Beyond the Surface: Endocytosis of Mosquito-Borne Flaviviruses
Abstract
:1. Mosquito-Borne Flaviviruses: Replication, Disease, and Epidemiology
1.1. Flavivirus Life Cycle
1.2. Dengue Virus (DENV)
1.3. Yellow Fever Virus (YFV)
1.4. Zika Virus (ZIKV)
1.5. West Nile Virus (WNV)
1.6. Japanese Encephalitis Virus (JEV)
2. Mosquito-Borne Flavivirus Receptors
3. Endocytic Pathways Mediating Mosquito-Borne Flavivirus Internalization
3.1. Clathrin-Mediated Endocytosis
3.2. Flaviviruses and Clathrin-Mediated Endocytosis
3.3. Flavivirus Entry: Beyond Traditional Clathrin-Mediated Endocytosis
4. Targeting Endocytic Entry: An Avenue for Flavivirus Antivirals
5. Summary
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Huang, Y.-J.S.; Higgs, S.; Horne, K.M.; van Landingham, D.L. Flavivirus-mosquito interactions. Viruses 2014, 6, 4703–4730. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kemenesi, G.; Bányai, K. Tick-borne flaviviruses, with a focus on powassan virus. Clin. Microbiol. Rev. 2018, 32, 106–117. [Google Scholar] [CrossRef] [Green Version]
- Moureau, G.; Cook, S.; Lemey, P.; Nougairede, A.; Forrester, N.L.; Khasnatinov, M.; Charrel, R.N.; Firth, A.E.; Gould, E.A.; de Lamballerie, X. New insights into flavivirus evolution, taxonomy and biogeographic history, extended by analysis of canonical and alternative coding sequences. PLoS ONE 2015, 10, e0117849. [Google Scholar] [CrossRef] [Green Version]
- Messina, J.P.; Brady, O.J.; Scott, T.W.; Zou, C.; Pigott, D.M.; Duda, K.A.; Bhatt, S.; Katzelnick, L.; Howes, R.E.; Battle, K.E.; et al. Global spread of dengue virus types: Mapping the 70-year history. Trends Microbiol. 2014, 22, 138–146. [Google Scholar] [CrossRef] [Green Version]
- Ganeshkumar, P.; Murhekar, M.; Poornima, V.; Saravanakumar, V.; Sukumaran, K.; Anandaselvasankar, A.; John, D.; Mehendale, S.M. Dengue infection in India: A systematic review and meta-analysis. PLoS Negl. Trop. Dis. 2018, 12, e0006618. [Google Scholar] [CrossRef] [Green Version]
- Hadfield, J.; Brito, A.F.; Swetnam, D.M.; Vogels, C.B.; Tokarz, R.E.; Andersen, K.G.; Smith, R.C.; Bedford, T.; Grubaugh, N. Twenty years of West Nile virus spread and evolution in the Americas visualized by Nextstrain. PLoS Pathog. 2019, 15, e1008042. [Google Scholar] [CrossRef] [Green Version]
- Giovanetti, M.; de Mendonça, M.C.L.; Fonseca, V.; Mares-Guia, M.A.; Fabri, A.; Xavier, J.; de Jesus, J.G.; Gräf, T.; Rodrigues, C.D.D.S.; dos Santos, C.C.; et al. Yellow fever virus reemergence and spread in Southeast Brazil, 2016–2019. J. Virol. 2019, 94, 01623-19. [Google Scholar] [CrossRef]
- Zhang, H.; Rehman, M.U.; Li, K.; Luo, H.; Lan, Y.; Nabi, F.; Zhang, L.; Iqbal, M.K.; Zhu, S.; Javed, M.T.; et al. Epidemiologic survey of Japanese encephalitis virus infection, Tibet, China, 2015. Emerg. Infect. Dis. 2017, 23, 1023–1024. [Google Scholar] [CrossRef]
- Metsky, H.C.; Matranga, C.B.; Wohl, S.; Schaffner, S.F.; Freije, C.A.; Winnicki, S.M.; West, K.; Quigley, J.E.; Baniecki, M.L.; Gladden-Young, A.; et al. Zika virus evolution and spread in the Americas. Nature 2017, 546, 411–415. [Google Scholar] [CrossRef] [Green Version]
- Halstead, S.B. Dengvaxia sensitizes seronegatives to vaccine enhanced disease regardless of age. Vaccine 2017, 35, 6355–6358. [Google Scholar] [CrossRef]
- Fatima, K.; Syed, N.I. Dengvaxia controversy: Impact on vaccine hesitancy. J. Glob. Health 2018, 8, 010312. [Google Scholar] [CrossRef] [PubMed]
- Wilder-Smith, A. The first licensed dengue vaccine: Can it be used in travelers? Curr. Opin. Infect. Dis. 2019, 32, 394–400. [Google Scholar] [CrossRef] [PubMed]
- Yun, S.I.; Lee, Y.M. Japanese encephalitis: The virus and vaccines. Hum. Vaccin. Immunother. 2014, 10, 263–279. [Google Scholar] [CrossRef] [Green Version]
- Li, X.; Ma, S.-J.; Liu, X.; Jiang, L.-N.; Zhou, J.-H.; Xiong, Y.-Q.; Ding, H.; Chen, Q. Immunogenicity and safety of currently available Japanese encephalitis vaccines: A systematic review. Hum. Vaccin. Immunother. 2014, 10, 3579–3593. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Barrett, A.D. Yellow fever live attenuated vaccine: A very successful live attenuated vaccine but still we have problems controlling the disease. Vaccine 2017, 35, 5951–5955. [Google Scholar] [CrossRef] [PubMed]
- Casey, R.M.; Harris, J.B.; Ahuka-Mundeke, S.; Dixon, M.G.; Kizito, G.M.; Nsele, P.M.; Umutesi, G.; Laven, J.; Kosoy, O.; Paluku, G.; et al. Immunogenicity of fractional-dose vaccine during a yellow fever outbreak—final report. N. Engl. J. Med. 2019, 381, 444–454. [Google Scholar] [CrossRef] [PubMed]
- Pierson, T.C.; Diamond, M.S. The continued threat of emerging flaviviruses. Nat. Microbiol. 2020, 5, 796–812. [Google Scholar] [CrossRef] [PubMed]
- Kaufmann, B.; Rossmann, M.G. Molecular mechanisms involved in the early steps of flavivirus cell entry. Microbes Infect. 2011, 13, 1–9. [Google Scholar] [CrossRef] [Green Version]
- Bhatt, S.; Gething, P.W.; Brady, O.J.; Messina, J.P.; Farlow, A.W.; Moyes, C.L.; Drake, J.M.; Brownstein, J.S.; Hoen, A.G.; Sankoh, O.; et al. The global distribution and burden of dengue. Nature 2013, 496, 504–507. [Google Scholar] [CrossRef]
- Vasilakis, N.; Cardosa, J.; Hanley, K.A.; Holmes, E.C.; Weaver, S.C. Fever from the forest: Prospects for the continued emergence of sylvatic dengue virus and its impact on public health. Nat. Rev. Microbiol. 2011, 9, 532–541. [Google Scholar] [CrossRef]
- Brady, O.J.; Hay, S.I. The global expansion of dengue: How Aedes aegypti mosquitoes enabled the first pandemic arbovirus. Annu. Rev. Entomol. 2020, 65, 191–208. [Google Scholar] [CrossRef] [Green Version]
- Gubler, D.J. Dengue, Urbanization and globalization: The unholy trinity of the 21st Century. Trop. Med. Health 2011, 39, S3–S11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kraemer, M.U.G.; Reiner, R.C.; Brady, O.J.; Messina, J.P.; Gilbert, M.; Pigott, D.M.; Yi, D.; Johnson, K.; Earl, L.; Marczak, L.B.; et al. Past and future spread of the arbovirus vectors Aedes aegypti and Aedes albopictus. Nat. Microbiol. 2019, 4, 854–863. [Google Scholar] [CrossRef] [PubMed]
- Katzelnick, L.C.; Fonville, J.M.; Gromowski, G.D.; Arriaga, J.B.; Green, A.M.; James, S.L.; Lau, L.; Montoya, M.; Wang, C.; VanBlargan, L.A.; et al. Dengue viruses cluster antigenically but not as discrete serotypes. Science 2015, 349, 1338–1343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guzman, M.G.; Gubler, D.J.; Izquierdo, A.; Martinez, E.; Halstead, S.B. Dengue infection. Nat. Rev. Dis. Primers 2016, 2, 16055. [Google Scholar] [CrossRef] [PubMed]
- Halstead, S.B.; Nimmannitya, S.; Cohen, S.N. Observations related to pathogenesis of dengue hemorrhagic fever. IV. Relation of disease severity to antibody response and virus recovered. Yale J. Biol. Med. 1970, 42, 311–328. [Google Scholar] [PubMed]
- Kliks, S.C.; Brandt, W.E.; Wahl, L.; Nisalak, A.; Burke, D.S. Antibody-dependent enhancement of dengue virus growth in human monocytes as a risk factor for dengue hemorrhagic fever. Am. J. Trop. Med. Hyg. 1989, 40, 444–451. [Google Scholar] [CrossRef]
- Katzelnick, L.C.; Gresh, L.; Halloran, M.E.; Mercado, J.C.; Kuan, G.; Gordon, A.; Balmaseda, A.; Harris, E. Antibody-dependent enhancement of severe dengue disease in humans. Science 2017, 358, 929–932. [Google Scholar] [CrossRef] [Green Version]
- Oh-Ainle, M.; Balmaseda, A.; Macalalad, A.R.; Tellez, Y.; Zody, M.C.; Saborío, S.; Nuñez, A.; Lennon, N.J.; Birren, B.W.; Gordon, A.; et al. Dynamics of dengue disease severity determined by the interplay between viral genetics and serotype-specific immunity. Sci. Transl. Med. 2011, 3, 114ra128. [Google Scholar] [CrossRef] [Green Version]
- Simmons, C.P.; Chau, T.N.; Thuy, T.T.; Tuan, N.M.; Hoang, D.M.; Thien, N.T.; le Lien, B.; Quy, N.T.; Hieu, N.T.; Hien, T.T.; et al. Maternal antibody and viral factors in the pathogenesis of dengue virus in infants. J. Infect. Dis. 2007, 196, 416–424. [Google Scholar] [CrossRef]
- World Health Organization. Global Strategy for Dengue Prevention and Control, 2012–2020. Available online: https://www.who.int/denguecontrol/9789241504034/en/ (accessed on 25 August 2020).
