1
|
Geng J, Ren N, Yang C, Wang F, Huang D, Rodriguez S, Yuan Z, Xia H. Favipiravir Treatment Prolongs Survival in a Lethal BALB/c Mouse Model of Ebinur Lake Virus Infection. Viruses 2024; 16:631. [PMID: 38675972 PMCID: PMC11054260 DOI: 10.3390/v16040631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/07/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Orthobunyavirus is the largest and most diverse genus in the family Peribunyaviridae. Orthobunyaviruses are widely distributed globally and pose threats to human and animal health. Ebinur Lake virus (EBIV) is a newly classified Orthobunyavirus detected in China, Russia, and Kenya. This study explored the antiviral effects of two broad-spectrum antiviral drugs, favipiravir and ribavirin, in a BALB/c mouse model. Favipiravir significantly improved the clinical symptoms of infected mice, reduced viral titer and RNA copies in serum, and extended overall survival. The median survival times of mice in the vehicle- and favipiravir-treated groups were 5 and 7 days, respectively. Favipiravir significantly reduced virus titers 10- to 100-fold in sera at all three time points compared to vehicle-treated mice. And favipiravir treatment effectively reduced the virus copies by approximately 10-fold across the three time points, relative to vehicle-treated mice. The findings expand the antiviral spectrum of favipiravir for orthobunyaviruses in vivo.
Collapse
Affiliation(s)
- Jingke Geng
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430200, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Nanjie Ren
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430200, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Cihan Yang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430200, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Fei Wang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430200, China
| | - Doudou Huang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430200, China
| | - Sergio Rodriguez
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77551, USA
| | - Zhiming Yuan
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430200, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Han Xia
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430200, China
- University of Chinese Academy of Sciences, Beijing 101408, China
- Hubei Jiangxia Laboratory, Wuhan 430207, China
| |
Collapse
|
2
|
Kim EH, Park SJ. Emerging Tick-Borne Dabie bandavirus: Virology, Epidemiology, and Prevention. Microorganisms 2023; 11:2309. [PMID: 37764153 PMCID: PMC10536723 DOI: 10.3390/microorganisms11092309] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/09/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Severe Fever with Thrombocytopenia Syndrome (SFTS), caused by Dabie bandavirus (SFTSV), is an emerging infectious disease first identified in China. Since its discovery, infections have spread throughout East Asian countries primarily through tick bites but also via transmission between animals and humans. The expanding range of ticks, the primary vectors for SFTSV, combined with migration patterns of tick-carrying birds, sets the stage for the global spread of this virus. SFTSV rapidly evolves due to continuous mutation and reassortment; currently, no approved vaccines or antiviral drugs are available. Thus, the threat this virus poses to global health is unmistakable. This review consolidates the most recent research on SFTSV, including its molecular characteristics, transmission pathways through ticks and other animals, as well as the progress in antiviral drug and vaccine development, encompassing animal models and clinical trials.
Collapse
Affiliation(s)
- Eun-Ha Kim
- Center for Study of Emerging and Re-Emerging Viruses, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea;
| | - Su-Jin Park
- Division of Life Science, Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| |
Collapse
|
3
|
Snyman J, Snyman LP, Buhler KJ, Villeneuve CA, Leighton PA, Jenkins EJ, Kumar A. California Serogroup Viruses in a Changing Canadian Arctic: A Review. Viruses 2023; 15:1242. [PMID: 37376542 DOI: 10.3390/v15061242] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/24/2023] [Accepted: 05/18/2023] [Indexed: 06/29/2023] Open
Abstract
The Arctic is warming at four times the global rate, changing the diversity, activity and distribution of vectors and associated pathogens. While the Arctic is not often considered a hotbed of vector-borne diseases, Jamestown Canyon virus (JCV) and Snowshoe Hare virus (SSHV) are mosquito-borne zoonotic viruses of the California serogroup endemic to the Canadian North. The viruses are maintained by transovarial transmission in vectors and circulate among vertebrate hosts, both of which are not well characterized in Arctic regions. While most human infections are subclinical or mild, serious cases occur, and both JCV and SSHV have recently been identified as leading causes of arbovirus-associated neurological diseases in North America. Consequently, both viruses are currently recognised as neglected and emerging viruses of public health concern. This review aims to summarise previous findings in the region regarding the enzootic transmission cycle of both viruses. We identify key gaps and approaches needed to critically evaluate, detect, and model the effects of climate change on these uniquely northern viruses. Based on limited data, we predict that (1) these northern adapted viruses will increase their range northwards, but not lose range at their southern limits, (2) undergo more rapid amplification and amplified transmission in endemic regions for longer vector-biting seasons, (3) take advantage of northward shifts of hosts and vectors, and (4) increase bite rates following an increase in the availability of breeding sites, along with phenological synchrony between the reproduction cycle of theorized reservoirs (such as caribou calving) and mosquito emergence.
Collapse
Affiliation(s)
- Jumari Snyman
- Department of Biochemistry, Microbiology and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Louwrens P Snyman
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Kayla J Buhler
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Carol-Anne Villeneuve
- Research Group on Epidemiology of Zoonoses and Public Health (GREZOSP), Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Patrick A Leighton
- Research Group on Epidemiology of Zoonoses and Public Health (GREZOSP), Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Emily J Jenkins
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Anil Kumar
- Department of Biochemistry, Microbiology and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| |
Collapse
|
4
|
Chen L, Chen T, Li R, Xu Y, Xiong Y. Recent Advances in the Study of the Immune Escape Mechanism of SFTSV and Its Therapeutic Agents. Viruses 2023; 15:v15040940. [PMID: 37112920 PMCID: PMC10142331 DOI: 10.3390/v15040940] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/04/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
Sever fever with thrombocytopenia syndrome (SFTS) is a new infectious disease that has emerged in recent years and is widely distributed, highly contagious, and lethal, with a mortality rate of up to 30%, especially in people with immune system deficiencies and elderly patients. SFTS is an insidious, negative-stranded RNA virus that has a major public health impact worldwide. The development of a vaccine and the hunt for potent therapeutic drugs are crucial to the prevention and treatment of Bunyavirus infection because there is no particular treatment for SFTS. In this respect, investigating the mechanics of SFTS-host cell interactions is crucial for creating antiviral medications. In the present paper, we summarized the mechanism of interaction between SFTS and pattern recognition receptors, endogenous antiviral factors, inflammatory factors, and immune cells. Furthermore, we summarized the current therapeutic drugs used for SFTS treatment, aiming to provide a theoretical basis for the development of targets and drugs against SFTS.
