Brief Report: Enhanced isolation of SARS-CoV-2 by TMPRSS2-expressing cells (original) (raw)

Proc Natl Acad Sci U S A. 2020 Mar 31; 117(13): 7001–7003.

Brief Report

Shutoku Matsuyama,a,1 Naganori Nao,a Kazuya Shirato,a Miyuki Kawase,a Shinji Saito,b Ikuyo Takayama,b Noriyo Nagata,c Tsuyoshi Sekizuka,d Hiroshi Katoh,a Fumihiro Kato,a Masafumi Sakata,a Maino Tahara,a Satoshi Kutsuna,e Norio Ohmagari,e Makoto Kuroda,d Tadaki Suzuki,c Tsutomu Kageyama,b and Makoto Takedaa,1

Shutoku Matsuyama

aDepartment of Virology 3, National Institute of Infectious Diseases, Tokyo 162-8640, Japan;

Naganori Nao

aDepartment of Virology 3, National Institute of Infectious Diseases, Tokyo 162-8640, Japan;

Kazuya Shirato

aDepartment of Virology 3, National Institute of Infectious Diseases, Tokyo 162-8640, Japan;

Miyuki Kawase

aDepartment of Virology 3, National Institute of Infectious Diseases, Tokyo 162-8640, Japan;

Shinji Saito

bInfluenza Virus Research Center, National Institute of Infectious Diseases, Tokyo 162-8640, Japan;

Ikuyo Takayama

bInfluenza Virus Research Center, National Institute of Infectious Diseases, Tokyo 162-8640, Japan;

Noriyo Nagata

cDepartment of Pathology, National Institute of Infectious Diseases, Tokyo 162-8640, Japan;

Tsuyoshi Sekizuka

dPathogen Genomics Center, National Institute of Infectious Diseases, Tokyo 162-8640, Japan;

Hiroshi Katoh

aDepartment of Virology 3, National Institute of Infectious Diseases, Tokyo 162-8640, Japan;

Fumihiro Kato

aDepartment of Virology 3, National Institute of Infectious Diseases, Tokyo 162-8640, Japan;

Masafumi Sakata

aDepartment of Virology 3, National Institute of Infectious Diseases, Tokyo 162-8640, Japan;

Maino Tahara

aDepartment of Virology 3, National Institute of Infectious Diseases, Tokyo 162-8640, Japan;

Satoshi Kutsuna

eDisease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo 162-8655, Japan

Norio Ohmagari

eDisease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo 162-8655, Japan

Makoto Kuroda

dPathogen Genomics Center, National Institute of Infectious Diseases, Tokyo 162-8640, Japan;

Tadaki Suzuki

cDepartment of Pathology, National Institute of Infectious Diseases, Tokyo 162-8640, Japan;

Tsutomu Kageyama

bInfluenza Virus Research Center, National Institute of Infectious Diseases, Tokyo 162-8640, Japan;

Makoto Takeda

aDepartment of Virology 3, National Institute of Infectious Diseases, Tokyo 162-8640, Japan;

aDepartment of Virology 3, National Institute of Infectious Diseases, Tokyo 162-8640, Japan;

bInfluenza Virus Research Center, National Institute of Infectious Diseases, Tokyo 162-8640, Japan;

cDepartment of Pathology, National Institute of Infectious Diseases, Tokyo 162-8640, Japan;

dPathogen Genomics Center, National Institute of Infectious Diseases, Tokyo 162-8640, Japan;

eDisease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo 162-8655, Japan

Edited by Yuan Chang, University of Pittsburgh, Pittsburgh, PA, and approved March 5, 2020 (received for review February 11, 2020)

Author contributions: S.M., N. Nao, K.S., and M. Takeda designed research; S.M., N. Nao, K.S., M. Kawase, S.S., I.T., N. Nagata, T. Sekizuka, H.K., F.K., M.S., M. Tahara, M. Kuroda, T. Suzuki, and T.K. performed research; S.K. and N.O. contributed new reagents/analytic tools; S.M., N. Nao, K.S., T. Sekizuka, M. Kuroda, T.K., and M. Takeda analyzed data; and S.M. and M. Takeda wrote the paper.

Copyright © 2020 the Author(s). Published by PNAS.

