functional RNAi based screening is affected by large numbers of false positive and negative hits due to prevalent sequence based off-target effects. We performed a druggable genome targeting siRNA screen intended to identify novel regulators of E-cadherin (CDH1) expression, a known key player in epithelial mesenchymal transition (EMT). Analysis of primary screening results indicated a large number of false-positive hits. To address these crucial difficulties we developed an analysis method, SENSORS, which, similar to published methods, is a seed enrichment strategy for analyzing siRNA off-targets in RNAi screens. Using our approach, we were able to demonstrate that accounting for seed based off-target effects stratifies primary screening results and enables the discovery of additional screening hits. While traditional hit detection methods are prone to false positive results which are undetected, we were able to identify false positive hits robustly. Transcription factor MYBL1 was identified as a putative novel target required for CDH1 expression and verified experimentally. No siRNA pool targeting MYBL1 was present in the used siRNA library. Instead, MYBL1 was identified as a putative CDH1 regulating target solely based on the SENSORS off-target score, i.e. as a gene that is a cause for off-target effects down regulating E-cadherin expression.
Collapse
|
88
|
Rodrigues AF, Guerreiro MR, Formas-Oliveira AS, Fernandes P, Blechert AK, Genzel Y, Alves PM, Hu WS, Coroadinha AS. Increased titer and reduced lactate accumulation in recombinant retrovirus production through the down-regulation of HIF1 and PDK. Biotechnol Bioeng 2015; 113:150-62. [PMID: 26134455 DOI: 10.1002/bit.25691] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 06/15/2015] [Accepted: 06/24/2015] [Indexed: 12/15/2022]
Abstract
Many mammalian cell lines used in the manufacturing of biopharmaceuticals exhibit high glycolytic flux predominantly channeled to the production of lactate. The accumulation of lactate in culture reduces cell viability and may also decrease product quality. In this work, we engineered a HEK 293 derived cell line producing a recombinant gene therapy retroviral vector, by down-regulating hypoxia inducible factor 1 (HIF1) and pyruvate dehydrogenase kinase (PDK). Specific productivity of infectious viral titers could be increased more than 20-fold for single gene knock-down (HIF1 or PDK) and more than 30-fold under combined down-regulation. Lactate production was reduced up to 4-fold. However, the reduction in lactate production, alone, was not sufficient to enhance the titer: high-titer clones also showed significant enrollment of metabolic routes not related to lactate production. Transcriptome analysis indicated activation of biological amines metabolism, detoxification routes, including glutathione metabolism, pentose phosphate pathway, glycogen biosynthesis and amino acid catabolism. The latter were validated by enzyme activity assays and metabolite profiling, respectively. High-titer clones also presented substantially increased transcript levels of the viral genes expression cassettes. The results herein presented demonstrate the impact of HIF1 and PDK down-regulation on the production performance of a mammalian cell line, reporting one of the highest fold-increase in specific productivity of infectious virus titers achieved by metabolic engineering. They additionally highlight the contribution of secondary pathways, beyond those related to lactate production, that can be also explored to pursue improved metabolic status favoring a high-producing phenotype.
Collapse
Affiliation(s)
- A F Rodrigues
- Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - M R Guerreiro
- Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - A S Formas-Oliveira
- Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - P Fernandes
- Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - A-K Blechert
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering Group, Magdeburg, Germany
| | - Y Genzel
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering Group, Magdeburg, Germany
| | - P M Alves
- Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - W S Hu
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, USA
| | - A S Coroadinha
- Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal. .,Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal.
| |
Collapse
|
89
|
Fujino T, Kogashi K, Okada K, Mattarella M, Suzuki T, Yasumoto K, Sogawa K, Isobe H. Chimeric RNA Oligonucleotides with Triazole and Phosphate Linkages: Synthesis and RNA Interference. Chem Asian J 2015; 10:2683-8. [PMID: 26248050 DOI: 10.1002/asia.201500765] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Indexed: 11/12/2022]
Abstract
Chimeric RNA oligonucleotides with an artificial triazole linker were synthesized using solution-phase click chemistry and solid-phase automated synthesis. Scalable synthesis methods for jointing units for the chimeric structure have been developed, and after click-coupling of the jointing units with triazole linkers, a series of chimeric oligonucleotides was prepared by utilizing the well-established phosphoramidite method for the elongation. The series of chimeric 21-mer oligonucleotides that possessed the triazole linker at different strands and positions allowed for a screening study of the RNA interference to clarify the preference of the triazole modifications in small-interfering RNA molecules.