- Monath, T.P. Yellow fever: An update. Lancet Infect. Dis. 2001, 1, 11–20. [Google Scholar] [CrossRef]
- Johansson, M.A.; Vasconcelos, P.F.C.; Staples, J.E. The whole iceberg: Estimating the incidence of yellow fever virus infection from the number of severe cases. Trans. R. Soc. Trop. Med. Hyg. 2014, 108, 482–487. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hamer, D.H.; Angelo, K.; Caumes, E.; van Genderen, P.J.; Florescu, S.A.; Popescu, C.P.; Perret, C.; McBride, A.; Checkley, A.; Ryan, J.; et al. Fatal yellow fever in travelers to Brazil, 2018. Morb. Mortal. Wkly. Report 2018, 67, 340–341. [Google Scholar] [CrossRef] [PubMed]
- Zanotto, P.M.; Gould, E.A.; Gao, G.F.; Harvey, P.H.; Holmes, E.C. Population dynamics of flaviviruses revealed by molecular phylogenies. Proc. Natl. Acad. Sci. USA 1996, 93, 548–553. [Google Scholar] [CrossRef] [Green Version]
- Bryant, J.E.; Holmes, E.C.; Barrett, A.D. Out of Africa: A molecular perspective on the introduction of yellow fever virus into the Americas. PLoS Pathog. 2007, 3, e75. [Google Scholar] [CrossRef] [Green Version]
- Patterson, K. Yellow fever epidemics and mortality in the United States, 1693–1905. Soc. Sci. Med. 1992, 34, 855–865. [Google Scholar] [CrossRef]
- Bryan, C.S.; Moss, S.W.; Kahn, R.J. Yellow fever in the Americas. Infect. Dis. Clin. North Am. 2004, 18, 275–292. [Google Scholar] [CrossRef]
- Theiler, M.; Smith, H.H. The use of yellow fever virus modified by in vitro cultivation for human immunization. J. Exp. Med. 1937, 65, 787–800. [Google Scholar] [CrossRef] [Green Version]
- Gubler, D.J. The changing epidemiology of yellow fever and dengue, 1900 to 2003: Full circle? Comp. Immunol. Microbiol. Infect. Dis. 2004, 27, 319–330. [Google Scholar] [CrossRef]
- Barrett, A.; Higgs, S. Yellow fever: A disease that has yet to be conquered. Annu. Rev. Entomol. 2007, 52, 209–229. [Google Scholar] [CrossRef] [Green Version]
- Jentes, E.S.; Poumerol, G.; Gershman, M.D.; Hill, D.R.; le Marchand, J.; Lewis, R.F.; Staples, J.E.; Tomori, O.; Wilder-Smith, A.; Monath, T.P. The revised global yellow fever risk map and recommendations for vaccination, 2010: Consensus of the informal WHO working group on geographic risk for yellow fever. Lancet Infect. Dis. 2011, 11, 622–632. [Google Scholar] [CrossRef]
- Hamlet, A.; Jean, K.K.; Perea, W.; Yactayo, S.; Biey, J.; van Kerkhove, M.; Ferguson, N.; Garske, T. The seasonal influence of climate and environment on yellow fever transmission across Africa. PLoS Negl. Trop. Dis. 2018, 12, e0006284. [Google Scholar] [CrossRef] [Green Version]
- Garske, T.; van Kerkhove, M.D.; Yactayo, S.; Ronveaux, O.; Lewis, R.F.; Staples, J.E.; Perea, W.; Ferguson, N.M. For the yellow fever expert committee yellow fever in Africa: Estimating the burden of disease and impact of mass vaccination from outbreak and serological data. PLoS Med. 2014, 11, e1001638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tuboi, S.H.; Costa, Z.G.; da Costa Vasconcelos, P.F.; Hatch, D. Clinical and epidemiological characteristics of yellow fever in Brazil: Analysis of reported cases 1998–2002. Trans. R. Soc. Trop. Med. Hyg. 2007, 101, 169–175. [Google Scholar] [CrossRef] [PubMed]
- de Rezende, I.M.; Sacchetto, L.; de Mello Érica, M.; Alves, P.A.; Iani, F.C.D.M.; Adelino, T.É.R.; Duarte, M.M.; Cury, A.L.F.; Bernardes, A.F.L.; Santos, T.A.; et al. Persistence of Yellow fever virus outside the Amazon Basin, causing epidemics in Southeast Brazil, from 2016 to 2018. PLoS Negl. Trop. Dis. 2018, 12, e0006538. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Paploski, I.A.D.; Souza, R.L.; Tauro, L.B.; Cardoso, C.W.; Mugabe, V.A.; Alves, A.B.P.S.; Gomes, J.D.J.; Kikuti, M.; Campos, G.S.; Sardi, S.; et al. Epizootic outbreak of yellow fever virus and risk for human disease in Salvador, Brazil. Ann. Intern. Med. 2017, 168, 301–302. [Google Scholar] [CrossRef] [PubMed]
- Faria, N.R.; Kraemer, M.U.G.; Hill, S.C.; de Jesus, J.G.; Aguiar, R.S.; Iani, F.C.M.; Xavier, J.; Quick, J.; Du Plessis, L.; Dellicour, S.; et al. Genomic and epidemiological monitoring of yellow fever virus transmission potential. Science 2018, 361, 894–899. [Google Scholar] [CrossRef] [Green Version]
- Chen, L.H.; Wilson, M.E. Yellow fever control: Current epidemiology and vaccination strategies. Trop. Dis. Travel Med. Vaccines 2020, 6, 1–10. [Google Scholar] [CrossRef]
- Dick, G.W.A.; Kitchen, S.F.; Haddow, A.J. Zika Virus (I). Isolations and serological specificity. Trans. R. Soc. Trop. Med. Hyg. 1952, 46, 509–520. [Google Scholar] [CrossRef]
- Duffy, M.R.; Chen, T.H.; Hancock, W.T.; Powers, A.M.; Kool, J.L.; Lanciotti, R.S.; Pretrick, M.; Marfel, M.; Holzbauer, S.; Dubray, C.; et al. Zika virus outbreak on Yap Island, Federated States of Micronesia. N. Engl. J. Med. 2009, 360, 2536–2543. [Google Scholar] [CrossRef]
- Faye, O.; Freire, C.C.M.; Iamarino, A.; Faye, O.; de Oliveira, J.V.C.; Diallo, M.; Zanotto, P.M.A.; A Sall, A. molecular evolution of Zika virus during its emergence in the 20th century. PLoS Negl. Trop. Dis. 2014, 8, e2636. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Musso, D.; Bossin, H.; Mallet, H.P.; Besnard, M.; Broult, J.; Baudouin, L.; Levi, J.E.; Sabino, E.C.; Ghawche, F.; Lanteri, M.C.; et al. Zika virus in French Polynesia 2013–14: Anatomy of a completed outbreak. Lancet Infect. Dis. 2018, 18, e172–e182. [Google Scholar] [CrossRef]
- Faria, N.R.; Azevedo, R.D.S.D.S.; Kraemer, M.U.G.; Souza, R.; Cunha, M.S.; Hill, S.C.; Thézé, J.; Bonsall, M.B.; Bowden, T.A.; Rissanen, I.; et al. Zika virus in the Americas: Early epidemiological and genetic findings. Science 2016, 352, 345–349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- dos Santos, T.; Rodriguez, A.; Almiron, M.; Sanhueza, A.; Ramon, P.; de Oliveira, W.K.; Coelho, G.E.; Badaro, R.; Cortez, J.; Ospina, M.; et al. Zika virus and the Guillain-Barre syndrome—Case series from seven countries. N. Engl. J. Med. 2016, 375, 1598–1601. [Google Scholar] [CrossRef]
- Mécharles, S.; Herrmann, C.; Poullain, P.; Tran, T.-H.; Deschamps, N.; Mathon, G.; Landais, A.; Breurec, S.; Lannuzel, A. Acute myelitis due to Zika virus infection. Lancet 2016, 387, 1481. [Google Scholar] [CrossRef] [Green Version]
- Carteaux, G.; Maquart, M.; Bedet, A.; Contou, D.; Brugières, P.; Fourati, S.; de Langavant, L.C.; de Broucker, T.; Brun-Buisson, C.; Leparc-Goffart, I.; et al. Zika Virus Associated with Meningoencephalitis. N. Engl. J. Med. 2016, 374, 1595–1596. [Google Scholar] [CrossRef]
- Sharp, T.M.; Muñoz-Jordán, J.; Perez-Padilla, J.; Bello-Pagán, M.I.; Rivera, A.; Pastula, D.M.; Salinas, J.L.; Mendez, J.H.M.; Méndez, M.; Powers, A.M.; et al. Zika virus infection associated with severe thrombocytopenia. Clin. Infect. Dis. 2016, 63, 1198–1201. [Google Scholar] [CrossRef] [Green Version]
- Foy, B.D.; Kobylinski, K.C.; Chilson-Foy, J.L.; Blitvich, B.J.; Travassos da Rosa, A.; Haddow, A.D.; Lanciotti, R.S.; Tesh, R.B. Probable non-vector-borne transmission of Zika virus, Colorado, USA. Emerg. Infect. Dis. 2011, 17, 880–882. [Google Scholar] [CrossRef]
- Musso, D.; Roche, C.; Robin, E.; Nhan, T.; Teissier, A.; Cao-Lormeau, V.-M. Potential sexual transmission of Zika virus. Emerg. Infect. Dis. 2015, 21, 359–361. [Google Scholar] [CrossRef]
- D’Ortenzio, E.; Matheron, S.; de Lamballerie, X.; Hubert, B.; Piorkowski, G.; Maquart, M.; Descamps, D.; Damond, F.; Yazdanpanah, Y.; Leparc-Goffart, I. Evidence of sexual transmission of Zika virus. N. Engl. J. Med. 2016, 374, 2195–2198. [Google Scholar] [CrossRef]
- Mansuy, J.-M.; Dutertre, M.; Mengelle, C.; Fourcade, C.; Marchou, B.; Delobel, P.; Izopet, J.; Martin-Blondel, G. Zika virus: High infectious viral load in semen, a new sexually transmitted pathogen? Lancet Infect. Dis. 2016, 16, 405. [Google Scholar] [CrossRef] [Green Version]
- Turmel, J.M.; Abgueguen, P.; Hubert, B.; Vandamme, Y.M.; Maquart, M.; le Guillou-Guillemette, H.; Leparc-Goffart, I. Late sexual transmission of Zika virus related to persistence in the semen. Lancet 2016, 387, 2501. [Google Scholar] [CrossRef] [Green Version]
- Mlakar, J.; Korva, M.; Tul, N.; Popovic, M.; Poljsak-Prijatelj, M.; Mraz, J.; Kolenc, M.; Resman Rus, K.; Vesnaver Vipotnik, T.; Fabjan Vodusek, V.; et al. Zika virus associated with microcephaly. N. Engl. J. Med. 2016, 374, 951–958. [Google Scholar] [CrossRef] [PubMed]
- Brasil, P.; Pereira, J.P., Jr.; Moreira, M.E.; Ribeiro-Nogueira, R.M.; Damasceno, L.; Wakimoto, M.; Rabello, R.S.; Valderramos, S.G.; Halai, U.A.; Salles, T.S.; et al. Zika virus infection in pregnant women in Rio de Janeiro. N. Engl. J. Med. 2016, 375, 2321–2334. [Google Scholar] [CrossRef]
- Sarno, M.; Sacramento, G.A.; Khouri, R.; Rosário, M.S.D.; Costa, F.; Archanjo, G.; Santos, L.A.; Nery, N.; Vasilakis, N.; Ko, A.I.; et al. Zika virus infection and stillbirths: A case of hydrops fetalis, hydranencephaly and fetal demise. PLoS Negl. Trop. Dis. 2016, 10, e0004517. [Google Scholar] [CrossRef] [Green Version]
- Besnard, M.; Eyrolle-Guignot, D.; Guillemette-Artur, P.; Lastère, S.; Bost-Bezeaud, F.; Marcelis, L.; Abadie, V.; Garel, C.; Moutard, M.-L.; Jouannic, J.-M.; et al. Congenital cerebral malformations and dysfunction in fetuses and newborns following the 2013 to 2014 Zika virus epidemic in French Polynesia. Eurosurveillance 2016, 21, 21. [Google Scholar] [CrossRef]
- de Araújo, T.V.B.; Rodrigues, L.C.; Ximenes, R.A.; Miranda-Filho, D.D.B.; Montarroyos, U.R.; de Melo, A.P.L.; Valongueiro, S.; Albuquerque, M.F.P.M.; Souza, W.V.; Braga, C.; et al. Association between Zika virus infection and microcephaly in Brazil, January to May 2016: Preliminary report of a case-control study. Lancet Infect. Dis. 2016, 16, 1356–1363. [Google Scholar] [CrossRef] [Green Version]
- Paploski, I.A.; Prates, A.P.P.; Cardoso, C.W.; Kikuti, M.; Silva, M.M.O.; Waller, L.A.; Reis, M.G.; Kitron, U.; Ribeiro, G.S. Time lags between exanthematous illness attributed to Zika virus, Guillain-Barré syndrome, and microcephaly, Salvador, Brazil. Emerg. Infect. Dis. 2016, 22, 1438–1444. [Google Scholar] [CrossRef] [Green Version]
- Moore, C.A.; Staples, J.E.; Dobyns, W.B.; Pessoa, A.; Ventura, C.V.; da Fonseca, E.B.; Ribeiro, E.M.; Ventura, L.O.; Neto, N.N.; Arena, J.F.; et al. Characterizing the Pattern of Anomalies in Congenital Zika Syndrome for Pediatric Clinicians. JAMA Pediatr. 2017, 171, 288–295. [Google Scholar] [CrossRef] [Green Version]
- Miner, J.J.; Cao, B.; Govero, J.; Smith, A.M.; Fernandez, E.; Cabrera, O.H.; Garber, C.; Noll, M.; Klein, R.S.; Noguchi, K.K.; et al. Zika virus infection during pregnancy in mice causes placental damage and fetal demise. Cell 2016, 165, 1081–1091. [Google Scholar] [CrossRef] [Green Version]
- Waldorf, K.M.A.; Stencel-Baerenwald, J.E.; Kapur, R.P.; Studholme, C.; Boldenow, E.; Vornhagen, J.; Baldessari, A.; Dighe, M.K.; Thiel, J.; Merillat, S.; et al. Fetal brain lesions after subcutaneous inoculation of Zika virus in a pregnant nonhuman primate. Nat. Med. 2016, 22, 1256–1259. [Google Scholar] [CrossRef] [PubMed]