Abstract

A novel betacoronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which caused a large respiratory outbreak in Wuhan, China in December 2019, is currently spreading across many countries globally. Here, we show that a TMPRSS2-expressing VeroE6 cell line is highly susceptible to SARS-CoV-2 infection, making it useful for isolating and propagating SARS-CoV-2. Our results reveal that, in common with SARS- and Middle East respiratory syndrome-CoV, SARS-CoV-2 infection is enhanced by TMPRSS2.

Keywords: coronavirus, SARS-CoV-2, VeroE6, outbreak, TMPRSS2

In December 2019 a respiratory outbreak from a novel betacoronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), occurred in Wuhan City, China (1, 2). As of 9 February 2020, 37,558 confirmed cases and 813 deaths had been recorded. Although the great majority of cases were reported in China, 24 countries had already been affected with 307 confirmed cases. On 15 January 2020, the first case was detected in Japan. As of 10 February 2020, Japan had 16 domestically confirmed cases and 9 returnees from Wuhan using government-chartered flights. In addition, 70 cases were confirmed on a SARS-CoV-2–quarantined cruise ship.

SARS-CoV-2 is isolatable using VeroE6, Huh7, or human airway epithelial cells (24), but here we show that an engineered cell line, VeroE6/TMPRSS2, is highly susceptible to SARS-CoV-2 infection, suggesting the important role for TMPRSS2 in SARS-CoV-2 infection and indicating its potential utility for isolating and propagating this virus.

Previous studies (5, 6) have shown that the phylogenetically related SARS-CoV is proteolytically activated by TMPRSS2 in vitro and in vivo. Therefore, we attempted to isolate SARS-CoV-2 using VeroE6/TMPRSS2 cells, which express TMPRSS2 constitutively. The messenger RNA expression level of TMPRSS2 in VeroE6/TMPRSS2 cells is ∼10-fold higher than in normal human lung tissue and other human cell lines (Fig. 1_A_). SARS-CoV-2 uses the same receptor, ACE2, as SARS-CoV (2), and ACE2 expression is very high in VeroE6 cells (7). Seven clinical specimens (throat swabs or sputum) obtained from seven SARS-CoV-2 infection cases were inoculated into VeroE6/TMPRSS2 cells, which were monitored daily for cytopathic effect (CPE). These clinical specimens were deidentified prior to use, and this study was approved by the ethics committee of the National Institute of Infectious Diseases, Japan (approval no. 1091). Informed consent was obtained from all participants, from which the subjects were obtained, or their legally acceptable representatives for sample donation. In five cases among the seven, clear CPE with detachment/floating (black arrows, Fig. 1_B_) and syncytium formation (white arrows, Fig. 1_B_) developed at 2 or 3 d postinfection (p.i.) (Table 1). The virus titers in culture supernatants of the five cases at 3 d p.i. were 4.6 × 106 to 6.8 × 107 median tissue culture infectious dose (TCID50) per mL (Table 1). Typical coronavirus particles were detected by electron microscopy (Fig. 1_C_). Next-generation sequencing (NGS) of case Wk-521 detected the nearly full-length genome sequence from SARS-CoV-2 with >99.9% homology (1, 2) (GISAID database ID EPI_ISL_408667). Unexpectedly, the NGS data showed contaminated mycoplasma sequences (Mycoplasma hyorhinis and Mycoplasma arginini) from VeroE6/TMPRSS2 cells. CPE in VeroE6 cells persistently infected with SARS-CoV was enhanced by infection with Mycoplasma fermentans (8), but whether a similar situation exists for SARS-CoV-2–related CPE in this cell line is unclear.

An external file that holds a picture, illustration, etc. Object name is pnas.2002589117fig01.jpg

(A) Expression of TMPRSS2 in total cellular RNA (0.2 µg) of indicated cells was compared with that in human lung RNA (catalog no. 636524; Clontech) by quantitative real-time PCR. ND, not detectable. (B) SARS-CoV-2–infected VeroE6/TMPRSS2 cells. Cell rounding (black arrows) and syncytium formation (white arrows) (C). Electron micrograph showing isolated virus particles with negative staining. (Scale bar, 200 nm.) (D) Viral RNA multiplication in various cells at 48 h postinoculation with the viral specimen, as determined by real-time RT-PCR using E and N primer/probe sets (9). Cq, quantitation cycle. (E) Real-time RT-PCR amplification plot using the E primer/probe set, corresponding to the data in C. RFU, relative fluorescence units. (F) Comparison of cell susceptibility to the isolated virus, detected with a patient’s serum and Alexa 488-conjugated goat anti-human IgG. Nuclei were stained with DAPI.

