Reverse Genetics of RNA Viruses Methods and Protocols
Reverse Genetics of RNA Viruses Methods and Protocols
Daniel R. Pérez
Editor
Reverse Genetics
of RNA Viruses
Methods and Protocols
Second Edition
METHODS IN MOLECULAR BIOLOGY
Series Editor
John M. Walker
School of Life and Medical Sciences
University of Hertfordshire
Hatfield, Hertfordshire, UK
Second Edition
Edited by
Daniel R. Pérez
College of Veterinary Medicine, University of Georgia, Athens, GA, USA
Editor
Daniel R. Pérez
College of Veterinary Medicine
University of Georgia
Athens, GA, USA
© The Editor(s) (if applicable) and The Author(s), under exclusive license to Springer Science+Business Media, LLC, part
of Springer Nature 2024
This work is subject to copyright. All rights are solely and exclusively licensed by the Publisher, whether the whole or part
of the material is concerned, specifically the rights of translation, reprinting, reuse of illustrations, recitation,
broadcasting, reproduction on microfilms or in any other physical way, and transmission or information storage and
retrieval, electronic adaptation, computer software, or by similar or dissimilar methodology now known or hereafter
developed.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this publication does not imply,
even in the absence of a specific statement, that such names are exempt from the relevant protective laws and regulations
and therefore free for general use.
The publisher, the authors, and the editors are safe to assume that the advice and information in this book are believed to
be true and accurate at the date of publication. Neither the publisher nor the authors or the editors give a warranty,
expressed or implied, with respect to the material contained herein or for any errors or omissions that may have been
made. The publisher remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
This Humana imprint is published by the registered company Springer Science+Business Media, LLC, part of Springer
Nature.
The registered company address is: 1 New York Plaza, New York, NY 10004, U.S.A.
Reverse Genetics of RNA Viruses: Methods and Protocols, Second Edition follows the footsteps
of the first edition with the incorporation of new chapters and modifications of some of the
old chapters to highlight alternative methods and approaches for the manipulation of RNA
viruses. This second edition complements and does not replace the first edition which
continues to be relevant to this date. The term “reverse genetics” is an approach to unravel
the function of a gene by establishing and analyzing the phenotypic effects of synthetically
engineered gene sequences. For RNA viruses, reverse genetics implies the de novo reconsti-
tution of the virus from a cDNA copy. Using molecular biology, cDNA copies of RNA
viruses are cloned into a variety of vectors, most typically and in order of preference,
plasmids, bacterial artificial chromosomes or bacmids, or recombinant viral vectors. Consid-
ered the holy grail to study RNA viruses, reverse genetics has truly revolutionized our
understanding of pathogenesis, host range, and transmission of a myriad of RNA viruses.
In recent years and despite the sounding alarms of those that warn against this type of
research, reverse genetics has been instrumental in identifying and characterizing virulent
markers in RNA viruses and in producing innocuous viral progenies and reporter systems
that can be safely handled in the lab as surrogates for development of vaccines and antivirals.
Reverse genetics and synthetic biology played a crucial role in the timely production of
protective vaccines against influenza during the 2009 H1N1 pandemic. And prior research
using reverse genetics systems of coronaviruses led to the realization and manipulation of
key elements of the Spike glycoprotein of the severe acute respiratory syndrome coronavirus
2 (SARS-CoV-2) to produce in record time the highly effective mRNAs vaccines that helped
curtail the effects of the COVID-19 pandemic.
Here we have compiled 15 chapters summarizing reverse genetics breakthroughs and
detailed protocols. As with the first edition, the book does not cover every reverse genetics
protocol for every RNA virus. And like the first edition, this book would not have been
possible without the outstanding and most generous contributions of our authors who are
leaders in their respective fields and that have shared their insights and step-by-step proto-
cols to help you, our colleagues, with your own research endeavors. Reverse genetics will
continue to play a fundamental role in studying RNA viruses, identifying markers of host
range, disease, and transmission, and in helping us with the further the development of in
silico computational biology tools and artificial intelligence algorithms that can better
predict the emergence of pathogens that threaten food security and/or carry greater
zoonotic and/or pandemic potential.
I hope you find this book helpful.
v
Contents
Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix
vii
viii Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 265
Contributors
ix
x Contributors
GRIFFIN HAAS • Icahn School of Medicine at Mount Sinai, New York, NY, USA
THOMAS HOENEN • Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-
Institut, Greifswald, Insel Riems, Germany
SUSANNE KESSLER • Institute of Virology, Medical Center – University of Freiburg, Freiburg,
Germany; Faculty of Medicine, University of Freiburg, Freiburg, Germany
BENHUR LEE • Icahn School of Medicine at Mount Sinai, New York, NY, USA
XIAO-FENG LI • Department of Virology, State Key Laboratory of Pathogen and Biosecurity,
Beijing Institute of Microbiology and Epidemiology, AMMS, Beijing, China
ZHONG-YU LIU • School of Medicine, Sun Yat-sen University, Shenzhen, China
SHINJI MAKINO • Departments of Microbiology and Immunology, Galveston, TX, USA;
Institute of Human Infection and Immunity, Galveston, TX, USA; Center for Biodefense
and Emerging Infectious Diseases, Galveston, TX, USA; UTMB Center for Tropical
Diseases, Galveston, TX, USA; The Sealy Institute for Vaccine Sciences, Galveston, TX, USA
LUIS MARTÍNEZ-SOBRIDO • Texas Biomedical Research Institute, San Antonio, TX, USA
IGNACIO MENA • Department of Microbiology, Icahn School of Medicine at Mount Sinai,
New York, NY, USA; Global Health and Emerging Pathogens Institute, Icahn School of
Medicine at Mount Sinai, New York, NY, USA; Department of Immunology and
Microbiology, The Scripps Research Institute, La Jolla, CA, USA
AITOR NOGALES • Centro de Investigacion en Sanidad Animal (CISA-INIA/CSIC),
Madrid, Spain
JOHN T. PATTON • Department of Biology, Indiana University, Bloomington, IN, USA
DANIEL R. PÉREZ • Department of Population Health, College of Veterinary Medicine,
University of Georgia, Athens, GA, USA
ASHA A. PHILIP • Department of Biology, Indiana University, Bloomington, IN, USA;
CSL Seqirus, Waltham, MA, USA
CHENG-FENG QIN • Department of Virology, State Key Laboratory of Pathogen and
Biosecurity, Beijing Institute of Microbiology and Epidemiology, AMMS, Beijing, China
MARTIN SCHWEMMLE • Institute of Virology, Medical Center – University of Freiburg,
Freiburg, Germany; Faculty of Medicine, University of Freiburg, Freiburg, Germany
JOSÉ VALTER JOAQUIM SILVA JÚNIOR • Virology Sector, Department of Preventive Veterinary
Medicine, Federal University of Santa Maria, Santa Maria, RS, Brazil; Virology Sector,
Laboratory of Immunopathology Keizo Asami, Federal University of Pernambuco, Recife,
PE, Brazil
RYOSUKE SUZUKI • Department of Virology II, National Institute of Infectious Diseases,
Tokyo, Japan
TETSURO SUZUKI • Department of Microbiology and Immunology, Hamamatsu University
School of Medicine, Hamamatsu, Japan
BREANNA TERCERO • Departments of Microbiology and Immunology, Galveston, TX, USA
DANIELA TORO-ASCUY • Laboratory of Virology, Department of Biology, Faculty of Sciences,
Universidad de Chile, Santiago, Chile
YESSENY VÁSQUEZ-MARTÍNEZ • Escuela de Medicina, Faculty of Health Sciences, University of
Santiago de Chile, Santiago, Chile
CHENGJIN YE • Texas Biomedical Research Institute, San Antonio, TX, USA
THINESSHWARY YOGARAJAH • Department of Microbiology and Immunology, Yong Loo Lin
School of Medicine, National University Health System, National University of Singapore,
Singapore, Singapore
Chapter 1
Abstract
Filoviruses are causative agents of severe hemorrhagic fevers with high case fatality rates in humans. For
studies of virus biology and the subsequent development of countermeasures, reverse genetic systems, and
especially those facilitating the generation of recombinant filoviruses, are indispensable. Here, we describe
the generation of recombinant filoviruses from cDNA.
Key words Ebola virus, Marburg virus, Filoviruses, Reverse genetics, Full-length clone system,
Infectious clone, Recombinant virus
1 Introduction
Daniel R. Pérez (ed.), Reverse Genetics of RNA Viruses: Methods and Protocols, Methods in Molecular Biology, vol. 2733,
https://doi.org/10.1007/978-1-0716-3533-9_1,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2024
1
2 Bianca S. Bodmer and Thomas Hoenen
2 Materials
2.1 Rescue and 1. Cells for initial rescue and passaging, e.g., VeroE6, HEK293T,
Passaging of Viruses or Huh7 cells (see Note 1).
4 Bianca S. Bodmer and Thomas Hoenen
Fig. 1 Schematic depiction of processes taking place during a filovirus rescue. Cells are transfected with
plasmids encoding a cDNA copy of the full-length viral genome (rgZ) and each of the viral ribonucleoprotein
complex (RNP) components, i.e., L, VP35, VP30, and NP. A plasmid encoding the T7 RNA polymerase is also
transfected. Initial transcription of the viral genome-encoding plasmid by T7 RNA polymerase (A) results in a
“naked” full-length antigenome (cRNA), which is subsequently illegitimately encapsidated (B) by the viral RNP
proteins. At this point, it can then serve as a template for genome replication (C) to produce genomic RNA
(vRNA), which is encapsidated to form RNPs. These vRNA-containing RNPs can undergo not only replication
but also transcription of viral mRNAs encoding each of the viral proteins (D). This results in translation of the
viral proteins (E), which further encapsidate newly replicated genomes and antigenomes (F) to mediate further
viral RNA synthesis, but also mediate additional steps in the morphogenesis and budding of recombinant virus
particles (G)
Filovirus Reverse Genetics 5
3 Methods
3.2 Passage of 1. Six days post transfection, seed p1 cells (see Note 6) in 4 mL
Recombinant Viruses DMEM10%FCS/PS/Q in 6-well plates for ~90% confluence on
the next day. Incubate cells at 37 °C and 5% CO2 in a humidi-
fied incubator.
Filovirus Reverse Genetics 7
2. On the next day (i.e., 7 days post transfection), take the p1 cells
into the BSL4 laboratory. Add 1 mL of supernatant from the
p0 cells to the medium of the p1 cells (see Note 7). Return the
cells to the incubator.
3. From now on, check for cytopathic effect (CPE) in p1 cells
daily (see Note 8). Alternatively, if reporter-expressing viruses
(e.g., viruses expressing GFP or luciferase) are being rescued,
reporter activity can be used as a read-out.
4. Once clear CPE is visible, passage 1 mL of p1 supernatant onto
a T150 flask with 90% confluent cells (p2 cells) in 60 mL
DMEM10%FCS/PS/Q (see Note 7). Check for CPE in p2 cells
daily.
3.3 Harvest of 1. Once p2 cells show clearly discernibly CPE, scrape the cells into
Recombinant Viruses the medium using a cell scraper. Then transfer ~30 mL of the
resulting cell suspension each into two 50 mL centrifuge tubes,
and spin down for 10 min at 1000× g at 4 °C.
2. Pour the clarified supernatant into fresh centrifuge tubes each
containing 3 mL FBS (heat-inactivated for 30 min at 56 °C;
i.e., 10% total sample volume), mix by inversion, and aliquot.
Store the aliquots in liquid nitrogen.
3.4 Sequence 1. To ensure that the rescued virus has the correct/expected
Confirmation of sequence (see Note 9), inactivate 140 μL virus stock by adding
Rescued Virus it to 560 μL AVL buffer containing carrier RNA (from the
QIAamp viral RNA Mini Kit). Vortex and incubate for 10 min.
2. Remove the inactivated virus from the biosafety cabinet and
transfer it into a clean tube containing 560 μL 96–100% etha-
nol (see Note 10). Mix by vortexing, incubate for a further
10 min, and then remove the sample from the BSL4 laboratory
following appropriate approved protocols.
3. Extract viral RNA using the QIAamp viral RNA Mini Kit,
following the manufacturer’s instructions.
4. In a PCR tube, combine 1 μL RNA, 1 μL RT primer #3185
(2 μM), 1 μL dNTPs, and 11 μL RNase-free water (see Note
11). Incubate for 5 min at 65 °C, and then place immediately
on ice for at least 1 min.
5. In the meantime, combine 4 μL 5× SSIV buffer, 1 μL 0.1 M
DTT, 1 μL RNaseOUT, and 1 μL SuperScript IV, vortex
briefly, and place on ice for at least 1 min.
6. Add 7 μL of this mixture to the RNA/primer/dNTP mix (see
Note 11), incubate for 10 min at 50 °C, and then heat-
inactivate for 10 min at 80 °C and cool to 4 °C.
7. Add 1 μL RNase H (see Note 11), incubate for 20 min at 37 °
C, and then store cDNA at 4 °C.
8 Bianca S. Bodmer and Thomas Hoenen
Table 1
Primer combinations for PCRs and subsequent sequencing
(continued)
Filovirus Reverse Genetics 9
Table 1
(continued)
Table 2
PCR conditions
4 Notes
1. Various cell lines and cell line mixtures have been used for the
rescue of recombinant filoviruses, e.g., a HEK293T/Vero cell
mix, Vero cells, or Huh-7 cells. These cell lines vary in their
susceptibility to filovirus infection and their transfectability,
both of which impacts rescue efficiency. For example,
HEK293 and HEK293T cells are highly transfectable, but
only poorly susceptible to filovirus infection (although this
can be overcome by expressing the virus attachment factor
Tim-1 in these cells) [17, 37]. In contrast, Vero cells are highly
susceptible to filovirus infection, but somewhat harder to
transfect—although in our experience, this also varies between
different Vero clones. Further, it has been shown that the
choice of cell line can influence sequence fidelity in the recom-
binant viruses that are rescued, with rescue in Vero cells result-
ing in certain mutations (particularly A insertions in poly-A
stretches) in the genome with a higher frequency than when
using other cells [38]. These properties should be taken into
consideration when choosing a cell line for rescue. The authors
recommend Huh-7 cells for initial rescue, and either Huh-7 or
VeroE6 cells for further passaging.
2. All primer sequences provided here are for the species Zaire
ebolavirus and are based on the strain Mayinga sequence (Gen-
Bank accession number NC_002549) [39].
3. The amount of Transit-LT1 may need to be adjusted to obtain
optimal transfection efficacy in the cell line used. As a starting
point for most cell types, a ratio of 3 μL Transit-LT1 per 1 ug
DNA can be used. For Vero cells, this ratio should be increased
to 6:1.
4. At this point, the supernatant should be considered to contain
infectious ebolaviruses, and standard practices should be used
to avoid cross-contaminating wells (i.e., changing of pipettes/
pipette tips between different wells).
Filovirus Reverse Genetics 11
Acknowledgments
References
1. Kuhn JH, Amarasinghe GK, Basler CF, 4. History of Ebola Virus Disease (EVD) Out-
Bavari S, Bukreyev A, Chandran K, Crozier I, breaks. CDC. https://www.cdc.gov/vhf/
Dolnik O, Dye JM, Formenty PBH, ebola/history/chronology.html. Accessed
Griffiths A, Hewson R, Kobinger GP, Leroy 30 Mar 2022
EM, Muhlberger E, Netesov SV, Palacios G, 5. WHO (2020) Ebola virus disease Democratic
Palyi B, Paweska JT, Smither SJ, Takada A, Republic of Congo: external situation report
Towner JS, Wahl V, Ictv Report C (2019) 9 8 / 2 0 2 0 . h t t p s : // w w w. w h o . i n t /
ICTV virus taxonomy profile: filoviridae. J publications/i/item/10665-332654.
Gen Virol 100(6):911–912. https://doi.org/ Accessed 30 Mar 2022
10.1099/jgv.0.001252 6. Goldstein T, Anthony SJ, Gbakima A, Bird
2. Burk R, Bollinger L, Johnson JC, Wada J, BH, Bangura J, Tremeau-Bravard A, Belagana-
Radoshitzky SR, Palacios G, Bavari S, Jahrling halli MN, Wells HL, Dhanota JK, Liang E,
PB, Kuhn JH (2016) Neglected filoviruses. Grodus M, Jangra RK, DeJesus VA, Lasso G,
FEMS Microbiol Rev 40(4):494–519. Smith BR, Jambai A, Kamara BO, Kamara S,
https://doi.org/10.1093/femsre/fuw010 Bangura W, Monagin C, Shapira S, Johnson
3. Yamaoka S, Ebihara H (2021) Pathogenicity CK, Saylors K, Rubin EM, Chandran K, Lipkin
and virulence of Ebolaviruses with species- WI, Mazet JAK (2018) The discovery of Bom-
and variant-specificity. Virulence 12(1): bali virus adds further support for bats as hosts
8 8 5 – 9 0 1 . h t t p s : // d o i . o r g / 1 0 . 1 0 8 0 / of ebolaviruses. Nat Microbiol 3(10):
21505594.2021.1898169 1084–1089. https://doi.org/10.1038/
s41564-018-0227-2
Filovirus Reverse Genetics 13
7. Negredo A, Palacios G, Vazquez-Moron S, system for Ebola virus shows a genome length-
Gonzalez F, Dopazo H, Molero F, Juste J, dependent role of VP24 in virus infectivity. J
Quetglas J, Savji N, de la Cruz MM, Herrera Virol 88(18):10511–10524. https://doi.org/
JE, Pizarro M, Hutchison SK, Echevarria JE, 10.1128/JVI.01272-14
Lipkin WI, Tenorio A (2011) Discovery of an 18. Reid SP, Leung LW, Hartman AL, Martinez O,
ebolavirus-like filovirus in europe. PLoS Shaw ML, Carbonnelle C, Volchkov VE,
Pathog 7(10):e1002304. https://doi.org/10. Nichol ST, Basler CF (2006) Ebola virus
1371/journal.ppat.1002304 VP24 binds karyopherin alpha1 and blocks
8. Yang XL, Tan CW, Anderson DE, Jiang RD, STAT1 nuclear accumulation. J Virol 80(11):
Li B, Zhang W, Zhu Y, Lim XF, Zhou P, Liu 5156–5167. https://doi.org/10.1128/JVI.
XL, Guan W, Zhang L, Li SY, Zhang YZ, Wang 02349-05
LF, Shi ZL (2019) Characterization of a filovi- 19. Feagins AR, Basler CF (2015) Lloviu virus
rus (Mengla virus) from Rousettus bats in VP24 and VP35 proteins function as innate
China. Nat Microbiol 4(3):390–395. https:// immune antagonists in human and bat cells.
doi.org/10.1038/s41564-018-0328-y Virology 485:145–152. https://doi.org/10.
9. FDA (2020) FDA approves first treatment for 1016/j.virol.2015.07.010
Ebola virus. https://www.fda.gov/news- 20. He FB, Khan H, Huttunen M, Kolehmainen P,
events/press-announcements/fda-approves- Melen K, Maljanen S, Qu M, Jiang M,
first-treatment-ebola-virus. Accessed Kakkola L, Julkunen I (2021) Filovirus VP24
30 Mar 2022 proteins differentially regulate RIG-I and
10. FDA (2020) FDA approves treatment for MDA5-dependent type I and III interferon
Ebola virus. https://www.fda.gov/drugs/ promoter activation. Front Immunol 12:
news-events-human-drugs/fda-approves-treat 694105. https://doi.org/10.3389/fimmu.
ment-ebola-virus. Accessed 30 Mar 2022 2021.694105
11. FDA (2019) First FDA-approved vaccine for 21. Noda T, Sagara H, Suzuki E, Takada A,
the prevention of Ebola virus disease, marking Kida H, Kawaoka Y (2002) Ebola virus VP40
a critical milestone in public health prepared- drives the formation of virus-like filamentous
ness and response. https://www.fda.gov/ particles along with GP. J Virol 76(10):
news-events/press-announcements/first-fda- 4855–4865. https://doi.org/10.1128/jvi.76.
approved-vaccine-prevention-ebola-virus-dis 10.4855-4865.2002
ease-marking-critical-milestone-public-health. 22. Valmas C, Grosch MN, Schumann M,
Accessed 30 Mar 2022 Olejnik J, Martinez O, Best SM, Krahling V,
12. EMA (2019) Ervebo. https://www.ema. Basler CF, Muhlberger E (2010) Marburg
europa.eu/en/medicines/human/EPAR/ virus evades interferon responses by a mecha-
ervebo. Accessed 30 Mar 2022 nism distinct from ebola virus. PLoS Pathog
13. EMA (2020) Zabdeno. https://www.ema. 6(1):e1000721. https://doi.org/10.1371/
europa.eu/en/medicines/human/EPAR/ journal.ppat.1000721
zabdeno. Accessed 30 Mar 2022 23. Moller-Tank S, Maury W (2015) Ebola virus
14. EMA (2020) Mvabea. https://www.ema. entry: a curious and complex series of events.
europa.eu/en/medicines/human/EPAR/ PLoS Pathog 11(4):e1004731. https://doi.
mvabea. Accessed 30 Mar 2022 org/10.1371/journal.ppat.1004731
15. Muhlberger E, Weik M, Volchkov VE, Klenk 24. Wendt L, Bostedt L, Hoenen T, Groseth A
HD, Becker S (1999) Comparison of the tran- (2019) High-throughput screening for
scription and replication strategies of marburg negative-stranded hemorrhagic fever viruses
virus and Ebola virus by using artificial replica- using reverse genetics. Antivir Res 170:
tion systems. J Virol 73(3):2333–2342. 104569. https://doi.org/10.1016/j.antiviral.
h t t p s : // d o i . o r g / 1 0 . 1 1 2 8 / J V I . 7 3 . 3 . 2019.104569
2333-2342.1999 25. Volchkov VE, Volchkova VA, Muhlberger E,
16. Banadyga L, Hoenen T, Ambroggio X, Kolesnikova LV, Weik M, Dolnik O, Klenk
Dunham E, Groseth A, Ebihara H (2017) HD (2001) Recovery of infectious Ebola virus
Ebola virus VP24 interacts with NP to facilitate from complementary DNA: RNA editing of
nucleocapsid assembly and genome packaging. the GP gene and viral cytotoxicity. Science
Sci Rep 7(1):7698. https://doi.org/10.1038/ 291(5510):1965–1969. https://doi.org/10.
s41598-017-08167-8 1126/science.1057269
17. Watt A, Moukambi F, Banadyga L, Groseth A, 26. Neumann G, Feldmann H, Watanabe S,
Callison J, Herwig A, Ebihara H, Feldmann H, Lukashevich I, Kawaoka Y (2002) Reverse
Hoenen T (2014) A novel life cycle modeling genetics demonstrates that proteolytic proces-
sing of the Ebola virus glycoprotein is not
14 Bianca S. Bodmer and Thomas Hoenen
essential for replication in cell culture. J Virol 35. Mittler E, Schudt G, Halwe S, Rohde C,
76(1):406–410. https://doi.org/10.1128/ Becker S (2018) A fluorescently labeled Mar-
jvi.76.1.406-410.2002 burg Virus glycoprotein as a new tool to study
27. Hoenen T, Shabman RS, Groseth A, viral transport and assembly. J Infect Dis 218
Herwig A, Weber M, Schudt G, Dolnik O, (suppl_5):S318–S326. https://doi.org/10.
Basler CF, Becker S, Feldmann H (2012) 1093/infdis/jiy424
Inclusion bodies are a site of ebolavirus replica- 36. Ebihara H, Theriault S, Neumann G, Alimonti
tion. J Virol 86(21):11779–11788. https:// JB, Geisbert JB, Hensley LE, Groseth A, Jones
doi.org/10.1128/JVI.01525-12 SM, Geisbert TW, Kawaoka Y, Feldmann H
28. Brandt J, Wendt L, Bodmer BS, Mettenleiter (2007) In vitro and in vivo characterization of
TC, Hoenen T (2020) The cellular protein recombinant Ebola viruses expressing
CAD is recruited into Ebola virus inclusion enhanced green fluorescent protein. J Infect
bodies by the nucleoprotein NP to facilitate Dis 196(Suppl 2):S313–S322. https://doi.
genome replication and transcription. Cells org/10.1086/520590
9(5). https://doi.org/10.3390/cells9051126 37. Kondratowicz AS, Lennemann NJ, Sinn PL,
29. Wendt L, Brandt J, Bodmer BS, Reiche S, Davey RA, Hunt CL, Moller-Tank S, Meyer-
Schmidt ML, Traeger S, Hoenen T (2020) holz DK, Rennert P, Mullins RF, Brindley M,
The Ebola virus nucleoprotein recruits the Sandersfeld LM, Quinn K, Weller M, McCray
nuclear RNA export factor NXF1 into inclu- PB Jr, Chiorini J, Maury W (2011) T-cell
sion bodies to facilitate viral protein expression. immunoglobulin and mucin domain
Cells 9(1). https://doi.org/10.3390/ 1 (TIM-1) is a receptor for Zaire Ebolavirus
cells9010187 and Lake Victoria Marburgvirus. Proc Natl
30. Groseth A, Marzi A, Hoenen T, Herwig A, Acad Sci U S A 108(20):8426–8431. https://
Gardner D, Becker S, Ebihara H, Feldmann doi.org/10.1073/pnas.1019030108
H (2012) The Ebola virus glycoprotein contri- 38. Tsuda Y, Hoenen T, Banadyga L, Weisend C,
butes to but is not sufficient for virulence Ricklefs SM, Porcella SF, Ebihara H (2015) An
in vivo. PLoS Pathog 8(8):e1002847. improved reverse genetics system to overcome
https://doi.org/10.1371/journal.ppat. cell-type-dependent Ebola virus genome plas-
1002847 ticity. J Infect Dis. https://doi.org/10.1093/
31. Prins KC, Delpeut S, Leung DW, Reynard O, infdis/jiu681
Volchkova VA, Reid SP, Ramanan P, Cardenas 39. Hoenen T, Groseth A, Feldmann F, Marzi A,
WB, Amarasinghe GK, Volchkov VE, Basler CF Ebihara H, Kobinger G, Gunther S, Feldmann
(2010) Mutations abrogating VP35 interac- H (2014) Complete genome sequences of
tion with double-stranded RNA render Ebola three ebola virus isolates from the 2014 out-
virus avirulent in guinea pigs. J Virol 84(6): break in west Africa. Genome Announc 2(6):
3004–3015. https://doi.org/10.1128/JVI. e01331–e01314. https://doi.org/10.1128/
02459-09 genomeA.01331-14
32. Ebihara H, Takada A, Kobasa D, Jones S, 40. Volchkova VA, Dolnik O, Martinez MJ,
Neumann G, Theriault S, Bray M, Reynard O, Volchkov VE (2011) Genomic
Feldmann H, Kawaoka Y (2006) Molecular RNA editing and its impact on Ebola virus
determinants of Ebola virus virulence in mice. adaptation during serial passages in cell culture
PLoS Pathog 2(7):e73. https://doi.org/10. and infection of guinea pigs. J Infect Dis 204
1371/journal.ppat.0020073 (Suppl 3):S941–S946. https://doi.org/10.
33. Neumann G, Geisbert TW, Ebihara H, Geis- 1093/infdis/jir321
bert JB, Daddario-DiCaprio KM, 41. Haddock E, Feldmann F, Feldmann H (2016)
Feldmann H, Kawaoka Y (2007) Proteolytic Effective chemical inactivation of Ebola virus.
processing of the Ebola virus glycoprotein is Emerg Infect Dis 22(7):1292–1294. https://
not critical for Ebola virus replication in non- doi.org/10.3201/eid2207.160233
human primates. J Virol 81(6):2995–2998. 42. Smither SJ, Weller SA, Phelps A, Eastaugh L,
https://doi.org/10.1128/JVI.02486-06 Ngugi S, O’Brien LM, Steward J, Lonsdale SG,
34. Schudt G, Kolesnikova L, Dolnik O, Sodeik B, Lever MS (2015) Buffer AVL alone does not
Becker S (2013) Live-cell imaging of Marburg inactivate Ebola virus in a representative clinical
virus-infected cells uncovers actin-dependent sample type. J Clin Microbiol 53(10):
transport of nucleocapsids over long distances. 3148–3154. https://doi.org/10.1128/JCM.
Proc Natl Acad Sci U S A 110(35): 01449-15
14402–14407. https://doi.org/10.1073/
pnas.1307681110
Chapter 2
Abstract
Paramyxoviruses place significant burdens on both human and wildlife health; while some paramyxoviruses
are established within human populations, others circulate within diverse animal reservoirs. Concerningly,
bat-borne paramyxoviruses have spilled over into humans with increasing frequency in recent years,
resulting in severe disease. The risk of future zoonotic outbreaks, as well as the persistence of paramyx-
oviruses that currently circulate within humans, highlights the need for efficient tools through which to
interrogate paramyxovirus biology. Reverse genetics systems provide scientists with the ability to rescue
paramyxoviruses de novo, offering versatile tools for implementation in both research and public health
settings. Reverse genetics systems have greatly improved over the past 30 years, with several key innovations
optimizing the success of paramyxovirus rescue. Here, we describe the significance of such advances and
provide a generally applicable guide for the development and use of reverse genetics systems for the rescue
of diverse members of Paramyxoviridae.
1 Introduction
Daniel R. Pérez (ed.), Reverse Genetics of RNA Viruses: Methods and Protocols, Methods in Molecular Biology, vol. 2733,
https://doi.org/10.1007/978-1-0716-3533-9_2,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2024
15
16 Griffin Haas and Benhur Lee
for traversing the species barrier and have caused severe disease
when they have spilled over into human hosts; there is increasing
concern that such zoonotic paramyxoviruses could establish them-
selves in the human population, resulting in outbreaks [6–11]. Sys-
tems to advance our understanding of paramyxoviruses are
increasingly needed, and the use of such tools will undoubtedly
bolster our ability to prepare for and respond to future (zoonotic)
pandemics.
The ability to rescue paramyxovirus species de novo from
cDNA provides numerous applications for both research and public
health. Reverse genetics systems allow for the generation of pure
viral stocks without a need for excessive passaging, preventing
adaptation of virus to cell culture; this is desirable to produce
vaccine or research strains in which passaging may introduce unex-
pected mutations. Plasmid-encoded viral cDNA can be easily
manipulated, allowing for the integration of reporter genes, the
generation of desired mutations, and the ability to knock out entire
genes from viral genomes; such versatility greatly expands the
toolkit of virologists and likewise the scope of questions that may
be investigated. Excitingly, reverse genetics systems even allow for
the de novo rescue and characterization of sequenced viral species
that have been identified by metagenomics, but have yet to be
isolated, such as a mumps-like virus from Epomophorus bats (Bat-
MuV) and a morbillivirus from Myotis bats (Fig. 2) [12, 13].
1.1 History and Minireplicon systems laid the foundation for Paramyxoviridae
Optimization of reverse genetics. Between 1991 and 1993, minireplicon systems
Paramyxovirus were constructed for Sendai virus (SeV), respiratory syncytial virus
Reverse Genetics (RSV), and HPIV-3 [14–16]. These constructs were used as tem-
Systems plate for the in vitro transcription of synthetic, viral-like RNAs for
each species; transfection of the respective viral-like RNA species
into cells, complemented by superinfection with respective full-
length virus, resulted in “rescue” of the viral-like RNAs, character-
ized by replication of the viral-like RNA and concomitant reporter
gene expression by the viral RNA-dependent RNA polymerase
(vRdRp). Further, such viral-like RNAs were observed to be pack-
aged into propagable, viral-like particles (VLPs). These works
Reverse Genetics Systems for Diverse Paramyxoviruses 17
Fig. 2 Phylogeny of fully sequenced members of Paramyxoviridae. The evolutionary history was inferred using
the neighbor-joining method for amino acid sequences of the large (L) vRdRp protein from each species. The
tree is drawn to scale, with branch lengths (next to the branches) in the same units as those of the evolutionary
distances used to infer the phylogenetic tree. The evolutionary distances were computed using the Poisson
correction method and are in the units of the number of amino acid substitutions per site. Evolutionary
analyses were conducted in MEGA X. An asterisk (*) next to a species name denotes that reverse genetics
systems have been successfully established for the respective species
Fig. 3 A Hh-Rbz allows for efficient transcription without compromising the rule
of six. (a) Minimal and optimal T7 promoter sequences. Templated G’s are
underlined. (b) A Hh-Rbz allows for implementation of the optimal T7 promoter
sequence, as it can be designed to cleave immediately upstream of the viral 3′
terminus (shown in blue)
(NCR) between viral genes. This approach has been successful for
the insertion of reporter genes upstream of N, between N and P/V,
between P/V and M, and between RBP and L [26, 34, 46, 72,
73]. Alternatively, a P2A sequence may be used to encode reporter
genes within the same transcript as some viral genes, such as matrix
(M) or the receptor binding protein (RBP) [26, 24, 71]. Regardless
of approach, such gene insertions must not violate the rule of six
nor disrupt the phasing of the terminal bipartite promoter ele-
ments. If manipulation of a paramyxovirus genome violates the
rule of six, we recommend correcting this by adding additional
nucleotides immediately following the stop codon of a gene, as
this will have minimal, if any, effect on virus replication or kinetics.
Hh-Rbz Design
The Hh-Rbz encoded upstream of the 3′ leader sequence (3′Ldr)
must be designed specifically for each respective virus. This is
because a region of the Hh-Rbz is required to base-pair with the
extreme terminal nucleotide sequence of the 3′ Ldr (Fig. 3b). We
have compared numerous Hh-Rbz designs and have identified at
least two designs that are efficiently cleaved [24]. Base-pairing of
the Hh-Rbz with between five and eight nucleotides of the viral 3′
Ldr sequence is often sufficient for proper folding and cleavage;
however, due to sequence variation, designs that work for one
species may be suboptimal for another. Thus, we recommend
22 Griffin Haas and Benhur Lee
Table 1
Replication complex ratios successfully applied for the rescue of diverse paramyxovirus species
Accessory
plasmid
Family Genus Species N:P:L promoter Source(s)
Orthoparamyxovirinae Morbillivirus Measles virus 5.0:1.0: CAG [36]
(MeV) 5.0
3.0:3.0: T7 [26]
1.0
Myotis bat 3.0:3.0: T7 [13]
morbillivirus 1.0
(MBaMV)
Canine distemper 3.0:3.0: T7 [37, 38]
virus (CDV) 1.0
2.0:2.0: T7 [38]
1.0
Rinderpest virus 10.0: T7 [39]
(RPV) 10.0:
1.0
Peste-des-petits- 2.0:1.0: CAG [40]
ruminants 1.0
virus (PPRV) 1.5:1.0: CAG [41, 42]
1.0
[
20.0: T7 43]
20.0:
1.0
Jeilongvirus J-virus (JV) 2.0:1.0: T7 [44]
75.0
[
Narmovirus Tupaia virus 1.0:1.0: CAG 45]
(TupV) 1.0
Henipavirus Nipah virus 3.1:2.0: T7 [26, 46]
(NiV) 1.0
5.0:1.0: T7 [27]
2.0
3.1:2.0: CAG [47]
1.0
2.0:2.0: CAG [21]
1.0
Hendra virus 3.1:2.0: T7 [48]
(HeV) 1.0
5.0:1.0: T7 [27]
2.0
Cedar virus 3.1:2.0: CMV [49]
(CedV) 1.0
3.1:2.0: T7 Lee lab
1.0 unpublished
data
Respirovirus Human 8.0:8.0: T7 [50]
parainfluenza 1.0
virus type
1 (HPIV-1)
(continued)
Reverse Genetics Systems for Diverse Paramyxoviruses 23
Table 1
(continued)
Accessory
plasmid
Family Genus Species N:P:L promoter Source(s)
Human 20.6: T7 [26]
parainfluenza 11.0:
virus type 1.0
3 (HPIV-3) 8.0:8.0: T7 [51]
1.0
Bovine 2.0:2.0: T7 [52]
parainfluenza 1.0
virus type
3 (BPIV3)
Sendai virus 20.6: T7 [26, 53]
(SeV) 11.0:
1.0
10.0: T7 [54]
10.0:
1.0
Avulavirinae Orthoavulavirus Newcastle disease 2.5:3.0: T7 [55]
virus (NDV) 1.0
(APMV-1) 2.0:1.0: T7 [26, 56–58]
1.0
2.0:1.0: CMV [20]
1.0
Paraavulavirus Avian 3.0:2.0: T7 [59]
paramyxovirus 1.0
type 3 (APMV-
3)
Metaavulavirus Avian 3.0:2.0: T7 [60]
paramyxovirus 1.0
type 2 (APMV-
2)
Avian 2.0:1.0: CAG [61]
paramyxovirus 1.0
type 6 (APMV-
6)
Avian 2.0:1.0: T7 [62]
paramyxovirus 1.0
type 7 (APMV-
7)
Avian 2.0:1.0: CAG [61]
paramyxovirus 1.0
type
10 (APMV-
10)
(continued)
24 Griffin Haas and Benhur Lee
Table 1
(continued)
Accessory
plasmid
Family Genus Species N:P:L promoter Source(s)
Rubulavirinae Orthorubulavirus Mumps virus 6.0:1.0: T7 [63–65]
(MuV) 4.0
6.0:1.0: T7 [66]
10.0
3.0:1.0: T7 [26]
2.0
Human 8.0:1.0: T7 [67]
parainfluenza 4.0
virus type 8.0:8.0: T7 [68]
2 (HPIV-2) 1.0
Parainfluenza 10.0: T7 [69]
virus type 1.0:
5 (PIV-5) 10.0
6.0:1.0: T7 [70]
7.5
Bat mumps virus 6.0:1.0: T7 [12]
(BatMuV) 4.6
Pararubulavirus Menangle virus 3.0:1.0: T7 Lee lab
(MenV) 2.0 unpublished
data
Sosuga virus 2.0:1.0: CAG [71]
(SosV) 4.0
Ratios of plasmids encoding N, P, and L were calculated from amounts of respective accessory plasmids employed for
rescue in the respective corresponding publication(s). Promoter type designates the promoter element driving transcrip-
tion of the accessory plasmids. T7 transcription by T7 polymerase, CMV transcription by RNA polymerase II, CAG
transcription by RNA polymerase II
2 Materials
2.2 Cell Preparation 1. Sterile tissue culture materials (plasticware, serological pipettes,
etc.).
2. BSRT7/5 cells (4 × 105 cells per well of a six-well plate), or
HEK-293T cells (1 × 106 cells per well of a six-well plate).
3. Media for mammalian cell maintenance: 1× DMEM (Gibco)
supplemented with 10% fetal bovine serum (Gibco).
3 Methods
3.1 Transformation 1. Thaw chemically competent bacterial cells on ice for 10 min.
and Validation of Full- Transfer thawed competent cells to pre-labeled tubes (50 μL
Length Paramyxovirus aliquots, one tube per plasmid). Use Stellar competent cells for
cDNA and Accessory the smaller, accessory rescue plasmids (e.g., pTM1-N, pTM1-
Rescue Plasmids P, pTM1-L, and pCAGGs-T7CoOpt). Use Max-Efficiency
Stbl2 competent cells for propagation/amplification of the
larger, antigenomic viral cDNA plasmids.
2. On ice, add 5–10 ng of plasmid per respective tube of compe-
tent cells and gently flick to mix. Incubate tube on ice for
30 min.
3. Heat-shock competent cells at 42 °C for either 45 s (Stellar
competent cells) or 30 s (Max-Efficiency Stbl2 competent
cells). Immediately return heat-shocked tubes to ice. Incubate
on ice for 2 min.
4. Add S.O.C. to each tube (with no antibiotic) to a final volume
of 500 μL.
5. Incubate tubes at 30 °C with gentle shaking for 1 h.
6. Using a sterile cell spreader (or pipette tip), inoculate appropri-
ate antibiotic-containing LB agar plate with 50–100 μL of
transformants. Use sterile cell spreader or pipette tips to streak
out transformants to isolate single colonies. Incubate for
24–48 h between 30 and 33 °C (see Note 1).
7. Pick several colonies (using sterile pipette tips) to inoculate
5 mL of LB supplemented with appropriate antibiotic. Mark
the agar plate to indicate which colonies were picked. Incubate
outgrowth cultures with shaking at between 30 and 33 °C
overnight.
8. Save an aliquot of the outgrowth (250–500 μL) to make a
glycerol stock by adding equal volume of 50% glycerol. Glyc-
erol stocks must be frozen at -80 °C. Alternatively, wrap the
transformation plate in parafilm and store at 4 °C; validated,
marked colonies may be regrown and picked again.
9. Pellet and miniprep the bacterial outgrowths.
10. Use appropriate restriction enzyme(s) to perform a diagnostic
digest of the derived clones for each plasmid.
11. Resolve restriction digests by gel electrophoresis to validate
plasmid integrity. If the diagnostic digest is as expected,
sequence-verify each construct (see Note 2).
28 Griffin Haas and Benhur Lee
12. For validated clones, use a scraping from the glycerol stock
(or re-pick respective colonies from the LB agar plate) to
inoculate 50–100 mL of TB supplemented with the appropri-
ate antibiotic. Incubate in an appropriately sized Erlenmeyer
flask overnight with shaking, at between 30 and 33 °C.
13. When the outgrowth is turbid and sufficient for harvest, pellet
the bacteria via centrifugation at 4000× g for 10 min, and
process the pellet using a midiprep kit according to the manu-
facturer’s instructions.
3.2 Cell Preparation 1. BSRT7/5 cells and/or HEK-293T cells should be 80–90%
confluent on the day of transfection. Cells are maintained in
DMEM supplemented with 10% FBS.
2. Immediately prior to transfection, replace the media on all cells
with fresh, pre-warmed DMEM + 10% FBS.
3.4 Virus Recovery 1. Monitor the rescue plate daily for cytopathic effect. If using a
and Amplification reporter virus (e.g., GFP), check for reporter gene expression
and monitor for secondary infection events.
2. If the cells become over-confluent or if the media becomes
acidic, replace the media as needed to prolong cell viability.
3. Upon observation of cytopathic effect or secondary infection
events, prepare for generation of passage 1 (P1) stocks:
Reverse Genetics Systems for Diverse Paramyxoviruses 29
Fig. 6 Overview of plasmid-mediated paramyxovirus rescue. (1) Plasmids encoding the viral antigenome/
cDNA, codon-optimized T7 polymerase, and the accessory viral N, P, and L proteins are transfected into cells.
(2) T7 polymerase is transcribed and translated by the cell. (3) T7 polymerase transcribes the accessory
viral N, P, and L proteins, as well as the viral antigenome. The Rbz elements encoded on the 5′ and 3′ ends of
the antigenomic vRNA are cleaved, resulting in viral antigenome without exogenous sequence on the termini.