Collapse
Affiliation(s)
- Tomoko Fujino
- Department of Chemistry, Tohoku University, Aoba-ku, Sendai, 980-8578, Japan
| | - Kanako Kogashi
- Department of Chemistry, Tohoku University, Aoba-ku, Sendai, 980-8578, Japan
| | - Koudai Okada
- Department of Chemistry, Tohoku University, Aoba-ku, Sendai, 980-8578, Japan
| | - Martin Mattarella
- Department of Chemistry, Tohoku University, Aoba-ku, Sendai, 980-8578, Japan
| | - Takeru Suzuki
- Department of Chemistry, Tohoku University, Aoba-ku, Sendai, 980-8578, Japan
| | - Kenichi Yasumoto
- Department of Biomolecular Sciences, Tohoku University, Aoba-ku, Sendai, 980-8578, Japan
| | - Kazuhiro Sogawa
- Department of Biomolecular Sciences, Tohoku University, Aoba-ku, Sendai, 980-8578, Japan
| | - Hiroyuki Isobe
- Department of Chemistry, Tohoku University, Aoba-ku, Sendai, 980-8578, Japan. .,JST, ERATO, Isobe Degenerate π-Integration Project and Advanced Institute for Materials Research, Tohoku University, Aoba-ku, Sendai, 980-8577, Japan.
| |
Collapse
|
90
|
Bhardwaj V. Villain of Molecular Biology: Why are we not reproducible in research? F1000Res 2015; 4:438. [PMID: 26339478 PMCID: PMC4544406 DOI: 10.12688/f1000research.6854.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/28/2015] [Indexed: 11/20/2022] Open
Abstract
Worldwide, there is an issue of irreproducibility in life science research. In the USA alone $28 billion per year spent on preclinical research is not reproducible. Within this opinion article, I provide a brief historical account of the discovery of the Watson-Crick DNA model and introduce another neglected model of DNA. This negligence may be one of the fundamental reasons behind irreproducibility in molecular biology research.
Collapse
Affiliation(s)
- Vikash Bhardwaj
- Molecular Biology and Genetics Domain, Lovely Professional University, Punjab, India
| |
Collapse
|
91
|
Mukherjee A, Bhattacharyya J, Sagar MV, Chaudhuri A. Liposomally encapsulated CDC20 siRNA inhibits both solid melanoma tumor growth and spontaneous growth of intravenously injected melanoma cells on mouse lung. Drug Deliv Transl Res 2015; 3:224-34. [PMID: 25788131 DOI: 10.1007/s13346-013-0141-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Cell division cycle homologue 20 (CDC20), a key cell cycle regulator required for the completion of mitosis in organisms from yeast to human, is highly expressed in several carcinomas. Recent studies have shown that specific knockdown of CDC20 expression is capable of significantly inhibiting the growth of human pancreatic carcinoma cells. However, preclinical studies aimed at demonstrating the therapeutic potential of CDC20 siRNA in combating tumor growth has not yet been reported. Herein, in a syngeneic C57BL/6J mouse tumor model, we show that intraperitoneal administration of a 19-bp synthetic CDC20 siRNA encapsulated within liposomes of guanidinylated cationic amphiphile with stearyl tails inhibits solid melanoma (B16F10) tumor growth. In addition, using a spontaneous lung metastasis model in C57BL/6J mice, we show that intravenous administration of the same liposomally encapsulated 19-bp synthetic CDC20 siRNA inhibits B16F10 melanoma growth on mouse lung. Liposomally bound CDC20 siRNA was found to be efficient in silencing the expression of CDC20 in B16F10 cells at both protein and mRNA levels. Findings in the flow cytometric studies confirmed the presence of significantly enhanced populations of G2/M phase in cells treated with liposomally bound CDC20 siRNA. To the best of our knowledge, the present findings demonstrate, for the first time, systemic use of CDC20 siRNA in inhibiting mouse tumor growth.