Collapse
Affiliation(s)
- Lei Chen
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi 563000, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China
| | - Tingting Chen
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi 563000, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China
| | - Ruidong Li
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi 563000, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China
| | - Yingshu Xu
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi 563000, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China
| | - Yongai Xiong
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi 563000, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China
| |
Collapse
|
5
|
Amodiaquine derivatives as inhibitors of severe fever with thrombocytopenia syndrome virus (SFTSV) replication. Antiviral Res 2023; 210:105479. [PMID: 36566117 DOI: 10.1016/j.antiviral.2022.105479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 11/27/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022]
Abstract
Severe fever with thrombocytopenia syndrome (SFTS) is an emerging tick-borne viral infection caused by a bandavirus in the family of Phenuiviridae, commonly known as SFTS virus (SFTSV). We have previously isolated SFTSV from blood samples of SFTS patients and established an antiviral assay system to identify selective inhibitors of SFTSV in vitro. Using the assay system, the antimalarial agent amodiaquine was identified as a selective inhibitor of SFTSV replication. However, due to its insufficient antiviral activity, 98 amodiaquine derivatives were newly synthesized and examined for their anti-SFTSV activity. Among the derivatives, some compounds showed selective inhibitory effect on SFTSV replication in vitro. The 50% effective concentration (EC50) and cytotoxic concentration (CC50) of the most active compound (C-90) were 2.6 ± 0.6 and >50 μM, respectively. This EC50 value was comparable to or slightly better than that of favipiravir (4.1 ± 0.6 μM). On the other hand, pharmacokinetic studies in vivo revealed that C-90 was poor in its oral bioavailability in mice. Therefore, we further designed and synthesized derivatives and obtained 2 compounds with selective anti-SFTSV activity in vitro and improved pharmacokinetics in vivo.
Collapse
|
6
|
Shimojima M, Sugimoto S, Umekita K, Onodera T, Sano K, Tani H, Takamatsu Y, Yoshikawa T, Kurosu T, Suzuki T, Takahashi Y, Ebihara H, Saijo M. Neutralizing mAbs against SFTS Virus Gn Protein Show Strong Therapeutic Effects in an SFTS Animal Model. Viruses 2022; 14:v14081665. [PMID: 36016286 PMCID: PMC9416629 DOI: 10.3390/v14081665] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 11/16/2022] Open
Abstract
Severe fever with thrombocytopenia syndrome (SFTS) is an infectious disease with a high case fatality rate caused by the SFTS virus, and currently there are no approved specific treatments. Neutralizing monoclonal antibodies (mAbs) against the virus could be a therapeutic agent in SFTS treatment, but their development has not sufficiently been carried out. In the present study, mouse and human mAbs exposed to the viral envelope proteins Gn and Gc (16 clones each) were characterized in vitro and in vivo by using recombinant proteins, cell culture with viruses, and an SFTS animal model with IFNAR-/- mice. Neutralization activities against the recombinant vesicular stomatitis virus bearing SFTS virus Gn/Gc as envelope proteins were observed with three anti-Gn and six anti-Gc mAbs. Therapeutic activities were observed among anti-Gn, but not anti-Gc mAbs with neutralizing activities. These results propose an effective strategy to obtain promising therapeutic mAb candidates for SFTS treatment, and a necessity to reveal precise roles of the SFTS virus Gn/Gc proteins.
Collapse
Affiliation(s)
- Masayuki Shimojima
- Department of Virology I, National Institute of Infectious Diseases, Tokyo 208-0011, Japan; (S.S.); (Y.T.); (T.Y.); (T.K.); (H.E.)
- Correspondence: shimoji-@niid.go.jp (M.S.); (M.S.)
| | - Satoko Sugimoto
- Department of Virology I, National Institute of Infectious Diseases, Tokyo 208-0011, Japan; (S.S.); (Y.T.); (T.Y.); (T.K.); (H.E.)
| | - Kunihiko Umekita
- Department of Respirology, Rheumatology, Infectious Diseases and Neurology, Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan;
| | - Taishi Onodera
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan; (T.O.); (Y.T.)
| | - Kaori Sano
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 162-8640, Japan; ka-- (K.S.); (T.S.)
| | - Hideki Tani
- Department of Virology, Toyama Institute of Health, Toyama 939-0363, Japan;
| | - Yuki Takamatsu
- Department of Virology I, National Institute of Infectious Diseases, Tokyo 208-0011, Japan; (S.S.); (Y.T.); (T.Y.); (T.K.); (H.E.)
- Department of Virology, Institute of Tropical Medicine, Nagasaki University, Nagasaki 852-8523, Japan
| | - Tomoki Yoshikawa
- Department of Virology I, National Institute of Infectious Diseases, Tokyo 208-0011, Japan; (S.S.); (Y.T.); (T.Y.); (T.K.); (H.E.)
| | - Takeshi Kurosu
- Department of Virology I, National Institute of Infectious Diseases, Tokyo 208-0011, Japan; (S.S.); (Y.T.); (T.Y.); (T.K.); (H.E.)
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 162-8640, Japan; ka-- (K.S.); (T.S.)
| | - Yoshimasa Takahashi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan; (T.O.); (Y.T.)
| | - Hideki Ebihara
- Department of Virology I, National Institute of Infectious Diseases, Tokyo 208-0011, Japan; (S.S.); (Y.T.); (T.Y.); (T.K.); (H.E.)
| | - Masayuki Saijo
- Department of Virology I, National Institute of Infectious Diseases, Tokyo 208-0011, Japan; (S.S.); (Y.T.); (T.Y.); (T.K.); (H.E.)