(4) N encapsidates the viral antigenome, facilitating the helical arrangement of the vRNP and proper
positioning of the antigenomic bipartite promoter (highlighted in green). (5) The vRdRp associates with the
vRNP via P and recognizes the bipartite promoter element. (6) The vRdRp replicates the antigenome, making
viral genome. The vRNP properly forms the genomic bipartite promoter (highlighted in blue). (7) The genomic
bipartite promoter is recognized by the vRdRp, and viral genome provides a template for vRdRp-mediated
transcription of viral gene products. (8) The accumulation of viral gene products facilitates replication of the
viral genome. (9) The accumulation of viral proteins facilitates the assembly of virions and packaging of the
viral genome. Matrix protein facilitates the budding of virions
4 Notes
References
1. Plemper RK, Lamb RA (2020) Paramyxoviri- 8. Albariño CG, Foltzer M, Towner JS, Rowe LA,
dae: the viruses and their replication. Fields Campbell S, Jaramillo CM, Bird BH, Reeder
Virol 1(7):504–558 DM, Vodzak ME, Rota P, Metcalfe MG, Spir-
2. Calain P, Roux L (1993) The rule of six, a basic opoulou CF, Knust B, Vincent JP, Frace MA,
feature for efficient replication of Sendai virus Nichol ST, Rollin PE, Ströher U (2014) Novel
defective interfering RNA. J Virol 67(8): paramyxovirus associated with severe acute
4822–4830. https://doi.org/10.1128/JVI. febrile disease, South Sudan and Uganda,
67.8.4822-4830.1993 2012. Emerg Infect Dis 20(2):211–216.
3. Virtue ER, Marsh GA, Wang L-F (2009) Para- https://doi.org/10.3201/eid2002.131620
myxoviruses infecting humans: the old, the 9. Philbey AW, Kirkland PD, Ross AD, Davis RJ,
new and the unknown. Future Microbiol Gleeson AB, Love RJ, Daniels PW, Gould AR,
4(5):537–554. https://doi.org/10.2217/ Hyatt AD (1998) An apparently new virus
fmb.09.26 (family Paramyxoviridae) infectious for pigs,
4. Drexler JF, Corman VM, Müller MA, humans, and fruit bats. Emerg Infect Dis
Maganga GD, Vallo P, Binger T, Gloza- 4(2):269–271. https://doi.org/10.3201/
Rausch F, Cottontail VM, Rasche A, eid0402.980214
Yordanov S, Seebens A, Knörnschild M, 10. Eaton BT, Broder CC, Middleton D, Wang
Oppong S, Adu Sarkodie Y, Pongombo C, L-F (2006) Hendra and Nipah viruses: differ-
Lukashev AN, Schmidt-Chanasit J, Stöcker A, ent and dangerous. Nat Rev Microbiol 4(1):
Carneiro AJB, Erbar S, Maisner A, Fronhoffs F, 2 3 – 3 5 . h t t p s : // d o i . o r g / 1 0 . 1 0 3 8 /
Buettner R, Kalko EKV, Kruppa T, Franke CR, nrmicro1323
Kallies R, Yandoko ERN, Herrler G, 11. Chua KB, Bellini WJ, Rota PA, Harcourt BH,
Reusken C, Hassanin A, Krüger DH, Tamin A, Lam SK, Ksiazek TG, Rollin PE, Zaki
Matthee S, Ulrich RG, Leroy EM, Drosten C SR, Shieh W, Goldsmith CS, Gubler DJ, Roeh-
(2012) Bats host major mammalian paramyx- rig JT, Eaton B, Gould AR, Olson J, Field H,
oviruses. Nat Commun 3:796. https://doi. Daniels P, Ling AE, Peters CJ, Anderson LJ,
org/10.1038/ncomms1796 Mahy BW (2000) Nipah virus: a recently emer-
5. Samal SK (2008) Paramyxoviruses of gent deadly paramyxovirus. Science
animals. In: Encyclopedia of virology, p 40 288(5470):1432–1435. https://doi.org/10.
6. Thibault PA, Watkinson RE, Moreira-Soto A, 1126/science.288.5470.1432
Drexler JF, Lee B (2017) Zoonotic potential of 12. Krüger N, Sauder C, Hüttl S, Papies J, Voigt K,
emerging paramyxoviruses: knowns and Herrler G, Hardes K, Steinmetzer T, Örvell C,
unknowns. Adv Virus Res 98:1–55. https:// Drexler JF, Drosten C, Rubin S, Müller MA,
doi.org/10.1016/bs.aivir.2016.12.001 Hoffmann M (2018) Entry, replication,
7. Amman BR, Albariño CG, Bird BH, immune evasion, and neurotoxicity of synthet-
Nyakarahuka L, Sealy TK, Balinandi S, Schuh ically engineered bat-borne mumps virus. Cell
AJ, Campbell SM, Ströher U, Jones MEB, Rep 25(2):312–320.e317. https://doi.org/
Vodzack ME, Reeder DM, Kaboyo W, Nichol 10.1016/j.celrep.2018.09.018
ST, Towner JS (2015) A recently discovered 13. Ikegame S, Carmichael JC, Wells H, Furler
pathogenic paramyxovirus, Sosuga virus, is O’Brien RL, Acklin JA, Chiu H-P, Oguntuyo
present in Rousettus aegyptiacus fruit bats at KY, Cox RM, Patel AR, Kowdle S, Stevens CS,
multiple locations in Uganda. J Wildl Dis Eckley M, Zhan S, Lim JK, Veit EC, Evans MJ,
51(3):774–779. https://doi.org/10.7589/ Hashiguchi T, Durigon E, Schountz T, Epstein
2015-02-044 JH, Plemper RK, Daszak P, Anthony SJ, Lee B
(2023) Metagenomics-enabled reverse-
32 Griffin Haas and Benhur Lee
genetics assembly and characterization of myo- not essential for virus replication in tissue cul-
tis bat morbillivirus. Nat Microbiol 8(6):1108– ture, and the human RSV leader region acts as a
1122. https://doi.org/10.1038/s41564- functional BRSV genome promoter. J Virol
023-01380-4 73(1):251–259. https://doi.org/10.1128/
14. Park KH, Huang T, Correia FF, Krystal M jvi.73.1.251-259.1999
(1991) Rescue of a foreign gene by Sendai 23. Beaty SM, Park A, Won ST, Hong P, Lyons M,
virus. Proc Natl Acad Sci U S A 88(13): Vigant F, Freiberg AN, tenOever BR, Duprex
5537–5541. https://doi.org/10.1073/pnas. WP, Lee B (2017) Efficient and robust para-
88.13.5537 myxoviridae reverse genetics systems. mSphere
15. Dimock K, Collins PL (1993) Rescue of syn- 2(2). https://doi.org/10.1128/mSphere.
thetic analogs of genomic RNA and replicative- 00376-16
intermediate RNA of human parainfluenza 24. Yun T, Park A, Hill TE, Pernet O, Beaty SM,
virus type 3. J Virol 67(5):2772–2778. Juelich TL, Smith JK, Zhang L, Wang YE,
h t t p s : // d o i . o r g / 1 0 . 1 1 2 8 / J V I . 6 7 . 5 . Vigant F, Gao J, Wu P, Lee B, Freiberg AN
2772-2778.1993 (2015) Efficient reverse genetics reveals genetic
16. Collins PL, Mink MA, Stec DS (1991) Rescue determinants of budding and fusogenic differ-
of synthetic analogs of respiratory syncytial ences between Nipah and Hendra viruses and
virus genomic RNA and effect of truncations enables real-time monitoring of viral spread in
and mutations on the expression of a foreign small animal models of henipavirus infection. J
reporter gene. Proc Natl Acad Sci U S A Virol 89(2):1242–1253. https://doi.org/10.
88(21):9663–9667. https://doi.org/10. 1128/JVI.02583-14
1073/pnas.88.21.9663 25. Li B-Y, Li X-R, Lan X, Yin X-P, Li Z-Y, Yang B,
17. Radecke F, Spielhofer P, Schneider H, Liu J-X (2011) Rescue of Newcastle disease
Kaelin K, Huber M, Dötsch C, virus from cloned cDNA using an RNA poly-
Christiansen G, Billeter MA (1995) Rescue of merase II promoter. Arch Virol 156(6):
measles viruses from cloned DNA. EMBO J 979–986. https://doi.org/10.1007/s00705-
14(23):5773–5784 011-0932-0
18. Garcin D, Pelet T, Calain P, Roux L, Curran J, 26. Griffin BD, Leung A, Chan M, Warner BM,
Kolakofsky D (1995) A highly recombinogenic Ranadheera C, Tierney K, Audet J, Frost KL,
system for the recovery of infectious Sendai Safronetz D, Embury-Hyatt C, Booth SA,
paramyxovirus from cDNA: generation of a Kobasa D (2019) Establishment of an RNA
novel copy-back nondefective interfering polymerase II-driven reverse genetics system
virus. EMBO J 14(24):6087–6094. https:// for Nipah virus strains from Malaysia and
doi.org/10.1002/j.1460-2075.1995. Bangladesh. Sci Rep 9(1):11171. https://doi.
tb00299.x org/10.1038/s41598-019-47549-y
19. Collins PL, Hill MG, Camargo E, Grosfeld H, 27. Chey S, Palmer JM, Doerr L, Liebert UG
Chanock RM, Murphy BR (1995) Production (2021) Dual promoters improve the rescue of
of infectious human respiratory syncytial virus recombinant measles virus in human cells.
from cloned cDNA confirms an essential role Viruses 13(9):1723. https://doi.org/10.
for the transcription elongation factor from the 3390/v13091723
5′ proximal open reading frame of the M2 28. Martin CT, Muller DK, Coleman JE (1988)
mRNA in gene expression and provides a capa- Processivity in early stages of transcription by
bility for vaccine development. Proc Natl Acad T7 RNA polymerase. Biochemistry 27(11):
Sci 92(25):11563–11567. https://doi.org/ 3966–3974. https://doi.org/10.1021/
10.1073/pnas.92.25.11563 bi00411a012
20. Borkotoky S, Murali A (2018) The highly effi- 29. Kolakofsky D, Pelet T, Garcin D, Hausmann S,
cient T7 RNA polymerase: a wonder macro- Curran J, Roux L (1998) Paramyxovirus RNA
molecule in biological realm. Int J Biol synthesis and the requirement for hexamer
Macromol 118(Pt A):49–56. https://doi. genome length: the rule of six revisited. J
org/10.1016/j.ijbiomac.2018.05.198 Virol 72(2):891–899. https://doi.org/10.
21. Fuerst TR, Earl PL, Moss B (1987) Use of a 1128/JVI.72.2.891-899.1998
hybrid vaccinia virus-T7 RNA polymerase sys- 30. Egelman EH, Wu SS, Amrein M, Portner A,
tem for expression of target genes. Mol Cell Murti G (1989) The Sendai virus nucleocapsid
Biol 7(7):2538–2544. https://doi.org/10. exists in at least four different helical states. J
1128/mcb.7.7.2538-2544.1987 Virol 63(5):2233–2243. https://doi.org/10.
22. Buchholz UJ, Finke S, Conzelmann K-K 1128/JVI.63.5.2233-2243.1989
(1999) Generation of bovine respiratory syncy- 31. Gutsche I, Desfosses A, Effantin G, Ling WL,
tial virus (BRSV) from cDNA: BRSV NS2 is Haupt M, Ruigrok RWH, Sachse C, Schoehn
Reverse Genetics Systems for Diverse Paramyxoviruses 33
Washington/1964 strain of human parain- 59. Elbehairy MA, Khattar SK, Samal SK (2021)
fluenza virus type 1 (HPIV1) and recovery Recovery of recombinant avian paramyxovirus
and characterization of wild-type recombinant type-3 strain wisconsin by reverse genetics and
HPIV1 produced by reverse genetics. Virus its evaluation as a vaccine vector for chickens.
Genes 24(1):77–92. https://doi.org/10. Viruses 13(2). https://doi.org/10.3390/
1023/a:1014042221888 v13020316
51. Durbin AP, Hall SL, Siew JW, Whitehead SS, 60. Subbiah M, Khattar SK, Collins PL, Samal SK
Collins PL, Murphy BR (1997) Recovery of (2011) Mutations in the fusion protein cleav-
infectious human parainfluenza virus type age site of avian paramyxovirus serotype
3 from cDNA. Virology 235(2):323–332. 2 increase cleavability and syncytium formation
https://doi.org/10.1006/viro.1997.8697 but do not increase viral virulence in chickens. J
52. Schmidt AC, McAuliffe JM, Huang A, Surman Virol 85(11):5394–5405. https://doi.org/10.
SR, Bailly JE, Elkins WR, Collins PL, Murphy 1128/jvi.02696-10
BR, Skiadopoulos MH (2000) Bovine parain- 61. Tsunekuni R, Hikono H, Tanikawa T,
fluenza virus type 3 (BPIV3) fusion and Kurata R, Nakaya T, Saito T (2017) Recombi-
hemagglutinin-neuraminidase glycoproteins nant avian paramyxovirus serotypes 2, 6, and
make an important contribution to the 10 as vaccine vectors for highly pathogenic
restricted replication of BPIV3 in primates. J avian influenza in chickens with antibodies
Virol 74(19):8922–8929. https://doi.org/10. against newcastle disease virus. Avian Dis
1128/jvi.74.19.8922-8929.2000 61(3):296–306. https://doi.org/10.1637/
53. Hou X, Suquilanda E, Zeledon A, Kacsinta A, 11512-100616-regr1
Moore A, Seto J, McQueen N (2005) Muta- 62. Xiao S, Khattar SK, Subbiah M, Collins PL,
tions in Sendai virus variant F1-R that correlate Samal SK (2012) Mutation of the F-protein
with plaque formation in the absence of tryp- cleavage site of avian paramyxovirus type
sin. Med Microbiol Immunol 194(3): 7 results in furin cleavage, fusion promotion,
129–136. https://doi.org/10.1007/s00430- and increased replication in vitro but not
004-0224-3 increased replication, tissue tropism, or viru-
54. Bajimaya S, Hayashi T, Takimoto T (2017) lence in chickens. J Virol 86(7):3828–3838.
Rescue of Sendai virus from cloned cDNA. https://doi.org/10.1128/jvi.06765-11
Methods Mol Biol 1602:103–110. https:// 63. Clarke DK, Sidhu MS, Johnson JE, Udem SA
doi.org/10.1007/978-1-4939-6964-7_7 (2000) Rescue of mumps virus from cDNA. J
55. Krishnamurthy S, Huang Z, Samal SK (2000) Virol 74(10):4831–4838. https://doi.org/10.
Recovery of a virulent strain of newcastle dis- 1128/jvi.74.10.4831-4838.2000
ease virus from cloned cDNA: expression of a 64. Lemon K, Rima BK, McQuaid S, Allen IV,
foreign gene results in growth retardation and Duprex WP (2007) The F gene of rodent
attenuation. Virology 278(1):168–182. brain-adapted mumps virus is a major determi-
https://doi.org/10.1006/viro.2000.0618 nant of neurovirulence. J Virol 81(15):
56. Nakaya T, Cros J, Park MS, Nakaya Y, 8293–8302. https://doi.org/10.1128/JVI.
Zheng H, Sagrera A, Villar E, Garcı́a-Sastre A, 00266-07
Palese P (2001) Recombinant Newcastle dis- 65. Sauder CJ, Zhang CX, Ngo L, Werner K,
ease virus as a vaccine vector. J Virol 75(23): Lemon K, Duprex WP, Malik T, Carbone K,
11868–11873. https://doi.org/10.1128/ Rubin SA (2011) Gene-specific contributions
JVI.75.23.11868-11873.2001 to mumps virus neurovirulence and neuroatte-
57. Peeters BP, de Leeuw OS, Koch G, Gielkens nuation. J Virol 85(14):7059–7069. https://
AL (1999) Rescue of Newcastle disease virus doi.org/10.1128/JVI.00245-11
from cloned cDNA: evidence that cleavability 66. Zhou D, Zhu M-Y, Wang Y-L, Hao X-Q,
of the fusion protein is a major determinant for Zhou D-M, Liu R-X, Zhang C-D, Qu C-F,
virulence. J Virol 73(6):5001–5009. https:// Zhao Z-Y (2019) Establishment of an efficient
doi.org/10.1128/JVI.73.6.5001-5009.1999 reverse genetic system of Mumps virus S79
58. Sun W, Leist SR, McCroskery S, Liu Y, from cloned DNA. World J Pediatr 15(5):
Slamanig S, Oliva J, Amanat F, Sch€afer A, Din- 499–505. https://doi.org/10.1007/s12519-
non KH 3rd, Garcı́a-Sastre A, Krammer F, 019-00286-8
Baric RS, Palese P (2020) Newcastle disease 67. Kawano M, Kaito M, Kozuka Y, Komada H,
virus (NDV) expressing the spike protein of Noda N, Nanba K, Tsurudome M, Ito M,
SARS-CoV-2 as a live virus vaccine candidate. Nishio M, Ito Y (2001) Recovery of infectious
EBioMedicine 62:103132. https://doi.org/ human parainfluenza type 2 virus from cDNA
10.1016/j.ebiom.2020.103132 clones and properties of the defective virus
without V-specific cysteine-rich domain.
Reverse Genetics Systems for Diverse Paramyxoviruses 35
Abstract
Reverse genetics allows for the generation of recombinant infectious viruses from viral sequences or
complete viral genomes cloned into plasmids. Using reverse genetics, it is then possible to introduce
changes in the genome of infectious viruses for multiple applications.
Newcastle disease virus (NDV) is a non-segmented, negative-sense RNA virus that has been amenable to
manipulation by reverse genetics for more than two decades. Since then, recombinant NDVs have been
extensively used as viral vectors to express heterologous proteins. We describe the key steps required to
design and introduce an additional transcription unit in the genome of the Newcastle disease virus for the
efficient expression of a heterologous gene.
Key words Reverse genetics, Mononegavirales, Recombinant Newcastle disease virus, Virus-vectored
vaccine, Codon optimization
1 Introduction
Daniel R. Pérez (ed.), Reverse Genetics of RNA Viruses: Methods and Protocols, Methods in Molecular Biology, vol. 2733,
https://doi.org/10.1007/978-1-0716-3533-9_3,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2024
37
38 Arantza Cobela-Garcı́a et al.
A) vTF7-3
P helper
plasmids
B) unique
restriction site T7 terminator
Hepatitis delta
SacII Ribozyme
NP P M F HN L
Fig. 1 (a) Basic steps followed for the rescue of a recombinant rabies virus (RV) in 1994. A similar approach is
still used nowadays. See text for details. (Not to scale). (b) Main features of the pNDV rescue plasmid. (Not to
scale)
GE+IG+GS GE+IG+GS
Fig. 2 Schematic representation of functional paramyxovirus transcription units. Each open reading frame
(white boxes) is flanked by conserved non-translated sequences (gray boxes): GE gene end, IG intergenic, GS
gene start
2 Materials
2.1 Design and 1. Software to perform simple sequence analysis such us translat-
Cloning of the ing nucleotide sequences into amino acid sequences or identi-
Additional fying restriction sites. We find BioEdit (https://bioedit.
Transcription Unit software.informer.com/7.2/) and SnapGene Viewer
(https://www.snapgene.com/snapgene-viewer/) very conve-
nient and easy to use, but there are many other options.
2. Rescue plasmid that contains the full-length cDNA of
the genome of the NDV vaccine strain LaSota, flanked by the
sequence of the promoter of the T7 RNA polymerase on the 5′
side (to transcribe the positive-sense antigenome) and by the
sequence of the ribozyme of the hepatitis delta virus and the T7
terminator on the 3′ side. The cDNA contains a unique restric-
tion site SacII engineered between the stop codon and the gene
end (GE) regulatory sequence of the P gene (Fig. 1b).
3. Reagents for cloning: restriction enzyme SacII (NEB, cat.
#R0157S) to linearize the vector. CloneAmp HiFi PCR Premix
(TaKaRa, cat. #639298). Specific primers to amplify the insert
by PCR and to confirm the insert by sequencing. Equipment
and reagents to run agarose gel electrophoresis. In-Fusion
Snap Assembly Master Mix (TaKaRa, cat. #638948). Stellar
competent bacteria and provided SOC medium (Takara Bio,
cat. #636766). Luria–Bertani (LB) agar plates with ampicillin
(100 μg/mL). Kits for DNA purification from agarose gel
bands (we use Omega Bio-Tek cat.#D2500-01) and for plas-
mid preparation (we use Omega Bio-Tek cat.#D6942-01 and
Invitrogen cat.#K210005).
4. Equipment for growing bacteria at 37 °C (incubator and
shaker).
Recombinant NDV Expressing a Foreign Gene 41
2.2 Rescue of the Rescue of lentogenic (avirulent) NDV, such as the vaccine strain
Recombinant NDV LaSota, may be performed in Biosafety Level 2 (BSL-2) conditions.
Expressing a Codon- Recombinant viruses with enhanced infectivity or pathogenicity
Optimized Influenza and virulent (mesogenic or velogenic) NDVs will require additional
Hemagglutinin biosafety and biocontainment measures.
All the tissue culture and egg inoculation work must be per-
formed in sterile conditions.
1. HEp2 cells.
2. CO2 incubator and biosafety cabinet.
3. Complete DMEM: Dulbecco’s modified Eagle medium
(DMEM) supplemented with 5% fetal bovine serum (FBS)
and antibiotics (penicillin (100 UI/mL)/streptomycin
(100 mg/mL)).
4. Phosphate-buffered saline (PBS) and PBS supplemented with
0.3% bovine serum albumin and antibiotics (PBS + BSA + P/
S).
5. Recombinant modified vaccinia virus Ankara expressing the T7
RNA polymerase (MVA-T7).
6. Opti-MEM, reduced serum medium.
7. Set of rescue plasmids: pNDV-H5 (containing the full-length
cDNA of the NDV genome with an additional transcription
unit encoding a codon-optimized H5 protein. Generated in
Subheading 3.1). Helper plasmids pTM1-N, pTM1-P, and
pTM1-L, expressing the viral proteins N, P, and L under the
control of the T7 promoter.
8. Mirus TransIT-LT1 Transfection Reagent.
9. Pathogen-free, 8–10-day-old, embryonated chicken eggs
(Charles River).
10. 0.5 mL syringes with 25G × 5/8″ needles (insulin syringes).
11. Turkey or chicken whole blood, diluted in PBS to 0.5% red
blood cells, to perform hemagglutination assay (HA).
3 Methods
3.1 In Silico Design 1. Download the coding sequence of the protein to be expressed
and Insertion into the by the recombinant NDV. As an example, we will use the
NDV Genome of an hemagglutinin (HA) from A/chicken/Iowa/04-20/2015
Additional (H5N2) (GenBank accession number: KR492974.3). Figure 3
Transcription Unit summarizes the modifications made to the original sequence.
Encoding a Chimeric 2. In the amino acid sequence, identify the multibasic cleavage
Influenza site (PQRERRRKR/GLF) and replace it by the low patho-
Hemagglutinin genic cleavage site from the same lineage (PQRETR/GLF)
(see Note 1).
42 Arantza Cobela-Garcı́a et al.
Addition of NDV
regulatory signals and
Kozak sequence (K)
codon optimization
GE+IG+GS
K HPAI H5 ectodomain L TM
SacII SacII
Flanking sequences Highly pathogenic cleavage
for cloning site replaced by low Transmembrane and cytoplasmic
pathogenic cleavage site domains (TM) of the HA replaced
by those of NDV F protein,
separated by a flexible linker (L)
Fig. 3 Modifications to the wild-type sequence of an avian influenza hemagglutinin gene in order to increase
safety, expression, and immunogenicity of the heterologous gene
cells
+
supernatant
MVA-T7
pNDV-H5
infection
pTM1-N 3 days
transfection Harvest
pTM1-P HA assay
characterization
pTM1-L
Fig. 4 Rescue of a recombinant NDV expressing influenza H5. On day 1, cells are infected with a recombinant
poxvirus expressing T7 RNA polymerase and transfected with the rescue plasmid containing the full-length
cDNA and the three helper plasmids (N, P, and L) all under the control of the T7 promoter. On day
2, supernatant and cells are inoculated into embryonated chicken eggs. On day 5, the allantoic fluid is
collected, and hemagglutination-positive samples are further analyzed
3.2 Use the In this section, we describe a basic protocol to rescue a recombinant
Sequence-Confirmed NDV from plasmids and amplification in embryonated chicken
Rescue Plasmid pNDV- eggs (Fig. 4). Detailed protocols can also be found in [14, 15].
H5 to Rescue a rNDV 1. The day before infection/transfection, plate HEp-2 cells in a
Expressing a Chimeric, 6-well plate. Cells should be around 80–90% confluent at the
Codon-Optimized H5 time of transfection.
2. Wash the cells with sterile PBS and infect with MVA-T7 diluted
in 0.5 mL of PBS + BSA + P/S, at a multiplicity of infection of
5 PFU/cell. Incubate 1 h at 37 °C, rocking the plate every
10–15 min. During this time, prepare the transfection mix.
3. Prepare the transfection mix: for each well in a sterile Eppen-
dorf tube, add 250 μL of Opti-MEM + 1 μg of pNDV-H5
rescue plasmid + 0.5 μg of pTM1-N + 0.25 μg of pTM1-
P + 0.25 μg of pTM1-L + 6 μL of TransIT-LT1 Transfection
Reagent. Mix well and incubate at room temperature for about
20 min (until the end of the 1 h MVA-T7 infection).
44 Arantza Cobela-Garcı́a et al.
4 Notes
References
1. Amarasinghe GK, Ayllon MA, Bao Y, Basler cDNA. EMBO J 13(18):4195–4203. https://
CF, Bavari S, Blasdell KR, Briese T, Brown doi.org/10.1002/j.1460-2075.1994.
PA, Bukreyev A, Balkema-Buschmann A, tb06739.x
Buchholz UJ, Chabi-Jesus C, Chandran K, 3. Pfaller CK, Cattaneo R, Schnell MJ (2015)
Chiapponi C, Crozier I, de Swart RL, Dietzgen Reverse genetics of Mononegavirales: how
RG, Dolnik O, Drexler JF, Durrwald R, Dun- they work, new vaccines, and new cancer ther-
don WG, Duprex WP, Dye JM, Easton AJ, apeutics. Virology 479-480:331–344. https://
Fooks AR, Formenty PBH, Fouchier RAM, doi.org/10.1016/j.virol.2015.01.029
Freitas-Astua J, Griffiths A, Hewson R, 4. Molouki A, Peeters B (2017) Rescue of recom-
Horie M, Hyndman TH, Jiang D, Kitajima binant Newcastle disease virus: a short history
EW, Kobinger GP, Kondo H, Kurath G, Kuz- of how it all started. Arch Virol 162(7):
min IV, Lamb RA, Lavazza A, Lee B, Lelli D, 1845–1854. https://doi.org/10.1007/
Leroy EM, Li J, Maes P, Marzano SL, s00705-017-3308-2
Moreno A, Muhlberger E, Netesov SV,
Nowotny N, Nylund A, Okland AL, 5. Calain P, Roux L (1993) The rule of six, a basic
Palacios G, Palyi B, Paweska JT, Payne SL, feature for efficient replication of Sendai virus
Prosperi A, Ramos-Gonzalez PL, Rima BK, defective interfering RNA. J Virol 67(8):
Rota P, Rubbenstroth D, Shi M, Simmonds P, 4822–4830. https://doi.org/10.1128/JVI.
Smither SJ, Sozzi E, Spann K, Stenglein MD, 67.8.4822-4830.1993
Stone DM, Takada A, Tesh RB, Tomonaga K, 6. Peeters BP, de Leeuw OS, Koch G, Gielkens
Tordo N, Towner JS, van den Hoogen B, AL (1999) Rescue of Newcastle disease virus
Vasilakis N, Wahl V, Walker PJ, Wang LF, from cloned cDNA: evidence that cleavability
Whitfield AE, Williams JV, Zerbini FM, of the fusion protein is a major determinant for
Zhang T, Zhang YZ, Kuhn JH (2019) Taxon- virulence. J Virol 73(6):5001–5009. https://
omy of the order Mononegavirales: update doi.org/10.1128/JVI.73.6.5001-5009.1999
2019. Arch Virol 164(7):1967–1980. 7. Romer-Oberdorfer A, Mundt E, Mebatsion T,
https://doi.org/10.1007/s00705-019- Buchholz UJ, Mettenleiter TC (1999) Gener-
04247-4 ation of recombinant lentogenic Newcastle dis-
2. Schnell MJ, Mebatsion T, Conzelmann KK ease virus from cDNA. J Gen Virol 80(Pt 11):
(1994) Infectious rabies viruses from cloned
46 Arantza Cobela-Garcı́a et al.
Abstract
Influenza A (FLUAV) and influenza B (FLUBV) viruses are human and/or animal pathogens widely
studied due to their importance to public health and animal production. Both FLUAV and FLUBV possess
a genome composed of eight viral gene segments. For reverse genetics of influenza viruses, transcription of
the mRNA for the viral proteins is typically done from a plasmid encoding an RNA polymerase II (pol II)
promoter element upstream of cloned viral cDNA and expressed like host mRNA. On the other side, the
synthesis of the negative-sense, single-stranded, uncapped vRNAs can be accomplished by the host’s RNA
polymerase I (pol I). The reverse genetics for influenza has allowed the manipulation of influenza genomes
incorporating heterogeneous sequences into different segments of the influenza genome, such as reporter
genes. In this chapter, we outline the protocol from the generation of reverse genetic plasmid that can be
applied for the cloning of any of the segments of FLUAV or FLUBV. Furthermore, we describe a protocol
for generating FLUAV or FLUBV recombinant viruses carrying Nanoluciferase (NLuc) in the PB1 gene
using reverse genetics. Finally, we delineate a microneutralization protocol using FLUAV-NLuc or
FLUBV-NLuc viruses optimized for the use of antibodies from different sources (mice, ferrets, avian,
etc.), which provides a more sensitive, reliable, and avidity-independent method to assess the presence of
neutralizing antibodies against FLUAV or FLUBV.
1 Introduction
Daniel R. Pérez (ed.), Reverse Genetics of RNA Viruses: Methods and Protocols, Methods in Molecular Biology, vol. 2733,
https://doi.org/10.1007/978-1-0716-3533-9_4,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2024
47
48 C. Joaquin Caceres et al.
Fig. 1 Schematic representation of the rescue of recombinant influenza viruses expressing NLuc using reverse
genetics. Eight plasmids (one per influenza segment) are transfected in a co-culture of 293T/MDCK cells.
Transfection supernatants are collected and used to amplify influenza in embryonated chicken eggs or MDCK
cells, generating recombinant influenza viruses expressing NLuc. Influenza-NLuc viruses can be subsequently
used to evaluate the presence of neutralizing antibodies. The NLuc activity is used as a readout of the viral
replication and is negatively correlated to the presence of neutralizing antibodies in the sample evaluated.
(Figure created with BioRender.com)
2 Materials
2.2 Amplification of 1. Influenza A primer set for cloning the eight gene segments into
Influenza A and B Gene pDP2002 bidirectional reverse genetic plasmid vector
Segments from vRNA (Table 1) (see Note 1).
2. Influenza B primer set for cloning the eight gene segments into
pDP2002 bidirectional reverse genetic plasmid vector
(Table 1).
3. Superscript III One-Step RT-PCR System with Platinum Taq
DNA Polymerase (Thermo Fisher Scientific).
4. Nuclease-free water (i.e., QIAGEN).
5. Agarose, LE analytical grade (i.e., Promega).
6. SYBR® Safe gel staining (Invitrogen).
7. 1 kb ladder (Thermo Scientific).
8. QIAquick gel extraction kit (QIAGEN).
9. 1.5 mL microcentrifuge tubes (i.e., USA Scientific).
10. 250 μL PCR tubes (i.e., USA Scientific).
11. Ice or cooling tube racks.
12. Laboratory equipment: pipettes and tips of appropriate sizes,
vortex, microcentrifuge with a rotor capable of reaching up to
12,000×g, thermocycler, DNA gel electrophoresis system,
transilluminator, water bath or heating block capable of reach-
ing 50 °C, scale capable of determining weight in milligrams,
NanoDrop (or similar spectrophotometer).
Forward Reverse
Gene primer primer
FLUAV
PB2 Bm-PB2 1F TATTCGTCTCAGGGAGCGAAAGCAGGTC Bm-PB2 ATATCGTCTCGTATTAGTAGAAACAAGGTCG
2341R TTT
PB1 Bm-PB1 1F TTATTCGTCTCAGGGAGCGAAAGCAGGCA Bm-PB1 ATATCGTCTCGTATTAGTAGAAACAAGGCA
2341R TTT
PA Bm-PA-1F TATTCGTCTCAGGGAGCGAAAGCAGGTAC Bm-PA 2233R ATATCGTCTCGTATTAGTAGAAACAAGGTAC
TT
HA Bm-HA 1F TATTCGTCTCAGGGAGCAAAAGCAGGGG Bm-NS 890R ATATCGTCTCGTATTAGTAGAAACAAGGGTG
TTTT
NP Bm-NP-1F TATTCGTCTCAGGGAGCAAAAGCAGGGTA Bm-NP 1565R ATATCGTCTCGTATTAGTAGAAACAAGGGTA
TTTTT
NA Bm-NA 1F TATTCGTCTCAGGGAGCAAAAGCAGGAGT Bm-NA 1413R ATATCGTCTCGTATTAGTAGAAACAAGGAG
TTTTTT
M Bm-M-1F TATTCGTCTCAGGGAGCAAAAGCAGGTAG Bm-M 1027R ATATCGTCTCGTATTAGTAGAAACAAGGTAG
TTTTT
NS Bm-NS 1F TATTCGTCTCAGGGAGCAAAAGCAGGGTG Bm-NS 890R ATATCGTCTCGTATTAGTAGAAACAAGGGTG
TTTT
FLUBV
PB2 Bm-PB2b 1F TATTCGTCTCAGGGAGCAGAAGCGGAGCG Bm-PB2b ATATCGTCTCGTATTAGTAGAAACACGAGCA
TTTTCAAGATG 2396R TT
Reverse Genetics for Influenza NanoLuc Viruses
(continued)
51
52
Table 1
(continued)
Forward Reverse
Gene primer primer
PA Bm-PAb-1F TATTCGTCTCAGGGAGCAGAAGCGGTGCG Bm-PAb ATATCGTCTCGTATTAGTAGAAACACGTGCA
TTTGA 2308R TT
HA MDV-B 5′ TATTCGTCTCAGGGAGCAGAAGCAGAGCA MDV-B 3′ ATATCGTCTCGTATTAGTAGTAACAAGAGCA
Bm-HA TTTTCTAATATC Bm-HA TTTTTC
NP MDV-B 5′ TATTCGTCTCAGGGAGCAGAAGCACAGCA MDV-B 5′ ATATCGTCTCGTATTAGTAGAAACAACAGCA
C. Joaquin Caceres et al.
2.5 Cell Culture 1. T75 tissue culture flasks, canted neck (VWR).
Growth and 2. Dulbecco’s Modified Eagle’s Medium (DMEM) high glucose
Maintenance (1×) liquid; with L-glutamine; sodium pyruvate, and sodium
bicarbonate (Sigma).
3. 100× antibiotic–antimycotic solution (100 units/mL penicil-
lin G, 0.1 mg/mL streptomycin sulfate, and 0.25 μg/mL
amphotericin B) (VWR).
4. L-Glutamine solution 200 mM (Sigma).
5. Phosphate-buffered saline (Thermo Fisher).
6. Fetal bovine serum (FBS), heat-inactivated, sterile-filtered
(Sigma).
54 C. Joaquin Caceres et al.
7. Trypsin–EDTA (Sigma).
8. Individually wrapped serological pipettes of the appropriate
size as needed.
9. Sterile tubes or bottles appropriate to prepare and store media
and reagents.
10. Laboratory equipment: cell culture incubator set at 37 °C with
5% CO2, water bath set at 37 °C, pipettor for serological
pipettes, biosafety cabinet class II.
2.7 Propagation of 1. Cleared infectious cell culture supernatant containing the res-
Rescued Influenza A cued FLUAV or FLUBV.
and B Viruses in MDCK 2. MDCK cells (ATCC, CCL-34).
Cells
3. T75 (or T175) tissue culture flasks, canted neck (VWR).
4. Dulbecco’s Modified Eagle’s Medium (DMEM) high glucose
(1×) liquid; with L-glutamine; sodium pyruvate, and sodium
bicarbonate (Sigma).
Reverse Genetics for Influenza NanoLuc Viruses 55
2.8 Propagation of 1. Cleared infectious cell culture supernatant containing the res-
Rescued Influenza A cued FLUAV-NLuc or FLUBV-NLuc.
and B NanoLuc Viruses 2. 9- to 11-day-old specific pathogen-free (SPF) embryonated
in Embryonated chicken eggs (ECEs).
Chicken Eggs 3. 70% ethanol
4. Egg puncher (i.e., Dremel Multipro 7.2 V, model 770) or
16G × 1.5″ needle with robber stopper.
5. 1 mL syringes with 25G × 5/8″ needles.
6. 10 mL syringes.
7. 18G × 1″ needles.
8. Glue to seal the eggs (i.e., Elmer’s glue).
9. 0.5% turkey or chicken red blood cells .
10. Phosphate-buffered saline (PBS) (Thermo Fisher).
11. 96-well V-bottom plates.
12. Multichannel pipette able to dispense 50 μL with
corresponding pipette tips.
13. Laboratory equipment: egg candler, egg incubator set at 33 °C
or 35 °C, tabletop minicentrifuge, biosafety cabinet class II,
refrigerated centrifuge capable of containing 15 and 50 mL
conical tubes set at 4 °C, ultra-freezer set at -80 °C.
56 C. Joaquin Caceres et al.
3 Methods
3.1 vRNA Extraction Dedicated equipment, supplies, and reagents solely for RNA work
from Influenza A and B are highly recommended; see Notes 3 and 4 for the precautions and
Viruses recommendations to be followed during the RNA extraction pro-
cedure. Below we describe the RNA extraction procedure using the
QIAamp Viral RNA kit (QIAGEN).
1. Equilibrate buffer AVE to room temperature.
2. Prepare buffer AW1 and Buffer AW2 according to manufac-
turer’s instructions.
3. Dispense 560 μL buffer AVL into a 1.5 mL microcentrifuge
tube. If the sample volume is >140 μL, then increase AVL
buffer proportionally.
4. Add 140 μL of the sample to buffer AVL and pulse-vortex for
15 s.
5. Incubate at room temperature for 10 min.
6. Briefly centrifuge to remove droplets from the lid.
7. Add 560 μL of ethanol (96–100%) and pulse-vortex for 15 s.
Briefly centrifuge to remove droplets from the lid.
8. Add 630 μL of the solution from step 5 to the column (within
a 2 mL collection tube provided with the kit). Close the cap
and centrifuge at 6000×g for 1 m. Transfer the column into a
clean 2 mL collection tube and discard the previous tube.
9. Repeat step 8 until all the lysate has been passed though the
column.
10. Add 500 μL of buffer AW1 to the column. Close the cap and
centrifuge at 6000×g for 1 min.
11. Transfer the column into a clean 2 mL collection tube and
discard the previous tube.
12. Add 500 μL of buffer AW2 to the column. Close the cap and
centrifuge at 6000×g for 1 min.
13. Transfer the column into a clean 2 mL collection tube and
centrifuge at full speed for 1 min.
58 C. Joaquin Caceres et al.
3.2 FLUAV or FLUBV 1. Amplification of FLUAV and FLUBV gene segments from
RT-PCR Amplification vRNA can be achieved with any RT-PCR system; however, we
recommend the use of Superscript III One-Step RT-PCR System
with Platinum Taq DNA Polymerase (Thermo Fisher Scientific)
as it has rendered the most robust results, and the protocol
described below has been optimized for the use of this
RT-PCR system. See Table 1 for gene-specific primers to be used.
2. In a PCR tube, add 2.5 μL of the RNA previously extracted and
keep the tubes on ice.
3. In a 1.5 mL microcentrifuge tube, prepare reaction mix 2 as
follows (adjust volume accordingly based on number of reac-
tions needed. 25 μL reactions can be prepared by cutting in half
the volumes for each reagent):
3.3 Cloning of FLUAV 1. Using the purified PCR products from step 11, Subheading
and FLUBV Gene 2.2, set up the enzymatic digestions as follows:
Segments into
pDP2002 (a) Purified PCR product 26 μL
(b) 10× restriction endonuclease buffer 3 μL
(c) Nuclease-free water 20 μL
(d) Endonuclease (BsmBI, BsaI, or AarI) 1 μL
Vector size in bp
= molecular ratio∴
Insert size in bp
Reverse Genetics for Influenza NanoLuc Viruses 61
50 ng of vector
= 1 part of insert in ng × 3
molecular ratio
= 3 parts of insert in ng
15. Calculate the volume of insert and vector needed based on the
concentrations obtained in steps 3 and 11 and the calculations
from step 13.
16. Set ligation reactions as follows for a total reaction volume of
15 μL (see Note 8):
17. Mix well, quick spin, and set to incubate at room temperature
for 5 min and then place the reaction in ice. Ligation reactions
can be stored at -20 °C if it cannot be used for transformation
on the same day.
18. Transform ligation reactions into TOP10 cells following the
manufacturer’s instructions.
19. Spread 100 μL of transformation rxn onto LB agar plates
supplemented with 50 μg of ampicillin.
20. Set to incubate overnight at 37 °C. Transformants can be
screened by colony PCR (cPCR) the following day.
3.4 Screening of After 16–18 h of incubation at 37 °C, plates from step 15, Sub-
Transformant Colonies heading 3.3, should have colonies with potential positive reverse
by cPCR genetic (RG) clones carrying the desired FLUAV and FLUBV
gene segments. It is recommended to screen between five and
ten transformant colonies. To screen for RG clones carried into
the pDP2002 vector, we recommend using the following
primer set: T7Seq (5′-TAATACGACTCACTATAGG-3′) and
SeqPol1 (5′-GAGGTATATCTTTCGCTCCG-3′).
1. Before starting, pre-warm an LB agar plate (with 50 μg/mL of
ampicillin). Draw a grid and label the compartments with the
corresponding colony ID numbers. This plate will serve to save
a copy of the colonies selected for screening.
2. In a 1.5 mL microcentrifuge tube, prepare cPCR reactions as
follows (amounts disclosed below are per reaction; adjust
depending on the number of reactions to be performed):
62 C. Joaquin Caceres et al.
13. The next day, pellet the bacterial cultures at 3000 rpm/10 m,
discard the supernatant, and proceed with plasmid purification
using the QIAprep Spin Mini kit (QIAGEN). Follow manu-
facturer’s instructions (see Note 9). Measure the concentration
using the spectrophotometer of your choice. DNA can be
stored at -20 °C if not used immediately.