Collapse
Affiliation(s)
- Anubhab Mukherjee
- Division of Lipid Science and Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India
| | | | | | | |
Collapse
|
92
|
Evaluation of canonical siRNA and Dicer substrate RNA for inhibition of hepatitis C virus genome replication--a comparative study. PLoS One 2015; 10:e0117742. [PMID: 25705875 PMCID: PMC4338191 DOI: 10.1371/journal.pone.0117742] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Accepted: 01/01/2015] [Indexed: 01/06/2023] Open
Abstract
Hepatitis C virus (HCV) frequently establishes persistent infections in the liver, leading to the development of chronic hepatitis and, potentially, to liver cirrhosis and hepatocellular carcinoma at later stages. The objective of this study was to test the ability of five Dicer substrate siRNAs (DsiRNA) to inhibit HCV replication and to compare these molecules to canonical 21 nt siRNA. DsiRNA molecules were designed to target five distinct regions of the HCV genome - the 5' UTR and the coding regions for NS3, NS4B, NS5A or NS5B. These molecules were transfected into Huh7.5 cells that stably harboured an HCV subgenomic replicon expressing a firefly luciferase/neoR reporter (SGR-Feo-JFH-1) and were also tested on HCVcc-infected cells. All of the DsiRNAs inhibited HCV replication in both the subgenomic system and HCVcc-infected cells. When DsiRNAs were transfected prior to infection with HCVcc, the inhibition levels reached 99.5%. When directly compared, canonical siRNA and DsiRNA exhibited similar potency of virus inhibition. Furthermore, both types of molecules exhibited similar dynamics of inhibition and frequencies of resistant mutants after 21 days of treatment. Thus, DsiRNA molecules are as potent as 21 nt siRNAs for the inhibition of HCV replication and may provide future approaches for HCV therapy if the emergence of resistant mutants can be addressed.
Collapse
|
93
|
Zhou N, Ding B, Agler M, Cockett M, McPhee F. Lethality of PAK3 and SGK2 shRNAs to human papillomavirus positive cervical cancer cells is independent of PAK3 and SGK2 knockdown. PLoS One 2015; 10:e0117357. [PMID: 25615606 PMCID: PMC4304782 DOI: 10.1371/journal.pone.0117357] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 12/22/2014] [Indexed: 01/23/2023] Open
Abstract
The p21-activated kinase 3 (PAK3) and the serum and glucocorticoid-induced kinase 2 (SGK2) have been previously proposed as essential kinases for human papillomavirus positive (HPV+) cervical cancer cell survival. This was established using a shRNA knockdown approach. To validate PAK3 and SGK2 as potential targets for HPV+ cervical cancer therapy, the relationship between shRNA-induced phenotypes in HPV+ cervical cancer cells and PAK3 or SGK2 knockdown was carefully examined. We observed that the phenotypes of HPV+ cervical cancer cells induced by various PAK3 and SGK2 shRNAs could not be rescued by complement expression of respective cDNA constructs. A knockdown-deficient PAK3 shRNA with a single mismatch was sufficient to inhibit HeLa cell growth to a similar extent as wild-type PAK3 shRNA. The HPV+ cervical cancer cells were also susceptible to several non-human target shRNAs. The discrepancy between PAK3 and SGK2 shRNA-induced apoptosis and gene expression knockdown, as well as cell death stimulation, suggested that these shRNAs killed HeLa cells through different pathways that may not be target-specific. These data demonstrated that HPV+ cervical cancer cell death was not associated with RNAi-induced PAK3 and SGK2 knockdown but likely through off-target effects.
Collapse
Affiliation(s)
- Nannan Zhou
- Department of Virology, R&D of Bristol-Myers Squibb Company, Wallingford, Connecticut, United States of America
| | - Bo Ding
- Department of Virology, R&D of Bristol-Myers Squibb Company, Wallingford, Connecticut, United States of America
| | - Michele Agler
- Department of Leads Discovery, R&D of Bristol-Myers Squibb Company, Wallingford, Connecticut, United States of America
| | - Mark Cockett
- Department of Virology, R&D of Bristol-Myers Squibb Company, Wallingford, Connecticut, United States of America
| | - Fiona McPhee
- Department of Virology, R&D of Bristol-Myers Squibb Company, Wallingford, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
94
|
Qiu L, Tan EK, Zeng L. microRNAs and Neurodegenerative Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 888:85-105. [PMID: 26663180 DOI: 10.1007/978-3-319-22671-2_6] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
microRNAs (miRNAs) are small, noncoding RNA molecules that through imperfect base-pairing with complementary sequences of target mRNA molecules, typically cleave target mRNA, causing subsequent degradation or translation inhibition. Although an increasing number of studies have identified misregulated miRNAs in the neurodegenerative diseases (NDDs) Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis, which suggests that alterations in the miRNA regulatory pathway could contribute to disease pathogenesis, the molecular mechanisms underlying the pathological implications of misregulated miRNA expression and the regulation of the key genes involved in NDDs remain largely unknown. In this chapter, we provide evidence of the function and regulation of miRNAs and their association with the neurological events in NDDs. This will help improve our understanding of how miRNAs govern the biological functions of key pathogenic genes in these diseases, which potentially regulate several pathways involved in the progression of neurodegeneration. Additionally, given the growing interest in the therapeutic potential of miRNAs, we discuss current clinical challenges to developing miRNA-based therapeutics for NDDs.