- Cugola, F.R.; Fernandes, I.R.; Russo, F.B.; Freitas, B.C.; Dias, J.L.M.; Guimarães, K.P.; Benazzato, C.; Almeida, N.; Pignatari, F.B.R.G.C.; Romero, S.; et al. The Brazilian Zika virus strain causes birth defects in experimental models. Nature 2016, 534, 267–271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Netto, E.M.; Moreira-Soto, A.; Pedroso, C.; Höser, C.; Funk, S.; Kucharski, A.J.; Rockstroh, A.; Kümmerer, B.M.; Sampaio, G.S.; Luz, E.; et al. High Zika Virus seroprevalence in Salvador, Northeastern Brazil limits the potential for further outbreaks. mBio 2017, 8, e01390-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Grubaugh, N.D.; Ladner, J.T.; Kraemer, M.U.G.; Dudas, G.; Tan, A.L.; Gangavarapu, K.; Wiley, M.R.; White, S.; Thézé, J.; Magnani, D.M.; et al. Genomic epidemiology reveals multiple introductions of Zika virus into the United States. Nature 2017, 546, 401–405. [Google Scholar] [CrossRef]
- Ali, S.; Gugliemini, O.; Harber, S.; Harrison, A.; Houle, L.; Ivory, J.; Kersten, S.; Khan, R.; Kim, J.; LeBoa, C.; et al. Environmental and social change drive the explosive emergence of Zika virus in the Americas. PLoS Negl. Trop. Dis. 2017, 11, e0005135. [Google Scholar] [CrossRef]
- Smithburn, K.C.; Hughes, T.P.; Burke, A.W.; Paul, J.H. A neurotropic virus isolated from the blood of a native of Uganda 1. Am. J. Trop. Med. Hyg. 1940, 20, 471–492. [Google Scholar] [CrossRef]
- Hubálek, Z.; Halouzka, J. West Nile fever–a reemerging mosquito-borne viral disease in Europe. Emerg. Infect. Dis. 1999, 5, 643–650. [Google Scholar] [CrossRef]
- Tsai, T.F.; Popovici, F.; Cernescu, C.; Campbell, G.L.; Nedelcu, N.I. West Nile encephalitis epidemic in southeastern Romania. Lancet 1998, 352, 767–771. [Google Scholar] [CrossRef]
- Centers for Disease Control and Prevention (CDC). Outbreak of West nile-like viral encephalitis—New York, 1999. MMWR Morb. Mortal. Wkly. Rep. 1999, 48, 845–849. [Google Scholar]
- Briese, T.; Jia, X.-Y.; Huang, C.; Grady, L.J.; Lipkin, W.L. Identification of a Kunjin/West Nile-like flavivirus in brains of patients with New York encephalitis. Lancet 1999, 354, 1261–1262. [Google Scholar] [CrossRef]
- Lanciotti, R.S. Origin of the West Nile virus responsible for an outbreak of encephalitis in the Northeastern United States. Science 1999, 286, 2333–2337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jia, X.-Y.; Briese, T.; Jordan, I.; Rambaut, A.; Chi, H.C.; MacKenzie, J.S.; Hall, R.A.; Scherret, J.; Lipkin, W.I. Genetic analysis of West Nile New York 1999 encephalitis virus. Lancet 1999, 354, 1971–1972. [Google Scholar] [CrossRef]
- Giladi, M.; Metzkor-Cotter, E.; Martin, D.A.; Siegman-Igra, Y.; Korczyn, A.D.; Rosso, R.; Berger, S.A.; Campbell, G.L.; Lanciotti, R.S. West Nile encephalitis in Israel, 1999: The New York connection. Emerg. Infect. Dis. 2001, 7, 659–661. [Google Scholar] [CrossRef] [PubMed]
- Davis, C.T.; Ebel, G.D.; Lanciotti, R.S.; Brault, A.C.; Guzman, H.; Siirin, M.; Lambert, A.; Parsons, R.E.; Beasley, D.W.; Novak, R.J.; et al. Phylogenetic analysis of North American West Nile virus isolates, 2001–2004: Evidence for the emergence of a dominant genotype. Virology 2005, 342, 252–265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Petersen, L.R.; Carson, P.J.; Biggerstaff, B.J.; Custer, B.; Borchardt, S.M.; Busch, M.P. Estimated cumulative incidence of West Nile virus infection in US adults, 1999–2010. Epidemiol. Infect. 2013, 141, 591–595. [Google Scholar] [CrossRef] [PubMed]
- Habarugira, G.; Suen, W.W.; Hobson-Peters, J.; Hall, R.A.; Bielefeldt-Ohmann, H. West Nile virus: An update on pathobiology, epidemiology, diagnostics, control and “one health” implications. Pathogens 2020, 9, 589. [Google Scholar] [CrossRef] [PubMed]
- Bakonyi, T.; Ivanics, É.; Erdélyi, K.; Ursu, K.; Ferenczi, E.; Weissenböck, H.; Nowotny, N. Lineage 1 and 2 strains of encephalitic West Nile virus, Central Europe. Emerg. Infect. Dis. 2006, 12, 618–623. [Google Scholar] [CrossRef]
- Veo, C.; Della-Ventura, C.; Moreno, A.; Rovida, F.; Percivalle, E.; Canziani, S.; Torri, D.; Calzolari, M.; Vezzoli, F.; Galli, M.; et al. Evolutionary dynamics of the lineage 2 West Nile virus that caused the largest European epidemic: Italy 2011–2018. Viruses 2019, 11, 814. [Google Scholar] [CrossRef] [Green Version]
- Kramer, L.D.; Styer, L.M.; Ebel, G.D. A global perspective on the epidemiology of West Nile virus. Annu. Rev. Entomol. 2008, 53, 61–81. [Google Scholar] [CrossRef] [Green Version]
- Higgs, S.; Schneider, B.S.; van Landingham, D.L.; Klingler, K.A.; Gould, E.A. Nonviremic transmission of West Nile virus. Proc. Natl. Acad. Sci. USA 2005, 102, 8871–8874. [Google Scholar] [CrossRef] [Green Version]
- Root, J.J.; Hall, J.S.; Nemeth, N.M.; Oesterle, P.T.; Gould, D.H.; Klenk, K.; Clark, L.; McLean, R.G. Experimental infection of fox squirrels (Sciurus niger) with West Nile virus. Am. J. Trop. Med. Hyg. 2006, 75, 697–701. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Webster, L.T. Japanese B encephalitis virus: Its differentiation from St. Louis encephalitis virus and relationship to Louping-Ill virus. Science 2006, 86, 402–403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Solomon, T.; Ni, H.; Beasley, D.W.C.; Ekkelenkamp, M.; Cardosa, M.J.; Barrett, A.D.T. Origin and evolution of Japanese encephalitis virus in Southeast Asia. J. Virol. 2003, 77, 3091–3098. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gao, X.; Liu, H.; Li, M.; Fu, S.; Liang, G. Insights into the evolutionary history of Japanese encephalitis virus (JEV) based on whole-genome sequences comprising the five genotypes. Virol. J. 2015, 12, 43–47. [Google Scholar] [CrossRef] [Green Version]
- Chen, W.-R.; Tesh, R.B.; Rico-Hesse, R. Genetic variation of Japanese encephalitis virus in nature. J. Gen. Virol. 1990, 71, 2915–2922. [Google Scholar] [CrossRef]
- Chen, W.R.; Rico-Hesse, R.; Tesh, R.B. A new genotype of Japanese encephalitis virus from Indonesia. Am. J. Trop. Med. Hyg. 1992, 47, 61–69. [Google Scholar] [CrossRef]
- Uchil, P.D.; Satchidanandam, V. Phylogenetic analysis of Japanese encephalitis virus: Envelope gene based analysis reveals a fifth genotype, geographic clustering, and multiple introductions of the virus into the Indian subcontinent. Am. J. Trop. Med. Hyg. 2001, 65, 242–251. [Google Scholar] [CrossRef]
- Li, M.-H.; Fu, S.-H.; Chen, W.-X.; Wang, H.-Y.; Guo, Y.-H.; Liu, Q.-Y.; Li, Y.-X.; Luo, H.-M.; Da, W.; Ji, D.Z.D.; et al. Genotype V Japanese encephalitis virus is emerging. PLoS Negl. Trop. Dis. 2011, 5, e1231. [Google Scholar] [CrossRef]
- Mohammed, M.A.; Galbraith, S.E.; Radford, A.; Dove, W.; Takasaki, T.; Kurane, I.; Solomon, T. Molecular phylogenetic and evolutionary analyses of Muar strain of Japanese encephalitis virus reveal it is the missing fifth genotype. Infect. Genet. Evol. 2011, 11, 855–862. [Google Scholar] [CrossRef]
- MacKenzie, J.S.; Williams, D.T.; Smith, D.W. Japanese encephalitis virus: The geographic distribution, incidence, and spread of a virus with a propensity to emerge in new areas. Perspect. Med. Virol. 2006, 16, 201–268. [Google Scholar] [CrossRef]
- Igarashi, A.; Tanaka, M.; Morita, K.; Takasu, T.; Ahmed, A.; Ahmed, A.; Akram, D.; Waqar, M.A. Detection of West Nile and Japanese Encephalitis Viral Genome Sequences in Cerebrospinal Fluid from Acute Encephalitis Cases in Karachi, Pakistan. Microbiol. Immunol. 1994, 38, 827–830. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hanna, J.N.; A Ritchie, S.; A Phillips, D.; Shield, J.; Bailey, M.C.; MacKenzie, J.S.; Poidinger, M.; McCall, B.J.; Mills, P.J. An outbreak of Japanese encephalitis in the Torres Strait, Australia, 1995. Med J. Aust. 1996, 165, 256–260. [Google Scholar] [CrossRef] [PubMed]
- Hanna, J.N.; Ritchie, S.A.; Hills, S.L.; Hurk, A.F.V.D.; Phillips, D.A.; Pyke, A.T.; Lee, J.M.; Johansen, C.A.; MacKenzie, J.S. Japanese encephalitis in north Queensland, Australia, 1998. Med J. Aust. 1999, 170, 533–536. [Google Scholar] [CrossRef] [PubMed]
- MacKenzie, J.S.; Johansen, C.A.; Ritchie, S.A.; Hurk, A.F.V.D.; Hall, R.A. Japanese encephalitis as an emerging virus: The emergence and spread of Japanese encephalitis virus in Australasia. Curr. Top. Microbiol. Immunol. 2002, 267, 49–73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Simon-Lorière, E.; Faye, O.; Prot, M.; Casademont, I.; Fall, G.; Fernandez-Garcia, M.D.; Diagne, M.M.; Kipela, J.-M.; Fall, I.S.; Holmes, E.C.; et al. Autochthonous Japanese encephalitis with yellow fever coinfection in Africa. N. Engl. J. Med. 2017, 376, 1483–1485. [Google Scholar] [CrossRef]
- Connor, B.; Bunn, W.B. The changing epidemiology of Japanese encephalitis and New data: The implications for New recommendations for Japanese encephalitis vaccine. Trop. Dis. Travel Med. Vaccines 2017, 3, 1–6. [Google Scholar] [CrossRef]
- Huang, Y.J.; Harbin, J.N.; Hettenbach, S.M.; Maki, E.; Cohnstaedt, L.W.; Barrett, A.D.; Higgs, S.; Vanlandingham, D.L. Susceptibility of a North American Culex quinquefasciatus to Japanese encephalitis virus. Vector Borne Zoonotic Dis. 2015, 15, 709–711. [Google Scholar] [CrossRef]
- Hennessy, S.; Strom, B.; Bilker, W.; Zhengle, L.; Chao-Min, W.; Hui-Lian, L.; Tai-Xiang, W.; Hong-Ji, Y.; Qi-Mau, L.; Tsai, T.; et al. Effectiveness of live-attenuated Japanese encephalitis vaccine (SA14-14-2): A case-control study. Lancet 1996, 347, 1583–1586. [Google Scholar] [CrossRef]
- van Gessel, Y.; Klade, C.S.; Putnak, R.; Formica, A.; Krasaesub, S.; Spruth, M.; Cena, B.; Tungtaeng, A.; Gettayacamin, M.; Dewasthaly, S. Correlation of protection against Japanese encephalitis virus and JE vaccine (IXIARO((R))) induced neutralizing antibody titers. Vaccine 2011, 29, 5925–5931. [Google Scholar] [CrossRef]
- Jelinek, T. IXIARO updated: Overview of clinical trials and developments with the inactivated vaccine against Japanese encephalitis. Expert Rev. Vaccines 2013, 12, 859–869. [Google Scholar] [CrossRef]
- Batchelor, P.; Petersen, K. Japanese encephalitis: A review of clinical guidelines and vaccine availability in Asia. Trop. Dis. Travel Med. Vaccines 2015, 1, 11. [Google Scholar] [CrossRef] [PubMed]
- Erlanger, T.E.; Weiss, S.; Keiser, J.; Utzinger, J.; Wiedenmayer, K. Past, present, and future of Japanese encephalitis. Emerg. Infect. Dis. 2009, 15, 1–7. [Google Scholar] [CrossRef] [PubMed]
- Solomon, T.; Dung, N.M.; Kneen, R.; Gainsborough, M.; Vaughn, D.W.; Khanh, V.T. Japanese encephalitis. J. Neurol. Neurosurg. Psychiatry 2000, 68, 405–415. [Google Scholar] [CrossRef] [PubMed]
- Solomon, T.; Kneen, R.; Dung, N.M.; Khanh, V.C.; Thuy, T.T.N.; Ha, D.Q.; Day, N.P.; Nisalak, A.; Vaughn, D.W.; White, N.J. Poliomyelitis-like illness due to Japanese encephalitis virus. Lancet 1998, 351, 1094–1097. [Google Scholar] [CrossRef]
- Centers for Disease Control and Prevention (CDC). Transmission of Japanese Encephalitis Virus. Available online: https://www.cdc.gov/japaneseencephalitis/transmission/index.html (accessed on 25 August 2020).