Table 1.

SARS-CoV-2 cases and virus isolation

Cq value of specimens
Case N set* N2 set* Virus isolation Days (CPE appearance) Virus titer at 3 d p.i., TCID50/mL
V-009 33.87 30.87 No >6 d u.d.
Wk-177 u.d. 35.08 No >6 d u.d.
I-004 34.52 31.66 Yes 3 d 2.2 × 107
V-029 32.85 28.80 Yes 3 d 4.6 × 106
Wk-012 33.53 29.60 Yes 3 d 2.2 × 107
Wk-501 27.35 21.76 Yes 2 d 6.8 × 106
Wk-521 29.68 24.41 Yes 2 d 6.8 × 107

The viral RNA copies in the clinical specimens used for virus isolation were estimated by real-time RT-PCR (9, 10). As expected, viral RNA copies in the clinical specimens in which CPE developed within 2 d p.i. were greater than those in the other specimens (Table 1).

VeroE6/TMPRSS2 cells are superior to other cell lines tested in this study for SARS-CoV-2 isolation. Consistent with previous reports (2, 4), the amount of SARS-CoV-2 RNAs in the culture supernatants of Vero, Calu-3, and A549 cells 48 h p.i. was low and was measurably higher when VeroE6 cells were used. However, the viral RNA copies in the VeroE6/TMPRSS2 cell culture supernatants were >100 times greater than those from VeroE6 cells (Fig. 1 D and E). Data for SARS-CoV show that TMPRSS2 enhances its entry efficiency (5, 11). VeroE6 and VeroE6/TMPRSS2 cells were infected with 10-fold serially diluted SARS-CoV-2 samples, and the infected cells were visualized by indirect immunofluorescent assays (Fig. 1_E_). The results showed that VeroE6/TMPRSS2 displayed ∼10-fold greater number of SARS-CoV-2–infected cells than the parental VeroE6 cells. These data suggest that, in common with SARS-CoV, TMPRSS2 may also play an important role in SARS-CoV-2 cell entry.

SARS-CoV and Middle East respiratory syndrome (MERS)-CoV can enter cells via endocytosis and use cathepsin in endosomes for activation (1214). However, TMPRSS2 expression greatly promotes replication and syncytium formation in these viruses in vitro and in vivo (5, 6, 11, 12, 15). Our findings suggest that TMPRSS2 is also likely to be a key protease for SARS-CoV-2 replication. Thus, developing TMPRSS2-related therapeutic agents may be a promising countermeasure against the current and new outbreaks of CoVs.

TMPRSS2-expressing cell lines are highly susceptible to SARS-CoV, MERS-CoV, and SARS-CoV-2, making the VeroE6/TMPRSS2 cell line a suitable contributor to the global surveillance of high-risk CoVs. VeroE6/TMPRSS2 cells are easily maintained, suitable for large-scale propagation, and now available from Japanese Collection of Research Bioresources (JCRB) Cell Bank in Japan (https://cellbank.nibiohn.go.jp/english/) (JCRB no. JCRB1819). Treatment for mycoplasma is now ongoing in the JCRB Cell Bank.

Data Availability

Data have been deposited in the Global Initiative on Sharing All Influenza Data (GISAID) database with accession ID EPI_ISL_408667. The cell line information is available from JCRB Cell Bank in Japan (https://cellbank.nibiohn.go.jp/english/) (JCRB no. JCRB1819).

Acknowledgments

We thank the staff of the National Center for Global Health and Medicine and the Aichi and Kyoto Prefectures for specimen collection and transport and the National Institute of Biomedical Innovation, Health and Nutrition for JCRB Cell Bank service. We thank Drs. T. Wakita and M. Ohnishi for supervision of this project; Drs. T. Chiba and K. Sadamasu (Tokyo Metropolitan Institute of Public Health) for their strong support; and Drs. M. Suzuki, H. Hasegawa, T. Matsui, T. Shimada, T. Sunagawa, M.Saijo, N. Iwata-Yoshikawa, M. Kataoka, T. Kubota, Y. Nakatsu, Y. Mori, K. Someya, N. Otsuki, and Mr. Y. Tanaka for invaluable support. We thank all the staff who care for patients and returnees from Wuhan. We also acknowledge all staff at the National Institute of Infectious Diseases, the Field Epidemiology Training Program team, Ministry of Health, Labour and Welfare, and local governments for administration, field investigation, data collection, and assistance with laboratory testing. This study was supported by a grant-in aid from the Japan Agency for Medical Research and Development under grants JP19fk0108104, JP19fk0108030j0403, JP19fk0108058j0802, and 19fk0108111j0001 and the Japan Society for the Promotion of Science under grants 17K08868 and 18H02665.

