Summary

MicroRNA-based Regulation of Picornavirus Tropism

Published: February 06, 2017
doi:

Summary

We describe here a method for regulating picornavirus tropism by incorporating sequences complementary to specific microRNAs into the viral genome. This protocol can be adapted to all different classes of viruses with modifications based upon the length and nature of their life cycle.

Abstract

Cell-specific restriction of viral replication without concomitant attenuation can benefit vaccine development, gene therapy, oncolytic virotherapy, and understanding the biological properties of viruses. There are several mechanisms for regulating viral tropism, however they tend to be virus class specific and many result in virus attenuation. Additionally, many viruses, including picornaviruses, exhibit size constraints that do not allow for incorporation of large amounts of foreign genetic material required for some targeting methods. MicroRNAs are short, non-coding RNAs that regulate gene expression in eukaryotic cells by binding complementary target sequences in messenger RNAs, preventing their translation or accelerating their degradation. Different cells exhibit distinct microRNA signatures and many microRNAs serve as biomarkers. These differential expression patterns can be exploited for restricting gene expression in cells that express specific microRNAs while maintaining expression in cells that do not. In regards to regulating viral tropism, sequences complementary to specific microRNAs are incorporated into the viral genome, generally in the 3′ non-coding regions, targeting them for destruction in the presence of the cognate microRNAs thus preventing viral gene expression and/or replication. MicroRNA-targeting is a technique that theoretically can be applied to all viral vectors without altering the potency of the virus in the absence of the corresponding microRNAs. Here we describe experimental methods associated with generating a microRNA-targeted picornavirus and evaluating the efficacy and specificity of that targeting in vitro. This protocol is designed for a rapidly replicating virus with a lytic replication cycle, however, modification of the time points analyzed and the specific virus titration readouts used will aid in the adaptation of this protocol to many different viruses.

Introduction

The development of a broadly applicable, easy and effective method for engineering a vector with restricted tropism provides a major opportunity to enhance the safety, biological understanding and therapeutic utility of viruses. Several mechanisms exist to target viral tropism including transductional, transcriptional, and translational-based techniques. However, these methods are not generally applicable to all vector systems, may require defective signaling pathways in targeted cells or require insertion of large coding sequences into the viral genome. Additionally, these methods can result in attenuation of the virus, significantly hindering their therapeutic activity and limiting insight into the unmodified system.

MicroRNAs are small (22-25 nucleotides), non-coding RNAs that mediate gene silencing in eukaryotic cells. MicroRNAs function by binding complementary target sequences (response elements) in messenger RNAs (mRNA) resulting in transcript destabilization, degradation or translational repression. MicroRNAs normally bind response elements with partial complementarity and yield small modifications in gene expression1,2,3,4,5. More significant alterations in gene expression can be achieved by increasing complementarity of the response element6. Thousands of mature microRNAs have been identified in a variety of species and many exhibit differential expression patterns in a variety of cell and tissue types7,8,9. These microRNA signatures can be exploited for cell-specific restriction of virus amplification by incorporating perfectly complementary response elements into the viral genome10,11,12,13. The overall goal of this microRNA-targeting technique is to control the tropism of a vector genome without additional attenuation.

The utility of this method for regulating viral tropism was originally demonstrated in lentiviral vectors to restrict transgene expression in specific tissues14,15,16. This technique has subsequently been applied to a vast array of replicating and non-replicating viral vectors for enhanced gene therapy as well as to improve the safety profiles of many oncolytic viruses by eliminating undesired toxicities in normal tissues10,11,12,13,17. It has also been utilized to generate safe and effective live-attenuated vaccines as well as to improve virus and vaccine manufacturing processes18,19,20,21. MicroRNA-targeting of a vector can allow for attenuation in vaccinated hosts or targeted systems while maintaining wild-type growth levels in producer systems. MicroRNA-targeting can also be used to improve the biosafety of viruses for research purposes by restricting transmission in one species (e.g. humans) while maintaining transmission in other hosts22. Finally, microRNA-targeting can allow for in-depth analyses of viral life cycles and specific roles of cell types in pathogenesis and immunity by segregating viral growth23,24,25,26.