The purified plasmids are now ready for sequencing. In
addition to cPCR screening, plasmids need to be sequenced to
confirm the presence of unexpected mutations in either the
gene segment or the plasmid vector, as well as the presence of a
complete FLUAV or FLUBV gene segment and its orientation. The
following primer set is recommended for Sanger sequencing:
pCMVF (5′-ATTACATGTTGCCAAGTACGCCCCCTATT
GACG-3′), T7Seq (5′-TAATACGACTCACTATAGG-3′), Seq-
Pol1 (5′-GAGGTATATCTTTCGCTCCG-3′) and BGH-R (5′-T
AGAAGGCACAGTCGAGG-3′).
3.5 Cell Culture If cells have been kept in liquid nitrogen, perform fast thawing
Growth and following source’s instructions. For general growth and care, fol-
Maintenance low the recommendation listed below (volumes adjusted for T75
flask).
1. After extracting a vial of cells from the liquid nitrogen, thaw
quickly in 37 °C water bath.
2. Dispense the vial’s contents into a T75 flask containing 10 ml
of pre-warmed DMEM supplemented with 10% FBS and 1×
penicillin/streptomycin solution, and 2 mM L-glutamine
(complete media).
3. Place the cells into a cell culture incubator set at 37 °C/5%
CO2.
4. The next day, remove the media; wash cells with 5 mL of
pre-warmed, sterile 1× PBS; and add 10 mL of fresh
pre-warmed complete media.
5. When the cells have reached confluence (about 48 h), remove
and discard the media.
6. Wash cells with 2 mL of pre-warmed, sterile 1× PBS.
7. Add 2 mL (MDCK) or 1 mL (HEK293T) of 0.25% trypsin–
EDTA solution and incubate it for 10 min (MDCK cells) or
3 min (HEK293T) at 37 °C (in the cell culture incubator) until
the cells have completely detached. You may tap the sides of the
flask to help with detachment. Incubation time can be
extended up to 20 min for MDCK cells if needed.
8. Add 8 mL of complete media to stop the trypsinization and
wash the bottom of the flask by pipetting up and down (see
Note 10).
64 C. Joaquin Caceres et al.
3.7 Propagation of 1. The day before infection, wash and treat MDCK cells with
FLUAV-NLuc and trypsin as described in Subheading 3.5. Resuspend cells with
FLUBV-NLuc in MDCK 10 mL of complete growth media (DMEM supplemented with
Cells 10% FBS and 1× penicillin/streptomycin solution, 2 mM
L-glutamine).
2. Into a new T75 flask, add 2 mL of cell suspension and 10 mL of
complete media.
3. Set cells to incubate at 37 °C/5% CO2 overnight (~16 to 24 h).
4. Once the cells have reached ~80% to 90% confluency, using
plain Opti-MEM, make serial dilutions of your samples (rang-
ing between 1/100 and 1/10,000) from the FLUAV-NLuc/
FLUBV-NLuc transfection or blind passage supernatant (from
Subheading 3.6), based on the CPE severity and of HA titer.
We recommend the propagation of each virus from two differ-
ent dilutions.
5. Wash cells 2× with 5 mL of 1× PBS.
6. Next, inoculate cells (in the T75 flask) with 3 mL of the
FLUAV-NLuc/FLUBV-NLuc dilutions and incubate for 1 h
at 35 °C/5% CO2, rocking the plate every 15 min.
7. Afterward, remove the inoculum and add 10 mL of Opti-
MEM-AB/AM+1 μg/mL TPCK-trypsin per well and monitor
daily for CPE and/or HA assay at 48–72 hpi. CPE may start
appearing at 48 hpi. Incubation may be prolonged beyond
72 hpi (up to 120 hpi) if deemed appropriate to maximize
Reverse Genetics for Influenza NanoLuc Viruses 67
3.8 Propagation of 1. Candle embryos and mark the limit between the air chamber
FLUAV-NLuc and and the allantoid cavity.
FLUBV-NLuc in ECEs 2. Label eggs accordingly.
3. Prepare tenfold dilutions of the virus starting by mixing 100 μL
of transfection or blind passage supernatant (from Subheading
3.6) with 900 μL of 1xPBS (supplemented with 1× antibiotic–
antimycotic solution) and keep in ice until needed.
4. Decontaminate eggshell with 70% ethanol or ethanol/iodine
solution.
5. Drill/punch a hole on the shell a few millimeters (~3 mm) from
the mark between the air chamber and the allantoid cavity.
6. Using 1 mL syringes with a needle (25G × 5/8″), inoculate
each dilution into three to five embryos with 100 μL/embryo.
Three different dilutions (ranging from 10-2 to 10-7) are
recommended.
7. Seal the hole with glue.
8. Set embryos to incubate at 33 °C (FLUBV-NLuc) or 35 °C
(FLUAV-NLuc) for 48 h candling daily to remove unspecific
mortality. No virus-induced mortality is expected from
embryos inoculated with low pathogenic FLUAV or FLUBV.
9. Once incubation has been completed, chill embryos overnight
at 4 °C.
10. Using 1 mL syringes with needle, collect 100 μL of allantoic
fluid (AF) from each chilled embryo into tubes or a 96-well
plate.
11. Prepare 96-well V-bottom plates (as many as needed) by add-
ing 50 μL of 1× PBS to each well.
12. Add 50 μL of AF to individual wells of column 1.
13. Perform twofold dilutions from columns 1 through 12.
14. Next, add 50 μL of 0.5% turkey red blood cells to each well.
15. Tap or shake the plate to homogenize and set to incubate for
30 min at room temperature.
68 C. Joaquin Caceres et al.
16. Select the highest dilution at which all the inoculated embryos
give the highest HA titer to avoid viral interfering particles.
17. Into a single 50 mL conical tube (set in ice), collect as much AF
as possible from each egg from the selected dilution.
18. Next, clear the AF by centrifugation at 2500 rpm for 10 min at
4 °C.
19. Use the cleared AF to prepare aliquots of the desired volume.
20. Store aliquots at -80 °C.
3.9 Titration of 1. The day before infection, prepare a 96-well plate with 1.5 × 104
FLUAV-NLuc and MDCK cells/well. Use a total volume of 100 μL of Opti-
FLUBV-NLuc by Tissue MEM-AB/AM.
Culture Infectious 2. Set cells to incubate at 37 °C/5% CO2 overnight (~16 to 24 h).
Dose 50 (TCID50) 3. Once the cells have reached ~80% to 90% confluency, use a new
96-well plate to prepare the virus dilutions. For this, prepare
tenfold serial dilutions of virus dispensing 216 μL of Opti-
MEM-AB/AM containing 1 μg/mL TPCK-trypsin to
each well.
4. Thaw an aliquot of the virus to be used on ice.
5. Add 24 μL of virus to the first four wells of the 96-well plate
(A1, B1, C1, and D1) prepared in step 3. If two viruses are
being titrated, repeat the same procedure with wells E1, F1,
G1, and H1.
6. Pipette up and down between seven and ten times to properly
mix the contents of the first column (A1–H1) and aspirate
24 μL into the next column (A2–H2). Do not pipette up and
down after the sample has been deposited into the next col-
umn. Ensure to change tips with each dilution.
7. Once the virus dilutions are completed, remove the media from
the cells prepared the day before. Aspirate 200 μL from the
dilution plate starting from column 12 and deposit the volume
into the plate containing cells. Repeat the same process with
each column until dilution -1 (Column A) has been added
(there is no need of changing tips).
8. Incubate cells at 35 °C/5% CO2 for 72 h.
9. After 72 h, remove plates from the incubator and aspirate 50 μL
of supernatant starting from the highest dilution into an HA
assay plate.
10. Next, add 50 μL of 0.5% turkey or chicken red blood cells.
11. Tap HA assay plate gently and incubate for 30 min at room
temperature.
12. Proceed to virus titer calculation using the Reed–Muench
method [26].
Reverse Genetics for Influenza NanoLuc Viruses 69
3.10 Use of FLUAV- 1. The day before performing the assay, prepare a 96-well plate
NLuc and FLUBV-NLuc with 1.5 × 104 MDCK cells/well. Use a total volume of 100 μL
Viruses for the of Opti-MEM-AB/AM. If sera from the mammalian origin will
Assessment of be evaluated, treat 1 volume of sera with 3 volumes of a fresh
Neutralizing aliquot of receptor-destroying enzyme (RDE, VWR) at 37 °C
Antibodies overnight.
2. The following day, stop the RDE activity by incubating the
samples at 56 °C in a water bath for 30 min. Allow the samples
to reach room temperature. Generate a 1/10 dilution of the
sera adding PBS-AB/A.
3. If sera from avian origin will be evaluated, treat the day of the
assay 1 volume of sera with 1 volume of a solution of chicken
red blood cells at 50%. Complete with PBS to make a 1/10
dilution of sera. Centrifuge at 1000 rpm for 1 min and repeat
the same process with the supernatant. Once the process is
repeated, collect the supernatant, and proceed with the assay.
4. Prepare a viral stock of concentration 100 TCID50 per 50 μL.
It is recommended to do 1/10 serial dilutions in PBS-AB/AM
of the stock until you reach the 100 TCID50 per 50 μL.
5. In a different 96-well plate, add 100 μL of serum sample to the
first column.
6. Next, add 50 μL of PBS-AB/AM to all the wells from columns
2–12. Then make serial dilutions aspirating 50 μL from column
1 into the next column. Do not pipette up and down after the
sample has been deposited into the next column. Ensure to
change tips with each dilution.
7. Once dilutions are completed, add 50 μL of FLUAV-NLuc or
FLUBV-NLuc virus (adjusted to 100 TCID50/50 μL) start-
ing from the highest dilution.
8. Incubate at 35 °C/5% CO2 for 1 h.
9. After the incubation is complete, remove the media from the
96-well MDCK plate prepared in step 1 and transfer the serum
dilutions to the MDCK plate.
10. Incubate for 15 min at 4 °C, followed by 45 m at 35 °C.
11. Discard the serum dilutions and add 200 μL Opti-MEM-AB/
AM containing 1 μg/mL TPCK-trypsin to each well.
12. Incubate at 35 °C/5% CO2 for 48 h.
13. Once incubation for 48 h is completed, proceed to the NLuc
activity reading using the Nano-Glo® Luciferase Assay System
(Promega).
14. Set the assay buffer to that until it reaches room temperature.
15. Turn on the 96-well luciferase reader that will be used to
record the luciferase data.
70 C. Joaquin Caceres et al.
3.11 Analysis of 1. For the data analysis and generations of graphs, we recommend
Luciferase Expression the use of the software PRISM (https://www.graphpad.com)
and Determination of (Fig. 2).
the Effective Serum 2. Export the luciferase data of your mock-infected control (virus
Dilution 50 (ESD50) in the absence of antibodies) and the samples of interest.
3. Plot the data showing the dilution factor of the serum of
interest in the X-axis and the luciferase values in log10 scale
on the Y-axis as shown in Fig. 2.
4. To calculate the serum dilution where 50% of neutralization is
observed (ESD50), analyze the data through a nonlinear
regression and obtain the EC50, which is equivalent to the
ESD50.
4 Notes
Fig. 2 Microneutralization assays using FLUAV- or FLUBV-NLuc viruses. (a–c) Mice antisera were used to
assess the presence of neutralizing antibodies using recombinant (a) FLUAV-NLuc (H1N1) against two different
homologous vaccines. (b) FLUAV-NLuc viruses (H3N1) carrying the HA of A/Hong Kong/1/68 (H3N2) (HK68) or
A/turkey/Ohio/313053/04 (H3N2) (OH04) were against homologous or heterologous antisera. (c) FLUBV-NLuc
viruses (homologous and heterologous virus) against sera raised after LAIV vaccination. (d, e) Different ferret
antisera were evaluated against (d) FLUAV-NLuc (H3N1) carrying the HA of A/Texas/1/77 (H3N2) or (e) the HA
of A/Switzerland/9715293/13 (H3N2). (f) Quail antisera raised against A/Guinea Fowl/Hong Kong/WF10/99
(H9N2) (WF10) was evaluated against FLUAV-NLuc (H9N1) carrying the homologous HA or phylogenetically
different HAs. (Figure created with BioRender.com)
HEK293T cells are less efficient for the rescue of FLUAV and
FLUBV than PK-15 cells when employing the spol I reverse
genetic system [21].
3. Dedicated equipment, supplies, and reagents solely for RNA
work are highly recommended. Disposable gloves must be
always worn while performing the RNA extraction procedure
and while handling RNA samples to avoid RNA degradation.
Extracted RNA samples should be always kept on ice and,
preferably, be used immediately for RT-PCR assays. The
remaining RNA can be kept frozen at -80 °C. Consult the
Kit’s manufacturer’s instructions for more details about
storage.
4. The vRNAs are extracted using silica-based (i.e., QIAamp Viral
RNA kit) or organic-based (TRIzol reagent, Invitrogen)
reagents or any other available RNA extraction method follow-
ing procedures recommended by the manufacturer. RNA extrac-
tion using TRIzol reagent is preferred when extracting RNA
from samples with low viral loads. We recommend the QIAamp
Viral RNA kit, which has provided consistent and reliable results,
with adequate vRNA purity and devoid of contaminants and
72 C. Joaquin Caceres et al.
Acknowledgments
References
1. King AMQ, Adams MJ, Carstens EB, Lefko- doi.org/10.1074/jbc.R110.129809.
witz EJ (2012) Ninth report of the interna- PubMed PMID: 20538598; PubMed Central
tional committee on taxonomy of viruses. PMCID: PMCPMC2937864
Elsevier Academic Press, San Diego 9. Gambaryan AS, Tuzikov AB, Piskarev VE,
2. Rajao DS, Perez DR (2018) Universal vaccines Yamnikova SS, Lvov DK, Robertson JS et al
and vaccine platforms to protect against influ- (1997) Specification of receptor-binding phe-
enza viruses in humans and agriculture. Front notypes of influenza virus isolates from differ-
Microbiol 9:123. Epub 2018/02/23. https:// ent hosts using synthetic sialylglycopolymers:
doi.org/10.3389/fmicb.2018.00123. non-egg-adapted human H1 and H3 influenza
PubMed PMID: 29467737; PubMed Central A and influenza B viruses share a common high
PMCID: PMCPMC5808216 binding affinity for 6′-sialyl(N-acetyllactosa-
3. Chen R, Holmes EC (2008) The evolutionary mine). Virology 232(2):345–350. Epub
dynamics of human influenza B virus. J Mol 1997/06/09. https://doi.org/10.1006/viro.
Evol 66(6):655–663. Epub 2008/05/28. 1997.8572
https://doi.org/10.1007/s00239-008-9119- 10. Liu C, Eichelberger MC, Compans RW, Air
z. PubMed PMID: 18504518; PubMed Cen- GM (1995) Influenza type A virus neuramini-
tral PMCID: PMCPMC3326418 dase does not play a role in viral entry, replica-
4. Palese P, Shaw M (2007) Orthomyxoviridae: tion, assembly, or budding. J Virol 69(2):
the viruses and their replication. Lippincott 1099–1106. Epub 1995/02/01. https://doi.
Williams & Wilkins, Philadelphia org/10.1128/JVI.69.2.1099-1106.1995.
5. Wright P, Nuemann G, Kawaoka Y (2007) PubMed PMID: 7815489; PubMed Central
Orthomhxoviruses. Fields virology, 5th edn. PMCID: PMCPMC188682
Lippincott Williams & Wilkins, Philadelphia 11. Webster RG, Laver WG (1967) Preparation
6. Nogales A, Perez DR, Santos J, Finch C, and properties of antibody directed specifically
Martinez-Sobrido L (2017) Reverse genetics against the neuraminidase of influenza virus. J
of influenza B viruses. In: Perez DR Immunol 99(1):49–55. Epub 1967/07/01
(ed) Reverse genetics of RNA viruses. Humana 12. Tong S, Li Y, Rivailler P, Conrardy C, Castillo
Press, New York DA, Chen LM, et al (2012) A distinct lineage
7. Palese P, Schulman JL (1976) Mapping of the of influenza A virus from bats. Proc Natl Acad
influenza virus genome: identification of the Sci U S A;109(11):4269–4274. Epub 2012/
hemagglutinin and the neuraminidase genes. 03/01. https://doi.org/10.1073/pnas.
Proc Natl Acad Sci U S A 73(6):2142–2146. 1116200109. PubMed PMID: 22371588;
Epub 1976/06/01. https://doi.org/10. PubMed Central PMCID:
1073/pnas.73.6.2142. PubMed PMID: PMCPMC3306675
1064882; PubMed Central PMCID: 13. Tong S, Zhu X, Li Y, Shi M, Zhang J,
PMCPMC430466 Bourgeois M, et al (2013) New world bats
8. Gamblin SJ, Skehel JJ (2010). Influenza hem- harbor diverse influenza A viruses. PLoS
agglutinin and neuraminidase membrane gly- Pathog 9(10):e1003657. Epub 2013/10/17.
coproteins. J Biol Chem 285(37): https://doi.org/10.1371/journal.ppat.
28403–28409. Epub 2010/06/12. https:// 1003657. PubMed PMID: 24130481;
74 C. Joaquin Caceres et al.
Abstract
New World fruit bats were recently found to harbor two distinct and previously unknown influenza A
viruses (IAVs) of the subtypes H17N10 and H18N11. Although viral genome sequences were detected in
the liver, intestine, lung, and kidney of infected bats and the complete genome sequences have been isolated
from their rectal swab samples, all attempts to isolate an infectious virus from bats in nature have failed. The
lack of an infectious bat IAV isolate was overcome by reverse genetic approaches that led to the generation
of an infectious virus in vitro. Using such synthetic bat IAVs enabled the identification of their unconven-
tional cell entry via major histocompatibility complex II (MCH-II) molecules and their ability to replicate in
mice, ferrets, and bats. Importantly, we also showed that these synthetic recombinant bat IAVs are not able
to reassort with conventional IAVs, preventing the acquisition of enhanced transmission properties in
non-bat species by reassortment with conventional IAVs. As authentic viruses are key for understanding the
molecular biology of bat IAVs, in this chapter, we describe our recently established reverse genetics protocol
for generating H17N10 and H18N11 in vitro. This step-by-step protocol starts with cloning of cDNA
copies of the viral RNA segments into reverse genetics plasmids, followed by the generation of a highly
concentrated stock and finally a method to determine viral titers.
Key words New World bat, H17N10, H18N11, Bat influenza A virus, Reverse genetics, cDNA,
HEK293T cells, RIE1495 cells
1 Introduction
Daniel R. Pérez (ed.), Reverse Genetics of RNA Viruses: Methods and Protocols, Methods in Molecular Biology, vol. 2733,
https://doi.org/10.1007/978-1-0716-3533-9_5,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2024
75
76 Susanne Kessler et al.
Fig. 1 Structure of a bat IAV virion. The eight gene segments of a bat IAV are packed inside the viral particle
and code for at least ten different proteins. The mRNA transcripts of the M segment and the NS segment have
alternative splice sites allowing their processing into the splice products M2 and nuclear export protein (NEP),
respectively. A host-derived lipid-bilayer forms the viral envelope that is stabilized by a subjacent layer of M1
proteins. The three transmembrane proteins HA, NA and M2 are embedded into the viral membrane. HA
mediates cell entry by receptor binding and membrane fusion. The ion channel M2 induces a pH-shift
releasing the genomic content. The function of bat IAV NA (N10 and N11), however, is still unknown
2 Materials
Fig. 2 Schematic workflow for generation of bat IAV stocks. HEK293T cells are transfected with eight different
pHW2000 plasmids, each coding for one gene segment of the virus. Additionally, pCAGGS expression
plasmids coding for the vRNP components PB2, PB1, PA, and NP are transfected to increase the rescue
efficiency. At 48 h post transfection, the supernatant is propagated in RIE1495 cells pretreated with DEAE in a
T75 flask. Subsequently, the supernatant from the T75 flask is collected 60 h post infection and viral particles
are concentrated via ultracentrifugation
2.2 Primers Primers used for cDNA synthesis (see Subheading 3.2) should be
designed based on the highly conserved noncoding regions of the
bat influenza vRNAs shown in Table 1. In general, primers should
be between 25 and 40 nucleotides in length and comprise some
random nucleotides (lower case), a restriction site (italics; see Note
1), the conserved terminal nucleotides (bold), and segment-specific
nucleotides (underlined):
gatcCGTCTCAGGGAGCAGAAGCAGGTCAGAGATTGTC
Primers for cloning the ORFS of PB2, PB1, PA, and NP into
pCAGGS plasmids should be between 25 and 40 nucleotides in
length and comprise some random nucleotides, followed by a
restriction site and the gene-specific nucleotides of the 5′ or 3′
end of the ORF.
2.3 Cells and Media 1. Human embryonic kidney 293T (HEK293T) cells (American
Type Culture Collection, ATCC, CRL-3216), Madin–Darby
canine kidney cells II (MDCK II), and canine RIE1495 cells
are grown in culture medium: Dulbecco’s modified Eagle’s
medium (DMEM; Gibco, Thermo Fisher Scientific) supple-
mented with 10% fetal calf serum (FCS), 100 U/mL penicillin,
and 100 mg/mL streptomycin. According to cell confluency,
cells were split two to three times per week at a ratio of 1:10.
2. HEK293T cells are transfected in Opti-MEM medium and
subsequently maintained in rescue medium: DMEM supple-
mented with 0.2% bovine serum albumin (BSA), 100 U/mL
penicillin, and 100 mg/mL streptomycin.
Table 1
Overview of the 5′ and 3′ terminal ends of the A/little yellow-shouldered bat/Guatemala/164/2009 (H17N10) and A/flat-faced bat/Peru/033/2010
(H18N11) genome segments
3 Methods
3.1 Extraction of Bat The genomic material of influenza viruses is RNA that is highly
Influenza A vRNA sensitive to degradation through RNAses found on work surfaces
and the skin. Therefore, it is highly recommended to wear dispo-
sables gloves and use sterile equipment and conditions while
handling RNA material. To further avoid RNA degradation, sam-
ples should be kept on ice during the entire process of RNA
isolation. Viral RNA (vRNA) is extracted from virus-containing
supernatant, swab samples, or organ tissue using TRIzol reagent
and then purified using the Direct-zol RNA kit (Zymo Research) or
a comparable kit according to the manufacturer’s instructions.
Purified RNA material can be stored at -80 °C until cDNA
synthesis.
3.2 cDNA Synthesis Reverse transcription (RT) of the purified vRNA (see Subheading
of Bat Influenza A 3.1) into complementary DNA (cDNA) is performed using the
Virus vRNA QIAGEN OneStep RT-PCR kit or a similar product. To generate
the cDNA of each of the eight genomic vRNA segments, an indi-
vidual RT-PCR must be done using segment-specific primers (see
Subheading 2.2). Mix the components and run the RT-PCR
according to the protocol listed in Table 2. Synthesized cDNA
can be stored at -20 °C until further use.
3.3 Cloning of Bat Bat influenza A viruses are recovered from cloned cDNA (see Sub-
Influenza A Virus heading 3.2) in vitro using bidirectional pHW2000 rescue plasmids
cDNAs into pHW2000 together with pCAGGS protein expression plasmids. We recom-
Rescue Plasmids and mend the additional use of pCAGGS helper plasmids for the rescue
pCAGGS Helper protocol as it increases viral yields. For this, the ORF of the cDNA
Plasmids sequences of the viral ribonucleoprotein components PB2, PB1,
PA, and NP are individually inserted into the pCAGGS plasmid
backbone. Similarly, the cDNA sequences of the eight viral gene
segments are inserted into the pHW2000 backbone.
1. Perform an enzymatic digest of the PCR products and the
vector plasmid in a reaction tube by mixing the following
components (see Note 3) and incubating them for 2 h at 37 °C:
Table 2
Components required for the OneStep RT-PCR and the conditions used for
the thermal cycler
Component Volume
5× buffer 10 μL
dNTP mix (10 mM) 1 μL
Primer A forward 2 μL
Primer B reverse 2 μL
OneStep RT-PCR enzyme mix 2 μL
RNase inhibitor 2 μL
Total RNA 1–10 μL (0.1–2 μg)
Sterile RNase-free H2O To 50 μL
Reverse transcription 30 min 50 °C
Initial PCR activation step 15 min 95 °C
Three-step cycle (32 cycles)
Denaturation 1 min 94 °C
Annealing 1 min 58 °C
Extension 1–2 min 72 °C
Final extension 10 min 72 °C
3.4 Rescue of Bat The rescue procedure is exemplarily described for one transfection
Influenza A Virus by reaction (one well) in a 6-well plate and can be easily adjusted to
Reverse Genetics more reactions and a different format (12-well or 24-well). One day
prior to transfection, 5 × 105 HEK293T cells are seeded per well of
a 6-well plate in 2 mL culture medium and are grown at 37 °C with
5% CO2.
1. Bring Opti-MEM® medium to room temperature before
transfection.
2. For one transfection reaction, prepare 100 μL of Opti-MEM®
medium containing 8 μL Lipofectamine 2000 transfection
reagent (2 μL/μg DNA) and incubate for 10 min at room
temperature. Meanwhile, proceed preparing the plasmid trans-
fection mixture.
3. For the plasmid transfection mixture, prepare 100 μL of Opti-
MEM® medium containing 300 ng of each of the eight
pHW2000 rescue plasmids and 400 ng of each of the four
pCAGGS expression plasmids in a 1.5 mL Eppendorf tube.
4. Add the Opti-MEM® medium–Lipofectamine 2000 mixture
to the plasmid transfection mixture, mix gently, and incubate
for 20 min at room temperature.
5. Meanwhile, replace the culture medium of the HEK293T cells
with 1.5 mL Opti-MEM® medium.
6. Carefully pipette the transfection mixture (200 μL/well) onto
the HEK293T cells in small droplets, then gently sway the
plate, and place it at 37 °C with 5% CO2.
7. At 6 to 8 h post transfection, replace the cell supernatant with
2 mL rescue medium.
Reverse Genetics of Bat Influenza A Viruses 83
3.5 Propagation of One day prior to infection, 1 × 106 RIE1495 cells are seeded in a
Bat Influenza A T-75 flask in 10 mL culture medium and are grown at 37 °C with
Viruses In Vitro 5% CO2 to reach 80–90% confluency. The following paragraph
exemplarily describes infection of a single T-75 flask. However,
to produce sufficient viral stock titers, it is recommended
to concentrate the virus-containing supernatant of multiple flasks
(see Note 5).
1. Remove culture medium from the cells and pretreat cells with
5 mL PBS containing 0.2% BSA and 1 mg/mL DEAE-dextran
for 15 min at room temperature (see Note 6). Occasionally,
sway the flask gently to prevent the cells from drying.
2. Wash cells with PBS containing 0.2% BSA.
3. Infect cells with 4 mL virus-containing rescue supernatant in
10 mL infection medium and incubate the flask at 37 °C with
5% CO2.
4. At 48 h post infection, 7 mL fresh infection medium was added
onto the cells.
5. At 60 h post infection, harvest the cell culture supernatant and
remove cell debris through centrifugation at 4000×g for
10 min at room temperature. Transfer the supernatant in a
fresh tube.
6. Prepare an ultracentrifugation tube by pipetting a 5 mL 30%
sucrose cushion, then carefully layer the 17 mL cell superna-
tant, and concentrate the propagated virus through ultracen-
trifugation at 25,000 rpm for 2 h at 4 °C (Beckman coulter,
Optima L-90K Ultracentrifuge, SW 32 Ti rotor).
7. After ultracentrifugation, carefully remove the culture medium
and the sucrose layer and resuspend the virus pellet in 1–2 mL
infection medium.
3.6 Titration of Bat Viral titers are determined as endpoint titers by immunofluores-
Influenza A Viruses cence assay. It is recommended to titrate virus stocks at least in
triplicates. One day prior to infection, 1 × 104 MDCK II cells per
well of a 96-well plate are seeded in 100 μL culture medium and are
grown at 37 °C with 5% CO2 to reach 90–100% confluency.
84 Susanne Kessler et al.
4 Notes
Acknowledgments
References
1. Long JS et al (2019) Host and viral determi- negative-sense-rna-viruses-2011/w/negrna_
nants of influenza A virus species specificity. viruses/209/orthomyxoviridae
Nat Rev Microbiol 17(2):67–81 3. Tong S et al (2012) A distinct lineage of influ-
2. ICTV (2022) Orthomyxoviridae. [Cited enza A virus from bats. Proc Natl Acad Sci U S
22 June 2022]. Available from: https://talk. A 109(11):4269–4274
ictvonline.org/ictv-reports/ictv_9th_report/
86 Susanne Kessler et al.
4. Tong S et al (2013) New world bats harbor harbouring the entry machinery of an influenza
diverse influenza A viruses. PLoS Pathog A virus. Nat Commun 5:4448
9(10):e1003657 15. Moreira EA et al (2016) A conserved influenza
5. Campos ACA et al (2019) Bat Influenza A A virus nucleoprotein code controls specific
(HL18NL11) virus in fruit bats, Brazil. viral genome packaging. Nat Commun 7:
Emerg Infect Dis 25(2):333–337 12861
6. Fournier E et al (2012) Interaction network 16. Sato M et al (2019) Generation of bat-derived
linking the human H3N2 influenza A virus influenza viruses and their reassortants. Sci Rep
genomic RNA segments. Vaccine 30(51): 9(1):1158
7359–7367 17. Sun X et al (2013) Bat-derived influenza hem-
7. Fournier E et al (2012) A supramolecular agglutinin H17 does not bind canonical avian
assembly formed by influenza A virus genomic or human receptors and most likely uses a
RNA segments. Nucleic Acids Res 40(5): unique entry mechanism. Cell Rep 3(3):
2197–2209 769–778
8. Gavazzi C et al (2013) An in vitro network of 18. Zhu X et al (2013) Hemagglutinin homologue
intermolecular interactions between viral RNA from H17N10 bat influenza virus exhibits
segments of an avian H5N2 influenza A virus: divergent receptor-binding and
comparison with a human H3N2 virus. pH-dependent fusion activities. Proc Natl
Nucleic Acids Res 41(2):1241–1254 Acad Sci U S A 110(4):1458–1463
9. Gavazzi C et al (2013) A functional sequence- 19. Karakus U et al (2019) MHC class II proteins
specific interaction between influenza A virus mediate cross-species entry of bat influenza
genomic RNA segments. Proc Natl Acad Sci U viruses. Nature 567(7746):109–112
S A 110(41):16604–16609 20. Moreira EA et al (2016) Synthetically derived
10. Lowen AC (2017) Constraints, drivers, and bat influenza A-like viruses reveal a cell type-
implications of influenza A virus reassortment. but not species-specific tropism. Proc Natl
Annu Rev Virol 4(1):105–121 Acad Sci U S A 113(45):12797–12802
11. White MC, Lowen AC (2018) Implications of 21. Hoffmann E et al (2000) A DNA transfection
segment mismatch for influenza virus evolu- system for generation of influenza A virus from
tion. J Gen Virol 99(1):3–16 eight plasmids. Proc Natl Acad Sci U S A
12. Ciminski K et al (2021) Influenza A viruses: 97(11):6108–6113
understanding human host determinants. 22. Ciminski K et al (2019) Bat influenza viruses
Trends Mol Med 27(2):104–112 transmit among bats but are poorly adapted to
13. Zhou B et al (2014) Characterization of uncul- non-bat species. Nat Microbiol 4(12):
tivable bat influenza virus using a replicative 2298–2309
synthetic virus. PLoS Pathog 10(10): 23. Zhong G et al (2020) Mutations in the
e1004420 neuraminidase-like protein of bat influenza
14. Juozapaitis M et al (2014) An infectious H18N11 virus enhance virus replication in
bat-derived chimeric influenza virus mammalian cells, mice, and ferrets. J Virol
94(5):e01416-19
Chapter 6
Abstract
The piscine orthomyxovirus called infectious salmon anemia virus (ISAV) is one of the most important
emerging pathogens affecting the salmon industry worldwide. The first reverse genetics system for ISAV,
which allows the generation of recombinant ISA virus (rISAV), is an important tool for the characterization
and study of this virus. The plasmid-based reverse genetics system for ISAV includes the use of a novel fish
promoter, the Atlantic salmon internal transcribed spacer region 1 (ITS-1). The salmon, viral, and
mammalian genetic elements included in the pSS-URG vectors allow the expression of the eight viral
RNA segments. In addition to four cytomegalovirus (CMV)-based vectors that express the four proteins of
the ISAV ribonucleoprotein complex, the eight pSS-URG vectors allowed the generation of infectious
rISAV in salmon cells.
Key words Infectious salmon anemia virus, ISAV, Viral RNA, Reverse genetics, Salmon cells, Trans-
fection, ITS-1, RNA polymerase I, RNA polymerase II
1 Introduction
Daniel R. Pérez (ed.), Reverse Genetics of RNA Viruses: Methods and Protocols, Methods in Molecular Biology, vol. 2733,
https://doi.org/10.1007/978-1-0716-3533-9_6,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2024
87
88 Daniela Toro-Ascuy et al.
Fig. 1 Schematic diagram of the reverse genetics system for ISA virus rescue. ASK cells are co-transfected
with 12 plasmids, 8 pSS-URG plasmids encoding each of the eight vRNA gene segments, and 4 plasmids
expressing the viral polymerase complex and the nucleoprotein (NP). After transcription of the vRNA segments
in the nucleus by the RNA polymerase I, they associate with the polymerase complex and NP to form viral
ribonucleoprotein (vRNP) complexes leading to de novo generation of ISA virus
2 Materials
2.1 Viral RNA 1. Commercial E.Z.N.A. total RNA kit I (Omega, Bio-Tek, Inc.).
Extraction Add β-mercaptoethanol to TRK buffer before use: 20 μL
β-mercaptoethanol per 1 mL of TRK buffer.
2. RNase Away.
3. 1.5 and 2 mL RNase-free tubes.
4. Molecular biology grade ethanol (100%).
5. RNase-free, DEPC-treated water.
6. Microcentrifuge.
Fig. 2 Generation of the universal reverse genetics plasmid for ISAV. (a) Schematic of the design of the
pSS-URG cassette. The universal vector contains the sequence of the Salmo salar promoter (RNA pol I pro), the
hammerhead ribozyme (HH ribozyme), the ribozyme of the hepatitis δ virus (HδV ribozyme), and the
transcription terminator of the rabbit β-globin (transcription Ter). (b) The schematic diagram illustrates the
two SapI restriction site sequences in the pSS-URG, which are cut twice by SapI, yielding the vector
fragments. The SapI sites in the vector are eliminated and the ISAV viral cDNAs can be inserted in only one
orientation by T4 DNA ligase
Table 1
Primers used for the amplification of the eight genomic segments of ISAV via RT-PCR
4. Temperature/PCR cycler.
5. Analytic grade agarose.
6. Wizard SV Gel and PCR Clean-Up System.
7. RNase-free, DEPC-treated water.
8. Chemocompetent Escherichia coli Novablue cells.
9. Luria–Bertani media.
10. Ampicillin 1 μg/mL.
11. Pure Yield TM Plasmid Miniprep system.
12. Pure Yield TM Plasmid Midiprep system.
2.5 Transfection of 1. Regular growth medium: Leibovitz medium (L-15), 10% fetal
Salmon Cells bovine serum, 50 μg/mL gentamicin, 40 μM
β-mercaptoethanol, and 6 mM L-glutamine.
2. Transfection medium: Medium L-15, 40 μM
β-mercaptoethanol, and 6 mM L-glutamine.
3. Phosphate-buffered saline (PBS): 137 mM NaCl, 2.7 mM KCl,
10 mM Na2HPO4, 2 mM KH2PO4, pH 7.4.
4. 12-well plates.
5. Fugene 6 (Promega).
6. Atlantic salmon kidney cells (ASK, ATCC_CRL-2747).
92 Daniela Toro-Ascuy et al.
Table 2
Primers used for the amplification that amplifies the PB2, PB1, PA, and NP ORFs of ISAV901_09 via
RT-PCR
3 Methods
3.1 Viral RNA 1. Mix 350 μL of virus stock with 350 mL of buffer TRK. The
Extraction protocol outlined here is based on the E.Z.N.A. total RNA kit
I; however, other RNA extraction kits or procedures may be
used (see Note 2).
Infectious Salmon Anemia Virus Reverse Genetics 93
3.2 cDNA Synthesis The following protocol is based on SuperScript™ III One-Step
and PCR RT-PCR System with Platinum® Taq DNA Polymerase. Other
commercially available enzymes or kits for reverse transcription
and PCR may be used.
1. Mix the following components on ice:
Extracted RNA 8 μL
Forward primer (10 μM) 0.5 μL
Reverse primer (10 μM) 0.5 μL
dNTPs (10 μM) 1.5 μL
RNase-free water 1 μL
3.3 Cloning of Viral The eight genomic segments of ISAV901_09 were synthesized by
cDNAs Genscript Co in the cloning vector pUC57 with each of its ends
containing a SapI restriction site. Alternatively, the pSS-URG plas-
mids containing each viral segment and the previously mentioned
elements that allow vRNA synthesis, and the expression plasmids,
can be synthesized by Genscript [15]. If that is the case, proceed
according to step 7.
For cloning of viral cDNAs into pSS-URG vector (see Note 3),
digest with restriction enzyme SapI (see Note 4).
1. 2 Digest 2 μg of each pUC57 vector containing the genomic
segments ISAV (cDNA) or the cDNA produced in the previous
step (Subheading 3.2) with the restriction enzyme SapI (5 U
enzyme/μg of DNA).
2. Digest 1 μg of pSS-URG vector with the restriction enzyme
SapI (5 U enzyme/μg of DNA).
3. Incubate digestion for 2 h at 37 °C.
4. Visualize the digestion products in a 0.8% agarose gel (w/v),
and then purify from the gel with the Wizard SV Gel and PCR
Clean-Up System according to the manufacturer’s
instructions.
5. Ligate the digested fragments using the following ligation mix:
3.4 Cloning of 1. In addition to the eight pSS-URG plasmids for the transcrip-
Expression Plasmids tion of the eight ISAV viral RNA segments, helper plasmids for
the expression of the ISAV viral proteins PB2, PB1, PA, and NP
must be prepared. As an alternative, pCDNA3.1 plasmids con-
taining the ORF of each protein can be directly purchased from
Genscript.
2. Design oligonucleotides that amplify the PB2, PB1, PA, and
NP open reading frames.
3. As a template for PCR amplification, use the respective (a) viral
cDNAs or (b) pSS-URG plasmids.
4. Clone PCR products into a protein expression vector, such as
pTriex-3 or pCI-neo or pCDNA3.1.
5. Sequence the resulting constructs to confirm the absence of
unwanted mutations introduced by PCR.
3.6 Isavirus Growth 1. Seed 2.5 × 104 ASK cells/cm2 in a 6- or 12-well plate in a
in ASK Cells regular growth medium.
2. Incubate cells for 1–2 days at 18 °C. Cells should be 80–90%
confluent at the time of infection.
3. Prepare tenfold dilutions of virus-containing supernatant col-
lected from transfected ASK cells. Dilute supernatant in infec-
tion medium.
4. Wash ASK cells twice with PBS.
5. Add 500 μL of (un)diluted virus-containing supernatant to
ASK cells. Infect ASK cells for 4 h at 15 °C.
6. Wash ASK cells three times with PBS to remove the inoculum.
7. Incubate cells with a regular growth medium.
8. Observe cells daily for CPE:
– CPE is indicative of virus replication.
– For undiluted virus-containing supernatant, CPE typically
appears within 7 days of infection.
– If no CPE is observed, analyze sample by RT-PCR in super-
natant (see Note 6).
9. When ~80% of ASK cells are lysed (typically 7 days post infec-
tion), harvest virus-containing supernatant.
10. Spin down supernatant for 5 min at 4 °C to pellet floating cells.
11. Transfer virus-containing supernatant to a new tube.
12. Store virus-containing supernatant at -80 °C until further use
(ideally store samples in aliquots).
13. Before the virus is used for further studies, its sequence should
be confirmed.
14. Before the virus is used for further studies, virus stocks should
be grown, and their titers should be determined by qPCR or
plaque assay [14].
4 Notes
Acknowledgments
References
1. Dannevig BH, Falk K, Namork E (1995) Iso- 7. Comai L (2004) Mechanism of RNA polymer-
lation of the causal virus of infectious salmon ase I transcription. Adv Protein Chem 67:123–
anaemia (ISA) in a long-term cell line from 155
Atlantic salmon head kidney. J Gen Virol 76 8. Castro J et al (1997) Molecular analysis of a
(Pt 6):1353–1359 NOR site polymorphism in brown trout
2. Garcia-Sastre A, Palese P (1993) Genetic (Salmo trutta): organization of rDNA inter-
manipulation of negative-strand RNA virus genic spacers. Genome 40(6):916–922
genomes. Annu Rev Microbiol 47:765–790 9. Reed KM, Hackett JD, Phillips RB (2000)
3. Fodor E et al (1999) Rescue of influenza A Comparative analysis of intra-individual and
virus from recombinant DNA. J Virol 73(11): inter-species DNA sequence variation in salmo-
9679–9682 nid ribosomal DNA cistrons. Gene 249(1–2):
4. Hoffmann E, Webster RG (2000) Unidirec- 115–125
tional RNA polymerase I-polymerase II tran- 10. Van Herwerden L, Caley MJ, Blair D (2003)
scription system for the generation of influenza Regulatory motifs are present in the ITS1 of
A virus from eight plasmids. J Gen Virol 81 some flatworm species. J Exp Zool B Mol Dev
(Pt 12):2843–2847 Evol 296(1):80–86
5. Neumann G et al (1999) Generation of influ- 11. Toro-Ascuy D et al (2015) Development of a
enza A viruses entirely from cloned cDNAs. reverse genetic system for infectious salmon
Proc Natl Acad Sci U S A 96(16):9345–9350 anemia virus: rescue of recombinant fluores-
6. Zobel A, Neumann G, Hobom G (1993) RNA cent virus by using salmon internal transcribed
polymerase I catalysed transcription of insert spacer region 1 as a novel promoter. Appl Envi-
viral cDNA. Nucleic Acids Res 21(16): ron Microbiol 81(4):1210–1224
3607–3614
Infectious Salmon Anemia Virus Reverse Genetics 99
12. Lanoix J, Acheson NH (1988) A rabbit beta- and titration in salmon ASK cells. J Fish Dis
globin polyadenylation signal directs efficient 37(11):989–995
termination of transcription of polyomavirus 15. Cárdenas M, Michelson S, Pérez DR,
DNA. EMBO J 7(8):2515–2522 Montoya M, Toledo J, Vásquez-Martı́nez Y,
13. Sambrook J, Fritsch EF, Maniatis T (1989) Cortez-San MM (2022) Infectious salmon
Molecular cloning: a laboratory manual. Cold anemia virus infectivity is determined by multi-
Spring Harbor Laboratory, Cold Spring ple segments with an important contribution
Harbor from segment 5. Viruses 14(3):631. https://
14. Castillo-Cerda MT et al (2014) Development doi.org/10.3390/v14030631
of plaque assay for Chilean infectious salmon 16. Munir K, Kibenge FS (2004) Detection of
anaemia virus, application for virus purification infectious salmon anaemia virus by real-time
RT-PCR. J Virol Methods 117(1):37–47
Chapter 7
Abstract
Rift Valley fever virus (RVFV) is an important mosquito-borne virus that can cause severe disease manifes-
tations in humans including ocular damage, vision loss, late-onset encephalitis, and hemorrhagic fever. In
ruminants, RVFV can cause high mortality rates in young animals and high rates of abortion in pregnant
animals resulting in an enormous negative impact on the economy of affected regions. To date, no licensed
vaccines in humans or anti-RVFV therapeutics for animal or human use are available. The development of
reverse genetics has facilitated the generation of recombinant infectious viruses that serve as powerful tools
for investigating the molecular biology and pathogenesis of RVFV. Infectious recombinant RVFV can be
rescued entirely from cDNAs containing predetermined mutations in their genomes to investigate virus–
host interactions and mechanisms of pathogenesis and generate live-attenuated vaccines. In this chapter, we
will describe the experimental procedures for the implementation of RVFV reverse genetics.