- Medical Affairs Department, Health and Welfare Bureau, Sapporo 060-0042, Japan
- Correspondence: shimoji-@niid.go.jp (M.S.); (M.S.)
| |
Collapse
|
7
|
Sun J, Min YQ, Li Y, Sun X, Deng F, Wang H, Ning YJ. Animal Model of Severe Fever With Thrombocytopenia Syndrome Virus Infection. Front Microbiol 2022; 12:797189. [PMID: 35087498 PMCID: PMC8787146 DOI: 10.3389/fmicb.2021.797189] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/29/2021] [Indexed: 12/30/2022] Open
Abstract
Severe fever with thrombocytopenia syndrome (SFTS), an emerging life-threatening infectious disease caused by SFTS bunyavirus (SFTSV; genus Bandavirus, family Phenuiviridae, order Bunyavirales), has been a significant medical problem. Currently, there are no licensed vaccines or specific therapeutic agents available and the viral pathogenesis remains largely unclear. Developing appropriate animal models capable of recapitulating SFTSV infection in humans is crucial for both the study of the viral pathogenic processes and the development of treatment and prevention strategies. Here, we review the current progress in animal models for SFTSV infection by summarizing susceptibility of various potential animal models to SFTSV challenge and the clinical manifestations and histopathological changes in these models. Together with exemplification of studies on SFTSV molecular mechanisms, vaccine candidates, and antiviral drugs, in which animal infection models are utilized, the strengths and limitations of the existing SFTSV animal models and some important directions for future research are also discussed. Further exploration and optimization of SFTSV animal models and the corresponding experimental methods will be undoubtedly valuable for elucidating the viral infection and pathogenesis and evaluating vaccines and antiviral therapies.
Collapse
Affiliation(s)
- Jiawen Sun
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Yuan-Qin Min
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Yunjie Li
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Xiulian Sun
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Fei Deng
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Hualin Wang
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Yun-Jia Ning
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
8
|
Kato H, Takayama-Ito M, Satoh M, Kawahara M, Kitaura S, Yoshikawa T, Fukushi S, Nakajima N, Komeno T, Furuta Y, Saijo M. Favipiravir treatment prolongs the survival in a lethal mouse model intracerebrally inoculated with Jamestown Canyon virus. PLoS Negl Trop Dis 2021; 15:e0009553. [PMID: 34214091 PMCID: PMC8281987 DOI: 10.1371/journal.pntd.0009553] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 07/15/2021] [Accepted: 06/08/2021] [Indexed: 11/18/2022] Open
Abstract
Background Jamestown Canyon virus (JCV) is a mosquito-borne orthobunyavirus that causes acute febrile illness, meningitis, and meningoencephalitis, primarily in North American adults. Currently, there are no available vaccines or specific treatments against JCV infections. Methodology/Principal findings The antiviral efficacy of favipiravir (FPV) against JCV infection was evaluated in vitro and in vivo in comparison with that of ribavirin (RBV) and 2’-fluoro-2’-deoxycytidine (2’-FdC). The in vitro inhibitory effect of these drugs on JCV replication was evaluated in Vero and Neuro-2a (N2A) cells. The efficacy of FPV in the treatment of JCV infection in vivo was evaluated in C57BL/6J mice inoculated intracerebrally with JCV, as per the survival, viral titers in the brain, and viral RNA load in the blood. The 90% inhibitory concentrations (IC90) of FPV, RBV, and 2’-FdC were 41.0, 61.8, and 13.6 μM in Vero cells and 20.7, 25.8, and 8.8 μM in N2A cells, respectively. All mice infected with 1.0×104 TCID50 died or were sacrificed within 10 days post-infection (dpi) without treatment. However, mice treated with FPV for 5 days [initiated either 2 days prior to infection (−2 dpi–2 dpi) or on the day of infection (0 dpi–4 dpi)] survived significantly longer than control mice, administered with PBS (p = 0.025 and 0.011, respectively). Moreover, at 1 and 3 dpi, the virus titers in the brain were significantly lower in FPV-treated mice (0 dpi–4 dpi) versus PBS-treated mice (p = 0.002 for both 1 and 3 dpi). Conclusions/Significance Although the intracerebral inoculation route is thought to be a challenging way to evaluate drug efficacy, FPV inhibits the in vitro replication of JCV and prolongs the survival of mice intracerebrally inoculated with JCV. These results will enable the development of a specific antiviral treatment against JCV infections and establishment of an effective animal model. Jamestown Canyon virus (JCV) is a mosquito-borne virus (arbovirus) classified into the California serogroup. JCV is distributed widely throughout North America and is considered one of the potentially re-emerging viruses due to the recent spurt in JCV cases in the region. JCV infection often leads to an acute febrile illness, meningitis, and meningoencephalitis mainly among adults. Currently, no antiviral therapy against JCV is approved. In this study, we evaluated the antiviral efficacy of favipiravir (FPV), ribavirin (RBV), and 2’-fluoro-2’-deoxycytidine (2’-FdC) against JCV infection in cultured cells and mice. As a result, FPV, RBV, and 2’-FdC effectively inhibited JCV replication in Vero and Neuro-2a cells. Furthermore, FPV delayed the onset of neurological symptoms in mice intracerebrally inoculated with JCV. Notably, although most patients infected with JCV do not present severe disease, neuroinvasive cases are not rare and may result in residual neurological sequelae such as persisting cognitive deficits. Therefore, this study contributes to the development of a specific antiviral treatment for patients with JCV infection.