This technique offers an alternative targeting method that is easily implemented and applicable for all virus systems. Additionally, the ever-expanding collection of mature microRNAs with differential expression patterns in specific cell types makes this technique highly versatile. MicroRNA-based targeting has proven efficacious for a variety of virus systems without compromising system function. The major limitations of this technique include trial and error optimization, the potential for escape mutations, and potential off-target effects on endogenous transcripts. However, these limitations can generally be overcome with optimized and rational response element design. Positive-sense RNA viruses tend to be particularly responsive to microRNA-targeting due to the positive-sense orientation of their genome and the availability of the transcripts to the microRNA machinery during the completely cytoplasmic replication cycle. Here we describe a protocol for generating a microRNA-targeted picornavirus and the experimental methods to verify the efficiency and specificity of that targeting in vitro.

Protocol

1. Cloning microRNA Response Elements into the Viral Genome Design microRNA response element inserts. Identify the desired microRNA and its corresponding target sequence. Several databases are available with mature microRNA sequences. Recommended: http://www.mirbase.org/9,27,28,29,30. Clo…

Representative Results

Table 1 represents results typical of a titration assay for a picornavirus and describes how to calculate the 50% tissue culture infectious dose. A schematic representation of the overall concept of microRNA-based regulation of viral tropism described in this manuscript is shown in Figure 1. The orientation of microRNA to response element during intracellular interactions, proper design of response element oligonucleotides for annealing and plasmid insert…

Discussion

The design, composition and localization of the microRNA response elements within the viral genome will dictate targeting efficacy and specificity. Optimizing these will require trial and error. However, rational design based on RNA structural analysis and previous studies of viral replication and microRNA signatures aids in the implementation of this technique with minimal optimization10,11,12,13</s…

Declarações

The authors have nothing to disclose.

Acknowledgements

Al and Mary Agnes McQuinn, the Richard M. Schulze Foundation, and an NIH Relief Grant from the Mayo Clinic funded representative work described here.

Materials

RE encoding Oligonucleotides IDT PAGE-Purified Ultramer Sequence Designed by Investigator
Oligonucleotides encoding unique restriction site IDT 25nM Sequence Designed by Investigator
Expand High Fidelity PCR Kit Sigma Aldrich 11732641001 Many other High Fidelity Polymerase PCR kits available
T4 DNA Ligase System NEB M0202S
MEGAscript Kit ThermoFisher Scientific AM1333
MEGAclear Kit ThermoFisher Scientific AM1908
0.5 M EDTA ThermoFisher Scientific AM9260G RNase-free
5 M NH4 Acetate ThermoFisher Scientific N/A Comes in MEGAclear Kit
Ethanol ThermoFisher Scientific BP2818100
Nuclease-free Water Fisher Scientific AM9938
TransIT-2020 Transfection Reagent Mirus MIR 5404
TransIT-mRNA Transfection Reagent Mirus MIR 2225
0.2 μm syringe filter Millipore SLGP033RS
2mL Screw-Cap Tubes Sarstedt 72.694.005
Cell Scrapers Fisher Scientific 08-100-241
MicroRNA Mimics Dharmacon Varied
MTT Cell Proliferation Assay ATCC 30-1010K
Subcloning Efficiency DH5α Competent Cells ThermoFisher Scientific 18265017
pBlueScript II Vectors Agilent Technologies Variable (e.g. 212205) There are different plasmids with T7 or T3 promoters and variable cloning sites to enable cloning and RNA transcription.