Key words Recombinant Rift Valley fever virus, Reverse genetics, T7 polymerase-driven rescue
system, Transfection of cDNAs, Plaque assay, Virus amplification
1 Introduction
Daniel R. Pérez (ed.), Reverse Genetics of RNA Viruses: Methods and Protocols, Methods in Molecular Biology, vol. 2733,
https://doi.org/10.1007/978-1-0716-3533-9_7,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2024
101
102 Breanna Tercero and Shinji Makino
Fig. 1 RVFV virion, coding strategy, and viral RNA species produced during infection. (a) RVFV virion structure:
RVFV envelope consists of a lipid bilayer and two glycoproteins, Gn (dark blue) and Gc (light blue). The core of
the virus consists of three viral RNA segments (black lines) encapsidated by the nucleocapsid, N protein
(yellow), and bound by the RNA-dependent RNA polymerase, L protein (purple), forming ribonucleoprotein
complexes. Created by Biorender.com. (b) RVFV coding strategy: RVFV carries a single-stranded, negative-
sense, tri-segmented RNA genome, consisting of the large (L), medium (M), and small (S) viral RNA segments.
The genomic strands of L and M segments are used as templates to transcribe L and M mRNAs, respectively.
The L mRNA encodes the L protein, and the M mRNA encodes the Gn/Gc, NSm, and 78 kDa proteins. The S
segment has an ambisense coding strategy, wherein the N mRNA, encoding the N protein, is transcribed from
the genomic RNA, and the NSs mRNA, encoding the NSs protein, is transcribed from the antigenomic RNA. IGR
intergenic region
Reverse Genetics System for Rift Valley Fever Virus 103
2 Materials
2.1 RVFV Reverse 1. pProT7-L (+), pProT7-M (+), and pProT7-S (+) [35]: The
Genetics Plasmids full-length antigenomic sense (+) L, M, and S RNA segments
were cloned into pProT7 RNA expression plasmids, wherein
2.1.1 Viral RNA
each viral RNA segment was placed between the T7 promoter
Expression Plasmids
and the hepatitis delta virus (HDV) ribozyme, followed by the
T7 terminator sequence. The HDV ribozyme is used to cleave
the viral RNAs and obtain authentic viral RNA 3′ ends. These
plasmids direct the synthesis of L, M, and S antigenomic RNAs
(see Notes 1 and 2).
2.2 Cell Lines 1. For virus rescue: BSR-T7/5 cells stably expressing T7 RNA
polymerase (see Note 4): BSR-T7/5 cells are a clone of the
baby hamster kidney (BHK-21) cell line that stably express the
T7 RNA polymerase under selection in media containing G418
(Geneticin) [38].
2. For virus amplification and titration: Vero E6 (African green
monkey kidney epithelial) cells.
2.3 Tissue Culture 1. BSR-T7/5 cell growth media: Glasgow’s minimal essential
Media medium (GMEM) supplemented with 10% fetal bovine
serum (FBS), 10% tryptose phosphate broth (TPB), 1 mg/
mL G418, 1X penicillin/streptomycin (100 unit/mL of peni-
cillin, 100 μg/mL of streptomycin), 1X MEM amino acid
solution, and sodium bicarbonate.
2. BSR-T7/5 cell transfection media: Same as growth media
without antibiotics.
3. Opti-MEM I (Gibco).
4. Vero E6 growth media: Dulbecco’s modified minimum essen-
tial medium (DMEM) containing 5% FBS and 1X penicillin/
streptomycin (100 unit/mL of penicillin, 100 μg/mL of
streptomycin).
5. Modified Eagle Media (MEM) 2X containing 10% FBS and
10% TPB.
6. Virus dilution media: DMEM without FBS and containing 1X
penicillin/streptomycin (100 unit/mL of penicillin, 100 μg/
mL of streptomycin).
3 Methods
Table 1
Amount of plasmid DNA, TransIT-LT1, and media for RVFV rescue
3.1.4 Virus Collection 1. Collect the tissue culture supernatant containing RVFV into a
and Clarification: P0 15 mL conical tube.
Rescue Stock 2. Remove cell debris by centrifugation (2500 × g, for 15 min at
4 °C).
3. Collect the supernatant containing the virus and discard the
cell pellet.
4. Aliquot the virus into cryotubes and store at -80 °C (see
Note 8).
3.2 Plaque Assay of It is possible that the RVFV titers from the initial rescue are low,
RVFV MP-12: P0 and, therefore, we recommend amplifying the virus to generate
Rescue Stock a working stock. Before the preparation of a working stock
of the virus (P1), the titer of the P0 rescue stock should
be determined by plaque assay. This can be used to evaluate
the transfection efficiency of the rescue system and to determine
the dilution of the P0 virus stock required for the generation
of the P1 working stock (see Note 9).
1. Seed ~5 × 105 Vero E6 cells/well in a 6-well plate in growth
medium approximately 24 h prior to infection.
2. Incubate cells at 37 °C and 5% CO2. Cells should be 95–100%
confluent at the time of infection (see Note 10).
3. Prepare a tenfold serial dilution of the P0 virus stock (10-1–
10-6) in duplicate wells: Add 450 μL of virus dilution media
into the wells of a 96-well deep-well plate and then add 50 μL
of P0 virus stock into the first dilution wells (10-1). Carefully
pipette up and down to mix and then discard the pipette tip (see
Note 11). Using a new pipette tip, transfer 50 μL of the first
dilution into the second dilution wells (10-2) and mix. Con-
tinue this process until the final dilution (10-6).
4. Remove media from Vero E6 cells.
5. Add 400 μL of the serially diluted virus stock to Vero E6 cells.
6. Incubate cells for 1 h at 37 °C and 5% CO2. Rock the plate
gently every 15 min to ensure the even distribution of the virus
inoculum on the cells.
108 Breanna Tercero and Shinji Makino
Fig. 2 Plaque formation by MP-12 in Vero E6 cells. Vero E6 cells were infected
with MP-12 virus and overlaid with medium containing Noble agar. Plaques were
stained with neutral red solution
3. Remove media.
4. Calculate the multiplicity of infection (MOI) for the genera-
tion of P1 working stock. We recommend an MOI of 0.01 for
the infection with P0 stock virus to generate P1 working stock
(see Note 14). MOI calculation: desired PFU of virus for
infection = number of cells at confluency × MOI 0.01.
5. Dilute P0 virus stock with the virus dilution media accordingly
to prepare the accurate PFU of virus for infection at the MOI of
0.01.
6. Add the diluted virus inoculum (2 mL) to Vero E6 cells.
7. Incubate cells for 1 h at 37 °C and 5% CO2. Rock the plate
gently every 15 min to ensure the even distribution of the virus
inoculum on the cells.
8. After incubation, wash cells with PBS or growth medium to
remove the virus inoculum.
9. Add growth medium (10 mL) to cells and incubate for
2–3 days at 37 °C and 5% CO2. Observe the formation of
cytopathic effect (CPE) daily (see Note 15).
10. Collect the supernatant containing virus when the CPE is
80–90%, and clarify the supernatant, as described in Subhead-
ing 3.1, step 4 (see Note 16).
11. Aliquot the P1 working stock of virus in cryotubes and store at
-80 °C.
12. Evaluate the titer of the P1 working stock by plaque assay, as
described in Subheading 3.2 (see Notes 17 and 18).
4 Notes
Acknowledgments
References
1. Fawzy M, Helmy YA (2019) The One Health 5. Alrajhi AA, Al-Semari A, Al-Watban J (2004)
approach is necessary for the control of Rift Rift Valley fever encephalitis. Emerg Infect Dis
Valley fever infections in Egypt: a comprehen- 10(3):554–555
sive review. Viruses 11(2). https://doi.org/10. 6. Deutman AF, Klomp HJ (1981) Rift Valley
3390/v11020139 fever retinitis. Am J Ophthalmol 92(1):38–42
2. Abdel-Wahab KS, El Baz LM, El-Tayeb EM, 7. Laughlin LW, Girgis NI, Meegan JM, Straus-
Omar H, Ossman MA, Yasin W (1978) Rift baugh LJ, Yassin MW, Watten RH (1978)
Valley fever virus infections in Egypt: patholog- Clinical studies on Rift Valley fever. Part 2:
ical and virological findings in man. Trans R Ophthalmologic and central nervous system
Soc Trop Med Hyg 72(4):392–396. https:// complications. J Egypt Public Health Assoc
doi.org/10.1016/0035-9203(78)90134-7 53(3–4):183–184
3. Adam AA, Karsany MS, Adam I (2010) Mani- 8. Maar SA, Swanepoel R, Gelfand M (1979) Rift
festations of severe Rift Valley fever in Sudan. Valley fever encephalitis. A description of a
Int J Infect Dis 14(2):e179–e180. https://doi. case. Cent Afr J Med 25(1):8–11
org/10.1016/j.ijid.2009.03.029 9. Schrire L (1951) Macular changes in rift valley
4. Al-Hazmi A, Al-Rajhi AA, Abboud EB, Ayoola fever. S Afr Med J 25(50):926–930
EA, Al-Hazmi M, Saadi R, Ahmed N (2005) 10. Siam AL, Meegan JM (1980) Ocular disease
Ocular complications of Rift Valley fever out- resulting from infection with Rift Valley fever
break in Saudi Arabia. Ophthalmology virus. Trans R Soc Trop Med Hyg
112(2):313–318 74(4):539–541. https://doi.org/10.1016/
0035-9203(80)90074-7
112 Breanna Tercero and Shinji Makino
11. Siam AL, Meegan JM, Gharbawi KF (1980) (Pt 5):1207–1214. https://doi.org/10.
Rift Valley fever ocular manifestations: observa- 1099/vir.0.18876-0
tions during the 1977 epidemic in Egypt. Br J 22. Dunn EF, Pritlove DC, Jin H, Elliott RM
Ophthalmol 64(5):366–374. https://doi.org/ (1995) Transcription of a recombinant bunya-
10.1136/bjo.64.5.366 virus RNA template by transiently expressed
12. Strausbaugh LJ, Laughlin LW, Meegan JM, bunyavirus proteins. Virology
Watten RH (1978) Clinical studies on Rift Val- 211(1):133–143. https://doi.org/10.1006/
ley fever. Part I: Acute febrile and hemorrhagic- viro.1995.1386
like diseases. J Egypt Public Health Assoc 23. Flick K, Hooper JW, Schmaljohn CS, Petters-
53(3–4):181–182 son RF, Feldmann H, Flick R (2003) Rescue of
13. Swanepoel R, Manning B, Watt JA (1979) Hantaan virus minigenomes. Virology
Fatal Rift Valley fever of man in Rhodesia. 306(2):219–224. https://doi.org/10.1016/
Cent Afr J Med 25(1):1–8 s0042-6822(02)00070-3
14. Yassin W (1978) Clinico-pathological picture 24. Bouloy M, Weber F (2010) Molecular biology
in five human cases died with Rift Valley fever. J of rift valley fever virus. Open Virol J 4:8–14.
Egypt Public Health Assoc 53(3–4):191–193 h t t p s : // d o i . o r g / 1 0 . 2 1 7 4 /
15. Balkhy HH, Memish ZA (2003) Rift Valley 1874357901004020008
fever: an uninvited zoonosis in the Arabian 25. Sun Y, Li J, Gao GF, Tien P, Liu W (2018)
Peninsula. Int J Antimicrob Agents Bunyavirales ribonucleoproteins: the viral rep-
21(2):153–157 lication and transcription machinery. Crit Rev
16. Andriamandimby SF, Randrianarivo- Microbiol 44(5):522–540. https://doi.org/
Solofoniaina AE, Jeanmaire EM, 10.1080/1040841X.2018.1446901
Ravololomanana L, Razafimanantsoa LT, 26. Gauliard N, Billecocq A, Flick R, Bouloy M
Rakotojoelinandrasana T, Razainirina J, (2006) Rift Valley fever virus noncoding
Hoffmann J, Ravalohery JP, Rafisandratantsoa regions of L, M and S segments regulate RNA
JT, Rollin PE, Reynes JM (2010) Rift Valley synthesis. Virology 351(1):170–179
fever during rainy seasons, Madagascar, 2008 27. Ikegami T, Won S, Peters CJ, Makino S (2007)
and 2009. Emerg Infect Dis 16(6):963–970. Characterization of Rift Valley fever virus tran-
https://doi.org/10.3201/eid1606.091266 scriptional terminations. J Virol
17. Madani TA, Al-Mazrou YY, Al-Jeffri MH, Mis- 81(16):8421–8438. https://doi.org/10.
hkhas AA, Al-Rabeah AM, Turkistani AM, 1128/JVI.02641-06
Al-Sayed MO, Abodahish AA, Khan AS, Ksia- 28. Albarino CG, Bird BH, Nichol ST (2007) A
zek TG, Shobokshi O (2003) Rift Valley fever shared transcription termination signal on neg-
epidemic in Saudi Arabia: epidemiological, ative and ambisense RNA genome segments of
clinical, and laboratory characteristics. Clin Rift Valley fever, sandfly fever Sicilian, and Tos-
Infect Dis 37(8):1084–1092 cana viruses. J Virol 81(10):5246–5256.
18. Gargan TP 2nd, Clark GG, Dohm DJ, Turell https://doi.org/10.1128/JVI.02778-06
MJ, Bailey CL (1988) Vector potential of 29. Lara E, Billecocq A, Leger P, Bouloy M (2011)
selected North American mosquito species for Characterization of wild-type and alternate
Rift Valley fever virus. Am J Trop Med Hyg transcription termination signals in the Rift
38(2):440–446 Valley fever virus genome. J Virol
19. Rolin AI, Berrang-Ford L, Kulkarni MA 85(23):12134–12145. https://doi.org/10.
(2013) The risk of Rift Valley fever virus intro- 1128/JVI.05322-11
duction and establishment in the United States 30. Olschewski S, Cusack S, Rosenthal M (2020)
and European Union. Emerg Microbes Infect The cap-snatching mechanism of bunyaviruses.
2(12):e81. https://doi.org/10.1038/emi. Trends Microbiol 28(4):293–303. https://doi.
2013.81 org/10.1016/j.tim.2019.12.006
20. Accardi L, Prehaud C, Di Bonito P, Mochi S, 31. Hopkins K, Cherry S (2013) Bunyaviral
Bouloy M, Giorgi C (2001) Activity of Toscana cap-snatching vs. decapping: recycling cell
and Rift Valley fever virus transcription com- cycle mRNAs. Cell Cycle 12(24):3711–3712.
plexes on heterologous templates. J Gen Virol https://doi.org/10.4161/cc.26878
82(Pt 4):781–785. https://doi.org/10.1099/ 32. Spiegel M, Plegge T, Pohlmann S (2016) The
0022-1317-82-4-781 role of phlebovirus glycoproteins in viral entry,
21. Blakqori G, Kochs G, Haller O, Weber F assembly and release. Viruses 8(7). https://doi.
(2003) Functional L polymerase of La Crosse org/10.3390/v8070202
virus allows in vivo reconstitution of recombi- 33. Billecocq A, Gauliard N, Le May N, Elliott
nant nucleocapsids. J Gen Virol 84 RM, Flick R, Bouloy M (2008) RNA
Reverse Genetics System for Rift Valley Fever Virus 113
polymerase I-mediated expression of viral RNA syncytial virus (BRSV) from cDNA: BRSV
for the rescue of infectious virulent and aviru- NS2 is not essential for virus replication in
lent Rift Valley fever viruses. Virology tissue culture, and the human RSV leader
378(2):377–384. https://doi.org/10.1016/j. region acts as a functional BRSV genome pro-
virol.2008.05.033 moter. J Virol 73(1):251–259. https://doi.
34. Habjan M, Penski N, Spiegel M, Weber F org/10.1128/JVI.73.1.251-259.1999
(2008) T7 RNA polymerase-dependent and 39. Terasaki K, Kalveram B, Johnson KN,
-independent systems for cDNA-based rescue Juelich T, Smith JK, Zhang L, Freiberg AN,
of Rift Valley fever virus. J Gen Virol 89 Makino S (2021) Rift Valley fever virus 78kDa
(Pt 9):2157–2166. https://doi.org/10. envelope protein attenuates virus replication in
1099/vir.0.2008/002097-0 macrophage-derived cell lines and viral viru-
35. Ikegami T, Won S, Peters CJ, Makino S (2006) lence in mice. PLoS Negl Trop Dis 15(9):
Rescue of infectious Rift Valley fever virus e0009785. https://doi.org/10.1371/journal.
entirely from cDNA, analysis of virus lacking pntd.0009785
the NSs gene, and expression of a foreign gene. 40. Ikegami T, Peters CJ, Makino S (2005) Rift
J Virol 80(6):2933–2940. https://doi.org/10. Valley fever virus nonstructural protein NSs
1128/Jvi.80.6.2933-2940.2006 promotes viral RNA replication and transcrip-
36. Caplen H, Peters CJ, Bishop DH (1985) tion in a minigenome system. J Virol
Mutagen-directed attenuation of Rift Valley 79(9):5606–5615. https://doi.org/10.1128/
fever virus as a method for vaccine develop- JVI.79.9.5606-5615.2005
ment. J Gen Virol 66(Pt 10):2271–2277. 41. Ito N, Takayama-Ito M, Yamada K,
https://doi.org/10.1099/0022-1317-66- Hosokawa J, Sugiyama M, Minamoto N
10-2271 (2003) Improved recovery of rabies virus
37. Ikegami T, Hill TE, Smith JK, Zhang L, Juelich from cloned cDNA using a vaccinia virus-free
TL, Gong B, Slack OA, Ly HJ, reverse genetics system. Microbiol Immunol
Lokugamage N, Freiberg AN (2015) Rift Val- 47(8):613–617. https://doi.org/10.1111/j.
ley fever virus MP-12 vaccine is fully attenuated 1348-0421.2003.tb03424.x
by a combination of partial attenuations in 42. Pathak KB, Nagy PD (2009) Defective
the S, M, and L segments. J Virol interfering RNAs: foes of viruses and friends
89(14):7262–7276. https://doi.org/10. of virologists. Viruses 1(3):895–919. https://
1128/JVI.00135-15 doi.org/10.3390/v1030895
38. Buchholz UJ, Finke S, Conzelmann KK
(1999) Generation of bovine respiratory
Chapter 8
Abstract
Several mammarenaviruses cause hemorrhagic fever (HF) disease in humans and pose a significant public
health problem in their endemic regions. The Old World (OW) mammarenavirus Lassa virus (LASV) is
estimated to infect several hundred thousand people yearly in West Africa, resulting in high numbers of
Lassa fever (LF) cases, a disease associated with high morbidity and mortality. No licensed vaccines are
available to combat LASV infection, and anti-LASV drug therapy is limited to the off-label use of ribavirin
whose efficacy remains controversial. The development of reverse genetics approaches has provided inves-
tigators with a powerful approach for the investigation of the molecular, cell biology and pathogenesis of
mammarenaviruses. The use of cell-based minigenome systems has allowed examining the cis- and trans-
acting factors involved in viral genome replication and gene transcription, assembly, and budding, which has
facilitated the identification of several anti-mammarenavirus candidate drugs. Likewise, it is possible now to
rescue infectious recombinant mammarenaviruses from cloned cDNAs containing predetermined muta-
tions in their genomes to investigate virus–host interactions and mechanisms of viral pathogenesis. Reverse
genetics have also allowed the generation of mammarenaviruses expressing foreign genes to facilitate virus
detection, to identify antiviral drugs, and to generate live-attenuated vaccine (LAV) candidates. Likewise,
reverse genetics techniques have allowed the generation of single-cycle infectious, reporter-expressing
mammarenaviruses to study some aspects of the biology of HF-causing human mammarenavirus without
the need of high security biocontainment laboratories. In this chapter, we describe the experimental
procedures to generate recombinant (r)LASV using state-of-the-art plasmid-based reverse genetics.
Key words Lassa virus, Recombinant Lassa virus, Lassa virus reverse genetics, Lassa virus rescue
systems
1 Introduction
Daniel R. Pérez (ed.), Reverse Genetics of RNA Viruses: Methods and Protocols, Methods in Molecular Biology, vol. 2733,
https://doi.org/10.1007/978-1-0716-3533-9_8,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2024
115
116 Luis Martı́nez-Sobrido et al.
A) B)
L vRNA
IGR
L
5’ Z 3’
UTR UTR
S vRNA
IGR
5’ GPC
NP 3’ GP1
UTR UTR L NP Z
GP2
Fig. 1 LASV genome replication and virion structure. (a) Genome organization: LASV has a bi-segmented
negative-sense single-stranded RNA genome. Each of the two viral (v)RNA genome segments uses an
ambisense coding strategy to direct the synthesis of two viral proteins in opposite orientation. The large (L,
top) vRNA segment (7.2 kb) encodes the viral L and Z proteins. The small (S, bottom) vRNA segment (3.5 kb)
encodes the viral NP and GPC. IGR intergenic region. UTR untranslated regions. (b) Virion structure: LASV are
enveloped viruses whose virion surface is decorated by the matured GP complex constituted of heterotrimers
of SSP/GP1/GP2. GP1 binds to cell surface receptor used by the virus and GP2 mediates the pH-dependent
fusion event at the late endosome resulting in release of the vRNP into the cell cytoplasm where it directs the
biosynthetic processes of replication and gene transcription of the viral genome. Underneath the viral
membrane is a layer made of the Z protein (orange). The core of the virus is made of two viral ribonucleopro-
tein (vRNP) complexes, composed of the viral genome segments (black lines) encapsidated by the viral NP
(red). NP-Z interaction mediates incorporation of the vRNPs into nascent LASV virions. Associated with the two
vRNP complexes is the viral RNA-dependent RNA polymerase L protein (green) that, together with NP, is the
minimal component required for LASV genome replication and gene transcription
protein (Fig. 1a, green) [12], and the matrix Z protein (Fig. 1a,
orange) [13], which is the major driving force of mammarenavirus
budding [13]. The small (S, bottom) segment encodes the viral
glycoprotein precursor (GPC) (Fig. 1a, blue) that is proteolytically
cleaved by signal peptidases and the proprotein convertase (PC)
subtilisin kexin isozyme-1/site-1 protease (SKI-1/S1P) to yield a
stable signal peptide (SSP), the N-terminal GP1, and the
membrane-anchored GP2. The mature mammarenavirus GP com-
plex, made of SSP/GP1/GP2 heterotrimers, forms the spikes that
decorate the virion surface and mediate virus cell entry (Fig. 1b).
GP1 binds to cellular receptors, whereas the transmembrane GP2
mediates viral fusion [1]. Segment S also encodes the viral nucleo-
protein (NP) (Fig. 1a, red), which is responsible of encapsidating
the viral RNA (Fig. 1b), and, together with the L polymerase
protein and the viral RNA, forms the viral ribonucleoproteins
(vRNPs) that are the minimal trans-acting factors required for
mammarenavirus genome replication and gene transcription
[12, 14] (Fig. 2). In addition, mammarenavirus NP mediates the
incorporation of the vRNPs into mature infectious virions by inter-
acting with Z [15]. Furthermore, NP has also been shown to
counteract the host type-I interferon (IFN-I) [16–21] and
118 Luis Martı́nez-Sobrido et al.
Genomic RNA
IGR
UTR UTR
5’ 3’
NP/L
GP/Z
TRANSCRIPTION
REPLICATION
TRANSLATION
NP/L NP L
Antigenomic RNA
3’ 5’
GP/Z
NP/L
TRANSCRIPTION IGR
GP1
TRANSLATION
GP/Z Z
GP2
2 Materials
2.1 LASV Reverse We have developed an RNA mPol-I /Pol-II reverse genetics system
Genetics Plasmids for the rescue of the highly pathogenic Josiah strain of LASV
lineage IV [43]. Plasmids used for the generation of rLASV
(Fig. 3) can be transformed and amplified in DH5α competent
bacteria (Invitrogen) using Luria broth (LB) media in the presence
of 100 μg/mL of ampicillin (Fisher Scientific) at 37 °C for 16–18 h
(h), with the exception of the mPol-I L plasmid (see Notes 1 and 2)
[43]. Plasmids can be purified using commercially available max-
iprep kits (e.g., Qiagen) following manufacturer’s recommenda-
tions and stored at -20 °C until being used (see Notes 3 and 4).
1. pCAGGS protein expression plasmids: The pCAGGS plasmids
use the polymerase II (Pol-II)-driven chicken β-actin promoter
and the rabbit β-globin polyadenylation (pA) signal sequence
to direct the synthesis of LASV NP and L, which are the
minimum viral components for LASV genome replication and
gene transcription (Fig. 3a) required for the generation of
rLASV (Fig. 4) [43] (see Note 5).
2. mPol-I vRNA expression plasmids: LASV reverse genetics use
the species-specific [34] mouse RNA polymerase I (mPol-I)
promoter to direct the synthesis of S and L genome or anti-
genome RNA species [43] and the mPol-I terminator to gen-
erate authentic LASV vRNA 3′ ends [43] (Fig. 3b) (see Notes
2 and 6). For the generation of rLASV (Fig. 4), two mPol-I
plasmids expressing the S (mPol-I S) and L (mPol-I L) geno-
mic or antigenomic vRNAs are used [43] (Figs. 3b and 4) (see
Notes 2 and 6).
LASV Reverse Genetics 121
A) GP B) pCAGGS L L
mPol-I S NP
mPol-I L L
Z pCAGGS NP NP
Fig. 3 Schematic representation of the LASV reverse genetics plasmids. (a) vRNA expression plasmids: vRNA
expression plasmids under the control of the mouse polymerase I (mPol-I) promoter (gray arrow) and
terminator (gray box) sequences direct the synthesis of LASV vRNA S (top) and L (bottom) segments. (b)
Protein expression plasmids: Protein expression pCAGGS plasmids use the chicken β-actin promoter (black
arrow) and the rabbit b-globin polyadenylation (pA) signal (black box) sequences to direct the synthesis of
LASV L (pCAGGS L, top) and NP (pCAGGS NP, bottom). These plasmids are required to provide the minimal
proteins required to initiate LASV gene transcription and genome replication. Both vRNA mPol-I and protein
pCAGGS expression plasmids are ampicillin resistant (Ampr) and have the origin of replication of Simian virus
40 (SV40)
2.2 Cell Lines for the The reverse genetics system we used for the rescue of infectious
Generation of rLASV rLASV is based on the use of the mPol-I promoter to direct
intracellular synthesis of the S and L genome or antigenome RNA
species (Figs. 3 and 4) [43] (see Note 6). The activity of the mPol-I
promoter exhibits species specificity and therefore is restricted to
rodent cells [34]. We use baby hamster kidney (BHK-21) cells since
they are easy to maintain, have high transfection efficiencies, and are
able to produce high viral titers [34, 35, 38, 39]. BHK-21 cells are
available from the American Type Culture Collection (ATCC).
Vero (African green monkey kidney epithelial) cells (LASV amplifi-
cation and titration) are available from the ATCC. Vero cells are
used to amplify and titrate rLASV from cell culture supernatants
collected from transfected BHK-21 cells [43]. Both cell lines are
maintained in T75 flasks at 37 °C in a 5% CO2 atmosphere.
2.3 Cell Culture 1. Cell culture medium: DMEM supplemented with 10% fetal
Media and Reagents bovine serum (FBS) and 1% penicillin/streptomycin (PS).
This cell culture media is used for maintenance of both BHK-
21 and Vero cells. Mix 445 mL DMEM (Invitrogen), 50 mL of
FBS (Atlanta Biologics), and 5 mL of 100X PS (Invitrogen).
Store at 4 °C.
2. Opti-MEM I medium: Opti-MEM I is used in the transfection
protocol to rescue rLASV.
122 Luis Martı́nez-Sobrido et al.
A) NP L
Day 1
plasmid
pCAGGS NP pCAGGS L transfecon
L
Z GP
NP
Day 7
virus
B) detecon
Mock rLASV
α-NP
Fig. 4 Generation of rLASV: BHK-21 cells are transiently co-transfected, using LPF2000, with the pCAGGS
protein expression plasmids encoding LASV NP and polymerase L (required to initiate viral gene transcription
and genome replication) together with the mPol-I vRNA expression plasmids encoding the viral L and S
segments (required to provide LASV vRNAs to initiate viral gene transcription and genome replication) (a). At
72 h post-transfection, BHK-21 cells are trypsinized and scaled-up into T75 flasks (a). After an additional
72–96-h incubation period, cell culture supernatants are collected and presence of LASV is determined by
immunofluorescence using LASV-specific antibodies (b). The chicken β-actin promoter (black arrow) and the
rabbit β-globin polyadenylation (pA) signal are indicated in the pCAGGS protein expression plasmids. The
mPol-I promoter and terminator sequences in the mPol-I vRNA expression plasmids are indicated by gray
arrows and boxes, respectively. Viral untranslated (UTR, black boxes) and intergenic (IGR) regions in the mPol-I
vRNA expression plasmids are indicated
3 Methods
Gently tap the cell culture T75 flask to confirm all the BHK-21
cells have been detached. After BHK-21 cells are completely
detached from the T75 flasks, carefully resuspend the cells in
9 mL of DMEM 10% FBS, 1% PS. Place the total media/cell
mixture in a 15 mL centrifuge tube and centrifuge the cells for
5 min at 1000 × g. Remove the media and resuspend the
BHK-21 cells in 10 mL of fresh DMEM 10% FBS, 1%
PS. Count the cells using a hemocytometer or cell counter
and adjust the cell concentration to approximately
1.0–1.2 × 106 cells/mL.
5. LPF2000/DNA incubation with BHK-21 cells: After approxi-
mately 20–30 min of incubation, pipette into each LPF2000/
DNA tube (step 3) 1 mL containing approximately
1.0–1.2 × 106 cells. Incubate the LPF2000/DNA/BHK-21
cell mixture for approximately 5 min at room temperature.
6. LPF2000/DNA/BHK-21 cell plating: Transfer the
LPF2000/DNA/BHK-21 cell mixture (step 5) into individual
wells of a 6-well cell culture plate. Gently tap the plate to
distribute the cells uniformly and incubate the cells in a 5%
CO2 humidified 37 °C incubator for approximately 6–12 h.
7. Transfection media change: After approximately 6–12 h of
incubation, replace the cell culture supernatant with 2 mL of
infection media and return the transfected BHK-21 cells to the
5% CO2 humidified 37 °C incubator and incubate for an addi-
tional 48–72 h [43].
8. Cell passage: After 48–72 h of incubation, transfected BHK-21
cells should reach approximately 100% confluence. Remove the
cell culture supernatant, wash the cells twice with 1X PBS, and
trypsinize them by adding 500 μL of trypsin-EDTA/well.
Return the 6-well plates to the 5% CO2 humidified 37 °C
incubator and let them incubate for approximately 5 min.
Gently tap the plates to complete BHK-21 cell detachment
from the plate. Carefully resuspend the BHK-21 cells with
1 mL of DMEM 10% FBS, 1% PS, and transfer to a 1.5 mL
microcentrifuge tube. Centrifuge the cells for 5 min at
5000 × g, at 4 °C in a microcentrifuge. Remove the cell culture
supernatant and resuspend the BHK-21 cells in 1 mL of infec-
tion media and transfer to a T75 cell culture flask. Bring up the
volume in the cell culture T75 flask to 10 mL with infection
media. Gently shake the T75 flask to allow uniform distribu-
tion of the BHK-21 cells and incubate the cells at 37 °C, 5%
CO2, for an additional 72–96 h (see Note 11).
9. LASV recovery from cell culture supernatants: After 72–96 h of
incubation, collect the cell culture supernatants from the T75
cell culture flask (step 8) into a 15 mL centrifuge tube.
LASV Reverse Genetics 125
4 Notes
Acknowledgments
References
1. Buchmeier MJ, Peter CJ, de la Torre JC (2007) challenge of antiviral therapy. Adv Virus Res
Arenaviridae: the viruses and their replication, 58:125–155
vol 2. Lippincott William and Wilkins, 12. Lee KJ, Novella IS, Teng MN, Oldstone MB,
Philadelphia de La Torre JC (2000) NP and L proteins of
2. Enria DA, Briggiler AM, Sanchez Z (2008) lymphocytic choriomeningitis virus (LCMV)
Treatment of Argentine hemorrhagic fever. are sufficient for efficient transcription and rep-
Antivir Res 78:132–139 lication of LCMV genomic RNA analogs. J
3. Barton LL, Mets MB, Beauchamp CL (2002) Virol 74:3470–3477
Lymphocytic choriomeningitis virus: emerging 13. Perez M, Craven RC, de la Torre JC (2003)
fetal teratogen. Am J Obstet Gynecol 187: The small RING finger protein Z drives arena-
1715–1716 virus budding: implications for antiviral strate-
4. Fischer SA, Graham MB, Kuehnert MJ, Kotton gies. Proc Natl Acad Sci U S A 100:12978–
CN, Srinivasan A, Marty FM, Comer JA, 12983
Guarner J, Paddock CD, DeMeo DL, Shieh 14. Ortiz-Riano E, Cheng BY, de la Torre JC,
WJ, Erickson BR, Bandy U, DeMaria A Jr, Martinez-Sobrido L (2012) Self-association of
Davis JP, Delmonico FL, Pavlin B, Likos A, lymphocytic choriomeningitis virus nucleopro-
Vincent MJ, Sealy TK, Goldsmith CS, Jernigan tein is mediated by its N-terminal region and is
DB, Rollin PE, Packard MM, Patel M, not required for its anti-interferon function. J
Rowland C, Helfand RF, Nichol ST, Fishman Virol 86:3307–3317
JA, Ksiazek T, Zaki SR (2006) Transmission of 15. Ortiz-Riano E, Cheng BY, de la Torre JC,
lymphocytic choriomeningitis virus by organ Martinez-Sobrido L (2011) The C-terminal
transplantation. N Engl J Med 354:2235– region of lymphocytic choriomeningitis virus
2249 nucleoprotein contains distinct and segregable
5. Borio L, Inglesby T, Peters CJ, Schmaljohn functional domains involved in NP-Z interac-
AL, Hughes JM, Jahrling PB, Ksiazek T, John- tion and counteraction of the type I interferon
son KM, Meyerhoff A, O’Toole T, Ascher MS, response. J Virol 85:13038–13048
Bartlett J, Breman JG, Eitzen EM Jr, 16. Pythoud C, Rodrigo WW, Pasqual G,
Hamburg M, Hauer J, Henderson DA, John- Rothenberger S, Martinez-Sobrido L, de la
son RT, Kwik G, Layton M, Lillibridge S, Torre JC, Kunz S (2012) Arenavirus nucleo-
Nabel GJ, Osterholm MT, Perl TM, protein targets interferon regulatory factor-
Russell P, Tonat K (2002) Hemorrhagic fever activating kinase IKKepsilon. J Virol 86:
viruses as biological weapons: medical and pub- 7728–7738
lic health management. JAMA 287:2391– 17. Martinez-Sobrido L, Emonet S, Giannakas P,
2405 Cubitt B, Garcia-Sastre A, de la Torre JC
6. Birmingham K, Kenyon G (2001) Lassa fever is (2009) Identification of amino acid residues
unheralded problem in West Africa. Nat Med critical for the anti-interferon activity of the
7:878 nucleoprotein of the prototypic arenavirus lym-
7. Gunther S, Lenz O (2004) Lassa virus. Crit phocytic choriomeningitis virus. J Virol 83:
Rev Clin Lab Sci 41:339–390 11330–11340
8. Freedman DO, Woodall J (1999) Emerging 18. Martinez-Sobrido L, Giannakas P, Cubitt B,
infectious diseases and risk to the traveler. Garcia-Sastre A, de la Torre JC (2007) Differ-
Med Clin North Am 83:865–883 ential inhibition of type I interferon induction
9. Richmond JK, Baglole DJ (2003) Lassa fever: by arenavirus nucleoproteins. J Virol 81:
epidemiology, clinical features, and social con- 12696–12703
sequences. BMJ 327:1271–1275 19. Martinez-Sobrido L, Zuniga EI, Rosario D,
10. Briese T, Paweska JT, McMullan LK, Hutch- Garcia-Sastre A, de la Torre JC (2006) Inhibi-
ison SK, Street C, Palacios G, Khristova ML, tion of the type I interferon response by the
Weyer J, Swanepoel R, Egholm M, Nichol ST, nucleoprotein of the prototypic arenavirus lym-
Lipkin WI (2009) Genetic detection and char- phocytic choriomeningitis virus. J Virol 80:
acterization of Lujo virus, a new hemorrhagic 9192–9199
fever-associated arenavirus from southern 20. Borrow P, Martinez-Sobrido L, de la Torre JC
Africa. PLoS Pathog 5:e1000455 (2010) Inhibition of the type I interferon anti-
11. Damonte EB, Coto CE (2002) Treatment of viral response during arenavirus infection.
arenavirus infections: from basic studies to the Viruses 2:2443–2480
130 Luis Martı́nez-Sobrido et al.
potent CD8+ T cell immunity. Nat Med 16: highly susceptible to lethal infection with
339–345 Lassa virus. J Virol 87:10908–10911
43. Yun NE, Seregin AV, Walker DH, Popov VL, 44. Albarino CG, Bird BH, Chakrabarti AK, Dodd
Walker AG, Smith JN, Miller M, de la Torre JC, KA, Erickson BR, Nichol ST (2011) Efficient
Smith JK, Borisevich V, Fair JN, Wauquier N, rescue of recombinant Lassa virus reveals the
Grant DS, Bockarie B, Bente D, Paessler S influence of S segment noncoding regions on
(2013) Mice lacking functional STAT1 are virus replication and virulence. J Virol 85:
4020–4024
Chapter 9
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a new member of the Coronaviridae
family responsible for the coronavirus disease 19 (COVID-19) pandemic. To date, SARS-CoV-2 has been
accountable for over 624 million infection cases and more than 6.5 million human deaths. The develop-
ment and implementation of SARS-CoV-2 reverse genetics approaches have allowed researchers to geneti-
cally engineer infectious recombinant (r)SARS-CoV-2 to answer important questions in the biology of
SARS-CoV-2 infection. Reverse genetics techniques have also facilitated the generation of rSARS-CoV-
2 expressing reporter genes to expedite the identification of compounds with antiviral activity in vivo and
in vitro. Likewise, reverse genetics has been used to generate attenuated forms of the virus for their
potential implementation as live-attenuated vaccines (LAV) for the prevention of SARS-CoV-2 infection.
Here we describe the experimental procedures for the generation of rSARS-CoV-2 using a well-established
and robust bacterial artificial chromosome (BAC)-based reverse genetics system. The protocol allows to
produce wild-type and mutant rSARS-CoV-2 that can be used to understand the contribution of viral
proteins and/or amino acid residues in viral replication and transcription, pathogenesis and transmission,
and interaction with cellular host factors.