Collapse
Affiliation(s)
- Hirofumi Kato
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Mutsuyo Takayama-Ito
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
- * E-mail:
| | - Masaaki Satoh
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Madoka Kawahara
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Satoshi Kitaura
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
- Department of Internal Medicine, The University of Tokyo, Graduate School of Medicine, Tokyo, Japan
| | - Tomoki Yoshikawa
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Shuetsu Fukushi
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | | | | | | | - Masayuki Saijo
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| |
Collapse
|
9
|
Dai S, Deng F, Wang H, Ning Y. Crimean-Congo Hemorrhagic Fever Virus: Current Advances and Future Prospects of Antiviral Strategies. Viruses 2021; 13:v13071195. [PMID: 34206476 PMCID: PMC8310003 DOI: 10.3390/v13071195] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/09/2021] [Accepted: 06/18/2021] [Indexed: 02/03/2023] Open
Abstract
Crimean-Congo hemorrhagic fever virus (CCHFV) is a widespread, tick-borne pathogen that causes Crimean-Congo hemorrhagic fever (CCHF) with high morbidity and mortality. CCHFV is transmitted to humans through tick bites or direct contact with patients or infected animals with viremia. Currently, climate change and globalization have increased the transmission risk of this biosafety level (BSL)-4 virus. The treatment options of CCHFV infection remain limited and there is no FDA-approved vaccine or specific antivirals, which urges the identification of potential therapeutic targets and the design of CCHF therapies with greater effort. In this article, we discuss the current progress and some future directions in the development of antiviral strategies against CCHFV.
Collapse
Affiliation(s)
- Shiyu Dai
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China;
| | - Fei Deng
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China;
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
- Correspondence: (F.D.); (H.W.); (Y.N.); Tel./Fax: +86-27-8719-8465 (F.D.); +86-27-8719-9353 (H.W.); +86-27-8719-7200 (Y.N.)
| | - Hualin Wang
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China;
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
- Correspondence: (F.D.); (H.W.); (Y.N.); Tel./Fax: +86-27-8719-8465 (F.D.); +86-27-8719-9353 (H.W.); +86-27-8719-7200 (Y.N.)
| | - Yunjia Ning
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China;
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
- Correspondence: (F.D.); (H.W.); (Y.N.); Tel./Fax: +86-27-8719-8465 (F.D.); +86-27-8719-9353 (H.W.); +86-27-8719-7200 (Y.N.)
| |
Collapse
|
10
|
Urata S, Yasuda J, Iwasaki M. Loperamide Inhibits Replication of Severe Fever with Thrombocytopenia Syndrome Virus. Viruses 2021; 13:v13050869. [PMID: 34068464 PMCID: PMC8150324 DOI: 10.3390/v13050869] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Severe fever with thrombocytopenia syndrome (SFTS) is an emerging tick-borne infectious disease caused by the SFTS virus (SFTSV). SFTS is mainly prevalent in East Asia. It has a mortality rate of up to 30%, and there is no approved treatment against the disease. In this study, we evaluated the effect of loperamide, an antidiarrheal and antihyperalgesic agent, on the propagation of SFTSV in a cell culture system. METHODS SFTSV-infected human cell lines were exposed to loperamide, and viral titers were evaluated. To clarify the mode of action of loperamide, several chemical compounds having shared targets with loperamide were used. Calcium imaging was also performed to understand whether loperamide treatment affected calcium influx. RESULTS Loperamide inhibited SFTSV propagation in several cell lines. It inhibited SFTSV in the post-entry step and restricted calcium influx into the cell. Furthermore, nifedipine, a calcium channel inhibitor, also blocked post-entry step of SFTSV infection. CONCLUSIONS Loperamide inhibits SFTSV propagation mainly by restraining calcium influx into the cytoplasm. This indicates that loperamide, a Food and Drug Administration (FDA)-approved drug, has the potential for being used as a treatment option against SFTS.
Collapse
Affiliation(s)
- Shuzo Urata
- National Research Center for the Control and Prevention of Infectious Diseases (CCPID), Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan;
- Department of Emerging Infectious Diseases, Institute of Tropical Medicine (NEKKEN), Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
- Correspondence: ; Tel.: +81-95-819-7970
| | - Jiro Yasuda
- National Research Center for the Control and Prevention of Infectious Diseases (CCPID), Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan;
- Department of Emerging Infectious Diseases, Institute of Tropical Medicine (NEKKEN), Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Masaharu Iwasaki
- Laboratory of Emerging Viral Diseases, International Research Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Osaka 565-0871, Japan;
| |
Collapse
|
11
|
García-Serradilla M, Risco C. Light and electron microscopy imaging unveils new aspects of the antiviral capacity of silver nanoparticles in bunyavirus-infected cells. Virus Res 2021; 302:198444. [PMID: 33961898 DOI: 10.1016/j.virusres.2021.198444] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 04/21/2021] [Accepted: 05/01/2021] [Indexed: 10/21/2022]
Abstract
Drug repurposing is an important source of new antivirals because many compounds used to treat a variety of pathologies also hamper viral infections. Habitually, silver nanoparticles (AgNPs) have been used to treat bacterial and fungal infections and their antiviral properties have been also reported. In this work, we have studied the antiviral capacity of AgNPs in cells infected with Bunyamwera virus (BUNV), the prototype of the Bunyavirales order. This group of viruses contains important pathogens for humans, animals and plants. Incubation of BUNV-infected Vero cells with non-toxic concentrations of AgNPs, reduced the production of extracellular infectious viruses in up to three orders of magnitude. With a combination of imaging techniques, we have visualized the intracellular distribution of AgNPs in mock- and BUNV-infected cells and studied their effects on intracellular organelles. In mock-infected cells and at short times post-incubation, AgNPs were detected inside nuclei and mitochondria by transmission electron microscopy (TEM). At long times post-treatment, they accumulated inside lysosome-like organelles. Cell compartments did not exhibit any appreciable ultrastructural alterations after incubation with AgNPs. In BUNV-infected cells, AgNPs attached to extracellular virions, that showed a disrupted morphology. Inside cells, they were detected inside the nucleus, in mitochondria and around characteristic Golgi-associated, single-membrane spherules. These membranous structures are the replication organelles (ROs) of bunyaviruses and contain active viral replication complexes (VRCs). Compared to normal spherules that are round, compact and have an electron-dense core, spherules in AgNPs-treated cells were deformed and their core was electron-lucent. Interestingly, in BUNV-infected cells treated with the typical antiviral ribavirin (RBV), spherules with VRCs exhibit also an anomalous morphology and an electron-lucent core. Both AgNPs and RBV might interfere with BUNV-induced dismantling of cell nucleoli and with the intercellular propagation of large groups of virions, a mechanism of BUNV transmission observed for the first time in cultured cells. Our results point to silver nanoparticles as good candidates for antiviral therapy, either alone or in combination with other antiviral drugs, such as RBV-related compounds.