Referências

  1. Wightman, B., Ha, I., Ruvkun, G. Posttranscriptional Regulation of the Heterochronic Gene Lin-14 By Lin-4 Mediates Temporal Pattern Formation in C. Elegans. Cell. 75 (5), 855-862 (1993).
  2. Lee, R. C., Feinbaum, R. L., Ambros, V. The C. Elegans Heterochronic Gene Lin-4 Encodes Small RNAs With Antisense Complementarity to Lin-14. Cell. 75 (5), 843-854 (1993).
  3. Ambros, V. The Functions of Animal MicroRNAs. Nature. 431 (7006), 350-355 (2004).
  4. Bartel, D. P. MicroRNAs: Genomics, Biogenesis, Mechanism, and Function. Cell. 116 (2), 281-297 (2004).
  5. Bartel, D. P. MicroRNAs: Target Recognition and Regulatory Functions. Cell. 136 (2), 215-233 (2009).
  6. Benitez, A. A., Spanko, L. A., Bouhaddou, M., Sachs, D., Tenoever, B. R. Engineered Mammalian RNAi Can Elicit Antiviral Protection That Negates the Requirement for the Interferon Response. Cell Rep. 13 (7), 1456-1466 (2015).
  7. Lagos-Quintana, M., Rauhut, R., Yalcin, A., Meyer, J., Lendeckel, W., Tuschl, T. Identification of Tissue-Specific MicroRNAs From Mouse. Curr Biol. 12 (9), 735-739 (2002).
  8. Landgraf, P., et al. A Mammalian MicroRNA Expression Atlas Based on Small RNA Library Sequencing. Cell. 129 (7), 1401-1414 (2007).
  9. Griffiths-Jones, S., Saini, H. K., Van Dongen, S., Enright, A. J. miRBase: Tools for MicroRNA Genomics. Nucleic Acids Res. 36, D154-D158 (2008).
  10. Kelly, E. J., Russell, S. J. MicroRNAs and the Regulation of Vector Tropism. Mol Ther. 17 (3), 409-416 (2009).
  11. Brown, B. D., Naldini, L. Exploiting and Antagonizing MicroRNA Regulation for Therapeutic and Experimental Applications. Nat Rev Genet. 10 (8), 578-585 (2009).
  12. Tenoever, B. R. RNA Viruses and the Host MicroRNA Machinery. Nat Rev Microbiol. 11 (3), 169-180 (2013).
  13. Ruiz, A. J., Russell, S. J. MicroRNAs and Oncolytic Viruses. Curr Opin Virol. 13, 40-48 (2015).
  14. Brown, B. D., Venneri, M. A., Zingale, A., Sergi Sergi, L., Naldini, L., L, Endogenous MicroRNA Regulation Suppresses Transgene Expression in Hematopoietic Lineages and Enables Stable Gene Transfer. Nat Med. 12 (5), 585-591 (2006).
  15. Brown, B. D., et al. A MicroRNA-Regulated Lentiviral Vector Mediates Stable Correction of Hemophilia B Mice. Blood. 110 (13), 4144-4152 (2007).
  16. Brown, B. D., et al. Endogenous MicroRNA Can be Broadly Exploited to Regulate Transgene Expression According to Tissue, Lineage and Differentiation State. Nat Biotechnol. 25 (12), 1457-1467 (2007).
  17. Ruiz, A. J., Hadac, E. M., Nace, R. A., Russell, S. J. MicroRNA-Detargeted Mengovirus for Oncolytic Virotherapy. J Virol. 90 (8), 4078-4092 (2016).
  18. Vignuzzi, M., Wendt, E., Andino, R. Engineering Attenuated Virus Vaccines By Controlling Replication Fidelity. Nat Med. 14 (2), 154-161 (2008).
  19. Barnes, D., Kunitomi, M., Vignuzzi, M., Saksela, K., Andino, R. Harnessing Endogenous MiRNAs to Control Virus Tissue Tropism as a Strategy for Developing Attenuated Virus Vaccines. Cell Host Microbe. 4 (3), 239-248 (2008).
  20. Perez, J. T., Pham, A. M., Lorini, M. H., Chua, M. A., Steel, J., Tenoever, B. R. MicroRNA-Mediated Species-Specific Attenuation of Influenza a Virus. Nat Biotechnol. 27 (6), 572-576 (2009).
  21. Saydaminova, K., et al. Efficient Genome Editing in Hematopoietic Stem Cells With Helper-Dependent Ad5/35 Vectors Expressing Site-Specific Endonucleases Under MicroRNA Regulation. Mol Ther Methods Clin Dev. 1, 14057 (2015).