Key words Coronavirus, SARS-CoV-2, COVID-19, Bacterial artificial chromosome, Reverse genet-
ics, Recombinant virus, Infectious clone
1 Introduction
Daniel R. Pérez (ed.), Reverse Genetics of RNA Viruses: Methods and Protocols, Methods in Molecular Biology, vol. 2733,
https://doi.org/10.1007/978-1-0716-3533-9_9,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2024
133
134 Kevin Chiem et al.
A)
NCR NCR
Ribosomal
5’ SAR2 nsp2 nsp3 nsp4 SAR3 6 7 8 9 10 11 S 3a E M 6 7a 7b 8 N 10 3’
frameshift
ORF1a nsp Endo-
nsp12 Helicase SAR4 SAR5
RNAase
ORF1b
B)
Gene Locus Protein Product Start End Proposed functions
5’ UTR 5’ UTR None 1 177 N/A
ORF1a SAR2 Leader protein 178 717 Induce host mRNA cleavage
ORF1a nsp2 nsp2 718 2,631 Binds to Prohibitin-1/2
ORF1a nsp3 nsp3 2,632 8,466 Release NSPs 1, 2, 3 (Papain like proteinase)
ORF1a nsp4 nsp4 8,467 9,966 Membrane rearrangement
ORF1a SAR3 3C-like proteinase 9,967 10,884 Cleaves at 11 sites of NSP polyprotein (3C-like proteinase)
ORF1a nsp6 nsp6 10,885 11,754 Generates autophagosomes
ORF1a nsp7 nsp7 11,755 12,003 Dimerizes with NSP8
ORF1a nsp8 nsp8 12,004 12,597 Stimulates NSP12
ORF1a nsp9 nsp9 12,598 12,936 Binds to helicase
ORF1a nsp10 nsp10 12,937 13,353 Stimulates NSP16
ORF1b nsp11 nsp11 13,354 13,392 Unknown
ORF1b nsp12 RdRp 13,352 16,148 Copies viral RNA methylation (RNA polymerase)
ORF1b helicase helicase 16,149 17,951 Unwinds duplex RNA (Helicase)
5ʹ-cap RNA (3ʹ to 5ʹ exonuclease, guanine N7-
ORF1b SAR4 3’-5’ exonuclease 17,952 19,532
methyltransferase)
Degrade RNA to evade host defense
ORF1b endoRNAse endoribonuclease 19,533 20,570
(endoRNAse/endoribonuclease)
2’-O-ribose 5ʹ-cap RNA methylation (2ʹ-O-ribose-methyltransferase—
ORF1b SAR5 20,571 21,464
methyltransferase potential antiviral drug target)
S S Spike (S) 21,475 25,296 Glycoprotein
ORF3a 3a 3a 25,305 26,132 Not fully defined, has Ion channel activity
E E Envelope (E) 26,157 26,384 Envelope protein
M M Membrane (M) 26,435 27,103 Membrane protein
ORF6 6 6 27,114 27,299 Interferon antagonist
ORF7a 7a 7a 27,306 27,671 Immunomodulator and interferon antagonist
ORF7b 7b 7b 27,668 27,799 Apoptosis activation
ORF8 8 8 27,806 28,171 Immunomodulator, down-regulator of MHC-1
N N Nucleocapsid (N) 28,186 29,445 Nucleocapsid protein
ORF10 10 10 29,470 29,586 Interferon antagonist
3’ UTR 3’ UTR None 29,587 29,930 N/A
C)
Spike
(S)
Membrane
(M)
Envelope
(E)
Viral RNA
genome
Nucleocapsid
(N)
Fig. 1 SARS-CoV-2 genomic organization and virion structure. (a) Genome organization. At the end of the
SARS-CoV-2 genome ( 29,930 nucleotides) are located the 5′ and 3′ non-coding regions (NCR). The 5′ end
136 Kevin Chiem et al.
Fig. 1 (continued) of the genome contains two overlapping ORFs (1a and 1b), which are translated in two
co-amino terminal polyproteins named pp1a and pp1ab. Both polyproteins are cleaved into 16 mature nsps:
leader protein (SAR2), nsp2, nsp4, nsp4, 3C-like proteinase (SAR3), nsp6, nsp7, nsp8, nsp9, nsp10, nsp11,
RNA-dependent RNA polymerase (RdRp, nsp12), helicase, 3′-to-5′ exonuclease (SAR4), endoRNase, and
2′-O-ribose methyltransferase (SAR5). The structural genes encoding the viral S (green), E (orange), M (red),
and N (blue) structural proteins and the genes encoding the accessory proteins 3a, 6, 7a, 7b, 8, and 10, are
located downstream the ORF1b. (b) SARS-CoV-2 genes, proteins, and their functions: SARS-CoV-2 genes and
their respective encoded proteins, including their location in the viral genome and functions, are indicated. (c)
Schematic representation of SARS-CoV-2 virion: SARS-CoV-2 virion is decorated by the spike (S, green),
membrane protein (M, red), and envelope (E, orange) proteins. Inside the virion is the viral nucleocapsid (N,
blue) protein that encapsulates the positive sense, single-stranded RNA viral genome
BAC-Based SARS-CoV-2 Reverse Genetics 137
2 Materials
2.2 SARS-CoV-2 SARS-CoV-2 WA-1 strain (NR-52281) was obtained from the
Biodefense and Emerging Infections Research Resources Reposi-
tory (BEI Resources), a National Institute of Allergy and Infectious
Diseases repository managed by the American Type Culture Col-
lection (ATCC). We selected SARS-CoV-2 WA-1 strain to develop
the reverse genetics approaches based on the use of the plasmid
pBeloBAC11 since it was isolated from a patient with respiratory
illness in Seattle, Washington, in 2020 and because the complete
sequence was available (No. MN985325) at the National Center
for Biotechnology Information (NCBI).
2.3 BAC and Plasmid The plasmid pBeloBAC11 (NEB) used for the generation of the
Extraction SARS-CoV-2 BAC-based reverse genetics has been previously
described [26, 53, 68, 72–74] and used to generate the pBelo-
BAC11-SARS-CoV-2 [30, 48, 66, 67]. The Qiagen Large-
Construct Kit was used for the preparation of ultrapure BACs (see
Note 1).
2.4 E. coli DH10B E. coli DH10B electrocompetent cells (Thermo Fisher Scientific)
Cells are used for efficient propagation of SARS-CoV-2 BAC (see Note
2). E. coli DH10B electrocompetent cells are stored at -80 °C to
avoid thaw-freeze cycles until use.
138 Kevin Chiem et al.
2.5 Culture Media for Reagents in this section could be purchased from different
DH10B Cells suppliers.
1. Luria Broth (LB) media: To prepare 1 L of LB media, mix 10 g
of Bacto Tryptone (BD), 5 g Bacto yeast extract (BD), and 10 g
NaCl (T Baker) in 900 mL of ddH2O. Adjust the volume to
1 L with ddH2O. Autoclave to sterilize and store at 4 °C.
2. Chloramphenicol (Thermo Fisher Scientific): Prepare a stock
of chloramphenicol at 12.5 mg/mL in 100% ethanol. Aliquot
and store at - 20 °C.
3. LB media with chloramphenicol: Dilute the working stock of
chloramphenicol to a final concentration of 12.5 μg/mL (1:
1000) in LB. LB media with chloramphenicol can be stored at
4 °C for up to 30 days.
4. LB agar plates with chloramphenicol: Prepare 1 L of LB media,
and add 15 g of Bacto agar (BD). Autoclave to sterilize, equili-
brate at 55 °C for 30 min, and add chloramphenicol to a final
concentration of 12.5 μg/mL. Dispense 25 mL of media to
petri dishes, and store at 4 °C.
5. SOC media: To prepare 1 L of SOC media, mix 20 g of Bacto
Tryptone (BD), 5 g Bacto yeast extract (BD), 0.5 g NaCl
(T Baker), and 2.5 mL of 1 M KCl in 900 mL of ddH2O,
and adjust the volume of the solution to 1 L with
ddH2O. Sterilize by autoclaving, and before use, add 10 mL
of 1 M MgCl2, 10 mL of 1 M MgSO4, and 20 mL of 1 M
glucose. SOC media can be stored at 4 °C.
2.6 Cell Line for the Vero E6 (African green monkey kidney epithelial cells) are available
Rescue of rSARS-CoV- from the ATCC (CRL-1586). Vero E6 cells are used in all experi-
2 ments outlined in the protocol to generate and characterize rSARS-
CoV-2 because of their acceptable transfection efficiency and their
ability to efficiently support SARS-CoV-2 replication. Vero E6 cells
are cultured and maintained in growth medium (described below in
Subheading 2.7) at 37 °C in a 5% CO2 atmosphere cell culture
humidified incubator (see Notes 3 and 4).
2.7 Cell Culture Reagents in this section could be purchased from other suppliers.
Media and Reagents
1. Cell growth medium: Dulbecco’s Modified Eagle’s Medium
for Rescue and
(DMEM, Gibco) supplemented with 5–10% fetal bovine serum
Propagation of rSARS- (FBS, Avantor Seradigm), 100 mg/mL penicillin/streptomy-
CoV-2 cin, and 2 mM L-glutamine (PSG, Sigma-Aldrich). Store at 4 °
C.
2. Virus growth medium: DMEM supplemented with 2% FBS,
100 mg/mL PSG (Sigma-Aldrich). Store at 4 °C.
3. Trypsin-EDTA solution (Sigma-Aldrich): 0.25% (w/v) trypsin,
0.02% (w/v) EDTA.
BAC-Based SARS-CoV-2 Reverse Genetics 139
2.8 Reagents for Reagents described in this section could be purchased for other
Plaque Assay and suppliers.
Immunostaining
1. Fixing solution: 10% (v/v) neutral buffered formalin solution
(Sigma-Aldrich) in PBS.
2. Permeabilization solution: 0.5% (v/v) Triton X-100 (Sigma-
Aldrich) in PBS.
3. Blocking solution: 2.5% (w/v) bovine serum albumin (BSA,
Sigma-Aldrich) in PBS.
4. Primary mouse anti-SARS nucleocapsid (N) protein 1C7C7
cross-reactive monoclonal antibody (mAb, Millipore Sigma)
(see Notes 7 and 8).
5. Vectastain ABC Kit, peroxidase mouse IgG (Vector Labora-
tories Inc.).
6. DAB Substrate Kit, Peroxidase (HRP), with Nickel (Vector
Laboratory Inc.).
2.9 Reagents for Reagents described in this section could be purchased for other
Immunofluorescence suppliers.
1. Fixing solution: 10% (v/v) neutral buffered formalin solution
(Sigma-Aldrich) in PBS.
140 Kevin Chiem et al.
2.10 Laboratory Equipment, reagents, and supplies in this section could be pur-
Equipment, Reagents, chased for other suppliers different to those recommended.
and Supplies 1. Benchtop refrigerated microcentrifuge (Thermo Fisher
Scientific).
2. High-speed refrigerated centrifuge (Thermo Fisher Scientific).
3. Water bath (VWR).
4. Power supply (Bio-Rad).
5. NanoDrop (Thermo Fisher Scientific) or similar
spectrophotometer.
6. MicroPulser electroporator (Bio-Rad).
7. Electroporation cuvettes, 0.2 cm gap (Bio-Rad).
8. Laboratory 5% CO2 cell culture humidified microbiological
incubator at 37 °C for maintenance of cells, virus rescue, and
amplification (PHC Corporation).
9. Class II biosafety cabinet (Baker).
10. Evos light microscope (Thermo Fisher Scientific).
11. Evos fluorescent microscope (Thermo Fisher Scientific).
12. Microwave oven to melt 2% Oxoid agar (Panasonic).
13. -80 °C freezer (PHC Corporation).
14. Autoclave.
15. MaxQ Large Incubated Benchtop Orbital Shaker (Thermo
Fisher Scientific).
16. Polystyrene T75 cell culture flask (Corning).
17. Cell culture plates (Greiner Bio-One).
18. Microcentrifuge tubes, 1.5 mL (VWR).
19. Nalgene® System 100 Cryogenic Vials (Thermo Fisher
Scientific).
20. Polypropylene sterile conical 15 mL and 50 mL conical tubes
(Greiner Bio-One).
21. Serological 5 mL, 10 mL, and 25 mL pipettes (Greiner
Bio-One).
BAC-Based SARS-CoV-2 Reverse Genetics 141
22. Universal 20 μL, 200 μL, and 1000 μL filtered pipette tips
(VWR).
23. Bacteria orbital shaker (Thermo Fisher Scientific).
3 Methods
ori2 Cmr
repE
pBeloBAC11
sopC
SARS-CoV-2
sopB sopA
Fig. 2 Generation of the pBeloBAC11-SARS-CoV-2. The full-length SARS-CoV-2 genome is cloned into the
pBeloBAC11 plasmid as a cDNA between the cytomegalovirus (CMV) polymerase II-driven promoter (gray
arrow) at the 5’ end and the hepatitis delta ribozyme (Rz) and bovine growth hormone (bGH) polyadenylation
signals (gray boxes) at the 3’ end. The pBeloBAC11 plasmid can accommodate large cDNA fragments as 1–2
copies into E. coli. The pBeloBAC11 regulatory genes (sopA, sopB, sopC, and repE), the F-factor replication
origin (Ori2), and the chloramphenicol resistance gene (Cmr) are indicated. Viral proteins are those previously
described in Fig. 1
142 Kevin Chiem et al.
pBeloBAC11
SARS-CoV-2
Media
change
Fig. 3 Rescue of rSARS-CoV-2 from the BAC infectious clone. Vero E6 cells in six-well plates are transfected in
suspension with 4 μg of pBeloBAC11-SARS-CoV-2 and LPF2000 as described in the method section. Two
days after transfection, cells are collected and scaled up into T75 flasks. After 48 h post-transfection, when
the CPE is clear, the cell culture supernatants are collected, and the presence of the virus is confirmed by the
CPE, IFA, and plaque assay
A) B)
Mock SARS-CoV-2 rSARS-CoV-2 Mock SARS-CoV-2 rSARS-CoV-2
FITC
DAPI
Fig. 4 Characterization of rescued rSARS-CoV-2. At 48–72 h after scaling up transfected Vero E6 cells into T75
flask, cell culture supernatants are collected and used to infect fresh monolayers of Vero E6 cells to confirm
the presence of rSARS-CoV-2 by CPE, IFA, and plaque assay. (a) CPE: Representative images of Vero E6 cells
mock-infected or treated with the cell culture supernatant from transfected Vero E6 cells are shown. As
internal control, Vero E6 cells were also infected with SARS-CoV-2 natural isolate. Scale bars, 400 μm. (b) IFA:
Presence of rSARS-CoV-2 is also evaluated by infecting fresh Vero E6 cells with cell culture supernatants
collected at 48–72 h after scaling up transfected Vero E6 cells into T75 flask and assessing the presence of
SARS-CoV-2 using the mAb 1C7C7 against the viral N protein. Vero E6 cells mock-infected or infected with
SARS-CoV-2 are included as internal controls. DAPI is used to stain the nucleus. Scale bars, 100 μm. (c)
Plaque assay: Confluent monolayers of Vero E6 cells in six-well plates were infected with tenfold serial
dilutions of the cell culture supernatants collected at 48–72 h after scaling up transfected Vero E6 cells into
T75 flask. After 1 h adsorption, cells are overlaid with agar, and 72 h after infection, the presence of the virus
was evaluated with the mAb 1C7C7 against the viral N protein by immunostaining. Mock-infected and Vero E6
cells infected with SARS-CoV-2 were used as controls to validate the plaque assay
BAC-Based SARS-CoV-2 Reverse Genetics 145
13. Remove the blocking solution, and incubate the cells with
500 μL/well of a 1 μg/mL solution of the anti-SARS N
protein mAb 1C7C7 (1 μg/mL) in a 37 °C incubator for 1 h
(see Note 7).
14. Remove the primary antibody solution, and wash the cells
three times with PBS.
15. Add 500 μL/well of a 1:200 dilution of an anti-mouse IgG-
FITC-conjugated antibody and 1 mg/mL of DAPI in blocking
solution. Place the plates in a 37 °C incubator for 1 h.
16. Remove the secondary antibody/DAPI solution, and wash the
cells three times with PBS. Leave 1 mL of PBS in each well.
Samples can be stored at 4 °C protected from light with
aluminum foil.
17. Observe the samples under a fluorescence microscope. Detec-
tion of SARS-CoV-2 N protein is seen in green (FITC), while
the cell nucleus is seen in blue (DAPI). Only Vero E6 cells
exposed to cell culture supernatants containing rSARS-CoV-
2 or infected with the SARS-CoV-2 natural isolate should give
a positive FITC signal. Mock-infected cells should only be
stained with DAPI (Fig. 4B) (see Note 23).
3.4 Plaque Assay for To determine the titer of rSARS-CoV-2 from our rescue approach
Viral Titration and and to assess the plaque phenotype of the recombinant virus, we
Visualization of Plaque recommend conducting a plaque assay in Vero E6 cells (Fig. 4c).
Sizes Mock-infected cells and cells infected with the SARS-CoV-2 WA-1
natural isolate should be included as internal negative and positive
controls, respectively, in the plaque assay. An agar overlay is placed
on top of the Vero E6 cells infected with tenfold serial dilutions of
the rSARS-CoV-2 to restrict the spread of the virus to only neigh-
boring cells. Thus, each viral plaque is produced by a single virus.
1. Seed six-well plates of Vero E6 cells (0.5 × 106 cells/well) in
cell culture media the day before the plaque assay. Vero E6 cells
should be ~80–90% confluency (~1 × 106 cells/well) at the
time of viral infection. We recommend preparing enough
six-well plates to titrate each sample in triplicates.
2. Thaw an aliquot of the rSARS-CoV-2 and SARS-CoV-2 natural
isolate, and make tenfold serial dilutions in virus growth media.
3. Wash Vero E6 cells three times with virus growth media, and
infect each of the wells with 1 mL of the tenfold viral serial
dilutions. We recommend using 10-2–10-7 serial dilutions of
the virus stocks.
4. Incubate the plates at 37 °C in a 5% CO2 incubator for 1 h.
Gently rock the plates every 15 min.
BAC-Based SARS-CoV-2 Reverse Genetics 147
3.5 Amplification of 1. Seed Vero E6 cells in T75 flasks (~5 × 106 cells/T75) in cell
rSARS-CoV-2 and growth media to reach ~80–90% confluent the day before
Generation of Virus infection.
Stocks 2. Wash the cells twice with virus growth media, and infect them
with a MOI of ~0.01–0.001 PFU in 5 mL of virus growth
media.
3. After viral adsorption, remove the virus inoculum, and add
15 mL of virus growth media. Place the T75 flask in a 37 °C
incubator with 5% CO2, and incubate the cells until CPE is
observed.
4. When CPE is ~25% (~48–72 h post-infection), collect the cell
culture supernatant into a 15 mL tube, and centrifuge at
2000 × g for 10 min at 4 °C to remove cell debris.
5. Collect the cell culture supernatant containing the virus in a
new 15 mL tube, and discard the cell pellet.
6. Aliquot the cell culture supernatant in labeled cryotubes, and
store it at - 80 °C (see Notes 25 and 26).
7. Thaw one cryotube aliquot, and assess the viral titer by plaque
assay as described previously in Subheading 3.4 (see Note 27).
4 Notes
Acknowledgments
References
1. Andersen KG et al (2020) The proximal origin 8. Gudbjartsson DF et al (2020) Spread of SARS-
of SARS-CoV-2. Nat Med 26(4):450–452 CoV-2 in the Icelandic population. N Engl J
2. Lu R et al (2020) Genomic characterisation Med 382(24):2302–2315
and epidemiology of 2019 novel coronavirus: 9. Petersen E et al (2020) Comparing SARS-
implications for virus origins and receptor CoV-2 with SARS-CoV and influenza pan-
binding. Lancet 395(10224):565–574 demics. Lancet Infect Dise 20(9):e238–e244
3. Ksiazek TG et al (2003) A novel coronavirus 10. Sikkema RS et al (2019) Global status of Mid-
associated with severe acute respiratory syn- dle East respiratory syndrome coronavirus in
drome. N Engl J Med 348(20):1953–1966 dromedary camels: a systematic review – COR-
4. Peiris JS et al (2003) Coronavirus as a possible RIGENDUM. Epidemiol Infect 147:e198
cause of severe acute respiratory syndrome. 11. Sarkar M, Madabhavi I (2022) SARS-CoV-
Lancet 361(9366):1319–1325 2 variants of concern: a review. Monaldi Arch
5. van Boheemen S et al (2012) Genomic charac- Chest Dis 93(3):2337
terization of a newly discovered coronavirus 12. Ahmad A, Fawaz MAM, Aisha A (2022) A
associated with acute respiratory distress syn- comparative overview of SARS-CoV-2 and its
drome in humans. MBio 3(6):e00473 variants of concern. Infez Med 30(3):328–343
6. Zaki AM et al (2012) Isolation of a novel coro- 13. Ashoor D et al (2022) How concerning is a
navirus from a man with pneumonia in Saudi SARS-CoV-2 variant of concern? Computa-
Arabia. N Engl J Med 367(19):1814–1820 tional predictions and the variants labeling sys-
7. Alfaraj SH et al (2019) Clinical predictors of tem. Front Cell Infect Microbiol 12:868205
mortality of Middle East Respiratory Syn- 14. Bian L et al (2022) Research progress on vac-
drome Coronavirus (MERS-CoV) infection: a cine efficacy against SARS-CoV-2 variants of
cohort study. Travel Med Infect Dis 29:48–50 concern. Hum Vaccin Immunother 18(5):
2057161
152 Kevin Chiem et al.
Abstract
The infectious clone has been constructed for years via various mechanisms using reverse genetics of viral
RNA into cDNA. The mechanism of construction has evolved to DNA-launch plasmids which simplify
infectious clone manipulation and expression in mammalian cells. Infectious clones have enormously
allowed manipulation of the enterovirus genome to discover antivirals, viral replication mechanisms, and
functions of essential viral proteins. Here we will be discussing methods for the production of DNA-launch
human enterovirus infectious clones and viral genome engineered with a fluorescent reporter gene.
Key words Enterovirus, DNA-launch plasmids, Infectious clone, Reverse genetics, Fluorescence
reporter gene
1 Introduction
Daniel R. Pérez (ed.), Reverse Genetics of RNA Viruses: Methods and Protocols, Methods in Molecular Biology, vol. 2733,
https://doi.org/10.1007/978-1-0716-3533-9_10,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2024
155
156 Thinesshwary Yogarajah and Justin Jang Hann Chu
Fig. 1 Approach for the construction of EV-D68 complete genome DNA-launch infectious cDNA clone. Virus is
cultured in RD cells, and virus culture supernatant is used for RNA extraction. Viral RNA is reverse transcribed
into cDNA. Three overlapping fragments were produced via PCR labeled Fragment 1, Fragment 2, and
Fragment 4. Fragment 4 was produced via fusion PCR with additional hepatitis delta virus sequence encoding
for a self-cleaving ribozyme (to be henceforth referred to as “HDV”). Each fragment was cloned into high-copy
number plasmid vector sequentially via unique restriction sites. The complete genome clone is transfected
into permissive mammalian cells for EV-D68 replication to rescue clone-derived virus. The fluorescence
reporter gene is then inserted into complete genome clone for obtaining the EV-D68 fluorescence reporter
gene expression virus
2 Materials
2.3 PCR PCR reaction: Q5 Hot Start High-Fidelity DNA 2X Master Mix
Amplification (see Note 2). 10 pmol oligonucleotide primer, nuclease-free water.
2.4 Agarose Gel 1. 0.8% TAE-agarose gel: 0.8 g agarose, 100 mL 1× TAE buffer
Electrophoresis and consists of 1 mM EDTA, 40 mM tris acetate.
Gel Extraction 0.5% TAE-agarose gel: 0.5 g agarose, 100 mL 1× TAE
buffer consists of 1 mM EDTA, 40 mM tris acetate.
2. 100 bp DNA ladder and 1 kb plus DNA ladder.
3. Ethidium bromide (EtBr) for DNA visualization.
4. Agarose gel documentation system with trans-UV lamp and a
blue light transilluminator.
5. QIAquick Gel Extraction Kit (see Note 1).
3 Methods
3.1 EV-D68 Viral RNA 1. Propagate virus in suitable cell culture until the cytopathic
Extraction and Viral effect is observed. Following that harvest virus culture super-
cDNA Synthesis natant from EV-D68-infected cells (see Note 4). Viral RNA is
extracted using QIAamp Viral RNA Mini Kit according to the
manufacturer’s instructions. Viral RNA is eluted in 50 μL of
AVE buffer. Quantitate RNA concentration in ng/μL using
NanoDrop 2000.
2. A first strand cDNA is synthesized covering the complete
genome of EV-D68 with Maxima H minus Reverse Transcrip-
tase (see Note 1). Assemble a reverse transcription reaction with
a total volume of 20 μL in a 0.2 mL tube. Mix appropriate
volume of viral RNA (1 μg) and 3 μL (30 pmol) of the oligo-dT
anchored primers (see Note 5), and heat for 5 min at 25 °C.
Then keep on ice for 5 min. Then add 200 U Maxima H Minus
Reverse Transcriptase, 5× RT buffer, 40 U ribonuclease inhibi-
tor, 0.5 mM dNTPs, 10 mM DTT, and makeup balance vol-
ume with nuclease-free water. Incubate the mixture at 55 °C
for 30 min, followed by incubation at 85 °C for 5 min for the
inactivation of the enzyme (see Note 5). Store the cDNA
product at -20 °C until required.
3.2 Viral cDNA 1. Amplify cDNA fragments using EV-D68-specific primers into
Fragment three fragments with different lengths. Each primer set is used
Amplification with PCR in PCR with Q5 Hot Start High-Fidelity DNA 2X Master Mix.
For the amplification of Fragment 1 from cDNA, NheI, a
unique restriction site, is inserted upstream of the 5′UTR
viral sequence, and NotI, also a unique restriction site, is
inserted downstream of the 2C viral sequence to ease cloning
into pcDNA3.1(+) plasmid vector (Fig. 2).
160 Thinesshwary Yogarajah and Justin Jang Hann Chu
Fig. 4 The construction of EV-D68 complete genome DNA-launch infectious cDNA clone using three
fragments. Three overlapping fragments starting from Fragment 1 followed by Fragment 2 and then Fragment
4 were sequentially inserted via specific RE digestion into pcDNA3.1(+) plasmid vector
3.3 Viral cDNAs 1. All purified fragments and pcDNA3.1(+) plasmid vector are
Cloning into a Plasmid quantitated using NanoDrop 2000. Double digestion by two
Vector restriction endonuclease, PCR purification kit, and ligated and
transformed into plasmid sequentially (Fig. 4). For the assem-
bly of Fragment 1, restriction endonucleases Nhel and NotI are
used for double digestion. Restriction endonuclease digestion
was set up according to New England BioLabs manufacturer’s
protocol. Restriction endonucleases are used to generate phos-
phorylated ends of the purified cDNA fragments and plasmid
vector for subsequent sticky-end cloning (see Note 8).
164 Thinesshwary Yogarajah and Justin Jang Hann Chu
3.4 Plasmid 1. Grow HEK293T cells in DMEM supplemented with 10% FBS,
Transfection to and then seed cells into cell-culture treated 6-well tissue culture
Recover Infectious plate at a seeding density of 600,000 cells per well to achieve
Clone-Derived EV-D68 60–70% confluency at the time of transfection. Incubate plate
Virus for 18–24 h before transfection (see Note 14).
2. Replace DMEM supplemented with 10% FBS with 2.5 mL of
fresh DMEM supplemented with 2% FBS about 30 min prior
to transfection. Keep plate in 37 °C until use.
3. Transfect purified plasmid DNA into cells using jetPRIME
reagent. Dilute 5 μL jetPRIME reagent in 200 μL of jetPRIME
buffer, then add 2–3 μg of plasmid DNA, and mix gently.
Incubate mixture for 10 min at room temperature. Add com-
plex by dropwise method into cells, and mix side by side before
incubation at 33 °C incubator for 4–7 days (see Note 15). Then
freeze the transfected six-well tissue culture tray in a -80 °C
freezer.
3.5 Propagation and 1. Seed RD cells in a T75 flask at a seeding density of 12 million
Characterization of cells/flask or 80% confluency. Incubate the cells 18–24 h before
Clone-Derived EV-D68 infection with clone-derived EV-D68 (see Note 14).
2. Thaw the transfected six-well tissue culture tray mentioned in
Subheading 3.4, step 3 in a 37 °C incubator until completely
thawed. Harvest the supernatant from transfected wells into a
cryovial. Infect 1 mL of the virus culture supernatant from
cryovials into the seeded T75 flask, and incubate at 33 °C for
60 min. Following that remove unbound virus by washing with
warm 1× PBS. Add 10 mL of DMEM with 2% FBS, and
incubate at 33 °C with 5% CO2 until cytopathic effect (CPE)
is visible. Virus harvest can be carried out upon observation of
80–90% CPE. Unless there is no visible CPE, upon consider-
ation of cell health in flask, harvest virus after infection upon
4–6 days (see Note 16).
3. Phenotypically characterize the clone-derived EV-D68 virus by
comparing plaque morphology on RD cells by performing
plaque assay, and then compare growth kinetics to the parental
virus.
Construction of Infectious Clones for Human Enteroviruses 167
3.6 Introduction of a 1. To insert a fluorescence reporter gene, PCR is carried out using
Fluorescence Reporter a pEGFP plasmid containing EGFP gene. The insertion of
Gene into the Complete EGFP gene in between 5′UTR and VP4 gene is accommo-
Genome cDNA Clone dated by producing three fragments of PCR and stitching them
together using fusion PCR. Each primer set is used in PCR
with Q5 Hot Start High-Fidelity DNA 2X Master Mix. For the
amplification of Fragment 1 from pcDNA3.1(+) EV-D68, a
unique restriction site, NheI is inserted upstream of the 5′UTR
of viral sequence, and 15 base of EGFP sequence is inserted
in the reverse primer in set 1 to ease fusion PCR (Fig. 5).
Fragment 2 is amplified from pEGFP plasmid using a 15 base
of 5′UTR sequence addition in the forward primer and an
addition of a unique 2A cleavage sequence in the reverse primer
168 Thinesshwary Yogarajah and Justin Jang Hann Chu
Fig. 6 The construction and propagation of EGFP EV-D68 DNA-launch infectious cDNA clone. Fragment 5 was
inserted via specific RE digestion into pcDNA3.1(+) EV-D68 plasmid vector. Clone-derived vector was
transfected into HEK293T cells, and EGFP expression was viewed using Olympus IX81 fluorescence
microscope
4 Notes
References
10. Qin Z, Chen Y, Yu J, He X, Liu X, Zhu L, 14. Meister SW, Hendrikse NM, Löfblom J (2019)
Wu Q, Wan C, Zhang B, Zhao W (2021) Directed evolution of the 3C protease from
Rapid construction of an infectious clone of coxsackievirus using a novel fluorescence-
the Zika virus, strain ZKC2. Front assisted intracellular method. Biol Chem
Microbiol 12 400(3):405–415. https://doi.org/10.1515/
11. Chin W-X, Lee RCH, Kaur P, Lew TS, hsz-2018-0362
Yogarajah T, Kong HY, Teo Z-Y, Salim CK, 15. Schürer H, Lang K, Schuster J, Mörl M (2002)
Zhang R-R, Li X-F, Alonso S, Qin C-F, Chu A universal method to produce in vitro tran-
JJH (2021) A single-dose live attenuated chi- scripts with homogeneous 3′ ends. Nucleic
meric vaccine candidate against Zika virus. NPJ Acids Res 30(12):e56. https://doi.org/10.
Vaccines 6(1):20–20. https://doi.org/10. 1093/nar/gnf055
1038/s41541-021-00282-y 16. Walker SC, Avis JM, Conn GL (2003) General
12. Zimina A, Viktorova EG, Moghimi S, plasmids for producing RNA in vitro tran-
Nchoutmboube J, Belov GA (2021) Interac- scripts with homogeneous ends. Nucleic Acids
tion of poliovirus capsid proteins with the cel- Res 31(15):e82. https://doi.org/10.1093/
lular autophagy pathway. Viruses 13(8). nar/gng082
https://doi.org/10.3390/v13081587 17. Al-Allaf FA, Tolmachov OE, Zambetti LP,
13. van der Schaar HM, Melia CE, van Bruggen Tchetchelnitski V, Mehmet H (2013) Remark-
JA, Strating JR, van Geenen ME, Koster AJ, able stability of an instability-prone lentiviral
Bárcena M, van Kuppeveld FJ (2016) Illumi- vector plasmid in Escherichia coli Stbl3. 3 Bio-
nating the sites of enterovirus replication in tech 3(1):61–70. https://doi.org/10.1007/
living cells by using a split-GFP-tagged viral s13205-012-0070-8
protein. mSphere 1(4). https://doi.org/10.
1128/mSphere.00104-16
Chapter 11
Abstract
The reverse genetics system commonly used for the production of hepatitis C virus (HCV), which is a major
causative agent of liver diseases, involves introduction of the viral genomic RNA synthesized in vitro into
human hepatoma cells by electroporation. As an alternative methodology, we describe a cell culture system
based on transfection with an expression plasmid containing a full-length HCV cDNA clone flanked by
RNA polymerase I promoter and terminator sequences to generate infectious virus particles from trans-
fected cells.
Key words Plasmid-based reverse genetics, Hepatitis C virus, RNA polymerase I, Promoter, Termi-
nator, Infectious cDNA clone
1 Introduction
Daniel R. Pérez (ed.), Reverse Genetics of RNA Viruses: Methods and Protocols, Methods in Molecular Biology, vol. 2733,
https://doi.org/10.1007/978-1-0716-3533-9_11,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2024
175
176 Ryosuke Suzuki and Tetsuro Suzuki
2 Materials
2.1 HCV This protocol is described for the rescue of HCV. Work with HCV
should follow biosafety laboratory rules and regulations appropri-
ate for this type of work. All material used for the rescue of HCV
should be sterilized before disposal.
Reverse genetic manipulation of HCV infectious cDNA clones
can be successfully performed using HCV strain that can propagate
in vitro. This method was described based on the experience using
HCV JFH-1 strain [17, 19].
2.2 Cloning Vector The plasmid containing the human RNA polymerase I promoter
(Pol I-P) and the mouse terminator (Pol I-T) sequences (e.g.,
pHH21 [5]) is used for plasmid-based HCV reverse genetics sys-
tems. The constructed plasmid allowed researchers to generate
wild-type and mutant viruses by transfection of plasmid without
RNA transcription. Viral genomic RNA is transcribed in the
nucleus of transfected cells, which is exported to the cytoplasm to
initiate RNA replication, following viral assembly and secretion
(Fig. 1).
E1 p7 NS4A NS5A
Pol I P Pol I T
C E2 NS2 NS3 NS4B NS5B
Transfection Release
Assembly
RNA
Transcription replication
nucleus
Translation
ER
Fig. 1 Overview of the reverse genetics of hepatitis C virus based on the Pol I-
mediated transcription
Huh-7 or Huh7.5.1 cells are transfected with plasmid containing the full-length
HCV cDNA under control of RNA polymerase I promoter (Pol I-P). A genomic viral
RNA generated in the nucleus is transported to the cytoplasm, leading to
expression of the viral proteins in a cap-independent manner. The HCV genome
is synthesized by the replication complex, which consists of viral and host cell-
derived proteins, in tight association with structurally rearranged vesicle-like
cytoplasmic membranes. The genomic RNA synthesized and the viral structural
proteins are assembled into the viral particle. Matured infectious virus is
released from cells
2.4 Cell Line and 1. The human hepatoma cell line, Huh-7, or its derivative cell
Reagents lines such as Huh7.5.1 [20].
2. Dulbecco’s Modified Eagle Medium (DMEM) supplemented
with nonessential amino acids, 100 U of penicillin/ml, 100 mg
of streptomycin/ml, and 10% fetal bovine serum (FBS).
3. OPTI-MEM I reduced serum medium (Invitrogen).
4. Phosphate-buffered saline (PBS).
178 Ryosuke Suzuki and Tetsuro Suzuki
3 Methods
3.1 Isolation of Viral Virus RNA is isolated from virus stock or virus-infected cells by
RNA standard procedures using silica-based (e.g., RNeasy Mini Kit,
Qiagen) or organic-based (e.g., TRIzol reagent, Invitrogen)
reagents following procedures recommended by the manufacturer.
Extracted RNA is eluted in RNase-free water.
3.3 Assembly of the 1. Digest the PCR products from cloning vector with
Full-Length cDNA corresponding restriction endonuclease.
Clone 2. Digest the vector containing the human Pol I-P and the mouse
Pol I-T with corresponding restriction endonuclease (e.g.,
BsmBI for pHH21 vector) (Fig. 2).
RNA Polymerase I-Mediated HCV Reverse Genetics 179
Amp
pHH21
Pol I P Pol I T
HCV cDNA
Pol I P BsmBI Pol I T BsmBI
-GGGTTATT GGAGACGGTACCGTCTCCT CCCCCCCA- CGTCTCNTATTACCTGCCC----GATCATGTCCCNGAGACG
-CCCAATAA CCTCTGCCATGGCAGAGGA GGGGGGGT- GCAGAGNATAATGGACGGG----CTAGTACAGGGNCTCTGC
BsmBI BsmBI
HCV cDNA
Pol I P Pol I T
-GGGTTATT ACCTGCCC----GATCATGT CCCCCCCA-
-CCCAATAA TGGACGGG----CTAGTACA GGGGGGGT-
4 Notes
Acknowledgments
References
16. Ikegami T (2021) Development of a simian versatile tool for study of virus assembly and
RNA polymerase I promoter-driven reverse infection. Virology 432:29–38
genetics for the rescue of recombinant rift val- 19. Wakita T, Pietschmann T, Kato T, Date T et al
ley fever virus from vero cells. J Virol 95: (2005) Production of infectious hepatitis C
e02004–e02020 virus in tissue culture from a cloned viral
17. Masaki T, Suzuki R, Saeed M et al (2010) genome. Nat Med 11:791–796
Production of infectious hepatitis C virus by 20. Zhong J, Gastaminza P, Cheng G et al (2005)
using RNA polymerase I-mediated transcrip- Robust hepatitis C virus infection in vitro. Proc
tion. J Virol 84:5824–5835 Natl Acad Sci U S A 102:9294–9299
18. Suzuki R, Saito K, Kato T et al (2012) Trans-
complemented hepatitis C virus particles as a
Chapter 12
Abstract
Zika virus (ZIKV) is a mosquito-borne member of the Flaviviridae family that has become a global threat to
human health. Although ZIKV has been known to circulate for decades causing mild febrile illness, the
more recent ZIKV outbreaks in the Americas and the Caribbean have been associated with severe neuro-
logical disorders and congenital abnormalities. The development of ZIKV reverse genetics approaches have
allowed researchers to address key questions on the biology of ZIKV by genetically engineering infectious
recombinant (r)ZIKV. This has resulted in a better understanding of the biology of ZIKV infections,
including viral pathogenesis, molecular mechanisms of viral replication and transcription, or the interaction
of viral and host factors, among others aspects. In addition, reverse genetics systems have facilitated the
identification of anti-ZIKV compounds and the development of new prophylactic approaches to combat
ZIKV infections. Different reverse genetics strategies have been implemented for the recovery of rZIKV. All
these reverse genetics systems have faced and overcome multiple challenges, including the viral genome
size, the toxicity of viral sequences in bacteria, etc. In this chapter we describe the generation of a ZIKV full-
length complementary (c)DNA infectious clone based on the use of a bacterial artificial chromosome
(BAC) and the experimental procedures for the successful recovery of rZIKV. Importantly, the protocol
described in this chapter provides a powerful method for the generation of infectious clones of other
flaviviruses with genomes that have stability problems during bacterial propagation.
Key words Zika virus, Reverse genetics, Recombinant virus, Bacterial artificial chromosome, cDNA
clone, Infectious clone
1 Introduction
Daniel R. Pérez (ed.), Reverse Genetics of RNA Viruses: Methods and Protocols, Methods in Molecular Biology, vol. 2733,
https://doi.org/10.1007/978-1-0716-3533-9_12,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2024
185
186 Aitor Nogales et al.
A)
Envelope (E) protein
Genomic RNA
B)
0 2 4 6 8 10 11 kb
5’ UTR 3’ UTR
Cap C prM E NS1 NS2A NS2B NS3 NS4A NS4B NS5
Fig. 1 ZIKV structure and genome organization. (a) Virion structure. The viral particle is constituted by a
nucleocapsid core composed by the C protein associated to the genomic RNA, surrounded by a lipid bilayer
that contains the structural proteins M and E arranged with icosahedral symmetry on the surface. (b) Genetic
structure of the ZIKV genome. The viral genome is a positive single-stranded RNA molecule that contains a cap
structure at the 5′-end and a single open reading frame (ORF) flanked by 5′- and 3′ untranslated regions
(UTR). The coding regions from the structural (C, prM/M, and E) and nonstructural (NS1, NS2A, NS2B, NS3,
NS4A, NS4B, and NS5) proteins are illustrated by colored boxes. A scale in kb is included at the top as
reference of the genomic position of the coding regions
CMV C prM E NS1 NS2A NS2B NS3 NS4A NS4B NS5 Rz BGH
Cmr
parC
Fig. 2 Strategy to assemble a ZIKV-RGN BAC cDNA clone. After the selection of appropriated restriction sites in
the viral genome (genomic positions indicated in parenthesis), four overlapping fragments (ZIKV 1 to ZIKV 4)
covering the entire viral genome and flanked by the selected restriction sites were chemically synthesized and
sequentially cloned into the pBeloBAC11-Afe I plasmid to generate the pBAC-ZIKV infectious clone. Fragment
ZIKV 1 contains the human cytomegalovirus (CMV) promoter to allow the expression of the viral RNA into
transfected cells. Fragment ZIKV 4 contains the last nucleotides of the viral genome fused to the hepatitis delta
virus (HDV) ribozyme (Rz) and the bovine growth hormone (BGH) termination and polyadenylation sequences to
produce viral RNAs with authentic 3′-ends. Both, the fusion of CMV-5′ end and 3′ end-Rz are shown, where
the transcription start and the Rz cleavage site are indicated. Acronyms for coding region and regulatory
element are as described in Fig. 1. The BAC regulatory genes (parA, parB, parC, and repE), the F-factor
replication origin (OriS), and the chloramphenicol resistance gene (Cmr) are also indicated
2 Materials
2.1 Biosafety This chapter describes the assembly of ZIKV-RGN strain [10, 29]
Recommendation (accession number KU527068) in the pBeloBAC11 BAC (see Note
1). The rescue and manipulation of the recovered rZIKV should be
performed in a biosafety level 2 (BSL2), BSL2+, or BSL3 labora-
tory according to the country and institutional regulations and
required permits regarding ZIKV handling and storage. All mate-
rial used for the recovery of rZIKV must be sterilized before dis-
posal, following the appropriated institutional biosafety
indications.
2.3 Culture Media for 1. Luria broth (LB) liquid media: 1% (w/v) tryptone, 0.5% (w/v)
E. coli yeast extract, 1% (w/v) NaCl, pH 7.0. It can be purchased from
suppliers or prepared in the laboratory. To prepare LB liquid
media, mix 10 g of Bacto Tryptone, 5 g Bacto Yeast Extract,
and 10 g NaCl in 900 mL of ddH2O. Adjust the pH to 7.0 with
5 N NaOH, and adjust the volume of the solution to 1 L with
ddH2O. Sterilize by autoclaving on liquid cycle, and store at 4 °
C.
2. LB liquid media with chloramphenicol: Add chloramphenicol
to a final concentration of 12.5 μg/mL from a stock solution of
34 mg/mL. LB liquid media with chloramphenicol can be
stored at 4 °C for 1 month.
3. LB agar plates with chloramphenicol: Prepare 500 mL LB
liquid media, and, just before autoclaving, add 15 g/L of
Bacto Agar. After autoclaving on liquid cycle, equilibrate at
55 °C for 30 min, and add chloramphenicol to a final concen-
tration of 12.5 μg/mL. Dispense 20–25 mL of media to
90 mm petri dishes, and store LB agar plates at 4 °C.
4. SOC liquid media: 2% (w/v) tryptone, 0.5% (w/v) yeast
extract, 0.05% (w/v) NaCl, 2.5 mM KCl, 10 mM MgCl2,
10 mM MgSO4, 20 mM glucose, pH 7.0. SOC liquid media
can be purchased from suppliers or prepared in the laboratory.
To prepare SOC media, mix 20 g of Bacto Tryptone, 5 g Bacto
Yeast Extract, 0.5 g NaCl, and 2.5 mL of 1 M KCl (final
concentration 2.5 mM) in 900 mL of ddH2O. Adjust the pH
to 7.0 with 5 N NaOH, and adjust the volume of the solution
to 1 L with ddH2O. Autoclave and, before use, add 10 mL of
1 M MgCl2, 10 mL of 1 M MgSO4, and 20 mL of 1 M glucose.
Store SOC liquid media at 4 °C.