Collapse
Affiliation(s)
- Moisés García-Serradilla
- Cell Structure Laboratory, National Center for Biotechnology, CNB-CSIC, Campus UAM, Cantoblanco, 28049, Madrid, Spain
| | - Cristina Risco
- Cell Structure Laboratory, National Center for Biotechnology, CNB-CSIC, Campus UAM, Cantoblanco, 28049, Madrid, Spain.
| |
Collapse
|
12
|
Jang Y, Shin JS, Lee MK, Jung E, An T, Kim UI, Kim K, Kim M. Comparison of Antiviral Activity of Gemcitabine with 2'-Fluoro-2'-Deoxycytidine and Combination Therapy with Remdesivir against SARS-CoV-2. Int J Mol Sci 2021; 22:ijms22041581. [PMID: 33557278 PMCID: PMC7915419 DOI: 10.3390/ijms22041581] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 01/28/2021] [Accepted: 02/01/2021] [Indexed: 12/15/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the coronavirus disease 2019 (COVID-19) pandemic. The virus still spreads globally through human-to-human transmission. Nevertheless, there are no specific treatments clinically approved. This study aimed to compare antiviral activity of gemcitabine and its analogue 2′-fluoro-2′-deoxycytidine (2FdC) against SARS-CoV-2 as well as cytotoxicity in vitro. Fluorescent image-based antiviral assays revealed that gemcitabine was highly potent, with a 50% effective concentration (EC50) of 1.2 μM, more active than the well-known nucleoside monophosphate remdesivir (EC50 = 35.4 μM). In contrast, 2FdC was marginally active (EC50 = 175.2 μM). For all three compounds, the 50% cytotoxic concentration (CC50) values were over 300 μM toward Vero CCL-81 cells. Western blot and quantitative reverse-transcription polymerase chain reaction analyses verified that gemcitabine blocked viral protein expression in virus-infected cells, not only Vero CCL-81 cells but also Calu-3 human lung epithelial cells in a dose-dependent manner. It was found that gemcitabine has a synergistic effect when combined with remdesivir. This report suggests that the difluoro group of gemcitabine is critical for the antiviral activity and that its combination with other evaluated antiviral drugs, such as remdesivir, could be a desirable option to treat SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Yejin Jang
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Korea; (Y.J.); (J.S.S.); (M.K.L.); (E.J.); (T.A.)
| | - Jin Soo Shin
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Korea; (Y.J.); (J.S.S.); (M.K.L.); (E.J.); (T.A.)
| | - Myoung Kyu Lee
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Korea; (Y.J.); (J.S.S.); (M.K.L.); (E.J.); (T.A.)
| | - Eunhye Jung
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Korea; (Y.J.); (J.S.S.); (M.K.L.); (E.J.); (T.A.)
| | - Timothy An
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Korea; (Y.J.); (J.S.S.); (M.K.L.); (E.J.); (T.A.)
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon 34134, Korea
| | - Uk-Il Kim
- Research and Development Center, ST Pharm Co., Ltd., Seoul 01694, Korea;
| | - Kyungjin Kim
- Research and Development Center, ST Pharm Co., Ltd., Seoul 01694, Korea;
- Correspondence: (K.K.); (M.K.)
| | - Meehyein Kim
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Korea; (Y.J.); (J.S.S.); (M.K.L.); (E.J.); (T.A.)
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon 34134, Korea
- Correspondence: (K.K.); (M.K.)
| |
Collapse
|
13
|
Yu P, Wang Y, Li Y, Li Y, Miao Z, Peppelenbosch MP, Pan Q. 2'-Fluoro-2'-deoxycytidine inhibits murine norovirus replication and synergizes MPA, ribavirin and T705. Arch Virol 2020; 165:2605-2613. [PMID: 32770483 PMCID: PMC7414258 DOI: 10.1007/s00705-020-04759-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 06/30/2020] [Indexed: 11/22/2022]
Abstract
Noroviruses are the main causative agents of acute viral gastroenteritis worldwide. However, no vaccine or specific antiviral treatment is available, imposing a heavy global health burden. The nucleoside analogue 2’-fluoro-2’-deoxycytidine (2’-FdC) has been reported to have broad antiviral activity. Here, we report that 2’-FdC significantly inhibits murine norovirus replication in macrophages. This effect was partially reversed by exogenous supplementation of cytidine triphosphate. The combination of 2’-FdC with mycophenolic acid, ribavirin or favipiravir (T705) exerts synergistic antiviral effects. These results indicate that 2’-FdC is a potential candidate for antiviral drug development against norovirus infection.