  22. Langlois, R. A., et al. MicroRNA-Based Strategy to Mitigate the Risk of Gain-of-function Influenza Studies. Nat Biotechnol. 31 (9), 844-847 (2013).
  23. Kelly, E. J., Hadac, E. M., Cullen, B. R., Russell, S. J. MicroRNA Antagonism of the Picornaviral Life Cycle: Alternative Mechanisms of Interference. PLoS Pathog. 6 (3), e1000820 (2010).
  24. Pham, A. M., Langlois, R. A., Tenoever, B. R. Replication in Cells of Hematopoietic Origin is Necessary for Dengue Virus Dissemination. PLoS Pathog. 8 (1), 1002465 (2012).
  25. Langlois, R. A., Varble, A., Chua, M. A., García-Sastre, A., Tenoever, B. R. Hematopoietic-Specific Targeting of Influenza a Virus Reveals Replication Requirements for Induction of Antiviral Immune Responses. Proc Natl Acad Sci U S A. 109 (30), 12117-12122 (2012).
  26. Chua, M. A., Schmid, S., Perez, J. T., Langlois, R. A., Tenoever, B. R. Influenza a Virus Utilizes Suboptimal Splicing to Coordinate the Timing of Infection. Cell Rep. 3 (1), 23-29 (2013).
  27. Griffiths-Jones, S. The MicroRNA Registry. Nucleic Acids Res. 32, D109-D111 (2004).
  28. Griffiths-Jones, S., Grocock, R. J., Van Dongen, S., Bateman, A., Enright, A. J. miRBase: MicroRNA Sequences, Targets and Gene Nomenclature. Nucleic Acids Res. 34, D140-D144 (2006).
  29. Kozomara, A., Griffiths-Jones, S. miRBase: Integrating MicroRNA Annotation and Deep-Sequencing Data. Nucleic Acids Res. 39, D152-D157 (2011).
  30. Kozomara, A., Griffiths-Jones, S. miRBase: Annotating High Confidence MicroRNAs Using Deep Sequencing Data. Nucleic Acids Res. 42, D68-D73 (2014).
  31. Heckman, K. L., Pease, L. R. Gene Splicing and Mutagenesis By PCR-Driven Overlap Extension. Nat Protoc. 2 (4), 924-932 (2007).
  32. . Basic Methods in Cellular and Molecular Biology. Gel Purification. Available from: https://www.jove.com/science-education/5063/gel-purification (2016)
  33. . Basic Methods in Cellular and Molecular Biology. DNA Ligation Reactions Available from: https://www.jove.com/science-education/5069/dna-ligation-reactions (2016)
  34. . Basic Methods in Cellular and Molecular Biology. Bacterial Transformation: The Heat Shock Method Available from: https://www.jove.com/science-education/5059/bacterial-transformation-the-heat-shock-method (2016)
  35. Zhang, S., Cahalan, M. D. Purifying Plasmid DNA From Bacterial Colonies Using the Qiagen Miniprep Kit. J Vis Exp. (6), e247 (2007).
  36. . Basic Methods in Cellular and Molecular Biology. Molecular Cloning. Available from: https://www.jove.com/science-education/5074/molecular-cloning (2016)
  37. Kueberuwa, G., Cawood, R., Tedcastle, A., Seymour, L. W. Tissue-Specific Attenuation of Oncolytic Sindbis Virus Without Compromised Genetic Stability. Hum Gene Ther Methods. 25 (2), 154-165 (2014).
  38. Grundhoff, A., Sullivan , C. S. Virus-Encoded MicroRNAs. Virology. 411 (2), 325-343 (2011).
  39. Kincaid, R. P., Sullivan, C. S. Virus-Encoded MicroRNAs: An Overview and a Look to the Future. PLoS Pathog. 8 (12), e1003018 (2012).
  40. Thomson, D. W., Bracken, C. P., Goodall, G. J. Experimental Strategies for MicroRNA Target Identification. Nucleic Acids Res. 39 (16), 6845-6853 (2011).
  41. Thomson, D. W., Dinger, M. E. Endogenous MicroRNA Sponges: Evidence and Controversy. Nat Rev Genet. 17 (5), 272-283 (2016).
  42. Mullokandov, G., et al. High-Throughput Assessment of MicroRNA Activity and Function Using MicroRNA Sensor and Decoy Libraries. Nat Methods. 9 (8), 840-846 (2012).