- Lord, J.S.; Gurley, E.S.; Pulliam, J.R.C. Rethinking Japanese encephalitis virus transmission: A framework for implicating host and vector species. PLoS Negl. Trop. Dis. 2015, 9, e0004074. [Google Scholar] [CrossRef] [Green Version]
- Lai, C.; Lemke, G. An extended family of protein-tyrosine kinase genes differentially expressed in the vertebrate nervous system. Neuron 1991, 6, 691–704. [Google Scholar] [CrossRef]
- O’Bryan, J.P.; Frye, R.A.; Cogswell, P.C.; Neubauer, A.; Kitch, B.; Prokop, C.; Espinosa, R., 3rd; le Beau, M.M.; Earp, H.S.; Liu, E.T. Axl, a transforming gene isolated from primary human myeloid leukemia cells, encodes a novel receptor tyrosine kinase. Mol. Cell. Biol. 1991, 11, 5016–5031. [Google Scholar] [CrossRef] [Green Version]
- Meertens, L.; Carnec, X.; Lecoin, M.P.; Ramdasi, R.; Guivel-Benhassine, F.; Lew, E.; Lemke, G.; Schwartz, O.; Amara, A. The TIM and TAM Families of phosphatidylserine receptors mediate dengue virus entry. Cell Host Microbe 2012, 12, 544–557. [Google Scholar] [CrossRef] [Green Version]
- Miner, J.J.; Daniels, B.P.; Shrestha, B.; Proenca-Modena, J.L.; Lew, E.D.; Lazear, H.M.; Gorman, M.J.; Lemke, G.; Klein, R.S.; Diamond, M.S. The TAM receptor Mertk protects against neuroinvasive viral infection by maintaining blood-brain barrier integrity. Nat. Med. 2015, 21, 1464–1472. [Google Scholar] [CrossRef] [Green Version]
- Hamel, R.; Dejarnac, O.; Wichit, S.; Ekchariyawat, P.; Neyret, A.; Luplertlop, N.; Perera-Lecoin, M.; Surasombatpattana, P.; Talignani, L.; Thomas, F.; et al. Biology of Zika Virus Infection in Human Skin Cells. J. Virol. 2015, 89, 8880–8896. [Google Scholar] [CrossRef] [Green Version]
- Liu, S.; de Lalio, L.J.; Isakson, B.E.; Wang, T.T. AXL-mediated productive infection of human endothelial cells by Zika virus. Circul. Res. 2016, 119, 1183–1189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Richard, A.S.; Shim, B.-S.; Kwon, Y.-C.; Zhang, R.; Otsuka, Y.; Schmitt, K.; Berri, F.; Diamond, M.S.; Choe, H. AXL-dependent infection of human fetal endothelial cells distinguishes Zika virus from other pathogenic flaviviruses. Proc. Natl. Acad. Sci. USA 2017, 114, 2024–2029. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tabata, T.; Petitt, M.; Puerta-Guardo, H.; Michlmayr, D.; Wang, C.; Fang-Hoover, J.; Harris, E.; Pereira, L. Zika virus targets different primary human placental cells, suggesting two routes for vertical transmission. Cell Host Microbe 2016, 20, 155–166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tang, H.; Hammack, C.; Ogden, S.C.; Wen, Z.; Qian, X.; Li, Y.; Yao, B.; Shin, J.; Zhang, F.; Lee, E.M.; et al. Zika virus infects human cortical neural progenitors and attenuates their growth. Cell Stem Cell 2016, 18, 587–590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nowakowski, T.J.; Pollen, A.A.; di Lullo, E.; Sandoval-Espinosa, C.; Bershteyn, M.; Kriegstein, A.R. Expression analysis highlights axl as a candidate Zika virus entry receptor in neural stem cells. Cell Stem Cell 2016, 18, 591–596. [Google Scholar] [CrossRef] [Green Version]
- Retallack, H.; di Lullo, E.; Arias, C.; Knopp, K.A.; Laurie, M.T.; Sandoval-Espinosa, C.; Mancia-Leon, W.R.; Krencik, R.; Ullian, E.M.; Spatazza, J.; et al. Zika virus cell tropism in the developing human brain and inhibition by azithromycin. Proc. Natl. Acad. Sci. USA 2016, 113, 14408–14413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wells, M.F.; Salick, M.R.; Wiskow, O.; Ho, D.J.; Worringer, K.A.; Ihry, R.J.; Kommineni, S.; Bilican, B.; Klim, J.R.; Hill, E.J.; et al. Genetic ablation of AXL does not protect human neural progenitor cells and cerebral organoids from Zika virus infection. Cell Stem Cell 2016, 19, 703–708. [Google Scholar] [CrossRef] [Green Version]
- Hastings, A.K.; Yockey, L.J.; Jagger, B.W.; Hwang, J.; Uraki, R.; Gaitsch, H.F.; Parnell, L.A.; Cao, B.; Mysorekar, I.U.; Rothlin, C.V.; et al. TAM receptors are not required for Zika virus infection in mice. Cell Rep. 2017, 19, 558–568. [Google Scholar] [CrossRef] [Green Version]
- Li, F.; Wang, P.R.; Qu, L.B.; Yi, C.H.; Zhang, F.C.; Tang, X.P.; Zhang, L.G.; Chen, L. AXL is not essential for Zika virus infection in the mouse brain. Emerg. Microbes Infect. 2017, 6, 1–2. [Google Scholar] [CrossRef] [Green Version]
- Wang, Z.-Y.; Wang, Z.; Zhen, Z.-D.; Feng, K.-H.; Guo, J.; Gao, N.; Fan, D.-Y.; Han, D.-S.; Wang, P.; An, J. Axl is not an indispensable factor for Zika virus infection in mice. J. Gen. Virol. 2017, 98, 2061–2068. [Google Scholar] [CrossRef]
- Rausch, K.; Hackett, B.A.; Weinbren, N.L.; Reeder, S.M.; Sadovsky, Y.; Hunter, C.A.; Schultz, D.C.; Coyne, C.B.; Cherry, S. Screening bioactives reveals nanchangmycin as a broad spectrum antiviral active against Zika virus. Cell Rep. 2017, 18, 804–815. [Google Scholar] [CrossRef] [PubMed]
- Meertens, L.; la Beau, A.; Dejarnac, O.; Cipriani, S.; Sinigaglia, L.; Bonnet-Madin, L.; Le Charpentier, T.; Hafirassou, M.L.; Zamborlini, A.; Cao-Lormeau, V.-M.; et al. Axl mediates ZIKA virus entry in human glial cells and modulates innate immune responses. Cell Rep. 2017, 18, 324–333. [Google Scholar] [CrossRef] [PubMed]
- Chen, J.; Yang, Y.-F.; Yang, Y.; Zou, P.; Chen, J.; He, Y.; Shui, S.-L.; Cui, Y.-R.; Bai, R.; Liang, Y.-J.; et al. AXL promotes Zika virus infection in astrocytes by antagonizing type I interferon signalling. Nat. Microbiol. 2018, 3, 302–309. [Google Scholar] [CrossRef] [PubMed]
- Sun, X.; Hua, S.; Chen, H.-R.; Ouyang, Z.; Einkauf, K.; Tse, S.; Ard, K.; Ciaranello, A.; Yawetz, S.; Sax, P.; et al. Transcriptional changes during naturally acquired Zika virus infection render dendritic cells highly conducive to viral replication. Cell Rep. 2017, 21, 3471–3482. [Google Scholar] [CrossRef] [Green Version]
- Bhattacharyya, S.; Zagórska, A.; Lew, E.D.; Shrestha, B.; Rothlin, C.V.; Naughton, J.; Diamond, M.S.; Lemke, G.; Young, J.A. Enveloped viruses disable innate immune responses in dendritic cells by direct activation of TAM receptors. Cell Host Microbe 2013, 14, 136–147. [Google Scholar] [CrossRef] [Green Version]
- Fan, W.; Qian, P.; Wang, D.; Zhi, X.; Wei, Y.; Chen, H.; Li, X. Integrin alphavbeta3 promotes infection by Japanese encephalitis virus. Res. Vet. Sci. 2017, 111, 67–74. [Google Scholar] [CrossRef]
- Chu, J.J.; Ng, M.L. Interaction of West Nile virus with alpha v beta 3 integrin mediates virus entry into cells. J. Biol. Chem. 2004, 279, 54533–54541. [Google Scholar] [CrossRef] [Green Version]
- Bogachek, M.V.; Zaitsev, B.N.; Sekatskii, S.K.; Protopopova, E.V.; Ternovoi, V.A.; Ivanova, A.V.; Kachko, A.V.; Ivanisenko, V.A.; Dietler, G.; Loktev, V.B. Characterization of glycoprotein E C-end of West Nile virus and evaluation of its interaction force with alphaVbeta3 integrin as putative cellular receptor. Biochemistry 2010, 75, 472–480. [Google Scholar] [CrossRef]
- Wang, S.; Zhang, Q.; Tiwari, S.K.; Lichinchi, G.; Yau, E.H.; Hui, H.; Li, W.; Furnari, F.; Rana, T.M. Integrin alphavbeta5 internalizes Zika virus during neural stem cells infection and provides a promising target for antiviral therapy. Cell Rep. 2020, 30, 969–983. [Google Scholar] [CrossRef] [Green Version]
- Dejarnac, O.; Hafirassou, M.L.; Chazal, M.; Versapuech, M.; Burlaud-Gaillard, J.; Perera-Lecoin, M.; Umaña-Diaz, C.; Bonnet-Madin, L.; Carnec, X.; Tinevez, J.-Y.; et al. TIM-1 ubiquitination mediates dengue virus entry. Cell Rep. 2018, 23, 1779–1793. [Google Scholar] [CrossRef]
- Savidis, G.; McDougall, W.M.; Meraner, P.; Perreira, J.M.; Portmann, J.M.; Trincucci, G.; John, S.P.; Aker, A.M.; Renzette, N.; Robbins, D.R.; et al. Identification of Zika virus and Dengue virus dependency factors using functional genomics. Cell Rep. 2016, 16, 232–246. [Google Scholar] [CrossRef] [Green Version]
- Zhang, R.; Miner, J.J.; Gorman, M.J.; Rausch, K.; Ramage, H.; White, J.P.; Zuiani, A.; Zhang, P.; Fernandez, E.; Zhang, Q.; et al. A CRISPR screen defines a signal peptide processing pathway required by flaviviruses. Nature 2016, 535, 164–168. [Google Scholar] [CrossRef] [Green Version]
- Marceau, C.D.; Puschnik, A.S.; Majzoub, K.; Ooi, Y.S.; Brewer, S.M.; Fuchs, G.; Swaminathan, K.; Mata, M.A.; Elias, J.E.; Sarnow, P.; et al. Genetic dissection of Flaviviridae host factors through genome-scale CRISPR screens. Nature 2016, 535, 159–163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, Y.; Muffat, J.; Javed, A.O.; Keys, H.R.; Lungjangwa, T.; Bosch, I.; Khan, M.; Virgilio, M.C.; Gehrke, L.; Sabatini, D.M.; et al. Genome-wide CRISPR screen for Zika virus resistance in human neural cells. Proc. Natl. Acad. Sci. USA 2019, 116, 9527–9532. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- la Beau, A.; Simon-Loriere, E.; Hafirassou, M.-L.; Bonnet-Madin, L.; Tessier, S.; Zamborlini, A.; Dupré, T.; Seta, N.; Schwartz, O.; Chaix, M.-L.; et al. A genome-wide CRISPR-Cas9 screen identifies the dolichol-phosphate mannose synthase complex as a host dependency factor for dengue virus infection. J. Virol. 2020, 94, 01751-19. [Google Scholar] [CrossRef]
- Kaksonen, M.; Roux, A. Mechanisms of clathrin-mediated endocytosis. Nat. Rev. Mol. Cell Biol. 2018, 19, 313–326. [Google Scholar] [CrossRef]
- Kirchhausen, T.; Owen, D.; Harrison, S.C. Molecular structure, function, and dynamics of clathrin-mediated membrane traffic. Cold Spring Harb. Perspect. Biol. 2014, 6, a016725. [Google Scholar] [CrossRef] [Green Version]
- Elkin, S.R.; Lakoduk, A.M.; Schmid, S.L. Endocytic pathways and endosomal trafficking: A primer. Wien. Med. Wochenschr. 2016, 166, 196–204. [Google Scholar] [CrossRef] [Green Version]
- Mettlen, M.; Chen, P.-H.; Srinivasan, S.; Danuser, G.; Schmid, S.L. Regulation of clathrin-mediated endocytosis. Annu. Rev. Biochem. 2018, 87, 871–896. [Google Scholar] [CrossRef]
- Hackett, B.A.; Yasunaga, A.; Panda, D.; Tartell, M.A.; Hopkins, K.C.; Hensley, S.E.; Cherry, S. RNASEK is required for internalization of diverse acid-dependent viruses. Proc. Natl. Acad. Sci. USA 2015, 112, 7797–7802. [Google Scholar] [CrossRef] [Green Version]
- Hackett, B.A.; Cherry, S. Flavivirus internalization is regulated by a size-dependent endocytic pathway. Proc. Natl. Acad. Sci. USA 2018, 115, 4246–4251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gollins, S.W.; Porterfield, J.S. Flavivirus infection enhancement in macrophages: An electron microscopic study of viral cellular entry. J. Gen. Virol. 1985, 66, 1969–1982. [Google Scholar] [CrossRef] [PubMed]