Collapse
Affiliation(s)
- Lifeng Qiu
- Neural Stem Cell Research Lab, Department of Research, National Neuroscience Institute, Singapore, 308433, Singapore
| | - Eng King Tan
- Department of Neurology, National Neuroscience Institute, SGH Campus, Singapore, 169856, Singapore
- Department of Research, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433, Singapore
- Neuroscience and Behavioral Disorders program, Duke-National University of Singapore, Graduate Medical School, Singapore, 169857, Singapore
| | - Li Zeng
- Neural Stem Cell Research Lab, Department of Research, National Neuroscience Institute, Singapore, 308433, Singapore.
- Department of Research, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433, Singapore.
- Neuroscience and Behavioral Disorders program, Duke-National University of Singapore, Graduate Medical School, Singapore, 169857, Singapore.
| |
Collapse
|
95
|
Sharma VK, Kushwaha N, Basu S, Singh AK, Chakraborty S. Identification of siRNA generating hot spots in multiple viral suppressors to generate broad-spectrum antiviral resistance in plants. PHYSIOLOGY AND MOLECULAR BIOLOGY OF PLANTS : AN INTERNATIONAL JOURNAL OF FUNCTIONAL PLANT BIOLOGY 2015; 21:9-18. [PMID: 25648440 PMCID: PMC4312327 DOI: 10.1007/s12298-014-0264-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 09/16/2014] [Accepted: 09/22/2014] [Indexed: 05/27/2023]
Abstract
Viruses are one of the most devastating plant pathogens causing severe economic losses worldwide. RNA silencing is a robust technology to knock down the expression of specific genes. This mechanism can be exploited to generate virus resistant plants through expression of the viral derived sequences. Viruses in turn have evolved to encode suppressors of RNA silencing to combat host defense. Mixed infection of plants is of common occurrence in nature and simultaneous targeting of suppressor(s) of multiple viruses offers an effective strategy. In this study, we have in silico designed siRNAs against suppressors of the two most devastating viruses of tomato, leaf curl causing tomato begomoviruses and Cucumber mosaic virus. Three different siRNA prediction programs were used to evaluate siRNAs generating capability of each sequence and common putative candidate siRNAs were selected fulfilling the stringent parameters. Our results indicated that in the case of each suppressor a particular region of 100-150 base pairs could be source of potent siRNAs referred as hotspots. Expression of these viral hot spots as a single construct in the plants would facilitate development of transgenic plants with a high degree of broad spectrum resistance against multiple viruses.
Collapse
Affiliation(s)
| | - Nirbhay Kushwaha
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Saumik Basu
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | | | | |
Collapse
|
96
|
Perez I, Blanco L, Sanz B, Errarte P, Ariz U, Beitia M, Fernández A, Loizate A, Candenas ML, Pinto FM, Gil J, López JI, Larrinaga G. Altered Activity and Expression of Cytosolic Peptidases in Colorectal Cancer. Int J Med Sci 2015; 12:458-67. [PMID: 26078706 PMCID: PMC4466510 DOI: 10.7150/ijms.11808] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 04/29/2015] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND AND OBJECTIVE The role of peptidases in carcinogenic processes and their potential usefulness as tumor markers in colorectal cancer (CRC) have been classically attributed to cell-surface enzymes. The objective of the present study was to analyze the activity and mRNA expression of three cytosolic peptidases in the CRC and to correlate the obtained results with classic histopathological parameters for tumor prognosis and survival. METHODS The activity and mRNA levels of puromycin-sensitive aminopeptidase (PSA), aminopeptidase B (APB) and pyroglutamyl-peptidase I (PGI) were measured by fluorimetric and quantitative RT-PCR methods in colorectal mucosa and tumor tissues and plasma samples from CRC patients (n=81). RESULTS 1) PSA and APB activity was higher in adenomas and carcinomas than in the uninvolved mucosa. 2) mRNA levels of PSA and PGI was lower in tumors. 3) PGI activity in CRC tissue correlated negatively with histological grade, tumor size and 5-year overall survival of CRC patients. 4) Higher plasmatic APB activity was independently associated with better 5-year overall survival. CONCLUSIONS Data suggest that cytosolic peptidases may be involved in colorectal carcinogenesis and point to the determination of this enzymes as a valuable method in the determination of CRC prognosis.