190 Aitor Nogales et al.
2.4 Enzymes, The enzymes, reagents, and kits described in this section could be
Reagents, and Kits for purchased from other suppliers different to those recommended
the Assembly and here.
Manipulation of BAC 1. Restriction endonucleases (New England Biolabs, Inc.): Afe I,
Clones ApaL I, Asc I, BamH I, BstB I, Mlu I, and Pml I (see Note 4).
2. Shrimp Alkaline Phosphatase (SAP, Promega).
3. T4 DNA ligase (Promega).
4. Expand high-fidelity DNA polymerase (Roche).
5. PCR Nucleotide Mix (dNTP).
6. UltraPure distilled water (ddH2O).
7. SeaKem LE Agarose (Lonza) (see Note 5).
8. QIAEX II Gel Extraction Kit (Qiagen).
9. QIAGEN Plasmid Midi Kit (Qiagen).
10. QIAGEN Large-Construct Kit (Qiagen) (see Note 6).
2.5 Cell Culture Other suppliers different to those recommended here could also be
Media and Reagents used.
for Rescue and
1. Dulbecco’s Modified Eagle’s Medium (DMEM, Gibco).
Propagation of rZIKV
2. Fetal bovine serum (FBS).
3. Growth medium: DMEM supplemented with 5% FBS,
2 mM L-glutamine (Sigma-Aldrich), 1% nonessential amino
acids (Sigma-Aldrich), and 100 mg/mL penicillin/streptomy-
cin (Sigma-Aldrich). Store at 4 °C.
4. Virus growth medium: DMEM supplemented with 2% FBS,
2 mM L-glutamine (Sigma-Aldrich), 1% nonessential amino
acids (Sigma-Aldrich), and 100 mg/mL penicillin/streptomy-
cin (Sigma-Aldrich). Store at 4 °C.
5. Trypsin-EDTA solution (Sigma-Aldrich): 0.25% (w/v) trypsin,
0.02% (w/v) EDTA.
6. Phosphate-buffered saline (PBS): PBS (138 mM NaCl, 3 mM
KCl, 8.1 mM of Na2HPO4, 1.5 mM KH2PO4, pH 7.4) can be
purchased from suppliers or prepared in the laboratory. To
prepare PBS, mix 8 g of NaCl, 0.2 g of KCl, 1.44 g of
Na2HPO4, and 0.24 g of KH2PO4 in 900 mL of
ddH2O. Adjust pH to 7.4, add ddH2O up to 1 L, sterilize by
autoclaving, and store at room temperature.
BAC-Based ZIKV Reverse Genetics 191
2.6 Cell Line for the Vero cells (African green monkey kidney epithelial cells) are avail-
Rescue of rZIKV able from the American Type Culture Collection, ATCC, CCL-81
(see Note 7). Vero cells have been shown to have a high transfection
efficiency, and they are the best cell line for producing high ZIKV
titers. Vero cells are growth and maintained in growth medium at
37 °C in a 5% CO2 atmosphere cell culture humidified incubator.
2.7 Reagents for the The reagent described in this section could be purchased from
Titration and Detection other suppliers different to those recommended here.
of ZIKV
2.7.1 Plaque Assay and 1. Overlay solution: Virus growth medium containing 1% DEAE-
Immunostaining dextran (Sigma-Aldrich) and 0.6% agar noble (Difco, Thermo
Fisher) (see Note 8).
2. 37% formaldehyde (Sigma-Aldrich).
3. Crystal violet solution: 0.5% crystal violet (Sigma-Aldrich),
20% methanol (Merck) (see Note 9).
4. Immunostaining blocking solution: 10% FBS in PBS.
5. Primary monoclonal antibody (mAb) pan-flavivirus E protein
4G2 (BEI Resources NR-50327) (see Note 10).
6. Biotinylated anti-mouse secondary antibody (Vector Labora-
tories, Inc.).
7. Vectastain ABC Kit (Vector Laboratories, Inc.).
8. DAB Substrate Kit, Peroxidase (HRP), with Nickel (Vector
Laboratory, Inc.).
3 Methods
3.1 Assembly of a The strategy for the assembly and generation of the ZIKV-RGN
ZIKV Infectious cDNA strain infectious clone using the BAC has been previously described
Clone in the BAC [10, 29] (Fig. 2).
1. The first step for the assembly of the full-length cDNA clone is
the choice of appropriate restriction endonuclease sites. For
that, select unique restriction sites in the viral genome that
are absent in the pBeloBAC11-AfeI (Fig. 2). In the case of
ZIKV-RGN, the unique restriction sites Pml I, Afe I, and
BstB I (genomic positions 3347, 5969, and 9127, respectively)
were selected to generate the infectious clone (Fig. 2) (see
Note 11).
2. Design and chemically synthesize four overlapping cDNA frag-
ments spanning the full-length viral genome (ZIKV 1 to ZIKV
4 fragments, Fig. 2) flanked by the appropriated regulatory
elements and restriction sites (see Note 12). The full-length
cDNA must be flanked by the CMV promoter at the 5′-end
(ZIKV 1) and by the HDV Rz and the BGH termination and
polyadenylation sequences at the 3′-end (ZIKV 4) (Fig. 2).
Fragment ZIKV 1 contains the CMV immediate-early pro-
moter precisely fused to the first 3350 nucleotides of the viral
genome (until restriction site Pml I) flanked at the 5′-end by
ApaL I (to clone into the pBeloBAC11-AfeI) and Asc I (absent
in the viral genome and in the vector) and at the 3′-end by the
BAC-Based ZIKV Reverse Genetics 193
10. Plate the cells onto LB agar plates containing 12.5 μg/mL
chloramphenicol, and incubate them at 37 °C for 16–24 h.
11. Pick about ten bacterial colonies, make a replica on a fresh LB
agar plate containing 12.5 μg/mL chloramphenicol, and test
whether they contain the correct insert by direct PCR analysis
using specific oligonucleotides and expand high-fidelity DNA
polymerase, following the manufacturer’s instruction (see
Note 18).
12. Pick a positive colony from the replica plate, and grow it in
100 mL of LB containing 12.5 μg/mL of chloramphenicol at
37 °C for 16 h with shaking (200–250 rpm). Isolate the BAC
plasmid using the QIAGEN Plasmid Midi Kit, following the
protocol for purification of large low-copy plasmids (see
Note 19).
13. Verify the genetic integrity of the isolated plasmid by restriction
analysis and sequencing using specific oligonucleotides.
14. After confirming the genetic identity of the plasmid pBAC-
ZIKV1, we recommend to prepare a glycerol stock of positive
bacteria. For that, mix 0.5 mL of LB freezing buffer with
0.5 mL of an overnight bacterial culture in a cryotube, freeze
in ethanol–dry ice, and store at -80 °C.
15. Starting from the generated plasmid pBAC-ZIKV1, sequen-
tially clone fragments ZIKV 2 to ZIKV 4 using the selected
restriction sites (Pml I /Afe I for ZIKV2, Afe I /BstB I for
ZIKV 3, and BstB I/Mlu I for ZIKV 4) to generate the full-
length infectious pBAC-ZIKV cDNA clone (Fig. 2), following
similar experimental approaches as described above for ZIKV
1 fragment.
3.2 Preparation of For the rescue of rZIKV using the infectious BAC cDNA clone, it is
Ultrapure pBAC-ZIKV recommended to prepare high-purity pBAC-ZIKV with no bacteria
Infectious Clone DNA contamination. Any commercial kit designed for BAC purifi-
cation is suitable, but we recommend using the QIAGEN Large-
Construct Kit. This kit includes an ATP-dependent exonuclease
digestion step that removes bacteria DNA contamination, allowing
the isolation of the BAC plasmid with a grade of purity similar to
that using cesium chloride method.
1. Inoculate a single colony of bacteria carrying the pBAC-ZIKV
infectious clone from a freshly streaked plate (LB agar plate
containing 12.5 μg/ml chloramphenicol) in 5 mL of LB con-
taining 12.5 μg/mL of chloramphenicol, and incubate at 37 °C
for 8 h with vigorous shaking (200–250 rpm).
2. Add 1 mL of the bacterial culture to 500 mL of LB with
12.5 μg/mL of chloramphenicol, and grow the bacteria at
37 °C with vigorous shaking (200–250 rpm) for 14–16 h
until an OD600 of 0.8–1 (late logarithmic growth phase) (see
Note 20).
BAC-Based ZIKV Reverse Genetics 195
3.3 Rescue of rZIKV Overall, rescue of rZIKV is carried out using experimental methods
similar to those previously described [23, 24, 29, 30, 50]. To
increase the likelihood of successfully recovering rZIKV, we recom-
mend three independent transfections. Infectious rZIKV is recov-
ered by the transfection of Vero cells with the generated pBAC-
ZIKV cDNA clone, using LPF2000 as transfection reagent. A
schematic representation of the protocol to generate rZIKV is
shown in Fig. 3. The following protocol is established for six-well
plates.
1. The day before transfection, plate 6 × 105 Vero cells/well on
six-well plates using growth medium without antibiotics to
raise 90% confluent cell monolayers on the day of transfection
(see Note 23). On the day of transfection, check the cells under
a light microscope to confirm the presence of a uniform cell
monolayer.
2. Just before transfection, equilibrate the Opti-MEM I medium
at room temperature, put the DNA and the LPF2000 reagent
on ice, and prepare sterile microfuge tubes.
3. Prepare Opti-MEM I-LPF2000 and plasmid transfection
mixtures.
4. Opti-MEM I-LPF2000 mixture: For each transfection sample,
mix 250 μL of Opti-MEM I medium and 12 μL of LPF2000
(gently vortex before use), and incubate for 5 min at room
temperature. Meanwhile, prepare the plasmid transfection
mixture.
5. Plasmid transfection mixture: Dilute 4 μg of pBAC-ZIKV plas-
mid in 250 μL of Opti-MEM I medium. Avoid prolonged
pipetting to prevent plasmid shearing.
6. Combine the Opti-MEM I-LPF2000 (step 4) with diluted
DNA (step 5), mix carefully, and incubate for 20–30 min at
room temperature.
7. During this incubation period, wash the Vero cell monolayers
twice with 4 mL of growth medium without antibiotics, and
leave the cells in 1 mL of the same medium (see Note 23).
196 Aitor Nogales et al.
CMV C prM E NS1 NS2A NS2B NS3 NS4A NS4B NS5 Rz BGH
pBAC-ZIKV
Cmr
parC
(18,308 bp)
TRANSFECTION
Vero cells
4-6 DAYS
rZIKV
VIRUS AMPLIFICATION
Fig. 3 Rescue of rZIKV from the pBAC cDNA clone. Vero cell monolayers at 90% confluence (six-well plate
format, triplicates) are transfected with 4 μg of the pBAC-ZIKV cDNA clone (Fig. 2) using LPF2000. When CPE
is evident (4–6 days post-transfection), cell cultured supernatants are collected and titrated in Vero cells, and
the presence of the rZIKV is confirmed using different experimental approaches. Finally, the rescued rZIKV is
amplified and titrated in Vero cells (Fig. 4) and stored at -80 °C
A) B) C)
MOCK CRYSTAL VIOLET MOCK
Fig. 4 In vitro characterization of the rescued rZIKV. (a) CPE. Confluent (~90%) monolayers of Vero cells
(six-well plate format, triplicates) were mock-transfected or transfected with pBAC-ZIKV, and CPE was
analyzed daily. Pictures show the presence of CPE at 4 days after transfection with pBAC-ZIKV (bottom) but
not in mock-transfected cells (top). Scale bars, 50 μm. (b) Plaque assays. Vero cells at 90% confluence
(six-well plate format, triplicates) were infected with tenfold serial dilutions of rZIKV, and at 3–4 days post-
infection, viral plaques were visualized by crystal violet staining (top) or by immunostaining using the
pan-flavivirus E protein mAb 4G2 (bottom). Representative images containing ~25–50 PFU are shown. (c)
Immunofluorescence assay (IFA). Vero cells at 90% confluence (24-well plate format, triplicates) were mock-
infected (top) or infected (bottom) with 0.5 PFU/cell of rescued rZIKV. At 18 h post-infection, the expression of
the viral E protein was assessed by fluorescent microscopy using the pan-flavivirus E protein mAb 4G2. Cell
nuclei were stained with DAPI. Scale bars and 20 μm
3.4.1 Plaque Assay and 1. The day before infection, seed six-well plates with approxi-
Crystal Violet Staining mately 6 × 105 Vero cells/well in growth medium to raise
90% confluent cell monolayers by the time of titration. We
recommend conducting the titration of the recovered rZIKV
in triplicates.
2. Next day, make tenfold serial dilutions (10-1–10-6) of the
virus sample in virus growth medium without FBS. For that,
add 900 μL of virus growth medium without FBS in six micro-
tubes. Then, add 100 μL of virus sample in the first microtube
(dilution 10-1), and vortex the tube for 5 s. Pipet 100 μL of the
10-1 dilution, and transfer into the second tube (dilution 10-
2
). Repeat these steps for the other dilutions. Change the tip to
avoid any cross-contamination between samples.
3. Wash the cells twice with PBS, and starting with the most
diluted sample, add 800 μL of the virus dilution to each of
the wells. Place plates in a 5% CO2 humidified incubator at 37 °
C for a 90 min adsorption period, rocking the plates every
15 min to prevent the cells from drying.
4. Remove the viral inoculum, and overlay the cells with 3 mL of
virus growth medium containing 1% DEAE-dextran and 0.6%
agar noble. Let the plate(s) sit for 10 min at room temperature,
as the agar overlay solidifies, and incubate the plates at 37 °C in
the 5% CO2 incubator for 3–4 days (see Note 8).
5. When viral plaques are visible, fix the plates with a 2 mL of 4%
formaldehyde in PBS for 1 h at room temperature.
6. Carefully remove the overlay media.
7. Add 1 mL of 0.5% crystal violet solution to each well, and
incubate for 30 min at room temperature.
8. Discard the crystal violet solution, wash the cells gently with
ddH2O, and allow the plates to air-dry.
9. Count the plaques in the lowest two dilutions with visual
plaques, and calculate the viral titer in plaque-forming units
(PFU)/mL using the following formula: PFU/mL = number
of plaques x virus dilution x 1/volume of inoculum (mL).
3.4.2 Plaque Assay and To perform the immunostaining, we recommend the use of a
Immunostaining commercial kit as Vectastain ABC Kit (Vector Laboratories, Inc.),
following the manufacture’s indications. The following protocol is
designed for a six-well plate format.
1. Perform cell infections and plaque assay as indicated above in
Subheading 3.4.1 (steps 1–6).
2. After fixing and removal of the overlay agar, wash the cells twice
with PBS.
3. Permeabilize the cells by incubation with 1 mL/well of 0.5%
Triton X-100 in PBS for 15 min at room temperature.
BAC-Based ZIKV Reverse Genetics 199
4. Wash the cells three times with PBS, and incubate them with
1 mL/well of blocking solution (10% FBS in PBS) for 1 h at
room temperature.
5. Remove the blocking solution, and incubate the cells with
1 mL/well of the pan-flavivirus E protein mAb 4G2 diluted
in blocking solution (1 μg/mL) for 1 h at 37 °C.
6. Wash the cells three times with PBS, add 1 mL of biotinylated
anti-mouse secondary antibody diluted (following the manu-
facturer’s recommendation) in blocking solution, and incubate
for 1 h at 37 °C.
7. Wash the cells three times with PBS to remove biotinylated Ab
solution thoroughly.
8. Prepare VECTASTAIN ABC Reagent by following manufac-
turer’s instructions. Add 50 μL of Reagent A (Avidin, ABC)
and 50 μL of Reagent B (Biotinylated HRP, ABC) to 5 mL of
PBS. Add 1 mL/well of VECTASTAIN ABC Reagent, and
incubate for 30 min at 37 °C.
9. Wash cells three times with PBS. Remove PBS, and dry the
plate.
10. Prepare developing solution by following manufacturer’s
instructions (DAB Substrate Kit with Nickel).
11. Add 1 mL of developing solution to each well and wait for
3–5 min to visualize viral plaques.
12. Stop the reaction by removing the developing solution, and
wash with PBS (see Note 25).
13. Take images of the different plates.
3.5 Confirmation of To confirm the identity of the rescued virus, Vero cells are infected
rZIKV Rescue by IFA with rZIKV, and the expression of ZIKV E protein is analyzed by
IFA using the pan-flavivirus E protein mAb 4G2 (Fig. 4c). Alterna-
tively, the virus identity can be confirmed directly by sequencing.
Using the IFA assay, the virus titers in the cell culture supernatants
can be also determined by immunofocus assay (fluorescent focus-
forming units (FFU)/ml) in 96-well plate format.
1. The day before performing the IFA, put sterile coverslips in
24-well plates, and seed 1 × 105 Vero cells/well in growth
medium to raise 90% confluent cell monolayers by the time of
infection. Place the plates at 37 °C in a 5% CO2 humidified
incubator.
2. The next day, wash the cells twice with PBS, and infect them in
triplicate with a multiplicity of infection (MOI) of 0.5 PFU/-
cell of the rescue rZIKV in virus growth medium without FBS
(final volume 250 μL/well). Incubate the plates at 37 °C for
90 min.
200 Aitor Nogales et al.
3.6 Amplification of 1. The day before infection, seed 6 × 106 Vero cells in 100 mm
ZIKV and Production of plates in growth medium to raise 90% confluent cell mono-
Virus Stocks layers by the time of infection.
2. The next day, wash the cells twice with PBS, and infect them
with an MOI of 0.1 PFU/cell as described before (see Note
28).
3. When the CPE is around 75% (approximately 2–3 days post-
infection), collect the cell culture supernatant into a 15 ml
falcon tube, and centrifuge at 2000 × g for 10 min at 4 °C to
remove cells debris.
4. Collect the supernatant containing rZIKV into a new 15 mL
falcon tube, and discard the cell pellet.
5. Aliquot the supernatant in cryotubes, and store them at -80 °
C (see Note 29).
6. Determine the viral titer by plaque assay as described before
(Subheading 3.4).
BAC-Based ZIKV Reverse Genetics 201
4 Notes
25. It is important not to wait for too long once the plaques are
visible to prevent the entire cells from turning black.
26. Cells can be blocked using alternative blocking solutions (e.g.,
10% FBS in PBS) or overnight at 4 °C.
27. For long-term storage, store samples at 4 °C, protected from
light.
28. Adjust the volumes according with the plaque size.
29. We recommend making small-volume aliquots to prevent mul-
tiple freeze-thaw cycles, which may reduce virus titers.
Acknowledgments
References
1. Pierson TC, Kielian M (2013) Flaviviruses: perspectives, mechanisms, and impact. BMC
braking the entering. Curr Opin Virol 3(1): Med 14(1):112. https://doi.org/10.1186/
3–12. https://doi.org/10.1016/j.coviro. s12916-016-0660-0. [pii]
2012.12.001. S1879-6257(12)00189-7 [pii] 6. Costello A, Dua T, Duran P, Gulmezoglu M,
2. Sirohi D, Chen Z, Sun L, Klose T, Pierson TC, Oladapo OT, Perea W et al (2016) Defining
Rossmann MG et al (2016) The 3.8 a resolu- the syndrome associated with congenital Zika
tion cryo-EM structure of Zika virus. Science virus infection. Bull World Health Organ
352(6284):467–470. https://doi.org/10. 94(6):406-A. https://doi.org/10.2471/BLT.
1126/science.aaf5316. [pii] 16.176990. [pii]
3. Calvet G, Aguiar RS, Melo ASO, Sampaio SA, 7. Cugola FR, Fernandes IR, Russo FB, Freitas
de Filippis I, Fabri A et al (2016) Detection and BC, Dias JL, Guimaraes KP et al (2016) The
sequencing of Zika virus from amniotic fluid of Brazilian Zika virus strain causes birth defects
fetuses with microcephaly in Brazil: a case in experimental models. Nature 534(7606):
study. Lancet Infect Dis 16(6):653–660. 2 6 7 – 2 7 1 . h t t p s : // d o i . o r g / 1 0 . 1 0 3 8 /
https://doi.org/10.1016/S1473-3099(16) nature18296. [pii]
00095-5. S1473-3099(16)00095-5 [pii] 8. do Rosario MS, de Jesus PA, Vasilakis N, Farias
4. Duffy MR, Chen TH, Hancock WT, Powers DS, Novaes MA, Rodrigues SG et al (2016)
AM, Kool JL, Lanciotti RS et al (2009) Zika Guillain-Barre syndrome after Zika virus infec-
virus outbreak on Yap Island, Federated States tion in Brazil. Am J Trop Med Hyg 95(5):
of Micronesia. N Engl J Med 360(24): 1157–1160. https://doi.org/10.4269/ajtmh.
2536–2543. https://doi.org/10.1056/ 16-0306. ajtmh.16-0306 [pii]
NEJMoa0805715. [pii] 9. Miner JJ, Cao B, Govero J, Smith AM,
5. Boeuf P, Drummer HE, Richards JS, Scoullar Fernandez E, Cabrera OH et al (2016) Zika
MJ, Beeson JG (2016) The global threat of virus infection during pregnancy in mice causes
Zika virus to pregnancy: epidemiology, clinical placental damage and fetal demise. Cell
204 Aitor Nogales et al.
Abstract
Zika virus (ZIKV) is a mosquito-borne flavivirus of the Flaviviridae family first isolated from a sentinel
monkey in the Zika Forest, Uganda, in 1947. Since 2007, the virus has had a vast geographic expansion that
extended to the Americas in 2015, leading to a series of large outbreaks. Although mainly transmitted by
the bite of Aedes mosquitoes, human infection of ZIKV can also happen through unconventional routes
such as sexual intercourse and, more importantly, vertical transmission. The genome of ZIKV is a single-
stranded, positive-sense RNA molecule about 11 kb in length. The genome contains a single opening
reading frame (ORF) flanked by highly structured 5′ and 3′ untranslated regions. To understand the
mechanisms about ZIKV replication, transmission, and pathogenesis, reverse genetic tools are of great
importance. In this chapter, a novel system is described for the generation and manipulation of a ZIKV
infectious clone stabilized by a self-splicing group II intron, a mobile element with ribozyme activity. The
intron can be spliced in vitro, and thus full-length vRNA can be prepared allowing virus genome manipula-
tion required for further studies.
Key words Zika virus, Reverse genetics, Recombinant virus, Ribozyme, Self-splicing, Intron, Infec-
tious clone
1 Introduction
Daniel R. Pérez (ed.), Reverse Genetics of RNA Viruses: Methods and Protocols, Methods in Molecular Biology, vol. 2733,
https://doi.org/10.1007/978-1-0716-3533-9_13,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2024
207
208 Zhong-Yu Liu et al.
2.1 Isolation of ZIKV 1. ZIKV strain GZ01 (GenBank: KU820898) was isolated from
RNA from Culture the serum samples of an imported case of Zika infection
Supernatants [24]. Baby hamster kidney cell line BHK-21 (ATCC
CCL-10) is cultured using DMEM supplemented with 5%
FBS, 25 mM HEPES, 100 U/mL penicillin, and 100 μg/mL
streptomycin.
2. DMEM medium, low glucose (Thermo Fisher Scientific).
A Stable Reverse Genetics System of Zika Virus Based on a Self-Splicing. . . 209
2.2 RT-PCR for ZIKV 1. Superscript III reverse transcriptase (Thermo Fisher Scientific).
cDNA Fragments 2. Pyrobest high-fidelity DNA polymerase (Takara).
3. LA Taq DNA polymerase (Takara).
4. dNTP mixture (2.5 mM and 10 mM for each dNTP) (Takara).
5. Nuclease-free water.
6. Agarose (Biowest).
7. 50×TAE (dissolve 242 g Tris-free base in ultrapure water, then
add 57.1 mL HOAc and 100 mL 0.5 M EDTA, pH 8.0, adjust
the volume to 1 L).
8. 10× loading solution (Takara).
9. Ethidium bromide (5 mg/mL).
10. Wizard SV Gel and PCR Clean-Up System (Promega).
14. 50× TAE (242 g Tris-free base, 57.1 mL HOAc, and 100 mL
0.5 M EDTA, pH 8.0 in 1 L of ultrapure water).
15. 10× loading buffer (Takara).
16. Wizard SV Gel and PCR Clean-Up System (Promega).
17. Restriction endonucleases NotI-HF, KpnI-HF, KasI, BstEII-
HF, XhoI, SacII, and HindIII-HF.
2.4 In Vitro 1. PureLink HiPure Plasmid Filter Maxiprep Kit (Thermo Fisher
Transcription Scientific).
2. Q5 high-fidelity DNA polymerase (NEB).
3. Wizard SV Gel and PCR Clean-Up System (Promega).
4. RiboMax SP6 large-scale RNA production system (Promega).
5. m7GpppG analogue (Promega).
6. Nuclease-free water.
7. RNeasy Mini Kit (Qiagen) or E.Z.N.A. Total RNA Kit I
(Omega Bio-tek).
3 Methods
3.1 Isolation of ZIKV 1. Seed BHK-21 cells into a 25 cm2 flask, and incubate in a 37 °C
RNA from Culture incubator with 5% CO2 until the cells reach approximately 90%
Supernatants confluency.
3.1.1 Propagation of ZIKV 2. Infect the BHK-21 cells using 0.1 MOI of ZIKV GZ01 strain,
in BHK-21 Cells and change the virus-containing medium with fresh DMEM
containing 2% FBS after a two-hour incubation. Culture the
infected cells at 37 °C with 5% CO2 until visible CPE is
apparent.
3. Freeze and thaw the infected cells together with the superna-
tant for one or two times, transfer the culture into a sterile
50 mL centrifuge tube, and centrifuge at 6000 g for 10 min at
4 °C. Discard the precipitation, aliquot the supernatant into
sterile screw cap cryotubes. The virus aliquot can be stored at -
80 °C for long-term storage. Steps 2 and 3 require a BSL-
2 facility.
3.1.2 ZIKV RNA Isolation The vRNA can be isolated from the virus-containing supernatants
from Culture Supernatants in Subheading 3.1.1. A brief procedure based on the RNeasy Mini
Kit is listed below.
212 Zhong-Yu Liu et al.
3.2 RT-PCR 1. Generation of cDNA from the isolated vRNA. Set up the
reaction as following:
3.2.1 Reverse
Transcription
Components Volume
Isolated vRNA 8 μL
dNTP mixture (10 mM each) 1 μL
Primer P-Z-R14B (20 μM, see Note 4) 1 μL
RNase-free H2O 2 μL
Total 12 μL
Components Volume
Denatured mixture in step 1 12 μL
5× first-strand buffer 4 μL
0.1 M DTT 2 μL
RNase inhibitor 1 μL
Superscript III reverse transcriptase 1 μL
Total 20 μL
A Stable Reverse Genetics System of Zika Virus Based on a Self-Splicing. . . 213
Table 1
Primers for the amplification of ZIKV GZ01 fragments
3.2.2 PCR Amplification 1. Using the above obtained cDNA as the template, amplify the
four DNA fragments (S1, S2, S3, S4′, see Note 5). Primer
sequences are listed in Table 1.
Set up the reactions as the following:
Components Volume
cDNA product 1 μL
Forward primer (20 μM) 1 μL
Reverse primer (20 μM) 1 μL
10× LA Taq Buffer 5 μL
dNTP mixture (2.5 mM each) 8 μL
LA Taq DNA polymerase (see Note 6) 0.5 μL
Nuclease-free water 33.5 μL
Total 50 μL
214 Zhong-Yu Liu et al.
3.3 Generation of the 1. Order the following oligonucleotides from a local supplier.
Infectious Clone P-ZSK-1: 5′-GGCCGCGCTAGATGCGGTACCTTTC
Plasmid AAGATGGGCGCCAATTACACTGCCGGCATATCATT
3.3.1 Construction of an
GGTAACCATTCCATTC-3′
Assemble Backbone P-ZSK-2: 5′-TCGAGAATGGAATGGTTACCAATGATAT
GCCGGCAGTGTAATTGGCGCCCATCTTGAAAGGT
ACCGCATCTAGCGC-3′
2. Dissolve the oligos into 20 μM solutions using nuclease-free
water (see Note 8). Mix 20 μL of the two oligos together, and
incubate the sample in boiling water for 5 min. Then left the
mixture in the water bath until it cools to below 30 °C (may
take a few hours). The oligos are now annealed to form a
dsDNA containing sticky ends of NotI and XhoI, respectively.
3. Digest the pACNR-E70 plasmid with NotI-HF/XhoI, separate
the digested plasmid DNA by agarose gel electrophoresis, and
retrieve the 2.3 kb length vector band by using a Wizard SV Gel
and PCR Clean-Up System. Ligate the annealed ZSK adaptor
dsDNA with the pACNR vector by using T4 DNA ligase at
4 °C for 16 h.
4. Transform the ligation reaction into E. coli TOP10 chemically
competent cells. Thaw the competent cell on ice. Add 15 μL of
the ligation reaction to 100 μL competent cell. Mix well, and
incubate on ice for 30 min.
5. Heat shock at 42 °C for 90 s. Then transfer the competent cell
to an ice bath immediately, and incubate for 2–3 min.
6. Add 500 μL LB broth, and rotate at 180–200 rpm under 37°C
for 50 min.
7. Transfer 200 μL of the transformed cells to a LB agar plate with
100 μg/mL ampicillin. Dispense evenly with a sterile spreader.
Incubate the agar plate inversely at 37 °C for 12–16 h when the
agar surface is dry.
8. Pick several clones, and incubate them into 3 mL of LB broth.
Culture in a shaker bath at 37 °C for 12–16 h. Isolate plasmid
from the clones. Then screen the recombinants by restriction
endonuclease analysis combined with Sanger sequencing to
obtain an aimed pACNR-GZ01-linker backbone plasmid.
3.3.2 Sub-cloning of ZIKV 1. Clone the PCR-prepared S2 fragment into the pACNR-GZ01-
cDNA Fragments linker by using KpnI-HF/KasI double digestion, which results
in the pACNR-GZ01-S2 recombinant plasmid. Follow the
basic cloning procedure in Subheading 3.3.1.
2. Clone the S1, S3, and S4′ fragments into pGEM-T Easy vector
by AT-cloning. Follow the basic procedure in Subheading
3.3.1, and add 20 μL of IPTG and X-gal before the spreading
of the bacterial suspension. Screen the resultant clones by blue-
216 Zhong-Yu Liu et al.
Components Volume
Cloned S4′ fragment (PCR prepared, 20–50 ng) 1 μL
S4ter (annealed, 20 μM) 1 μL
P-Z-BstEII-F 1 μL
P-Z-Tail 1 μL
10× LA Taq Buffer 5 μL
dNTP mixture (2.5 mM each) 8 μL
LA Taq DNA polymerase 0.5 μL
Nuclease-free water 32.5 μL
Total 50 μL
(continued)
A Stable Reverse Genetics System of Zika Virus Based on a Self-Splicing. . . 217
3.3.3 Design of the Order a full-gene synthesis service for the Pylaiella littoralis mito-
Modified P.li.LSUI2 Intron chondrial rRNA intron 2 (P.li.LSUI2) from a local supplier. Substi-
tute the exon-binding sequences (EBS) 1, 2, and 3 with the
complement sequence to the 2460–2464, 2467–2472, and 2473
positions in ZIKV GZ01 genome (Fig. 1). In addition, ZIKV
sequences were added to the upstream and downstream of the
self-splicing intron sequence to facilitate further molecular manip-
ulations. The secondary structure of the resultant P.li.LSUI2-
mZika is shown in Fig. 1.
Fuse the S1 fragment with the P.li.LSUI2-mZika sequence by
using overlapping PCR method as described in Subheading 3.3.2,
step 3. Clone the resultant S1-intron fragment into pGEM-T Easy,
and perform screening as described in Subheading 3.3.2.
3.3.4 Assembly of the 1. Stepwise assemble the S3-, S4-, and S1-intron fragments into
Full-Length pACNR-GZ01- the pACNR-GZ01-S2 plasmid by sub-cloning using the
Intron-IC corresponding restriction sites. Perform these sub-cloning
steps as described in Subheading 3.3.1. For the last round of
the assembly, shifting the culturing temperature from 37 °C to
30 °C may increase the positive rate of recombinants.
2. Identify the obtained pACNR-GZ01-Intron-IC by using
restriction endonuclease analysis (HindIII, SacII, and
NotI-HF/KpnI), and confirm the full-length genomic
sequence in the correct clone by Sanger sequencing.
3. Cryo-store the E. coli TOP10 strain containing the pACNR-
GZ01-Intron-IC. Add 200 μL of 50% sterile glycerol to 800 μL
of fresh overnight culture. Mix well, seal, and label the Eppen-
dorf tubes before storing them at -80 °C.
Fig. 1 Structure model of the P.li.LSUI2-mZika intron. Magenta, the EBS1-IBS1 sequence; cyan, the EBS2 and
IBS2 sequence; red, the EBS3 and IBS3 sequence
Components Volume
Plasmid DNA 35 μL (approximately 5–15 μg)
10× NEB CutSmart Buffer 5 μL
XhoI 1 μL
Nuclease-free water 9 μL
Total 50 μL
Volume
Control
Components Sample reaction
Template* 8 μL 2 μL
5× SP6 Buffer 6 μL 2 μL
NTP mixture (A:C:U:G=3:3:3:1, 100 mM in 6 μL 2 μL
total)
m7GpppG Cap analogue 1 μL 0.33 μL
(continued)
220 Zhong-Yu Liu et al.
Volume
Control
Components Sample reaction
SP6 Enzyme Mix 3 μL 1 μL
Nuclease-free water 6 μL 2.67 μL
Total 30 μL 10 μL
*The total amount of the template DNA should be at least 500 ng in a 30-L reaction. 1–
2 g of template DNA usually works well
3.5 In Vitro Splicing 1. Incubate the 2× splicing buffer (80 mM Tris–HCl, pH 7.4,
2 M NH4Cl, 20 mM MgCl2, and 0.04% SDS) at 45 °C for
several minutes to dissolve any salt precipitations.
2. Mix with the RNA preparation with an equal volume of 2×
splicing buffer, and incubate at 45 °C for 1 h.
A Stable Reverse Genetics System of Zika Virus Based on a Self-Splicing. . . 221
Components Volume
In vitro spliced or unspliced RNA 2 μL
Primer P-Z-R4.1 (20 μM) 1 μL
RNase-free H2O 9 μL
Total 12 μL
Components Volume
5X M-MLV Buffer 4 μL
dNTP Mix (10 mM each) 1 μL
RNase inhibitor (40 U/μL) 1 μL
RTase M-MLV (RNase H-) (200 U/μL) 1 μL
RNase-free H2O 1 μL
Total (together with the RNA/primer mix) 20 μL
Components Volume
cDNA product 1 μL
Forward primer (20 μM) 1 μL
Reverse primer (20 μM) 1 μL
10× LA Taq Buffer 5 μL
dNTP mixture (2.5 mM each) 8 μL
(continued)
222 Zhong-Yu Liu et al.
Components Volume
LA Taq DNA polymerase (see Note 6) 0.5 μL
Nuclease-free water 33.5 μL
Total 50 μL
3.6 RNA Transfection 1. One day prior to transfection, plate BHK-21 cells into 24-well
plates at a density of 5 × 104 cells/well, and culture at 37 °C
with 5% CO2 for 12–14 h.
2. Perform the transfection by using the Lipofectamine 2000
reagent (Thermo Fisher Scientific). Use 500 ng RNA for each
well of BHK-21 cells. Perform the transfection in triplicates.
3. Dilute 2 μg spliced RNA in 200 μL of Opti-MEM I.
4. Dilute 4 μL Lipofectamine 2000 in 200 μL of Opti-MEM
I. Incubate at room temperature for 5 min.
5. Mix the diluted RNA and Lipofectamine 2000 by pipetting
gently. Do not centrifuge. Incubate at room temperature for
20 min.
6. Add 100 μL RNA/liposome complex to the cells in the 24-well
plate. Incubate for 6 h in a 37 °C CO2 incubator.
7. Discard the medium containing RNA/liposome complex at
6 h post-transfection, and culture the transfected cells in
DMEM with 5% FBS for 2–3 days.
8. Collect the culture supernatants of the transfected cells at 72 h
post-infection, and seed 100 μL onto C6/36 cell monolayer in
a 12-well plate.
9. Continue to culture the blind-infected C6/36 cells at 28 °C for
4–5 days, and then collect the supernatants for virus titer
determination.
3.7 Titration 1. One day prior to plaque assay, seed BHK-21 cells into 12-well
plates at a density of approximately 2.5 × 105 cells/well. Cul-
ture the cells in a CO2 incubator at 37 °C overnight.
2. At the next day, check the confluency of the BHK-21 cells
under microscopy. The confluency should be >90% at the
time of plaque assay. Perform a tenfold serial dilution of the
A Stable Reverse Genetics System of Zika Virus Based on a Self-Splicing. . . 223
3.8 Mutagenesis Mutants targeting the cyclization sequences and 5′-SLA element
Analysis of cis-Acting will be analyzed in this protocol (see Note 11). For replication
RNA Elements Based analysis of (+)ssRNA viruses, a mutant containing an inactive
on pACNR-GZ01- vRNA polymerase usually serves as the negative control group.
Intron-IC 1. Perform the in vitro transcription and in vitro splicing as
described above.
2. Seed BHK-21 cells into 48-well plates at a density of 2 × 104
cells per well.
3. When the cells reach 50% confluency at the next day, perform
the transfection in triplicates using Lipofectamine 2000.
224 Zhong-Yu Liu et al.
3.9 qRT-PCR The qRT-PCR will be performed using the One-Step PrimeScript
RT-PCR Kit (Perfect Real Time) on a LightCycler 2.0 real-time
PCR system.
The primer and probe sequence are listed in Table 2.
Table 2
Primer and probe for qRT-PCR
Components Volume
2× One-Step RT-PCR Buffer III 10 μL
TaKaRa Ex Taq HS (5 U/μL) 0.4 μL
PrimeScript RT Enzyme Mix II 0.4 μL
P-ZIKV-AS-F (20 μM) 0.4 μL
P-ZIKV-AS-R (20 μM) 0.4 μL
ZIKV-AS-Probe (20 μM) 0.4 μL
RNA sample 2 μL
RNase-free water 6 μL
Total 20 μL
Temperature
Cycles/ transition
Steps Temperatures Durations detection rates
Stage 1 ×1
Stage 2 ×40
Stage 3 ×1
3.10 IFA 1. Transfer the desired amounts of sterile cover slips into wells of
24-well plates using a sterile ophthalmic forceps.
2. Seed BHK-21 cells onto the cover slips, and perform RNA
transfection as described in Subheading 3.6.
3. At 24, 48, or 72 h post-transfection, wash the cover slips with
sterile PBS once, and add 1.5 mL fixation solution to each well
containing a cover slip. Fix at -20 °C for 1 h.
4. Aspire the fixation solution completely, and air-dry the fixed
cover slips.
5. Transfer the cover slips to a new 24-well plate. Add 160 μL of
1:2000 diluted mouse anti-ZIKV E mAb, and incubate at 37 °
C for 1 h. Make sure that the antibody work solution covers the
fixed cells completely and evenly.
6. Discard the primary antibody working solution, and wash the
cells with 1 mL of PBS for 5 min on a mini-shaker. Perform the
washing for three times. Air-dry the cover slips.
7. Add 160 μL of 1:200~1:400 diluted goat anti-mouse IgG
conjugated with Alexa Fluor 488. Incubate at 37 °C for 1 h.
Wash the cover slips as described in step 6.
8. Add 200 μL of 1:2000~1:5000 diluted DAPI (using deionized
water), and incubate at room temperature for 5–10 min. Dis-
card the DAPI dilutes, and wash once with 1 mL deionized
water.
9. Check the results using an Olympus BX51 fluorescence
microscope.
4 Notes
References
1. Dick GW, Kitchen SF, Haddow AJ (1952) Zika Decam C, Choumet V, Halstead SK, Willison
virus. I. Isolations and serological specificity. HJ, Musset L, Manuguerra JC, Despres P,
Trans R Soc Trop Med Hyg 46(5):509–520 Fournier E, Mallet HP, Musso D, Fontanet A,
2. Macnamara FN (1954) Zika virus: a report on Neil J, Ghawche F (2016) Guillain-Barre Syn-
three cases of human infection during an epi- drome outbreak associated with Zika virus
demic of jaundice in Nigeria. Trans R Soc Trop infection in French Polynesia: a case-control
Med Hyg 48(2):139–145 study. Lancet 387(10027):1531–1539.
3. Simpson DI (1964) Zika virus infection in https://doi.org/10.1016/S0140-6736(16)
man. Trans R Soc Trop Med Hyg 58:335–338 00562-6
4. Fagbami AH (1979) Zika virus infections in 12. Brasil P, Pereira JP Jr, Moreira ME, Ribeiro
Nigeria: virological and seroepidemiological Nogueira RM, Damasceno L, Wakimoto M,
investigations in Oyo State. J Hyg 83(2): Rabello RS, Valderramos SG, Halai UA, Salles
213–219 TS, Zin AA, Horovitz D, Daltro P, Boechat M,
Raja Gabaglia C, Carvalho de Sequeira P,
5. Olson JG, Ksiazek TG, Suhandiman T (1981) Pilotto JH, Medialdea-Carrera R, Cotrim da
Zika virus, a cause of fever in Central Java, Cunha D, Abreu de Carvalho LM, Pone M,
Indonesia. Trans R Soc Trop Med Hyg 75(3): Machado Siqueira A, Calvet GA, Rodrigues
389–393 Baiao AE, Neves ES, Nassar de Carvalho PR,
6. Duffy MR, Chen TH, Hancock WT, Powers Hasue RH, Marschik PB, Einspieler C,
AM, Kool JL, Lanciotti RS, Pretrick M, Janzen C, Cherry JD, Bispo de Filippis AM,
Marfel M, Holzbauer S, Dubray C, Nielsen-Saines K (2016) Zika virus infection in
Guillaumot L, Griggs A, Bel M, Lambert AJ, pregnant women in Rio de Janeiro. N Engl J
Laven J, Kosoy O, Panella A, Biggerstaff BJ, Med 375(24):2321–2334. https://doi.org/
Fischer M, Hayes EB (2009) Zika virus out- 10.1056/NEJMoa1602412
break on Yap Island, Federated States of Micro- 13. Liu ZY, Qin CF (2020) Structure and function
nesia. N Engl J Med 360(24):2536–2543. of cis-acting RNA elements of flavivirus. Rev
https://doi.org/10.1056/NEJMoa0805715 Med Virol 30(1):e2092. https://doi.org/10.