- Mizutani, T.; Kobayashi, M.; Eshita, Y.; Shirato, K.; Kimura, T.; Ako, Y.; Miyoshi, H.; Takasaki, T.; Kurane, I.; Kariwa, H.; et al. Involvement of the JNK-like protein of the Aedes albopictus mosquito cell line, C6/36, in phagocytosis, endocytosis and infection of West Nile virus. Insect Mol. Biol. 2003, 12, 491–499. [Google Scholar] [CrossRef] [PubMed]
- Chu, J.J.; Leong, P.W.; Ng, M.L. Analysis of the endocytic pathway mediating the infectious entry of mosquito-borne flavivirus West Nile into Aedes albopictus mosquito (C6/36) cells. Virology 2006, 349, 463–475. [Google Scholar] [CrossRef] [Green Version]
- Liou, M.-L.; Hsu, C.Y. Japanese encephalitis virus is transported across the cerebral blood vessels by endocytosis in mouse brain. Cell Tissue Res. 1998, 293, 389–394. [Google Scholar] [CrossRef]
- Ishak, R.; Tovey, D.G.; Howard, C.R. Morphogenesis of yellow fever virus 17D in infected cell cultures. J. Gen. Virol. 1988, 69, 325–335. [Google Scholar] [CrossRef]
- Ng, M.L.; Lau, L.C.L. Possible involvement of receptors in the entry of Kunjin virus into Vero cells. Arch. Virol. 1988, 100, 199–211. [Google Scholar] [CrossRef]
- Van der Schaar, H.M.; Rust, M.J.; Chen, C.; van der Ende-Metselaar, H.; Wilschut, J.; Zhuang, X.; Smit, J.M. Dissecting the cell entry pathway of dengue virus by single-particle tracking in living cells. PLoS Pathog. 2008, 4, e1000244. [Google Scholar] [CrossRef] [Green Version]
- Se-Thoe, S.; Ling, A.; Ng, M. Alteration of virus entry mode: A neutralisation mechanism for dengue-2 virus. J. Med Virol. 2000, 62, 364–376. [Google Scholar] [CrossRef]
- Lim, H.-Y.; Ng, M.L. A different mode of entry by dengue-2 neutralisation escape mutant virus. Arch. Virol. 1999, 144, 989–995. [Google Scholar] [CrossRef]
- Mosso, C.; Galvan-Mendoza, I.J.; Ludert, J.E.; del Angel, R.M. Endocytic pathway followed by dengue virus to infect the mosquito cell line C6/36 HT. Virology 2008, 378, 193–199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Acosta, E.G.; Castilla, V.; Damonte, E.B. Functional entry of dengue virus into Aedes albopictus mosquito cells is dependent on clathrin-mediated endocytosis. J. Gen. Virol. 2008, 89, 474–484. [Google Scholar] [CrossRef] [PubMed]
- Suksanpaisan, L.; Susantad, T.; Smith, D.R. Characterization of dengue virus entry into HepG2 cells. J. Biomed. Sci. 2009, 16, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peng, T.; Wang, J.-L.; Chen, W.; Zhang, J.-L.; Gao, N.; Chen, Z.; Xu, X.; Fan, D.-Y.; An, J. Entry of dengue virus serotype 2 into ECV304 cells depends on clathrin-dependent endocytosis, but not on caveolae-dependent endocytosis. Can. J. Microbiol. 2009, 55, 139–145. [Google Scholar] [CrossRef] [PubMed]
- Alhoot, M.A.; Wang, S.M.; Sekaran, S.D. Inhibition of dengue virus entry and multiplication into monocytes using RNA interference. PLoS Negl. Trop. Dis. 2011, 5, e1410. [Google Scholar] [CrossRef] [PubMed]
- Acosta, E.G.; Castilla, V.; Damonte, E.B. Infectious dengue-1 virus entry into mosquito C6/36 cells. Virus Res. 2011, 160, 173–179. [Google Scholar] [CrossRef] [PubMed]
- Alhoot, M.A.; Wang, S.M.; Sekaran, S.D. RNA interference mediated inhibition of dengue virus multiplication and entry in HepG2 cells. PLoS ONE 2012, 7, e34060. [Google Scholar] [CrossRef] [Green Version]
- Fernandez-Garcia, M.D.; Meertens, L.; Chazal, M.; Hafirassou, M.L.; Dejarnac, O.; Zamborlini, A.; Despres, P.; Sauvonnet, N.; Arenzana-Seisdedos, F.; Jouvenet, N.; et al. Vaccine and wild-type strains of yellow fever virus engage distinct entry mechanisms and differentially stimulate antiviral immune responses. mBio 2016, 7, e01956-15. [Google Scholar] [CrossRef] [Green Version]
- Acosta, E.G.; Castilla, V.; Damonte, E.B. Alternative infectious entry pathways for dengue virus serotypes into mammalian cells. Cell. Microbiol. 2009, 11, 1533–1549. [Google Scholar] [CrossRef] [Green Version]
- Piccini, L.E.; Castilla, V.; Damonte, E.B. Dengue-3 virus entry into Vero cells: Role of clathrin-mediated endocytosis in the outcome of infection. PLoS ONE 2015, 10, e0140824. [Google Scholar] [CrossRef] [Green Version]
- Rinkenberger, N.; Schoggins, J.W. Comparative analysis of viral entry for Asian and African lineages of Zika virus. Virology 2019, 533, 59–67. [Google Scholar] [CrossRef] [PubMed]
- Owczarek, K.; Chykunova, Y.; Jassoy, C.; Maksym, B.; Rajfur, Z.; Pyrc, K. Zika virus: Mapping and reprogramming the entry. Cell Commun. Signal. 2019, 17, 41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhu, Y.Z.; Xu, Q.Q.; Wu, D.G.; Ren, H.; Zhao, P.; Lao, W.G.; Wang, Y.; Tao, Q.Y.; Qian, X.J.; Wei, Y.H.; et al. Japanese encephalitis virus enters rat neuroblastoma cells via a pH-dependent, dynamin and caveola-mediated endocytosis pathway. J. Virol. 2012, 86, 13407–13422. [Google Scholar] [CrossRef] [Green Version]
- Kalia, M.; Khasa, R.; Sharma, M.; Nain, M.; Vrati, S. Japanese encephalitis virus infects neuronal cells through a clathrin-independent endocytic mechanism. J. Virol. 2013, 87, 148–162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xu, Q.; Cao, M.; Song, H.; Chen, S.; Qian, X.; Zhao, P.; Ren, H.; Tang, H.; Wang, Y.; Wei, Y.; et al. Caveolin-1-mediated Japanese encephalitis virus entry requires a two-step regulation of actin reorganization. Future Microbiol. 2016, 11, 1227–1248. [Google Scholar] [CrossRef] [PubMed]
- Das, S.; Chakraborty, S.; Basu, A. Critical role of lipid rafts in virus entry and activation of phosphoinositide 3′ kinase/Akt signaling during early stages of Japanese encephalitis virus infection in neural stem/progenitor cells. J. Neurochem. 2010, 115, 537–549. [Google Scholar] [CrossRef]
- Chuang, C.K.; Yang, T.H.; Chen, T.H.; Yang, C.F.; Chen, W.J. Heat shock cognate protein 70 isoform D is required for clathrin-dependent endocytosis of Japanese encephalitis virus in C6/36 cells. J. Gen. Virol. 2015, 96, 793–803. [Google Scholar] [CrossRef] [Green Version]
- Yang, S.; He, M.; Liu, X.; Li, X.; Fan, B.; Zhao, S. Japanese encephalitis virus infects porcine kidney epithelial PK15 cells via clathrin- and cholesterol-dependent endocytosis. Virol. J. 2013, 10, 258. [Google Scholar] [CrossRef] [Green Version]
- Liu, C.-C.; Zhang, Y.-N.; Li, Z.-Y.; Hou, J.-X.; Zhou, J.; Kan, L.; Zhou, B.; Chen, P.-Y. Rab5 and Rab11 are required for clathrin-dependent endocytosis of Japanese encephalitis virus in BHK-21 Cells. J. Virol. 2017, 91, 01113-17. [Google Scholar] [CrossRef] [Green Version]
- Khasa, R.; Vaidya, A.; Vrati, S.; Kalia, M. Membrane trafficking RNA interference screen identifies a crucial role of the clathrin endocytic pathway and ARP2/3 complex for Japanese encephalitis virus infection in HeLa cells. J. Gen. Virol. 2019, 100, 176–186. [Google Scholar] [CrossRef]
- Acosta, E.G.; Piccini, L.E.; Talarico, L.B.; Castilla, V.; Damonte, E.B. Changes in antiviral susceptibility to entry inhibitors and endocytic uptake of dengue-2 virus serially passaged in Vero or C6/36 cells. Virus Res. 2014, 184, 39–43. [Google Scholar] [CrossRef] [PubMed]
- Krishnan, M.N.; Sukumaran, B.; Pal, U.; Agaisse, H.; Murray, J.L.; Hodge, T.W.; Fikrig, E. Rab 5 is required for the cellular entry of dengue and West Nile viruses. J. Virol. 2007, 81, 4881–4885. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Krishnan, M.N.; Ng, A.; Sukumaran, B.; Gilfoy, F.D.; Uchil, P.D.; Sultana, H.; Brass, A.L.; Adametz, R.; Tsui, M.; Qian, F.; et al. RNA interference screen for human genes associated with West Nile virus infection. Nature 2008, 455, 242–245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schoggins, J.W.; Wilson, S.J.; Panis, M.; Murphy, M.Y.; Jones, C.T.; Bieniasz, P.; Rice, C.M. A diverse range of gene products are effectors of the type I interferon antiviral response. Nature 2011, 472, 481–485. [Google Scholar] [CrossRef]
- Schoggins, J.W.; Dorner, M.; Feulner, M.; Imanaka, N.; Murphy, M.Y.; Ploss, A.; Rice, C.M. Dengue reporter viruses reveal viral dynamics in interferon receptor-deficient mice and sensitivity to interferon effectors in vitro. Proc. Natl. Acad. Sci. USA 2012, 109, 14610–14615. [Google Scholar] [CrossRef] [Green Version]
- Yu, J.; Liu, S.-L. Emerging role of LY6E in virus–host interactions. Viruses 2019, 11, 1020. [Google Scholar] [CrossRef] [Green Version]
- Noda, S.; Kosugi, A.; Saitoh, S.; Narumiya, S.; Hamaoka, T. Protection from anti-TCR/CD3-induced apoptosis in immature thymocytes by a signal through thymic shared antigen-1/stem cell antigen-2. J. Exp. Med. 1996, 183, 2355–2360. [Google Scholar] [CrossRef] [Green Version]
- Kosugi, A.; Saitoh, S.-I.; Narumiya, S.; Miyake, K.; Hamaoka, T. Activation-induced expression of thymic shared antigen-1 on T lymphocytes and its inhibitory role for TCR-mediated IL-2 production. Int. Immunol. 1994, 6, 1967–1976. [Google Scholar] [CrossRef]
- Saitoh, S.; Kosugi, A.; Noda, S.; Yamamoto, N.; Ogata, M.; Minami, Y.; Miyake, K.; Hamaoka, T. Modulation of TCR-mediated signaling pathway by thymic shared antigen-1 (TSA-1)/stem cell antigen-2 (Sca-2). J. Immunol. 1995, 155, 5574–5581. [Google Scholar]
- Mar, K.B.; Rinkenberger, N.R.; Boys, I.N.; Eitson, J.L.; McDougal, M.B.; Richardson, R.B.; Schoggins, J.W. LY6E mediates an evolutionarily conserved enhancement of virus infection by targeting a late entry step. Nat. Commun. 2018, 9, 1–14. [Google Scholar] [CrossRef]
- Ramanathan, H.N.; Zhang, S.; Douam, F.; Mar, K.B.; Chang, J.; Yang, P.L.; Schoggins, J.W.; Ploss, A.; Lindenbach, B.D. A sensitive yellow fever virus entry reporter identifies valosin-containing protein (VCP/p97) as an essential host factor for flavivirus uncoating. mBio 2020, 11, 00467-20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sharonov, G.; Balatskaya, M.N.; Tkachuk, V.A. Glycosylphosphatidylinositol-anchored proteins as regulators of cortical cytoskeleton. Biochemistry 2016, 81, 636–650. [Google Scholar] [CrossRef] [PubMed]