Collapse
Affiliation(s)
- Itxaro Perez
- 1. Department of Nursing I, School of Nursing, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain ; 2. Department of Physiology, Faculty of Medicine and Dentistry, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain ; 6. BioCruces Health Research Institute, Barakaldo, Bizkaia, Spain
| | - Lorena Blanco
- 2. Department of Physiology, Faculty of Medicine and Dentistry, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain ; 6. BioCruces Health Research Institute, Barakaldo, Bizkaia, Spain
| | - Begoña Sanz
- 2. Department of Physiology, Faculty of Medicine and Dentistry, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain ; 6. BioCruces Health Research Institute, Barakaldo, Bizkaia, Spain
| | - Peio Errarte
- 2. Department of Physiology, Faculty of Medicine and Dentistry, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain ; 6. BioCruces Health Research Institute, Barakaldo, Bizkaia, Spain
| | - Usue Ariz
- 2. Department of Physiology, Faculty of Medicine and Dentistry, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain ; 6. BioCruces Health Research Institute, Barakaldo, Bizkaia, Spain
| | - Maider Beitia
- 2. Department of Physiology, Faculty of Medicine and Dentistry, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain ; 6. BioCruces Health Research Institute, Barakaldo, Bizkaia, Spain
| | - Ainhoa Fernández
- 2. Department of Physiology, Faculty of Medicine and Dentistry, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain ; 6. BioCruces Health Research Institute, Barakaldo, Bizkaia, Spain
| | - Alberto Loizate
- 3. Department of Surgery, Basurto University Hospital, University of the Basque Country (UPV/EHU), Bilbao, Bizkaia, Spain
| | - M Luz Candenas
- 4. Institute for Chemical Research, CSIC-Isla de la Cartuja, Sevilla, Spain
| | - Francisco M Pinto
- 4. Institute for Chemical Research, CSIC-Isla de la Cartuja, Sevilla, Spain
| | - Javier Gil
- 2. Department of Physiology, Faculty of Medicine and Dentistry, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain ; 6. BioCruces Health Research Institute, Barakaldo, Bizkaia, Spain
| | - José I López
- 5. Department of Anatomic Pathology, Cruces University Hospital, University of the Basque Country (UPV/EHU), Barakaldo, Bizkaia, Spain ; 6. BioCruces Health Research Institute, Barakaldo, Bizkaia, Spain
| | - Gorka Larrinaga
- 1. Department of Nursing I, School of Nursing, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain ; 2. Department of Physiology, Faculty of Medicine and Dentistry, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain ; 6. BioCruces Health Research Institute, Barakaldo, Bizkaia, Spain
| |
Collapse
|
97
|
Frazier KS. Antisense oligonucleotide therapies: the promise and the challenges from a toxicologic pathologist's perspective. Toxicol Pathol 2014; 43:78-89. [PMID: 25385330 DOI: 10.1177/0192623314551840] [Citation(s) in RCA: 231] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Many antisense oligonucleotides (ASOs) from several classes of molecules are currently in drug development. Despite over 20 years of pharmaceutical research, few ASOs have been marketed due to problems with clinical efficacy or preclinical toxicologic challenges. However, a number of recent developments have renewed interest in this class including the registration of mipomersen, the advent of successful screening strategies to eliminate more toxic molecules, and new understanding of the risks of off-target nucleotide binding and mitigation of potential off-target effects. Recent advances in backbone chemistries, conjugation to other moieties, and new delivery systems have allowed better tissue penetration, enhanced intracellular targeting, and less frequent dosing, resulting in fewer toxicities. While these new developments provide invigorated interest in these platforms, a few lingering challenges and preclinical/clinical toxicity issues remain to be completely resolved, including: (1) proinflammatory effects (vasculitis/inflammatory infiltrates); (2) nephrotoxicity and hepatotoxicity unrelated to lysosomal accumulation; and (3) thrombocytopenia. Recent investigative work by several laboratories have helped elucidate mechanisms for these issues, allowing a better understanding of the clinical relevance and implications of particular toxicities. It is important for toxicologists, pathologists, and regulatory reviewers to be familiar with new developments in the ASO field and their implications, as a greater number of new types of antisense molecules undergo preclinical toxicity testing.