Collapse
Affiliation(s)
- Peifa Yu
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Room Na-1005, 's-Gravendijkwal 230, 3015 CE, Rotterdam, The Netherlands
| | - Yining Wang
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Room Na-1005, 's-Gravendijkwal 230, 3015 CE, Rotterdam, The Netherlands
| | - Yunlong Li
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Room Na-1005, 's-Gravendijkwal 230, 3015 CE, Rotterdam, The Netherlands
| | - Yang Li
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Room Na-1005, 's-Gravendijkwal 230, 3015 CE, Rotterdam, The Netherlands
| | - Zhijiang Miao
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Room Na-1005, 's-Gravendijkwal 230, 3015 CE, Rotterdam, The Netherlands
| | - Maikel P Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Room Na-1005, 's-Gravendijkwal 230, 3015 CE, Rotterdam, The Netherlands
| | - Qiuwei Pan
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Room Na-1005, 's-Gravendijkwal 230, 3015 CE, Rotterdam, The Netherlands.
| |
Collapse
|
14
|
Efficient functional screening of a cellular cDNA library to identify severe fever with thrombocytopenia syndrome virus entry factors. Sci Rep 2020; 10:5996. [PMID: 32265454 PMCID: PMC7138800 DOI: 10.1038/s41598-020-62876-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 03/18/2020] [Indexed: 01/15/2023] Open
Abstract
The identification of host cell factors for virus entry is useful for the molecular explanation of viral tropisms and often leads to a more profound understanding of virus-induced diseases. Severe fever with thrombocytopenia syndrome (SFTS) is an emerging infectious disease caused by SFTS virus. No countermeasures against the disease exist. In this report, we show an efficient method using virus-like particles for the functional screening of a cellular cDNA library to identify SFTS virus entry factors. Two variants encoding dendritic cell-specific ICAM-3 grabbing non-integrin related (DC-SIGNR), a calcium-dependent lectin known to enhance SFTS virus infection, were successfully identified from a human liver cDNA library. We will discuss applications for yet unidentified factor(s) for SFTS virus entry and for entry factor(s) for other viruses related to SFTS virus.
Collapse
|
15
|
Bugert JJ, Hucke F, Zanetta P, Bassetto M, Brancale A. Antivirals in medical biodefense. Virus Genes 2020; 56:150-167. [PMID: 32076918 PMCID: PMC7089181 DOI: 10.1007/s11262-020-01737-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 01/20/2020] [Indexed: 02/07/2023]
Abstract
The viruses historically implicated or currently considered as candidates for misuse in bioterrorist events are poxviruses, filoviruses, bunyaviruses, orthomyxoviruses, paramyxoviruses and a number of arboviruses causing encephalitis, including alpha- and flaviviruses. All these viruses are of concern for public health services when they occur in natural outbreaks or emerge in unvaccinated populations. Recent events and intelligence reports point to a growing risk of dangerous biological agents being used for nefarious purposes. Public health responses effective in natural outbreaks of infectious disease may not be sufficient to deal with the severe consequences of a deliberate release of such agents. One important aspect of countermeasures against viral biothreat agents are the antiviral treatment options available for use in post-exposure prophylaxis. These issues were adressed by the organizers of the 16th Medical Biodefense Conference, held in Munich in 2018, in a special session on the development of drugs to treat infections with viruses currently perceived as a threat to societies or associated with a potential for misuse as biothreat agents. This review will outline the state-of-the-art methods in antivirals research discussed and provide an overview of antiviral compounds in the pipeline that are already approved for use or still under development.
Collapse
Affiliation(s)
- J J Bugert
- Bundeswehr Institute for Microbiology, Neuherbergstraße 11, 80937, Munich, Germany.
| | - F Hucke
- Bundeswehr Institute for Microbiology, Neuherbergstraße 11, 80937, Munich, Germany
| | - P Zanetta
- Bundeswehr Institute for Microbiology, Neuherbergstraße 11, 80937, Munich, Germany
| | - M Bassetto
- Department of Chemistry, Swansea University, Swansea, SA2 8PP, UK
| | - A Brancale
- Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, CF10 3NB, UK
| |
Collapse
|
16
|
Takayama-Ito M, Saijo M. Antiviral Drugs Against Severe Fever With Thrombocytopenia Syndrome Virus Infection. Front Microbiol 2020; 11:150. [PMID: 32117168 PMCID: PMC7026129 DOI: 10.3389/fmicb.2020.00150] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 01/22/2020] [Indexed: 12/15/2022] Open
Abstract
Severe fever with thrombocytopenia syndrome (SFTS) is an emerging tick-borne infectious disease caused by SFTS virus (SFTSV), which is a novel bunyavirus. SFTSV was first isolated from patients who presented with fever, thrombocytopenia, leukocytopenia, and multiorgan dysfunction in China. Subsequently, it was found to be widely distributed in Southeast Asia (Korea, Japan, and Vietnam). SFTSV can be transmitted not only from ticks but also from domestic animals, companion animals, and humans. Because the case fatality rate of SFTS is high (6–30%), development of specific and effective treatment for SFTS is required. Studies of potential antiviral drugs for SFTS-specific therapy have been conducted on existing or newly discovered agents in vitro and in vivo, with ribavirin and favipiravir being the most promising candidates. While animal experiments and retrospective studies have demonstrated the limited efficacy of ribavirin, it was also speculated that ribavirin would be effective in patients with a viral load <1 × 106 copies/mL. Favipiravir showed higher efficacy than ribavirin against SFTSV in in vitro assays and greater efficacy in animal models, even administrated 3 days after the virus inoculation. Although clinical trials evaluating the efficacy of favipiravir in SFTS patients in Japan are underway, this has yet to be confirmed. Other drugs, including hexachlorophene, calcium channel blockers, 2′-fluoro-2′-deoxycytidine, caffeic acid, amodiaquine, and interferons, have also been evaluated for their inhibitory efficacy against SFTSV. Among them, calcium channel blockers are promising because in addition to their efficacy in vitro and in vivo, retrospective clinical data have indicated that nifedipine, one of the calcium channel blockers, reduced the case fatality rate by >5-fold. Although further research is necessary to develop SFTS-specific therapy, considerable progress has been achieved in this area. Here we summarize and discuss recent advances in antiviral drugs against SFTSV.