- Saha, S.; Anilkumar, A.A.; Mayor, S. GPI-anchored protein organization and dynamics at the cell surface. J. Lipid Res. 2016, 57, 159–175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bacquin, A.; Bireau, C.; Tanguy, M.; Romanet, C.; Vernochet, C.; Dupressoir, A.; Heidmann, T. A cell fusion-based screening method identifies glycosylphosphatidylinositol-anchored protein Ly6e as the receptor for mouse endogenous retroviral envelope Syncytin-A. J. Virol. 2017, 91, 00832-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Langford, M.B.; Outhwaite, J.E.; Hughes, M.; Natale, D.R.C.; Simmons, D. Deletion of the Syncytin A receptor Ly6e impairs syncytiotrophoblast fusion and placental morphogenesis causing embryonic lethality in mice. Sci. Rep. 2018, 8, 1–11. [Google Scholar] [CrossRef] [PubMed]
- Perreira, J.M.; Aker, A.M.; Savidis, G.; Chin, C.R.; McDougall, W.M.; Portmann, J.M.; Meraner, P.; Smith, M.C.; Rahman, M.; Baker, R.E.; et al. RNASEK Is a V-ATPase-associated factor required for endocytosis and the replication of rhinovirus, influenza A virus, and dengue virus. Cell Rep. 2015, 12, 850–863. [Google Scholar] [CrossRef] [Green Version]
- Kozik, P.; Hodson, N.A.; Sahlender, D.A.; Simecek, N.; Soromani, C.; Wu, J.; Collinson, L.; Robinson, M.S. A human genome-wide screen for regulators of clathrin-coated vesicle formation reveals an unexpected role for the V-ATPase. Nat. Cell Biol. 2012, 15, 50–60. [Google Scholar] [CrossRef] [Green Version]
- Hopkins, K.C.; McLane, L.M.; Maqbool, T.; Panda, D.; Gordesky-Gold, B.; Cherry, S. A genome-wide RNAi screen reveals that mRNA decapping restricts bunyaviral replication by limiting the pools of Dcp2-accessible targets for cap-snatching. Genes Dev. 2013, 27, 1511–1525. [Google Scholar] [CrossRef] [Green Version]
- Yasunaga, A.; Hanna, S.L.; Li, J.; Cho, H.; Rose, P.P.; Spiridigliozzi, A.; Gold, B.; Diamond, M.S.; Cherry, S. Genome-wide RNAi screen identifies broadly-acting host factors that inhibit arbovirus infection. PLoS Pathog. 2014, 10, e1003914. [Google Scholar] [CrossRef] [Green Version]
- Lafourcade, C.; Sobo, K.; Kieffer-Jaquinod, S.; Garin, J.; van der Goot, F.G. Regulation of the V-ATPase along the endocytic pathway occurs through reversible subunit association and membrane localization. PLoS ONE 2008, 3, e2758. [Google Scholar] [CrossRef] [Green Version]
- Maxson, M.E.; Grinstein, S. The vacuolar-type H+-ATPase at a glance—More than a proton pump. J. Cell Sci. 2014, 127, 4987–4993. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Abbas, Y.M.; Wu, D.; Bueler, S.A.; Robinson, C.V.; Rubinstein, J.L. Structure of V-ATPase from the mammalian brain. Science 2020, 367, 1240–1246. [Google Scholar] [CrossRef] [PubMed]
- Toei, M.; Saum, R.; Forgac, M. Regulation and isoform function of the V-ATPases. Biochemistry 2010, 49, 4715–4723. [Google Scholar] [CrossRef] [Green Version]
- Granger, E.; McNee, G.; Allan, V.J.; Woodman, P. The role of the cytoskeleton and molecular motors in endosomal dynamics. Semin. Cell Dev. Biol. 2014, 31, 20–29. [Google Scholar] [CrossRef] [PubMed]
- le Sommer, C.; Barrows, N.J.; Bradrick, S.S.; Pearson, J.L.; Garcia-Blanco, M.A. G protein-coupled receptor kinase 2 promotes Flaviviridae entry and replication. PLoS Negl. Trop. Dis. 2012, 6, e1820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ban, T.A. Fifty years chlorpromazine: A historical perspective. Neuropsychiatr. Dis. Treat. 2007, 3, 495–500. [Google Scholar] [PubMed]
- Miller, C.L. On the mechanism of action of antipsychotic drugs: A chemical reaction not receptor blockade. Curr. Drug Discov. Technol. 2013, 10, 195–208. [Google Scholar] [CrossRef]
- Wang, L.H.; Rothberg, K.G.; Anderson, R.G. Mis-assembly of clathrin lattices on endosomes reveals a regulatory switch for coated pit formation. J. Cell Biol. 1993, 123, 1107–1117. [Google Scholar] [CrossRef]
- Nawa, M.; Takasaki, T.; Yamada, K.-I.; Kurane, I.; Akatsuka, T. Interference in Japanese encephalitis virus infection of Vero cells by a cationic amphiphilic drug, chlorpromazine. J. Gen. Virol. 2003, 84, 1737–1741. [Google Scholar] [CrossRef]
- Persaud, M.; Martinez-Lopez, A.; Buffone, C.; Porcelli, S.A.; Diaz-Griffero, F. Infection by Zika viruses requires the transmembrane protein AXL, endocytosis and low pH. Virology 2018, 518, 301–312. [Google Scholar] [CrossRef]
- Carro, A.C.; Piccini, L.E.; Damonte, E.B. Blockade of dengue virus entry into myeloid cells by endocytic inhibitors in the presence or absence of antibodies. PLoS Negl. Trop. Dis. 2018, 12, e0006685. [Google Scholar] [CrossRef]
- Ang, F.; Wong, A.P.Y.; Ng, M.M.-L.; Chu, J.J.H. Small interference RNA profiling reveals the essential role of human membrane trafficking genes in mediating the infectious entry of dengue virus. Virol. J. 2010, 7, 24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Candurra, N.A.; Maskin, L.; Damonte, E.B. Inhibition of arenavirus multiplication in vitro by phenotiazines. Antivir. Res. 1996, 31, 149–158. [Google Scholar] [CrossRef]
- de Wilde, A.; Jochmans, D.; Posthuma, C.; Zevenhoven-Dobbe, J.; van Nieuwkoop, S.; Bestebroer, T.; Hoogen, B.V.D.; Neyts, J.; Snijder, E.J. Screening of an FDA-approved compound library identifies four small-molecule inhibitors of Middle East respiratory syndrome coronavirus replication in cell culture. Antimicrob. Agents Chemother. 2014, 58, 4875–4884. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ferraris, O.; Moroso, M.; Pernet, O.; Emonet, S.; Rembert, A.F.; Paranhos-Baccalà, G.; Peyrefitte, C. Evaluation of Crimean-Congo hemorrhagic fever virus in vitro inhibition by chloroquine and chlorpromazine, two FDA approved molecules. Antivir. Res. 2015, 118, 75–81. [Google Scholar] [CrossRef] [PubMed]
- Cong, Y.; Hart, B.J.; Gross, R.; Zhou, H.; Frieman, M.; Bollinger, L.; Wada, J.; Hensley, L.E.; Jahrling, P.B.; Dyall, J.; et al. MERS-CoV pathogenesis and antiviral efficacy of licensed drugs in human monocyte-derived antigen-presenting cells. PLoS ONE 2018, 13, e0194868. [Google Scholar] [CrossRef]
- Plaze, M.; Attali, D.; Petit, A.-C.; Blatzer, M.; Simon-Loriere, E.; Vinckier, F.; Cachia, A.; Chrétien, F.; Gaillard, R. Repurposing chlorpromazine to treat COVID-19: The reCoVery study. L’Encéphale 2020, 46, 169–172. [Google Scholar] [CrossRef]
- Simanjuntak, Y.; Liang, J.-J.; Lee, Y.-L.; Lin, Y.-L. Repurposing of prochlorperazine for use against dengue virus infection. J. Infect. Dis. 2014, 211, 394–404. [Google Scholar] [CrossRef]
- Ho, M.-R.; Tsai, T.-T.; Chen, C.-L.; Jhan, M.-K.; Tsai, C.-C.; Lee, Y.-C.; Chen, C.-H.; Lin, C.-F. Blockade of dengue virus infection and viral cytotoxicity in neuronal cells in vitro and in vivo by targeting endocytic pathways. Sci. Rep. 2017, 7, 6910. [Google Scholar] [CrossRef] [Green Version]
- Simanjuntak, Y.; Liang, J.-J.; Lee, Y.-L.; Lin, Y.-L. Japanese encephalitis virus exploits dopamine D2 receptor-phospholipase c to target dopaminergic human neuronal cells. Front. Microbiol. 2017, 8, 651. [Google Scholar] [CrossRef]
- Smith, J.L.; Stein, D.A.; Shum, D.; Fischer, M.A.; Radu, C.; Bhinder, B.; Djaballah, H.; Nelson, J.A.; Früh, K.; Hirsch, A.J. Inhibition of dengue virus replication by a class of small-molecule compounds that antagonize dopamine receptor D4 and downstream mitogen-activated protein kinase signaling. J. Virol. 2014, 88, 5533–5542. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, T.; Tan, L.; Cederquist, G.Y.; Fan, Y.; Hartley, B.J.; Mukherjee, S.; Tomishima, M.; Brennand, K.J.; Zhang, Q.; Schwartz, R.E.; et al. High-content screening in hPSC-neural progenitors identifies drug candidates that inhibit Zika virus infection in fetal-like organoids and adult brain. Cell Stem Cell 2017, 21, 274–283. [Google Scholar] [CrossRef] [PubMed]
- Han, Y.; Mesplède, T.; Xu, H.; Quan, Y.; Wainberg, M.A. The antimalarial drug amodiaquine possesses anti-ZIKA virus activities. J. Med. Virol. 2018, 90, 796–802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Boonyasuppayakorn, S.; Reichert, E.D.; Manzano, M.; Nagarajan, K.; Padmanabhan, R. Amodiaquine, an antimalarial drug, inhibits dengue virus type 2 replication and infectivity. Antivir. Res. 2014, 106, 125–134. [Google Scholar] [CrossRef] [PubMed]
- Nelson, E.A.; Dyall, J.; Hoenen, T.; Barnes, A.B.; Zhou, H.; Liang, J.Y.; Michelotti, J.; Dewey, W.H.; Dewald, L.E.; Bennett, R.S.; et al. The phosphatidylinositol-3-phosphate 5-kinase inhibitor apilimod blocks filoviral entry and infection. PLoS Negl. Trop. Dis. 2017, 11, e0005540. [Google Scholar] [CrossRef] [PubMed]
- Hulseberg, C.E.; Fénéant, L.; Wijs, K.M.S.-D.; Kessler, N.P.; Nelson, E.A.; Shoemaker, C.J.; Schmaljohn, C.S.; Polyak, S.J.; White, J.M. Arbidol and other low-molecular-weight drugs that inhibit Lassa and Ebola viruses. J. Virol. 2019, 93, e02185-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kang, Y.L.; Chou, Y.Y.; Rothlauf, P.W.; Liu, Z.; Soh, T.K.; Cureton, D.; Case, J.B.; Chen, R.E.; Diamond, M.S.; Whelan, S.P.J.; et al. Inhibition of PIKfyve kinase prevents infection by Zaire ebolavirus and SARS-CoV-2. Proc. Natl. Acad. Sci. USA 2020, 117, 20803–20813. [Google Scholar] [CrossRef]
- Dittmar, M.; Lee, J.S.; Whig, K.; Segrist, E.; Li, M.; Jurado, K.; Samby, K.; Ramage, H.; Schultz, D.; Cherry, S. Drug repurposing screens reveal FDA approved drugs active against SARS-CoV-2. bioRxiv 2020. [Google Scholar] [CrossRef]
- Kang, S.; Shields, A.R.; Jupatanakul, N.; Dimopoulos, G. Suppressing dengue-2 infection by chemical inhibition of Aedes aegypti host factors. PLoS Negl. Trop. Dis. 2014, 8, e3084. [Google Scholar] [CrossRef] [Green Version]
- Sabino, C.; Basic, M.; Bender, D.; Elgner, F.; Himmelsbach, K.; Hildt, E. Bafilomycin A1 and U18666A efficiently impair ZIKV infection. Viruses 2019, 11, 524. [Google Scholar] [CrossRef] [Green Version]
- Nawa, M. Japanese encephalitis virus infection in Vero cells: The involvement of intracellular acidic vesicles in the early phase of viral infection was observed with the treatment of a specific vacuolar type H+-ATPase inhibitor, bafilomycin A1. Microbiol. Immunol. 1997, 41, 537–543. [Google Scholar] [CrossRef] [PubMed]
- Yang, C.-C.; Hu, H.-S.; Lin, H.-M.; Wu, P.-S.; Wu, R.-H.; Tian, J.-N.; Wu, S.-H.; Tsou, L.K.; Song, J.-S.; Chen, H.-W.; et al. A novel flavivirus entry inhibitor, BP34610, discovered through high-throughput screening with dengue reporter viruses. Antivir. Res. 2019, 172, 104636. [Google Scholar] [CrossRef] [PubMed]
- Li, C.; Zhu, X.; Ji, X.; Quanquin, N.; Deng, Y.-Q.; Tian, M.; Aliyari, R.; Zuo, X.; Yuan, L.; Afridi, S.K.; et al. Chloroquine, an FDA-approved drug, prevents Zika virus infection and its associated congenital microcephaly in mice. EBioMedicine 2017, 24, 189–194. [Google Scholar] [CrossRef] [PubMed]