Collapse
|
98
|
Off-target effects of the septin drug forchlorfenuron on nonplant eukaryotes. EUKARYOTIC CELL 2014; 13:1411-20. [PMID: 25217460 DOI: 10.1128/ec.00191-14] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The septins are a family of GTP-binding proteins that form cytoskeletal filaments. Septins are highly conserved and evolutionarily ancient but are absent from land plants. The synthetic plant cytokinin forchlorfenuron (FCF) was shown previously to inhibit budding yeast cell division and induce ectopic septin structures (M. Iwase, S. Okada, T. Oguchi, and A. Toh-e, Genes Genet. Syst. 79:199-206, 2004, http://dx.doi.org/10.1266/ggs.79.199). Subsequent studies in a wide range of eukaryotes have concluded that FCF exclusively inhibits septin function, yet the mechanism of FCF action in nonplant cells remains poorly understood. Here, we report that the cellular effects of FCF are far more complex than previously described. The reported growth arrest of budding yeast cells treated with 1 mM FCF partly reflects sensitization caused by a bud4 mutation present in the W303 strain background. In wild-type (BUD4(+)) budding yeast, growth was inhibited at FCF concentrations that had no detectable effect on septin structure or function. Moreover, FCF severely inhibited the proliferation of fission yeast cells, in which septin function is nonessential. FCF induced fragmentation of budding yeast mitochondrial reticula and the loss of mitochondrial membrane potential. Mitochondria also fragmented in cultured mammalian cells treated with concentrations of FCF that previously were assumed to target septins only. Finally, FCF potently inhibited ciliation and motility and induced mitochondrial disorganization in Tetrahymena thermophila without apparent alterations in septin structure. None of these effects was consistent with the inhibition of septin function. Our findings point to nonseptin targets as major concerns when using FCF.
Collapse
|
99
|
Combination of hepatocyte specific delivery and transformation dependent expression of shRNA inducing transcriptional gene silencing of c-Myc promoter in hepatocellular carcinoma cells. BMC Cancer 2014; 14:582. [PMID: 25108398 PMCID: PMC4153911 DOI: 10.1186/1471-2407-14-582] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 07/28/2014] [Indexed: 12/11/2022] Open
Abstract
Background A specific targeting modality for hepatocellular carcinoma (HCC) could ideally encompass a liver cell specific delivery system of a transcriptional unit that is active only in neoplastic cells. Sendai virosomes, derived from Sendai viral envelopes, home to hepatocytes based on the liver specific expression of asialoglycoprotein receptors (ASGPRs) which are recognized by the Sendai virosomal fusion (F) proteins. As reported earlier by us and other groups, transcriptional gene silencing (TGS) does not require continuous presence of the effector siRNA/shRNA molecule and is heritable, involving epigenetic modifications, leading to long term transcriptional repression. This could be advantageous over conventional gene therapy approaches, since continuous c-Myc inactivation is required to suppress hepatocarcinoma cells. Methods Exploiting such virosomal delivery, the alpha-fetoprotein (AFP) promoter, in combination with various tumour specific enhancers, was used to drive the expression of shRNA directed against ME1a1 binding site of the proto-oncogene c-Myc P2 promoter, in order to induce TGS in neoplastic liver cells. Results The dual specificity achieved by the Sendai virosomal delivery system and the promoter/enhancer guided expression ensured that the shRNA inducing TGS was active only in liver cells that had undergone malignant transformation. Our results indicate that such a bimodal therapeutic system induced specific activation of apoptosis in hepatocarcinoma cells due to heterochromatization and increased DNA methylation of the CpG islands around the target loci. Conclusions The Sendai virosomal delivery system, combined with AFP promoter/enhancer expression machinery, could serve as a generalized mechanism for the expression of genes deleterious to transformed hepatocarcinoma cells. In this system, the epigenetic suppression of c-Myc could have an added advantage for inducing cell death in the targeted cells. Electronic supplementary material The online version of this article (doi:10.1186/1471-2407-14-582) contains supplementary material, which is available to authorized users.
Collapse
|
100
|
|