  43. Thomson, D. W., et al. Assessing the Gene Regulatory Properties of Argonaute-Bound Small RNAs of Diverse Genomic Origin. Nucleic Acids Res. 43 (1), 470-481 (2015).
  44. Wu, S., et al. Multiple MicroRNAs Modulate P21cip1/waf1 Expression By Directly Targeting Its 3′ Untranslated Region. Oncogene. 29 (15), 2302-2308 (2010).
  45. Vo, N. K., Dalton, R. P., Liu, N., Olson, E. N., Goodman, R. H. Affinity Purification of MicroRNA-133a With the Cardiac Transcription Factor, Hand2. Proc Natl Acad Sci U S A. 107 (45), 19231-19236 (2010).
  46. Arvey, A., Larsson, E., Sander, C., Leslie, C. S., Marks, D. S. Target mRNA Abundance Dilutes MicroRNA and siRNA Activity. Mol Syst Biol. 6, 363 (2010).
  47. Garcia, D. M., Baek, D., Shin, C., Bell, G. W., Grimson, A., Bartel, D. P. Weak Seed-Pairing Stability and High Target-Site Abundance Decrease the Proficiency of Lsy-6 and Other MicroRNAs. Nat Struct Mol Biol. 18 (10), 1139-1146 (2011).
  48. Jinek, M., Doudna, J. A. A Three-Dimensional View of the Molecular Machinery of RNA Interference. Nature. 457 (7228), 405-412 (2009).
  49. Pasquinelli, A. E. MicroRNAs and Their Targets: Recognition, Regulation and an Emerging Reciprocal Relationship. Nat Rev Genet. 13 (4), 271-282 (2012).
  50. Finnegan, E. F., Pasquinelli, A. E. MicroRNA Biogenesis: Regulating the Regulators. Crit Rev Biochem Mol Biol. 48 (1), 51-68 (2013).
  51. Ha, M., Kim, V. N. Regulation of MicroRNA Biogenesis. Nat Rev Mol Cell Biol. 15 (8), 509-524 (2014).
  52. Zuker, M. Mfold Web Server for Nucleic Acid Folding and Hybridization Prediction. Nucleic Acids Res. 31 (13), 3406-3415 (2003).
  53. Reuter, J. S., Mathews, D. H. RNAstructure: Software for RNA Secondary Structure Prediction and Analysis. BMC Bioinformatics. 11, 129 (2010).
  54. Khan, A. A., Betel, D., Miller, M. L., Sander, C., Leslie, C. S., Marks, D. S. Transfection of Small RNAs Globally Perturbs Gene Regulation By Endogenous MicroRNAs. Nat Biotechnol. 27 (6), 549-555 (2009).
  55. Skalsky, R. L., Cullen, B. R. Viruses, MicroRNAs, and Host Interactions. Annu Rev Microbiol. 64, 123-141 (2010).
  56. Sugio, K., et al. Enhanced Safety Profiles of the Telomerase-Specific Replication-Competent Adenovirus By Incorporation of Normal Cell-Specific MicroRNA-Targeted Sequences. Clin Cancer Res. 17 (9), 2807-2818 (2011).
  57. Fu, X., Rivera, A., Tao, L., De Geest, B., Zhang, X. Construction of an Oncolytic Herpes Simplex Virus That Precisely Targets Hepatocellular Carcinoma Cells. Mol Ther. 20 (2), 339-346 (2012).
  58. Yao, W., Guo, G., Zhang, Q., Fan, L., Wu, N., Bo, Y. The Application of Multiple MiRNA Response Elements Enables Oncolytic Adenoviruses to Possess Specificity to Glioma Cells. Virology. 458-459, 69-82 (2014).
  59. Bofill-De Ros, X., Gironella, M., Fillat, C. Mir-148a- and Mir-216a-regulated Oncolytic Adenoviruses Targeting Pancreatic Tumors Attenuate Tissue Damage Without Perturbation of MiRNA Activity. Mol Ther. 22 (9), 1665-1677 (2014).
  60. Baertsch, M. A., et al. MicroRNA-Mediated Multi-Tissue Detargeting of Oncolytic Measles Virus. Cancer Gene Ther. 21 (9), 373-380 (2014).
check_url/pt/55033?article_type=t

Play Video

Citar este artigo
Ruiz, A. J., Russell, S. J. MicroRNA-based Regulation of Picornavirus Tropism. J. Vis. Exp. (120), e55033, doi:10.3791/55033 (2017).

View Video