- Shiryaev, S.A.; Mesci, P.; Pinto, A.; Fernandes, I.; Sheets, N.; Shresta, S.; Farhy, C.; Huang, C.-T.; Strongin, A.Y.; Muotri, A.R.; et al. Repurposing of the anti-malaria drug chloroquine for Zika virus treatment and prophylaxis. Sci. Rep. 2017, 7, 15771. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Del Vecchio, R.; Higa, L.M.; Pezzuto, P.; Valadão, A.L.; Garcez, P.P.; Monteiro, F.L.; Loiola, E.C.; Dias, A.A.; Silva, F.J.M.; Aliota, M.; et al. Chloroquine, an endocytosis blocking agent, inhibits Zika virus infection in different cell models. Viruses 2016, 8, 322. [Google Scholar] [CrossRef] [Green Version]
- Han, Y.; Pham, H.T.; Xu, H.; Quan, Y.; Mesplèdes, T. Antimalarial drugs and their metabolites are potent Zika virus inhibitors. J. Med Virol. 2019, 91, 1182–1190. [Google Scholar] [CrossRef]
- Li, M.; Zhang, D.; Li, C.; Zheng, Z.; Fu, M.; Ni, F.; Liu, Y.; Du, T.; Wang, H.; Griffin, G.E.; et al. Characterization of Zika virus endocytic pathways in human glioblastoma cells. Front. Microbiol. 2020, 11, 242. [Google Scholar] [CrossRef]
- Wang, C.-Y.; Hour, M.-J.; Lai, H.-C.; Chen, C.-H.; Chang, P.-J.; Huang, S.-H.; Lin, C.-W. Epigallocatechin-3-gallate inhibits the early stages of Japanese encephalitis virus infection. Virus Res. 2018, 253, 140–146. [Google Scholar] [CrossRef]
- Carneiro, B.M.; Batista, M.N.; Braga, A.C.S.; Nogueira, M.L.; Rahal, P. The green tea molecule EGCG inhibits Zika virus entry. Virology 2016, 496, 215–218. [Google Scholar] [CrossRef]
- Raekiansyah, M.; Buerano, C.C.; Luz, M.A.D.; Morita, K. Inhibitory effect of the green tea molecule EGCG against dengue virus infection. Arch. Virol. 2018, 163, 1649–1655. [Google Scholar] [CrossRef]
- Wang, L.-F.; Lin, Y.-S.; Huang, N.-C.; Mayumi, M.; Tsai, W.-L.; Chen, J.-J.; Kubota, T.; Matsuoka, M.; Chen, S.-R.; Yang, C.-S.; et al. Hydroxychloroquine-inhibited dengue virus is associated with host defense machinery. J. Interf. Cytokine Res. 2015, 35, 143–156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gaudry, A.; Bos, S.; Viranaicken, W.; Roche, M.; Krejbich-Trotot, P.; Gadea, G.; Desprès, P.; el Kalamouni, C. The flavonoid isoquercitrin precludes initiation of Zika virus infection in human cells. Int. J. Mol. Sci. 2018, 19, 1093. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mazzon, M.; Ortega-Prieto, A.M.; Imrie, D.; Luft, C.; Hess, L.; Czieso, S.; Grove, J.; Skelton, J.K.; Farleigh, L.; Bugert, J.J.; et al. Identification of broad-spectrum antiviral compounds by targeting viral entry. Viruses 2019, 11, 176. [Google Scholar] [CrossRef] [PubMed]
- Xu, M.; Lee, E.M.; Wen, Z.; Cheng, Y.; Huang, W.-K.; Qian, X.; Tcw, J.; Kouznetsova, J.; Ogden, S.C.; Hammack, C.; et al. Identification of small-molecule inhibitors of Zika virus infection and induced neural cell death via a drug repurposing screen. Nat. Med. 2016, 22, 1101–1107. [Google Scholar] [CrossRef] [PubMed]
- Kao, J.-C.; Huangfu, W.-C.; Tsai, T.-T.; Ho, M.-R.; Jhan, M.-K.; Shen, T.-J.; Tseng, P.-C.; Wang, Y.-T.; Lin, C.-F. The antiparasitic drug niclosamide inhibits dengue virus infection by interfering with endosomal acidification independent of mTOR. PLoS Negl. Trop. Dis. 2018, 12, e0006715. [Google Scholar] [CrossRef] [PubMed]
- Li, Z.; Brecher, M.; Deng, Y.-Q.; Zhang, J.; Sakamuru, S.; Liu, B.; Huang, R.; A Koetzner, C.; Allen, C.A.; Jones, S.A.; et al. Existing drugs as broad-spectrum and potent inhibitors for Zika virus by targeting NS2B-NS3 interaction. Cell Res. 2017, 27, 1046–1064. [Google Scholar] [CrossRef] [Green Version]
- Fang, J.; Sun, L.; Peng, G.; Xu, J.; Zhou, R.; Cao, S.; Chen, H.; Song, Y. Identification of three antiviral inhibitors against Japanese encephalitis virus from library of pharmacologically active compounds 1280. PLoS ONE 2013, 8, e78425. [Google Scholar] [CrossRef]
- Garrison, A.R.; Radoshitzky, S.R.; Kota, K.P.; Pegoraro, G.; Ruthel, G.; Kuhn, J.H.; Altamura, L.A.; Kwilas, S.; Bavari, S.; Haucke, V.; et al. Crimean–Congo hemorrhagic fever virus utilizes a clathrin- and early endosome-dependent entry pathway. Virology 2013, 444, 45–54. [Google Scholar] [CrossRef] [Green Version]
- Archer, M.A.; Brechtel, T.M.; Davis, L.E.; Parmar, R.C.; Hasan, M.H.; Tandon, R. Inhibition of endocytic pathways impacts cytomegalovirus maturation. Sci. Rep. 2017, 7, 46069. [Google Scholar] [CrossRef] [Green Version]
- Herrscher, C.; Pastor, F.; Burlaud-Gaillard, J.; Dumans, A.; Seigneuret, F.; Moreau, A.; Patient, R.; Eymieux, S.; de Rocquigny, H.; Hourioux, C.; et al. Hepatitis B virus entry into HepG2-NTCP cells requires clathrin-mediated endocytosis. Cell. Microbiol. 2020, 22, e13205. [Google Scholar] [CrossRef]
- Von Kleist, L.; Stahlschmidt, W.; Bulut, H.; Gromova, K.; Puchkov, D.; Robertson, M.J.; MacGregor, K.A.; Tomilin, N.; Pechstein, A.; Chau, N.; et al. Role of the clathrin terminal domain in regulating coated pit dynamics revealed by small molecule inhibition. Cell 2011, 146, 471–484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Basic, M.; Elgner, F.; Bender, D.; Sabino, C.; Herrlein, M.-L.; Roth, H.; Glitscher, M.; Fath, A.; Kerl, T.; Schmalz, H.-G.; et al. A synthetic derivative of houttuynoid B prevents cell entry of Zika virus. Antivir. Res. 2019, 172, 104644. [Google Scholar] [CrossRef] [PubMed]
- Macia, E.; Ehrlich, M.; Massol, R.; Boucrot, E.; Brunner, C.; Kirchhausen, T. Dynasore, a cell-permeable inhibitor of dynamin. Dev. Cell 2006, 10, 839–850. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gayle, S.; Landrette, S.; Beeharry, N.; Conrad, C.; Hernandez, M.; Beckett, P.; Ferguson, S.M.; Mandelkern, T.; Zheng, M.; Xu, T.; et al. Identification of apilimod as a first-in-class PIKfyve kinase inhibitor for treatment of B-cell non-Hodgkin lymphoma. Blood 2017, 129, 1768–1778. [Google Scholar] [CrossRef]
- Sbrissa, D.; Ikonomov, O.C.; Shisheva, A. PIKfyve, a mammalian ortholog of yeast Fab1p lipid kinase, synthesizes 5-phosphoinositides. J. Biol. Chem. 1999, 274, 21589–21597. [Google Scholar] [CrossRef] [Green Version]
- Ikonomov, O.C.; Sbrissa, D.; Mlak, K.; Deeb, R.; Fligger, J.; Soans, A.; Finley, R.L.; Shisheva, A. Active PIKfyve associates with and promotes the membrane attachment of the late endosome-to-trans-Golgi network transport factor Rab9 effector p40. J. Biol. Chem. 2003, 278, 50863–50871. [Google Scholar] [CrossRef] [Green Version]
- Rutherford, A.C.; Traer, C.; Wassmer, T.; Pattni, K.; Bujny, M.V.; Carlton, J.G.; Stenmark, H.; Cullen, P.J. The mammalian phosphatidylinositol 3-phosphate 5-kinase (PIKfyve) regulates endosome-to-TGN retrograde transport. J. Cell Sci. 2006, 119, 3944–3957. [Google Scholar] [CrossRef] [Green Version]
- Burakoff, R.; Barish, C.F.; Riff, D.; Pruitt, R.; Chey, W.Y.; Farraye, F.A.; Shafran, I.; Katz, S.; Krone, C.L.; Vander-Vliet, M.; et al. A phase 1/2A trial of STA 5326, an oral interleukin-12/23 inhibitor, in patients with active moderate to severe Crohn’s disease. Inflamm. Bowel Dis. 2006, 12, 558–565. [Google Scholar] [CrossRef]
- Sands, B.E.; Jacobson, E.W.; Sylwestrowicz, T.; Younes, Z.; Dryden, G.; Fedorak, R.; Greenbloom, S. Randomized, double-blind, placebo-controlled trial of the oral interleukin-12/23 inhibitor apilimod mesylate for treatment of active Crohn’s disease. Inflamm. Bowel Dis. 2010, 16, 1209–1218. [Google Scholar] [CrossRef]
- Krausz, S.; Boumans, M.J.H.; Gerlag, D.M.; Lufkin, J.; van Kuijk, A.W.R.; Bakker, A.; de Boer, M.; Lodde, B.M.; Reedquist, K.A.; Jacobson, E.W.; et al. Brief Report: A phase IIa, randomized, double-blind, placebo-controlled trial of apilimod mesylate, an interleukin-12/interleukin-23 inhibitor, in patients with rheumatoid arthritis. Arthritis Rheum. 2012, 64, 1750–1755. [Google Scholar] [CrossRef] [Green Version]
- Sharma, N.; Murali, A.; Singh, S.K.; Giri, R. Epigallocatechin gallate, an active green tea compound inhibits the Zika virus entry into host cells via binding the envelope protein. Int. J. Biol. Macromol. 2017, 104, 1046–1054. [Google Scholar] [CrossRef] [PubMed]
- Singh, P.; Chakraborty, P.; He, D.-H.; Mergia, A. Extract prepared from the leaves of Ocimum basilicum inhibits the entry of Zika virus. Acta Virol. 2019, 63, 316–321. [Google Scholar] [CrossRef] [PubMed]
- Talarico, L.B.; Pujol, C.A.; Zibetti, R.G.M.; Faría, P.C.S.; Noseda, M.D.; Duarte, M.E.R.; Damonte, E.B. The antiviral activity of sulfated polysaccharides against dengue virus is dependent on virus serotype and host cell. Antivir. Res. 2005, 66, 103–110. [Google Scholar] [CrossRef] [PubMed]
- Rees, C.R.; Costin, J.M.; Fink, R.C.; McMichael, M.; A Fontaine, K.; Isern, S.; Michael, S.F. In vitro inhibition of dengue virus entry by p-sulfoxy-cinnamic acid and structurally related combinatorial chemistries. Antivir. Res. 2008, 80, 135–142. [Google Scholar] [CrossRef]
- Fang, C.-Y.; Chen, S.-J.; Wu, H.-N.; Ping, Y.; Lin, C.-Y.; Shiuan, D.; Chen, C.-L.; Lee, Y.; Huang, K.-J. Honokiol, a Lignan Biphenol Derived from the Magnolia Tree, Inhibits Dengue Virus Type 2 Infection. Viruses 2015, 7, 4894–4910. [Google Scholar] [CrossRef] [Green Version]
- Vázquez-Calvo, Á.; de Oya, N.J.; Martín-Acebes, M.A.; Garcia-Moruno, E.; Saiz, J.-C. Antiviral properties of the natural polyphenols delphinidin and epigallocatechin gallate against the flaviviruses West Nile virus, Zika virus, and dengue virus. Front. Microbiol. 2017, 8, 1314. [Google Scholar] [CrossRef]
- Park, S.J.; Park, Y.J.; Shin, J.H.; Kim, E.S.; Hwang, J.J.; Jin, N.-H.; Kim, J.C.; Cho, N.-H. A receptor tyrosine kinase inhibitor, Tyrphostin A9 induces cancer cell death through Drp1 dependent mitochondria fragmentation. Biochem. Biophys. Res. Commun. 2011, 408, 465–470. [Google Scholar] [CrossRef]
- Banbury, D.N.; Oakley, J.D.; Sessions, R.B.; Banting, G. Tyrphostin A23 inhibits internalization of the transferrin receptor by perturbing the interaction between tyrosine motifs and the medium chain subunit of the AP-2 adaptor complex. J. Biol. Chem. 2003, 278, 12022–12028. [Google Scholar] [CrossRef] [Green Version]
- Savarino, A.; Boelaert, J.R.; Cassone, A.; Majori, G.; Cauda, R. Effects of chloroquine on viral infections: An old drug against today’s diseases? Lancet Infect. Dis. 2003, 3, 722–727. [Google Scholar] [CrossRef]