Collapse
Affiliation(s)
- Mutsuyo Takayama-Ito
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Masayuki Saijo
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| |
Collapse
|
17
|
Javelle E, Lesueur A, Pommier de Santi V, de Laval F, Lefebvre T, Holweck G, Durand GA, Leparc-Goffart I, Texier G, Simon F. The challenging management of Rift Valley Fever in humans: literature review of the clinical disease and algorithm proposal. Ann Clin Microbiol Antimicrob 2020; 19:4. [PMID: 31969141 PMCID: PMC6977312 DOI: 10.1186/s12941-020-0346-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 01/12/2020] [Indexed: 01/01/2023] Open
Abstract
Rift Valley Fever (RVF) is an emerging zoonotic arbovirus with a complex cycle of transmission that makes difficult the prediction of its expansion. Recent outbreaks outside Africa have led to rediscover the human disease but it remains poorly known. The wide spectrum of acute and delayed manifestations with potential unfavorable outcome much complicate the management of suspected cases and prediction of morbidity and mortality during an outbreak. We reviewed literature data on bio-clinical characteristics and treatments of RVF human illness. We identified gaps in the field and provided a practical algorithm to assist clinicians in the cases assessment, determination of setting of care and prolonged follow-up.
Collapse
Affiliation(s)
- Emilie Javelle
- Laveran Military Teaching Hospital, CS500413384, Marseille Cedex 13, France. .,IRD, AP-HM, SSA, VITROME, IHU-Méditerranée Infection, Aix Marseille Univ, Marseille, France.
| | - Alexandre Lesueur
- Laveran Military Teaching Hospital, CS500413384, Marseille Cedex 13, France
| | - Vincent Pommier de Santi
- IRD, AP-HM, SSA, VITROME, IHU-Méditerranée Infection, Aix Marseille Univ, Marseille, France.,French Armed Forces Centre for Epidemiology and Public Health (CESPA), Marseille, France
| | - Franck de Laval
- French Armed Forces Centre for Epidemiology and Public Health (CESPA), Marseille, France.,INSERM, IRD, SESSTIM, Sciences Economiques & Sociales de la Santé & Traitement de l'Information Médicale, Aix Marseille Univ, Marseille, France
| | - Thibault Lefebvre
- French Military Health Service, RSMA Medical Unit, Paris, Mayotte, France
| | - Guillaume Holweck
- Laveran Military Teaching Hospital, CS500413384, Marseille Cedex 13, France
| | - Guillaume André Durand
- French Armed Forces Biomedical Research Institute (IRBA)-CNR des arbovirus-IHU Méditerranée Infection, Marseille, France.,IRD 190, Inserm 1207, IHU Méditerranée Infection, AP-HM, UVE, Aix-Marseille Univ, Marseille, France
| | - Isabelle Leparc-Goffart
- French Armed Forces Biomedical Research Institute (IRBA)-CNR des arbovirus-IHU Méditerranée Infection, Marseille, France.,IRD 190, Inserm 1207, IHU Méditerranée Infection, AP-HM, UVE, Aix-Marseille Univ, Marseille, France
| | - Gaëtan Texier
- IRD, AP-HM, SSA, VITROME, IHU-Méditerranée Infection, Aix Marseille Univ, Marseille, France.,French Armed Forces Centre for Epidemiology and Public Health (CESPA), Marseille, France
| | - Fabrice Simon
- Laveran Military Teaching Hospital, CS500413384, Marseille Cedex 13, France.,IRD 190, Inserm 1207, IHU Méditerranée Infection, AP-HM, UVE, Aix-Marseille Univ, Marseille, France
| |
Collapse
|
18
|
Calcium channel blockers reduce severe fever with thrombocytopenia syndrome virus (SFTSV) related fatality. Cell Res 2019; 29:739-753. [PMID: 31444469 DOI: 10.1038/s41422-019-0214-z] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 07/23/2019] [Indexed: 11/09/2022] Open
Abstract
Severe fever with thrombocytopenia syndrome (SFTS), an emerging tick-borne infectious disease caused by a novel phlebovirus (SFTS virus, SFTSV), was listed among the top 10 priority infectious diseases by the World Health Organization due to its high fatality of 12%-50% and possibility of pandemic transmission. Currently, effective anti-SFTSV intervention remains unavailable. Here, by screening a library of FDA-approved drugs, we found that benidipine hydrochloride, a calcium channel blocker (CCB), inhibited SFTSV replication in vitro. Benidipine hydrochloride was revealed to inhibit virus infection through impairing virus internalization and genome replication. Further experiments showed that a broad panel of CCBs, including nifedipine, inhibited SFTSV infection. The anti-SFTSV effect of these two CCBs was further analyzed in a humanized mouse model in which CCB treatment resulted in reduced viral load and decreased fatality rate. Importantly, by performing a retrospective clinical investigation on a large cohort of 2087 SFTS patients, we revealed that nifedipine administration enhanced virus clearance, improved clinical recovery, and remarkably reduced the case fatality rate by >5-fold. These findings are highly valuable for developing potential host-oriented therapeutics for SFTS and other lethal acute viral infections known to be inhibited by CCBs in vitro.
Collapse
|
19
|
In silico structural elucidation of RNA-dependent RNA polymerase towards the identification of potential Crimean-Congo Hemorrhagic Fever Virus inhibitors. Sci Rep 2019; 9:6809. [PMID: 31048746 PMCID: PMC6497722 DOI: 10.1038/s41598-019-43129-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 04/17/2019] [Indexed: 01/05/2023] Open
Abstract
The Crimean-Congo Hemorrhagic Fever virus (CCHFV) is a segmented negative single-stranded RNA virus (-ssRNA) which causes severe hemorrhagic fever in humans with a mortality rate of ~50%. To date, no vaccine has been approved. Treatment is limited to supportive care with few investigational drugs in practice. Previous studies have identified viral RNA dependent RNA Polymerase (RdRp) as a potential drug target due to its significant role in viral replication and transcription. Since no crystal structure is available yet, we report the structural elucidation of CCHFV-RdRp by in-depth homology modeling. Even with low sequence identity, the generated model suggests a similar overall structure as previously reported RdRps. More specifically, the model suggests the presence of structural/functional conserved RdRp motifs for polymerase function, the configuration of uniform spatial arrangement of core RdRp sub-domains, and predicted positively charged entry/exit tunnels, as seen in sNSV polymerases. Extensive pharmacophore modeling based on per-residue energy contribution with investigational drugs allowed the concise mapping of pharmacophoric features and identified potential hits. The combination of pharmacophoric features with interaction energy analysis revealed functionally important residues in the conserved motifs together with in silico predicted common inhibitory binding modes with highly potent reference compounds.