7. Cao-Lormeau VM, Roche C, Teissier A, 1002/rmv.2092
Robin E, Berry AL, Mallet HP, Sall AA, 14. Villordo SM, Carballeda JM, Filomatori CV,
Musso D (2014) Zika virus, French polynesia, Gamarnik AV (2016) RNA structure duplica-
South pacific, 2013. Emerg Infect Dis 20(6): tions and flavivirus host adaptation. Trends
1085–1086. https://doi.org/10.3201/ Microbiol 24(4):270–283. https://doi.org/
eid2006.140138 10.1016/j.tim.2016.01.002
8. Musso D, Roche C, Robin E, Nhan T, 15. Shan C, Xie X, Muruato AE, Rossi SL, Roundy
Teissier A, Cao-Lormeau VM (2015) Potential CM, Azar SR, Yang Y, Tesh RB, Bourne N,
sexual transmission of Zika virus. Emerg Infect Barrett AD, Vasilakis N, Weaver SC, Shi PY
Dis 21(2):359–361. https://doi.org/10. (2016) An infectious cDNA clone of Zika
3201/eid2102.141363 virus to study viral virulence, mosquito trans-
9. Mlakar J, Korva M, Tul N, Popovic M, Poljsak- mission, and antiviral inhibitors. Cell Host
Prijatelj M, Mraz J, Kolenc M, Resman Rus K, Microbe 19(6):891–900. https://doi.org/10.
Vesnaver Vipotnik T, Fabjan Vodusek V, 1016/j.chom.2016.05.004
Vizjak A, Pizem J, Petrovec M, Avsic Zupanc 16. Schwarz MC, Sourisseau M, Espino MM, Gray
T (2016) Zika virus associated with microceph- ES, Chambers MT, Tortorella D, Evans MJ
aly. N Engl J Med 374(10):951–958. https:// (2016) Rescue of the 1947 Zika virus proto-
doi.org/10.1056/NEJMoa1600651 type strain with a cytomegalovirus promoter-
10. Oehler E, Watrin L, Larre P, Leparc-Goffart I, driven cDNA clone. mSphere 1(5):e00246–
Lastere S, Valour F, Baudouin L, Mallet H, e00216. https://doi.org/10.1128/mSphere.
Musso D, Ghawche F (2014) Zika virus infec- 00246-16
tion complicated by Guillain-Barre syndrome – 17. Widman DG, Young E, Yount BL, Plante KS,
case report, French Polynesia, December 2013. Gallichotte EN, Carbaugh DL, Peck KM,
Euro Surv Bulletin Europeen sur les maladies Plante J, Swanstrom J, Heise MT, Lazear
transmissibles = Eur Commun Dis Bull 19(9): HM, Baric RS (2017) A reverse genetics plat-
20720 form that spans the Zika virus family tree.
11. Cao-Lormeau VM, Blake A, Mons S, Lastere S, MBio 8(2):e02014–e02016. https://doi.org/
Roche C, Vanhomwegen J, Dub T, 10.1128/mBio.02014-16
Baudouin L, Teissier A, Larre P, Vial AL,
A Stable Reverse Genetics System of Zika Virus Based on a Self-Splicing. . . 229
Abstract
Dengue virus (DENV) is one of the most important and widespread arthropod-borne viruses, causing
millions of infections over the years. Considering its epidemiological importance, efforts have been directed
towards understanding various aspects of DENV biology, which have been facilitated by the development of
different molecular strategies for engineering viral genomes, such as reverse genetics approaches. Reverse
genetic systems are a powerful tool for investigating virus–host interaction, for vaccine development, and
for high-throughput screening of antiviral compounds. However, stable manipulation of DENV genomes is
a major molecular challenge, especially when using conventional cloning systems. To circumvent this issue,
we describe a simple and efficient yeast-based reverse genetics system to recover infectious DENV clones.
Key words Dengue, Reverse genetics, Infectious clone, Homologous recombination, Yeast, Shuttle
vector
1 Introduction
Daniel R. Pérez (ed.), Reverse Genetics of RNA Viruses: Methods and Protocols, Methods in Molecular Biology, vol. 2733,
https://doi.org/10.1007/978-1-0716-3533-9_14,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2024
231
232 José Valter Joaquim Silva Júnior et al.
2 Materials
2.3 Vector 1. Shuttle vector pSVJS01 [16], which carries the TRP1 gene
Preparation involved in the tryptophan biosynthesis pathway. TRP1 is
used as an auxotrophic marker for selection in trp1-Δ1yeast
strains (see Note 2).
2. NotI restriction enzyme (New England Biolabs).
3. Antarctic Phosphatase (New England Biolabs).
4. Agarose, molecular biology grade.
5. QIAquick Gel Extraction Kit (QIAGEN).
6. 2-Propanol (isopropanol), molecular biology grade.
7. Water bath or thermal block at 37 and 50 °C.
2.6 Full-Length PCR 1. Platinum SuperFi DNA Polymerase (Thermo Fisher Scientific).
and In Vitro 2. UltraPure phenol–chloroform–isoamyl alcohol (25:24:1, v/v)
Transcription (Thermo Fisher Scientific).
3. Chloroform–isoamyl alcohol 24:1 (Sigma-Aldrich).
4. 3 M sodium acetate, pH 5.2.
5. Pellet Paint Co-Precipitant (Millipore).
6. Ethanol, molecular biology grade.
7. Cap Analog [m7G(5′)ppp(5′)G] (Thermo Fisher Scientific).
8. MEGAscript T7 Transcription Kit (Thermo Fisher Scientific).
3 Methods
3.1 DENV RNA 1. Pipette 560 μL buffer AVL into a 1.5 mL microcentrifuge tube
Extraction (nuclease-free), and add 5.6 μL carrier RNA.
2. Add 140 μL of DENV-infected cell culture supernatant, and
mix by pulse-vortexing for 15 s.
3. Incubate at room temperature for 10 min.
4. Briefly centrifuge the microtube to remove drops from the lid.
5. Add 560 μL ethanol (96–100%) to the microtube, and mix by
pulse-vortexing (15 s). After, briefly centrifuge the tube to
remove drops from the lid.
6. Carefully apply 630 μL of the solution from the above step to
the QIAamp Mini Column (in a 2 mL vial collection tube).
Close the lid, and centrifuge at 6,000× g (8,000 rpm) for
1 min. Place the QIAamp Mini Column in a clean collection
tube, and discard the tube containing the filtrate.
7. Carefully open the QIAamp Mini Column, and repeat step 6
until all the lysate has been loaded onto the spin column.
8. Carefully open the QIAamp Mini Column, and add 500 μL
buffer AW1. Close the cap, and centrifuge at 6,000×
g (8,000 rpm) for 1 min. Discard the tube containing the
filtrate.
9. Carefully open the QIAamp Mini Column, and add 500 μL
buffer AW2. Close the lid, and centrifuge at a maximum speed
(20,000× g; 14,000 rpm) for 3 min.
10. It is recommended to place the QIAamp Mini Column in a
new 2 mL collection tube and centrifuge at full speed for
1 min.
11. Place the QIAamp Mini Column in a clean 1.5 mL microcen-
trifuge tube (nuclease-free), and add 60 μL AVE equilibrated
to room temperature. Close the lid, and incubate at room
temperature for 1 min.
12. Centrifuge at 6,000× g (8,000 rpm) for 1 min.
13. Viral RNA is stable for 1 year when stored at -80 °C.
3.2 Amplification of 1. After extracting virus RNA, amplify the viral genome into five
Genomic Fragments by complementary and partially homologous genomic fragments
RT-PCR using the primers shown in Table 1 (see Note 4).
2. Perform RT-PCR reactions using the SuperScript III One-Step
RT-PCR System with Platinum Taq High-Fidelity DNA
Polymerase.
(i) Set up the thermocycler with the following conditions:
1 cycle at 55 °C for 30 min and 94 °C for 2 min (for cDNA
Table 1
Primers to assemble the full-length infectious clone of DENV-2 by homologous recombination in yeast
Primer annealing
Genomic Amplicon
fragment Primer Sequence (5′-3′) size (bp) Position (nt)a Genomic region
F1 pSVJS01-F cggtccgtaatacgactcactataGAGTTGTTAR 2070 1–23 5′UTR
TCTACGTGGACCGA
DENV2-2045-R TCTGCTTCTATGTTGACTGGG 2025–2045 E
F2 DENV2-1810-F CTACAGCTCAAAGGAATGTCAT 1762 1810–1831 E
DENV2-3571-R CATGTTTCGTTCCTACTCGGGTCC 3548–3571 NS2A
F3 DENV2-3502-F TCACTAGGAGTCTTGGGAATGGC 2078 3502–3524 NS2A
DENV2-5579-R TCCGTGACCCATTCATGTCC 5560–5579 NS3
F4 DENV2-5492-F CATTTCCTCAGAGCAATGCACCAATC 1879 5492–5517 NS3
DENV2-7370-R AATACTTGAGTCACGCAGAGG 7350–7370 NS4B
F5 DENV2-7191-F GCCAGGACTTCAAGCAAAAGC 3560 7191–7211 NS4B
pSVJS01-R ttcaacatttccgtgtcgcgcggccgcAG 10,697– 3′UTR
AACCRGTTGATTCAACAGCACCATT 10,723
a
Positions referring to isolate 3311/BR-PE/95 (DENV-2) (GenBank access JX669481). The RsrII restriction site is in bold. The T7 RNA polymerase promoter is in italics. A
single G to start transcription is underlined. The homology region with the NotI-linearized pSVJS01 vector is in lowercase. E envelope, F fragment, NS nonstructural, UTR
untranslated region
Reverse Genetics of Dengue Virus
237
238 José Valter Joaquim Silva Júnior et al.
3.5 Assembly of the 1. Seed S. cerevisiae YPH252 onto YPD agar plate, and incubate
Full-Length cDNA at 30 °C until individual colonies grow (2–3 days).
Clone in a Single-Step 2. Select a colony, and inoculate into 5 mL of YPD.
Cloning (Fig. 1)
3. Grow the yeast at 30 °C for 16–18 h at 130 rpm.
4. Count the yeast cells in a Neubauer chamber, and prepare a
20 mL suspension at a concentration of 5 × 106 cells/mL. Scale
up suspension volume based on the number of transformations
(see Note 6).
5. Grow the suspension described above to a concentration of 107
cells/mL (approximately for 2–4 h).
6. Centrifuge the cells at 522× g (or 1,500 rpm) for 10 min at 4 °
C.
7. Wash the cell pellet with 50 mL of deionized water.
8. Centrifuge the cells at 522× g (or 1,500 rpm) for 10 min at 4 °
C.
9. Resuspend the pellet with 20 mL of DTT (25 mM), and
incubate for 10 min at room temperature.
10. Centrifuge the cells at 522× g (or 1,500 rpm) at 4 °C.
11. Wash the pellet twice with 20 mL of EPB.
240 José Valter Joaquim Silva Júnior et al.
Fig. 1 Assembly of the full-length infectious clone of DENV-2 by homologous recombination in yeast. The
complete genome of DENV-2 can be assembled into the pSVJS01 vector in a single-step cloning by
homologous recombination of five complementary and partially homologous genomic fragments. The dotted
area corresponds to the region of overlap (homology) between fragment–fragment and fragment–vector. The
numbers refer to the initial and final nucleotides of each fragment, according to their position in the complete
genome (GeneBank access JX669481). The T7 RNA polymerase promoter, F fragment, UTR untranslated
region, C capsid, prM pre-membrane, E envelope, NS nonstructural. *Region of homology between F1-vector
and F5-vector inserted by primers pSVJS01-F and pSVJS01-R, respectively
3.6 Yeast Plasmid 1. Pick individual colonies into 10 mL of YNB broth containing
Isolation (for Cloning yeast synthetic dropout medium supplement (without
Screening) tryptophan).
2. Grow the yeast at 30 °C for 16–18 h at 130 rpm.
3. Centrifuge the cells at 2,000× g (3,000 rpm) for 5 min at 4 °C.
4. Wash the pellet twice in 20 mL of deionized water (centrifuge
at 2,000× g or 3,000 rpm for 5 min).
5. Resuspend the pellet in 400 μL buffer SCE, and transfer the
suspension to a clean 1.5 mL microfuge tube (nuclease-free).
6. Add 30 U of Zymolyase (Zymo Research), and incubate the
mixture at 37 °C for 1 h.
7. Centrifuge at 17,900×g (13,000 rpm) for 10 min.
8. Resuspend the pellet in 250 μL buffer P1 containing RNAse A.
9. Add 250 μL buffer P2, mix well by inverting vigorously four to
six times, and incubate at room temperature for up to 5 min.
10. Add 350 μL buffer N3, and mix immediately and thoroughly
by inverting the tube four to six times.
11. Centrifuge for 10 min at 17,900× g (13,000 rpm) in a
microcentrifuge.
12. Apply 800 μL supernatant from the step above to the QIAprep
2.0 spin column.
13. Centrifuge for 1 min at 17,900× g (13,000 rpm), and discard
the flow-through.
14. Wash the spin column by adding 0.5 mL buffer PB. Centrifuge
for 1 min at 17,900× g (13,000 rpm), and discard the flow-
through.
15. Wash the spin column by adding 0.75 mL buffer PE. Centri-
fuge for 1 min at 17,900× g (13,000 rpm), and discard the
flow-through.
16. Centrifuge for 1 min at 17,900× g (13,000 rpm) to remove
residual wash buffer.
17. Place the column in a clean 1.5 mL microcentrifuge tube
(nuclease-free).
18. To elute DNA, add 50 μL buffer EB (10 mM Tris-Cl, pH 8.5)
or water to the center of column, let stand for 1 min, and
centrifuge for 1 min at 17,900× g (13,000 rpm).
19. Use the eluted DNA for cloning screening.
(a) Confirm cloning by amplifying adjacent fragments
together, for instance, using primers pSVJS01-F and
DENV2-3571-R to amplify the F1-F2 cassette (Table 1).
Furthermore, full genome sequencing can be performed
from eluted DNA to confirm the identity of the assembled
amplicons and the lack of unwanted mutations.
242 José Valter Joaquim Silva Júnior et al.
3.7 Yeast Plasmid 1. Inoculate selected clones into 20 mL of YNB broth containing
Isolation (for Full- yeast synthetic dropout medium supplement (without
Length PCR Template) tryptophan).
2. Incubate the yeast at 30 °C and 130 rpm for 16–18 h.
3. Inoculate all overnight cultured yeasts into 500 mL of YNB
broth containing yeast synthetic dropout medium supplement
(without tryptophan).
4. Incubate the yeast at 30 °C and 130 rpm for 48 h.
5. Centrifuge the cells at 6,000× g for 15 min at 4 °C.
6. Wash the pellet twice in 50 mL of deionized water (centrifuge
at 6,000× g for 15 min at 4 °C).
7. Resuspend the pellet in 4 mL buffer SCE.
8. Add 200 U of Zymolyase, and incubate the mixture at 37 °C
for 2 h.
9. Centrifuge at 6,000× g for 15 min at 4 °C.
10. Resuspend the pellet in 10 mL buffer P1 containing RNAse A.
11. Add 10 mL buffer P2, mix thoroughly by vigorously inverting
four to six times, and incubate at room temperature for up to
5 min.
12. Add 10 mL chilled buffer P3, mix immediately and thoroughly
by vigorously inverting four to six times, and incubate on ice
for 20 min.
13. Centrifuge at ≥20,000× g for 30 min at 4 °C.
14. Promptly remove supernatant containing plasmid DNA.
15. Centrifuge the supernatant again at ≥20,000× g for 15 min at
4 °C. Again, remove supernatant containing plasmid DNA
promptly.
16. Equilibrate a QIAGEN-tip 100 by applying 4 mL buffer QBT,
and allow the column to empty by gravity flow.
17. Apply the supernatant from step 15 to the QIAGEN-tip, and
allow it to enter by gravity flow.
18. Wash the QIAGEN-tip with 2 × 10 mL buffer QC.
19. Add 15 mL buffer QF, and allow it to elute by gravity.
20. Precipitate DNA by adding 10.5 mL room-temperature iso-
propanol to the eluted DNA. Mix and centrifuge immediately
at ≥15,000× g for 30 min at 4 °C. Carefully decant the
supernatant.
21. Wash DNA pellet with 5 mL of room-temperature 70% etha-
nol, and centrifuge at ≥15,000× g for 10 min. Carefully decant
the supernatant without disturbing the pellet.
22. Air-dry the pellet for 5–10 min, and redissolve the DNA in a
suitable volume of buffer (TE buffer (pH 8.0) or 10 mM Tris-
Cl (pH 8.5)).
Reverse Genetics of Dengue Virus 243
3.8 Full-Length PCR 1. Thaw, mix, and briefly centrifuge PCR reagents.
2. Prepare the master mix: 10 μL of 5X SuperFiTM Buffer, 1 μL of
10 mM dNTPs mix, 0.5 μL of PlatinumTM SuperFiTM DNA
Polymerase, and water (nuclease-free) q.s.p 50 μL (here, con-
sider additional volume of primers and template).
3. Add 2.5 μL of primer pSVJS01-F (Table 1) and 2.5 μL of
primer pSVJS01-R2 (5′- AGAACCRGTTGATTCAACAG
CACCATT-3′) (both at 10 μM) and 200 ng of template.
4. Set up the thermocycler with the following conditions: 95 °C
for 2 min (initial denaturation), 35 cycles of 95 °C for 10 s
(denaturation), 56 °C for 10 s (annealing), 68 °C for 7 min
(extension), and final extension at 68 °C for 5 min. Proceed
with the PCR reaction.
5. Analyze amplicon on 1% agarose gel.
3.9 Purification and 1. Measure full-length PCR product volume, and complete to
Precipitation of the 200 μL with nuclease-free water.
Full Amplicon 2. Add 200 μL of UltraPure phenol–chloroform–isoamyl alcohol
(25:24:1), and vortex for 10 s.
3. Centrifuge for 5 min at 17,900× g (13,000 rpm).
4. Carefully collect the upper phase, and measure its volume.
5. Add the same volume of chloroform–isoamyl alcohol (24:1),
and vortex for 10 s.
6. Centrifuge for 5 min at 17,900× g (13,000 rpm).
7. Carefully collect the upper phase, and measure its volume.
8. Add 1/10 of the volume of 3 M sodium acetate, 2 μL of Pellet
Paint Co-Precipitant, and 2 volumes of ethanol.
9. Vortex and incubate for 30 min at -20 °C.
10. Wash the pellet twice with 80% ethanol.
11. Centrifuge for 5 min at 17,900× g (13,000 rpm).
12. Carefully remove all ethanol with a pipette.
13. Air-dry pellet at 37 °C for 5 min.
14. Proceed with in vitro transcription.
3.10 In Vitro 1. Perform in vitro transcription with the DNA from Subheading
Transcription 3.9, step 13.
2. Resuspend the DNA pellet in 1.25 μL of nuclease-free water
(see Note 8).
3. Add in vitro transcription mix: 0.5 μL of 10X Reaction Buffer,
2 μL of rNTPs (75 mM ATP, CTP, UTP, and 15 mM GTP),
0.75 μL of cap analog [m7G(5′)ppp(5′)G] (40 mM), and
0.5 μL of enzyme mix.
244 José Valter Joaquim Silva Júnior et al.
3.11 Cell 1. Split BHK-21 cells the day before transfection, and maintain
Transfection the cell under routine culture conditions (e.g., in MEM sup-
plemented with 10% heat-inactivated FBS, at 37 °C and 5%
CO2).
2. On the day of transfection, harvest BHK-21 cells, and prepare a
suspension with 2 × 106 cells per transfection.
3. Wash the cell suspension twice, using ice-cold PBS or pure
MEM (i.e., without antibiotic and FBS).
4. For washes, centrifuge the cells at 334× g (1,200 rpm) for
5 min at 4 °C.
5. After the last wash, resuspend cells in 100 μL ice-cold cytomix
solution.
6. Add 5 μg of in vitro transcribed RNA. As a negative control,
transfect BHK-21 under the same conditions without RNA.
7. Transfer the transfection mixture into a 2 mm cuvette.
8. Proceed with transfection using the following electroporation
settings: 2 pulses of 100 μs, 1,200 V, and 1 s interval (see Note
10).
9. After electroporation, allow cells to recover for 10 min at room
temperature, then resuspend in complete growth medium, and
seed as recommended below:
(i) Plate approximately 104 cells on NuncTM Lab-TekTM
chamber slide to monitor viral rescue by IFA as described
in the next section.
(ii) Seed the rest of the cells into 25 cm2 culture flasks for virus
rescue amplification.
3.12 Viral Rescue 1. Three days after transfection, proceed with IFA of the seeded
Monitoring by Indirect cells on NuncTM Lab-TekTM chamber slide.
Immunofluorescence (a) Include negative control IFA for cell background
differentiation.
(b) Include DENV-infected BHK-21 as a positive control.
2. Fix cells with cold acetone for 5 min at 4 °C and air-dry.
3. Incubate cells with the monoclonal antibody D1-4G2-4-15 (1:
100 dilution in PBS) as the primary antibody for 1 h at 37 °C,
using a humid chamber.
4. Wash the slides twice with PBS (pH 7.4) using a Coplin jar. It is
recommended to let the slide rest in PBS for 10 min with
each wash.
Reverse Genetics of Dengue Virus 245
4 Notes
References
1. Harapan H, Michie A, Sasmono RT, Imrie A Baker TS, Strauss JH (2002) Structure of den-
(2020) Dengue: a minireview. Viruses 12(8): gue virus: implications for flavivirus organiza-
829. https://doi.org/10.3390/v12080829 tion, maturation, and fusion. Cell 108(5):
2. International Committee on Taxonomy of 717–725. https://doi.org/10.1016/s0092-
Viruses-ICTV (2022) https://ictv.global/ 8674(02)00660-8
taxonomy/ 9. Gebhard LG, Filomatori CV, Gamarnik AV
3. Diamond MS, Pierson TC (2015) Molecular (2011) Functional RNA elements in the den-
insight into dengue virus pathogenesis and its gue virus genome. Viruses 3:1739–1756.
implications for disease control. Cell 162(3): https://doi.org/10.3390/v3091739
488–492. https://doi.org/10.1016/j.cell. 10. Izmirly AM, Alturki SO, Alturki SO,
2015.07.005 Connors J, Haddad EK (2020) Challenges in
4. World Health Organization- WHO (2022) dengue vaccines development: pre-existing
https://www.who.int/news-room/fact- infections and cross-reactivity. Front Immunol
sheets/detail/dengue-and-severe-dengue 11:1055. https://doi.org/10.3389/fimmu.
5. Bhatt P, Sabeena SP, Varma M, Arunkumar G 2020.01055
(2021) Current understanding of the patho- 11. Wang W-H, Urbina AN, Lin C-Y, Yang Z-S,
genesis of dengue virus infection. Curr Micro- Assavalapsakul W, Thitithanyanont A,
biol 78(1):17–32. https://doi.org/10.1007/ Po-Liang L, Chen Y-H, Wang S-F (2021) Tar-
s00284-020-02284-w gets and strategies for vaccine development
6. Bell SM, Katzelnick L, Bedford T (2019) Den- against dengue viruses. Biomed Pharmacother
gue genetic divergence generates within- 144:112304. https://doi.org/10.1016/j.bio
serotype antigenic variation, but serotypes pha.2021.112304
dominate evolutionary dynamics. elife 8: 12. Ward R, Davidson AD (2008) Reverse genetics
e42496. https://doi.org/10.7554/eLife. and the study of dengue virus. Futur Virol
42496 3(3):279–290. https://doi.org/10.2217/
7. Halstead SB (1979) In vivo enhancement of 17460794.3.3.279
dengue virus infection in rhesus monkeys by 13. Perez DR (2017) Preface. In: Perez DR
passively transferred antibody. J Infect Dis (ed) Reverse genetics of RNA viruses: methods
140:527–533. https://doi.org/10.1093/ and protocols, Methods in molecular biology,
infdis/140.4.527 vol 1602. © Springer Science+Business Media
8. Kuhn RJ, Zhang W, Rossmann MG, Pletnev LLC. https://doi.org/10.1007/978-1-4939-
SV, Corver J, Lenches E, Jones CT, 6964-7_3
Mukhopadhyay S, Chipman PR, Strauss EG,
Reverse Genetics of Dengue Virus 247
14. Bhat AI, Rao GP (2020) Development of number plasmid. J Virol Methods 92(1):
infectious clone of virus. In: Characterization 71–82. https://doi.org/10.1016/s0166-
of plant viruses, Springer protocols handbooks. 0934(00)00277-9
Humana, New York. https://doi.org/10. 24. Suzuki R, de Borba L, Duarte dos Santos CN,
1007/978-1-0716-0334-5_46 Mason PW (2007) Construction of an infec-
15. Stobart CC, Moore M (2014) RNA virus tious cDNA clone for a Brazilian prototype
reverse genetics and vaccine design. Viruses strain of dengue virus type 1: characterization
6(7):2531–2550. https://doi.org/10.3390/ of a temperature-sensitive mutation in NS1.
v6072531 Virology 362(2):374–383. https://doi.org/
16. Santos JJ, Cordeiro MT, Bertani GR, Marques 10.1016/j.virol.2006.11.026
ET, Gil LH (2013) Construction and charac- 25. Kapoor M, Zhang L, Mohan PM, Padmanab-
terization of a complete reverse genetics system han R (1995) Synthesis and characterization of
of dengue virus type 3. Mem Inst Oswaldo an infectious dengue virus type-2 RNA
Cruz 108(8):983–991. https://doi.org/10. genome (New Guinea C strain). Gene 162(2):
1590/0074-0276130298 175–180. https://doi.org/10.1016/0378-
17. Santos JJ, Cordeiro MT, Bertani GR, Marques 1119(95)00332-z
ET, Gil LH (2014) A two-plasmid strategy for 26. Tamura T, Zhang J, Madan V, Biswas A,
engineering a dengue virus type 3 infectious Schwoerer MP, Cafiero TR, Heller BL,
clone from primary Brazilian isolate. An Acad Wang W, Ploss A (2022) Generation and char-
Bras Cienc 86(4):1749–1759. https://doi. acterization of genetically and antigenically
org/10.1590/0001-3765201420130332 diverse infectious clones of dengue virus sero-
18. Santos JJ, Magalhães T, Silva Junior JV, Silva types 1-4 Emerg. Microbes Infect 11(1):
AN, Cordeiro MT, Gil LH (2015) Full-length 2 2 7 – 2 3 9 . h t t p s : // d o i . o r g / 1 0 . 1 0 8 0 /
infectious clone of a low passage dengue virus 22221751.2021.2021808
serotype 2 from Brazil. Mem Inst Oswaldo 27. Blaney JE Jr, Hanson CT, Firestone CY, Han-
Cruz 110(5):677–683. https://doi.org/10. ley KA, Murphy BR, Whitehead SS (2004)
1590/0074-02760150053 Genetically modified, live attenuated dengue
19. Usme-Ciro JA, Lopera JA, Enjuanes L, virus type 3 vaccine candidates. Am J Trop
Almazán F, Gallego-Gomez JC (2014) Devel- Med Hyg 71(6):811–821
opment of a novel DNA-launched dengue 28. Gebhard LG, Iglesias NG, Byk LA, Filomatori
virus type 2 infectious clone assembled in a CV, De Maio FA, Gamarnik AV (2016) A
bacterial artificial chromosome. Virus Res proline-rich N-terminal region of the dengue
180:12–22. https://doi.org/10.1016/j. virus NS3 is crucial for infectious particle pro-
virusres.2013.12.001 duction. J Virol 90(11):5451–5461. https://
20. Pu SY, Wu RH, Yang CC, Jao TM, Tsai MH, doi.org/10.1128/JVI.00206-16
Wang JC, Lin HM, Chao YS, Yueh A (2011) 29. Matusan AE, Pryor MJ, Davidson AD, Wright
Successful propagation of flavivirus infectious PJ (2001) Mutagenesis of the Dengue virus
cDNAs by a novel method to reduce the cryp- type 2 NS3 protein within and outside helicase
tic bacterial promoter activity of virus genomes. motifs: effects on enzyme activity and virus
J Virol 85(6):2927–2941. https://doi.org/10. replication. J Virol 75(20):9633–9643.
1128/JVI.01986-10 h t t p s : // d o i . o r g / 1 0 . 1 1 2 8 / J V I . 7 5 . 2 0 .
21. Aubry F, Nougairède A, Gould EA, de Lam- 9633-9643.2001
ballerie X (2015) Flavivirus reverse genetic sys- 30. Silveira GF, Strottmann DM, de Borba L,
tems, construction techniques and Mansur DS, Zanchin NI, Bordignon J, dos
applications: a historical perspective. Antivir Santos CN (2016) Single point mutations in
Res 114:67–85. https://doi.org/10.1016/j. the helicase domain of the NS3 protein
antiviral.2014.12.007 enhance dengue virus replicative capacity in
22. Polo S, Ketner G, Levis R, Falgout B (1997) human monocyte-derived dendritic cells and
Infectious RNA transcripts from full-length circumvent the type I interferon response.
dengue virus type 2 cDNA clones made in Clin Exp Immunol 183(1):114–128. https://
yeast. J Virol 71(7):5366–5374. https://doi. doi.org/10.1111/cei.12701
org/10.1128/JVI.71.7.5366-5374.1997 31. Duan X, Lu X, Li J, Liu Y (2008) Novel bind-
23. Sriburi R, Keelapang P, Duangchinda T, ing between pre-membrane protein and vacuo-
Pruksakorn S, Maneekarn N, Malasit P, Sitti- lar ATPase is required for efficient dengue virus
sombut N (2001) Construction of infectious secretion. Biochem Biophys Res Commun
dengue 2 virus cDNA clones using high copy
248 José Valter Joaquim Silva Júnior et al.
Abstract
Rotaviruses are the primary cause of severe gastroenteritis in infants and young children throughout the
world. To combat rotavirus illness, several live oral vaccines have been developed, or are under develop-
ment, that are formulated from attenuated human or human–animal reassortant strains of rotavirus. While
the effectiveness of these vaccines is generally high in developed countries, the same vaccines are signifi-
cantly less effective in many developing countries, where the need for rotavirus vaccines is greatest.
Recently, reverse genetics systems have been developed that allow modification of the segmented double-
stranded (ds)RNA genome of rotavirus, including reprogramming the genome to allow expression of
additional proteins that may stimulate expanded neutralizing antibody responses in vaccinated children.
The use of reverse genetics systems may not only lead to the development of more potent classes of vaccines
but can be used to better explore the intricacies of rotavirus molecular biology and pathogenesis. In this
article, we share protocols that can be used to generate recombinant rotaviruses, including modified strains
that express foreign proteins.
Key words Rotavirus, Reverse genetics, Double-stranded RNA, dsRNA, Segmented genome,
Gastroenteritis
1 Introduction
Daniel R. Pérez (ed.), Reverse Genetics of RNA Viruses: Methods and Protocols, Methods in Molecular Biology, vol. 2733,
https://doi.org/10.1007/978-1-0716-3533-9_15,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2024
249
250 Chantal A. Agbemabiese et al.
Fig. 1 Rotavirus reverse genetics system. (a) T7 (pT7) plasmids used in generating recombinant rotaviruses
contain a full-length cDNA of a rotavirus genome segment, including its 5′- and 3′-untranslated region (UTR)
and open reading frame. Positioned upstream of the cDNA is a T7 promoter (prm) sequence, and downstream
is an HDV ribozyme (Rz) sequence and a T7 termination (term) signal. The T7 transcriptional start site and HDV
ribozyme cleavage site are indicated, along with origin of plasmid replication (Ori) and ampicillin resistance
gene (AmpR). (b) Recombinant rotavirus is prepared by transfecting BHK-T7 cells with the pT7 plasmids, along
with a plasmid expressing the pCMV-NP868R capping enzyme. The BHK-T7 cells are overseeded 3 days post-
transfection with MA104 cells to facilitate the spread and amplification of recombinant rotavirus. Five days
Rotavirus Reverse Genetics 253
2 Materials
2.1 Cell Lines 1. Baby hamster kidney (BHK-21) cells constitutively expressing
bacteriophage T7 RNA polymerase (BHK-T7) can be obtained
from Dr. Ulla Buchholz at the Laboratory of Infectious Dis-
eases, NIAID, NIH.
2. African green monkey kidney MA104 cells (American Type
Culture Collection).
2.2 Cell Culture 1. Cell culture medium is prepared under sterile conditions in a
Media biological safety cabinet and stored at 4 °C.
2. Prepare GMEM incomplete medium by combining 500 mL of
Glasgow minimal essential medium (MEM) with 50 mL of
tryptose–phosphate broth (TPB) (Gibco), 5 mL of 100X non-
essential amino acids (NEAA) (Gibco), 5 mL of 100X
L-glutamine (Gibco), and 5 mL of 100X penicillin–streptomy-
cin solution (Corning).
3. Prepare GMEM complete medium by adding 25 mL of heat-
inactivated fetal bovine serum to 500 mL of GMEM incom-
plete medium. To supplement with Geneticin, add 10 mL of a
50 mg/mL stock of Geneticin G418 (Gibco) to 500 mL of
GMEM complete medium.
Fig. 1 (continued) after overseeding, recombinant virus in cell lysates is amplified on MA104 cells and the
dsRNA genome of the viruses analyzed by RNA gel electrophoresis and sequencing. Subsequently, the
recombinant virus is plaque isolated and further amplified on MA104 cells. (c) Schematics of T7 plasmids
used to recover recombinant wild-type SA11 (pT7/SA11-NSP3-wt) and mutant form of the virus expressing
NSP3 fused to FLAG-tagged (3X FL) UnaG fluorescent protein (pT7/SA11-NSP3-fUnaG). The NSP3 +RNA
produced from pT7/SA11-NSP3-wt is 1.1 kb, while the modified NSP3 +RNA produced is 1.6 kb
254 Chantal A. Agbemabiese et al.
Fig. 2 Analysis of recombinant SA11 rotaviruses. (a) Electrophoretic profiles of the genomes of wild-type
recombinant SA11 rotavirus (rSA11-wt) and rSA11 with a modified segment 7 (NSP3) RNA expressing FLAG-
tagged UnaG (rSA11-NSP3-fUnaG). The RNAs were recovered by TRIzol extraction, analyzed by electrophore-
sis on a 10% polyacrylamide gel, and detected by ethidium–bromide staining. The 11 genome segments are
numbered with the position of the segment 7 RNAs indicated with an arrowhead. (b) Detection of UnaG
fluorescence by rSA11-NSP3-fUnaG-infected cells at 8 h post-infection using a Zoe fluorescent digital cell
imaging system (Bio-Rad), at 20× magnification (Scale bar = 100 μm). (c) Plaques produced on MA104 cell
monolayers (4 days post-infection) by rSA11-wt and rSA11-NSP3-fUnaG viruses and detected by neutral red
Rotavirus Reverse Genetics 255
2.3 Reverse Genetics 1. The 11 pT7 plasmids expressing rotavirus SA11 (+)RNA
Plasmids (pT7/VP1SA11, pT7/VP2SA11, pT7/VP3SA11,
pT7/VP4SA11, pT7/VP6SA11, pT7/VP7SA11,
pT7/NSP1SA11, pT7/NSP2SA11, pT7/NSP3SA11,
pT7/NSP4SA11, and pT7/NSP5SA11) can be obtained
from Addgene [19] or commercially synthesized (see Note 1).
{https://www.addgene.org/browse/article/25158/}
2. The plasmid pCMV/NP868R expressing the African swine
fever virus (ASFV) capping enzyme (GenBank MH212166)
can be synthesized commercially or obtained from the
authors [20].
3. Modified pT7/NSP3SA11 plasmids that express NSP3 fused
to FLAG-tagged UnaG (MK868472), or NSP3 and FLAG-
tagged UnaG (MK851042), as separate protein products, can
be synthesized commercially or obtained from the authors.
Modified pT7/NSP3SA11 plasmids expressing NSP3 and
other fluorescent reporter proteins (e.g., mRUBY,
mCHERRY, etc.), as separate products, are described by Philip
et al. [22, 23]. Sequences of modified pT7 plasmids should be
confirmed prior to use in reverse genetics experiments.
4. Plasmids are prepared using endo-free Qiagen or equivalent
plasmid purification kits, diluted to final concentrations of
1 mg/mL in ultrapure water, and stored at -20 °C in 5 μL
aliquots in 0.2 mL microfuge tubes.
ä
Fig. 2 (continued) staining. (d) Detection of VP6 and NSP3 in rSA11-wt-infected MA104 cells and VP6 and the
fused product NSP3-3XFLAG-UnaG in rSA11-NSP3-fUnaG-infected MA104 cells by immunoblot analysis with
anti-VP6, anti-NSP3, and anti-FLAG antibodies [21]. The positions of molecular weight markers (MWM) are
indicated
256 Chantal A. Agbemabiese et al.
3 Methods
3.1 Maintaining Cell 1. BHK-T7 cells are propagated in T75 flasks containing 15 mL
Lines of GMEM complete media. Geneticin is included in the cell
culture media with every other passage. To passage BHK-T7
cells, monolayers are rinsed with PBS, disrupted using trypsin–
EDTA solution, and resuspended in 5 mL of complete
GMEM/FBS. Four drops of resuspended cells are placed in
fresh T75 flasks containing 15 mL of complete GMEM/FBS.
The cells typically reach confluency in 3 days. BHK-T7 cells
beyond passage 20 are not used in reverse genetics experiments
(see Note 2).
2. MA104 cells are propagated in T75 flasks containing 15 mL of
DMEM complete medium. To passage MA104 cells, mono-
layers are rinsed with PBS, disrupted using trypsin–EDTA
solution, and resuspended in 5 mL of DMEM complete
medium. One mL of resuspended MA104 cells is added to a
T75 flask containing 15 mol of DMEM complete medium. The
newly seeded cells will reach confluency in 4–5 days. MA104
cells beyond passage 25 are not used in reverse genetics
experiments.
3. Cell lines are grown in humidified 37 °C incubators in the
presence of 5% CO2 (see Note 3).
3.3 Detection of 1. To recover viral RNA from clarified MA104 cell lysates, com-
Recombinant bine 600 μL volumes of lysates with 400 μL of TRIzol reagent
Rotavirus RNA by Gel (Invitrogen) in 1.5 mL microfuge tubes, vortex the mixtures
Electrophoresis for 30 s, and then incubate at room temperature for 5 min.
2. Add 200 μL of chloroform to the TRIzol-treated mixtures,
vortex for 30 s, and then incubate for 3 min at room
temperature.
3. Centrifuge mixtures at 12,000× g for 15 min at 4 °C, and
transfer the upper aqueous phase into new microfuge tubes.
Add 2 volumes of cold isopropanol to the aqueous phase, mix
by inverting the tubes six to eight times, and incubate the
mixtures at room temperature for 10 min.
4. Centrifuge mixtures for 15 min at 12,000× g to pellet viral
RNA. Look for a white pellet at the bottom of the microfuge
tube, and carefully discard the supernatant.
5. Rinse RNA pellets by adding 500 μL of 75% ethanol to the
tubes, gently inverting the tubes a single time, followed by
centrifugation at 7500× g for 5 min at 4 °C. Discard the
supernatant, and air-dry the pellet at room temperature for
5–10 min.
6. Thoroughly dissolve the RNA pellet in 20 μL of nuclease-free
water, and store at -20 °C.
7. Mix 10 μL of extracted RNA with 2 μL of 6X DNA loading
buffer (Thermo Scientific). Carefully load the mixture onto
10% pre-cast polyacrylamide minigels (e.g., Invitrogen Novex
XV00100PK20), or hand-cast equivalents, and resolve the
RNA by electrophoresis at constant current (16 mA for one
gel, 32 mA for two gels) in Tris–glycine running buffer for 2 h
(see Note 7).
8. Stain gels by incubating in ultrapure water containing ~1 μg/
mL ethidium bromide for 5-10 min. Detect the rotavirus
genome segments using a UV transilluminator or equivalent
gel imaging system (e.g., BioRad ChemiDoc system) (see
Note 8).
Rotavirus Reverse Genetics 259
3.4 Plaque Isolation 1. Seed six-well plates with 1.5 × 105 MA104 cells in 2 mL
of Recombinant DMEM complete medium per well. Incubate cells at 37 °C in
Rotaviruses a humidified 5% CO2 incubator until MA104 monolayers have
fully formed (typically 3–4 days).
2. Activate rotavirus in 100 μL portions of clarified cell lysates by
adding 1 μL of 1 mg/mL porcine trypsin stock and incubating
mixtures for 1 h at 37 °C.
3. Prepare tenfold (10-1–10-7) dilution series of trypsin-treated
lysates by mixing 100 uL volumes of lysate samples into 900 μL
of prewarmed DMEM incomplete medium in 1.5 mL micro-
fuge tubes. Vortex to mix serial dilutions at each step before
subsequent transfer. Use fresh pipettor tips to prepare each
dilution.
4. Rinse confluent monolayers in six-well plates twice with 2 mL
of prewarmed 1X PBS. Rinse monolayers one additional time
with 2 mL of prewarmed DMEM incomplete medium, leaving
100 μL of medium in the wells to prevent the cells from drying.
5. Gently add 400 μL of the 10-1–10-7 serially diluted virus
samples to the MA104 monolayers in duplicate starting from
the most dilute to the least dilute. For the negative control, add
400 μL of DMEM incomplete media to wells.
6. Adsorb the virus to the cells by incubating the plates in an
incubator for 1 h, accompanied by gentle rocking of the inoc-
ulum covering the cells every 15 min.
7. While virus is adsorbing, prepare a solution of 1.5% SeaPlaque
agarose (Lonza) by melting in water using a microwave. Place
melted agar in a 56 °C water bath until ready to use. Prepare
the necessary volume of 1:1 agar–EMEM mixture (3 mL per
well) by mixing equal volumes of the melted agarose and pre-
warmed 2X EMEM incomplete medium, for a final concentra-
tion of 0.75% agar. Place agar–EMEM mixture in a 42 °C water
bath until ready to use. Adjust agar–EMEM solution to a final
concentration of 0.5 μg/mL porcine trypsin immediately
before placing on monolayers.
8. Carefully pipet off inoculum from the MA104 monolayers, and
rinse once with 2 mL of prewarmed DMEM. Adjust the dis-
charge setting of a pipet-aid to slow speed, and use it to
carefully overlay the MA104 monolayers with 3 mL of the
agar–EMEM mixture. Briefly and slowly swirl agar–EMEM
mixture over monolayer, and then let the six-well plates rest
at room temperature until agar has solidified.