- Al-Bari, A.A. Targeting endosomal acidification by chloroquine analogs as a promising strategy for the treatment of emerging viral diseases. Pharmacol. Res. Perspect. 2017, 5, e00293. [Google Scholar] [CrossRef]
- Farias, K.J.S.; Machado, P.R.L.; Muniz, J.A.P.C.; Imbeloni, A.A.; da Fonseca, B.A.L. Antiviral activity of chloroquine against dengue virus type 2 replication in Aotus monkeys. Viral Immunol. 2015, 28, 161–169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Miller, B.R.; Tsai, T.F.; Mitchell, C.J. Aedes aegypti and yellow fever virus: The effect of chloroquine on infection and transmission rates. Trans. R. Soc. Trop. Med. Hyg. 1987, 81, 111–112. [Google Scholar] [CrossRef]
- Cao, B.; Sheth, M.N.; Mysorekar, I. To Zika and destroy: An antimalarial drug protects fetuses from Zika infection. Future Microbiol. 2018, 13, 137–139. [Google Scholar] [CrossRef] [PubMed]
- Farias, K.J.S.; Machado, P.R.L.; Junior, R.F.D.A.; de Aquino, A.A.; da Fonseca, B.A.L. Chloroquine interferes with dengue-2 virus replication in U937 cells. Microbiol. Immunol. 2014, 58, 318–326. [Google Scholar] [CrossRef] [PubMed]
- Brandriss, M.W.; Schlesinger, J.J. Antibody-mediated infection of P388D1 cells with 17D yellow fever virus: Effects of chloroquine and cytochalasin B. J. Gen. Virol. 1984, 65, 791–794. [Google Scholar] [CrossRef] [PubMed]
- Fantini, J.; di Scala, C.; Chahinian, H.; Yahi, N. Structural and molecular modelling studies reveal a new mechanism of action of chloroquine and hydroxychloroquine against SARS-CoV-2 infection. Int. J. Antimicrob. Agents 2020, 55, 105960. [Google Scholar] [CrossRef] [PubMed]
- Randolph, V.B.; Winkler, G.; Stollar, V. Acidotropic amines inhibit proteolytic processing of flavivirus prM protein. Virology 1990, 174, 450–458. [Google Scholar] [CrossRef]
- Kumar, A.; Liang, B.; Aarthy, M.; Singh, S.K.; Garg, N.; Mysorekar, I.U.; Giri, R. Hydroxychloroquine inhibits Zika virus NS2B-NS3 protease. ACS Omega 2018, 3, 18132–18141. [Google Scholar] [CrossRef] [Green Version]
- Schrezenmeier, E.; Dörner, T. Mechanisms of action of hydroxychloroquine and chloroquine: Implications for rheumatology. Nat. Rev. Rheumatol. 2020, 16, 155–166. [Google Scholar] [CrossRef]
- Marmor, M.F.; Kellner, U.; Lai, T.Y.; Melles, R.B.; Mieler, W.F. Recommendations on screening for chloroquine and hydroxychloroquine retinopathy (2016 Revision). Ophthalmology 2016, 123, 1386–1394. [Google Scholar] [CrossRef] [Green Version]
- Chatre, C.; Roubille, F.; Vernhet, H.; Jorgensen, C.; Pers, Y.-M. Cardiac complications attributed to chloroquine and hydroxychloroquine: A systematic review of the literature. Drug Saf. 2018, 41, 919–931. [Google Scholar] [CrossRef] [PubMed]
- Arnaout, A.; Robertson, S.J.; Pond, G.R.; Lee, H.; Jeong, A.; Ianni, L.; Kroeger, L.; Hilton, J.; Coupland, S.; Gottlieb, C.; et al. A randomized, double-blind, window of opportunity trial evaluating the effects of chloroquine in breast cancer patients. Breast Cancer Res. Treat. 2019, 178, 327–335. [Google Scholar] [CrossRef] [PubMed]
- Pereira, B.B. Challenges and cares to promote rational use of chloroquine and hydroxychloroquine in the management of coronavirus disease 2019 (COVID-19) pandemic: A timely review. J. Toxicol. Environ. Health Part B 2020, 23, 177–181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Borba, M.G.S.; Val, F.F.A.; Sampaio, V.S.; Alexandre, M.A.A.; Melo, G.C.; Brito, M.; Mourao, M.P.G.; Brito-Sousa, J.D.; Baia-da-Silva, D.; Guerra, M.V.F.; et al. Effect of high vs low doses of chloroquine diphosphate as adjunctive therapy for patients hospitalized with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection: A randomized clinical trial. JAMA Netw. Open 2020, 3, e208857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tricou, V.; Minh, N.N.; Van, T.P.; Lee, S.J.; Farrar, J.; Wills, B.; Tran, H.T.; Simmons, C.P. A randomized controlled trial of chloroquine for the treatment of dengue in Vietnamese adults. PLoS Negl. Trop. Dis. 2010, 4, e785. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Borges, M.C.; Castro, L.A.; da Fonseca, B.A.L. Chloroquine use improves dengue-related symptoms. Memór. Inst. Oswaldo Cruz 2013, 108, 596–599. [Google Scholar] [CrossRef]
- Andoh, T.; Kawamata, H.; Umatake, M.; Terasawa, K.; Takegami, T.; Ochiai, H. Effect of bafilomycin A1 on the growth of Japanese encephalitis virus in Vero cells. J. Neurovirol 1998, 4, 627–631. [Google Scholar] [CrossRef]
- Nawa, M. Effects of bafilomycin A1 on Japanese encephalitis virus in C6/36 mosquito cells. Arch. Virol. 1998, 143, 1555–1568. [Google Scholar] [CrossRef]
- Dröse, S.; Altendorf, K. Bafilomycins and concanamycins as inhibitors of V-ATPases and P-ATPases. J. Exp. Biol. 1997, 200, 1–8. [Google Scholar]
- Bowman, E.J.; Siebers, A.; Altendorf, K. Bafilomycins: A class of inhibitors of membrane ATPases from microorganisms, animal cells, and plant cells. Proc. Natl. Acad. Sci. USA 1988, 85, 7972–7976. [Google Scholar] [CrossRef] [Green Version]
- Yoshimori, T.; Yamamoto, A.; Moriyama, Y.; Futai, M.; Tashiro, Y. Bafilomycin A1, a specific inhibitor of vacuolar-type H(+)-ATPase, inhibits acidification and protein degradation in lysosomes of cultured cells. J. Biol. Chem. 1991, 266, 17707–17712. [Google Scholar] [PubMed]
- Furuchi, T.; Aikawa, K.; Arai, H.; Inoue, K. Bafilomycin A1, a specific inhibitor of vacuolar-type H(+)-ATPase, blocks lysosomal cholesterol trafficking in macrophages. J. Biol. Chem. 1993, 268, 27345–27348. [Google Scholar] [PubMed]
- Oda, K.; Nishimura, Y.; Ikehara, Y.; Kato, K. Bafilomycin A1 inhibits the targeting of lysosomal acid hydrolases in cultured hepatocytes. Biochem. Biophys. Res. Commun. 1991, 178, 369–377. [Google Scholar] [CrossRef]
- Mauvezin, C.; Neufeld, T.P. Bafilomycin A1 disrupts autophagic flux by inhibiting both V-ATPase-dependent acidification and Ca-P60A/SERCA-dependent autophagosome-lysosome fusion. Autophagy 2015, 11, 1437–1438. [Google Scholar] [CrossRef] [Green Version]
- Lyszkiewicz, M.; Zietara, N.; Frey, L.; Pannicke, U.; Stern, M.; Liu, Y.; Fan, Y.; Puchalka, J.; Hollizeck, S.; Somekh, I.; et al. Human FCHO1 deficiency reveals role for clathrin-mediated endocytosis in development and function of T cells. Nat. Commun. 2020, 11, 1031. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jung, N.; Haucke, V. Clathrin-mediated endocytosis at synapses. Traffic 2007, 8, 1129–1136. [Google Scholar] [CrossRef] [PubMed]
Name | Mechanism of Action | EC50 | IC50 | Tested Against | Assay | References |
---|---|---|---|---|---|---|
Amodiaquine | Inhibits endosomal acidification Other functions? | n/a | 2.28 μM | ZIKV | Plaque | [224] |
3.07 ± 0.36 μM | n/a | ZIKV | MTT | [225] | ||
4.40 ± 0.51 μM | n/a | Plaque | ||||
7–11 μM | n/a | DENV-2 | Replicon | [226] | ||
2.69 ± 0.40 μM | n/a | Plaque | ||||
17.67 ± 1.74 μM | n/a | DENV-4 | Replicon | |||
8–15 μM | n/a | WNV | ||||
Apilimod * | Inhibition of endosomal trafficking via PI(3)P-5-kinase | n/a | 9–136 nM | EBOV | ELISA | [227] |
n/a | 10–140 nM | MARV | ||||
n/a | 40 nM | LASV | Pseudovirus | [228] | ||
n/a | 30 nM | EBOV | ||||
n/a | 50 nM | EBOV | Pseudovirus | [229] | ||
n/a | 50 nM | SARS-CoV-2 | ||||
n/a | 10 nM | SARS-CoV-2 | Focus-forming | |||
3–7 nM | n/a | SARS-CoV-2 | Infectivity | [230] | ||
Bafilomycin A1 | Inhibition of V-ATPase-mediated acidification | ? | ? | DENV-2, JEV, WNV, YFV, ZIKV | n/a | [155,193,221,231,232,233] |
BP34610 | Targets viral envelope protein | 480 ± 60 nM | n/a | DENV-2 | Plaque | [234] |
Chloroquine | Inhibits endosomal acidification | n/a | 1.7–4.2 μM | ZIKV | qRT-PCR | [235] |
n/a | 10 μM | ZIKV | IF | [236] | ||
4.95 ± 0.47 μM | n/a | ZIKV | MTT | [225] | ||
5.11 ± 0.62 μM | n/a | Plaque | ||||
9.82–14.2 μM | n/a | ZIKV | Cell viability | [237] | ||
5–10 μM | n/a | ZIKV | MTT | [238] | ||
5.12 ± 0.66 μM | n/a | Plaque | ||||
Chlorpromazine | Blocks binding of AP-2 to the plasma membrane, D2R antagonist | ? | ? | DENV, JEV, WNV, ZIKV | n/a | [156,164,166,168,171,175,211,212,213] |
Dynasore | Inhibitor of the GTPase dynamin | ? | ? | DENV, JEV, ZIKV | n/a | [168,171,175,213,239] |
Epigallocatechin gallate | ? | n/a | 7.0 μM | JEV | Plaque | [240] |
n/a | 7.9 μM | Attachment | ||||
n/a | 9.4 μM | Entry | ||||
21.4 μM | n/a | ZIKV | Focus-forming | [241] | ||
14.8 ± 2.6 μM | n/a | DENV-1 | ELISA | [242] | ||
18.0 ± 1.0 μM | n/a | DENV-2 | ||||
11.2 ± 1.7 μM | n/a | DENV-3 | ||||
13.6 ± 0.0 μM | n/a | DENV-4 | ||||
Hydroxychloroquine | Activates host innate immunity, inhibitor of viral protease | n/a | 10–13 μM | DENV-2 | Fluorescent intensity | [243] |
Isoquercitrin | ? | n/a | 10–15 μM | ZIKV | Plaque | [244] |
Nanchangmycin | ? | n/a | 100–400 nM | ZIKV | Infectivity | [133] |
n/a | 158 nM | WNV | Infectivity | [153] | ||
Niclosamide | Prevents endosomal acidification | n/a | 15 μM | DENV-1 | Infectivity | [245] |
n/a | 400 nM | DENV-2 | ||||
n/a | 1.6 μM | DENV-3 | ||||
n/a | 700 nM | ZIKV | ||||
n/a | 220–280 nM | ZIKV | Focus-forming | [246] | ||
10 μM | n/a | DENV-2 | Plaque | [247] | ||
480 ± 60 nM | n/a | ZIKV | Plaque-reduction | [248] | ||
550 ± 50 nM | n/a | DENV-2 | ||||
540 ± 170 nM | n/a | WNV | ||||
840 ± 20 nM | n/a | YFV | ||||
1.02 ± 80 μM | n/a | JEV | ||||
5.80 μM | n/a | JEV | Plaque-reduction | [249] | ||
Pitstops * | Block ligand association with clathrin terminal domain | ? | ? | CCHFV, HCMV, HBV, HIV | n/a | [250,251,252,253] |
Prochlorperazine | Interferes with clathrin associated pathways, D2R antagonist | 88 nM | n/a | DENV-2 | Plaque | [220] |
137 nM | n/a | |||||
TK1023 | ? | 1.55–1.68 μM | n/a | ZIKV | Plaque | [254] |
Tyrphostin A9 | RTK inhibitor, inhibitor of AP-2 complexes? | n/a | 1.4 μM | DENV-1 | Infectivity | [245] |
n/a | 400 nM | DENV-2 | ||||
n/a | 1.2 μM | DENV-3 | ||||
n/a | 300 nM | YFV | ||||
n/a | 300 nM | ZIKV |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Carro, S.D.; Cherry, S. Beyond the Surface: Endocytosis of Mosquito-Borne Flaviviruses. Viruses 2021, 13, 13. https://doi.org/10.3390/v13010013
Carro SD, Cherry S. Beyond the Surface: Endocytosis of Mosquito-Borne Flaviviruses. Viruses. 2021; 13(1):13. https://doi.org/10.3390/v13010013
Chicago/Turabian StyleCarro, Stephen D., and Sara Cherry. 2021. "Beyond the Surface: Endocytosis of Mosquito-Borne Flaviviruses" Viruses 13, no. 1: 13. https://doi.org/10.3390/v13010013