Footnotes

The authors declare no competing interest.

Data deposition: Data have been deposited in the Global Initiative on Sharing All Influenza Data (GISAID) database, https://www.gisaid.org/ (accession ID EPI_ISL_408667).

References

1. Wu F., et al., A new coronavirus associated with human respiratory disease in China. Nature, 10.1038/s41586-020-2008-3 (2020). [PMC free article] [PubMed] [CrossRef] [Google Scholar]

2. Zhou P., et al., A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature, 10.1038/s41586-020-2012-7 (2020). [PMC free article] [PubMed] [CrossRef] [Google Scholar]

3. Lu R., et al., Genomic characterisation and epidemiology of 2019 novel coronavirus: Implications for virus origins and receptor binding. Lancet 395, 565–574 (2020). [PMC free article] [PubMed] [Google Scholar]

4. Zhu N. et al..; China Novel Coronavirus Investigating and Research Team , A novel coronavirus from patients with pneumonia in China, 2019. N. Engl. J. Med. 382, 727–733 (2020). [PMC free article] [PubMed] [Google Scholar]

5. Matsuyama S., et al., Efficient activation of the severe acute respiratory syndrome coronavirus spike protein by the transmembrane protease TMPRSS2. J. Virol. 84, 12658–12664 (2010). [PMC free article] [PubMed] [Google Scholar]

6. Iwata-Yoshikawa N., et al., TMPRSS2 contributes to virus spread and immunopathology in the airways of murine models after coronavirus infection. J. Virol. 93, e01815-18 (2019). [PMC free article] [PubMed] [Google Scholar]

7. Ren X., et al., Analysis of ACE2 in polarized epithelial cells: Surface expression and function as receptor for severe acute respiratory syndrome-associated coronavirus. J. Gen. Virol. 87, 1691–1695 (2006). [PubMed] [Google Scholar]

8. Mizutani T., et al., Enhancement of cytotoxicity against Vero E6 cells persistently infected with SARS-CoV by Mycoplasma fermentans. Arch. Virol. 152, 1019–1025 (2007). [PMC free article] [PubMed] [Google Scholar]

9. Corman V. M., et al., Detection of 2019 novel coronavirus (2019-nCoV) by real-time RT-PCR. Euro. Surveill. 25, 2000045 (2020). [PMC free article] [PubMed] [Google Scholar]

10. Shirato K., et al., Development of genetic diagnostic methods for novel coronavirus 2019 (nCoV-2019) in Japan. Jpn. J. Infect. Dis., 10.7883/yoken.JJID.2020.061 (2020). [PubMed] [CrossRef] [Google Scholar]

11. Shulla A., et al., A transmembrane serine protease is linked to the severe acute respiratory syndrome coronavirus receptor and activates virus entry. J. Virol. 85, 873–882 (2011). [PMC free article] [PubMed] [Google Scholar]

12. Shirato K., Kawase M., Matsuyama S., Middle East respiratory syndrome coronavirus infection mediated by the transmembrane serine protease TMPRSS2. J. Virol. 87, 12552–12561 (2013). [PMC free article] [PubMed] [Google Scholar]

13. Simmons G., et al., Inhibitors of cathepsin L prevent severe acute respiratory syndrome coronavirus entry. Proc. Natl. Acad. Sci. U.S.A. 102, 11876–11881 (2005). [PMC free article] [PubMed] [Google Scholar]

14. Matsuyama S., et al., Middle East respiratory syndrome coronavirus spike protein is not activated directly by cellular furin during viral entry into target cells. J. Virol. 92, e00683-18 (2018). [PMC free article] [PubMed] [Google Scholar]

15. Gierer S., et al., The spike protein of the emerging betacoronavirus EMC uses a novel coronavirus receptor for entry, can be activated by TMPRSS2, and is targeted by neutralizing antibodies. J. Virol. 87, 5502–5511 (2013). [PMC free article] [PubMed] [Google Scholar]


Articles from Proceedings of the National Academy of Sciences of the United States of America are provided here courtesy of National Academy of Sciences