9. Place the plates in a humidified 37 °C CO2 incubator for
3–4 days, and leave until plaques are visible as whitish/opaque
circular patches on the MA104 monolayer when the plate is
observed using a light box.
260 Chantal A. Agbemabiese et al.
3.5 Amplification of 1. Trypsin activate 250 μL portions of virus samples eluted from
Plaque Isolated Virus agarose plugs by adjusting to 10 μg/mL porcine trypsin and
incubating for 1 h at 37 °C.
2. Following procedures detailed in Subheading 3.4 above, pre-
pare MA104 monolayers in six-well plates. After rinsing twice
with prewarmed 1X PBS and once with prewarmed DMEM
incomplete media, add activated virus samples to the mono-
layers, and incubate the plates for 1 h, rocking every 15 min.
Afterwards, add 2 mL of DMEM incomplete medium contain-
ing 0.5 μg/mL porcine trypsin to the wells.
3. Monitor monolayers daily for the presence of CPE. Once CPE
has reached 100%, subject the plates to three rounds of freeze-
thawing, and then collect the lysates in 2 mL microfuge tubes.
Clarify the lysates by centrifugation at 500× g for 10 min at 4 °
C.
4. Aliquot the clarified lysates (0.5 mL per 1.5 mL microfuge
tube), and freeze at -20 °C.
5. Viruses contained in the samples can be further amplified in
T75 flasks containing MA104 cell monolayers, prepared as
described in Subheading 3.1.
6. The titer of viruses contained in lysates can be determined by
plaque assay following procedures provided in Subheading 3.4.
Rotavirus Reverse Genetics 261
4 Notes
Acknowledgment
Our appreciation goes out to the many past and present members
of the laboratory for their efforts in developing an efficient rotavi-
rus reverse genetics system. This work was supported by funds
provided by the National Institutes of Health (R21AI144881),
GIVAX, Indiana Clinical and Translational Sciences Institute, and
the Lawrence M. Blatt Endowment.
References
6. Desselberger U (2020) Potential of plasmid 17. Sánchez-Tacuba L, Feng N, Meade NJ, Mellits
only based reverse genetics of rotavirus for the KH, Jaı̈s PH, Yasukawa LL, Resch TK, Jiang B,
development of next-generation vaccines. Curr López S, Ding S, Greenberg HB (2020) An
Opin Virol 44:1–6 optimized reverse genetics system suitable for
7. Philip AA, Patton JT (2021) Rotavirus as an efficient recovery of simian, human, and
expression platform of domains of the SARS- murine-like rotaviruses. J Virol 94(18):
CoV-2 spike protein. Vaccines (Basel) 9(5):449 e01294–e01220
8. Philip AA, Patton JT (2022) Generation of 18. Komoto S, Fukuda S, Kugita M, Hatazawa R,
recombinant rotaviruses expressing human Koyama C, Katayama K, Murata T, Taniguchi
norovirus capsid proteins. J Virol 96(22): K (2019) Generation of infectious recombi-
e0126222 nant human rotaviruses from just 11 cloned
9. Kawagishi T, Sánchez-Tacuba L, Feng N, cDNAs encoding the rotavirus genome. J
Costantini VP, Tan M, Jiang X, Green KY, Virol 93(8):e02207–e02218
Vinjé J, Ding S, Greenberg HB. Mucosal and 19. Kanai Y, Komoto S, Kawagishi T, Nouda R,
systemic neutralizing antibodies to norovirus Nagasawa N, Onishi M, Matsuura Y,
induced in infant mice orally inoculated with Taniguchi K, Kobayashi T (2017) Entirely
recombinant rotaviruses. Proc Natl Acad Sci U plasmid-based reverse genetics system for rota-
S A 2023 120(9):e2214421120. viruses. Proc Natl Acad Sci U S A 114(9):
10. Komoto S, Fukuda S, Murata T, Taniguchi K 2349–2354
(2021) Human Rotavirus Reverse Genetics 20. Philip AA, Perry JL, Eaton HE, Shmulevitz M,
Systems to Study Viral Replication and Patho- Hyser JM, Patton JT (2019) Generation of
genesis. Viruses 13(9):1791 recombinant rotavirus expressing NSP3-
11. Antia A, Pinski AN, Ding S (2022) UnaG fusion protein by a simplified reverse
Re-examining rotavirus innate immune eva- genetics system. J Virol 93(24):e01616–
sion: potential applications of the reverse e01619
genetics system. MBio 13(4):e0130822 21. Arnold MM, Brownback CS, Taraporewala ZF,
12. Desselberger U (2020) What are the limits of Patton JT (2012) Rotavirus variant replicates
the packaging capacity for genomic RNA in the efficiently although encoding an aberrant
cores of rotaviruses and of other members of NSP3 that fails to induce nuclear localization
the Reoviridae? Virus Res 15(276):197822 of poly(A)-binding protein. J Gen Virol 93
(Pt 7):1483–1494
13. Settembre EC, Chen JZ, Dormitzer PR,
Grigorieff N, Harrison SC (2011) Atomic 22. Philip AA, Herrin BE, Garcia ML, Abad AT,
model of an infectious rotavirus particle. Katen SP, Patton JT (2019) Collection of
EMBO J 30:408–416 recombinant rotaviruses expressing fluorescent
reporter proteins. Microbiol Resour Announc
14. Trask SD, McDonald SM, Patton JT (2012) 8(27):e00523–e00519
Structural insights into the coupling of virion
assembly and rotavirus replication. Nat Rev 23. Philip AA, Patton JT (2020) Expression of
Microbiol 10:165–177 separate heterologous proteins from the rota-
virus NSP3 genome segment using a transla-
15. Matthijnssens J, Ciarlet M, McDonald SM, tional 2A stop-restart element. J Virol 94(18):
Attoui H, Bányai K, Brister JR, Buesa J, e00959–e00920
Esona MD, Estes MK, Gentsch JR, Iturriza-
Gómara M, Johne R, Kirkwood CD, 24. Chang-Graham AL, Perry JL, Strtak AC,
Martella V, Mertens PP, Nakagomi O, Ramachandran NK, Criglar JM, Philip AA,
Parreño V, Rahman M, Ruggeri FM, Saif LJ, Patton JT, Estes MK, Hyser JM (2019) Rota-
Santos N, Steyer A, Taniguchi K, Patton JT, virus calcium dysregulation manifests as
Desselberger U, Van Ranst M (2011) Unifor- dynamic calcium signaling in the cytoplasm
mity of rotavirus strain nomenclature proposed and endoplasmic reticulum. Sci Rep 9(1):
by the Rotavirus Classification Working Group 10822
(RCWG). Arch Virol 156(8):1397–1413 25. Gratia M, Sarot E, Vende P, Charpilienne A,
16. Kanda M, Fukuda S, Hamada N, Nishiyama S, Baron CH, Duarte M, Pyronnet S, Poncet D
Masatani T, Fujii Y, Izumi F, Okajima M, (2015) Rotavirus NSP3 is a translational surro-
Taniguchi K, Sugiyama M, Komoto S, Ito N gate of the poly(A) binding protein-poly(A)
(2022) Establishment of a reverse genetics sys- complex. J Virol 89(17):8773–8782
tem for avian rotavirus A strain PO-13. J Gen 26. Luke GA, Ryan MD (2018) Therapeutic appli-
Virol 103(6) cations of the ‘NPGP’ family of viral 2As. Rev
Med Virol 28(6):e2001
INDEX
Daniel R. Pérez (ed.), Reverse Genetics of RNA Viruses: Methods and Protocols, Methods in Molecular Biology, vol. 2733,
https://doi.org/10.1007/978-1-0716-3533-9,
© The Editor(s) (if applicable) and The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer
Nature 2024
265
REVERSE GENETICS OF RNA VIRUSES: METHODS AND PROTOCOLS
266 Index
C DH5α........................................................... 120, 176, 179
4′,6′-Diamidino-2-2phenylindole
Canada ........................................................................... 116 (DAPI) ............................................. 191, 197, 200
Canine RIE1495 cells ..................................................... 78 Dianlovirus ........................................................................ 1
Cap Analog .................................................................... 235 Direct-zol RNA kit ...................................................77, 80
Capsid (C) ............................................................ 208, 232 DI RNAs........................................................................ 110
Carbenicillin ..............................................................25, 26 Dithiothreitol (DTT).......................................... 5, 7, 158,
CCL-81 ......................................................................... 191 159, 209, 212, 224, 226, 234
cDNA clone................................................ 136, 178, 179, Double-stranded RNA (dsRNAs) .............. 250, 261, 262
188, 189, 192–196, 202, 232 DMEM/F-12....................................................... 139, 149
cDNA synthesis ............................................................. 156 DMEM10%FCS/PS/Q ...................................................... 6, 7
Chemocompetent Escherichia coli.................................. 91 DMSO ........................................................................... 5, 9
Chicken Pol-II β-actin promoter ................................... 76 DNA-dependent RNA polymerase ................................ 18
Chicken polymerase II .................................................. 120 DNA ladder ................................................................... 158
Chicken polymerase II-driven β-actin promoter......... 104 DNA-launch plasmids.......................................... 156, 170
Chloramphenicol............... 138, 142, 188–190, 193, 194 DNA ligation................................................................. 158
Chloroform-isoamyl...................................................... 235 DNA polymerase .................................................. 190, 194
Chronic liver disease ..................................................... 175 dNTP mix ........................................................................ 81
Cirrhosis ........................................................................ 175 dNTPs.......... 5, 7, 9, 158, 159, 209, 212, 213, 216, 221
Cis-elements .................................................................. 175 Dulbecco’s Modified Eagle Medium (DMEM).......5, 25,
Codon optimization ....................................................... 45 28, 31, 41, 44, 78, 80, 105, 121, 122, 124, 127,
Colony PCR .................................................................... 61 138, 139, 147, 149, 159, 164, 167, 169, 177,
Competent cells.................................................... 176, 179 178, 180, 190, 208, 210, 211, 222–224
Complementary DNA (cDNA)........................ 16, 18–21, Dulbecco’s modified minimum essential medium ...... 105
27–29, 37, 38, 40, 41, 43, 76, 80–82, 87, 94, 103,
156, 157, 159–161, 163, 164, 166, 167, E
170–172, 187–189, 192–196, 201, 202, 208,
209, 212, 213, 215–217, 221, 227 E. coli DH5α cells ........................................................... 82
Complementary RNA (cRNA)..................................... 119 E70........................................................................ 209, 215
Congenital Zika syndrome ........................................... 208 Ebola virus (EBOV)................................................1–3, 11
Coronaviridae................................................................ 134 EcoRI...................................................158, 160, 165, 166
Coronavirus ................................................. 133, 134, 185 Effective serum dilution 50 ............................................ 70
Coronavirus disease 19 (COVID-19)................ 133, 134, eGFP .................................................................................. 6
137, 185 Elongation ...........................................214, 216, 217, 225
Cricetid rodents ............................................................ 116 Embryonated chicken eggs ............................... 38, 41–44
Crystal violet........................................191, 201, 210, 223 Encephalomyocarditis virus (EMCV) .......................... 104
Cuevavirus ......................................................................... 1 Enhanced BSL-3 ........................................................... 104
Cytomegalovirus (CMV) ..................................... 141, 156 Enteroviruses (EVs) .....................................155–157, 171
Cytomix solution .......................................................... 235 Envelope (E) ..............................102, 134, 136, 208, 232
Cytopathic effect (CPE) ...................................65, 66, 96, Envelope glycoproteins........................................ 103, 104
97, 109, 111, 124, 136, 143–145, 148, 150 Escherichia coli (E. coli) ..................... 137, 142, 148, 149,
Cytotoxic effects.............................................................. 96 156, 164–166, 176, 208, 209, 215, 217, 232
ESD50 ............................................................................. 70
D Ethanol .......................................... 5, 7, 11, 89, 209, 210,
212, 214, 218, 220, 222, 224, 226, 233–235
DAB Substrate............................................. 139, 191, 199 Ethidium bromide (EtBr).................................... 158, 209
DEAE-Dextran.............................................................. 139 EtOH ............................................................................... 93
Denaturation ............................................... 214, 216, 225 Eukaryotic promoter..................................................... 156
Dengue ................................................................. 231–246 Europe ........................................................................... 116
Dengue hemorrhagic fever (DHF) .............................. 231 European Medicines Agency (EMA) ............................... 2
Dengue shock syndrome (DSS) ................................... 231 EV-D68 ..............................156–161, 163–167, 169–173
Dengue virus (DENV) ........................................ 231–246 Exon-binding sequences (EBS).................................... 217
DENV-1 ........................................................................ 231 Extract-Peptone-Dextrose ............................................ 234
DENV-4 ........................................................................ 231 E.Z.N.A. ............................................................... 209–211
DH10B ............ 137, 138, 142, 148, 149, 189, 193, 201 E.Z.N.A. Total RNA Kit I.............................................. 89
REVERSE GENETICS OF RNA VIRUSES: METHODS AND PROTOCOLS
Index 267
F HDV-Rbz ..................................................................20, 21
HEK293T constitutively expressing hACE2............... 149
FastAP alkaline phosphatase ........................................... 81 HEK293T-hACE2 ........................................................ 149
Fetal bovine serum (FBS) ........................ 5, 7, 25, 41, 91, Hemagglutination assay (HA)..........................41, 44, 48,
105, 108, 122, 124, 127, 138, 159, 164, 167, 51, 52, 64–68, 71, 72, 75, 79
169, 177, 178, 180, 208, 209, 211, 222–224, 235 Hemagglutinin (HA) ...................................................... 48
Fetal calf serum (FCS) .................................................... 78 Hemorrhagic fever (HF) ..................................... 101, 116
Filoviruses ..................................................................1–4, 9 Henipavirus ...............................................................15, 22
Flaviviridae ................................................. 175, 207, 231 HEp2 cells ....................................................................... 41
Flavivirus.....................................185, 207, 208, 227, 231 Hepacivirus .................................................................... 175
FLUAV-NLuc .................................................... 64, 67, 71 Hepatitis C virus (HCV) ..................................... 175–182
FLUBV-NLuc ..........................55, 56, 64, 65, 67–69, 71 Hepatitis delta virus (HDV)................................. 40, 104,
Fluorescence reporter gene ........................ 157, 159, 167 141, 157, 161, 165, 171
Fluorescent microscope ................................................ 192 Hepatocellular carcinoma ............................................. 175
Focus-forming units (FFU).......................................... 181 HEPES........................................................................... 208
Food and Drug Administration (FDA) ...................2, 116 HiBind RNA column...................................................... 93
Formaldehyde.............................................. 210, 221, 223 High-fidelity DNA polymerase .................................... 177
Formalin ........................................................................ 122 Highly pathogenic avian influenza (HPAI) .............39, 40
Fugene 6 ....................................................................91, 96 Hind III-HF.................................................................. 210
Full-length cDNA ....................................... 192, 232, 239 HKU1 ............................................................................ 133
Full-length vRNA ......................................................... 208 Homologous recombination ...................... 233, 240, 245
Fusion PCR .........................................161, 162, 168, 169 Huh-7 ...............................................................3, 177, 180
Huh7.5.1 .............................................................. 177, 180
G
Human angiotensin converting enzyme 2
G418 .............................................................................. 105 (hACE2) ............................................................ 149
Gastroentiritis................................................................ 249 Human cytomegalovirus (CMV) ................................... 76
Gc.......................................................................... 102–104 Human embryonic kidney.............................................. 78
GC buffer .......................................................................... 9 Human embryonic kidney 293T cells (HEK293T)
Gel Extraction Kit ....................................... 190, 193, 202 cells................................................ 3, 9, 25, 28, 54,
Gene end (GE)................................................... 39, 40, 42 78, 82, 159, 164, 169, 170, 173
Gene start (GS) .........................................................39, 42 Human enteroviruses........................................... 155–173
Geneticin ....................................................................... 105 Human parainfluenza viruses (HPIV) ........15, 16, 22–24
Gentamicin ................................................................91, 92 Human polymerase I promoter (hPol-I).................50, 76
Glasgow’s minimal essential medium (GMEM) ......... 105 Human rhabdomyosarcoma (RD) cells ....................... 159
Glycoprotein (GP) ......................................................2, 11 Human RNA polymerase I............................................. 76
Glycoprotein precursor (GPC)............................ 117–119 Human transferrin receptor I ....................................... 118
Gn ......................................................................... 102–104
Gn/Gc .................................................................. 102, 103 I
GoTaq ................................................................... 209, 216 Immediate-early promoter.............................................. 76
GP complex ................................................................... 117 Immunofluorescence .................................................... 191
GP1 ....................................................................... 117, 119 Immunofluorescent assay (IFA) .........136, 143, 144, 150
GP2 ....................................................................... 117, 118 Immunofocus assay ....................................................... 199
Guillain-Barre syndrome (GBS) .......................... 186, 208 Immunostaining...........................................191, 196–198
GZ01 ............... 208, 209, 211, 213, 215, 217, 223, 227 Infection medium ........................................................... 92
Infectious cDNA clone ........................................ 156, 176
H
Infectious clones ........................................ 143, 156, 157,
H17............................................................................75, 76 169, 187, 188, 192, 193, 201, 208, 209, 215–
H17N10 ............................................................. 76, 77, 79 218, 227, 232, 233, 240
H18..................................................................... 75, 76, 84 Infectious DNA............................................................. 208
H18N11 ............................................................. 76, 77, 79 Infectious Salmon Anemia Virus (ISAV) ................87–90,
H5 ..............................................................................39–44 92, 94, 95, 97, 98
Haemagglutinin .............................................................. 75 Influenza .................................. 39, 41–43, 47–50, 52–55,
Hammerhead ribozyme (Hh-Rbz) ..........................20, 21 57, 64, 65, 70, 73, 75, 78–85, 87, 150, 151, 176
Hartmanivirus .............................................................. 115
REVERSE GENETICS OF RNA VIRUSES: METHODS AND PROTOCOLS
268 Index
Influenza A (FLUAV) .......................................47, 48, 51, LPF2000...................................................... 122, 124, 127
52, 54–56, 58–61, 63–65, 67–69, 71, 72 Luciferase..................................................... 48, 56, 69, 70
Influenza B (FLUBV) ................................ 47, 48, 51, 52, Lujo virus (LUJV)......................................................... 116
54–56, 58, 59, 61, 63–65, 67, 69, 71, 72 Luria Bertani (LB) .......................................................... 40
Influenza viruses........................................................47, 49 Luria broth (LB) ............................... 109, 120, 138, 142,
In-Fusion Snap Assembly Master Mix ........................... 40 158, 164–166, 172, 176, 179, 209, 215, 217
Interferon (IFN) ............................................................... 2 Lymphocytic choriomeningitis virus (LCMV)............ 116
Intergenic (IG).................................................39, 42, 103 Lysogeny broth (LB) .................... 25–28, 109, 138, 142,
Intergenic region (IGR) ............ 102, 103, 116–119, 122 158, 164–166, 172, 176, 179, 209, 215, 217
Internal ribosomal entry site (IRES) ................. 104, 106,
109, 155, 175 M
Internal transcribed spacer region 1 (ITS-1) ..........88, 89
Madagascar .................................................................... 101
iProof DNA polymerase ........................................ 5, 9, 12 Madin-Darby canine kidney ........................................... 54
IPTG ..................................................................... 209, 215 Madin-Darby canine kidney cells II ............................... 78
IRES-vL ................................................................ 104, 106
Mammarenavirus .........................................115–120, 128
IRES-vN ............................................................... 104, 106 Mammarenaviruses .............................115, 116, 118, 119
ISAV901_09 ....................................................... 91, 92, 94 Marburg virus.................................................................... 1
ITS-1................................................................................ 88
Matrix ............................................................................ 117
Max-Efficiency Stbl2 competent cells ......................25, 27
J
Maxiprep........................................................................ 120
Japan .............................................................................. 116 Mayinga ............................................................................. 9
jetPRIME ............................................................. 159, 164 MDCK HLA-DR cells .................................................... 84
JFH-1 strain .................................................................. 176 MDCK II............................................................ 78, 80, 83
Josiah strain ................................................................... 120 Measles virus (MeV) ................................... 15, 18, 20, 22
Junin virus (JUNV)....................................................... 116 Media without amino acids .......................................... 234
MEGAscript T7 Transcription ..................................... 235
K MEM Amino acids solution ......................................... 105
Kas I ...................................................................... 210, 215 Membrane ............................................................ 134, 136
Kpn I-HF.............................................................. 210, 215 Měnglà virus ...................................................................... 1
Mesogenic........................................................................ 41
L Methanol ....................................................................... 178
MgCl2 ............................................................................ 5, 9
L-15 ...........................................................................91, 92 M7GpppG analogue ..................................................... 210
LaSota ........................................................................40–42 Microneutralization assays ........................................48, 71
Lassa fever (LF) ............................................................. 116 Middle East respiratory syndrome coronavirus
Lassa–lymphocytic choriomeningitis serocomplex ..... 116 (MERS-CoV)............................................ 133, 134
Lassa virus (LASV) .............................................. 116–118, Minimum essential media (MEM)...................... 235, 244
120–122, 124, 126–128 Minireplicon ..............................................................16, 29
Lassa virus rescue systems............................................. 122 Mirus TransIT-LT1...................................................25, 41
Lassa virus reverse genetics........................................... 120 Mitochondrial rRNA intron 2...................................... 217
LA Taq DNA polymerase ................................... 209, 210, MluI ...........................................................................91, 92
213, 216, 222, 227 M-MLV Reverse transcriptase ...................................... 210
Late-onset encephalitis ................................................. 101 Modified Eagle Media .................................................. 105
LB agar ............................................................... 25, 27, 28 Monarch DNA Gel Extraction Kit............................... 178
Leibovitz medium ........................................................... 91 Monoclonal antibody (mAb) .................... 139, 140, 143,
Lentogenic.................................................................40, 41 144, 146, 147, 149
L-glutamine ........................................................ 5, 91, 138 Mononegavirales .............................................1, 15, 37, 39
Ligase-T4......................................................................... 94 Morbillivirus ..............................................................16, 22
Lipofectamine 2000..................................... 82, 122, 136, Mosquito-borne ................................................... 101, 207
139, 191, 210, 211, 222, 223 MP-12...........................................................104, 106–109
Lipofectamine LTX/PLUS ............................................ 25 mPol-I............................................................................ 127
Live-attenuated RVFV .................................................. 104 mPol-I L ...................................................... 120, 122, 126
Live-attenuated vaccines (LAV) .......................... 119, 134 mPol-I S......................................................................... 120
Lloviu virus........................................................................ 1 mRNAs ..................................................3, 4, 76, 102, 103
REVERSE GENETICS OF RNA VIRUSES: METHODS AND PROTOCOLS
Index 269
Mumps-like virus............................................................. 16 O
Mumps virus (MuV) .................................................15, 24
Murid rodents ............................................................... 116 OC43 ............................................................................. 133
Murine polymerase I transcription terminator .............. 50 Ocular damage .............................................................. 101
Murine terminator sequence .......................................... 76 Old World (OW)......................................... 115, 116, 118
MVA-T7 ............................................................. 38, 41, 42 Oligonucleotide primer ................................................ 158
Oligonucleotides ........................................................... 215
N One Shot TOP10.......................................................... 109
OneStep RT-PCR .............................................. 77, 80, 81
NanoDrop ..................................................................... 192 Open reading frames (ORFs) ...........................76, 78, 80,
NanoDrop 2000 .................................158, 159, 163, 164 102, 104, 134, 136, 155, 208, 232
Nano Glo ......................................................................... 70 Opti-MEM ........................................ 5, 6, 25, 28, 41, 42,
NanoLuc (NLuc) ........48, 49, 54–56, 64, 65, 67, 69, 71 54–56, 64–66, 68, 69, 78, 82, 139, 143, 180
NcoI ...........................................................................91, 92 Opti-MEM I............................................... 105, 106, 121,
NEBuffer ....................................................................... 164 122, 124, 127, 177, 191, 195, 210, 211
Negative-sense................................................................... 2 ORF1a ........................................................................... 134
Neuraminidase (NA)................48, 51, 52, 64, 72, 75, 79 ORF1b.................................................................. 134, 136
Neutral buffered formalin ..................139, 143, 147, 150 Orthomyxoviridae ............................................... 47, 75, 87
Neutralizing antibodies ........................48, 49, 56, 69, 71
Neutral red solution...................................................... 105 P
Newcastle disease virus (NDVs)...............................37–44
New World (NW) ................................................ 116, 118 PA.................................................... 76, 78–80, 91, 92, 95
NheI.....................................................158, 159, 164, 167 PACNR ................................................209, 215, 217, 223
Nidovirales..................................................................... 134 Panhandle structure ...................................................... 103
Nitrogen Base................................................................ 234 Paramyxoviridae .................................... 15–17, 29, 37, 38
NL63 ............................................................................. 133 Paramyxoviruses ............................. 15–23, 25, 27, 29, 39
Noble agar ................................................... 105, 108, 110 Pararubulavirus ........................................................15, 24
Non-coding intergenic region...................................... 116 PB1 ................................................. 76, 78–80, 91, 92, 95
Non-coding regions (NCRs)................................. 76, 102 PB1-NLuc ....................................................................... 64
Nonessential amino acids..................................... 177, 180 PB2 ................................................. 76, 78–80, 91, 92, 95
Non-polyadenylated viral mRNAs ............................... 119 pBeloBAC11 .............................................. 137, 141–143,
Non-structural proteins .............................. 103, 134, 232 149, 151, 187, 188, 192, 193, 201
North America .............................................................. 101 PBS + BSA +P/S.......................................................41, 42
NotI ................................... 158–161, 163–165, 234, 239 pCAGGS...........3, 6, 76, 78, 80–82, 104, 106, 109, 120
Not I-HF ..................................................... 210, 215, 217 pCAGGs-T7CoOpt ..................................................27, 28
NR-50327 ............................................................ 191, 203 pcDNA3.1 .................................................................95, 97
NS1 ....................................................................... 208, 232 pcDNA3.1(+) ............................................. 156, 158, 159,
NS2A .................................................................... 208, 232 161, 163, 166, 167, 169, 170, 172, 173
NS2B .................................................................... 208, 232 pCI-neo .....................................................................91, 95
NS3 .............................................................. 208, 232, 233 PCR...............................................................89–95, 97, 98
NS4A .................................................................... 208, 232 PCR Clean-Up ..................................................... 209, 210
NS4B .................................................................... 208, 232 PCR polymerase ............................................................ 156
NS5 ....................................................................... 208, 232 pDP2002 ............................................... 50–54, 59–61, 64
NSm ...................................................................... 102–104 pEGFP .................................................................. 158, 167
nsp.................................................................................. 134 Pellet Paint ........................................................... 235, 243
Nuclease-free water .................................... 158, 209–211, Penicillin ................. 41, 78, 80, 105, 177, 178, 180, 208
213, 216, 218, 220, 222 Penicillin/streptomycin (PS)............................5, 77, 105,
Nucleobond Xtra Midiprep .......................................... 109 122, 124, 127, 138
Nucleocapsid ..................... 102, 104, 134, 136, 139, 140 Peroxidase (HRP) ......................................................... 139
Nucleoprotein (NP)................ 2, 4, 6, 17, 37–39, 41–43, Peroxidase mouse IgG .................................................. 139
76, 78–80, 84, 91, 92, 95, 117–122, 127, 128 PGEM-Teasy ............................................... 209, 215, 217
Nunc Lab-Tek Chamber Slide ..................................... 235 Phage T7 ......................................................................... 38
Phenuiviridae ................................................................ 102
pHH21 ................................................176, 178, 179, 181
Phlebovirus ..................................................................... 102
REVERSE GENETICS OF RNA VIRUSES: METHODS AND PROTOCOLS
270 Index
Phosphate buffered saline (PBS)......................41, 42, 91, Q
92, 96, 97, 122, 124, 126, 128, 139, 140, 143,
144, 146, 147, 159, 177, 181, 209, 211, 224, Q5 high fidelity DNA polymerase ...................... 178, 210
226, 227 Q5 Hot Start High-Fidelity Master Mix ... 160, 161, 169
Phosphoprotein............................................................... 17 QIAamp Viral RNA Mini Kit (QIAGEN)..................5, 7,
pHW2000........................................................... 76, 80–82 158, 159, 233
Picornaviridae ............................................................... 155 QIAGEN Plasmid midi ................................................ 234
Plaque assays...........................................30, 97, 107, 109, QIAGEN Plasmid Plus Midi Kit......................... 158, 166
110, 128, 136, 137, 139, 143, 144, 146–150, QIAprep Spin Miniprep Kit ........ 26, 158, 164, 165, 234
164, 167, 169, 191, 196–200, 222, 260 QIAquick Gel Extraction kit .....158, 161, 169, 233, 234
Plasmid-based................................................................ 119 QIAquick PCR purification kit .................. 158, 164, 233
Plasmid-based reverse genetics..................................... 120
R
Plasmid DNA ................................................................ 218
Plasmid Midi Kit .................................................. 190, 194 Rabbit β-globin ............................................................... 89
Platinum Pfx DNA Polymerase ...................................... 91 Rabbit β-globin polyadenylation.................................. 120
Platinum SuperFi DNA Polymerase ............................ 235 Rabbit β-globin polyadenylation signal ......................... 77
Platinum Taq High Fidelity DNA polymerase... 233, 236 Rabies virus (RV) ............................................................ 37
Pneumoviridae................................................................. 18 Receptor-destroying enzyme.......................................... 69
Pol I ..............................................................176–179, 181 Recombinant ISA virus (rISAV)..................................... 88
Pol I-T ......................................................... 176, 178, 179 Recombinant Newcastle Disease Virus....................37–45
Poly (A) tail ................................................................... 155 Recombinant Rift Valley fever virus............................. 102
Polyadenylation ............................................................. 141 Recombinant virus ......................... 4, 142, 146, 250, 253
Polyadenylation signal .................................................... 76 Red blood cells at 50%.................................................... 69
Positive-sense........................................................ 208, 232 Reed-Muench method.................................................... 68
Positive-sense RNA ....................................................... 155 Regular growth medium ..........................................95, 97
pProT7......................................................... 104, 106, 109 Rhabdomyosarcoma (RD).................................. 157, 159,
Pre-denaturation .................................................. 214, 216 164, 167, 169, 171, 173
Pre-membrane (prM).................................................... 232 Reporter genes ................................................................ 48
Premembrane/membrane ............................................ 208 Reptarenavirus .............................................................. 115
PrM/M.......................................................................... 208 Respiratory syncytial virus (RSV).............................16, 18
Promoters ............................................. 16–21, 24, 29, 37, Reston virus....................................................................... 1
38, 40, 48, 50, 76, 87–89, 103, 118, 120–122, Restriction endonuclease ...........158, 163, 169, 177, 210
126, 127, 136, 141, 156, 176, 177, 179, 187, Reverse genetics .............................................. 1–4, 16–18,
188, 192, 232, 240, 251, 252 20, 21, 25, 37, 39, 48–50, 54, 59, 61, 64, 70, 71,
Propanol ............................................................... 233, 234 82, 87, 88, 92, 103, 104, 106–107, 110,
Proprotein-convertase (PC) ......................................... 117 119–121, 127, 128, 134, 136, 137, 141, 151,
Protein expression vector ............................................... 95 175–182, 187, 201, 207–227, 231–246, 249–262
P-S4ter-1 ....................................................................... 216 Reverse transcriptase ................................... 158, 177, 178
P-S4TER-2 .................................................................... 216 Reverse transcriptase and polymerase chain reaction
pSS-URG ...............................................89, 92, 94, 95, 97 (RT-PCR) ............................................................ 80
PSVJS01 ..................................... 234, 239–241, 243, 245 Reverse transcription.......................................80, 81, 156,
pT7............................................................... 104, 106, 109 158, 159, 164, 166, 167, 170
pTriex-3 .....................................................................91, 95 Ribavirin ........................................................................ 116
pUC57............................................................................. 94 RiboMax ........................................................................ 210
PureLink HiPure Plasmid Filter Maxiprep ......... 210, 218 Ribonuclease inhibitor ......................................... 158, 159
PureLink HiPure Plasmid Midiprep Kit ........................ 26 Ribonucleoprotein ............................................... 2, 4, 102
Pure Yield TM Plasmid Miniprep system ................91, 95 Ribonucleoprotein complex (RNP).................... 2–4, 102
Pylaiella littoralis .......................................................... 217 Ribosomal frameshift .................................................... 134
Pyrobest high fidelity DNA polymerase ...................... 209 Ribozyme........................................ 40, 95, 104, 141, 208
P-Z-F4 ........................................................................... 221 RIE1495 ...................................................... 78, 80, 83, 84
P-Z-R14B .................................................... 212, 213, 227 Rift Valley Fever (RVFV) .................... 101–104, 106–111
P-Z-R4........................................................................... 221 rLASV .................................................. 120–122, 124–127
P-ZSK-1......................................................................... 215 RNA ........................................93, 95, 208–212, 220–226
P-ZSK-2......................................................................... 215
REVERSE GENETICS OF RNA VIRUSES: METHODS AND PROTOCOLS
Index 271
RNA-dependent RNA polymerase SP6 ................................................................................. 156
(RdRp) ................................................76, 102, 116 Spike...................................................................... 134, 136
RNA genome ................................. 2, 116, 117, 175, 232 SSIV Buffer.................................................................... 5, 7
RNA mPol-I /Pol-II .................................................... 120 SSP/GP1/GP2 .................................................... 117, 119
RNA polymerase ......................................... 38, 40, 41, 43 Stable signal peptide (SSP) .................................. 117, 119
RNA polymerase I........... 48, 87–89, 103, 120, 175–182 STBL3..................................................156, 158, 165, 172
RNA polymerase I-mediated transcription......... 175–182 Stellar ............................................................................... 40
RNA polymerase I terminator........................................ 48 Stellar competent cells ..............................................25, 27
RNA polymerase I/II system ....................................... 103 Stop codon ................................................................40, 42
RNA polymerase II ............... 18, 24, 38, 48, 76, 87, 187 Streptomycin .......... 41, 78, 80, 105, 177, 178, 180, 208
RNase free H2O ................................................... 212, 221 Subgenomic mRNAs .................................................... 134
RNase-free water .......................................................93, 94 Sub-Saharan Africa ........................................................ 101
RNase H ............................................................... 5, 7, 210 Subtilisin kexin isozyme-1/site-1 protease ................. 117
RNase inhibitor .................................................... 212, 221 Sudan virus ........................................................................ 1
RNA transcription promoter ........................................ 156 Super optimal broth with catabolite repression ............ 25
RNaseOUT ................................................................... 5, 7 Supernatant ...............................................................96, 97
RNAses ............................................................................ 80 SuperScript III one-step RT-PCR system .......... 233, 236
RNeasy Mini Kit................................................... 209, 210 Superscript III reverse transcriptase .................... 209, 212
rNTPs ............................................................................ 243 SuperScript IV ................................................................... 7
Rotavirus..................................... 249–257, 259, 261, 262 SuperScript IV First-Strand Synthesis System ............. 178
rSARS-CoV-2 .....................136–138, 141–146, 148–151 Superscript IV Reverse Transcriptase ............................... 5
Rule of six ..................................................................38, 42 SuperScript™ III One-Step RT-PCR ............................ 93
Ruminants ..................................................................... 101 Synthetic drop-out medium ........................234, 240–242
S T
SacII ............................................40, 42, 43, 45, 210, 217 Tacaribe serocomplex.................................................... 116
Saccharomyces cerevisiae........................................ 234, 239 TAE buffer........158, 161, 171, 209, 210, 214, 218–222
SalI ................................................................................... 80 Taı̈ Forest virus.................................................................. 1
Salmo salar ...................................................................... 88 Termination ................................................................... 141
Salmon cells ...............................................................91, 95 Terminator.........................................................21, 76, 89,
SapI ..................................................................... 89, 94, 98 120–122, 167, 170, 176, 179
SARS-CoV ........ 133–138, 141–144, 146, 147, 149–151 Terrific broth (TB) ....................................................26, 28
Saudi Arabia................................................................... 101 T4 DNA ligase ............................................. 81, 158, 164,
SCE .............................................................. 234, 241, 242 165, 177, 179, 190, 193, 202, 209, 215
SDS ....................................................................... 210, 220 TMPRSS2...................................................................... 149
Segmented RNA virus .................................................. 102 T150 tissue culture flasks.................................................. 5
Self-cleaving ribozyme .................................................... 38 TOP10 cells...........................................61, 209, 215, 217
Self-splicing group II intron................................ 207–227 TPCK-treated trypsin ........................................ 77, 80, 83
Sendai virus (SeV) .............................................. 16, 18, 23 TPCK-trypsin ................................. 54–56, 65, 66, 68, 69
Severe acute respiratory syndrome coronavirus 2 Transcription promoter .................................................. 88
(SARS-CoV-2)...................................40, 133–138, Transfection medium ...................................................... 91
141–144, 146, 149–151, 185 Transfection mixture....................................................... 96
Sexual intercourse ......................................................... 207 Transfection of cDNAs ........................................ 103, 250
Shuttle vector ................................................................ 234 Transfections .............................................. 5–7, 9, 16, 25,
Sialic acid ......................................................................... 76 28, 29, 41, 42, 44, 49, 50, 54, 64, 65, 67, 72, 82,
Signal peptidases ........................................................... 117 83, 91, 92, 95, 96, 98, 105–108, 110, 121–124,
Single-stranded.......................................2, 102, 116, 117, 126, 127, 136, 138, 139, 142, 143, 149, 156,
155, 156, 175, 208, 232, 246 157, 159, 164, 173, 176, 178–181, 191,
SKI-1/S1P..................................................................... 117 195–197, 202, 210, 222, 223, 226, 232, 235,
SmaI ...........................................................................91, 92 244, 246, 250, 251, 256, 257, 261
SnapGene Viewer ............................................................ 40 Transferrin receptor I (Tfr1) ........................................ 118
SOC ...... 25, 27, 40, 138, 142, 158, 164, 165, 189, 193 TransIT-LT1..................5, 6, 9, 105, 106, 110, 178, 180
Sodium acetate ..................................................... 235, 243 Transmembrane serine protease 2................................ 149
Sodium bicarbonate ...................................................... 105 Triton-X100 .................................................................. 139
REVERSE GENETICS OF RNA VIRUSES: METHODS AND PROTOCOLS
272 Index
TRIzol...............................................................77, 80, 122 155–159, 167, 170, 171, 175, 177, 178, 188,
TRP1.............................................................................. 234 208, 211, 212, 220, 223, 232, 233, 236, 257
Trp1-Δ1yeast strains ...................................................... 234 Viral RNA-dependent RNA polymerase
Trypsin ........................................................................... 209 (vRdRp) ................................................16, 19, 116
Trypsin-EDTA....................... 25, 29, 123, 138, 178, 235 Virus amplification ............................................... 105, 111
Tryptone ........................................................................ 158 Virus rescues.........................................11, 25, 28, 65, 72,
T7......................................................................3, 4, 6, 156 88, 103, 105, 109–111, 140, 201, 244
T7 bacteriophage ............................................................ 18 Virus vectored vaccine .................................................... 39
T7 polymerase .................................................... 18–20, 25 Vision loss...................................................................... 101
T7 polymerase-driven rescue system............................ 103 VP1 ................................................................................ 155
T7 promoter.............................38, 41, 43, 103, 104, 136 VP2 ................................................................................ 155
T7 RNA polymerase .................... 3, 4, 38, 103, 105, 110 VP24 .................................................................................. 2
T7 terminator ......................................................... 40, 104 VP3 ................................................................................ 155
Type-I interferon (IFN-I) ............................................ 117 VP30 ..........................................................................2, 4, 6
Typtose phosphate broth (TPB) ......................... 105, 108 VP35 ..........................................................................2, 4, 6
VP4 ....................................................................... 155, 167
U VPg ................................................................................ 155
Uganda .......................................................................... 207
W
UltraPure phenol-chloroform-isoamyl
alcohol....................................................... 235, 243 WA-1 strain.................................136, 137, 141, 146, 150
UltraPure Salmon Sperm DNA Solution ........... 234, 240 Western Africa ............................................................... 116
United States ................................................................. 116 Wizard Plus SV Minipreps............................................ 209
Untranslated regions (UTRs)....117, 118, 175, 208, 232 Wizard SV Gel............................209, 210, 214, 215, 218
USA................................................................................ 102 Wizard SV Gel and PCR Clean-Up System ..... 89, 91, 94
USA-WA1/2020........................................................... 136
X
V
XbaI ...........................................................................91, 92
Variants of concern (VoC) ................................... 134, 136 X-gal...................................................................... 209, 215
Variants of interest (VoI) ..................................... 134, 136 XhoI ....................................... 91, 92, 210, 215, 218, 219
Vectastain ABC Kit..............................139, 147, 191, 198
Velogenic ......................................................................... 41 Y
Vero. 121, 123, 124, 126–128, 187, 191, 195–200, 202 Yeast artificial chromosomes (YACs) ........................... 232
Vero CCL81 cells ......................................................25, 29
Yeast-E. coli shuttle vectors .......................................... 232
Vero E6 (African green monkey kidney epithelial) cells 3, Yeast extract................................................................... 158
9, 104, 105, 107–109, 111, 136, 138, 142–144, Yeast Extract-Peptone-Dextrose (YPD) .... 234, 239, 240
146, 148–150
Yeast nitrogen base (YNB) ..........................234, 240–242
Vertical transmission ..................................................... 207 Yeasts.................................................. 138, 158, 189, 209,
Vesicular stomatitis virus................................................... 2 232–234, 239–242, 245, 246
vG.......................................................................... 104, 106 Yemen ............................................................................ 101
Viral cDNAs .................................................................... 95 YPH252....................................................... 234, 239, 245
Viral-like particles (VLPs)............................................... 16
Viral nucleoprotein ....................................................... 117 Z
Viral polymerase L ............................................................ 2
Viral protein 30 ................................................................. 2 Zero Blunt TOPO PCR Cloning ................................ 178
Viral replication complex ................................... 17, 18, 20 ZH501........................................................................... 104
Viral ribonucleoprotein complex ZH548........................................................................... 104
(vRNP)................................................76, 117, 118 Zika forest...................................................................... 207
Viral ribonucleoproteins ............................................... 117 Zika virus (ZIKV) ............................................... 185–189,
Viral RNA (vRNA)............2–5, 7, 18–20, 29, 44, 48–50, 191–197, 199–202, 207–227
57, 58, 64, 71, 72, 76, 80, 89, 92, 94–96, 98, Zymoclean .................................................................77, 81
102–104, 110, 116–122, 127, 134, 136, Zymolyase.................................................... 234, 241, 242
ZymoPURE...............................................................77, 82