Collapse
|
20
|
Yuan S, Chan JFW, Ye ZW, Wen L, Tsang TGW, Cao J, Huang J, Chan CCY, Chik KKH, Choi GKY, Cai JP, Yin F, Chu H, Liang M, Jin DY, Yuen KY. Screening of an FDA-Approved Drug Library with a Two-Tier System Identifies an Entry Inhibitor of Severe Fever with Thrombocytopenia Syndrome Virus. Viruses 2019; 11:v11040385. [PMID: 31027241 PMCID: PMC6520937 DOI: 10.3390/v11040385] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 04/23/2019] [Accepted: 04/24/2019] [Indexed: 12/27/2022] Open
Abstract
Severe fever with thrombocytopenia syndrome virus (SFTSV) is an emerging tick-borne bunyavirus that causes severe disease in humans with case-fatality rates of up to 30%. There are currently very limited treatment options for SFTSV infection. We conducted a drug repurposing program by establishing a two-tier test system to rapidly screen a Food and Drug Administration- (FDA)-approved drug library for drug compounds with anti-SFTSV activity in vitro. We identified five drug compounds that inhibited SFTSV replication at low micromolar concentrations, including hexachlorophene, triclosan, regorafenib, eltrombopag, and broxyquinoline. Among them, hexachlorophene was the most potent with an IC50 of 1.3 ± 0.3 µM and a selectivity index of 18.7. Mechanistic studies suggested that hexachlorophene was a virus entry inhibitor, which impaired SFTSV entry into host cells by interfering with cell membrane fusion. Molecular docking analysis predicted that the binding of hexachlorophene with the hydrophobic pocket between domain I and domain III of the SFTSV Gc glycoprotein was highly stable. The novel antiviral activity and mechanism of hexachlorophene in this study would facilitate the use of hexachlorophene as a lead compound to develop more entry inhibitors with higher anti-SFTSV potency and lower toxicity.
Collapse
Affiliation(s)
- Shuofeng Yuan
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Jasper Fuk-Woo Chan
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Hainan Medical University, Haikou 571101, China, and The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Zi-Wei Ye
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Lei Wen
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Terance Gi-Wai Tsang
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Jianli Cao
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Jingjing Huang
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Chris Chun-Yiu Chan
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Kenn Ka-Heng Chik
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Garnet Kwan-Yue Choi
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Jian-Piao Cai
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Feifei Yin
- Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Hainan Medical University, Haikou 571101, China, and The University of Hong Kong, Pokfulam, Hong Kong, China.
- Department of Pathogen Biology, Hainan Medical University, Haikou 571101, China.
- Key Laboratory of Translational Tropical Medicine, Hainan Medical University, Haikou 571101, China.
| | - Hin Chu
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Mifang Liang
- Key Laboratory for Medical Virology and National Institute for Viral Disease Control and Prevention, Chinese Centre for Disease Control and Prevention, Beijing 102206, China.
| | - Dong-Yan Jin
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Kwok-Yung Yuen
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Hainan Medical University, Haikou 571101, China, and The University of Hong Kong, Pokfulam, Hong Kong, China.
- The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong, China.
| |
Collapse
|
21
|
Flint M, Chatterjee P, Lin DL, McMullan LK, Shrivastava-Ranjan P, Bergeron É, Lo MK, Welch SR, Nichol ST, Tai AW, Spiropoulou CF. A genome-wide CRISPR screen identifies N-acetylglucosamine-1-phosphate transferase as a potential antiviral target for Ebola virus. Nat Commun 2019; 10:285. [PMID: 30655525 PMCID: PMC6336797 DOI: 10.1038/s41467-018-08135-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 12/19/2018] [Indexed: 12/28/2022] Open
Abstract
There are no approved therapies for Ebola virus infection. Here, to find potential therapeutic targets, we perform a screen for genes essential for Ebola virus (EBOV) infection. We identify GNPTAB, which encodes the α and β subunits of N-acetylglucosamine-1-phosphate transferase. We show that EBOV infection of a GNPTAB knockout cell line is impaired, and that this is reversed by reconstituting GNPTAB expression. Fibroblasts from patients with mucolipidosis II, a disorder associated with mutations in GNPTAB, are refractory to EBOV, whereas cells from their healthy parents support infection. Impaired infection correlates with loss of the expression of cathepsin B, known to be essential for EBOV entry. GNPTAB activity is dependent upon proteolytic cleavage by the SKI-1/S1P protease. Inhibiting this protease with the small-molecule PF-429242 blocks EBOV entry and infection. Disruption of GNPTAB function may represent a strategy for a host-targeted therapy for EBOV. Genetic screens are important tools to identify host factors associated with viral infections. Here, Flint et al. perform a genome-wide CRISPR screen using infectious Ebola virus (EBOV) and show that the host transferase GNPTAB is required for EBOV infection and a potential target for antiviral therapies
Collapse
Affiliation(s)
- Mike Flint
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, MS G-14, Atlanta, GA, 30329, USA.
| | - Payel Chatterjee
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, MS G-14, Atlanta, GA, 30329, USA
| | - David L Lin
- Department of Microbiology & Immunology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Laura K McMullan
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, MS G-14, Atlanta, GA, 30329, USA
| | - Punya Shrivastava-Ranjan
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, MS G-14, Atlanta, GA, 30329, USA
| | - Éric Bergeron
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, MS G-14, Atlanta, GA, 30329, USA
| | - Michael K Lo
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, MS G-14, Atlanta, GA, 30329, USA
| | - Stephen R Welch
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, MS G-14, Atlanta, GA, 30329, USA
| | - Stuart T Nichol
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, MS G-14, Atlanta, GA, 30329, USA
| | - Andrew W Tai
- Department of Microbiology & Immunology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA.,Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Christina F Spiropoulou
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, MS G-14, Atlanta, GA, 30329, USA.
| |
Collapse
|